You are on page 1of 9

Seminars in Immunology 46 (2019) 101333

Contents lists available at ScienceDirect

Seminars in Immunology
journal homepage: www.elsevier.com/locate/ysmim

Review

Immunologic mechanisms in asthma T


a,b a,c,d a,c
Tadech Boonpiyathad , Zeynep Celebi Sözener , Pattraporn Satitsuksanoa ,
Cezmi A. Akdisa,c,*
a
Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
b
Allergy and Clinical Immunology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
c
Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
d
Ankara University School of Medicine, Department of Chest Diseases Division of Clinical Immunology and Allergic Diseases, Ankara, Turkey

A R T I C LE I N FO A B S T R A C T

Keywords: Asthma is a chronic airway disease, which affects more than 300 million people. The pathogenesis of asthma
Asthma exhibits marked heterogeneity with many phenotypes defining visible characteristics and endotypes defining
Allergic molecular mechanisms. With the evolution of novel biological therapies, patients, who do not-respond to con-
Nonallergic ventional asthma therapy require novel biologic medications, such as anti-IgE, anti-IL-5 and anti-IL4/IL13 to
Eosinophilic
control asthma symptoms. It is increasingly important for physicians to understand immunopathology of asthma
Neutrophilic
Phenotype
and to characterize asthma phenotypes. Asthma is associated with immune system activation, airway hyperre-
Immunopathology sponsiveness (AHR), epithelial cell activation, mucus overproduction and airway remodeling. Both innate and
Mechanism adaptive immunity play roles in immunologic mechanisms of asthma. Type 2 asthma with eosinophilia is a
common phenotype in asthma. It occurs with and without visible allergy. The type 2 endotype comprises; T
helper type 2 (Th2) cells, type 2 innate lymphoid cells (ILC2), IgE-secreting B cells and eosinophils. Eosinophilic
nonallergic asthma is ILC2 predominated, which produces IL-5 to recruit eosinophil into the mucosal airway.
The second major subgroup of asthma is non-type 2 asthma, which contains heterogeneous group of endoypes
and phenotypes, such as exercise-induced asthma, obesity induced asthma, etc. Neutrophilic asthma is not in-
duced by allergens but can be induced by infections, cigarette smoke and pollution. IL-17 which is produced by
Th17 cells and type 3 ILCs, can stimulate neutrophilic airway inflammation. Macrophages, dendritic cells and
NKT cells are all capable of producing cytokines that are known to contribute in allergic and nonallergic asthma.
Bronchial epithelial cell activation and release of cytokines, such as IL-33, IL-25 and TSLP play a major role in
asthma. Especially, allergens or environmental exposure to toxic agents, such as pollutants, diesel exhaust,
detergents may affect the epithelial barrier leading to asthma development. In this review, we focus on the
immunologic mechanism of heterogenous asthma phenotypes.

1. Introduction airway lumen is one of the possible cause of the persistent airflow ob-
struction. Another mechanism of persistent airflow obstruction is
Asthma is a common chronic airway disease characterized by airway remodeling including pathologies such as goblet cell hyper-
variable airflow limitation secondary to airway narrowing, airway wall plasia, excessive subepithelial collagen deposition, decreased epithelial
thickening and increased mucus [1]. Airway narrowing results from and cartilage integrity, airway smooth muscle hyperplasia and in-
chronic airway inflammation secondary to plasma extravasation and creased vascularity [3].
influx of the inflammatory cells such as eosinophils, neutrophils, lym- Currently, asthma is considered as an umbrella diagnosis that con-
phocytes, macrophages and mast cells. Airway hyperresponsiveness sists several variable clinical presentations (phenotypes) and distinct
(AHR) is an important physiologic feature of asthma. AHR is an ex- pathophysiological mechanisms (endotypes) [4–6]. Allergic asthma is
aggerated response of the airways to a nonspecific stimuli, that would the most common asthma phenotype with an early onset, sensitization
produce little or no effect in healthy ones. Although asthma is often to an allergen, IgE related Th2-mediated background [7–9]. Type 2
defined as a reversible airway obstruction, it can evolve into irrever- immune response involves, Th2 cells, IgE-producing B cells, group 2
sible lung function impairment [2]. Increasing mucus production in the innate lymphoid cells (ILC2) and a small fraction of IL-4-producing NK


Corresponding author at: Swiss Institute of Allergy and Asthma Research (SIAF), Obere Str. 22 CH-7270, Davos Platz, Switzerland.
E-mail address: akdisac@siaf.uzh.ch (C.A. Akdis).

https://doi.org/10.1016/j.smim.2019.101333
Received 21 October 2019; Accepted 21 October 2019
Available online 06 November 2019
1044-5323/ © 2019 Elsevier Ltd. All rights reserved.
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

Fig. 1. Cellular compartments and pathways participates in immune mechanisms of allergic and nonallergic asthma. Airway epithelial cells contribute to type 2 and
non-type 2 inflammation, due to allergic or nonallergic (e.g. cigarette smoking and pollution) exposure. Epithelial cells secrete IL-25, IL-33 and thymic stromal
lymphopoietin (TSLP) together with dendritic cells, macrophages and NKT cells. These cytokines stimulate type 2 immune response via activated Th2 cells and ILC2.
IgE is involved in the early inflammatory process as a cause of allergic asthma. IL-5-secreting Th2 and ILC2 stimulates eosinophil-mediated inflammation. Th9
produces IL-9 which induce airway eosinophilic and allergic inflammation. Increasing numbers of CRTH2 expression on Th, Tc and Treg cells are found in the
immunopathology of allergic asthma. IL-1β and IL-23 secreted by epithelial cells induce neutrophilic inflammation stimulated by Th17 and ILC3.

cells and NK-T cells, basophils, eosinophils, mast cells and their major 2.1. Th2 cells
cytokines. There is a complex network between type 2 cytokines (IL-4,
IL-5, IL-9 and IL-13) which are mainly secreted from type 2 immune Th2 cells mediate these functions by producing various cytokines
cells and alarmins (IL-25, IL-33 and TSLP) which are released from such as IL-4, IL-5, and IL-13. They are considered as important mole-
tissue cells, particularly epithelial cells. Nonallergic or intrinsic asthma cules for management of allergic asthma. IL-4 is a cytokine that induces
includes a subset of patients with non-Th2 inflammation [2,10]. The differentiation of naïve T cells to Th2 cells [15]. IL-5 is responsible for
major mechanism leading to a non-type 2 response is thought to result the maturation and release of eosinophils in the bone marrow [16]. IL-
from an irregular innate immune response, including intrinsic neu- 13 induces proliferation of IgE-producing B cells and endothelial cells
trophil abnormalities and activation of the IL-17-mediated pathway. [17]. Recent developments revealed that the induction of allergen-re-
The immunologic mechanisms of asthma are shown in Fig. 1. The active Th2 cells requires a two-step procedure including initial exposure
molecular and cellular mechanisms of asthma have been well-char- or “sensitization” and reactivation with second exposure. T cell induc-
acterized, however, not fully understood. Therefore, this review aims to tion with an allergen is a complicated process that requires numerous
summarize the heterogenous asthma mechanisms and to highlight new interactions between several cell types in the lungs and lymph nodes.
findings of the research that have helped to better understand and Another Th2 cytokine, IL-31 and IL-31R concentrations are also in-
manage asthma over the last few years creased in the serum of allergic asthma patients together with stem cell
factor [18]. Indeed, IL-33 is demonstrated to induce the expression of
Th2 cytokine and activation of basophils and eosinophils [12]. Che-
2. Allergic asthma mokine receptors such as CCR4, CCR8, CXCR4 and CCR3 are expressed
on Th2 cells. CCR4 regulates chemotaxis of Th2 cells and its ligands
Allergic asthma is considered as the most common type of asthma, CCL17 and CCL22, are increased in the patients with allergic asthma.
which usually induced by sensitization to environmental allergens [11]. CCR8 can induce eosinophilia and AHR and may be elevate in Th2 cells
These environmental aeroallergens are house dust mites (HDM), grass, in the lungs and airways of allergic asthmatics [13]. CXCR4 which is
weed and tree pollens, fungal spores, animal dander, etc. After sensi- participated in Th2 cell migration into the lungs and treatment of al-
tization, symptoms of allergic asthma usually occur due to the sub- lergic mice with selective CXCR4 inhibitors, significantly reduces AHR
sequent exposures to the allergens. A recent study indicated that dif- and inflammatory responses [14].
ferent types of aeroallergens and specific sensitization profiles are
related to different clinical manifestations of allergic respiratory dis- 2.2. Type 2 innate lymphoid cells
eases (rhinitis with/without asthma), different clinical symptoms, and
different levels of severity [12,13]. Initiation of immune response, often Type 2 innate lymphoid cells (ILC2s) are derived from common
begins with the activation and differentiation of allergen-specific Th2 lymphoid progenitor and belong to the lymphoid lineage. These cells
cells, and triggered by allergens. Later on, immunoglobulin E (IgE) is produce type 2 cytokines such as IL-4, IL-5, IL-9, and IL-13. ILC2s and
produced against allergens. IgE dependent mast cells are activated and IL-33-ST2 receptor pathway play an important role in the development
eosinophils recruit into the lungs, and eventually persistent airway in- of allergic diseases and asthma [15]. ILC2s under the control of the
flammation and asthma symptoms occur [14]. transcription factors RORα and GATA3 [15]. In a mice model, lack of T

2
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

and B cells, activated ILC2s can induce eosinophilia and AHR [16]. A 2.5. Natural killer T cells
recent study indicated that induced Tregs (iTregs) suppress the pro-
duction of ILC2-driven, pro-inflammatory cytokines IL-5 and IL-13. The Natural killer T cells represents an important innate T cell subset.
suppression of Tregs is blocked by preventing direct cellular contact or They involve cell-cell interaction and down-stream modulation of DCs.
by inhibiting the ICOS-ICOS-ligand (ICOSL) pathway [15,17]. There- Invariant natural killer T (iNKT) cells, a sub group of NKT cells, are
fore, the regulation of ILC2 function is very important via the interac- suggested to modulate Th1/Th2 balance via the engagement of en-
tion of ICOS-ICOSL [15]. In other words, both paths are very important dogeneous or exogeneous ligands to Toll-like receptor 4 (TLR4) in iNKT
in the regulation of ILC2 function [18]. In addition, the decrease in cells. iNKT cells and mucosal-associated invariant T (MAIT) cells re-
cytokine production from ILC2s is controlled by IL-10 and TGF-β. spond rapidly to antigens bound to CD1d and MR1 molecules, respec-
Therefore, ILC2s play a pivotal role in the initiation, enhancement, and tively, and immediately exert effector functions by producing various
steroid resistance of allergic airway inflammation [18]. cytokines and granules [31]. Sixty percent of CD4+ T cells in the
bronchoalveolar lavage from severe asthmatic patients are iNKT cells
2.3. Th9 cells [41]. The number of iNKT cells were significantly increased in per-
ipheral blood and sputum of patients with severe asthma, too [42,43].
Th9 cells produce IL-9 which have the potential to induce eosino- Th2-like iNKT cells might be involved in development of asthma ex-
philic inflammation, mucus hypersecretion and hyperreactivity in the acerbation. MAIT cells exist in the lungs and contribute to produce Th2
airways [21]. Airway inflammation induced by Th9 cells, is strongly cytokines but several reports have indicated the different roles for these
stimulated by OX40 signaling in T cells [22]. OX40 activates the ubi- cells in asthma [44].
quitin ligase TRAF6, which triggered the induction of NF-kB-inducing Signaling via TLR4 in innate immune cells results in the generation
kinase (NIK) in CD4+ T cells and the non-canonical NF-kB pathway of type 1 cytokines and consequently the development of Th1/Th17 cell
which subsequently lead to Th9 differentiation [23]. Moreover, mTOR immunity [45]. Polymorphism of the components that involved in TLR4
complex 2 controls Th9 differentiation during allergic airway in- signaling might be correlated with asthma, allergy and AHR [46]. It was
flammation [24]. Patients with allergic asthma have higher Th9 cells demonstrated that the presence of TLR4 and MyD88 molecules are
and IL-9 concentrations in their circulation [25,26]. IL-9, impairs IFN-γ necessary for development of allergic responses. [47]. TLR4 has been
production and synergistically promotes IL-4-induced IgE secretion shown to be essential for the induction of airway inflammation by fungi
[25]. IL-9 plays a role in steroid-resistant asthma but anti-IL-9 mono- derived allergens and that, they bind to TLR4 on epithelial airway cells
clonal antibody has not been found beneficial in controlling asthma and macrophages supporting inflammatory signals and Th2 cell de-
symptoms and reducing asthma exacerbation [27]. velopment [48]. On the other hand, MyD88 is known to be critical for
induction of experimental asthma induced by Alternaria extract [49].
2.4. Dendritic cells MyD88 is shared by all TLRs except TLR3 as well as IL-33 which also
plays a role in inducing Th2 responses [50]. TLR4 ligands regulate iNKT
Airway dendritic cells (DCs) play a crucial role in providing capacity cell functions in pulmonary diseases.
to integrate signals from exposure to allergens and translate them di-
rectly into allergen-specific T cell responses [28]. Regulation of DC 2.6. Eosinophils
activation is a key mediated immune response to allergens. Activation
of DCs can be triggered by direct interaction with the foreign allergen; Eosinophils are effector cells of the innate immune system and they
however, it is recognized that functional activation of DCs is critically contain cytotoxic granules. By degranulation of eosinophils, numerous
influenced by interactions with other cells of innate immune system toxic proteins such as eosinophil-derived neurotoxin (EDN), eosinophil
[29]. The DC lineage consists of conventional DCs (cDC) and plasma- cationic protein (ECP), eosinophil peroxidase (EPO), major basic pro-
cytoid DCs (pDC). Lung DCs are heterogeneous cell population which tein (MBP), cytokine-mediated activation (IL-5) and lipid mediators,
comprises two different cDCs; type 1 (cDC1s) and type 2 cDCs (cDC2s). such as cysteinyl leukotrienes (cysLTs) are released [51]. Beside blood
Compared to other DC subsets, cDC1s have less capacity to take up eosinophilia, tissue eosinophilia is also considered as an important
allergens and are associated with tolerogenic function [30]. cDC1s can feature of allergic inflammation and asthma. Eosinophils tend to ac-
induce the differentiation of Tregs when expose to HDM through re- cumulate at sites of allergic inflammation and contribute to the devel-
tinoic acid (RA) and peroxisome proliferator-activated receptor-gamma opment of bronchial asthma [52]. They may have a role in airway re-
(PPARγ) induction [31]. modeling via producing transforming growth factor (TGF)-β and cysLTs
cDC2s are important for induction of Th2 and Th17 cell differ- and they induce AHR with MBP production [53]. CysLTs may involve in
entiation in HDM-induced asthma [32]. HDM can be recognized by the accumulation of eosinophils in the airways of asthmatics. Inhalation
innate receptors on the cell membrane of DCs, including C-type lectin of LTE4 stimulates the accumulation of eosinophils in asthmatic airways
receptors, such as Dectin-2 [33]. Dectin-1−/− mice did not develop [54]. LTD4 induces transendothelial migration of eosinophils and re-
eosinophilic inflammation and also did not show an induction of Th2 or lease of specific granule proteins primarily through β2 integrin and the
Th17 cytokines in an HDM-mediated asthma model [34]. In addition, cysLT1 receptor [55]. Development and maintenance of the eosino-
cDC2s express OX-40 ligand (OX-40 L) which is essential for Th2 cell philic inflammation in the airways is contribution of cysLTs, together
differentiation [35]. OX-40 L and CCL17 are higher on cDCs of patients with the Th2 network. Two independent studies showed that serum IL-
with asthma than healthy controls [36]. Allergen inhalation induces 5, EDN, and ECP were modulated following benralizumab treatment.
only DC2s to migrate to bronchial tissue and causes an increase of IL- The decrease seen in eosinophil count after benralizumab, results in a
25-receptor on both cDCs and pDCs [37]. In an allergic reaction, IgE- significant reduction also in EDN and ECP concentrations. This in-
bound antigens are rapidly introduced into antigen-specific CD4+ T dicates that cytotoxic granule proteins are not released after the re-
cells by DCs. Moreover, TSLP-stimulated cDCs of allergic asthmatic duction of eosinophils. [56]. The eosinophil recruitment is likely with
patients produce IL-9 that leads to polorization of Th9 [38]. ADAM10 is the adhesion of eosinophils to endothelial cells, through the α4 in-
a glycosylated type I membrane protein that expressed on DCs and tegrin/vascular cell adhesion molecule (VCAM)-1 [57]. VCAM-1 is
involved in the development of Th2 immune response [39]. Further- upregulated by IL-4 and IL-13 in endothelial cells. The interaction of
more, cDC1 has been shown to induce Th2 differentiation after ex- eosinophils with VCAM-1 induces eosinophil activation [57]. Eotaxin
posure to live-attenuated viral particles, this indicates that, in asth- and its receptor, CCR3, are expressed higher in the airways of asthmatic
matics, cDC1s shift towards a phenotype that promotes Th2 during viral patients than the controls [58]. Basophils may be particularly important
infections [40]. in eosinophilic asthma. Increasing sputum basophil number in the

3
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

patients with eosinophilic asthma was reported [59,60]. asthma. Barrier functions of the lung epithelium is mainly provided by
tight junctions (TJ). These heteromeric protein complexes form the
2.7. Mast cells sealing interface between adjacent epithelial cells [75].
Bronchial epithelial cells release significantly greater amounts of IL-
Mast cells are essential in the development of asthma and may have 8, GM-CSF, RANTES, and sICAM-1 in asthmatic patients than in non-
substantial effects on smooth muscles, mucous hyper-secretion and allergic controls [76]. When the epithelial barrier is damaged, epithelial
tissue remodeling, particularly in the airways, by releasing proteases cytokines such as TSLP, IL-25, and IL-33 are released [77]. IL-25 and IL-
such as tryptase and growth factors [61]. Mast cells express high levels 33 activate ILC2s directly to produce Th2-cytokines [78]. Viral infec-
of IL-33 receptor ST2 and have been shown to be activated by IL-33. IL- tions like rhinovirus, can also induce IL-33 and promotes type 2 in-
33 stimulates mast cells to produce Th2 cytokines, particularly IL-13 via flammatory pattern [79]. IL-4 and IL-13 cause barrier dysfunction in
5-/12-Lipoxygenase cascade [62]. Number of mast cells are increased in human airway epithelial cells [80]. IL-13-producing ILC2s significantly
allergic and non-allergic asthma but the accumulation of mast cells is impair the epithelial barrier of human bronchial epithelial cells [81].
more evidenced for the allergic asthmatics [63]. Moreover, mast cells Meanwhile, CPG-DNA administration enhances the tight junction in-
are more active in bronchial mucosa of the allergic patients than non- tegrity of the bronchial epithelial barrier [82]. Moreover, CD11c+ DCs
allergic ones [64]. stimulated by TSLP can activate CRTH2+ Th2 effector memory cells
They are the major cellular sources of PGD2. Mast cell numbers and and they undergo further Th2 polarization to amplify their roles in
PGD2 concentrations are increased in the airways of patients with se- allergic inflammation [83]. In addition, TSLP is a hematopoietic B cell
vere asthma [65]. D prostanoid (DP) and chemoattractant receptor- proliferation and differentiation factor [84]. TSLP also directly promote
homologous molecule (CRTH2), receptors for PGD2, are expressed on naive CD4+ and CD8+ T cells to develop into Th2 cells [85]. TSLP
Th2 cells. Recently, the role of CRTH2 in the pathogenesis of asthma further enhances GATA3 expression in human ILC2s and induces them
has been highlighted. It was demonstrated that CRTH2 expression on T to produce IL-4 and other Th2 cytokines [86]. TSLP can significantly
cells (Th2, Tc and Treg), ILC2s and eosinophils is higher in eosinophilic enhances eosinophilic inflammation, promotes activity and chemotaxis
allergic asthma than nonallergic asthma and healthy controls [66]. of eosinophils by delaying the eosinophil apoptosis, upregulating the
Moreover, CRTH2+ Treg cells have been shown to have defective expression of adhesion molecule CD18, intercellular adhesion mole-
suppressor functions and decreased numbers of CRTH2+ Treg cells in cule-1 and down-regulating the L-selectin [87,88]. Periostin is an ex-
patients with allergic asthma are found to be correlated with improved tracellular matrix protein which secreted by activated airway epithelial
asthma control [66]. cells. It recognizes as a biomarker of type 2 inflammation [89]. Peri-
ostin gene expression is up-regulated in bronchial epithelial cells by IL-
2.8. Immunoglobulin E 13 and IL-4 [90]. Periostin acts on fibroblasts to promote airway re-
modeling, enhance mucus secretion and recruit eosinophils [91].
IgE antibodies are responsible for the “early phase” of an allergic
reaction and considered to have a minor role in the “late phase” reac- 3. Nonallergic asthma
tion [67]. The biological role of IgE is complex and related to its ability
to influence the functions of several immune and structural cells that 3.1. Neutrophilic asthma
involved the pathogenesis of chronic allergic inflammation via specific
receptors, such as high-affinity (FcεRI) and low-affinity CD23 (or Many of the immunopathologic features of nonallergic asthma are
FcεRII) receptors [67]. FcεRI is expressed by mast cells, basophils, DCs, similar to those observed in allergic asthma [92]. Patients with non-
airway smooth muscle cells, epithelial cells, endothelial cells, and eo- allergic asthma have a high expression of RANTES and GM-CSF re-
sinophils. IgE facilitates antigen presentation by DCs. The allergen ceptor alpha in mucosa and bronchoalveolar lavage [93]. In the sputum
captured by DCs, binds to FCεRI receptors and is then presented to of non-eosinophilic asthmatics, three signature genes related to innate
memory Th2 cells [68]. Cross linked IgE on DCs, leads to 1000-fold immunity: IL-1β, alkaline phosphatase tissue-nonspecific isozyme
increase in activation of T cells and production of CCL28 (chemokine (ALPL) and CXCR2 were identified. [94]. While type 2 immune re-
attracts Th2 lymphocytes) [69]. Moreover, FcεRI activation blocks, or sponse cells mainly involve in the development of eosinophilic allergic
at least reduces, intracellular signals involved in the production of type asthma, other Th cell subsets like Th1 and Th17 cells which produced
I IFN that are degraded by the anti-viral response [70]. IL-17, IL-21 and IL-22 are dominant in neutrophilic asthma. IL-17 is
CD23 expression on B cells is a major part of the adaptive immune responsible for the recruitment of neutrophils into the lungs. Activation
response to inhaled HDM allergen to induce allergen-specific Th2 re- of the neutrophils can be either directly through CXCL8 production or
sponse [71]. IgE has a direct effect on eosinophil functions such as indirectly by production of IL-6, G-CSF, GM-CSF, IL-8, CXCL1 and
activation, release of eosinophil peroxidase, increase expression of in- CXCL5 from airway epithelial cells [4]. IL-17 is often associated with
tegrin and release of TNF-α [67]. IgE directly activates airway smooth neutrophilic inflammation and AHR. IL-17A and IL-17 F are upregu-
muscles to produce IL-4, IL-5, IL-13, TNF-α, TSLP and chemokines lated in the lungs of asthmatic patients and this is correlated with se-
(CCL5, CCL11, CXCL8, CXCL10) These causes contraction and pro- verity of asthma and steroid-resistant [95]. Moreover, IL-17–related
liferation of the airway smooth muscles, leading to airway remodeling cytokine levels were increased in bronchial/nasal mucosa of patients
[72]. CD23 which also expressed on airway epithelial cells involved in with exacerbation prone neutrophilic asthma [96]. Also, IL-17A and
the transport of IgE-allergen complexes across the polarized airway IFN-γ have significantly increased in patients with steroid-resistant
mucosal barrier [73]. IgE+ memory B cells and plasmablasts were asthma [97]. DCs play an important role to bridge innate and adaptive
elevated in allergic patients and correlated with Th2 cell numbers [74]. immunity, and they trigger Th17 cell differentiation by providing an-
tigenic, costimulatory and cytokine signals. IL-6, IL-23 and TGF-β are
2.9. Epithelial cells required for Th17 differentiation and are responsible for upregulation
of the transcription factor ROR??t as well as IL-23 receptor in T cells
Airway epithelium forms the first line of defense against airborne [98]. IL-23 is crucial for maintaining the Th17 cell functionally in ac-
pathogens and prevents organism from infectious particles. Airway tive state [99]. Another mechanism in neutrophilic airway inflamma-
epithelial damage following exposure to environmental factors causes tion is based on inflammasome activation. The nucleotide-binding oli-
inflammation. Subsequent to the histological changes in the airway gomerization domain-like receptor family, pyrin domain containing 3
mucosal epithelium, functional abnormalities occur. Such changes are (NLRP3) inflammasome, an intracellular multiprotein complex, facil-
believed to have a significant association with the pathophysiology of itates the autoactivation of proinflammatory cysteine protease caspase-

4
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

1 [100]. Then the activated caspase-1 cleaves pro-IL-1β and pro-IL-18 proinflammatory mediators, including eicosanoids, cytokines, and ad-
into their mature forms. Active IL-1β was found to promote Th17 cell- hesion molecules. Exposure to NO2 and O3 results in a significant re-
dependent inflammation [101]. NLRP3 is activated by serum amyloid A lease of LTC4 and a variety of cytokines, including IL-8, TNF-α, ICAM-1
(SAA) protein, which is produced upon exposure of airway epithelial and GM-CSF from the cultures [119,120]. In non-allergic inflammatory
cells to microbes and can be detected at high levels in both serum and response, bronchial airway epithelial cells can produce IL-1β, TGF-β
induced sputum of asthmatic patients [102]. NLRP3 activation can be and IL-6, which recruit neutrophils via stimulation of IL-17 release from
triggered by danger-associated molecular patterns (DAMP) that gener- Th17 cells. Cigarette smoke and ambient pollution have an adverse
ated in epithelial damage caused by oxidative stress such as air pollu- effect on airway epithelial cells. A recent study showed that, epithelial
tion and cigarette smoke. Gene expression of NLRP3, IL-1β and caspase- cell genes which are mostly related to cell growth and transcription
1 is detected at high levels in sputum, and IL-1β levels correlate with regulation, are significantly irregular in associated with smoking and
sputum IL-8 levels in patients with neutrophilic asthma [103]. In neu- PM2.5, [121]. Moreover, cigarette smoke induces MMP-1 release from
trophilic asthma, the ability of alveolar macrophages to phagocytize bronchial epithelial cells [122].
apoptotic cells is significantly impaired compared to eosinophilic TJ, mucin, and inflammasome-related molecules are expressed dif-
asthma [104]. ferently in distinct inflammatory phenotypes of asthma, Zo-1 and
Th1 cells and Th1 related cytokines such as IFN-γ and TNF-α, are Cldn18 were found to be downregulated in all phenotypes, while in-
increased in patients with severe neutrophilic asthma [105]. High levels creased Cldn4 expression was characteristic for neutrophilic airway
of IFN-γ and low levels of secretory leukocyte protease inhibitor (SLPI) inflammation [123]. Mucins Clca1 (Gob5) and Muc5ac were also up-
are found in patients with severe asthma [106]. High IFN-γ levels in the regulated in neutrophilic asthma phenotype. Moreover,increased ex-
airways, promote AHR through the suppression of SLPI. In mouse ex- pression of inflammasome-related molecules such as Nlrp3, Nlrc4,
periments it was shown that, high IFN-γ levels in the airways induces Casp-1, and IL-1beta for neutrophilic asthma have been identified
neutrophilic lung inflammation, emphysema and AHR [107]. TNF-α is [123].
mainly produced by macrophages and mast cells and it promotes neu-
trophil chemotaxis [108]. Administration of inhaled recombinant TNF- 3.2. Eosinophilic nonallergic asthma
α, to healthy controls results in AHR and airway neutrophilia [109].
AHR may result from the direct effect of TNF-α on the airway smooth Some patients with eosinophilic asthma are not allergic [124]. In-
muscle or indirectly the release of cysteinyl leukotrienes C4 and D4 deed, late-onset eosinophilic asthma which is frequently associated
[110]. The efficacy of anti-TNF-α treatment in moderate to severe with chronic rhinosinusitis and nasal polyps is often severe and non-
asthma is questionable. allergic [125]. In this phenotype, despite high doses of inhaled, and
Type-3 innate lymphoid cells (ILC3) and IL-17 secreting ILCs are systemic corticosteroids, lung and blood eosinophilia persists. ILC2s
potentially important in neutrophilic asthma. ILC3s, Th17 cells, M1 play a vital role in eosinophilic non-allergic asthma. The number of
macrophages and neutrophils are associated with steroid-resistant and ILC2s increased in the lungs and peripheral blood of the asthmatic
obesity-related asthma [111]. In the lungs of obese mice, increased IL- patients who have chronic rhinosinusitis with nasal polyps and have an
17A producing CCR6+ ILC3s were found to be associated with AHR and increased eosinophil count [126]. ILC2s can produce high amounts of
neutrophilic inflammation. [112]. These study also demonstrated that, IL-5, that might be explain this severe eosinophilic inflammation in the
IL-1β produced by M1 macrophages increased in both lungs and adi- absence of classical Th2-mediated allergic response in late-onset eosi-
pose tissue of obese mice, and stimulates ILC3s to produce IL-17A nophilic asthma. Lipoxin A4, a natural pro-resolving ligand for ALX/
[112]. IL-17+ ILC3 and ILC2 levels were found to be increased in mice FPR2 receptors, significantly increases NK cell-mediated eosinophil
on high-fat diet following HDM challenge and decreased using anti- apoptosis and decreases IL-13-secreting ILC2s [127]. Through, lipoxin
CD90 antibody [113]. M1-mediated inflammation in adipose tissue A4 decreases eosinophilic inflammation. In severe asthma, lipoxin A4
enhances M2-mediated inflammation in the asthmatic lung [114]. Gene concentrations have been shown to decrease and this may clarify eo-
signatures of ILC3 are enriched in total RNA of patients with adult-onset sinophilia and chronic airway inflammation that characterize the dis-
asthma [115]. ease. In an asthma genome study, ILC2 related genes, ROR-α and IL-33
IL-6 is secreted by non-leukocytes such as endothelial cells, fibro- receptor complex (ST2), were found to be associated with development
blasts, astrocytes, epithelial cells. IL-6 levels are also affected by viral of asthma [128]. In mouse asthma model induced by Alternaria, ILC2s
infections and obesity and increase in intrinsic asthma compared to together with IL-33, IL-5 and IL-13 are sufficient to drive eosinophilia
allergic asthma [104,105]. IL-6 levels in sputum are inversely corre- and AHR [129]. Moreover, ILC2s together with NKT cells, IL13 and IL-
lated with the predictive percentage of FEV1 [106]. In addition, in- 33 can induce AHR in mice exposed to glycolipid antigens [130]. TSLP
creased levels of IL-6 in serum are correlated with impaired lung confers, partial ILC2 resistance to corticosteroid through STAT5 sig-
function in obese asthmatic patients [107]. naling [131]. A recent study has shown that, glucocorticoid therapy
Environmental stimuli such as diesel exhaust particles and cigarette may reduce ILC2 activation in patients with asthma via MEK/JAK-STAT
smoke can trigger Th17-mediated airway inflammation in asthmatic signaling pathways [132].
patients [116]. Along with asthma inflammation, cigarette smoke can GM-CSF plays an important role in eosinophil activation after mi-
induce a change in the phenotype of asthma through the predominance gration process, even without IL-5. GM-CSF induces eosinophil super-
of activated macrophages and neutrophils in sputum, airways and lung oxide anion generation and the release of specific granule proteins in
parenchyma. In addition, cigarette smoke activates macrophages and vitro, when incubated with VCAM-1 or intercellular cell adhesion mo-
they produce oxygen species (ROS), matrix metalloproteinase (MMP) lecule (ICAM)-1 [133]. Even in the absence of IL-5, Th2 network,
and cytokines that prolonged survival of neutrophils in the lung tissue containing a cascade of VCAM-1/CC chemokines/GM-CSF, is likely the
(e.g. IL-8) [117]. Activated macrophages are also stimulate Th1 and primary pathway for maintaining eosinophilic infiltration and activa-
Th17, that can promote bronchial epithelial cells and goblet cell acti- tion in asthma [134].
vation to enhance airway inflammation, mucus production and airway
remodeling [117]. Exposure to fungal cell-wall beta-glucans, increases 4. Conclusion
the levels of IL-17A and IL-13 and contributes to severe steroid-resistant
asthma [118]. In this review, authors have described the immunologic mechanisms
Various studies have demonstrated that, exposure of bronchial in asthma (Table 1). Asthma is a heterogeneous disease, comprising
epithelial cells to nitrogen dioxide (NO2), ozone (O3), and diesel ex- distinct phenotypic spectrums of patient populations. The current
haust particles (DEPs) results in significant synthesis and release of availability of biotherapeutic agents has provided individualized

5
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

Table 1
Asthma phenotypes and cellular mechanism.
Asthma phenotype Clinical Inflammatory response

Allergic Early-onset, children, often associated with allergic rhinitis, positive skin prick test to Epithelial cells secreted IL-25, IL-33 and TSLP
aeroallergens or presence of allergen-specific Ig E, Eosiniphilic or noneosinophilic, DC2 express IL-4, OX-40 L, CCL17 and PGE2
eosinophilic inflammation in sputum ( > 3% of the total cell count), allergen induced TLR4
exacerbate NKT cells secreted type 2 cytokines
Th2 secreted IL-4, IL-5, IL-13 and IL-31
ILC2 secreted type 2 cytokines
Th9 secreted IL-9
IgE class-switched B cells
Mast cells secreted proteas and PGD2
Eosinophils secreted IL-4, IL-5, IL-13, granule proteins
(EDN, ECP, EPO, MBP) and cysteinyl leukotriens
Neutrophilic nonallergic Late-onset asthma, adult, corticosteroid resistant, absent of eosinophilia, paucigranulocytic/ Epithelial cells secreted IL-β and IL-23
neutrophilic, cigarette smoke, pollution and viral induced exacerbate DCs secreted IL-6, IL-23 and TGF-β
Th17 secreted IL-17
Th1secreted IFN-γ and TNF-α
ILC3s secreted IL-17
Monocyte and NKT cells secreted IL-8
Eosinophilic nonallergic Late-onset eosinophilic asthma, associated with chronic rhinosinusitis and nasal polyps, Epithelial cells secreted IL-25, IL-33 and TSLP
eosinophilia and eosinophilic inflammation in sputum ( > 3% of the total cell count) ILC2 secreted IL-5 and Lipoxin A4
NKT cells
GM-CSF

treatment for patients who do not respond to conventional asthma [7] J. Genuneit, A.M. Seibold, C.J. Apfelbacher, G.N. Konstantinou, J.J. Koplin, S. La
therapy. Novel developing medications, such as, anti-CRTH2 receptor, Grutta, K. Logan, M.R. Perkin, C. Flohr, E.I.G.o.E, Task Force’ overview of sys-
tematic reviews in allergy epidemiology’ of the, overview of systematic reviews in
anti-endothelin receptor, anti-TSLP, anti-IL-8, anti-IL-17 and the cur- allergy epidemiology, Allergy 72 (6) (2017) 849–856.
rent ones which are targeting the downstream allergy mediators, such [8] M. Odling, N. Andersson, S. Ekstrom, E. Melen, A. Bergstrom, I. Kull,
as IgE, IL-5 and IL-4/IL-13 have been studied for administration in Characterization of asthma in the adolescent population, Allergy 73 (8) (2018)
1744–1746.
different asthma phenotypes. To get the greatest benefit from these [9] M.W. Su, W.C. Lin, C.H. Tsai, B.L. Chiang, Y.H. Yang, Y.T. Lin, L.C. Wang, J.H. Lee,
agents, all asthmatic patients should undergo an aeroallergen skin prick C.C. Chou, Y.F. Wu, Y.L. Yeh, Y.L. Lee, Childhood asthma clusters reveal neu-
test or allergen-specific IgE measurement to assess allergic status, and trophil-predominant phenotype with distinct gene expression, Allergy 73 (10)
(2018) 2024–2032.
complete blood counts to assess eosinophil levels. These biomarkers are [10] L. Worth, S. Michel, V.D. Gaertner, M. Kabesch, M. Schieck, Asthma- and IgE-
both non-invasive and useful to provide accurate clinical information associated polymorphisms affect expression of TH 17 genes, Allergy 73 (6) (2018)
about the underlying asthma phenotype. The knowledge of molecular 1342–1347.
[11] S.T. Holgate, S. Wenzel, D.S. Postma, S.T. Weiss, H. Renz, P.D. Sly, Asthma, Nat.
process involved in disease pathogenesis will be further investigated to
Rev. Dis. Primers 1 (2015) 15025.
bring novel therapeutic options for asthmatic patient. [12] A. Valero, S. Quirce, I. Davila, J. Delgado, J. Dominguez-Ortega, Allergic re-
spiratory disease: different allergens, different symptoms, Allergy 72 (9) (2017)
Acknowledgments 1306–1316.
[13] J. Bousquet, S. Arnavielhe, A. Bedbrook, J. Fonseca, M. Morais Almeida, A. Todo
Bom, et al., The Allergic Rhinitis and its Impact on Asthma (ARIA) score of allergic
The authors' laboratories are supported by the Swiss National rhinitis using mobile technology correlates with quality of life: the MASK study,
Foundation grant 320030_140772 and Christine Kühne - Center for Allergy 73 (2) (2018) 505–510.
[14] S.R. Del Giacco, A. Bakirtas, E. Bel, A. Custovic, Z. Diamant, E. Hamelmann,
Allergy Research and Education (CK-CARE), European 7th frame work E. Heffler, O. Kalayci, S. Saglani, S. Sergejeva, S. Seys, A. Simpson, L. Bjermer,
projects MeDALL: Mechanisms of the Development of Allergy (No: Allergy in severe asthma, Allergy 72 (2) (2017) 207–220.
261357) and PREDICTA: Post-Infectious Immune Reprogramming and [15] D. Russkamp, A. Aguilar-Pimentel, F. Alessandrini, V. Gailus-Durner, H. Fuchs,
C. Ohnmacht, A. Chaker, M.H. de Angelis, M. Ollert, C.B. Schmidt-Weber, S. Blank,
Its Association with Persistence and Chronicity of Respiratory Allergic IL-4 receptor alpha blockade prevents sensitization and alters acute and long-
Diseases (No: 260895). lasting effects of allergen-specific immunotherapy of murine allergic asthma,
Allergy (2019).
[16] M. Hassani, L. Koenderman, Immunological and hematological effects of IL-
References 5(Ralpha)-targeted therapy: an overview, Allergy 73 (10) (2018) 1979–1988.
[17] Y. Mitamura, S. Nunomura, Y. Nanri, M. Ogawa, T. Yoshihara, M. Masuoka,
[1] M. Schatz, L. Rosenwasser, The allergic asthma phenotype, J. Allergy Clin. G. Tsuji, T. Nakahara, A. Hashimoto-Hachiya, S.J. Conway, M. Furue, K. Izuhara,
Immunol. Pract. 2 (6) (2014) 645–648 quiz 649. The IL-13/periostin/IL-24 pathway causes epidermal barrier dysfunction in al-
[2] R.M. Dunn, P.J. Busse, M.E. Wechsler, Asthma in the elderly and late-onset adult lergic skin inflammation, Allergy 73 (9) (2018) 1881–1891.
asthma, Allergy 73 (2) (2018) 284–294. [18] Z. Lei, G. Liu, Q. Huang, M. Lv, R. Zu, G.M. Zhang, Z.H. Feng, B. Huang, SCF and
[3] Y.C. Wang, M.S. Jaakkola, T.K. Lajunen, C.H. Lai, J.J.K. Jaakkola, Asthma-COPD IL-31 rather than IL-17 and BAFF are potential indicators in patients with allergic
Overlap Syndrome among subjects with newly diagnosed adult-onset asthma, asthma, Allergy 63 (3) (2008) 327–332.
Allergy 73 (7) (2018) 1554–1557. [21] T. Ohtomo, O. Kaminuma, J. Yamada, N. Kitamura, A. Abe, N. Kobayashi,
[4] I. Agache, C.A. Akdis, Endotypes of allergic diseases and asthma: an important step M. Suko, A. Mori, Eosinophils are required for the induction of bronchial hy-
in building blocks for the future of precision medicine, Allergol. Int. 65 (3) (2016) perresponsiveness in a Th transfer model of BALB/c background, Int. Arch. Allergy
243–252. Immunol. 152 (Suppl. 1) (2010) 79–82.
[5] H.T. Tan, S. Hagner, F. Ruchti, U. Radzikowska, G. Tan, C. Altunbulakli, [22] X. Xiao, Y. Fan, J. Li, X. Zhang, X. Lou, Y. Dou, X. Shi, P. Lan, Y. Xiao, L. Minze,
A. Eljaszewicz, M. Moniuszko, M. Akdis, C.A. Akdis, H. Garn, M. Sokolowska, X.C. Li, Guidance of super-enhancers in regulation of IL-9 induction and airway
Tight junction, mucin, and inflammasome-related molecules are differentially inflammation, J. Exp. Med. 215 (2) (2018) 559–574.
expressed in eosinophilic, mixed, and neutrophilic experimental asthma in mice, [23] X. Xiao, S. Balasubramanian, W. Liu, X. Chu, H. Wang, E.J. Taparowsky, Y.X. Fu,
Allergy (2018). Y. Choi, M.C. Walsh, X.C. Li, OX40 signaling favors the induction of T(H)9 cells
[6] A. Muraro, R.F. Lemanske Jr., M. Castells, M.J. Torres, D. Khan, H.U. Simon, and airway inflammation, Nat. Immunol. 13 (10) (2012) 981–990.
C. Bindslev-Jensen, W. Burks, L.K. Poulsen, H.A. Sampson, M. Worm, K.C. Nadeau, [24] H. Chen, L. Zhang, P. Wang, H. Su, W. Wang, Z. Chu, L. Zhang, X. Zhang, Y. Zhao,
Precision medicine in allergic disease-food allergy, drug allergy, and anaphylaxis- mTORC2 controls Th9 polarization and allergic airway inflammation, Allergy 72
PRACTALL document of the European Academy of Allergy and Clinical (10) (2017) 1510–1520.
Immunology and the American Academy of Allergy, Asthma and Immunology, [25] L. Jia, Y. Wang, J. Li, S. Li, Y. Zhang, J. Shen, W. Tan, C. Wu, Detection of IL-9
Allergy 72 (7) (2017) 1006–1021. producing T cells in the PBMCs of allergic asthmatic patients, BMC Immunol. 18
(1) (2017) 38.

6
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

[26] D. Hoppenot, K. Malakauskas, S. Lavinskiene, I. Bajoriuniene, V. Kalinauskaite, E. Seberova, P. Panzner, V. Zindr, Asthma management: a new phenotype-based
R. Sakalauskas, Peripheral blood Th9 cells and eosinophil apoptosis in asthma approach using presence of eosinophilia and allergy, Allergy 72 (9) (2017)
patients, Medicina Kaunas (Kaunas) 51 (1) (2015) 10–17. 1279–1287.
[27] M. Saeki, O. Kaminuma, T. Nishimura, N. Kitamura, A. Mori, T. Hiroi, Th9 cells [53] D.T. Wong, A. Elovic, K. Matossian, N. Nagura, J. McBride, M.Y. Chou,
induce steroid-resistant bronchial hyperresponsiveness in mice, Allergol. Int. 66S J.R. Gordon, T.H. Rand, S.J. Galli, P.F. Weller, Eosinophils from patients with
(2017) S35–S40. blood eosinophilia express transforming growth factor beta 1, Blood 78 (10)
[28] M.A. Gill, The role of dendritic cells in asthma, J. Allergy Clin. Immunol. 129 (4) (1991) 2702–2707.
(2012) 889–901. [54] L.A. Laitinen, A. Laitinen, T. Haahtela, V. Vilkka, B.W. Spur, T.H. Lee, Leukotriene
[29] H. Vroman, R.W. Hendriks, M. Kool, Dendritic cell subsets in asthma: impaired E4 and granulocytic infiltration into asthmatic airways, Lancet 341 (8851) (1993)
tolerance or exaggerated inflammation? Front. Immunol. 8 (2017) 941. 989–990.
[30] M. Plantinga, M. Guilliams, M. Vanheerswynghels, K. Deswarte, F. Branco- [55] K. Saito, M. Nagata, I. Kikuchi, Y. Sakamoto, Leukotriene D4 and eosinophil
Madeira, W. Toussaint, L. Vanhoutte, K. Neyt, N. Killeen, B. Malissen, H. Hammad, transendothelial migration, superoxide generation, and degranulation via beta2
B.N. Lambrecht, Conventional and monocyte-derived CD11b(+) dendritic cells integrin, Ann. Allergy Asthma Immunol. 93 (6) (2004) 594–600.
initiate and maintain T helper 2 cell-mediated immunity to house dust mite al- [56] T.H. Pham, G. Damera, P. Newbold, K. Ranade, Reductions in eosinophil bio-
lergen, Immunity 38 (2) (2013) 322–335. markers by benralizumab in patients with asthma, Respir. Med. 111 (2016) 21–29.
[31] A. Khare, N. Krishnamoorthy, T.B. Oriss, M. Fei, P. Ray, A. Ray, Cutting edge: [57] M. Nagata, J.B. Sedgwick, R. Vrtis, W.W. Busse, Endothelial cells upregulate eo-
inhaled antigen upregulates retinaldehyde dehydrogenase in lung CD103+ but sinophil superoxide generation via VCAM-1 expression, Clin. Exp. Allergy 29 (4)
not plasmacytoid dendritic cells to induce Foxp3 de novo in CD4+ T cells and (1999) 550–561.
promote airway tolerance, J. Immunol. 191 (1) (2013) 25–29. [58] S. Ying, D.S. Robinson, Q. Meng, J. Rottman, R. Kennedy, D.J. Ringler,
[32] A. Norimoto, K. Hirose, A. Iwata, T. Tamachi, M. Yokota, K. Takahashi, S. Saijo, C.R. Mackay, B.L. Daugherty, M.S. Springer, S.R. Durham, T.J. Williams, A.B. Kay,
Y. Iwakura, H. Nakajima, Dectin-2 promotes house dust mite-induced T helper Enhanced expression of eotaxin and CCR3 mRNA and protein in atopic asthma.
type 2 and type 17 cell differentiation and allergic airway inflammation in mice, Association with airway hyperresponsiveness and predominant co-localization of
Am. J. Respir. Cell Mol. Biol. 51 (2) (2014) 201–209. eotaxin mRNA to bronchial epithelial and endothelial cells, Eur. J. Immunol. 27
[33] N.A. Barrett, A. Maekawa, O.M. Rahman, K.F. Austen, Y. Kanaoka, Dectin-2 re- (12) (1997) 3507–3516.
cognition of house dust mite triggers cysteinyl leukotriene generation by dendritic [59] Y. Suzuki, K. Wakahara, T. Nishio, S. Ito, Y. Hasegawa, Airway basophils are in-
cells, J. Immunol. 182 (2) (2009) 1119–1128. creased and activated in eosinophilic asthma, Allergy 72 (10) (2017) 1532–1539.
[34] T. Ito, K. Hirose, A. Norimoto, T. Tamachi, M. Yokota, A. Saku, H. Takatori, [60] C.R. Brooks, C.J. van Dalen, I.F. Hermans, P.G. Gibson, J.L. Simpson, J. Douwes,
S. Saijo, Y. Iwakura, H. Nakajima, Dectin-1 plays an important role in house dust Sputum basophils are increased in eosinophilic asthma compared with non-eosi-
mite-induced allergic airway inflammation through the activation of CD11b+ nophilic asthma phenotypes, Allergy 72 (10) (2017) 1583–1586.
dendritic cells, J. Immunol. 198 (1) (2017) 61–70. [61] K. Amin, The role of mast cells in allergic inflammation, Respir. Med. 106 (1)
[35] J. Han, A. Dakhama, Y. Jia, M. Wang, W. Zeng, K. Takeda, Y. Shiraishi, (2012) 9–14.
M. Okamoto, S.F. Ziegler, E.W. Gelfand, Responsiveness to respiratory syncytial [62] M. Ro, A.J. Lee, J.H. Kim, 5-/12-Lipoxygenase-linked cascade contributes to the
virus in neonates is mediated through thymic stromal lymphopoietin and OX40 IL-33-induced synthesis of IL-13 in mast cells, thus promoting asthma develop-
ligand, J. Allergy Clin. Immunol. 130 (5) (2012) 1175–1186 e9. ment, Allergy 73 (2) (2018) 350–360.
[36] B. Bleck, A. Kazeros, K. Bakal, L. Garcia-Medina, A. Adams, M. Liu, R.A. Lee, [63] K. Amin, D. Ludviksdottir, C. Janson, O. Nettelbladt, E. Bjornsson, G.M. Roomans,
D.B. Tse, A. Chiu, G. Grunig, J.P. Egan 3rd, J. Reibman, Coexpression of type 2 G. Boman, L. Seveus, P. Venge, Inflammation and structural changes in the air-
immune targets in sputum-derived epithelial and dendritic cells from asthmatic ways of patients with atopic and nonatopic asthma. BHR Group, Am. J. Respir.
subjects, J. Allergy Clin. Immunol. 136 (3) (2015) 619–627 e5. Crit. Care Med. 162 (6) (2000) 2295–2301.
[37] D. Tworek, S.G. Smith, B.M. Salter, A.J. Baatjes, T. Scime, R. Watson, C. Obminski, [64] P. Bradding, C. Brightling, Mast cell infiltration of airway smooth muscle in
G.M. Gauvreau, P.M. O’Byrne, IL-25 receptor expression on airway dendritic cells asthma, Respir. Med. 101 (5) (2007) 1045 author reply 1046-7.
after allergen challenge in subjects with asthma, Am. J. Respir. Crit. Care Med. 193 [65] M.L. Fajt, S.L. Gelhaus, B. Freeman, C.E. Uvalle, J.B. Trudeau, F. Holguin,
(9) (2016) 957–964. S.E. Wenzel, Prostaglandin D(2) pathway upregulation: relation to asthma se-
[38] A. Froidure, C. Shen, D. Gras, J. Van Snick, P. Chanez, C. Pilette, Myeloid dendritic verity, control, and TH2 inflammation, J. Allergy Clin. Immunol. 131 (6) (2013)
cells are primed in allergic asthma for thymic stromal lymphopoietin-mediated 1504–1512.
induction of Th2 and Th9 responses, Allergy 69 (8) (2014) 1068–1076. [66] T. Boonpiyathad, G. Capova, H.W. Duchna, A.L. Croxford, H. Farine, A. Dreher,
[39] S.R. Damle, R.K. Martin, C.L. Cockburn, J.C. Lownik, J.A. Carlyon, A.D. Smith, M. Clozel, J. Schreiber, P. Kubena, N. Lunjani, D. Mirer, B. Ruckert,
D.H. Conrad, ADAM10 and Notch1 on murine dendritic cells control the devel- P. Satitsuksanoa, G. Tan, P.M.A. Groenen, M. Akdis, D.S. Strasser, E.D. Renner,
opment of type 2 immunity and IgE production, Allergy 73 (1) (2018) 125–136. C.A. Akdis, Impact of high altitude therapy on Type-2 immune responses in asthma
[40] C. Pilette, M.R. Jacobson, C. Ratajczak, B. Detry, G. Banfield, J. VanSnick, patients, Allergy (2019).
S.R. Durham, K.T. Nouri-Aria, Aberrant dendritic cell function conditions Th2-cell [67] A. Matucci, A. Vultaggio, E. Maggi, I. Kasujee, Is IgE or eosinophils the key player
polarization in allergic rhinitis, Allergy 68 (3) (2013) 312–321. in allergic asthma pathogenesis? Are we asking the right question? Respir. Res. 19
[41] C. Iwamura, T. Nakayama, Role of CD1d- and MR1-Restricted t cells in asthma, (1) (2018) 113.
Front. Immunol. 9 (2018) 1942. [68] J.T. Schroeder, A.P. Bieneman, K.L. Chichester, R.G. Hamilton, H. Xiao, S.S. Saini,
[42] Y.I. Koh, J.U. Shim, Association between sputum natural killer T cells and eosi- M.C. Liu, Decreases in human dendritic cell-dependent T(H)2-like responses after
nophilic airway inflammation in human asthma, Int. Arch. Allergy Immunol. 153 acute in vivo IgE neutralization, J. Allergy Clin. Immunol. 125 (4) (2010)
(3) (2010) 239–248. 896–901 e6.
[43] J.C. Carpio-Pedroza, G. Vaughan, B.E. del Rio-Navarro, J.M. del Rio-Chivardi, [69] S.H. Khan, M.H. Grayson, Cross-linking IgE augments human conventional den-
A. Vergara-Castaneda, L.A. Jimenez-Zamudio, A. Morales-Flores, G. Rodriguez- dritic cell production of CC chemokine ligand 28, J. Allergy Clin. Immunol. 125
Moreno, K. Ruiz-Tovar, S. Fonseca-Coronado, L.M. Goncalves Rossi, A. Escobar- (1) (2010) 265–267.
Gutierrez, Participation of CD161(+) and invariant natural killer T cells in pe- [70] J.P. Lynch, S.B. Mazzone, M.J. Rogers, J.J. Arikkatt, Z. Loh, A.L. Pritchard,
diatric asthma exacerbations, Allergy Asthma Proc. 34 (1) (2013) 84–92. J.W. Upham, S. Phipps, The plasmacytoid dendritic cell: at the cross-roads in
[44] Y. Endo, K. Hirahara, T. Iinuma, K. Shinoda, D.J. Tumes, H.K. Asou, N. Matsugae, asthma, Eur. Respir. J. 43 (1) (2014) 264–275.
K. Obata-Ninomiya, H. Yamamoto, S. Motohashi, K. Oboki, S. Nakae, H. Saito, [71] M. Dullaers, M.J. Schuijs, M. Willart, K. Fierens, J. Van Moorleghem, H. Hammad,
Y. Okamoto, T. Nakayama, The interleukin-33-p38 kinase axis confers memory T B.N. Lambrecht, House dust mite-driven asthma and allergen-specific T cells de-
helper 2 cell pathogenicity in the airway, Immunity 42 (2) (2015) 294–308. pend on B cells when the amount of inhaled allergen is limiting, J. Allergy Clin.
[45] A. Zakeri, M. Russo, Dual role of toll-like receptors in human and experimental Immunol. 140 (1) (2017) 76–88 e7.
asthma models, Front. Immunol. 9 (2018) 1027. [72] D.S. Ferreira, R.M. Carvalho-Pinto, M.G. Gregorio, R. Annoni, A.M. Teles,
[46] D. Vercelli, Discovering susceptibility genes for asthma and allergy, Nat. Rev. M. Buttignol, B.B. Araujo-Paulino, E.H. Katayama, B.L. Oliveira, H.S. Del Frari,
Immunol. 8 (3) (2008) 169–182. A. Cukier, M. Dolhnikoff, R. Stelmach, K.F. Rabe, T. Mauad, Airway pathology in
[47] A. Trompette, S. Divanovic, A. Visintin, C. Blanchard, R.S. Hegde, R. Madan, severe asthma is related to airflow obstruction but not symptom control, Allergy
P.S. Thorne, M. Wills-Karp, T.L. Gioannini, J.P. Weiss, C.L. Karp, Allergenicity 73 (3) (2018) 635–643.
resulting from functional mimicry of a Toll-like receptor complex protein, Nature [73] S. Palaniyandi, E. Tomei, Z. Li, D.H. Conrad, X. Zhu, CD23-dependent transcytosis
457 (7229) (2009) 585–588. of IgE and immune complex across the polarized human respiratory epithelial
[48] V.O. Millien, W. Lu, J. Shaw, X. Yuan, G. Mak, L. Roberts, L.Z. Song, J.M. Knight, cells, J. Immunol. 186 (6) (2011) 3484–3496.
C.J. Creighton, A. Luong, F. Kheradmand, D.B. Corry, Cleavage of fibrinogen by [74] J.J. Heeringa, L. Rijvers, N.J. Arends, G.J. Driessen, S.G. Pasmans, J.J.M. van
proteinases elicits allergic responses through Toll-like receptor 4, Science 341 Dongen, J.C. de Jongste, M.C. van Zelm, IgE-expressing memory B cells and
(6147) (2013) 792–796. plasmablasts are increased in blood of children with asthma, food allergy, and
[49] O. Denis, M. Vincent, X. Havaux, S. De Prins, G. Treutens, K. Huygen, Induction of atopic dermatitis, Allergy 73 (6) (2018) 1331–1336.
the specific allergic immune response is independent of proteases from the fungus [75] O.H. Wittekindt, Tight junctions in pulmonary epithelia during lung inflammation,
Alternaria alternata, Eur. J. Immunol. 43 (4) (2013) 907–917. Pflugers Arch. 469 (1) (2017) 135–147.
[50] H.J. McSorley, N.F. Blair, K.A. Smith, A.N. McKenzie, R.M. Maizels, Blockade of IL- [76] N.V. Chemuturi, P. Hayden, M. Klausner, M.D. Donovan, Comparison of human
33 release and suppression of type 2 innate lymphoid cell responses by helminth tracheal/bronchial epithelial cell culture and bovine nasal respiratory explants for
secreted products in airway allergy, Mucosal Immunol. 7 (5) (2014) 1068–1078. nasal drug transport studies, J. Pharm. Sci. 94 (9) (2005) 1976–1985.
[51] K. Nakagome, M. Nagata, Involvement and possible role of eosinophils in asthma [77] P. Licona-Limon, L.K. Kim, N.W. Palm, R.A. Flavell, TH2, allergy and group 2
exacerbation, Front. Immunol. 9 (2018) 2220. innate lymphoid cells, Nat. Immunol. 14 (6) (2013) 536–542.
[52] M. Terl, V. Sedlak, P. Cap, R. Dvorakova, V. Kasak, T. Koci, B. Novotna, [78] F. Roan, K. Obata-Ninomiya, S.F. Ziegler, Epithelial cell-derived cytokines: more

7
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

than just signaling the alarm, J. Clin. Invest. 129 (4) (2019) 1441–1451. [104] J.L. Simpson, P.G. Gibson, I.A. Yang, J. Upham, A. James, P.N. Reynolds, S. Hodge,
[79] D.J. Jackson, H. Makrinioti, B.M. Rana, B.W. Shamji, M.B. Trujillo-Torralbo, A.S.R. Group, Impaired macrophage phagocytosis in non-eosinophilic asthma,
J. Footitt, D.-R. Jerico, A.G. Telcian, A. Nikonova, J. Zhu, J. Aniscenko, Clin. Exp. Allergy 43 (1) (2013) 29–35.
L. Gogsadze, E. Bakhsoliani, S. Traub, J. Dhariwal, J. Porter, D. Hunt, T. Hunt, [105] M.A. Berry, B. Hargadon, M. Shelley, D. Parker, D.E. Shaw, R.H. Green,
T. Hunt, L.A. Stanciu, M. Khaitov, N.W. Bartlett, M.R. Edwards, O.M. Kon, P. Bradding, C.E. Brightling, A.J. Wardlaw, I.D. Pavord, Evidence of a role of
P. Mallia, N.G. Papadopoulos, C.A. Akdis, J. Westwick, M.J. Edwards, tumor necrosis factor alpha in refractory asthma, N. Engl. J. Med. 354 (7) (2006)
D.J. Cousins, R.P. Walton, S.L. Johnston, IL-33-dependent type 2 inflammation 697–708.
during rhinovirus-induced asthma exacerbations in vivo, Am. J. Respir. Crit. Care [106] M. Raundhal, C. Morse, A. Khare, T.B. Oriss, J. Milosevic, J. Trudeau, R. Huff,
Med. 190 (12) (2014) 1373–1382. J. Pilewski, F. Holguin, J. Kolls, S. Wenzel, P. Ray, A. Ray, High IFN-gamma and
[80] B. Saatian, F. Rezaee, S. Desando, J. Emo, T. Chapman, S. Knowlden, S.N. Georas, low SLPI mark severe asthma in mice and humans, J. Clin. Invest. 125 (8) (2015)
Interleukin-4 and interleukin-13 cause barrier dysfunction in human airway epi- 3037–3050.
thelial cells, Tissue Barriers 1 (2) (2013) e24333. [107] B.J. Lee, H.G. Moon, T.S. Shin, S.G. Jeon, E.Y. Lee, Y.S. Gho, C.G. Lee, Z. Zhu,
[81] K. Sugita, C.A. Steer, I. Martinez-Gonzalez, C. Altunbulakli, H. Morita, F. Castro- J.A. Elias, Y.K. Kim, Protective effects of basic fibroblast growth factor in the
Giner, T. Kubo, P. Wawrzyniak, B. Ruckert, K. Sudo, S. Nakae, K. Matsumoto, development of emphysema induced by interferon-gamma, Exp. Mol. Med. 43 (4)
L. O’Mahony, M. Akdis, F. Takei, C.A. Akdis, Type 2 innate lymphoid cells disrupt (2011) 169–178.
bronchial epithelial barrier integrity by targeting tight junctions through IL-13 in [108] T. Nabe, Tumor necrosis factor alpha-mediated asthma? Int. Arch. Allergy
asthmatic patients, J. Allergy Clin. Immunol. 141 (1) (2018) 300–310 e11. Immunol. 160 (2) (2013) 111–113.
[82] T. Kubo, P. Wawrzyniak, H. Morita, K. Sugita, K. Wanke, J.I. Kast, C. Altunbulakli, [109] P.S. Thomas, D.H. Yates, P.J. Barnes, Tumor necrosis factor-alpha increases airway
B. Ruckert, B. Jakiela, M. Sanak, M. Akdis, C.A. Akdis, CpG-DNA enhances the responsiveness and sputum neutrophilia in normal human subjects, Am. J. Respir.
tight junction integrity of the bronchial epithelial cell barrier, J. Allergy Clin. Crit. Care Med. 152 (1) (1995) 76–80.
Immunol. 136 (5) (2015) 1413–1416 e1-8. [110] C. Brightling, M. Berry, Y. Amrani, Targeting TNF-alpha: a novel therapeutic ap-
[83] Y.H. Wang, T. Ito, Y.H. Wang, B. Homey, N. Watanabe, R. Martin, C.J. Barnes, proach for asthma, J. Allergy Clin. Immunol. 121 (1) (2008) 5–10 quiz 11-2.
B.W. McIntyre, M. Gilliet, R. Kumar, Z. Yao, Y.J. Liu, Maintenance and polariza- [111] H.Y. Kim, D.T. Umetsu, R.H. Dekruyff, Innate lymphoid cells in asthma: Will they
tion of human TH2 central memory T cells by thymic stromal lymphopoietin-ac- take your breath away? Eur. J. Immunol. 46 (4) (2016) 795–806.
tivated dendritic cells, Immunity 24 (6) (2006) 827–838. [112] H.Y. Kim, H.J. Lee, Y.J. Chang, M. Pichavant, S.A. Shore, K.A. Fitzgerald,
[84] R.J. Ray, C. Furlonger, D.E. Williams, C.J. Paige, Characterization of thymic Y. Iwakura, E. Israel, K. Bolger, J. Faul, R.H. DeKruyff, D.T. Umetsu, Interleukin-
stromal-derived lymphopoietin (TSLP) in murine B cell development in vitro, Eur. 17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate
J. Immunol. 26 (1) (1996) 10–16. obesity-associated airway hyperreactivity, Nat. Med. 20 (1) (2014) 54–61.
[85] M. Kitajima, H.C. Lee, T. Nakayama, S.F. Ziegler, TSLP enhances the function of [113] L. Everaere, S. Ait-Yahia, O. Molendi-Coste, H. Vorng, S. Quemener, P. LeVu,
helper type 2 cells, Eur. J. Immunol. 41 (7) (2011) 1862–1871. S. Fleury, E. Bouchaert, Y. Fan, C. Duez, P. de Nadai, B. Staels, D. Dombrowicz,
[86] J. Mjosberg, J. Bernink, K. Golebski, J.J. Karrich, C.P. Peters, B. Blom, et al., The A. Tsicopoulos, Innate lymphoid cells contribute to allergic airway disease ex-
transcription factor GATA3 is essential for the function of human type 2 innate acerbation by obesity, J. Allergy Clin. Immunol. 138 (5) (2016) 1309–1318 e11.
lymphoid cells, Immunity 37 (4) (2012) 649–659. [114] N. Sharma, M. Akkoyunlu, R.L. Rabin, Macrophages-common culprit in obesity
[87] A. Cianferoni, J. Spergel, The importance of TSLP in allergic disease and its role as and asthma, Allergy 73 (6) (2018) 1196–1205.
a potential therapeutic target, Expert Rev. Clin. Immunol. 10 (11) (2014) [115] P.P. Hekking, M.J. Loza, S. Pavlidis, B. de Meulder, D. Lefaudeux, F. Baribaud,
1463–1474. C. Auffray, A.H. Wagener, P.I. Brinkman, R.I. Lutter, A.T. Bansal, A.R. Sousa,
[88] C.K. Wong, S. Hu, P.F. Cheung, C.W. Lam, Thymic stromal lymphopoietin induces S.A. Bates, Y. Pandis, L.J. Fleming, D.E. Shaw, S.J. Fowler, Y. Guo, A. Meiser,
chemotactic and prosurvival effects in eosinophils: implications in allergic in- K. Sun, J. Corfield, P.H. Howarth, E.H. Bel, I.M. Adcock, K.F. Chung,
flammation, Am. J. Respir. Cell Mol. Biol. 43 (3) (2010) 305–315. R. Djukanovic, P.J. Sterk, U.B.S. Group, Pathway discovery using transcriptomic
[89] A. James, C. Janson, A. Malinovschi, C. Holweg, K. Alving, J. Ono, S. Ohta, A. Ek, profiles in adult-onset severe asthma, J. Allergy Clin. Immunol. 141 (4) (2018)
R. Middelveld, B. Dahlen, B. Forsberg, K. Izuhara, S.E. Dahlen, Serum periostin 1280–1290.
relates to type-2 inflammation and lung function in asthma: data from the large [116] E.B. Brandt, M.B. Kovacic, G.B. Lee, A.M. Gibson, T.H. Acciani, T.D. Le Cras,
population-based cohort Swedish GA(2)LEN, Allergy 72 (11) (2017) 1753–1760. P.H. Ryan, A.L. Budelsky, G.K. Khurana Hershey, Diesel exhaust particle induction
[90] J.M. Lopez-Guisa, C. Powers, D. File, E. Cochrane, N. Jimenez, J.S. Debley, Airway of IL-17A contributes to severe asthma, J. Allergy Clin. Immunol. 132 (5) (2013)
epithelial cells from asthmatic children differentially express proremodeling fac- 1194–1204 e2.
tors, J. Allergy Clin. Immunol. 129 (4) (2012) 990–997 e6. [117] R. Polosa, N.C. Thomson, Smoking and asthma: dangerous liaisons, Eur. Respir. J.
[91] K. Izuhara, S.J. Conway, B.B. Moore, H. Matsumoto, C.T. Holweg, J.G. Matthews, 41 (3) (2013) 716–726.
J.R. Arron, Roles of periostin in respiratory disorders, Am. J. Respir. Crit. Care [118] Z. Zhang, J.M. Biagini Myers, E.B. Brandt, P.H. Ryan, M. Lindsey, R.A. Mintz-Cole,
Med. 193 (9) (2016) 949–956. et al., Beta-Glucan exacerbates allergic asthma independent of fungal sensitization
[92] S.P. Peters, Asthma phenotypes: nonallergic (intrinsic) asthma, J. Allergy Clin. and promotes steroid-resistant TH2/TH17 responses, J. Allergy Clin. Immunol.
Immunol. Pract. 2 (6) (2014) 650–652. 139 (1) (2017) 54–65 e8.
[93] N. Novak, T. Bieber, Allergic and nonallergic forms of atopic diseases, J. Allergy [119] J.L. Devalia, R.J. Sapsford, D.R. Cundell, C. Rusznak, A.M. Campbell, R.J. Davies,
Clin. Immunol. 112 (2) (2003) 252–262. Human bronchial epithelial cell dysfunction following in vitro exposure to ni-
[94] K.J. Baines, J.L. Simpson, L.G. Wood, R.J. Scott, N.L. Fibbens, H. Powell, trogen dioxide, Eur. Respir. J. 6 (9) (1993) 1308–1316.
D.C. Cowan, D.R. Taylor, J.O. Cowan, P.G. Gibson, Sputum gene expression sig- [120] C. Rusznak, J.L. Devalia, R.J. Sapsford, R.J. Davies, Ozone-induced mediator re-
nature of 6 biomarkers discriminates asthma inflammatory phenotypes, J. Allergy lease from human bronchial epithelial cells in vitro and the influence of nedo-
Clin. Immunol. 133 (4) (2014) 997–1007. cromil sodium, Eur. Respir. J. 9 (11) (1996) 2298–2305.
[95] W. Al-Ramli, D. Prefontaine, F. Chouiali, J.G. Martin, R. Olivenstein, C. Lemiere, [121] S.L. O’Beirne, S.A. Shenoy, J. Salit, Y. Strulovici-Barel, R.J. Kaner, S. Visvanathan,
Q. Hamid, T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma, J. J.S. Fine, J.G. Mezey, R.G. Crystal, Ambient pollution-related reprogramming of
Allergy Clin. Immunol. 123 (5) (2009) 1185–1187. the human small airway epithelial transcriptome, Am. J. Respir. Crit. Care Med.
[96] F.L.M. Ricciardolo, V. Sorbello, A. Folino, F. Gallo, G.M. Massaglia, G. Favata, 198 (11) (2018) 1413–1422.
S. Conticello, D. Vallese, F. Gani, M. Malerba, G. Folkerts, G. Rolla, M. Profita, [122] C. Mathis, C. Poussin, D. Weisensee, S. Gebel, A. Hengstermann, A. Sewer,
T. Mauad, A. Di Stefano, G. Ciprandi, Identification of IL-17F/frequent exacerbator V. Belcastro, Y. Xiang, S. Ansari, S. Wagner, J. Hoeng, M.C. Peitsch, Human
endotype in asthma, J. Allergy Clin. Immunol. 140 (2) (2017) 395–406. bronchial epithelial cells exposed in vitro to cigarette smoke at the air-liquid in-
[97] E.S. Chambers, A.M. Nanzer, P.E. Pfeffer, D.F. Richards, P.M. Timms, terface resemble bronchial epithelium from human smokers, Am. J. Physiol. Lung
A.R. Martineau, C.J. Griffiths, C.J. Corrigan, C.M. Hawrylowicz, Distinct en- Cell Mol. Physiol. 304 (7) (2013) L489–503.
dotypes of steroid-resistant asthma characterized by IL-17A(high) and IFN-gamma [123] H.T. Tan, S. Hagner, F. Ruchti, U. Radzikowska, G. Tan, C. Altunbulakli,
(high) immunophenotypes: potential benefits of calcitriol, J. Allergy Clin. A. Eljaszewicz, M. Moniuszko, M. Akdis, C.A. Akdis, H. Garn, M. Sokolowska,
Immunol. 136 (3) (2015) 628–637 e4. Tight junction, mucin, and inflammasome-related molecules are differentially
[98] G. Huang, Y. Wang, H. Chi, Regulation of TH17 cell differentiation by innate expressed in eosinophilic, mixed, and neutrophilic experimental asthma in mice,
immune signals, Cell. Mol. Immunol. 9 (4) (2012) 287–295. Allergy 74 (2) (2019) 294–307.
[99] M.J. McGeachy, Y. Chen, C.M. Tato, A. Laurence, B. Joyce-Shaikh, [124] I.D. Pavord, S. Korn, P. Howarth, E.R. Bleecker, R. Buhl, O.N. Keene, H. Ortega,
W.M. Blumenschein, T.K. McClanahan, J.J. O’Shea, D.J. Cua, The interleukin 23 P. Chanez, Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre,
receptor is essential for the terminal differentiation of interleukin 17-producing double-blind, placebo-controlled trial, Lancet 380 (9842) (2012) 651–659.
effector T helper cells in vivo, Nat. Immunol. 10 (3) (2009) 314–324. [125] G.G. Brusselle, T. Maes, K.R. Bracke, Eosinophils in the spotlight: eosinophilic
[100] T.H. Lee, H.J. Song, C.S. Park, Role of inflammasome activation in development airway inflammation in nonallergic asthma, Nat. Med. 19 (8) (2013) 977–979.
and exacerbation of asthma, Asia Pac. Allergy 4 (4) (2014) 187–196. [126] J.M. Mjosberg, S. Trifari, N.K. Crellin, C.P. Peters, C.M. van Drunen, B. Piet,
[101] G.J. Martinez, R.I. Nurieva, X.O. Yang, C. Dong, Regulation and function of W.J. Fokkens, T. Cupedo, H. Spits, Human IL-25- and IL-33-responsive type 2
proinflammatory TH17 cells, Ann. N. Y. Acad. Sci. 1143 (2008) 188–211. innate lymphoid cells are defined by expression of CRTH2 and CD161, Nat.
[102] F. Ozseker, S. Buyukozturk, B. Depboylu, D. Yilmazbayhan, E. Karayigit, Immunol. 12 (11) (2011) 1055–1062.
A. Gelincik, S. Genc, B. Colakoglu, M. Dal, H. Issever, Serum amyloid A (SAA) in [127] C. Barnig, M. Cernadas, S. Dutile, X. Liu, M.A. Perrella, S. Kazani, M.E. Wechsler,
induced sputum of asthmatics: a new look to an old marker, Int. E. Israel, B.D. Levy, Lipoxin A4 regulates natural killer cell and type 2 innate
Immunopharmacol. 6 (10) (2006) 1569–1576. lymphoid cell activation in asthma, Sci. Transl. Med. 5 (174) (2013) 174ra26.
[103] J.L. Simpson, S. Phipps, K.J. Baines, K.M. Oreo, L. Gunawardhana, P.G. Gibson, [128] M.F. Moffatt, I.G. Gut, F. Demenais, D.P. Strachan, E. Bouzigon, S. Heath, E. von
Elevated expression of the NLRP3 inflammasome in neutrophilic asthma, Eur. Mutius, M. Farrall, M. Lathrop, W. Cookson, G. Consortium, A large-scale, con-
Respir. J. 43 (4) (2014) 1067–1076. sortium-based genomewide association study of asthma, N. Engl. J. Med. 363 (13)

8
T. Boonpiyathad, et al. Seminars in Immunology 46 (2019) 101333

(2010) 1211–1221. regulation of allergy, Allergol. Int. 68 (1) (2019) 9–16.


[129] K.R. Bartemes, K. Iijima, T. Kobayashi, G.M. Kephart, A.N. McKenzie, H. Kita, IL- [132] Q.N. Yu, Y.B. Guo, X. Li, C.L. Li, W.P. Tan, X.L. Fan, Z.L. Qin, D. Chen, W.P. Wen,
33-responsive lineage- CD25+ CD44(hi) lymphoid cells mediate innate type 2 S.G. Zheng, Q.L. Fu, ILC2 frequency and activity are inhibited by glucocorticoid
immunity and allergic inflammation in the lungs, J. Immunol. 188 (3) (2012) treatment via STAT pathway in patients with asthma, Allergy 73 (9) (2018)
1503–1513. 1860–1870.
[130] H.Y. Kim, Y.J. Chang, S. Subramanian, H.H. Lee, L.A. Albacker, [133] M. Nagata, J.B. Sedgwick, H. Kita, W.W. Busse, Granulocyte macrophage colony-
P. Matangkasombut, P.B. Savage, A.N. McKenzie, D.E. Smith, J.B. Rottman, stimulating factor augments ICAM-1 and VCAM-1 activation of eosinophil func-
R.H. DeKruyff, D.T. Umetsu, Innate lymphoid cells responding to IL-33 mediate tion, Am. J. Respir. Cell Mol. Biol. 19 (1) (1998) 158–166.
airway hyperreactivity independently of adaptive immunity, J. Allergy Clin. [134] K. Nakagome, M. Nagata, Pathogenesis of airway inflammation in bronchial
Immunol. 129 (1) (2012) 216–227 e1-6. asthma, Auris Nasus Larynx 38 (5) (2011) 555–563.
[131] T.A. Doherty, D.H. Broide, Airway innate lymphoid cells in the induction and

You might also like