You are on page 1of 13

Leading Edge

Review

Innate Lymphoid Cells: 10 Years On


Eric Vivier,1,2,* David Artis,3 Marco Colonna,4 Andreas Diefenbach,5,6,7 James P. Di Santo,8,9 Gérard Eberl,10,11
Shigeo Koyasu,12,13 Richard M. Locksley,14 Andrew N.J. McKenzie,15 Reina E. Mebius,16 Fiona Powrie,17
and Hergen Spits18
1Innate Pharma Research Labs, Innate Pharma, Marseille, France
2Aix Marseille University, CNRS, INSERM, APHM, CIML, Hôpital de la Timone, Immunologie, Marseille-Immunopole, Marseille, France
3Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine and Department of

Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
4Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
5Laboratory of Innate Immunity, Department of Microbiology and Infection Immunology, Charité – Universitätsmedizin, Berlin, Germany
6Berlin Institute of Health, Berlin, Germany
7Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
8Innate Immunity Unit, Institut Pasteur, Paris, France
9Inserm U1223, Paris, France
10Microenvironment & Immunity Unit, Institut Pasteur, Paris, France
11INSERM U1224, Paris, France
12Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
13Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
14HHMI and Department of Medicine, University of California San Francisco, San Francisco, CA, USA
15Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
16Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, the Netherlands
17Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
18Department of Experimental Immunology Academic Medical Center of the University of Amsterdam, Amsterdam, the Netherlands

*Correspondence: vivier@ciml.univ-mrs.fr
https://doi.org/10.1016/j.cell.2018.07.017

Innate lymphoid cells (ILCs) are lymphocytes that do not express the type of diversified antigen
receptors expressed on T cells and B cells. ILCs are largely tissue-resident cells and are deeply
integrated into the fabric of tissues. The discovery and investigation of ILCs over the past decade
has changed our perception of immune regulation and how the immune system contributes to the
maintenance of tissue homeostasis. We now know that cytokine-producing ILCs contribute to mul-
tiple immune pathways by, for example, sustaining appropriate immune responses to commensals
and pathogens at mucosal barriers, potentiating adaptive immunity, and regulating tissue inflam-
mation. Critically, the biology of ILCs also extends beyond classical immunology to metabolic
homeostasis, tissue remodeling, and dialog with the nervous system. The last 10 years have also
contributed to our greater understanding of the transcriptional networks that regulate lymphocyte
commitment and delineation. This, in conjunction with the recent advances in our understanding of
the influence of local tissue microenvironments on the plasticity and function of ILCs, has led to a
re-evaluation of their existing categorization. In this review, we distill the advances in ILC biology
over the past decade to refine the nomenclature of ILCs and highlight the importance of ILCs in
tissue homeostasis, morphogenesis, metabolism, repair, and regeneration.

Innate lymphoid cells (ILCs), which lack adaptive antigen recep- bacteria and fungi. Myeloid and non-hematopoietic cells instruct
tors generated by the recombination of genetic elements, are the the ILCs and T cells, which belong to the lymphoid lineage, about
innate counterparts of T lymphocytes (Spits et al., 2013; Eberl the type of threat they will confront. The ILCs and T cells react by
et al., 2015; Artis and Spits, 2015). ILC1s, ILC2s, and ILC3s mirror providing positive and negative feedbacks and through immune
CD4+ T helper (Th)1, Th2, and Th17 cells, respectively, in terms of regulatory and effector functions. This initial description of a
function, whereas natural killer (NK) cells mirror the functions of simple phenotypic and functional trichotomy has been greatly
CD8+ cytotoxic T cells. ILCs and T cells play key roles in orches- enriched and has become much more complex since the discov-
trating the most appropriate immune response to the threat faced ery of these subsets 10 years ago. This review aims to clarify this
by the individual. ILC1s and Th1 cells react to intracellular patho- complexity and to propose an updated and operational classifi-
gens, such as viruses, and to tumors; ILC2s and Th2 cells cation and nomenclature for ILCs.
respond to large extracellular parasites and allergens; and ILCs act early in the immune response by reacting promptly to
ILC3s and Th17 cells combat extracellular microbes, such as signals, or inducer cytokines, expressed by tissue-resident cells.

1054 Cell 174, August 23, 2018 ª 2018 Elsevier Inc.


By contrast, the T cell response takes several days, as these The presence of ILCs in non-lymphoid tissues is ensured by
cells must undergo clonal expansion to become operational the recruitment of ILC progenitors from the blood, and main-
and develop antigen-specific memory. Nevertheless, several tained by the expression of local survival factors, such as IL-7.
types of T cells are ‘‘pre-programmed’’ and function like innate ILCs have slightly different phenotypes in different tissues, and
cells in certain tissues. These cells include invariant NKT cells, different local microenvironments may induce ILC progenitors
mucosa-associated invariant T (MAIT) cells, diverse subsets of from the same subset to adopt tissue-specific expression pat-
gd T cells and resident memory T (TRM) cells selected and terns. Most ILCs are tissue-resident cells (Gasteiger et al.,
expanded during earlier encounters with antigen (Mueller and 2015), but ILC3s can migrate from the intestinal lamina propria
Mackay, 2016). A few days after the initiation of an immune reac- to the draining mesenteric lymph node, and NK cells and inflam-
tion, both ILCs and T cells are active, and they cross-regulate matory ILC2s circulate in the bloodstream.
each other. ILCs can express major histocompatibility complex We first proposed a common nomenclature for ILCs and ILC
(MHC) class II molecules and process antigens, thereby regu- subsets in 2013 (Spits et al., 2013). The use of single-cell RNA
lating the activity of antigen-specific T cells (Oliphant et al., sequencing has led to the identification of more than 50 distinct
2014). In turn, these T cells produce interleukin-2 (IL-2), promot- ILC clusters (in t-distributed stochastic neighbor embedding
ing ILC activities. Both types of cell generate positive-feedback [t-SNE] projection analysis) in different tissues (Gury-BenAri
loops that amplify their responses, but they also compete for et al., 2016). We have re-evaluated the original nomenclature,
the same inducer cytokines and survival factors. Thus, these taking into account the identification of ILC developmental
two types of lymphoid cells mirror one another’s activities only lineages, transcriptional networks coordinating ILC effector func-
partially, thereby ensuring the timely orchestration of the immune tions and phenotypes, activation states, and tissue-specific ad-
response. aptations, and we propose here an updated and comprehensive,
ILCs begin functioning during fetal development, as lymphoid but simple and practical, nomenclature for ILCs. We also provide
tissue-inducer cells (LTi cells) (Mebius et al., 1997). These cells an overview of the newly discovered roles of ILCs in defense,
induce the development of most of the secondary lymphoid or- metabolism, repair and interactions with the nervous system.
gans. They instruct mesenchymal stromal cells to produce the
factors required to attract hematopoietic cells to the developing Nomenclature
lymphoid structure and retain them there. The interaction between The nomenclature for ILCs proposed in 2013 classified these
ILCs and the non-hematopoietic microenvironment is an impor- cells into groups 1, 2, and 3, each of which contained one, two
tant facet of ILC function maintained from birth into adulthood. It or three subsets. The subsets within each group had the same
includes the activation of stromal cells for the recruitment, reten- output, including cytokines in particular, and their development
tion, and activation of lymphocytes, and for their regeneration and function were dependent on the same transcription factors.
and the activation of defensive and anti-apoptotic programs in Group 1 ILCs comprised NK cells and ILC1s. These cells are
epithelial cells. Thus, through their generation early during the for- dependent on the T-box transcription factor Tbet for their devel-
mation of the immune system and their prompt reactivity, ILCs play opment and function, and they produce interferon-gamma
an important role in the crosstalk of lymphoid cells with non-he- (IFN-g). Group 2 contains a single subset, ILC2s, which are
matopoietic cells and in the spatial organization of immunity. dependent on GATA3 and RORa (Wong et al., 2012), and pro-
ILCs are clearly the innate counterpart of T cell effector sub- duce type 2 cytokines, predominantly IL-5 and IL-13. Group 3
sets, but research into ILCs has opened up an unprecedented ILCs include natural cytotoxicity receptor (NCR) ILC3s, NCR+
appreciation of the profound involvement of immune cells in tis- ILC3s, and LTi cells, all of which are dependent on the transcrip-
sue homeostasis, whether morphogenesis, metabolism, regen- tion factor RORgt and can produce IL-17 and/or IL-22.
eration, and growth. ILCs are largely tissue-resident cells and The last decade has provided a wealth of data on the mecha-
are integrated deep into the fabric of tissues, and studies of nisms of development and the molecules involved in the
these cells have revealed ever more intriguing relationships functions of T helper cell subsets, which has improved our un-
with basic developmental and biological processes. ILCs play derstanding of the development of ILC subsets (Eberl et al.,
a key role in homeostasis, due to the rapidity with which they 2015; Artis and Spits, 2015). Within the three initially proposed
react and their presence in normal healthy tissues, including ILC groups, we now appreciate the existence of distinct subsets
the intestine, lung and adipose tissues, in particular. An absence based on developmental trajectories. As such, we now propose
of ILC3s in the intestine can lead to a loss of control over the to classify ILCs into five subsets—NK cells, ILC1s, ILC2s, ILC3s,
symbiotic microbiota. In adipose tissues, ILC2s are involved in and LTi cells—based on their development (Figure 1) and func-
thermogenesis in the context of a cold shock. ILCs are also tion (Figure 2). The ILC nomenclature presented here is approved
involved in tissue tolerance and regeneration in response to by the International Union of Immunological Societies (IUIS).
tissue damage. In the intestine and thymus, ILC3s mediate toler- NK Cells and ILC1s
ance to chemical toxins and irradiation through the activation of NK cells are dedicated cytotoxic cells that circulate in the blood-
epithelial cells, whereas ILC2s produce amphiregulin (AREG), stream (Vivier et al., 2011). They can kill virus-infected normal
a member of the EGF family, which is involved in epithelial and tumor cells. In the mouse thymus, conventional NK cells
cell regulation. However, these functions can also favor the (cNK) coexist with ILC1s that express CD127 (IL-7Ra) and
development of carcinoma during chronic inflammation and GATA3, unlike cNK cells, produce IFN-g and granulocyte-
injury, and increase the severity of inflammation induced by macrophage colony-stimulating factor (GM-CSF) at much higher
pathogens. levels than cNK cells, and depend on the cytokine IL-7 and the

Cell 174, August 23, 2018 1055


Figure 1. Development of ILCs
ILC development, mainly based on mouse ILCs
differentiation paths, is schematized. ILCs develop
from CILPs (common innate lymphoid pro-
genitors), which themselves differentiate from
CLPs (common lymphoid progenitors). CILPs can
differentiate into NK cell precursors (NKP) cells or
into CHILPs (common helper innate lymphoid
progenitors), which themselves give rise to LTiPs
(lymphoid tissue inducer progenitors) and ILCP
(innate lymphoid cell precursors). LTiPs differen-
tiate into LTis and ILCPs into ILC1, ILC2, or ILC3.
Each stage of differentiation is dependent on the
expression of the indicated transcription factors:
NFIL3 (nuclear factor IL-3 induced), Id2 (inhibitor of
DNA binding 2), TOX (thymocyte selection-asso-
ciated high mobility group box protein), TCF-1
(T cell factor 1), ETS1 (avian erythroblastosis virus
E26 homolog-1), GATA3 (GATA binding protein 3),
PLZF (promyelocytic leukemia zinc finger), T-bet
(T-box transcription factor), Eomes (Eomeso-
dermin), RUNX3 (runt-related transcription fac-
tor 3), RORa (RAR-related orphan receptor a),
Bcl11b (B cell lymphoma/leukemia 11B), Gfi1
(growth factor independent 1), RORgt (RAR-
related orphan receptor gt), and AhR (Aryl hydro-
carbon receptor). It has been shown in humans
that ILC1 subsets may originate from precursors
other than ILCPs (Fuchs et al., 2013), but the
identity of which remains unknown at this time.

shown to distinguish ILC1s from NK cells


in mice (Weizman et al., 2017). Another
marker of human NK cells is NKp80,
transcription factor GATA3 for their differentiation (Vosshenrich which is not expressed on ILC1s (Freud et al., 2016). ILC1s are
et al., 2006). ILC1s are generally non-cytotoxic or weakly cyto- tissue-resident cells, whereas NK cells circulate in the blood-
toxic and function as a first line of defense against infections stream (Peng et al., 2013; Gasteiger et al., 2015). In mice,
with viruses and certain bacteria, such as T. gondii (Klose ILC1s are first detected before birth, whereas NK cells emerge
et al., 2014) or C. difficile (Abt et al., 2015). NK cells and ILC1s two to three weeks after birth (Diefenbach et al., 2014). There
have several features in common. Both these cell types produce are also differences in the production of and dependence on
IFN-g as their principal cytokine output and require Tbet for this transcription factors. In mice, ILC1s are strictly dependent on
function. However, they have different developmental paths Tbet, whereas NK cells are present in Tbet-deficient hosts
(Figure 1). In both humans and in mice, NK cells develop from (Daussy et al., 2014). In addition, NK cells require the T-box fac-
a common innate lymphoid progenitor (CILP) via an NK cell pre- tor Eomes, whereas ILC1s can develop in the absence of this
cursor (NKP), whereas ILC1s develop from CILPs via an innate transcription factor. Eomes expression is, therefore, often used
lymphoid cell precursor (ILCP) (Constantinides et al., 2014; Klose as a marker for NK cells, although it can be expressed on a pro-
et al., 2014; Lim et al., 2017; Renoux et al., 2015; Scoville et al., portion of ILC1s.
2016). NK cells and ILC1s are functionally different, as NK cells ILC2s
are dedicated cytotoxic cells strongly expressing perforin, ILC2s are defined by their capacity to produce the type 2 cyto-
whereas ILC1s have low levels of perforin expression. However, kines IL-4, IL-5, and IL-13 (Moro et al., 2010; Neill et al., 2010;
regardless of these developmental and functional differences, Price et al., 2010) and are tissue resident (Gasteiger et al.,
the phenotypic characterization of ILC1s is often problematic. 2015; Moro et al., 2016). They respond to the cytokines IL-25,
ILC1s preferentially express CD49a and TRAIL in both humans TSLP, and IL-33. ILC2s are involved in the innate immune
and mice, but the specificity of these markers is often lost response to parasites, such as the helminth Nippostrongulus
upon cell activation and is tissue-dependent. ILC1s have some brasiliensis. After resolving the infection, ILC2s help to repair
phenotypic markers in common with NK cells and ILC3s, in at damaged tissues by producing AREG (Monticelli et al., 2011,
least some organs. These markers include NK1.1 in mice, and 2015). ILC2s contain larger amounts of the transcription factor
NCRs, such as NKp44 in humans, and NKp46 in both humans GATA3 than the other ILC subsets, and absence of GATA3
and mice (Table 1). In humans, ILC1s express CD127 whereas inhibits the development and function of these cells (Hoyler
highly cytotoxic CD16+CD56+ NK cells do not express this et al., 2012; Klein Wolterink et al., 2013; Furusawa et al., 2013;
marker. However, CD127 may not be an absolute ILC marker Mjösberg et al., 2012). Although a classical marker for tissue-
in humans because a majority of CD16 CD56bright NK cells in pe- resident ILC2s in mice is ST2, a component of the IL-33 receptor,
ripheral blood do express CD127. CD200R expression has been some tissue ILC2s do not express ST2 (e.g., skin), as ST2

1056 Cell 174, August 23, 2018


Figure 2. Immune Function of ILCs
Some of the most well-known immune function of
each ILC subset is shown: NK cells and ILC1s
react to intracellular pathogens, such as viruses,
and to tumors; ILC2s respond to large extracellular
parasites and allergens; ILC3s combat extracel-
lular microbes, such as bacteria and fungi; and
LTis are involved in the formation of secondary
lymphoid structures. For each ILC subset, effector
molecules that can be produced upon activation
are indicated.

difficult to separate on the basis of marker


expression, but neuropilin (NRP-1), which
can be induced on human postnatal
NCR ILC3s may mark the capacity of
these cells to mediate the formation of ter-
tiary lymphoid structures (Robinette et al.,
2015; Shikhagaie et al., 2017). However,
lymphoid tissue-forming activity is not
unique to postnatal ILC3s/LTi cells, as
other lymphotoxin-producing cell types
expression can be impacted by the tissue of origin and the state may also be involved in this activity. LTi cells and ILC3s have
of the microenvironment. ILC2s present in human peripheral different developmental paths (Figure 1).
blood lack ST2 (Bal et al., 2016), but human ILC2s, whether in
the peripheral blood or in tissues, selectively express the chemo- ILC Plasticity: A Key to ILC Heterogeneity
attractant receptor expressed on Th2 cells (CRTH2) and high There is increasing evidence to suggest that like T helper cell
levels of CD161 (Mjösberg et al., 2011). subsets, ILC subsets also display a certain degree of plasticity.
ILC3s Thus, ILC subsets can change their phenotype and functional
ILC3s are abundant at mucosal sites and are involved in the capacities and this requires accessible polarizing signals in the
innate immune response to extracellular bacteria and the tissue in which conversion occurs, together with the expression
containment of intestinal commensals (Cella et al., 2009; Luci of cognate cytokine receptors and key transcription factors in
et al., 2009; Cupedo et al., 2009; Satoh-Takayama et al., the responding ILCs. For example, fate-mapping and adoptive
2008; Buonocore et al., 2010; Sonnenberg et al., 2011, 2012; transfer studies in mice have shown that gut CCR6–NKp46–
Rankin et al., 2016). The predominant homeostatic cytokine ILC3s can convert into IFN-g-producing NK1.1+NKp46+ ILC1s
produced by ILC3s is IL-22, by which they maintain intestinal via a CCR6–NKp46+ intermediate (Vonarbourg et al., 2010).
homeostasis and promote the proliferation of intestinal stem This process requires a decrease in RORgt expression (Vonar-
cells. In addition, ILC3s can regulate adaptive Th17 cell re- bourg et al., 2010), with a parallel increase in T-bet (Sciumé
sponses (Hepworth et al., 2013). In mice, ILC3s are dependent et al., 2012; Klose et al., 2013; Rankin et al., 2013) and Notch
on RORgt for their development and function (Satoh-Takayama signaling (Lee et al., 2011; Viant et al., 2016; Chea et al., 2016).
et al., 2008; Sanos et al., 2009; Buonocore et al., 2010), whereas IL-1b, IL-15 and IL-12 are likely the critical factors because
the IL-17A-producing ILC3 subset (but not the IL-22-producing human IL-22-producing ILC3s can be converted into IFN-g-pro-
subset) is absent in RORC-deficient humans (Lim et al., 2017). ducing ILC1-like cells if cultured in vitro with these cytokines
Two subsets of ILC3 can be distinguished on the basis of the (Cella et al., 2010; Bernink et al., 2015). Recent studies indicate
cell surface expression of NKp46 (in mice) and NKp44 (in hu- that ILC2s can also convert into IFN-g-producing ILC1s both
mans) (Luci et al., 2009; Cupedo et al., 2009; Sanos et al., in vitro and in vivo (Ohne et al., 2016; Bal et al., 2016; Silver
2009), i.e., the NCR+ ILC3. Both mouse and human ILC3s can et al., 2016). T-bet and the IL-12 receptor (IL-12R) must be
produce GM-CSF. In mice, NKp46+ ILC3s are dependent on induced for this conversion to occur (Lim et al., 2016). Further-
Tbet and can produce IFN-g (Sciumé et al., 2012; Klose et al., more, in vivo exposure of ILC2 to the cytokines IL-25 and IL-33
2013; Rankin et al., 2013). These cells develop after birth, in mice appear to elicit distinct functional responses. Systemic
following microbial stimulation. Ex-vivo-isolated human ILC3s injections of IL-25 promote the generation of ILC2s, which pro-
do not produce IFN-g. duce IL-17 in addition to type 2 cytokines and were called inflam-
LTi Cells matory ILC2s (Huang et al., 2015). This transformation is depen-
LTi cells are crucial for the formation of secondary lymph nodes dent on Notch signaling and RORgt expression (Zhang et al.,
and Peyer’s patches during embryonic development, through 2017). IL-25-responding ILC2s arise from resting intestinal
the action of lymphotoxin (Mebius et al., 1997). These cells ex- ILC2s in response to infection with helminths (Huang et al.,
press c-Kit and CCR6, but not NCRs. Like ILC3s, they are strictly 2015; Neill et al., 2010). Unlike IL-33-responsive ILC2s, which
dependent on RORgt. LTi cells and postnatal NCR- ILC3s are are tissue resident (Gasteiger et al., 2015; Moro et al., 2016),

Cell 174, August 23, 2018 1057


Table 1. Main Phenotypic Markers of the Different Subsets of Murine and Human ILCs
Mouse Human
NKp46 NKp46+ NKp44 NKp44+
NK ILC1 ILC2 LTi ILC3 ILC3 NK ILC1 ILC2 LTi ILC3 ILC3
Cell-surface CD45 + + + int + + CD45 + + ++ int + +
molecules CD127 (IL-7Ra) a b
+ ++ W W CD127 (IL-7Ra) –/+ e
+ + + +
CD161 (NK1.1) + + – – – –/+ CD161 (NK1.1) +/ + + +/ + +
ST2 (IL-33R) – nd +/ nd nd nd ST2 (IL-33R) +/ – +/ nd nd –
CD278 (ICOS) W nd ++ nd W W CD278 (ICOS) – nd + nd nd +
IL-17Rb (IL25R) – nd + – – – IL-17Rb (IL25R) – – + – nd –
CD294 (CRTH2) – nd + nd nd nd CD294 (CRTH2) – – + – – –
KLRG1 ++ – + – – – KLRG1 + – + – – –
CD117 (c-kit) - +/ +/ ++ W W CD117 (c-kit) –/W – +/ + + +
c
CD69 + nd nd nd nd CD69 W +/ nd nd nd nd
CD254 (RANKL) nd nd nd + + + CD254 (RANKL) – nd nd + + +
CD196 (CCR6) – nd – + +/ – CD196 (CCR6) – +/ +/ + +/ +/
CD335 (NKp46) + + – – – + CD335 (NKp46) + – – – -/W W/+
d
CD25 (IL-2Ra) –/W + +/ +/ +/ CD25 (IL-2Ra) +/ W + W +/ –/W
MHC-II – – + + + – MHC-II +/ nd +/ nd nd +/
IL23R – – nd + + + IL23R +/ +/ –/W + + +
IL1Rb – + nd + + + IL1R +/ + W + + +
CD122 + + W – – – CD122 + nd nd –/W W W
CD314 (NKG2D) + nd – – – + CD314 (NKG2D) + nd nd – -W –/W
Ly49 +/ +/ – – – – KIR +/ – – – – –
CD94 +/ nd +/ nd – +/ CD94 +/ – – – – –
Perforin + W – – – – Perforin + – – – – –
CD253 (TRAIL) – + nd nd nd nd IL12Rb + + – – – +/
d
Sca-1 (Ly6a) + + – nd + CD194 (CCR4) nd +/ + nd nd nd
CD49d nd nd – nd + + CD56 + – – –/W +/ +/
(integrin a4b7)
a
CD49a + nd nd nd nd CD183 (CXCR3) nd + nd nd nd nd
(integrin a1b1)
CD90 (Thy1) +/ + + + + + CD337 (NKp30) + + + +/ +/ +/
f d
CD160 + nd nd nd nd CD336 (NKp44) – – – – +
g
CD103 – nd nd nd nd CD16 +/ – – – – –
CD200R – + nd nd nd nd NKp80 + – nd nd nd nd
Transcription Tbet + + – – +/ + Tbet + + – – – –
factors Eomes + +/– – – – – Eomes + +/– – – – –
RORgt – – – + + + RORgt – –/W –/W + + +
GATA3 –/W –/W + –/W –/W –/W GATA3 –/W –/W + –/W –/W –/W
AhR – nd nd + + + AhR –/W W + + + +
RORa nd nd + nd nd nd
The expression of certain markers varies according to the organs. int, intermediate expression; W, weak expression; nd, not determined. ‘‘+/ ’’ means
that expression is detected in some, but not all, cells.aNot expressed by NK cells of liver, intestine, skin, uterus, salivary gland, bone marrow, or lymph
nodes. Expressed by NK cells in the thymus and in some spleen populations.
b
Expressed by the majority of ILC1, except for liver populations and intraepithelial gut ILC1.
c
Expressed by NK cells of the intestine, uterus, and thymus, but not the spleen and liver.
d
Expressed by activated cells.
e
Expressed by the majority of ILC1, except for tonsil and intraepithelial ILC1.
f
Not expressed by NK cells of the spleen, liver, uterus, or blood. Expressed by NK cells in the gut.
g
Not expressed by liver or spleen NK cells. Expressed by NK cells in the blood.

1058 Cell 174, August 23, 2018


IL-25-responsive ILC2s circulate in the bloodstream and can the aryl hydrocarbon receptor (AhR) transcription factor in
migrate to effector sites, where they help to protect tissues response to tryptophan metabolites is required to maintain
following infection (Huang et al., 2015). It remains unclear IL-22 expression and intestinal homeostasis (Lee et al., 2011;
whether these ILC2s constitute two different subsets or different Kiss et al., 2011). Consequently, an absence of AhR ligation im-
activation states of the same plastic cell type. Along this line, a pairs immunity to intestinal bacterial infections (Lee et al., 2011;
subset of IL-10-producing ILCs has been identified as regulatory Kiss et al., 2011). Similarly, a deficiency of dietary vitamin A re-
ILCs (ILCregs) (Wang et al., 2017). These cells are claimed to sults in abnormally small numbers of ILC3s, a shortfall in IL-22
develop directly from CHILP rather than from ILCp in an ID3- production and greater susceptibility to gastrointestinal Citro-
dependent manner and to be distinct from other ILC subsets. bacter rodentium infection (Spencer et al., 2014). The vitamin A
As ILC2s can develop into IL-10-producing cells (Gury-BenAri metabolite retinoic acid (RA), which is produced predominantly
et al., 2016; Seehus et al., 2017), further investigation is needed by dendritic cells, at least in the intestinal lamina propria, pro-
to fully understand the nature and potential significance of motes the expression of gut-homing receptors on ILC1s and
ILCregs. Finally, human and mouse cytotoxic NK cells also ac- ILC3s (Kim et al., 2015), and enhances ILC3 function by upregu-
quire ILC1-like features in response to enhanced TGF-b lating RORgt (van de Pavert et al., 2014) and IL-22 (Mielke et al.,
signaling (Gao et al., 2017; Cortez et al., 2017). 2013). Conversely, RA suppresses ILC2 proliferation by downre-
In addition to cytokine receptors and their downstream gulating IL-7Ra (Figure 3A). Accordingly, ILC2 numbers increase
transcription factors, ILC plasticity requires defined chromatin during periods of vitamin A deficiency, increasing the efficiency
regions, such as cis-acting enhancers and silencers of gene of parasitic helminth expulsion (Spencer et al., 2014). It has
expression, to be accessible to transcription factors. Recent been suggested that during periods of micronutrient scarcity,
studies have investigated chromatin status in human and mouse ILC2s are sustained by fatty-acid metabolism, which maintains
ILCs, comparing the results obtained to published findings for the production of IL-13 (Wilhelm et al., 2016). Thus, host dietary
the CD4 T helper cell counterparts of these cells (Koues et al., status can substantially modify the balance of the ILC response
2016; Shih et al., 2016). Accessible chromatin regions were and alter the predisposition to infection, selectively optimizing
assessed by ATAC-seq. It was then determined whether these immune responses to promote survival during periods of nutrient
regions were active or simply poised for activation, by assessing limitation. There are presumably subtle local adjustments to
histone H3-K27 acetylation or the binding of the regulatory factor the balance of available nutrients within tissues, to allow the im-
p300. One distinctive feature of the ILC regulome is that mune system to respond appropriately to the relentless stream
regulatory regions controlling the expression of signature of diverse immune challenges posed by co-infections and the
cytokine genes are already poised or active in ILCs, whereas commensal microbiome. A more thorough understanding of
identical regulatory regions in CD4 T helper cells become acces- these complex processes might facilitate the therapeutic manip-
sible only after activation by cytokine signals or infection (Shih ulation of immune responses through dietary refinements and
et al., 2016). These results suggest that ILC regulomes are nutrient supplementation.
more prone to dynamic changes in response to the microenvi- ILCs in Metabolic Tissues
ronment. Recently it was shown that transition of ILC2s to Adipose tissues contain many types of immune cells, which
ILC1s by IL-1b and IL-12 is marked by an atypical chromosomal reside among the adipocytes. Recent studies have demon-
landscape in which the locus encoding IFN-g and the locus strated that ILC2s contribute to the maintenance of metabolic
encoding IL-5 and IL-13 are simultaneously accessible (Ohne homeostasis by supporting the type-2 immune environment
et al., 2016). characteristic of the adipose tissue of lean individuals. Dysregu-
It is becoming clear that this malleability of ILCs directs the lation of this pathway leads to obesity with persistent, low-grade
heterogeneity of these cells in the tissues and this may be impor- type 1 inflammation. ILC2s reside constitutively in visceral
tant for effective immune responses and may also play roles adipose tissue (VAT), where they are maintained by the expres-
in inflammatory diseases, such as Crohn’s disease (Bernink sion of IL-33 (Brestoff et al., 2015). Here, they produce IL-5 to
et al., 2015) and chronic obstructive pulmonary disease (Silver sustain eosinophils, and IL-13 to polarize M2 macrophages
et al., 2016; Bal et al., 2016). However, the exact functional and regulate adiposity and insulin resistance (Brestoff et al.,
impact of ILC plasticity is still incompletely understood and re- 2015; Molofsky et al., 2013) (Figure 3A). ILC2s appear to regulate
mains an active area of research. adiposity and caloric expenditure through several different
mechanisms. For example, their production of IL-4 and IL-13 is
ILCs and Metabolism reported to induce pre-adipocyte differentiation into beige adi-
There is increasing evidence to link ILCs with metabolic homeo- pocytes (Lee et al., 2015). Additionally, ILC2s have been shown
stasis, dietary stress and obesity. Malnutrition and gluttony can to express the endopeptidase proprotein convertase subtilisin/
lead to dysregulated ILC-mediated responses both through the kexin 1 (Pcsk1), which can process the proenkephalin A
availability of specific dietary nutrients, and by altering immune produced by ILC2s to yield methionine-enkephalin (MetEnk)
homeostasis as adipose tissue energy stores expand and con- peptides, which induce UCP1 in adipocytes (Brestoff et al.,
tract with nutrient availability. 2015). The treatment of mice with exogenous MetEnk induces
Regulation of ILCs by Dietary Nutrients beiging, thus increasing caloric consumption, although the phys-
The ratios of ILC populations, and their activation states, can be iological contribution of ILC2-derived MetEnk remains unclear
profoundly influenced by the availability of environmental micro- (Figure 3A). Furthermore, in addition to the roles of ILC2s in
nutrients and diet-derived metabolites (Figure 3A). Signaling via VAT, IL-33-stimulated ILC2s in the pancreas can drive dendritic

Cell 174, August 23, 2018 1059


Figure 3. Functions of ILCs beyond Immunity
(A–C) The regulation of ILC function by nutrients (A),
their role in tissue repair (B), and the links between
ILCs and the nervous system (C) are schematized.

to maintain the delicate immune balance


within adipose tissue and are dysregu-
lated in obesity.

ILCs in Tissue Remodeling and


Repair
ILCs are largely tissue-resident cells
(Gasteiger et al., 2015). This remarkable
tissue dwelling property of ILC provides
an interesting framework for understand-
ing the biology of ILCs that seem to fulfill
functions in organ homeostasis and
repair, not conventionally assigned to
the immune system. Tissue residency
does not preclude local migration how-
ever, and ILC3s have been observed to
crawl into and out of cryptopatches at
steady state, with increased egress into
the tissue during the onset of inflamma-
tion (Pearson et al., 2016). ILCs may
follow local chemokine gradients and,
in this manner, migrate to local inflamma-
tory foci, returning to newly formed
lymphoid structures at the end of an infec-
tion. One puzzle has been how the low
abundance of ILCs in certain tissues
can exert any effect in vivo. Localization
and concentration within immune clusters
and local migration in response to envi-
ronmental cues may allow an exponential
increase in the potency of ILC-secreted
cytokines at sites of tissue damage or
infection.
cells to secrete retinoic acid, which promotes the secretion of In addition to migration within adult tissues, LTi cells, that are
insulin by b cells and glucose regulation (Dalmas et al., 2017). required for lymph node and Peyer’s patch formation, actively
During the maintenance of a lean state, heterogeneous ILC1/ migrate to the lymphoid organ primordia to promote lymphoid
NK populations may also be found in the VAT, in which they are tissue development (Veiga-Fernandes et al., 2007). It is impor-
thought to help maintain homeostasis. It has been suggested tant to note that ILCs are therefore populating organs and tissues
that this may involve the targeted cytotoxic-killing of adipose often early in ontogeny. LTi cells seed tissues already early
tissue macrophages, although, counter-intuitively, the target- during embryonal development (Mebius et al., 1997). The inter-
ing of beneficial M2 macrophages has also been reported dependency of the co-development of organs and components
(Boulenouar et al., 2017), and modulations in M2 macrophages of the innate immune system has not been addressed in much
have not been reported in lean ILC1-deficient mice. With the detail and will be an important trajectory for future research.
onset of obesity, there is a shift toward an inflammatory envi- The deep rooting of ILCs in tissues has led to a new appreciation
ronment in which IFN-g-producing ILC1/NK cells accumulate of unsuspected roles of components of the innate immune
in the VAT and prime M1 pro-inflammatory macrophage polar- system in basic developmental and physiological processes
ization, promoting insulin resistance (Lee et al., 2016; O’Sulli- such as tissue homeostasis, morphogenesis, growth, and
van et al., 2016; Wensveen et al., 2015) (Figure 3A). regeneration. On these aspects, ILCs and tissue-resident mac-
The extent to which ILCs sense and orchestrate changes in the rophages might share potential similarities, as they both develop
adipose tissues remains unclear, as does the extent to which in the mouse, both duringembryonic life and home in tissues
their activity simply reinforces and stabilizes established immu- where they can proliferate and differentiate into mature effector
noregulatory pathways. Nevertheless, ILC subsets clearly help cells (Bando et al., 2015).

1060 Cell 174, August 23, 2018


ILC2s, Tissue Repair, Fibrosis, and Tissue Remodeling and engulf and digest dying cells and extracellular matrix.
for Helminth Expulsion Such responses come therefore at a cost for the host’s affected
Type 2 immunity has been largely discussed in the context of im- tissue, and must be limited in time by repair responses that are
munity to worm infections and of pathogenesis of allergies. It has coordinated by type 2 responses. Nevertheless, in order to be re-
been proposed that a fundamental property of type 2 immunity is paired, a tissue has to be cleansed of microbes, dying cells and
to deal with organisms too large to ingest, break down and digest debris, and therefore, destructive responses that involve ILC1s
(Palm et al., 2012). Therefore, they need to be expelled across and ILC3s are necessary to proceed to repair before the tissue
barrier surfaces, a process requiring extensive tissue remodeling. can return to homeostasis (Figure 3B).
Recently a new regulatory loop for ILC2 function has been found Paradoxically at first sight, type 3 responses play an important
that is required to expel worms in process referred to as ‘‘weep- role, not in tissue repair, but in protection from tissue damage. In
and-sweep.’’ In the intestine, a secretory lineage of epithelial particular, epithelial barriers are reinforced by type 3 responses
cells, tuft cells or brush cells, secrete high levels of IL-25 following in order to resist damage by microbes. IL-22, produced mainly
helminth infection that activates ILC2. ILC2, in turn, secrete IL-13 by ILC3s, induces epithelial cells to express anti-bacterial
driving the differentiation of epithelial cells toward the secretory peptides such as Reg3g and Reg3b, as well as antiviral proteins
lineage likely by modulating Notch signaling in epithelial cells. through reinforcement of interferon-l signaling (Hernández et al.,
Thus, this feedforward loop instructs tissue remodeling required 2015). In addition, IL-22 stabilizes the epithelial barrier by pro-
for expulsion of large multicellular pathogens (von Moltke et al., tecting epithelial cells and Lgr5+ stem cells from apoptosis
2016; Howitt et al., 2016; Gerbe et al., 2016). induced by irradiation, chemotherapy or graft versus host dis-
Immune activation comes at the price of collateral tissue dam- ease through the activation of STAT3 (Aparicio-Domingo et al.,
age. The best studied example may be influenza infection where 2015; Lindemans et al., 2015). IL-22 also protects thymic epithe-
most of the clinical sequelae are believed to be a consequence of lial cells from irradiation (Dudakov et al., 2012) and hepatocytes
immune system activation. ILC2s have important roles in limiting from acute inflammation (Zenewicz et al., 2007) (Figure 3B).
tissue damage after infection by the production of ligands for the While constitutive absence of IL-22 enhances inflammation-
epithelial growth factor receptor, such as AREG. AREG pro- driven colorectal cancer (Huber et al., 2012), the proliferation-
duced by ILC2s has been shown to control proliferation and promoting effect of IL-22 on epithelial cells can also sustain the
differentiation of epithelial cells which was required for epithelial progression of colon cancer (Kirchberger et al., 2013; Hernandez
repair following influenza infection (Monticelli et al., 2011; Zaiss et al., 2018).
et al., 2015). Similarly, the extensive tissue damages after hel-
minth migration and expulsion also lead to the production of Neuronal Regulation of ILC Responses
AREG, an important factor for timely repair (Monticelli et al., The regulation of immune responses has been shown to involve
2015) (Figure 3B). It is possible that the same signaling circuitry the brain and nervous system. The autonomic nervous system,
and the same cytokines (IL-13 and AREG) that promote tissue through its neurotransmitters and neuropeptides, and the
repair can become dysregulated in the presence of chronic hypothalamic-pituitary-adrenal axis work in parallel to modulate
inflammation and an altered cytokine milieu to promote fibrotic inflammation and maintain homeostasis. Recent studies have
responses, including myofibroblast differentiation, proliferation, described a complexity of interactions between ILCs and the
and extracellular matrix deposition. nervous system.
It may be a fundamental property of type 2 immunity and in Regulation of NK Cells and ILC1s by the Hypothalamic-
particular of ILC2s to instruct remodeling of extracellular matrix. Pituitary-Adrenal Axis
Indeed, ILC2s are a relevant source of IL-5 early during immune The neuroendocrine system can sense changes in the environ-
responses and are indispensable for the recruitment of eosino- ment. It then acts to restore homeostasis by modulating the
phils and the programing of alternatively activated macrophages strength and duration of inflammatory reactions through rapid,
(also referred to as M2 macrophages) (Molofsky et al., 2013). pleiotropic mechanisms. Activation of the hypothalamic-pitui-
Upon muscle injury, eosinophil-derived IL-4 induces the prolifer- tary-adrenal axis leads to glucocorticoid release in the blood.
ation of fibro/adipogenic progenitors (FAPs), thereby preventing NK cells and ILC1s express the glucocorticoid receptor, a tran-
their differentiation into adipocytes. In this way, FAPs supported scription factor involved in the regulation of multiple pathways
myogenesis and the regeneration of skeletal muscle after injury (Quatrini et al., 2017). Glucocorticoid receptor signaling in these
driven by a type 2 immune program (Heredia et al., 2013). Mech- cells inhibits IFN-g production by NK cells in the liver and spleen
anisms controlling ILC2 activities are also important in regaining and by ILC1s in the liver and is required for host resistance to
homeostasis after inflammation. STAT1 signals downstream of endotoxic shock. This regulation is required for the development
type I and type II interferons and of IL-27 are critical for suppress- of IL-10-dependent endotoxin tolerance. By contrast, disruption
ing ILC2 functions (Duerr et al., 2016; Molofsky et al., 2015; Moro of this pathway results in high levels of IFN-g culminating in
et al., 2016), whereas STAT5 signals downstream of IL-2, IL-9, endotoxin-induced septic shock and death (Quatrini et al.,
and TSLP enhance activation of ILC2, leading to steroid resis- 2017). During mouse cytomegalovirus infection, regulation of
tance (Kabata et al., 2016). NK cell functions by glucocorticoid receptors is also required
ILC1s and ILC3s in Repair for host resistance to infection. This regulation occurs through
Type 1 and type 3 responses that are orchestrated by ILC1s and the tissue-specific expression of PD1, an inhibitory receptor,
ILC3s, involve phagocytes that release oxygen radicals and on spleen NK cells. Binding of PD1 to its ligands downregulates
enzymes to lethally damage pathogens, kill infected cells, IFN-g production. This inhibition prevents the excessive

Cell 174, August 23, 2018 1061


production that would otherwise result in a lethal IFN-g-depen- patch formation and the development of the gut nervous system
dent spleen immunopathology, without impairing an effective (Patel et al., 2012; Veiga-Fernandes et al., 2007). RET is strongly
antiviral response (S. Ugolini et al., personal communication) expressed in the ILC3s of the lamina propria in the adult gut.
(Figure 3C). These cells are located within cryptopatches, close to stellate
Neuropeptides, Neurotransmitters, and ILC2s projections of the lamina propria glial cells. These glial cells are
Neurotransmitters and neuropeptides play a crucial role in the principal producers of RET ligands, which are induced in
communication between the nervous and immune systems, response to commensal products and alarmins. This essential
and many of these molecules are also produced directly by im- glial cell-ILC3 pathway conditions the reactivity of epithelial cells
mune cells. The ILCs of the intestine and lungs express receptors and regulates intestinal defenses against Citrobacter rodentium
for the neurotransmitters and neuropeptides produced in infection by inducing the production of IL-22 by ILC3s in response
these mucosal tissues. Indeed, various ILC subsets express to RET ligands (Ibiza et al., 2016) (Figure 3C). Communications
the b2AR (b2-adrenergic receptor) for epinephrine and norepi- between the central nervous system and the intestine are also
nephrine, the CHRM (cholinergic receptor muscarinic) for acetyl- mediated by the so-called ‘‘gut-brain axis,’’ through projections
choline, the VPAC1/2 (vasoactive intestinal peptide receptor) from the sympathetic and parasympathetic systems, and via
for VIP, the NMUR1 for neuromedin U (NMU) and CALCRL the hypothalamic pituitary adrenal axis. The vagus nerve is the
(calcitonin receptor-like) for CGRP. ILC2s are subject to tight principal extrinsic parasympathetic nerve connecting the brain-
regulation by neuropeptides. The principal role of VIP is the main- stem and gut. ILC3s are located close to choline acetyltransfer-
tenance of homeostasis at mucosal barriers through the regula- ase-positive cells in the greater omentum (Dalli et al., 2017),
tion of IL-5 production (Nussbaum et al., 2013). CGRP signaling suggesting that crosstalk between ILC3s and cholinergic vagus
in ILC2s is required for a full Th2 immune response in models of nerve fibers occurs not only in the intestine but also in the perito-
allergen-induced asthma (Sui et al., 2018). NMU plays no signifi- neum. ILC3s can also be regulated by the cholinergic receptors
cant role in homeostatic conditions, but its induction upon hel- muscarinic (Chrm)1, 2, 4 and 5, and both mouse and human
minth infection leads to a type 2 protective immune response in ILC3s respond to acetyl-chloline stimulation by producing the
the intestine through intrinsic ILC2 regulation. NMU has also lipid mediator PCTR1 (16R-glutathionyl, 17S-hydroxy-4Z, 7Z,
been shown to act in synergy with IL-25 in the induction of 10Z, 12E, 14E, 19Z-docosahexaenoic acid) (Dalli et al., 2017)
cytokine production by lung ILC2s (Cardoso et al., 2017; Klose (Figure 3C). PCTR1 belongs to a family of proresolving molecules
et al., 2017; Wallrapp et al., 2017) (Figure 3C). While the role of that control the clearance of bacterial infections by phagocytes
catecholamines on NK cells appears complex and remains to and attenuate collateral tissue injury and inflammation. Thus, tis-
be dissected in depth, it has been recently demonstrated that sue-resident ILC3s actively regulate macrophage and granulo-
b2AR stimulation intrinsically suppresses the proliferation and cyte responses to infection through the release of cytokines,
effector functions of ILC2s. ILC2 responses and type 2 inflamma- such as GM-CSF (Mortha et al., 2014), and by the production of
tion are, thus, downregulated following exposure to the parasite proresolving mediators under vagal system control. The direct
N. brasiliensis in the gut and by intranasal IL-33 administration or role of vagus stimulation in ILC3 control remains to be shown.
exposure to Alternaria alternata extract in the lungs (Moriyama
et al., 2018) (Figure 3C). Other interactions between ILC2s and Conclusions and Perspectives
neurons also occur in the mucosal tissues of the respiratory tract. After 10 years of intensive research, the field of ILC biology
Indeed, ILC2s have been observed very close to SNAP-25+ nerve has provided a new vision on the organization of the immune
fibers on lung sections (Wallrapp et al., 2017), and lung IL-5-pro- response. Nevertheless, several key questions remain to be
ducing ILC2s have been identified in collagen-rich regions close addressed.
to the confluence of medium-sized blood vessels and airways ILC plasticity may be essential to shape and calibrate ILC
(Nussbaum et al., 2013). IL-5-producing ILC2s have also been responses to different types of pathogenic stimuli. However,
found close to pulmonary neuroendocrine cells in the airway several fundamental questions remain to be addressed:
branch junctions at which particles entering the airways become (1) When does ILC plasticity arise? Does it occur at all stages of
concentrated (Sui et al., 2018). Thus, ILC2s appear to be located ILC differentiation or is it progressively lost toward the final
at strategic points in the mucosal tissues of the airways, acting stages? How to discriminate cell plasticity from various cell acti-
with the neuroendocrine system to patrol the airways, recruiting vation states? (2) Given that Th cells often display mixed Th1/
circulating immune cells to sites of damage or in response to Th17 polarization, are there ILCs with a mixed cytokine secretion
pathogen invasion. pattern? (3) What epigenetic circuits are activated and silenced
ILC3s and Neuroimmune Cell Units during plastic ILC responses to a changing microenvironment?
Neuroimmune cell units (NICUs) have recently been described as (4) Is ILC conversion irreversible, or can ILCs repeatedly trans-
defined small cellular networks in which immune and neuronal form after restimulation with polarizing cytokines? (5) Given
cells are found at the same anatomic site, interact functionally, that infections, inflammation, and autoimmunity induce the
and participate in tissue homeostasis and integrity (Veiga-Fer- release of inflammatory cytokines into the tissue environment,
nandes and Artis, 2018). The RET neurotrophic factor receptor is ILC conversion enhanced under these conditions? (6) Is this
is a typical example of such communication between the cells plasticity—the functional capacity of converted ILCs—essential
of the nervous and immune systems. RET is a receptor tyrosine to fight infections and autoimmunity?
kinase activated by glial cell-derived neurotrophic factor and Compelling data in human and in the mouse indicate that ILCs
related proteins. It plays crucial roles in hematopoiesis, Peyer’s are involved in both inflammatory disorders and immunological

1062 Cell 174, August 23, 2018


repair (Ebbo et al., 2017). The absence of immunodeficiency in two ILC subsets are critical for protection against acute clostridium difficile
ILC-deficient patients in conditions of modern hygiene and med- infection. Cell Host Microbe 18, 27–37.
icine led to the proposal that ILCs are dispensable for protective Aparicio-Domingo, P., Romera-Hernandez, M., Karrich, J.J., Cornelissen, F.,
immunity if T and B cell functions are preserved (Vély et al., Papazian, N., Lindenbergh-Kortleve, D.J., Butler, J.A., Boon, L., Coles, M.C.,
Samsom, J.N., and Cupedo, T. (2015). Type 3 innate lymphoid cells maintain
2016). Thus, the precise roles of ILCs in immunity and their dialog
intestinal epithelial stem cells after tissue damage. J. Exp. Med. 212, 1783–
with the other components of the multilayered immune response 1791.
await further analysis. Along this line, the role and importance of Artis, D., and Spits, H. (2015). The biology of innate lymphoid cells. Nature 517,
ILCs in protective immunity in adult humans or wild-type mice, 293–301.
where many tissue-resident adaptive memory cells share space Bal, S.M., Bernink, J.H., Nagasawa, M., Groot, J., Shikhagaie, M.M., Golebski,
with ILCs, remains to be established. However, the roles of ILCs K., van Drunen, C.M., Lutter, R., Jonkers, R.E., Hombrink, P., et al. (2016). IL-
in adapting tissue physiology to changes in the environment 1b, IL-4 and IL-12 control the fate of group 2 innate lymphoid cells in human
have inspired research into processes that are not convention- airway inflammation in the lungs. Nat. Immunol. 17, 636–645.
ally associated with the immune system. Further exploration of Bando, J.K., Liang, H.E., and Locksley, R.M. (2015). Identification and distribu-
these pathways may likely reveal unsuspected pathways by tion of developing innate lymphoid cells in the fetal mouse intestine. Nat.
which immune system workings might be harnessed to improve Immunol. 16, 153–160.

health and health span. Bernink, J.H., Krabbendam, L., Germar, K., de Jong, E., Gronke, K., Kofoed-
Nielsen, M., Munneke, J.M., Hazenberg, M.D., Villaudy, J., Buskens, C.J.,
Finally, the role of ILCs in the onset and/or maintenance of
et al. (2015). Interleukin-12 and -23 control plasticity of CD127(+) group 1
inflammation, their preferential homing to mucosal tissues, and and group 3 innate lymphoid cells in the intestinal lamina propria. Immunity
the expression of several checkpoint receptors on their surface 43, 146–160.
should encourage researchers to explore the ILCs’ manipulation Boulenouar, S., Michelet, X., Duquette, D., Alvarez, D., Hogan, A.E., Dold, C.,
in innovative therapies. O’Connor, D., Stutte, S., Tavakkoli, A., Winters, D., et al. (2017). Adipose type
one innate lymphoid cells regulate macrophage homeostasis through targeted
cytotoxicity. Immunity 46, 273–286.
ACKNOWLEDGMENTS
Brestoff, J.R., Kim, B.S., Saenz, S.A., Stine, R.R., Monticelli, L.A., Sonnenberg,
We thank the members of all our laboratories for critically reading the G.F., Thome, J.J., Farber, D.L., Lutfy, K., Seale, P., and Artis, D. (2015). Group 2
manuscript, Adeline Crinier for help with Table 1, Linda Quatrini and Sophie innate lymphoid cells promote beiging of white adipose tissue and limit
Ugolini (CIML) for sharing unpublished results, and Marie-Alice Rarivoson obesity. Nature 519, 242–246.
(Innate-Pharma) for help in formatting the manuscript. Buonocore, S., Ahern, P.P., Uhlig, H.H., Ivanov, I.I., Littman, D.R., Maloy, K.J.,
Research in the Artis lab is supported by the NIH, the Crohn’s and Colitis and Powrie, F. (2010). Innate lymphoid cells drive interleukin-23-dependent
Foundation, the Burroughs Wellcome Fund, the Cure for IBD, and the Jill innate intestinal pathology. Nature 464, 1371–1375.
Roberts Institute. M.C. is supported by the US NIH (UO1 AI095542, RO1 Cardoso, V., Chesné, J., Ribeiro, H., Garcı́a-Cassani, B., Carvalho, T., Bouch-
DE025884, and RO1 DK103039).The Diefenbach laboratory is supported by ery, T., Shah, K., Barbosa-Morais, N.L., Harris, N., and Veiga-Fernandes, H.
the European Research Council (ERC) (311377 - NUTRIMMUNE), the Priority (2017). Neuronal regulation of type 2 innate lymphoid cells via neuromedin
Program 1937 ‘‘Innate Lymphoid Cells’’ of the Deutsche Forschungsgemein- U. Nature 549, 277–281.
schaft (DFG), the Einstein Foundation Berlin, and the Berlin Institute of Health.
Cella, M., Fuchs, A., Vermi, W., Facchetti, F., Otero, K., Lennerz, J.K., Doherty,
The Eberl lab is supported by grants from ANR, FRM, CCFA, Rainin Foundation
J.M., Mills, J.C., and Colonna, M. (2009). A human natural killer cell subset pro-
and the Institut Pasteur. Research in the Koyasu laboratory is supported by a
vides an innate source of IL-22 for mucosal immunity. Nature 457, 722–725.
Grant-in-Aid for Scientific Research (A) (16H02631). Research in the Locksley
laboratory is supported by the HHMI, the NIH, and the SABRE Center at Cella, M., Otero, K., and Colonna, M. (2010). Expansion of human NK-22 cels
UCSF. Research in the McKenzie laboratory is supported by funding from with IL-17, IL-2 and IL-1beta reveals intrinsic funciotnal plasticity. Proc. Natl.
the MRC (U105178805) and the Wellcome Trust (100963/Z/13/Z). The Di Santo Sci. USA 107, 10961–10966.
laboratory receives grants from the Institut Pasteur, Inserm, the Agence Natio- Chea, S., Perchet, T., Petit, M., Verrier, T., Guy-Grand, D., Banchi, E.G.,
nale de la Recherche, the Ligue Nationale contre le Cancer, and the European Vosshenrich, C.A., Di Santo, J.P., Cumano, A., and Golub, R. (2016). Notch
Research Council (ERC) under the European Union’s Horizon 2020 Research signaling in group 3 innate lymphoid cells modulates their plasticity. Sci.
and Innovation Program (695467 - ILC_REACTIVITY). The Spits lab is sup- Signal. 9, ra45.
ported by an advanced ERC grant (341038-AsthmaVir). Research in the Vivier Constantinides, M.G., McDonald, B.D., Verhoef, P.A., and Bendelac, A. (2014).
laboratory is supported by funding from the European Research Council (ERC) A committed precursor to innate lymphoid cells. Nature 508, 397–401.
under the European Union’s Horizon 2020 Research and Innovation Program
Cortez, V.S., Ulland, T.K., Cervantes-Barragan, L., Bando, J.K., Robinette,
(694502-TILC), the Agence Nationale de la Recherche; Equipe Labellisée ‘‘La
M.L., Wang, Q., White, A.J., Gilfillan, S., Cella, M., and Colonna, M. (2017).
Ligue,’’ Ligue Nationale contre le Cancer, MSDAvenir, and Innate Pharma
SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing
and institutional grants to the CIML (INSERM, CNRS, and Aix-Marseille Univer-
non-canonical TGF-b signaling. Nat. Immunol. 18, 995–1003.
sity) and Marseille Immunopôle.
Cupedo, T., Crellin, N.K., Papazian, N., Rombouts, E.J., Weijer, K., Grogan,
J.L., Fibbe, W.E., Cornelissen, J.J., and Spits, H. (2009). Human fetal lymphoid
DECLARATION OF INTERESTS tissue-inducer cells are interleukin 17-producing precursors to RORC+
CD127+ natural killer-like cells. Nat. Immunol. 10, 66–74.
E.V. is an employee of Innate-Pharma; the other authors declare no competing
Dalli, J., Colas, R.A., Arnardottir, H., and Serhan, C.N. (2017). Vagal regulation
financial interests.
of group 3 innate lymphoid cells and the immunoresolvent PCTR1 controls
infection resolution. Immunity 46, 92–105.
REFERENCES Dalmas, E., Lehmann, F.M., Dror, E., Wueest, S., Thienel, C., Borsigova, M.,
Stawiski, M., Traunecker, E., Lucchini, F.C., Dapito, D.H., et al. (2017). Inter-
Abt, M.C., Lewis, B.B., Caballero, S., Xiong, H., Carter, R.A., Susac, B., Ling, leukin-33-activated islet-resident innate lymphoid cells promote insulin secre-
L., Leiner, I., and Pamer, E.G. (2015). Innate immune defenses mediated by tion through myeloid cell retinoic acid production. Immunity 47, 928–942.e7.

Cell 174, August 23, 2018 1063


Daussy, C., Faure, F., Mayol, K., Viel, S., Gasteiger, G., Charrier, E., Bienvenu, Hoyler, T., Klose, C.S., Souabni, A., Turqueti-Neves, A., Pfeifer, D., Rawlins,
J., Henry, T., Debien, E., Hasan, U.A., et al. (2014). T-bet and Eomes instruct E.L., Voehringer, D., Busslinger, M., and Diefenbach, A. (2012). The transcrip-
the development of two distinct natural killer cell lineages in the liver and in tion factor GATA-3 controls cell fate and maintenance of type 2 innate
the bone marrow. J. Exp. Med. 211, 563–577. lymphoid cells. Immunity 37, 634–648.
Diefenbach, A., Colonna, M., and Koyasu, S. (2014). Development, differenti- Huang, Y., Guo, L., Qiu, J., Chen, X., Hu-Li, J., Siebenlist, U., Williamson, P.R.,
ation, and diversity of innate lymphoid cells. Immunity 41, 354–365. Urban, J.F., Jr., and Paul, W.E. (2015). IL-25-responsive, lineage-negative
Dudakov, J.A., Hanash, A.M., Jenq, R.R., Young, L.F., Ghosh, A., Singer, N.V., KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells.
West, M.L., Smith, O.M., Holland, A.M., Tsai, J.J., et al. (2012). Interleukin-22 Nat. Immunol. 16, 161–169.
drives endogenous thymic regeneration in mice. Science 336, 91–95. Huber, S., Gagliani, N., Zenewicz, L.A., Huber, F.J., Bosurgi, L., Hu, B., Hedl,
Duerr, C.U., McCarthy, C.D., Mindt, B.C., Rubio, M., Meli, A.P., Pothlichet, J., M., Zhang, W., O’Connor, W., Jr., Murphy, A.J., et al. (2012). IL-22BP is regu-
Eva, M.M., Gauchat, J.F., Qureshi, S.T., Mazer, B.D., et al. (2016). Type I inter- lated by the inflammasome and modulates tumorigenesis in the intestine. Na-
feron restricts type 2 immunopathology through the regulation of group 2 ture 491, 259–263.
innate lymphoid cells. Nat. Immunol. 17, 65–75.
Ibiza, S., Garcı́a-Cassani, B., Ribeiro, H., Carvalho, T., Almeida, L., Marques,
Ebbo, M., Crinier, A., Vély, F., and Vivier, E. (2017). Innate lymphoid cells: major R., Misic, A.M., Bartow-McKenney, C., Larson, D.M., Pavan, W.J., et al.
players in inflammatory diseases. Nat. Rev. Immunol. 17, 665–678. (2016). Glial-cell-derived neuroregulators control type 3 innate lymphoid cells
Eberl, G., Colonna, M., Di Santo, J.P., and McKenzie, A.N. (2015). Innate and gut defence. Nature 535, 440–443.
lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science
Kabata, H., Moro, K., Koyasu, S., Fukunaga, K., Asano, K., and Betsuyaku, T.
348, aaa6566.
(2016). Mechanisms to suppress ilc2-induced airway inflammation. Ann Am
Freud, A.G., Keller, K.A., Scoville, S.D., Mundy-Bosse, B.L., Cheng, S., Thorac Soc. 13, 95.
Youssef, Y., Hughes, T., Zhang, X., Mo, X., Porcu, P., et al. (2016). NKp80
Kim, M.H., Taparowsky, E.J., and Kim, C.H. (2015). Retinoic acid differentially
defines a critical step during human natural killer cell development. Cell Rep.
regulates the migration of innate lymphoid cell subsets to the gut. Immunity 43,
16, 379–391.
107–119.
Fuchs, A., Vermi, W., Lee, J.S., Lonardi, S., Gilfillan, S., Newberry, R.D., Cella,
M., and Colonna, M. (2013). Intraepithelial type 1 innate lymphoid cells are a Kirchberger, S., Royston, D.J., Boulard, O., Thornton, E., Franchini, F., Sza-
unique subset of IL-12- and IL-15-responsive IFN-g-producing cells. Immunity bady, R.L., Harrison, O., and Powrie, F. (2013). Innate lymphoid cells sustain
38, 769–781. colon cancer through production of interleukin-22 in a mouse model. J. Exp.
Med. 210, 917–931.
Furusawa, J., Moro, K., Motomura, Y., Okamoto, K., Zhu, J., Takayanagi, H.,
Kubo, M., and Koyasu, S. (2013). Critical role of p38 and GATA3 in natural help- Kiss, E.A., Vonarbourg, C., Kopfmann, S., Hobeika, E., Finke, D., Esser, C., and
er cell function. J. Immunol. 191, 1818–1826. Diefenbach, A. (2011). Natural aryl hydrocarbon receptor ligands control
Gao, Y., Souza-Fonseca-Guimaraes, F., Bald, T., Ng, S.S., Young, A., Ngiow, organogenesis of intestinal lymphoid follicles. Science 334, 1561–1565.
S.F., Rautela, J., Straube, J., Waddell, N., Blake, S.J., et al. (2017). Tumor im- Klein Wolterink, R.G., Serafini, N., van Nimwegen, M., Vosshenrich, C.A., de
munoevasion by the conversion of effector NK cells into type 1 innate lymphoid Bruijn, M.J., Fonseca Pereira, D., Veiga Fernandes, H., Hendriks, R.W., and
cells. Nat. Immunol. 18, 1004–1015. Di Santo, J.P. (2013). Essential, dose-dependent role for the transcription fac-
Gasteiger, G., Fan, X., Dikiy, S., Lee, S.Y., and Rudensky, A.Y. (2015). Tissue tor Gata3 in the development of IL-5+ and IL-13+ type 2 innate lymphoid cells.
residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Sci- Proc. Natl. Acad. Sci. USA 110, 10240–10245.
ence 350, 981–985. Klose, C.S., Kiss, E.A., Schwierzeck, V., Ebert, K., Hoyler, T., d’Hargues, Y.,
Gerbe, F., Sidot, E., Smyth, D.J., Ohmoto, M., Matsumoto, I., Dardalhon, V., Göppert, N., Croxford, A.L., Waisman, A., Tanriver, Y., and Diefenbach, A.
Cesses, P., Garnier, L., Pouzolles, M., Brulin, B., et al. (2016). Intestinal epithe- (2013). A T-bet gradient controls the fate and function of CCR6-RORgt+ innate
lial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature 529, lymphoid cells. Nature 494, 261–265.
226–230.
Klose, C.S.N., Flach, M., Möhle, L., Rogell, L., Hoyler, T., Ebert, K., Fabiunke,
Gury-BenAri, M., Thaiss, C.A., Serafini, N., Winter, D.R., Giladi, A., Lara-As- C., Pfeifer, D., Sexl, V., Fonseca-Pereira, D., et al. (2014). Differentiation of type
tiaso, D., Levy, M., Salame, T.M., Weiner, A., David, E., et al. (2016). The spec- 1 ILCs from a common progenitor to all helper-like innate lymphoid cell line-
trum and regulatory landscape of intestinal innate lymphoid cells are shaped ages. Cell 157, 340–356.
by the microbiome. Cell 166, 1231–1246.e13.
Klose, C.S.N., Mahlakõiv, T., Moeller, J.B., Rankin, L.C., Flamar, A.L., Kabata,
Hepworth, M.R., Monticelli, L.A., Fung, T.C., Ziegler, C.G., Grunberg, S., H., Monticelli, L.A., Moriyama, S., Putzel, G.G., Rakhilin, N., et al. (2017). The
Sinha, R., Mantegazza, A.R., Ma, H.L., Crawford, A., Angelosanto, J.M., neuropeptide neuromedin U stimulates innate lymphoid cells and type 2
et al. (2013). Innate lymphoid cells regulate CD4+ T-cell responses to intestinal inflammation. Nature 549, 282–286.
commensal bacteria. Nature 498, 113–117.
Koues, O.I., Collins, P.L., Cella, M., Robinette, M.L., Porter, S.I., Pyfrom, S.C.,
Heredia, J.E., Mukundan, L., Chen, F.M., Mueller, A.A., Deo, R.C., Locksley,
Payton, J.E., Colonna, M., and Oltz, E.M. (2016). Distinct gene regulatory path-
R.M., Rando, T.A., and Chawla, A. (2013). Type 2 innate signals stimulate
ways for human innate versus adaptive lymphoid cells. Cell 165, 1134–1146.
fibro/adipogenic progenitors to facilitate muscle regeneration. Cell 153,
376–388. Lee, J.S., Cella, M., McDonald, K.G., Garlanda, C., Kennedy, G.D., Nukaya,
Hernandez, P., Gronke, K., and Diefenbach, A. (2018). A catch-22: Interleukin- M., Mantovani, A., Kopan, R., Bradfield, C.A., Newberry, R.D., and Colonna,
22 and cancer. Eur J Immunol. 48, 15–31. M. (2011). AHR drives the development of gut ILC22 cells and postnatal
lymphoid tissues via pathways dependent on and independent of Notch.
Hernández, P.P., Mahlakoiv, T., Yang, I., Schwierzeck, V., Nguyen, N.,
Nat. Immunol. 13, 144–151.
Guendel, F., Gronke, K., Ryffel, B., Hoelscher, C., Dumoutier, L., et al.
(2015). Interferon-l and interleukin 22 act synergistically for the induction of Lee, M.W., Odegaard, J.I., Mukundan, L., Qiu, Y., Molofsky, A.B., Nussbaum,
interferon-stimulated genes and control of rotavirus infection. Nat. Immunol. J.C., Yun, K., Locksley, R.M., and Chawla, A. (2015). Activated type 2 innate
16, 698–707. lymphoid cells regulate beige fat biogenesis. Cell 160, 74–87.
Howitt, M.R., Lavoie, S., Michaud, M., Blum, A.M., Tran, S.V., Weinstock, J.V., Lee, B.C., Kim, M.S., Pae, M., Yamamoto, Y., Eberlé, D., Shimada, T., Kamei,
Gallini, C.A., Redding, K., Margolskee, R.F., Osborne, L.C., et al. (2016). Tuft N., Park, H.S., Sasorith, S., Woo, J.R., et al. (2016). Adipose natural killer cells
cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the regulate adipose tissue macrophages to promote insulin resistance in obesity.
gut. Science 351, 1329–1333. Cell Metab. 23, 685–698.

1064 Cell 174, August 23, 2018


Lim, A.I., Menegatti, S., Bustamante, J., Le Bourhis, L., Allez, M., Rogge, L., Mueller, S.N., and Mackay, L.K. (2016). Tissue-resident memory T cells: local
Casanova, J.L., Yssel, H., and Di Santo, J.P. (2016). IL-12 drives functional specialists in immune defence. Nat. Rev. Immunol. 16, 79–89.
plasticity of human group 2 innate lymphoid cells. J. Exp. Med. 213, 569–583. Neill, D.R., Wong, S.H., Bellosi, A., Flynn, R.J., Daly, M., Langford, T.K., Bucks,
Lim, A.I., Li, Y., Lopez-Lastra, S., Stadhouders, R., Paul, F., Casrouge, A., Ser- C., Kane, C.M., Fallon, P.G., Pannell, R., et al. (2010). Nuocytes represent a
afini, N., Puel, A., Bustamante, J., Surace, L., et al. (2017). Systemic human ILC new innate effector leukocyte that mediates type-2 immunity. Nature 464,
precursors provide a substrate for tissue ILC differentiation. Cell 168, 1086– 1367–1370.
1100.e10. Nussbaum, J.C., Van Dyken, S.J., von Moltke, J., Cheng, L.E., Mohapatra, A.,
Lindemans, C.A., Calafiore, M., Mertelsmann, A.M., O’Connor, M.H., Duda- Molofsky, A.B., Thornton, E.E., Krummel, M.F., Chawla, A., Liang, H.E., and
kov, J.A., Jenq, R.R., Velardi, E., Young, L.F., Smith, O.M., Lawrence, G., Locksley, R.M. (2013). Type 2 innate lymphoid cells control eosinophil homeo-
et al. (2015). Interleukin-22 promotes intestinal-stem-cell-mediated epithelial stasis. Nature 502, 245–248.
regeneration. Nature 528, 560–564.
O’Sullivan, T.E., Rapp, M., Fan, X., Weizman, O.E., Bhardwaj, P., Adams, N.M.,
Luci, C., Reynders, A., Ivanov, I.I., Cognet, C., Chiche, L., Chasson, L., Hard- Walzer, T., Dannenberg, A.J., and Sun, J.C. (2016). Adipose-resident group 1
wigsen, J., Anguiano, E., Banchereau, J., Chaussabel, D., et al. (2009). Influ- innate lymphoid cells promote obesity-associated insulin resistance. Immunity
ence of the transcription factor RORgammat on the development of NKp46+ 45, 428–441.
cell populations in gut and skin. Nat. Immunol. 10, 75–82.
Ohne, Y., Silver, J.S., Thompson-Snipes, L., Collet, M.A., Blanck, J.P., Can-
Mebius, R.E., Rennert, P., and Weissman, I.L. (1997). Developing lymph nodes tarel, B.L., Copenhaver, A.M., Humbles, A.A., and Liu, Y.J. (2016). IL-1 is a crit-
collect CD4+CD3- LTbeta+ cells that can differentiate to APC, NK cells, and ical regulator of group 2 innate lymphoid cell function and plasticity. Nat. Im-
follicular cells but not T or B cells. Immunity 7, 493–504. munol. 17, 646–655.
Mielke, L.A., Jones, S.A., Raverdeau, M., Higgs, R., Stefanska, A., Groom, Oliphant, C.J., Hwang, Y.Y., Walker, J.A., Salimi, M., Wong, S.H., Brewer,
J.R., Misiak, A., Dungan, L.S., Sutton, C.E., Streubel, G., et al. (2013). Retinoic J.M., Englezakis, A., Barlow, J.L., Hams, E., Scanlon, S.T., et al. (2014).
acid expression associates with enhanced IL-22 production by gd T cells and MHCII-mediated dialog between group 2 innate lymphoid cells and CD4(+)
innate lymphoid cells and attenuation of intestinal inflammation. J. Exp. Med. T cells potentiates type 2 immunity and promotes parasitic helminth expulsion.
210, 1117–1124. Immunity 41, 283–295.
Mjösberg, J.M., Trifari, S., Crellin, N.K., Peters, C.P., van Drunen, C.M., Piet, Palm, N.W., Rosenstein, R.K., and Medzhitov, R. (2012). Allergic host de-
B., Fokkens, W.J., Cupedo, T., and Spits, H. (2011). Human IL-25- and IL- fences. Nature 484, 465–472.
33-responsive type 2 innate lymphoid cells are defined by expression of
Patel, A., Harker, N., Moreira-Santos, L., Ferreira, M., Alden, K., Timmis, J.,
CRTH2 and CD161. Nat. Immunol. 12, 1055–1062.
Foster, K., Garefalaki, A., Pachnis, P., Andrews, P., et al. (2012). Differential
Mjösberg, J., Bernink, J., Golebski, K., Karrich, J.J., Peters, C.P., Blom, B., te RET signaling pathways drive development of the enteric lymphoid and ner-
Velde, A.A., Fokkens, W.J., van Drunen, C.M., and Spits, H. (2012). The tran- vous systems. Sci. Signal. 5, ra55.
scription factor GATA3 is essential for the function of human type 2 innate
lymphoid cells. Immunity 37, 649–659. Pearson, C., Thornton, E.E., McKenzie, B., Schaupp, A.L., Huskens, N., Gri-
seri, T., West, N., Tung, S., Seddon, B.P., Uhlig, H.H., and Powrie, F. (2016).
Molofsky, A.B., Nussbaum, J.C., Liang, H.E., Van Dyken, S.J., Cheng, L.E., ILC3 GM-CSF production and mobilisation orchestrate acute intestinal inflam-
Mohapatra, A., Chawla, A., and Locksley, R.M. (2013). Innate lymphoid type mation. eLife 5, e10066. https://doi.org/10.7554/eLife.10066.
2 cells sustain visceral adipose tissue eosinophils and alternatively activated
macrophages. J. Exp. Med. 210, 535–549. Peng, H., Jiang, X., Chen, Y., Sojka, D.K., Wei, H., Gao, X., Sun, R., Yokoyama,
W.M., and Tian, Z. (2013). Liver-resident NK cells confer adaptive immunity in
Molofsky, A.B., Van Gool, F., Liang, H.E., Van Dyken, S.J., Nussbaum, J.C.,
skin-contact inflammation. J. Clin. Invest. 123, 1444–1456.
Lee, J., Bluestone, J.A., and Locksley, R.M. (2015). Interleukin-33 and inter-
feron-g counter-regulate group 2 innate lymphoid cell activation during im- Price, A.E., Liang, H.E., Sullivan, B.M., Reinhardt, R.L., Eisley, C.J., Erle, D.J.,
mune perturbation. Immunity 43, 161–174. and Locksley, R.M. (2010). Systemically dispersed innate IL-13-expressing
cells in type 2 immunity. Proc. Natl. Acad. Sci. USA 107, 11489–11494.
Monticelli, L.A., Sonnenberg, G.F., Abt, M.C., Alenghat, T., Ziegler, C.G., Doer-
ing, T.A., Angelosanto, J.M., Laidlaw, B.J., Yang, C.Y., Sathaliyawala, T., et al. Quatrini, L., Wieduwild, E., Guia, S., Bernat, C., Glaichenhaus, N., Vivier, E.,
(2011). Innate lymphoid cells promote lung-tissue homeostasis after infection and Ugolini, S. (2017). Host resistance to endotoxic shock requires the neuro-
with influenza virus. Nat Immunol 12, 1045–1054. endocrine regulation of group 1 innate lymphoid cells. J. Exp. Med. 214,
3531–3541.
Monticelli, L.A., Osborne, L.C., Noti, M., Tran, S.V., Zaiss, D.M., and Artis, D.
(2015). IL-33 promotes an innate immune pathway of intestinal tissue protec- Rankin, L.C., Groom, J.R., Chopin, M., Herold, M.J., Walker, J.A., Mielke, L.A.,
tion dependent on amphiregulin-EGFR interactions. Proc Natl Acad Sci USA McKenzie, A.N., Carotta, S., Nutt, S.L., and Belz, G.T. (2013). The transcription
112, 10762–10767. factor T-bet is essential for the development of NKp46+ innate lymphocytes
via the Notch pathway. Nat. Immunol. 14, 389–395.
Moriyama, S., Brestoff, J.R., Flamar, A.L., Moeller, J.B., Klose, C.S.N., Rankin,
L.C., Yudanin, N.A., Monticelli, L.A., Putzel, G.G., Rodewald, H.R., and Artis, Rankin, L.C., Girard-Madoux, M.J., Seillet, C., Mielke, L.A., Kerdiles, Y., Fenis,
D. (2018). b2-adrenergic receptor-mediated negative regulation of group 2 A., Wieduwild, E., Putoczki, T., Mondot, S., Lantz, O., et al. (2016). Comple-
innate lymphoid cell responses. Science 359, 1056–1061. mentarity and redundancy of IL-22-producing innate lymphoid cells. Nat. Im-
munol. 17, 179–186.
Moro, K., Yamada, T., Tanabe, M., Takeuchi, T., Ikawa, T., Kawamoto, H., Fur-
usawa, J., Ohtani, M., Fujii, H., and Koyasu, S. (2010). Innate production of Renoux, V.M., Zriwil, A., Peitzsch, C., Michaëlsson, J., Friberg, D., Soneji, S.,
T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. and Sitnicka, E. (2015). Identification of a human natural killer cell lineage-
Nature 463, 540–544. restricted progenitor in fetal and adult tissues. Immunity 43, 394–407.
Moro, K., Kabata, H., Tanabe, M., Koga, S., Takeno, N., Mochizuki, M., Fuku- Robinette, M.L., Fuchs, A., Cortez, V.S., Lee, J.S., Wang, Y., Durum, S.K., Gil-
naga, K., Asano, K., Betsuyaku, T., and Koyasu, S. (2016). Interferon and IL-27 fillan, S., and Colonna, M.; Immunological Genome Consortium (2015). Tran-
antagonize the function of group 2 innate lymphoid cell function and type 2 scriptional programs define molecular characteristics of innate lymphoid cell
innate immune responses. Nat. Immunol. 17, 76–86. classes and subsets. Nat. Immunol. 16, 306–317.
Mortha, A., Chudnovskiy, A., Hashimoto, D., Bogunovic, M., Spencer, S.P., Sanos, S.L., Bui, V.L., Mortha, A., Oberle, K., Heners, C., Johner, C., and Die-
Belkaid, Y., and Merad, M. (2014). Microbiota-dependent crosstalk between fenbach, A. (2009). RORgammat and commensal microflora are required for
macrophages and ILC3 promotes intestinal homeostasis. Science 343, the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat. Im-
1249288. munol. 10, 83–91.

Cell 174, August 23, 2018 1065


Satoh-Takayama, N., Vosshenrich, C.A., Lesjean-Pottier, S., Sawa, S., Loch- Vély, F., Barlogis, V., Vallentin, B., Neven, B., Piperoglou, C., Ebbo, M., Per-
ner, M., Rattis, F., Mention, J.J., Thiam, K., Cerf-Bensussan, N., Mandelboim, chet, T., Petit, M., Yessaad, N., Touzot, F., et al. (2016). Evidence of innate
O., et al. (2008). Microbial flora drives interleukin 22 production in intestinal lymphoid cell redundancy in humans. Nat. Immunol. 17, 1291–1299.
NKp46+ cells that provide innate mucosal immune defense. Immunity 29, Viant, C., Rankin, L.C., Girard-Madoux, M.J., Seillet, C., Shi, W., Smyth, M.J.,
958–970. Bartholin, L., Walzer, T., Huntington, N.D., Vivier, E., and Belz, G.T. (2016).
Sciumé, G., Hirahara, K., Takahashi, H., Laurence, A., Villarino, A.V., Singleton, Transforming growth factor-b and Notch ligands act as opposing environ-
K.L., Spencer, S.P., Wilhelm, C., Poholek, A.C., Vahedi, G., et al. (2012). mental cues in regulating the plasticity of type 3 innate lymphoid cells. Sci.
Distinct requirements for T-bet in gut innate lymphoid cells. J. Exp. Med. Signal. 9, ra46.
209, 2331–2338.
Vivier, E., Raulet, D.H., Moretta, A., Caligiuri, M.A., Zitvogel, L., Lanier, L.L., Yo-
Scoville, S.D., Mundy-Bosse, B.L., Zhang, M.H., Chen, L., Zhang, X., Keller, koyama, W.M., and Ugolini, S. (2011). Innate or adaptive immunity? The
K.A., Hughes, T., Chen, L., Cheng, S., Bergin, S.M., et al. (2016). A progenitor example of natural killer cells. Science 331, 44–49.
cell expressing transcription factor RORgt generates all human innate
lymphoid cell subsets. Immunity 44, 1140–1150. von Moltke, J., Ji, M., Liang, H.E., and Locksley, R.M. (2016). Tuft-cell-derived
IL-25 regulates an intestinal ILC2-epithelial response circuit. Nature 529,
Seehus, C.R., Kadavallore, A., Torre, B., Yeckes, A.R., Wang, Y., Tang, J., and
221–225.
Kaye, J. (2017). Alternative activation generates IL-10 producing type 2 innate
lymphoid cells. Nat. Commun. 8, 1900. Vonarbourg, C., Mortha, A., Bui, V.L., Hernandez, P.P., Kiss, E.A., Hoyler, T.,
Flach, M., Bengsch, B., Thimme, R., Hölscher, C., et al. (2010). Regulated
Shih, H.Y., Sciumè, G., Mikami, Y., Guo, L., Sun, H.W., Brooks, S.R., Urban,
expression of nuclear receptor RORgt confers distinct functional fates to NK
J.F., Jr., Davis, F.P., Kanno, Y., and O’Shea, J.J. (2016). Developmental acqui-
cell receptor-expressing RORgt(+) innate lymphocytes. Immunity 33,
sition of regulomes underlies innate lymphoid cell functionality. Cell 165,
736–751.
1120–1133.
Shikhagaie, M.M., Bjӧrklund, Å.K., Mjӧsberg, J., Erjefält, J.S., Cornelissen, Vosshenrich, C.A., Garcı́a-Ojeda, M.E., Samson-Villéger, S.I., Pasqualetto, V.,
A.S., Ros, X.R., Bal, S.M., Koning, J.J., Mebius, R.E., Mori, M., et al. (2017). Enault, L., Richard-Le Goff, O., Corcuff, E., Guy-Grand, D., Rocha, B., Cu-
Neuropilin-1 is expressed on lymphoid tissue residing lti-like group 3 innate mano, A., et al. (2006). A thymic pathway of mouse natural killer cell develop-
lymphoid cells and associated with ectopic lymphoid aggregates. Cell Rep ment characterized by expression of GATA-3 and CD127. Nat. Immunol. 7,
18, 1761–1773. 1217–1224.

Silver, J.S., Kearley, J., Copenhaver, A.M., Sanden, C., Mori, M., Yu, L., Pritch- Wallrapp, A., Riesenfeld, S.J., Burkett, P.R., Abdulnour, R.E., Nyman, J., Dio-
ard, G.H., Berlin, A.A., Hunter, C.A., Bowler, R., et al. (2016). Inflammatory trig- nne, D., Hofree, M., Cuoco, M.S., Rodman, C., Farouq, D., et al. (2017). The
gers associated with exacerbations of COPD orchestrate plasticity of group 2 neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature
innate lymphoid cells in the lungs. Nat Immunol 17, 626–635. 549, 351–356.
Sonnenberg, G.F., Monticelli, L.A., Elloso, M.M., Fouser, L.A., and Artis, D. Wang, S., Xia, P., Chen, Y., Qu, Y., Xiong, Z., Ye, B., Du, Y., Tian, Y., Yin, Z., Xu,
(2011). CD4(+) lymphoid tissue-inducer cells promote innate immunity in the Z., and Fan, Z. (2017). Regulatory innate lymphoid cells control innate intestinal
gut. Immunity 34, 122–134. inflammation. Cell 171, 201–216.e18.
Sonnenberg, G.F., Monticelli, L.A., Alenghat, T., Fung, T.C., Hutnick, N.A., Ku- Weizman, O.E., Adams, N.M., Schuster, I.S., Krishna, C., Pritykin, Y., Lau, C.,
nisawa, J., Shibata, N., Grunberg, S., Sinha, R., Zahm, A.M., et al. (2012). Degli-Esposti, M.A., Leslie, C.S., Sun, J.C., and O’Sullivan, T.E. (2017). ILC1
Innate lymphoid cells promote anatomical containment of lymphoid-resident confer early host protection at initial sites of viral infection. Cell 171, 795–
commensal bacteria. Science 336, 1321–1325. 808.e12.
Spencer, S.P., Wilhelm, C., Yang, Q., Hall, J.A., Bouladoux, N., Boyd, A., Nut- Wensveen, F.M., Jelenc  ic
, V., Valentic 
, S., Sestan, M., Wensveen, T.T., Theur-
man, T.B., Urban, J.F., Jr., Wang, J., Ramalingam, T.R., et al. (2014). Adapta- 
ich, S., Glasner, A., Mendrila, D., Stimac, D., Wunderlich, F.T., et al. (2015). NK
tion of innate lymphoid cells to a micronutrient deficiency promotes type 2 bar- cells link obesity-induced adipose stress to inflammation and insulin resis-
rier immunity. Science 343, 432–437. tance. Nat. Immunol. 16, 376–385.
Spits, H., Artis, D., Colonna, M., Diefenbach, A., Di Santo, J.P., Eberl, G.,
Wilhelm, C., Harrison, O.J., Schmitt, V., Pelletier, M., Spencer, S.P., Urban,
Koyasu, S., Locksley, R.M., McKenzie, A.N., Mebius, R.E., et al. (2013). Innate
J.F., Jr., Ploch, M., Ramalingam, T.R., Siegel, R.M., and Belkaid, Y. (2016).
lymphoid cells–a proposal for uniform nomenclature. Nat. Rev. Immunol. 13,
Critical role of fatty acid metabolism in ILC2-mediated barrier protection during
145–149.
malnutrition and helminth infection. J. Exp. Med. 213, 1409–1418.
Sui, P., Wiesner, D.L., Xu, J., Zhang, Y., Lee, J., Van Dyken, S., Lashua, A., Yu,
Wong, S.H., Walker, J.A., Jolin, H.E., Drynan, L.F., Hams, E., Camelo, A.,
C., Klein, B.S., Locksley, R.M., et al. (2018). Pulmonary neuroendocrine cells
Barlow, J.L., Neill, D.R., Panova, V., Koch, U., et al. (2012). Transcription factor
amplify allergic asthma responses. Science 360, eaan8546. https://doi.org/
RORa is critical for nuocyte development. Nat. Immunol. 13, 229–236.
10.1126/science.aan8546.
van de Pavert, S.A., Ferreira, M., Domingues, R.G., Ribeiro, H., Molenaar, R., Zaiss, D.M.W., Gause, W.C., Osborne, L.C., and Artis, D. (2015). Emerging
Moreira-Santos, L., Almeida, F.F., Ibiza, S., Barbosa, I., Goverse, G., et al. functions of amphiregulin in orchestrating immunity, inflammation, and tissue
(2014). Maternal retinoids control type 3 innate lymphoid cells and set the repair. Immunity 42, 216–226.
offspring immunity. Nature 508, 123–127. Zenewicz, L.A., Yancopoulos, G.D., Valenzuela, D.M., Murphy, A.J., Karow,
Veiga-Fernandes, H., and Artis, D. (2018). Neuronal-immune system cross-talk M., and Flavell, R.A. (2007). Interleukin-22 but not interleukin-17 provides pro-
in homeostasis. Science 359, 1465–1466. tection to hepatocytes during acute liver inflammation. Immunity 27, 647–659.
Veiga-Fernandes, H., Coles, M.C., Foster, K.E., Patel, A., Williams, A., Natar- Zhang, K., Xu, X., Pasha, M.A., Siebel, C.W., Costello, A., Haczku, A., MacNa-
ajan, D., Barlow, A., Pachnis, V., and Kioussis, D. (2007). Tyrosine kinase re- mara, K., Liang, T., Zhu, J., Bhandoola, A., et al. (2017). Cutting edge: notch
ceptor RET is a key regulator of Peyer’s patch organogenesis. Nature 446, signaling promotes the plasticity of group-2 innate lymphoid cells.
547–551. J. Immunol. 198, 1798–1803.

1066 Cell 174, August 23, 2018

You might also like