You are on page 1of 12

Immunity

Review

Barrier Epithelial Cells


and the Control of Type 2 Immunity
Hamida Hammad1,2,* and Bart N. Lambrecht1,2,3,*
1VIB Inflammation Research Center, Technologiepark 927, 9052 Ghent, Belgium
2Laboratory of Immunoregulation and Mucosal Immunology, Department of Respiratory Medicine, Ghent University, De Pintelaan 185, 9000
Ghent, Belgium
3Department of Pulmonary Medicine, ErasmusMC, Dr Molewaterplein 50, 3015 GE Rotterdam, the Netherlands

*Correspondence: hamida.hammad@irc.vib-ugent.be (H.H.), bart.lambrecht@irc.vib-ugent.be (B.N.L.)


http://dx.doi.org/10.1016/j.immuni.2015.07.007

Type-2-cell-mediated immunity, rich in eosinophils, basophils, mast cells, CD4+ T helper 2 (Th2) cells, and
type 2 innate lymphoid cells (ILC2s), protects the host from helminth infection but also drives chronic allergic
diseases like asthma and atopic dermatitis. Barrier epithelial cells (ECs) represent the very first line of defense
and express pattern recognition receptors to recognize type-2-cell-mediated immune insults like proteolytic
allergens or helminths. These ECs mount a prototypical response made up of chemokines, innate cytokines
such as interleukin-1 (IL-1), IL-25, IL-33, and thymic stromal lymphopoietin (TSLP), as well as the alarmins uric
acid, ATP, HMGB1, and S100 proteins. These signals program dendritic cells (DCs) to mount Th2-cell-medi-
ated immunity and in so doing boost ILC2, basophil, and mast cell function. Here we review the general mech-
anisms of how different stimuli trigger type-2-cell-mediated immunity at mucosal barriers and how this leads
to protection or disease.

Introduction class switching in B cells, resulting in the production of parasite-


More than 3 billion people suffer from helminth infection or or allergen-specific IgE that arms effector mast cells and baso-
allergic diseases, like asthma, rhinitis, atopic dermatitis (AD), phils that carry the high-affinity IgE receptor FcεRI; IL-5 is
food allergy, and anaphylaxis. Before the advent of sanitation responsible for the generation and activation of eosinophils in
and access to modern medicine, infection with intestinal nema- the bone marrow and tissues; IL-9 causes mast cell and ILC2
todes was ubiquitous, and it continues to be so in the absence of activation; IL-13 (and to a lesser extent IL-4) causes smooth
sanitation. Probably the greatest evolutionary driving force for muscle hyperreactivity, goblet cell metaplasia, mucus hyperse-
type-2-cell-mediated immunity to exist is the ancient co-evolu- cretion, and alternatively activated macrophages (AAM) differen-
tion of the mammalian mucosal immune system with these hel- tiation of monocytes; and Th2 cells also make amphiregulin
minths (Grencis et al., 2014). In some parts of the world, tick, (Areg) that promotes epithelial repair from injury (Zaiss et al.,
insect, and snake bites and stings are also a significant threat. 2015). The cytokine IL-3 is made by naive T cells early after
These infectious, toxic, and inflammatory disorders resemble T cell receptor TCR stimulation and enforces mast cell and baso-
each other closely in that affected tissues, often the mucosal phil functions but is not generally seen as a Th2-cell-associated
barriers and the skin, mount a ‘‘type 2’’ cell-mediated immune cytokine (Shen et al., 2008). Of note, not all Th2 cells make all Th2
response, rich in mast cells, basophils, eosinophils, and alterna- cytokines, and unique production of IL-4 is more typical of Th2
tively activated (or M2) macrophages that together release bene- cells in lymph nodes, where they boost IgE production; whereas
ficial pro-inflammatory mediators, toxin-neutralizing enzymes, both IL-4- and IL-13-producing Th2 cells are found in tissues
and helminth-killing toxins that, however, also cause significant (Liang et al., 2012). In addition to CD4+ Th2 cells, it is now also
problems of itching, redness, swelling, and altered function of clear that ILC2s that lack antigen-specific recognition receptors
the entire organ (Pulendran and Artis, 2012; Voehringer, 2013). are an important and much earlier source of the type-2-cell-
This type-2-cell-induced altered functional response can be associated cytokines IL-5, IL-13, Areg, and (to a lesser extent)
very appropriate; for example, in the case of helminth infection, IL-4 (Artis and Spits, 2015; Nussbaum et al., 2013). Even in the
mediator-induced smooth muscle contraction and increased complete absence of the adaptive immune system, ILC2s can
mucus production and epithelial cell proliferation favor worm control a type-2-cell-mediated innate immune response, leading
expulsion (Maizels et al., 2012). Nevertheless, the type-2-cell- to eosinophilic airway inflammation after protease allergen ex-
mediated response can also be very detrimental; for example, posure or to expulsion of helminths. In many cases, however,
the smooth muscle contraction and mucus production after ILC2s will have to conspire with CD4+ Th2 cells to sustain
inhalation of harmless allergens leads to chronic airway narrow- type-2-cell-mediated immunity and acquire full-blown type 2
ing in asthma and can also cause acute death in anaphylaxis effector cell function such as parasite expulsion (Oliphant
(Lambrecht and Hammad, 2015). et al., 2014). Here, we will review the pathways shared by barrier
Integrated type-2-cell-mediated immune responses are en- epithelial cells (ECs) to recruit and activate various players of
forced by the cytokines interleukin-4 (IL-4), IL-5, IL-9, and the type-2-cell-mediated immune response. We will focus on
IL-13, all known to be produced by CD4+ T helper 2 (Th2) lym- EC-derived factors (chemokines, cytokines, alarmins) induced
phocytes of the adaptive immune system. IL-4 drives isotype by diverse pattern recognition receptors and the importance of

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 29


Immunity

Review

peptides to T cells in the draining lymph nodes. Several studies


have reported that DCs are necessary and sufficient for inducing
adaptive Th2 cell responses to HDM allergen, papain, the hel-
minth Schistosoma mansoni, and proteins in inorganic crystals
(Hammad et al., 2010; Kool et al., 2008; Ohnmacht et al.,
2010). Although the exact DC subset initiating Th2 cell responses
to helminths has not been formally identified yet, in the lung and
in the skin, CD11b+CD172 (SIRP1a)+ conventional (c)DCs that
depend on the transcription factor IRF4 and KLF4 are necessary
and sufficient to induce adaptive Th2-cell-associated immune
responses (Akbari et al., 2014; Gao et al., 2013; Kumamoto
et al., 2013; Plantinga et al., 2013; Tussiwand et al., 2015; Wil-
liams et al., 2013). The mechanisms by which these DCs induce
Th2 cell immunity are poorly understood, compared with Th1,
Th17, and T regulatory (Treg) cell responses, but several studies
have shown the importance of the affinity of the major histocom-
patibility complex-T cell receptor (MHC-TCR) interaction, the
tumor necrosis factor (TNF) ligand family member OX40L, the
Figure 1. Epithelial Cells and Dendritic Cell Crosstalk Control Th2
Cell Responses in the Lung Notch ligand Jagged1, the cytokine IL-6, and the IRF4-induced
Upon allergen exposure, airway epithelial cells get activated to produce absence of IL-12 production (Akbari et al., 2014; Amsen et al.,
chemokines and innate cytokines that will instruct immature dendritic cells 2004). Another reason why the initiation of Th2 cell immunity is
(iDCs) and activate ILC2s. ILC2-derived IL-5 contributes to airway eosino-
philia, whereas IL-13 drives goblet cell metaplasia (GCM) and cDC2 migration
so obscure is that DCs do not produce the polarizing IL-4 cyto-
to MLNs. In the MLN, cDC2, with the help of IL-4 derived from basophils, will kine that drives Th2 cell immunity in many models. Basophils are
induce the differentiation of Th2 cells. IL-4 derived from Th2 cells promotes IgE recruited to the lymph nodes very early in the course of a type-2-
production by B cells. Th2 cells also produce IL-9, which causes ILC2 and
cell-associated immune response and help DCs to polarize naive
mast cell (MC) activation. Th2 cells also make amphiregulin (Areg) that pro-
motes epithelial repair from injury. T cells to the Th2 cell fate, by producing IL-4 (Figure 1; Hammad
et al., 2010; Sokol et al., 2008). After exposure to papain, DCs are
indeed not sufficient to polarize Th2 cell responses, but this also
barrier integrity and mucus quality in murine models and in pa- depends on the experimental setting (Ohnmacht et al., 2010).
tients with Th2-cell-associated disorders. The basophils boost ILC2 effector function in a STAT-6 tran-
scription factor-dependent manner by producing IL-4 (Doherty
Initiation of Type-2-Cell-Mediated Immunity Requires et al., 2012; Motomura et al., 2014), and basophils help eosino-
More than Just Dendritic Cells phil recruitment via IL-4-induced endothelial cell expression of
Although we have a good idea of what constitutes a type-2-cell- the integrin VCAM-1. Similarly, ILC2s are recruited early in a
mediated immune response, the mechanisms that initiate, type-2-cell-mediated immune response and might provide IL-
perpetuate, and resolve it remain enigmatic. This is partly caused 13 to boost DC function (mainly migration) and promote Th2
by the fact that the triggers that cause type-2-cell-associated im- cell responses. For adaptive Th2-cell-mediated immunity to
munity are diverse, ranging from large extracellular multicellular papain, IL-13 (not IL-4) is the polarizing cytokine (Halim et al.,
organisms (e.g., nematodes), venoms, tick saliva, excreta of 2014). Eosinophils recruited into tissues and lymph nodes can
mites and cockroaches, mold and bacterial cell wall compo- also be a source of IL-4 and can promote DC-driven Th2-cell-
nents, and enzymes, food components, animal dander, plant mediated immunity via release of eosinophil peroxidase released
pollen, and even the inorganic alum and uric acid crystals from granules (Chu et al., 2014). Under certain conditions, baso-
(Kool et al., 2008, 2011; Pulendran and Artis, 2012). In the labo- phils, eosinophils, and ILC2s can also directly function as anti-
ratory mouse, different models of parasite infection have been gen-presenting cells (APCs), although they are clearly less
used to study type-2-cell-mediated immunity, such as the hook- potent than DCs (Kambayashi and Laufer, 2014; Oliphant
worm Nippostrongylus brasiliensis, the roundworm Heligmoso- et al., 2014).
moides polygyrus, the whipworm Trichuris muris, and Trichinella
spiralis, a nematode with an atypical life cycle. As experimental Epithelial Cells Produce Chemokines that Recruit DCs,
allergens, most groups now use the complex house dust mite Basophils, and ILC2s
(HDM) extracts or cockroach extracts (containing a multitude Initiation of type-2-cell-mediated immunity in mucosal barriers
of proteins, lipids, glycoconjugates, and microbiome-derived and skin is highly complex, requiring many cell types to respond
molecules), the enzyme papain, or the particulate polysaccha- to type 2 immune stimuli in a rapid and coordinated manner, and
ride polymer chitin, a constituent of the cell wall of fungi and at the same time the barrier epithelial cells are very impermeable
the exoskeleton of insects and crustaceans. due to the physical barrier of luminal mucus or epidermal kera-
From these experimental studies, it has emerged that dendritic tins, on account of the tight junction and adherens junction belt
cells (DCs), ILC2s, and basophils act very collaboratively and up- connecting the ECs with one another. Many pathogens that
stream in the type-2-cell-mediated immune response cascade cause a type-2-cell-mediated immune response, like nema-
(Figure 1; Lambrecht and Hammad, 2014). Antigen-presenting todes, have evolved mechanisms to penetrate through tissues,
DCs that recognize an allergen or helminth present antigenic via production of proteases that cleave mucus and tight

30 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.


Immunity

Review

PGD2 and CCL17 (Staples et al., 2012). Production of CCL11


(eotaxin), CCL17, and CCL22 by ECs is further enforced by IL-
4 and IL-13 in a STAT-6-dependent manner and inhibited by
the type 1 cytokine interferon-g (IFN-g). This observation ex-
plains the profound influence of STAT6 deficiency in ECs on in-
duction of type-2-cell-mediated immunity in the lung and gut
(Doherty et al., 2012).

Epithelial Cells Produce Cytokines that Activate DCs,


Basophils, and ILC2s
Barrier ECs can also produce several immunoregulatory and
instructive cytokines that will shape the immune response. In
many instances, antigens are cleared without the induction of in-
flammatory responses. In the gut and in the lung, EC-derived
transforming growth factor-b (TGF-b) production promotes the
differentiation of DCs with a tolerogenic potential, important for
the control of tolerance to food and environmental inhaled anti-
Figure 2. Epithelial Cells Are Upstream of the Th2 Cell Cascade
Exposure to type 2 cell immune stimuli (proteases, b-glucans, chitin, etc.) gens (Iliev et al., 2009; Wang et al., 2009a). However, in response
favored by a barrier defect leads to the triggering of a wide range of pattern to most type-2-cell-mediated stimuli, barrier ECs produce a set
recognition receptors on barrier epithelial cells. ECs have evolved receptors of prototypical cytokines (IL-33, thymic stromal lymphopoietin
like the protease-activated receptors (PARs), Toll-like receptors (TLRs), C-type
lectins (CLR), and inflammasomes to sense invasion and penetration. The
[TSLP], and IL-25) that activate DCs to promote adaptive Th2
activation of these receptors will induce NF-kB activation, ROS production, cell immunity, and license the innate type 2 cell response by acti-
and the release of innate pro-Th2 cell cytokines like TSLP, GM-CSF, IL-25, and vation of ILC2s, basophils, eosinophils, and mast cells (Figure 2;
IL-33. In addition, barrier ECs have the capacity to regulate type 2 immunity
Allakhverdi et al., 2007; Fallon et al., 2006; Halim et al., 2012;
through the production of endogenous danger-associated molecular patterns
(DAMPs) or alarmins like IL-1a, HMGB1, uric acid, ATP, or S100 proteins. The Schmitz et al., 2005). This had led to the view that these cyto-
release of DAMPs can amplify the production of TSLP, GM-CSF, IL-25, and IL- kines constitute an innate ‘‘pro-Th2’’ cell cluster of cytokines
33 by ECs. In response to these stimuli, immature DCs (iDCs) upregulate that induce an almost stereotypical response, despite their
OX40L and Notch ligand expression, downregulate IL-12 production, and
produce Th2-cell-attracting chemokines like CCL17 and CCL22. EC-derived lack of structural homology and use of different receptor families
factors stimulate ILC2 proliferation and survival and their production of IL-5 (Saenz et al., 2008). Indeed, DCs that recognize antigens in the
and IL-13. presence of TSLP, IL-25, or IL-33 express Ox40L and produce
CCL17 and CCL22, but in the absence of IL-12 production (Bes-
junctions. Also many allergens, like the HDM allergen Dermato- nard et al., 2011; Chu et al., 2013). The fact that ILC2s have been
phagoides pteronyssinus (Der p 1) are proteinases. Not surpris- discovered is also mainly due to their expansion in vivo after
ingly, the barrier ECs have evolved receptors (like the prote- administration of IL-25, IL-33, and TSLP (Fallon et al., 2006; Neill
ase-activated receptors or PARs) or pathways (reactive oxygen et al., 2010; Saenz et al., 2010; Salimi et al., 2013). IL-25 and IL-
species [ROS] sensing) to sense invasion and penetration and 33 can induce different types of ILCs. IL-33 is found to induce
have integrated this response to the initiation of type-2-cell- ILC2s whereas IL-25 preferentially elicits multipotent progenitor
mediated immunity to clear the insult and initiate the repair pro- (MPP) type 2 cells, a new population of innate cells promoting
cess of the damaged barrier, e.g., via epithelial proliferation, type 2 cell immunity even in the absence of ILC2s (Saenz et al.,
collagen deposition, or altered mucus production (Pulendran 2010, 2013). IL-25 has recently been shown to also drive inflam-
and Artis, 2012). Type 2 cell stimuli on barrier ECs have in com- matory ILC2s, which can develop into natural IL-33-responsive
mon that they recruit DCs, ILC2s, basophils, mast cell progeni- ILC2s and contribute to helminth expulsion (Huang et al.,
tors, and eosinophils almost simultaneously. This happens 2015). The production of CCL17 and CCL22 chemokines is simi-
through an epithelial production of chemokines like CCL17 and larly boosted when ECs are exposed to TSLP or IL-25, again
CCL22 (acting on CCR4+ ILC2s, basophils, Th2, and Treg cells) leading to a feed forward loop that enforces type 2 immunity.
and of eotaxins like CCL-11, -24, and -26 (acting on CCR3+ eo- Some effects on type-2-cell-mediated immunity are not shared
sinophils and Th2 cells). In support, transgenic overexpression of by these cytokines, and there are some subtle tissue-specific ef-
CCL17 by keratinocytes is sufficient to induce an atopic derma- fects of the pro-Th2-cell-associated cytokines. TSLP, for
titis (AD)-like disorder. In many type-2-cell-mediated disease example, has the unique property of boosting the hematopoiesis
states like asthma, there is constitutive overexpression of of basophils (Siracusa et al., 2011). IL-33 is not always pro-in-
CCL17 in ECs (Sekiya et al., 2000). The barrier ECs also make flammatory and has also been shown to be involved in epithelial
prostaglandin D2 that acts on the CRTH2 receptor to recruit repair from injury. In the gut, epithelial IL-33 drives the activation
ILC2s, basophils, mast cells, and Th2 cells (Xue et al., 2014). A of IL-33R-expressing Treg cells, converting them into fully sup-
striking feature is that many of the recruited innate type 2 cells pressive cells, an effect that could be overcome by IL-23 (Schier-
will start producing CCL17, CCL22, and prostaglandin D2 ing et al., 2014).
(PGD2) themselves, leading to a feed-forward loop that enforces Another important actor of barrier type 2 immunity is granu-
the response. As only one example, production of CCL17 and locyte macrophage colony stimulating factor (GM-CSF), which
CCL22 are defining characteristics of AAMs, and the alveolar is released by ECs and stimulates the proliferation, maturation,
macrophages of human asthmatics are known to overproduce and function of APCs like DCs and macrophages and also

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 31


Immunity

Review

promotes eosinophil survival (Figure 2). Transgenic GM-CSF asthma, via interleukin-1 receptor antagonist (IL-1RA)-mediated
overexpression in the lungs of mice induces spontaneous suppression of IL-1a and HMGB1 (Duong et al., 2015). Increased
Th2 cell sensitization to the inhaled innocuous protein oval- amounts of HMGB1 are found in the skin of mice with AD lesions
bumin (OVA), via activation of IL-33 and Ox40L+ DCs (Llop- (Karuppagounder et al., 2014, 2015). Similarly, induction of type-
Guevara et al., 2014). In contrast, the neutralization of GM- 2-cell-mediated immunity to nematodes in the gut and to aller-
CSF abolishes sensitization to HDM allergen and attenuates gens in the skin requires IL-1. The IL-1R can also be triggered
the adjuvant effects of diesel particles on allergic sensitization by IL-1b, whose release often depends on the Nlrp3 inflamma-
(Bleck et al., 2006; Willart et al., 2012). More recent data show some. In keratinocytes, HDM has been shown to lead to Nlrp3-
that GM-CSF lowers the threshold for allergen to induce fea- dependent production of IL-1b (Dai et al., 2011). In the lung,
tures of allergic asthma (Llop-Guevara et al., 2014). Moreover, type-2-cell-mediated immunity to HDM is, however, not affected
ECs from asthmatic patients overproduce GM-CSF, suggest- by loss of caspase-1, Nlrp3, or Asc-1 (Kool et al., 2011).
ing that, in such patients, its production could be epigeneti- Psoriasin (S100A7) is a keratinocyte-derived anti-microbial
cally regulated (Hackett et al., 2011). Whether GM-CSF plays protein that is highly expressed in the skin of patients with
an equally important role in other barrier epithelia remains to atopic dermatitis. More recently, the expression of other mem-
be shown. bers of the S100 protein family, S100A8 and S100A9, has also
Finally, the TNF family member cytokine TL1A (TNFSF15) is been found to be increased in lesional AD skin (Gittler et al.,
made under conditions of type-2-cell-mediated immunity like 2012) and by keratinocytes after exposure to HDM (Jin et al.,
helminth infection and HDM exposure. ILC2s express high 2014). These proteins function as intracellular differentiation
amounts of DR3 (TNFRSF15), the receptor for TL1A, and ligation markers, and upon secretion, they act as amplifiers of inflam-
of DR3 leads to activation and proliferation of ILC2s, necessary matory responses in several inflammatory diseases via two re-
for worm expulsion and airway allergy, and to differentiation of ceptors, Toll-like receptor 4 (TLR4) and receptor of advanced
Th9 cells (Meylan et al., 2014; Richard et al., 2015; Yu et al., glycation endproducts (RAGE), that also binds to HMGB1.
2014). S100A9-treated keratinocytes show a dramatic RAGE-depen-
dent increase in the amounts of IL-33 mRNA, whereas no sig-
Epithelial-Derived DAMPs and Alarmins Promote nificant changes in TSLP or TNF-a mRNA expression are
Th2-Cell-Mediated Immunity observed (Jin et al., 2014). The role of these S100 proteins
In addition to the polarizing cytokines IL-25, IL-33, and TSLP, in vivo in relevant type-2-mediated diseases still needs to be
barrier ECs have the capacity to regulate type-2-cell-mediated fully addressed.
immunity through the production of endogenous danger-associ- Many of the alarmins and DAMPs are released by necrotic cell
ated molecular patterns (DAMPs) or alarmins. We have previ- death. However, there is little evidence for massive cell death of
ously shown that exposure of asthmatic patients to a single ECs after exposure to type-2-cell-mediated immune stimuli in
HDM administration via the airways leads to the release of ATP the lungs. Lung ECs have been shown, however, to recognize
and uric acid in the bronchoalveolar lavage (Idzko et al., 2007; and internalize apoptotic cells in a pathway requiring the actin
Kool et al., 2011). Although the exact cellular source of these binding protein Rac1. Recognition of apoptotic cells leads to a
DAMPs has not been formally identified in these patients, several dampened IL-33 response in ECs, and epithelial-cell-specific
reports show that ECs directly release ATP when stimulated loss of Rac1 exacerbates type-2-cell-mediated immune inflam-
by allergens like grass pollen or Alternaria alternata (O’Grady mation in an asthma model (Juncadella et al., 2013).
et al., 2013). In mice, a single exposure to HDM is sufficient to
trigger uric acid (UA) production by airway ECs (Kool et al., Pattern Recognition Receptors Prime Type-2-Cell-
2011), which is crucial in boosting the production of HDM- Mediated Immunity in Epithelial Cells
induced TSLP, GM-CSF, IL-25, and IL-33. The degradation of Pattern recognition receptor (PRR) triggering on ECs induces the
uric acid by uricase completely prevents allergic sensitization release of cytokines and chemokines that attract and activate
(Kool et al., 2011). The protease allergens bromelain and papain immune cells. As an example, HDM allergen extract leads to a
similarly induce UA production in the lung, leading to IL-33 and chemokine response in isolated lung ECs in a process requiring
TSLP induction (Hara et al., 2014). b-glucans, suggesting involvement of the dectin receptor (Na-
The alarmins High Mobility Group Box 1 (HMGB1), S100 family than et al., 2009). We and others have shown a few years ago,
proteins, IL-33, and IL-1a are normally localized in the nucleus. by using radiation-induced bone marrow chimeric mice, that
These can, however, be released by non-programmed cell TLR4 triggering on lung structural cells is required for the recog-
death, or they can be actively secreted in a process not requiring nition of HDM (and cockroach) and for the development of Th2-
the classical cellular secretion pathway involving vesicular trans- cell-associated asthma features (Hammad et al., 2009; Ullah
port from the Golgi complex. IL-1a and HMGB1 are expressed et al., 2014). More recently, via the Cre-Lox technology, it has
by lung ECs (Figure 2), and exposure to HDM has been shown been confirmed that Th2-cell-mediated immunity to HDM de-
to induce release in the lung fluid. Neutralization of IL-1a or velops only when TLR4 is triggered on Clara cell secretory pro-
HMGB1 is able to block production of the pro-Th2-cell-derived tein (CCSP)+ airway epithelial cells (McAlees et al., 2015).
cytokines IL-33, IL-25, and GM-CSF in the lung after HDM Indeed, HDM-induced ECs produce the chemokines CCL2 and
administration, showing that the epithelial alarmins act very up- CCL20, as well as IL-1a, HMGB1, uric acid, TSLP, GM-CSF,
stream in the allergic type 2 cascade (Hara et al., 2014; Ullah IL-25, and IL-33, in a TLR4-dependent manner (Hammad et al.,
et al., 2014; Willart et al., 2012). Adoptive transfer of mesen- 2009; Kool et al., 2011; Willart et al., 2012). How exactly TLR4
chymal stem cells has been shown to suppress features of gets activated on ECs is a matter of intense study. Some

32 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.


Immunity

Review

allergens like Der p 2 bind to TLR4 by mimicking the MD2 The Threshold of EC Activation Influences the Induction
molecule (Trompette et al., 2009). Cat and dog allergens facili- of Type-2-Cell-Mediated Immunity
tate TLR4 signaling by binding to LPS directly (Herre et al., The activation threshold of ECs and their capacity to activate
2013). The protease allergens from Aspergillus or endogenously immune responses can be profoundly influenced by prior ex-
released proteases lead to cleavage of fibrinogen into smaller posure to PAMPs or DAMPs, and this is certainly relevant for
fragments that can directly trigger TLR4 on lung ECs to boost barrier ECs, exposed to both the outside world and organismal
type-2-cell-mediated immunity (Millien et al., 2013). microbiome. This could lead to either increased PRR expression
Just like ECs in the lung, keratinocytes express a wide range of or modulation of signaling molecules downstream of PRR. As
PRRs and can therefore also be activated by numerous PAMPs such, exposure of ECs to respiratory syncitial virus (RSV) or to
or DAMPs. A recent study reported that papain, a cysteine pro- cigarette smoke (which causes damage to cells and the release
tease used as a model allergen, or Der p 2, a major allergen of of reactive oxygen species [ROS]) can sensitize the airway
HDM, are able to induce Th2-cell-mediated responses when epithelium to a subsequent environmental exposure (LPS) by
applied epicutaneously, in a TLR4-independent manner (Strem- altered expression and membrane localization of TLR4 (Pace
nitzer et al., 2014, 2015), despite earlier reports of TLR4 depen- et al., 2008). These data could explain why early exposure to
dence when the antigen was injected subcutaneously. Nickel RSV or cigarette smoke predisposes some individuals to
allergy is one of the most frequent forms of contact hypersensi- develop allergic inflammation and asthma. The activation
tivity (CHS). Only the human TLR4 molecule is able to bind to threshold of ECs can also be regulated at the level of signaling
nickel, so mice do not develop CHS. However, mice carrying a molecules downstream of PRRs. Triggering of most PRRs leads
transgenic human TLR4 became fully sensitive to CHS to nickel to the activation of the transcription factor NF-kB, and many of
(Schmidt et al., 2010). The exposure of human keratinocytes to the innate pro-Th2-cell-associated cytokines have an NF-kB
double-stranded RNA (TLR3 ligand), flagellin (TLR5 ligand), binding site in the promotor region (Swamy et al., 2010). The acti-
and diacylated lipopeptide (TLR2-TLR6 ligand) stimulates these vation of the classical NF-kB pathway within the lung epithelium
cells to express TSLP (Takai et al., 2014). Of note, Staphylo- has been shown to play a crucial role in allergic airway inflamma-
coccus aureus, a Gram-positive opportunistic bacterium that is tion. The constitutive activation of NF-kB in airway ECs has been
known to colonize the skin of patients with atopic dermatitis, is found to be sufficient to activate DCs, breach inhalational toler-
also able to stimulate TSLP release by keratinocytes. This could, ance, and promote sensitization to the harmless inhaled allergen
for instance, explain how the environment or infections can ovalbumin (OVA) (Ather et al., 2011), whereas inhibition of epithe-
contribute to the initiation and/or amplification of Th2-cell-type lial NF-kB reduced Th2 cell recruitment and airway remodeling
skin inflammation including atopic dermatitis (AD) and the (Broide et al., 2005). A more recent study confirmed the impor-
‘‘atopic march,’’ a process describing the progression from tance of this signaling pathway in ECs after exposure to HDM
AD to other allergic disorders such as asthma, rhinitis, or food (Tully et al., 2013). An early gene induced after NF-kB activation
allergy. is the ubiquitin-editing enzyme A20 (also known as TNFAIP3),
Many allergens are proteases and many nematodes release which acts as a negative regulator of inflammatory gene induc-
proteases. Protease-activated receptors (PAR) are character- tion in the epithelium, by deubiquitinating K63-linked ubiquitin
ized by a mechanism of self-activation after specific proteolytic chains on TRAF6, a protein mediating signal transduction from
cleavage of their extracellular domain. Four PARs have been TNF/Toll/IL-1 family members (Kelly et al., 2011). Delivery of
identified, and PAR2 is the only member activated by serine pro- the A20 binding protein ABIN-1 to lung ECs is associated with
teases. Almost all skin cell types express PAR2, including kera- a considerable reduction of all features of allergic asthma (El
tinocytes. Lesional skin of AD patients express high amounts of Bakkouri et al., 2005). The expression of A20 in the gut epithe-
PAR2, and the activation of PAR2 is thought to be involved in the lium is strongly correlated with the appearance of the gut micro-
pruritus observed in AD (Lee et al., 2010) through the induction of biome (Wang et al., 2009b). In the lung, induction of A20 might be
the innate pro-Th2-cell-associated cytokine TSLP by keratino- induced in the early postnatal period as well, requiring instruction
cytes (Moniaga et al., 2013), suggesting that PAR-2 antagonists from the microbial world. Future studies are required to fully un-
might represent a potential therapeutic for treating skin allergic derstand how NF-kB is activated or de-activated in ECs. Also,
disorders. Although in the lung, PAR2 activation by the proteo- the existence of epigenetic modifications in this pathway,
lytic activities of some allergens like mold (Chiu et al., 2007), caused by previous environmental triggers (viruses, pollutants),
cockroach (Page et al., 2010), or fungi (Boitano et al., 2011) which would alter the threshold of allergen recognition, need to
seems crucial for generating type responses, in the skin, this be further investigated.
phenomenon has not been well studied. In the skin, barrier homeostasis is also maintained by Notch
Even if C-type lectin receptors, TLRs, and PARs are crucial for signaling. Keratinocyte-specific loss of the metalloproteinase
type-2-cell-mediated immunity to develop, we urgently need to ADAM17 leads to reduced ligand-independent Notch-1 and
understand better how these receptors promote type-2-cell- -2 signaling, known to suppress production of TSLP, and these
associated immune responses when triggered on ECs, whereas mice develop a severe form of atopic dermatitis, driven by
they tend to stimulate Th1 and/or Th17 cell immunity when they TSLP, as well as skin barrier defects (Murthy et al., 2012).
are triggered on hematopoietic cells. It could be that down- The amounts of Notch target genes are severely reduced in
stream signaling from these receptors is fundamentally different. skin of patients with AD. How exactly Notch gets activated
It has also been insufficiently studied what impact binding to LPS on keratinocytes is a matter of intense study, but the skin
(e.g., by allergen components) or the presence of a concomitant microbiome has a suppressive effect on TSLP expression
ROS response has on PRR signaling in barrier ECs. (Yockey et al., 2013).

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 33


Immunity

Review
Figure 3. Altered Epithelial Barrier Function
Contributes to Type 2 Immunity
In the gut or in the lung (left), exposure to allergen-
or helminth-derived proteases induces a rapid and
transient destabilization of tight junction proteins
(claudin, occludin) and a loss of E-cadherin
expression by epithelial cells. In the skin and the
esophagus (right), genetic polymorphisms have
been described in several genes involved in
epithelial barrier integrity (filaggrin [FLG], SPINK5,
CAPN14). All these processes lead to a facilitated
penetration of antigens, an increased access to
DCs, and Th2 cell sensitization.

Normal mucosal barriers (skin, GI


tract, lung) can be viewed as very tight
barriers, whose function is to retain mois-
ture and to prevent the penetration of
allergens and microbes. Increasing evi-
dence from genome-wide association
studies (GWASs) and from experimental
models have suggested that a defective
Whatever the mechanism by which ECs can downregulate barrier function along with an exaggerated or abnormal immune
responses to type 2 immune stimuli, it will be important to response might contribute to the pathophysiology of common
further investigate the therapeutic potential of dampening EC re- allergic disorders like AD and to less common disorders of
sponses. The evolutionary driving pressure to dampen type-2- type-2-cell-mediated immunity like eosinophilic esophagitis
cell-mediated immunity might stem from the chronic relationship (Figure 3, right). As such, filaggrin (FLG) protein is crucial for
between host, its microbiome, parasitic nematodes (which often the terminal differentiation of keratinocytes. In case of FLG mu-
occupy the host for life), and the ubiquitous nature of nematode tations, the epidermal barrier is disrupted due to decreased tight
infection before the advent of modern medicine. junction protein expression, and this might lead to the develop-
ment of percutaneous allergen sensitization and Th2 cell re-
Altered Epithelial Barrier Function Contributes to sponses (Mócsai et al., 2014). FLG mutations are correlated
Type-2-Cell-Mediated Immunity with a higher risk of developing allergic diseases like AD, allergic
The relationship between exposure to some inhaled antigens rhinitis, food allergies, and atopic asthma even though FLG is not
and barrier dysfunction is complex because some proteolytically expressed in airway ECs, illustrating that asthma development in
active allergens like HDM or fungal proteases could be at the individuals with AD might be secondary to allergic sensitization
origin of barrier dysfunction but, at the same time, barrier that occurs after the breakdown of the epidermal skin barrier
dysfunction might facilitate allergens to gain access to immune (Ring et al., 2012). Tight junction (TJ) proteins like claudins are
cells. In lung biopsies of asthmatic patients, tight junctions located on keratinocytes of the stratum granulosum, situated
(mostly E-cadherin) are defective, and when ECs from asth- just below the stratum corneum (Kuo et al., 2013). Reduced
matics are cultured ex vivo, they also show defective tight junc- expression of the TJ proteins claudin-1 and claudin-23 is
tion development (Xiao et al., 2011). Allergen extracts can also observed only in patients with AD (De Benedetto et al., 2011).
induce a rapid and transient destabilization of tight junctions Once these two physical barriers (filaggrin and tight junctions)
and a loss of E-cadherin expression (Figure 3, left). This effect are breached, there is an increased risk for AD patients to get
has been shown to be dependent on the enzymatic activity of colonized by Staphylococcus aureus. Because S. aureus can
HDM, cockroach, pollen, and fungi. It is important to note that produce high amounts of serine proteases that can degrade
DCs receive a tonic inhibitory signal through homotypic E-cad- the skin barrier further (Schlievert et al., 2010), this can contribute
herin interactions between DCs and ECs (Nawijn et al., 2011). to exacerbation of Th2-cell-associated symptoms of the dis-
A defective E-cadherin expression could activate DCs, and ease. The balance between proteases and protease inhibitors
therefore facilitate allergic sensitization. Moreover, a loss of E- therefore seems to be important in maintaining homeostasis in
cadherin in cultured ECs results in the increased production of the skin. Lymphoepithelial Kazal-type-related inhibitor (LEKTI)
TSLP (Heijink et al., 2007), and this event could constitute an is an inhibitor of several serine proteases in the skin, thereby con-
early step in the break of inhalational tolerance leading to Th2 trolling epidermal barrier function (Ovaere et al., 2009). A prema-
cell sensitization and asthma development. E-cadherin is also ture stop codon mutation in SPINK5, which encodes LEKTI, is
a ligand of KLRG-1 expressed by human and mouse ILC2s. Liga- associated with Netherton syndrome, a rare autosomal-reces-
tion of KLRG-1 by E-cadherin suppresses the production of IL-5 sive skin disease with severe skin inflammation and constant
and IL-13 by ILC2s, and in this way, a loss of E-cadherin might allergic manifestations (Di et al., 2009). In addition, several
promote type-2-cell-mediated innate immunity (Salimi et al., studies have reported that the polymorphisms in SPINK5 are
2013). This is relevant in the skin, because the expression of E- associated with AD (Walley et al., 2001). In eosinophilic esopha-
cadherin is reduced in patients with AD. gitis, recent studies have also shown evidence for defective

34 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.


Immunity

Review

epithelial barrier function, with decreased expression of LEKTI modimer. In the gut and in the lung, RELMb is induced by the Th2
and filaggrin, and SNPs in the CAPN14 gene that codes for an cell cytokines IL-4 and IL-13 produced during a helminth infec-
intracellular cysteine protease belonging to the calpain family tion or after exposure to fungal allergens (Herbert et al., 2009).
(Simon et al., 2015; Sleiman et al., 2014). Calpains have been RELMb has been shown to have very distinct effects in the gut
associated with asthma development and the blockade of these and in the lung. In the gut, RELMb is crucial in promoting protec-
proteins in an asthma model results in a marked improvement tion against gastrointestinal worm infection and against colitis
of asthma features (Aich et al., 2012). Altogether, these data development by regulating mucous secretion, thus contributing
suggest that a very fine balance between proteases, protease in- to barrier integrity (Krimi et al., 2008). In the lung, RELMb favors
hibitors, and junctions contributing to optimal barrier function leukocyte recruitment, collagen deposition, and lung remodeling
therefore seems to be crucial in maintaining immune homeosta- (Fang et al., 2015).
sis at barrier sites.
Alterations in EC-Repairing Factors Might Promote
Production of Mucus and Mucus-Associated Bioactive Type-2-Cell-Mediated Immunity
Molecules Promote Type-2-Cell-Mediated Immunity Given the complex composition of mucus and the metabolites
The airways and the intestinal mucosa are covered with a thick and antimicrobial peptides contained in it, it will be important
layer of mucous, consisting of a highly ordered well-hydrated to study whether type 2 immune cells sense mucus composition
gel composed of high-molecular-weight glycosylated proteins, directly, and whether mucus composition is altered by type 2 im-
produced by goblet cells, and containing antimicrobial peptides mune cells. In this regard, IL-22-producing ILC3s have already
and metabolites derived from commensal microbiota, and form- been shown to alter the glycosylation state of gut epithelial cells
ing yet another barrier to the entry of type 2 cell stimuli. One of the in a microbiome-dependent manner, leading to protection from
features of allergic asthma, and one of the major defense mech- invasive pathogens (Goto et al., 2014). IL-22 is also found in
anisms against intestinal nematodes, is IL-13-driven goblet cell the airways of asthmatic humans and mice, but the exact source
metaplasia (GCM), an increase in goblet cell numbers mostly (Th22 cells or NKp46+ ILCs) and function requires further study.
due to the transdifferentiation of airway ciliated and Clara cells Whereas Il22 / mice have severely reduced type 2 immunity in
or enterocytes to goblet cells, resulting in more mucus produc- an OVA-alum model of asthma (Besnard et al., 2011), recombi-
tion, of a different quality. In a healthy gut, expression of the nant IL-22 was also shown to reduce airway inflammation by
mucins Muc2 and Muc3 is predominant (Buisine et al., 2001). reducing the epithelial production of IL-25 (Takahashi et al.,
Exposure of enterocytes to IL-13 and GCM leads to the produc- 2011). It is unclear at present whether alterations in mucus con-
tion of more highly charged glycoproteins and of Mucin 5AC, an tent or glycosylation would contribute to the suppression of IL-
important factor for nematode expulsion in the gut (Hasnain 25. During nematode infection, IL-22 has been found responsible
et al., 2011a). Several transcription factors have been involved for the induction of GCM and worm expulsion (Turner et al.,
in this process (Hasnain et al., 2011c; Le Cras et al., 2011). A 2013). Amphiregulin (made by Th2, ILC2, and mast cells) has
recent paper has described that SPDEF and Foxa3 overexpres- also been shown to alter the mucin composition in the lung
sion in ECs is sufficient to induce airway goblet cell differentiation and gut (Okumura et al., 2005). Amphiregulin also causes a hy-
in neonatal and adult lungs, but strikingly, this also leads to perproliferative state in the gut epithelium, which eventually
eosinophilic airway inflammation and the recruitment of group leads to parasite expulsion. Undoubtedly, this hyperproliferative
2 ILCs and DCs (Rajavelu et al., 2015). This is caused by response will also alter cytokine production by ECs.
the SPDEF- and Foxa3-induced release of EC-derived IL-33, Trefoil factors are epithelial-derived cytokines that promote
TSLP, and GM-CSF. SPDEF- and Foxa3-induced GCM might epithelial healing, mucosal secretion, and intestinal integrity.
therefore play a role in the establishment of Th2 cell polarization, Allergens like HDM, Aspergillus, and OVA lead to increased pro-
influencing subsequent responses to different environmental duction of TFF2 in lung ECs, in a STAT6-dependent manner (Ni-
exposures. Similarly, Muc5AC-deficient mice infected with the kolaidis et al., 2003). Although TFF-2 is required for repair of lung
nematode T. muris had increased concentrations of IFN-g (Has- injury after N. brasiliensis passage through the lungs, TFF2 is also
nain et al., 2011a). Clearly more research is needed before a potent inducer of IL-33. TFF2 is thus able to promote allergen-
we understand the full impact of altered secretory capacity of driven airway type-2-cell-mediated immunity.
ECs on immune polarization in the mucosa. One possibility is
that secretion of mucins interferes with the unfolded protein Epithelial Cytokines in Human Type-2-Cell-Associated
response ([UPR], a cellular response to stress), that can intersect Chronic Inflammatory Diseases
with inflammatory response pathways, including NF-kB activa- Several recent reviews deal with epithelial pathways in chronic
tion (Janssens et al., 2014). In airway epithelial cells of human type-2-cell-mediated immune diseases, like asthma, atopic
asthmatics, IL-13-induced GCM leads to induction of Xbp1 dermatitis, and chronic helminth infections, so we will only briefly
splicing (Martino et al., 2013). This process is very similar to illustrate some studies on the importance of ECs in controlling
mucus production in the gut and depends on the mucosal- these diseases (Grencis et al., 2014; Hallstrand et al., 2014; Lam-
restricted endoplasmic reticulum (ER) stress sensor IRE1b, lead- brecht and Hammad, 2012). The evidence for increased produc-
ing to induction of the protein disulfide isomerase Agr2. Genetic tion of pro-Th2 cell cytokines by ECs is overwhelming in asthma,
polymorphisms in XBP1 and AGR2 are associated with IBD and and many genetic association studies point to an important role
could act far upstream in the immunological cascade. for ECs in this disease. In mice, the overexpression of TSLP in the
RELMb is produced by goblet cells in the intestine and in the lung epithelium leads to the spontaneous development of an
lung and is secreted into the lumen at high concentration as a ho- asthma-like disease (Zhou et al., 2005). A wide variety of stimuli,

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 35


Immunity

Review

including proteolytic allergens, diesel-exhaust particles, ciga- at the time of epicutaneous sensitization strongly reduces the
rette smoke, and respiratory viruses can induce the epithelial ability of skin-derived DCs to induce the proliferation and Th2-
release of TSLP (Bleck et al., 2010). The lung epithelium of asth- cell-associated cytokine production by allergen-specific CD4+
matics contains a higher number of TSLP+ cells, and the bron- T cells (Tordesillas et al., 2014).
choalveolar lavage of these patients had higher concentrations
of TSLP protein compared to healthy individuals (Bleck et al., Conclusions
2015). Genetic polymorphisms in the promoter region of TSLP There has been great progress in the study of type-2-cell-medi-
and IL33R (IL1RL1) are associated with increased risk of asthma ated immune responses driven by allergens and nematodes in
(Harada et al., 2011). Of note, airway ECs from asthmatic pa- barrier sites of the body. From these studies, a picture emerges
tients secrete more TSLP when exposed to dsRNA (viral analog) whereby epithelial cells constitute a first physical, chemical, and
(Brandelius et al., 2011). This might partly explain why patients immunological barrier that can be seen almost as an integral
with asthma get an exacerbation of their disease after being part of the innate defense mechanism. The ECs express pattern
exposed to such triggers (Jackson and Johnston, 2010). It is recognition receptors, integrate information from many receptors
therefore very likely that an aberrant production of TSLP might simultaneously, and talk to the innate and adaptive immune sys-
influence both the susceptibility of certain individuals to develop tem cells by releasing chemokines, cytokines, and alarmins. This
asthma and the clinical complications developing after exposure recognition event is tightly regulated and influenced by prior expo-
to some environmental triggers. Although IL-25 is produced by sure history and is terminated appropriately when the type-2-cell-
ECs of patients with Th2-cell-high corticosteroid-responsive mediated immune stimulus disappears. In certain chronic inflam-
asthma (Cheng et al., 2014), the precise role of this cytokine is matory diseases like asthma and AD, dysregulated EC responses
still unclear. Recent studies have linked IL-25 and IL-33 to vi- or barrier function could be the driver of the disease process. In the
rus-induced asthma exacerbations (Bousquet et al., 2014). future, the challenge will be to translate this new knowledge to
Although there are no reports showing virus-induced increased novel preventive or curative strategies for these disorders.
IL-33 production by ECs in asthmatic patients, it is very plausible
that the injury caused by respiratory viruses to ECs might lead to REFERENCES
enhanced IL-25 and IL-33 production, thereby driving an acute
exacerbation of allergic asthma via DC and/or ILC2 activation Aich, J., Mabalirajan, U., Ahmad, T., Agrawal, A., and Ghosh, B. (2012). Loss-
(Kumar et al., 2014). Also, primary ECs from allergic children of-function of inositol polyphosphate-4-phosphatase reversibly increases the
severity of allergic airway inflammation. Nat. Commun. 3, 877.
exposed to RSV are found to produce low amounts of type I
IFN and had higher viral shedding compared to healthy ECs Akbari, M., Honma, K., Kimura, D., Miyakoda, M., Kimura, K., Matsuyama, T.,
and Yui, K. (2014). IRF4 in dendritic cells inhibits IL-12 production and controls
(Spann et al., 2014). Of note, these features are absent when Th1 immune responses against Leishmania major. J. Immunol. 192, 2271–
adult ECs were used (Patel et al., 2014). 2279.
TSLP has also emerged as a key cytokine in the pathogenesis
Allakhverdi, Z., Comeau, M.R., Jessup, H.K., Yoon, B.R., Brewer, A., Chartier,
of Th2-cell-associated AD (Ziegler and Artis, 2010). Early studies S., Paquette, N., Ziegler, S.F., Sarfati, M., and Delespesse, G. (2007). Thymic
have shown that TSLP overexpression in keratinocytes leads to stromal lymphopoietin is released by human epithelial cells in response to mi-
the spontaneous development of AD-like disease in mice (Li crobes, trauma, or inflammation and potently activates mast cells. J. Exp.
Med. 204, 253–258.
et al., 2005). Moreover, TSLP overexpression in keratinocytes
during epicutaneous OVA or HDM sensitization has the potential Amsen, D., Blander, J.M., Lee, G.R., Tanigaki, K., Honjo, T., and Flavell, R.A.
(2004). Instruction of distinct CD4 T helper cell fates by different notch ligands
to aggravate features of asthma upon re-exposure to the allergen on antigen-presenting cells. Cell 117, 515–526.
via the airways. Of note, epicutaneous sensitization with peanut
Artis, D., and Spits, H. (2015). The biology of innate lymphoid cells. Nature 517,
promotes intestinal food allergy through a mechanism involving
293–301.
TSLP production by keratinocytes (Noti et al., 2014). These
data strongly suggest that TSLP (over)produced in the skin could Ather, J.L., Hodgkins, S.R., Janssen-Heininger, Y.M., and Poynter, M.E.
(2011). Airway epithelial NF-kB activation promotes allergic sensitization to
be a risk factor for the development of Th2-cell-mediated dis- an innocuous inhaled antigen. Am. J. Respir. Cell Mol. Biol. 44, 631–638.
eases like allergic airway inflammation or food allergy. Increased
Besnard, A.G., Togbe, D., Guillou, N., Erard, F., Quesniaux, V., and Ryffel, B.
expression of TSLP has been demonstrated in skin keratinocytes
(2011). IL-33-activated dendritic cells are critical for allergic airway inflamma-
from AD patients, and elevated serum TSLP concentrations have tion. Eur. J. Immunol. 41, 1675–1686.
been reported in AD children. The association of AD with a poly-
Bleck, B., Tse, D.B., Jaspers, I., Curotto de Lafaille, M.A., and Reibman, J.
morphism of the IL1RL1 gene (Shimizu et al., 2005) suggests that (2006). Diesel exhaust particle-exposed human bronchial epithelial cells
the IL-33-ST2 system might contribute to the development or induce dendritic cell maturation. J. Immunol. 176, 7431–7437.
worsening of the disease. IL-33 expression is upregulated in ker- Bleck, B., Tse, D.B., Gordon, T., Ahsan, M.R., and Reibman, J. (2010). Diesel
atinocytes and endothelial cells in AD (Oboki et al., 2010; Savinko exhaust particle-treated human bronchial epithelial cells upregulate Jagged-1
et al., 2012), as well as in the serum of AD patients (Tamagawa- and OX40 ligand in myeloid dendritic cells via thymic stromal lymphopoietin.
J. Immunol. 185, 6636–6645.
Mineoka et al., 2014). The specific overexpression of IL-33 by
keratinocytes leads to the spontaneous development of derma- Bleck, B., Kazeros, A., Bakal, K., Garcia-Medina, L., Adams, A., Liu, M., Lee,
titis with eosinophilic infiltration and activation of IL-5-producing R.A., Tse, D.B., Chiu, A., Grunig, G., et al. (2015). Coexpression of type 2 im-
mune targets in sputum-derived epithelial and dendritic cells from asthmatic
ILC2s (Imai et al., 2013). Moreover, IL-33 release by keratinocytes patients. J. Allergy Clin. Immunol. Published online March 23, 2015. http://
has been shown to drive the pro-Th2 cell capacities of skin DCs in dx.doi.org/10.1016/j.jaci.2014.12.1950.
a food allergy model based on the epicutaneous application of Boitano, S., Flynn, A.N., Sherwood, C.L., Schulz, S.M., Hoffman, J., Gruzinova,
peanut extracts (Tordesillas et al., 2014). The blockade of ST2 I., and Daines, M.O. (2011). Alternaria alternata serine proteases induce lung

36 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.


Immunity

Review
inflammation and airway epithelial cell activation via PAR2. Am. J. Physiol. airways fibroblasts to effect remodelling in asthma: from mouse to man. Clin.
Lung Cell. Mol. Physiol. 300, L605–L614. Exp. Allergy 45, 940–952.

Bousquet, J., Gern, J.E., Martinez, F.D., Anto, J.M., Johnson, C.C., Holt, P.G., Gao, Y., Nish, S.A., Jiang, R., Hou, L., Licona-Limón, P., Weinstein, J.S., Zhao,
Lemanske, R.F., Jr., Le Souëf, P.N., Tepper, R.S., von Mutius, E.R., et al. H., and Medzhitov, R. (2013). Control of T helper 2 responses by transcription
(2014). Birth cohorts in asthma and allergic diseases: report of a NIAID/ factor IRF4-dependent dendritic cells. Immunity 39, 722–732.
NHLBI/MeDALL joint workshop. J. Allergy Clin. Immunol. 133, 1535–1546.
Gittler, J.K., Shemer, A., Suárez-Fariñas, M., Fuentes-Duculan, J., Gulewicz,
Brandelius, A., Yudina, Y., Calvén, J., Bjermer, L., Andersson, M., Persson, C., K.J., Wang, C.Q., Mitsui, H., Cardinale, I., de Guzman Strong, C., Krueger,
and Uller, L. (2011). dsRNA-induced expression of thymic stromal lympho- J.G., and Guttman-Yassky, E. (2012). Progressive activation of T(H)2/T(H)22
poietin (TSLP) in asthmatic epithelial cells is inhibited by a small airway cytokines and selective epidermal proteins characterizes acute and chronic
relaxant. Pulm. Pharmacol. Ther. 24, 59–66. atopic dermatitis. J. Allergy Clin. Immunol. 130, 1344–1354.

Broide, D.H., Lawrence, T., Doherty, T., Cho, J.Y., Miller, M., McElwain, K., Goto, Y., Obata, T., Kunisawa, J., Sato, S., Ivanov, I.I., Lamichhane, A., Take-
McElwain, S., and Karin, M. (2005). Allergen-induced peribronchial fibrosis yama, N., Kamioka, M., Sakamoto, M., Matsuki, T., et al. (2014). Innate
and mucus production mediated by IkappaB kinase beta-dependent genes lymphoid cells regulate intestinal epithelial cell glycosylation. Science 345,
in airway epithelium. Proc. Natl. Acad. Sci. USA 102, 17723–17728. 1254009.

Buisine, M.P., Desreumaux, P., Leteurtre, E., Copin, M.C., Colombel, J.F., Grencis, R.K., Humphreys, N.E., and Bancroft, A.J. (2014). Immunity to gastro-
Porchet, N., and Aubert, J.P. (2001). Mucin gene expression in intestinal intestinal nematodes: mechanisms and myths. Immunol. Rev. 260, 183–205.
epithelial cells in Crohn’s disease. Gut 49, 544–551.
Hackett, T.L., Singhera, G.K., Shaheen, F., Hayden, P., Jackson, G.R., Hegele,
Cheng, D., Xue, Z., Yi, L., Shi, H., Zhang, K., Huo, X., Bonser, L.R., Zhao, J., Xu, R.G., Van Eeden, S., Bai, T.R., Dorscheid, D.R., and Knight, D.A. (2011).
Y., Erle, D.J., and Zhen, G. (2014). Epithelial interleukin-25 is a key mediator in Intrinsic phenotypic differences of asthmatic epithelium and its inflammatory
Th2-high, corticosteroid-responsive asthma. Am. J. Respir. Crit. Care Med. responses to respiratory syncytial virus and air pollution. Am. J. Respir. Cell
190, 639–648. Mol. Biol. 45, 1090–1100.

Chiu, L.L., Perng, D.W., Yu, C.H., Su, S.N., and Chow, L.P. (2007). Mold Halim, T.Y., Krauss, R.H., Sun, A.C., and Takei, F. (2012). Lung natural helper
allergen, pen C 13, induces IL-8 expression in human airway epithelial cells cells are a critical source of Th2 cell-type cytokines in protease allergen-
by activating protease-activated receptor 1 and 2. J. Immunol. 178, 5237– induced airway inflammation. Immunity 36, 451–463.
5244.
Halim, T.Y., Steer, C.A., Mathä, L., Gold, M.J., Martinez-Gonzalez, I.,
Chu, D.K., Llop-Guevara, A., Walker, T.D., Flader, K., Goncharova, S., Bou- McNagny, K.M., McKenzie, A.N., and Takei, F. (2014). Group 2 innate
dreau, J.E., Moore, C.L., Seunghyun In, T., Waserman, S., Coyle, A.J., et al. lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated
(2013). IL-33, but not thymic stromal lymphopoietin or IL-25, is central to allergic lung inflammation. Immunity 40, 425–435.
mite and peanut allergic sensitization. J. Allergy Clin. Immunol. 131, 187–
Hallstrand, T.S., Hackett, T.L., Altemeier, W.A., Matute-Bello, G., Hansbro,
200.e1, 8.
P.M., and Knight, D.A. (2014). Airway epithelial regulation of pulmonary im-
mune homeostasis and inflammation. Clin. Immunol. 151, 1–15.
Chu, D.K., Jimenez-Saiz, R., Verschoor, C.P., Walker, T.D., Goncharova, S.,
Llop-Guevara, A., Shen, P., Gordon, M.E., Barra, N.G., Bassett, J.D., et al.
Hammad, H., Chieppa, M., Perros, F., Willart, M.A., Germain, R.N., and Lam-
(2014). Indigenous enteric eosinophils control DCs to initiate a primary Th2 im-
brecht, B.N. (2009). House dust mite allergen induces asthma via Toll-like re-
mune response in vivo. J. Exp. Med. 211, 1657–1672.
ceptor 4 triggering of airway structural cells. Nat. Med. 15, 410–416.
Dai, X., Sayama, K., Tohyama, M., Shirakata, Y., Hanakawa, Y., Tokumaru, S., Hammad, H., Plantinga, M., Deswarte, K., Pouliot, P., Willart, M.A., Kool, M.,
Yang, L., Hirakawa, S., and Hashimoto, K. (2011). Mite allergen is a danger Muskens, F., and Lambrecht, B.N. (2010). Inflammatory dendritic cells–not ba-
signal for the skin via activation of inflammasome in keratinocytes. J. Allergy sophils–are necessary and sufficient for induction of Th2 immunity to inhaled
Clin. Immunol. 127, 806–14.e1, 4. house dust mite allergen. J. Exp. Med. 207, 2097–2111.
De Benedetto, A., Rafaels, N.M., McGirt, L.Y., Ivanov, A.I., Georas, S.N., Chea- Hara, K., Iijima, K., Elias, M.K., Seno, S., Tojima, I., Kobayashi, T., Kephart,
dle, C., Berger, A.E., Zhang, K., Vidyasagar, S., Yoshida, T., et al. (2011). Tight G.M., Kurabayashi, M., and Kita, H. (2014). Airway uric acid is a sensor of
junction defects in patients with atopic dermatitis. J. Allergy Clin. Immunol. inhaled protease allergens and initiates type 2 immune responses in respira-
127, 773–86.e1, 7. tory mucosa. J. Immunol. 192, 4032–4042.
Di, W.L., Hennekam, R.C., Callard, R.E., and Harper, J.I. (2009). A heterozy- Harada, C., Kawaguchi, T., Ogata-Suetsugu, S., Yamada, M., Hamada, N.,
gous null mutation combined with the G1258A polymorphism of SPINK5 Maeyama, T., Souzaki, R., Tajiri, T., Taguchi, T., Kuwano, K., and Nakanishi,
causes impaired LEKTI function and abnormal expression of skin barrier pro- Y. (2011). EGFR tyrosine kinase inhibition worsens acute lung injury in mice
teins. Br. J. Dermatol. 161, 404–412. with repairing airway epithelium. Am. J. Respir. Crit. Care Med. 183, 743–751.
Doherty, T.A., Khorram, N., Chang, J.E., Kim, H.K., Rosenthal, P., Croft, M., Hasnain, S.Z., Evans, C.M., Roy, M., Gallagher, A.L., Kindrachuk, K.N., Barron,
and Broide, D.H. (2012). STAT6 regulates natural helper cell proliferation dur- L., Dickey, B.F., Wilson, M.S., Wynn, T.A., Grencis, R.K., and Thornton, D.J.
ing lung inflammation initiated by Alternaria. Am. J. Physiol. Lung Cell. Mol. (2011a). Muc5ac: a critical component mediating the rejection of enteric nem-
Physiol. 303, L577–L588. atodes. J. Exp. Med. 208, 893–900.

Duong, K.M., Arikkatt, J., Ullah, M.A., Lynch, J.P., Zhang, V., Atkinson, K., Sly, Hasnain, S.Z., Thornton, D.J., and Grencis, R.K. (2011c). Changes in the
P.D., and Phipps, S. (2015). Immunomodulation of airway epithelium cell acti- mucosal barrier during acute and chronic Trichuris muris infection. Parasite
vation by mesenchymal stromal cells ameliorates HDM-induced airway inflam- Immunol. 33, 45–55.
mation in mice. Am. J. Respir. Cell Mol. Biol. Published online March 19, 2015.
http://dx.doi.org/10.1165/rcmb.2014-0431OC. Heijink, I.H., Kies, P.M., Kauffman, H.F., Postma, D.S., van Oosterhout, A.J.,
and Vellenga, E. (2007). Down-regulation of E-cadherin in human bronchial
El Bakkouri, K., Wullaert, A., Haegman, M., Heyninck, K., and Beyaert, R. epithelial cells leads to epidermal growth factor receptor-dependent Th2
(2005). Adenoviral gene transfer of the NF-kappa B inhibitory protein ABIN-1 cell-promoting activity. J. Immunol. 178, 7678–7685.
decreases allergic airway inflammation in a murine asthma model. J. Biol.
Chem. 280, 17938–17944. Herbert, D.R., Yang, J.Q., Hogan, S.P., Groschwitz, K., Khodoun, M., Munitz,
A., Orekov, T., Perkins, C., Wang, Q., Brombacher, F., et al. (2009). Intestinal
Fallon, P.G., Ballantyne, S.J., Mangan, N.E., Barlow, J.L., Dasvarma, A., He- epithelial cell secretion of RELM-beta protects against gastrointestinal worm
wett, D.R., McIlgorm, A., Jolin, H.E., and McKenzie, A.N. (2006). Identification infection. J. Exp. Med. 206, 2947–2957.
of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and
IL-13 at the onset of helminth expulsion. J. Exp. Med. 203, 1105–1116. Herre, J., Grönlund, H., Brooks, H., Hopkins, L., Waggoner, L., Murton, B.,
Gangloff, M., Opaleye, O., Chilvers, E.R., Fitzgerald, K., et al. (2013). Allergens
Fang, C.L., Yin, L.J., Sharma, S., Kierstein, S., Wu, H.F., Eid, G., Haczku, A., as immunomodulatory proteins: the cat dander protein Fel d 1 enhances TLR
Corrigan, C.J., and Ying, S. (2015). Resistin-like molecule-b (RELM-b) targets activation by lipid ligands. J. Immunol. 191, 1529–1535.

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 37


Immunity

Review
Huang, Y., Guo, L., Qiu, J., Chen, X., Hu-Li, J., Siebenlist, U., Williamson, P.R., Kuo, I.H., Yoshida, T., De Benedetto, A., and Beck, L.A. (2013). The cutaneous
Urban, J.F., Jr., and Paul, W.E. (2015). IL-25-responsive, lineage-negative innate immune response in patients with atopic dermatitis. J. Allergy Clin. Im-
KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. munol. 131, 266–278.
Nat. Immunol. 16, 161–169.
Lambrecht, B.N., and Hammad, H. (2012). The airway epithelium in asthma.
Idzko, M., Hammad, H., van Nimwegen, M., Kool, M., Willart, M.A., Muskens, Nat. Med. 18, 684–692.
F., Hoogsteden, H.C., Luttmann, W., Ferrari, D., Di Virgilio, F., et al. (2007).
Extracellular ATP triggers and maintains asthmatic airway inflammation by Lambrecht, B.N., and Hammad, H. (2014). Innate immune cells to the help. Im-
activating dendritic cells. Nat. Med. 13, 913–919. munity 40, 313–314.

Lambrecht, B.N., and Hammad, H. (2015). The immunology of asthma. Nat.


Iliev, I.D., Spadoni, I., Mileti, E., Matteoli, G., Sonzogni, A., Sampietro, G.M.,
Immunol. 16, 45–56.
Foschi, D., Caprioli, F., Viale, G., and Rescigno, M. (2009). Human intestinal
epithelial cells promote the differentiation of tolerogenic dendritic cells. Gut Le Cras, T.D., Acciani, T.H., Mushaben, E.M., Kramer, E.L., Pastura, P.A., Har-
58, 1481–1489. die, W.D., Korfhagen, T.R., Sivaprasad, U., Ericksen, M., Gibson, A.M., et al.
(2011). Epithelial EGF receptor signaling mediates airway hyperreactivity and
Imai, Y., Yasuda, K., Sakaguchi, Y., Haneda, T., Mizutani, H., Yoshimoto, T., remodeling in a mouse model of chronic asthma. Am. J. Physiol. Lung Cell.
Nakanishi, K., and Yamanishi, K. (2013). Skin-specific expression of IL-33 ac- Mol. Physiol. 300, L414–L421.
tivates group 2 innate lymphoid cells and elicits atopic dermatitis-like inflam-
mation in mice. Proc. Natl. Acad. Sci. USA 110, 13921–13926. Lee, S.E., Jeong, S.K., and Lee, S.H. (2010). Protease and protease-activated
receptor-2 signaling in the pathogenesis of atopic dermatitis. Yonsei Med. J.
Jackson, D.J., and Johnston, S.L. (2010). The role of viruses in acute exacer- 51, 808–822.
bations of asthma. J. Allergy Clin. Immunol. 125, 1178–1187, quiz 1188–1189.
Li, M., Messaddeq, N., Teletin, M., Pasquali, J.L., Metzger, D., and Chambon,
Janssens, S., Pulendran, B., and Lambrecht, B.N. (2014). Emerging functions P. (2005). Retinoid X receptor ablation in adult mouse keratinocytes generates
of the unfolded protein response in immunity. Nat. Immunol. 15, 910–919. an atopic dermatitis triggered by thymic stromal lymphopoietin. Proc. Natl.
Acad. Sci. USA 102, 14795–14800.
Jin, S., Park, C.O., Shin, J.U., Noh, J.Y., Lee, Y.S., Lee, N.R., Kim, H.R., Noh,
S., Lee, Y., Lee, J.H., and Lee, K.H. (2014). DAMP molecules S100A9 and Liang, H.E., Reinhardt, R.L., Bando, J.K., Sullivan, B.M., Ho, I.C., and Locks-
S100A8 activated by IL-17A and house-dust mites are increased in atopic ley, R.M. (2012). Divergent expression patterns of IL-4 and IL-13 define unique
dermatitis. Exp. Dermatol. 23, 938–941. functions in allergic immunity. Nat. Immunol. 13, 58–66.

Juncadella, I.J., Kadl, A., Sharma, A.K., Shim, Y.M., Hochreiter-Hufford, A., Llop-Guevara, A., Chu, D.K., Walker, T.D., Goncharova, S., Fattouh, R., Silver,
Borish, L., and Ravichandran, K.S. (2013). Apoptotic cell clearance by bron- J.S., Moore, C.L., Xie, J.L., O’Byrne, P.M., Coyle, A.J., et al. (2014). A GM-CSF/
chial epithelial cells critically influences airway inflammation. Nature 493, IL-33 pathway facilitates allergic airway responses to sub-threshold house
547–551. dust mite exposure. PLoS ONE 9, e88714.

Maizels, R.M., Hewitson, J.P., and Smith, K.A. (2012). Susceptibility and im-
Kambayashi, T., and Laufer, T.M. (2014). Atypical MHC class II-expressing an-
munity to helminth parasites. Curr. Opin. Immunol. 24, 459–466.
tigen-presenting cells: can anything replace a dendritic cell? Nat. Rev. Immu-
nol. 14, 719–730. Martino, M.B., Jones, L., Brighton, B., Ehre, C., Abdulah, L., Davis, C.W., Ron,
D., O’Neal, W.K., and Ribeiro, C.M. (2013). The ER stress transducer IRE1b is
Karuppagounder, V., Arumugam, S., Thandavarayan, R.A., Pitchaimani, V., required for airway epithelial mucin production. Mucosal Immunol. 6, 639–654.
Sreedhar, R., Afrin, R., Harima, M., Suzuki, H., Nomoto, M., Miyashita, S.,
et al. (2014). Resveratrol attenuates HMGB1 signaling and inflammation in McAlees, J.W., Whitehead, G.S., Harley, I.T., Cappelletti, M., Rewerts, C.L.,
house dust mite-induced atopic dermatitis in mice. Int. Immunopharmacol. Holdcroft, A.M., Divanovic, S., Wills-Karp, M., Finkelman, F.D., Karp, C.L.,
23, 617–623. and Cook, D.N. (2015). Distinct Tlr4-expressing cell compartments control
neutrophilic and eosinophilic airway inflammation. Mucosal Immunol. 8,
Karuppagounder, V., Arumugam, S., Thandavarayan, R.A., Pitchaimani, V., 863–873.
Sreedhar, R., Afrin, R., Harima, M., Suzuki, H., Nomoto, M., Miyashita, S.,
et al. (2015). Modulation of HMGB1 translocation and RAGE/NFkB cascade Meylan, F., Hawley, E.T., Barron, L., Barlow, J.L., Penumetcha, P., Pelletier,
by quercetin treatment mitigates atopic dermatitis in NC/Nga transgenic M., Sciumè, G., Richard, A.C., Hayes, E.T., Gomez-Rodriguez, J., et al.
mice. Exp. Dermatol. 24, 418–423. (2014). The TNF-family cytokine TL1A promotes allergic immunopathology
through group 2 innate lymphoid cells. Mucosal Immunol. 7, 958–968.
Kelly, C., Shields, M.D., Elborn, J.S., and Schock, B.C. (2011). A20 regulation
of nuclear factor-kB: perspectives for inflammatory lung disease. Am. J. Re- Millien, V.O., Lu, W., Shaw, J., Yuan, X., Mak, G., Roberts, L., Song, L.Z.,
spir. Cell Mol. Biol. 44, 743–748. Knight, J.M., Creighton, C.J., Luong, A., et al. (2013). Cleavage of fibrinogen
by proteinases elicits allergic responses through Toll-like receptor 4. Science
Kool, M., Soullié, T., van Nimwegen, M., Willart, M.A., Muskens, F., Jung, S., 341, 792–796.
Hoogsteden, H.C., Hammad, H., and Lambrecht, B.N. (2008). Alum adjuvant
boosts adaptive immunity by inducing uric acid and activating inflammatory Mócsai, G., Gáspár, K., Nagy, G., Irinyi, B., Kapitány, A., Bı́ró, T., Gyimesi, E.,
dendritic cells. J. Exp. Med. 205, 869–882. Tóth, B., Maródi, L., and Szegedi, A. (2014). Severe skin inflammation and fi-
laggrin mutation similarly alter the skin barrier in patients with atopic dermatitis.
Kool, M., Willart, M.A., van Nimwegen, M., Bergen, I., Pouliot, P., Virchow, Br. J. Dermatol. 170, 617–624.
J.C., Rogers, N., Osorio, F., Reis e Sousa, C., Hammad, H., and Lambrecht,
Moniaga, C.S., Jeong, S.K., Egawa, G., Nakajima, S., Hara-Chikuma, M.,
B.N. (2011). An unexpected role for uric acid as an inducer of T helper 2 cell
Jeon, J.E., Lee, S.H., Hibino, T., Miyachi, Y., and Kabashima, K. (2013). Prote-
immunity to inhaled antigens and inflammatory mediator of allergic asthma.
ase activity enhances production of thymic stromal lymphopoietin and baso-
Immunity 34, 527–540.
phil accumulation in flaky tail mice. Am. J. Pathol. 182, 841–851.
Krimi, R.B., Kotelevets, L., Dubuquoy, L., Plaisancié, P., Walker, F., Lehy, T., Motomura, Y., Morita, H., Moro, K., Nakae, S., Artis, D., Endo, T.A., Kuroki, Y.,
Desreumaux, P., Van Seuningen, I., Chastre, E., Forgue-Lafitte, M.E., and Ohara, O., Koyasu, S., and Kubo, M. (2014). Basophil-derived interleukin-4
Marie, J.C. (2008). Resistin-like molecule beta regulates intestinal mucous controls the function of natural helper cells, a member of ILC2s, in lung inflam-
secretion and curtails TNBS-induced colitis in mice. Inflamm. Bowel Dis. 14, mation. Immunity 40, 758–771.
931–941.
Murthy, A., Shao, Y.W., Narala, S.R., Molyneux, S.D., Zúñiga-Pflücker, J.C.,
Kumamoto, Y., Linehan, M., Weinstein, J.S., Laidlaw, B.J., Craft, J.E., and Iwa- and Khokha, R. (2012). Notch activation by the metalloproteinase ADAM17
saki, A. (2013). CD301b+ dermal dendritic cells drive T helper 2 cell-mediated regulates myeloproliferation and atopic barrier immunity by suppressing
immunity. Immunity 39, 733–743. epithelial cytokine synthesis. Immunity 36, 105–119.

Kumar, R.K., Foster, P.S., and Rosenberg, H.F. (2014). Respiratory viral infec- Nathan, A.T., Peterson, E.A., Chakir, J., and Wills-Karp, M. (2009). Innate im-
tion, epithelial cytokines, and innate lymphoid cells in asthma exacerbations. mune responses of airway epithelium to house dust mite are mediated through
J. Leukoc. Biol. 96, 391–396. beta-glucan-dependent pathways. J. Allergy Clin. Immunol. 123, 612–618.

38 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.


Immunity

Review
Nawijn, M.C., Hackett, T.L., Postma, D.S., van Oosterhout, A.J., and Heijink, ated allergic immunopathology by enhancing differentiation and pathogenicity
I.H. (2011). E-cadherin: gatekeeper of airway mucosa and allergic sensitiza- of IL-9-producing T cells. J. Immunol. 194, 3567–3582.
tion. Trends Immunol. 32, 248–255.
Ring, J., Möhrenschlager, M., and Weidinger, S. (2012). Molecular genetics of
Neill, D.R., Wong, S.H., Bellosi, A., Flynn, R.J., Daly, M., Langford, T.K., Bucks, atopic eczema. Chem. Immunol. Allergy 96, 24–29.
C., Kane, C.M., Fallon, P.G., Pannell, R., et al. (2010). Nuocytes represent a
new innate effector leukocyte that mediates type-2 immunity. Nature 464, Saenz, S.A., Taylor, B.C., and Artis, D. (2008). Welcome to the neighborhood:
1367–1370. epithelial cell-derived cytokines license innate and adaptive immune re-
sponses at mucosal sites. Immunol. Rev. 226, 172–190.
Nikolaidis, N.M., Zimmermann, N., King, N.E., Mishra, A., Pope, S.M., Finkel-
man, F.D., and Rothenberg, M.E. (2003). Trefoil factor-2 is an allergen-induced Saenz, S.A., Siracusa, M.C., Perrigoue, J.G., Spencer, S.P., Urban, J.F., Jr.,
gene regulated by Th2 cytokines and STAT6 in the lung. Am. J. Respir. Cell Tocker, J.E., Budelsky, A.L., Kleinschek, M.A., Kastelein, R.A., Kambayashi,
Mol. Biol. 29, 458–464. T., et al. (2010). IL25 elicits a multipotent progenitor cell population that pro-
motes T(H)2 cytokine responses. Nature 464, 1362–1366.
Noti, M., Kim, B.S., Siracusa, M.C., Rak, G.D., Kubo, M., Moghaddam, A.E.,
Sattentau, Q.A., Comeau, M.R., Spergel, J.M., and Artis, D. (2014). Exposure Saenz, S.A., Siracusa, M.C., Monticelli, L.A., Ziegler, C.G., Kim, B.S., Brestoff,
to food allergens through inflamed skin promotes intestinal food allergy J.R., Peterson, L.W., Wherry, E.J., Goldrath, A.W., Bhandoola, A., and Artis, D.
through the thymic stromal lymphopoietin-basophil axis. J. Allergy Clin. Immu- (2013). IL-25 simultaneously elicits distinct populations of innate lymphoid
nol. 133, 1390–1399, 1399.e1–1399.e6. cells and multipotent progenitor type 2 (MPPtype2) cells. J. Exp. Med. 210,
1823–1837.
Nussbaum, J.C., Van Dyken, S.J., von Moltke, J., Cheng, L.E., Mohapatra, A.,
Molofsky, A.B., Thornton, E.E., Krummel, M.F., Chawla, A., Liang, H.E., and Salimi, M., Barlow, J.L., Saunders, S.P., Xue, L., Gutowska-Owsiak, D., Wang,
Locksley, R.M. (2013). Type 2 innate lymphoid cells control eosinophil homeo- X., Huang, L.C., Johnson, D., Scanlon, S.T., McKenzie, A.N., et al. (2013). A
stasis. Nature 502, 245–248. role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic derma-
titis. J. Exp. Med. 210, 2939–2950.
O’Grady, S.M., Patil, N., Melkamu, T., Maniak, P.J., Lancto, C., and Kita, H.
(2013). ATP release and Ca2+ signalling by human bronchial epithelial cells Savinko, T., Matikainen, S., Saarialho-Kere, U., Lehto, M., Wang, G., Lehti-
following Alternaria aeroallergen exposure. J. Physiol. 591, 4595–4609. mäki, S., Karisola, P., Reunala, T., Wolff, H., Lauerma, A., and Alenius, H.
(2012). IL-33 and ST2 in atopic dermatitis: expression profiles and modulation
Oboki, K., Ohno, T., Kajiwara, N., Arae, K., Morita, H., Ishii, A., Nambu, A., Abe, by triggering factors. J. Invest. Dermatol. 132, 1392–1400.
T., Kiyonari, H., Matsumoto, K., et al. (2010). IL-33 is a crucial amplifier of
innate rather than acquired immunity. Proc. Natl. Acad. Sci. USA 107, Schiering, C., Krausgruber, T., Chomka, A., Fröhlich, A., Adelmann, K., Wohl-
18581–18586. fert, E.A., Pott, J., Griseri, T., Bollrath, J., Hegazy, A.N., et al. (2014). The alar-
min IL-33 promotes regulatory T-cell function in the intestine. Nature 513,
Ohnmacht, C., Schwartz, C., Panzer, M., Schiedewitz, I., Naumann, R., and 564–568.
Voehringer, D. (2010). Basophils orchestrate chronic allergic dermatitis and
protective immunity against helminths. Immunity 33, 364–374. Schlievert, P.M., Strandberg, K.L., Lin, Y.C., Peterson, M.L., and Leung, D.Y.
(2010). Secreted virulence factor comparison between methicillin-resistant
Okumura, S., Sagara, H., Fukuda, T., Saito, H., and Okayama, Y. (2005). Fcep- and methicillin-sensitive Staphylococcus aureus, and its relevance to atopic
silonRI-mediated amphiregulin production by human mast cells increases dermatitis. J. Allergy Clin. Immunol. 125, 39–49.
mucin gene expression in epithelial cells. J. Allergy Clin. Immunol. 115,
272–279. Schmidt, M., Raghavan, B., Müller, V., Vogl, T., Fejer, G., Tchaptchet, S., Keck,
S., Kalis, C., Nielsen, P.J., Galanos, C., et al. (2010). Crucial role for human Toll-
Oliphant, C.J., Hwang, Y.Y., Walker, J.A., Salimi, M., Wong, S.H., Brewer, like receptor 4 in the development of contact allergy to nickel. Nat. Immunol.
J.M., Englezakis, A., Barlow, J.L., Hams, E., Scanlon, S.T., et al. (2014). 11, 814–819.
MHCII-mediated dialog between group 2 innate lymphoid cells and CD4(+)
T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Schmitz, J., Owyang, A., Oldham, E., Song, Y., Murphy, E., McClanahan, T.K.,
Immunity 41, 283–295. Zurawski, G., Moshrefi, M., Qin, J., Li, X., et al. (2005). IL-33, an interleukin-1-
like cytokine that signals via the IL-1 receptor-related protein ST2 and induces
Ovaere, P., Lippens, S., Vandenabeele, P., and Declercq, W. (2009). The T helper type 2-associated cytokines. Immunity 23, 479–490.
emerging roles of serine protease cascades in the epidermis. Trends Biochem.
Sci. 34, 453–463. Sekiya, T., Miyamasu, M., Imanishi, M., Yamada, H., Nakajima, T., Yamaguchi,
M., Fujisawa, T., Pawankar, R., Sano, Y., Ohta, K., et al. (2000). Inducible
Pace, E., Ferraro, M., Siena, L., Melis, M., Montalbano, A.M., Johnson, M., expression of a Th2-type CC chemokine thymus- and activation-regulated
Bonsignore, M.R., Bonsignore, G., and Gjomarkaj, M. (2008). Cigarette smoke chemokine by human bronchial epithelial cells. J. Immunol. 165, 2205–2213.
increases Toll-like receptor 4 and modifies lipopolysaccharide-mediated re-
sponses in airway epithelial cells. Immunology 124, 401–411. Shen, T., Kim, S., Do, J.S., Wang, L., Lantz, C., Urban, J.F., Le Gros, G., and
Min, B. (2008). T cell-derived IL-3 plays key role in parasite infection-induced
Page, K., Ledford, J.R., Zhou, P., Dienger, K., and Wills-Karp, M. (2010). basophil production but is dispensable for in vivo basophil survival. Int. Immu-
Mucosal sensitization to German cockroach involves protease-activated re- nol. 20, 1201–1209.
ceptor-2. Respir. Res. 11, 62.
Shimizu, M., Matsuda, A., Yanagisawa, K., Hirota, T., Akahoshi, M., Inomata,
Patel, D.A., You, Y., Huang, G., Byers, D.E., Kim, H.J., Agapov, E., Moore, N., Ebe, K., Tanaka, K., Sugiura, H., Nakashima, K., et al. (2005). Functional
M.L., Peebles, R.S., Jr., Castro, M., Sumino, K., et al. (2014). Interferon SNPs in the distal promoter of the ST2 gene are associated with atopic derma-
response and respiratory virus control are preserved in bronchial epithelial titis. Hum. Mol. Genet. 14, 2919–2927.
cells in asthma. J. Allergy Clin. Immunol. 134, 1402–1412.e7.
Simon, D., Radonjic-Hösli, S., Straumann, A., Yousefi, S., and Simon, H.U.
Plantinga, M., Guilliams, M., Vanheerswynghels, M., Deswarte, K., Branco- (2015). Active eosinophilic esophagitis is characterized by epithelial barrier de-
Madeira, F., Toussaint, W., Vanhoutte, L., Neyt, K., Killeen, N., Malissen, B., fects and eosinophil extracellular trap formation. Allergy 70, 443–452.
et al. (2013). Conventional and monocyte-derived CD11b(+) dendritic cells
initiate and maintain T helper 2 cell-mediated immunity to house dust mite Siracusa, M.C., Saenz, S.A., Hill, D.A., Kim, B.S., Headley, M.B., Doering, T.A.,
allergen. Immunity 38, 322–335. Wherry, E.J., Jessup, H.K., Siegel, L.A., Kambayashi, T., et al. (2011). TSLP
promotes interleukin-3-independent basophil haematopoiesis and type 2
Pulendran, B., and Artis, D. (2012). New paradigms in type 2 immunity. Science inflammation. Nature 477, 229–233.
337, 431–435.
Sleiman, P.M., Wang, M.L., Cianferoni, A., Aceves, S., Gonsalves, N., Nadeau,
Rajavelu, P., Chen, G., Xu, Y., Kitzmiller, J.A., Korfhagen, T.R., and Whitsett, K., Bredenoord, A.J., Furuta, G.T., Spergel, J.M., and Hakonarson, H. (2014).
J.A. (2015). Airway epithelial SPDEF integrates goblet cell differentiation and GWAS identifies four novel eosinophilic esophagitis loci. Nat. Commun. 5,
pulmonary Th2 inflammation. J. Clin. Invest. 125, 2021–2031. 5593.

Richard, A.C., Tan, C., Hawley, E.T., Gomez-Rodriguez, J., Goswami, R., Sokol, C.L., Barton, G.M., Farr, A.G., and Medzhitov, R. (2008). A mechanism
Yang, X.P., Cruz, A.C., Penumetcha, P., Hayes, E.T., Pelletier, M., et al. for the initiation of allergen-induced T helper type 2 responses. Nat. Immunol.
(2015). The TNF-family ligand TL1A and its receptor DR3 promote T cell-medi- 9, 310–318.

Immunity 43, July 21, 2015 ª2015 Elsevier Inc. 39


Immunity

Review
Spann, K.M., Baturcam, E., Schagen, J., Jones, C., Straub, C.P., Preston, Ullah, M.A., Loh, Z., Gan, W.J., Zhang, V., Yang, H., Li, J.H., Yamamoto, Y.,
F.M., Chen, L., Phipps, S., Sly, P.D., and Fantino, E. (2014). Viral and host fac- Schmidt, A.M., Armour, C.L., Hughes, J.M., et al. (2014). Receptor for
tors determine innate immune responses in airway epithelial cells from children advanced glycation end products and its ligand high-mobility group box-1
with wheeze and atopy. Thorax 69, 918–925. mediate allergic airway sensitization and airway inflammation. J. Allergy Clin.
Immunol. 134, 440–450.
, R.
Staples, K.J., Hinks, T.S., Ward, J.A., Gunn, V., Smith, C., and Djukanovic
(2012). Phenotypic characterization of lung macrophages in asthmatic pa- Voehringer, D. (2013). Protective and pathological roles of mast cells and ba-
tients: overexpression of CCL17. J. Allergy Clin. Immunol. 130, 1404–12.e7. sophils. Nat. Rev. Immunol. 13, 362–375.

Stremnitzer, C., Manzano-Szalai, K., Starkl, P., Willensdorfer, A., Schrom, S., Walley, A.J., Chavanas, S., Moffatt, M.F., Esnouf, R.M., Ubhi, B., Lawrence, R.,
Singer, J., Reichart, U., Akira, S., and Jensen-Jarolim, E. (2014). Epicutane- Wong, K., Abecasis, G.R., Jones, E.Y., Harper, J.I., et al. (2001). Gene poly-
ously applied Der p 2 induces a strong TH 2-biased antibody response in morphism in Netherton and common atopic disease. Nat. Genet. 29, 175–178.
C57BL/6 mice, independent of functional TLR4. Allergy 69, 741–751.
Wang, H., Su, Z., and Schwarze, J. (2009a). Healthy but not RSV-infected lung
epithelial cells profoundly inhibit T cell activation. Thorax 64, 283–290.
Stremnitzer, C., Manzano-Szalai, K., Willensdorfer, A., Starkl, P., Pieper, M.,
König, P., Mildner, M., Tschachler, E., Reichart, U., and Jensen-Jarolim, E. Wang, J., Ouyang, Y., Guner, Y., Ford, H.R., and Grishin, A.V. (2009b). Ubiq-
(2015). Papain degrades tight junction proteins of human keratinocytes uitin-editing enzyme A20 promotes tolerance to lipopolysaccharide in entero-
in vitro and sensitizes C57BL/6 mice via the skin independent of its enzymatic cytes. J. Immunol. 183, 1384–1392.
activity or TLR4 activation. J. Invest. Dermatol. 135, 1790–1800.
Willart, M.A., Deswarte, K., Pouliot, P., Braun, H., Beyaert, R., Lambrecht,
Swamy, M., Jamora, C., Havran, W., and Hayday, A. (2010). Epithelial decision B.N., and Hammad, H. (2012). Interleukin-1a controls allergic sensitization to
makers: in search of the ‘epimmunome’. Nat. Immunol. 11, 656–665. inhaled house dust mite via the epithelial release of GM-CSF and IL-33.
J. Exp. Med. 209, 1505–1517.
Takahashi, K., Hirose, K., Kawashima, S., Niwa, Y., Wakashin, H., Iwata, A.,
Tokoyoda, K., Renauld, J.C., Iwamoto, I., Nakayama, T., and Nakajima, H. Williams, J.W., Tjota, M.Y., Clay, B.S., Vander Lugt, B., Bandukwala, H.S.,
(2011). IL-22 attenuates IL-25 production by lung epithelial cells and inhibits Hrusch, C.L., Decker, D.C., Blaine, K.M., Fixsen, B.R., Singh, H., et al.
antigen-induced eosinophilic airway inflammation. J. Allergy Clin. Immunol. (2013). Transcription factor IRF4 drives dendritic cells to promote Th2 differen-
128, 1067–76.e1, 6. tiation. Nat. Commun. 4, 2990.

Takai, T., Chen, X., Xie, Y., Vu, A.T., Le, T.A., Kinoshita, H., Kawasaki, J., Ka- Xiao, C., Puddicombe, S.M., Field, S., Haywood, J., Broughton-Head, V., Pux-
mijo, S., Hara, M., Ushio, H., et al. (2014). TSLP expression induced via Toll-like eddu, I., Haitchi, H.M., Vernon-Wilson, E., Sammut, D., Bedke, N., et al. (2011).
receptor pathways in human keratinocytes. Methods Enzymol. 535, 371–387. Defective epithelial barrier function in asthma. J. Allergy Clin. Immunol. 128,
549–56.e1, 12.
Tamagawa-Mineoka, R., Okuzawa, Y., Masuda, K., and Katoh, N. (2014).
Increased serum levels of interleukin 33 in patients with atopic dermatitis. Xue, L., Salimi, M., Panse, I., Mjösberg, J.M., McKenzie, A.N., Spits, H., Kle-
J. Am. Acad. Dermatol. 70, 882–888. nerman, P., and Ogg, G. (2014). Prostaglandin D2 activates group 2 innate
lymphoid cells through chemoattractant receptor-homologous molecule ex-
Tordesillas, L., Goswami, R., Benedé, S., Grishina, G., Dunkin, D., Järvinen, pressed on TH2 cells. J. Allergy Clin. Immunol. 133, 1184–1194.
K.M., Maleki, S.J., Sampson, H.A., and Berin, M.C. (2014). Skin exposure pro-
motes a Th2-dependent sensitization to peanut allergens. J. Clin. Invest. 124, Yockey, L.J., Demehri, S., Turkoz, M., Turkoz, A., Ahern, P.P., Jassim, O.,
4965–4975. Manivasagam, S., Kearney, J.F., Gordon, J.I., and Kopan, R. (2013). The
absence of a microbiota enhances TSLP expression in mice with defective
Trompette, A., Divanovic, S., Visintin, A., Blanchard, C., Hegde, R.S., Madan, skin barrier but does not affect the severity of their allergic inflammation.
R., Thorne, P.S., Wills-Karp, M., Gioannini, T.L., Weiss, J.P., and Karp, C.L. J. Invest. Dermatol. 133, 2714–2721.
(2009). Allergenicity resulting from functional mimicry of a Toll-like receptor
Yu, X., Pappu, R., Ramirez-Carrozzi, V., Ota, N., Caplazi, P., Zhang, J., Yan, D.,
complex protein. Nature 457, 585–588.
Xu, M., Lee, W.P., and Grogan, J.L. (2014). TNF superfamily member TL1A
elicits type 2 innate lymphoid cells at mucosal barriers. Mucosal Immunol. 7,
Tully, J.E., Hoffman, S.M., Lahue, K.G., Nolin, J.D., Anathy, V., Lundblad, L.K.,
730–740.
Daphtary, N., Aliyeva, M., Black, K.E., Dixon, A.E., et al. (2013). Epithelial NF-
kB orchestrates house dust mite-induced airway inflammation, hyperrespon- Zaiss, D.M., Gause, W.C., Osborne, L.C., and Artis, D. (2015). Emerging func-
siveness, and fibrotic remodeling. J. Immunol. 191, 5811–5821. tions of amphiregulin in orchestrating immunity, inflammation, and tissue
repair. Immunity 42, 216–226.
Turner, J.E., Stockinger, B., and Helmby, H. (2013). IL-22 mediates goblet cell
hyperplasia and worm expulsion in intestinal helminth infection. PLoS Pathog. Zhou, B., Comeau, M.R., De Smedt, T., Liggitt, H.D., Dahl, M.E., Lewis, D.B.,
9, e1003698. Gyarmati, D., Aye, T., Campbell, D.J., and Ziegler, S.F. (2005). Thymic stromal
lymphopoietin as a key initiator of allergic airway inflammation in mice. Nat. Im-
Tussiwand, R., Everts, B., Grajales-Reyes, G.E., Kretzer, N.M., Iwata, A., Bag- munol. 6, 1047–1053.
aitkar, J., Wu, X., Wong, R., Anderson, D.A., Murphy, T.L., et al. (2015). Klf4
expression in conventional dendritic cells is required for T helper 2 cell re- Ziegler, S.F., and Artis, D. (2010). Sensing the outside world: TSLP regulates
sponses. Immunity 42, 916–928. barrier immunity. Nat. Immunol. 11, 289–293.

40 Immunity 43, July 21, 2015 ª2015 Elsevier Inc.

You might also like