You are on page 1of 15

Review

Innate
Journal of J Innate Immun Received: December 18, 2017
Immunity DOI: 10.1159/000487057 Accepted after revision: January 18, 2018
Published online: February 13, 2018

Innate Immunity of the Lung: From Basic


Mechanisms to Translational Medicine
Dominik Hartl a, b Rabindra Tirouvanziam c Julie Laval a Catherine M. Greene d
       

David Habiel e Lokesh Sharma f Ali Önder Yildirim g Charles S. Dela Cruz h


       

Cory M. Hogaboam e  

a Department of Pediatrics I, Children’s Hospital, University of Tübingen, Tübingen, Germany; b Roche Pharma
 

Research and Early Development (pRED), Immunology, Inflammation and Infectious Diseases (I3) Discovery
and Translational Area, Roche Innovation Center Basel, Basel, Switzerland; c Department of Pediatrics, Emory
 

University School of Medicine, Center for Cystic Fibrosis and Airways Disease Research, Children’s Healthcare of
Atlanta, Atlanta, GA, USA; d Department of Clinical Microbiology, Royal College of Surgeons in Ireland Education
 

and Research Centre, Beaumont Hospital, Dublin, Ireland; e Division of Pulmonary and Critical Care Medicine,
 

Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA; f Section of Pulmonary, Critical Care
 

and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA;
g Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München,
 

Neuherberg, Germany; h Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
 

and Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA

Keywords host-pathogen, host-allergen, and host-particle interactions


Lung · Immunity · Immune cells · Neutrophils · within the mucosal airway compartment. Here, we summa-
Macrophages · Innate lymphoid cells · Epithelial cells rize and discuss recent findings on pulmonary innate immu-
nity and highlight key pathways relevant for biomarker and
therapeutic targeting strategies for acute and chronic dis-
Abstract eases of the respiratory tract. © 2018 S. Karger AG, Basel
The respiratory tract is faced daily with 10,000 L of inhaled
air. While the majority of air contains harmless environmen-
tal components, the pulmonary immune system also has to
cope with harmful microbial or sterile threats and react rap- The Lung as a Critical Immune Interface
idly to protect the host at this intimate barrier zone. The air-
ways are endowed with a broad armamentarium of cellular The human respiratory tract is faced daily with 10,000
and humoral host defense mechanisms, most of which be- L of inhaled air containing harmless environmental com-
long to the innate arm of the immune system. The complex ponents but also potentially airborne pathogens. This con-
interplay between resident and infiltrating immune cells and stant exposure requires a fine-tuned and rapidly acting
secreted innate immune proteins shapes the outcome of pulmonary immune system in order to immediately sense

© 2018 S. Karger AG, Basel Prof. Dr. Dominik Hartl


Department of Pediatrics I, Children’s Hospital, University of Tübingen
Hoppe-Seyler-Strasse 1
E-Mail karger@karger.com
DE–72076 Tübingen (Germany)
www.karger.com/jin E-Mail dominik.hartl @ med.uni-tuebingen.de
harmful microbial or sterile threats, and to protect the host tions for drug development. Because it is impossible to
at this intimate contact zone [1–3]. For that purpose, the cover this vast domain of research within a single review,
airways are endowed with a broad armamentarium of cel- we chose instead to highlight certain areas based on recent,
lular and humoral host defense mechanisms, most of exciting advances, and translational potential.
which belong to the innate arm of the immune system [1,
3]. The complex interplay between resident (airway epi-
thelial cells) and infiltrating immune cells acting in concert Airway Epithelial Cells as the First Line of Innate
with secreted innate immune proteins, such as defensins, Immune Sensing
mucins, or collectins, shapes the outcome of host-patho-
gen, host-allergen, and host-particle interactions within The defense functions of the airway epithelium and
the airway microenvironment. Airway epithelial cells, submucosa are well recognized and include regulation of
dendritic cells, and (in the lower airways) alveolar macro- barrier tightness, secretion of mucus and antimicrobials,
phages (AM) are the initial checkpoints that encounter in- and cooperation with various immune subsets via cyto-
haled antigens and trigger proinflammatory or tolerogen- kine, chemokine, and growth factor production. At the
ic/anti-inflammatory downstream immune responses. root of innate defense in the lung, however, is the ability
Major mediators communicating between airway sentinel of the airway epithelium to sense pathogens via a variety
cells and bone marrow-derived immune cells are che- of receptors. These include various families of pattern
moattractants, such as lipid mediators (prototypically recognition receptors (PRR) such as Toll-like receptors
eicosanoids/leukotrienes), complement factors, and che- (TLR), RIG-I-like receptors (RLR), protease-activated re-
mokines [4–7]. Of particular importance for the recruit- ceptors (PAR), Nod-like receptors (NLR), C-type lectin
ment of leukocytes into the airway microenvironment are receptors, and the bitter- and sweet-taste receptors.
the CXC chemokines CXCL1-8 and CXCL12, and the CC
chemokines CCL2, CCL17 (TARC), CCL18 (PARC), and TLR and RLR
CCL20. The biological effect of chemokines is relayed by TLR expression and function in airway epithelium
distinct cytokines that are released by local airway cells and have been widely studied. These PRR are expressed by
induce microenvironmentally tailored immune contexts, epithelial cells throughout the respiratory tract and re-
particularly IL-1- α and IL-1-β, IL-10, IL-17, IL-23, IL-25, spond rapidly to local microbial and host-derived factors
IL-33, and thymic stromal lymphopoietin. Through the present as a result of infection or tissue damage. Follow-
integrated action of proinflammatory lipid, chemokine, ing ligand recognition, TLR activate intracellular down-
complement, and cytokine mediators, different immune stream signaling cascades leading to changes in gene ex-
cell populations are sequentially recruited into the bron- pression associated with typical innate immune respons-
choalveolar and lung parenchymal compartments. In an es and activation of adaptive immunity. Tengroth et al.
acute inflammatory reaction, neutrophils are attracted [12] recently described the importance of the nucleic ac-
first and localize mainly in the bronchoalveolar space, id-sensing TLR (TLR3, TLR7, and TLR9) and the RLR
where they engage in short-term host-pathogen interac- RIG-I and MDA-5, which recognize replicating RNA vi-
tions [8], followed by monocytes and lymphocytes for ruses in infected cells, in nasal epithelium. These studies
more sustained/chronic host defense functions, with the using nasal biopsies and primary human nasal epithelial
latter having a clear tissue predominance for infiltrating cells stimulated with artificial TLR and RLR agonists
the lung parenchyma rather than the bronchoalveolar demonstrated how these receptors generate robust IL-6,
space [9, 10]. Within the pulmonary tissue, lymphocytes IL-8, and interferon (IFN)-β responses and act as a first
can form organized ectopic tertiary lymphoid organs, line of defense against viruses invading the respiratory
termed bronchus-associated lymphoid tissue, which can tract. In another study, reactive oxygen species produced
be permanent or inducible (the latter for humans) [11]. via Duox2 in influenza A virus-infected nasal epithelial
Besides these well-characterized components of the pul- cells were shown to increase RIG-I and MDA-5 mRNA in
monary immune system, recent studies highlight novel a process believed to resist IAV infection [13]. Kim et al.
immune cells and mediators, such as innate lymphoid cells [14] also evaluated inflammatory and antiviral responses
(ILC), mucosal-associated invariant T cells (MAIT), chi- to human virus infection in human nasal epithelial cells
tinase-like proteins (CLP), and others. Here we aim to from chronic rhinosinusitis patients, wherein decreased
summarize the different components of the innate pulmo- MDA-5 and IFN-β expressions were evident and human
nary immune response, with an emphasis on novel direc- rhinovirus clearance was slightly impaired.

2 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
C-Type Lectin Receptors gram-negative opportunistic bacterium Pseudomonas ae-
Another type of PRR essential for innate immunity in ruginosa, causing an increase in nitric oxide production
the pulmonary environment are C-type lectin receptors and enhanced mucociliary clearance [27]. Lee et al. [28]
that sense fungal patterns. On bronchial epithelial cells studied the regulation of bitter- and sweet-taste receptors
this is mainly orchestrated by dectin-1, which mediates in the upper airways and found that antimicrobial peptide
recognition of β-glucan motifs in Aspergillus fumigatus induction by T2R is repressed by activation of the sweet-
and the house dust mite (HDM) [15], leading to secretion taste receptor T1R2/3. Thus, activation of T1R2/3 and re-
of the dendritic cell chemokine CCL20. Other nonfungal pression of T2R due to increased intranasal glucose may
allergens, and specifically those with proteolytic proper- be responsible for chronic rhinosinusitis.
ties such as Derp1 and cockroach allergen, can elicit al-
lergic airway inflammation via PAR-2 when adminis- Other Recently Identified Receptors
tered through the mucosa [16]. In a study of allergic sen- Beyond these major families of PRR, new roles in
sitization and HDM-induced allergic airway inflammation innate immunity have emerged recently for other recep-
[17], PAR-2 was found to contribute to IgE responses but tors expressed by airway epithelium. Selected examples
was dispensable for proinflammatory cytokine secretion include: (1) the short-chain fatty acid receptor GPR41,
induced by HDM. In another study, it was demonstrated which is present at higher-than-normal levels in bron-
that, in addition to producing cytokines, allergen-activat- chial epithelium of cystic fibrosis (CF) patients and is re-
ed airway epithelial cells can also release uric acid [18], sponsible for exaggerated IL-8 induction in these cells in
thereby promoting TH2 sensitization and amplifying al- response to short-chain fatty acids from anaerobic bacte-
lergic inflammation [reviewed in 19]. ria present in CF lungs [29], (2) the fractalkine receptor
CX3CR1, which was recently reported to mediate respira-
Nod-Like Receptors tory syncytial virus-induced cytokine production in pri-
Among NLR, NOD1, NOD2, and NLRP3 are ex- mary human airway epithelial cells [30], and (3) an iso-
pressed by airway epithelial cells. The expression of form of the coxsackievirus and adenovirus receptor
NOD1 has been shown to be downregulated during pol- CAREx8a, expressed on the apical surface of airway epi-
len season among patients with allergic rhinitis [20], and thelial cells in culture, that enhances adenovirus entry
its normal activation can reduce airway hyperresponsive- into cells by coopting neutrophils [31].
ness accompanied by a reduction of allergen-specific T-
cell proliferation in allergen-induced lung inflammation
[21]. NLRP3 mediates cellular responses to inhaled par- Neutrophils
ticular matter (e.g., PM10) and has recently been elegant-
ly shown to have an important role in innate but not Neutrophils are the most abundant subset of leuko-
adaptive immune responses in airway epithelial cells [22]. cytes patrolling in blood in search of potential injuries
A novel NLR termed NLRX-1 has been identified in nasal and stimuli, e.g., pathogen- and damage-associated mo-
epithelium that is activated by double-stranded RNA and lecular pattern molecules (PAMP and DAMP), chemo-
participates in rhinoviral-mediated disruption of polar- kines, cytokines, and lipid mediators, which induce their
ized airway epithelial cell barrier function [23]. rapid recruitment to peripheral sites of inflammation.
The primary role fulfilled by neutrophils is pathogen kill-
Bitter- and Sweet-Taste Receptors ing and removal. Neutrophils are conventionally thought
One particularly exciting new finding relevant to sens- of as short-living cells with a limited capacity for de novo
ing capabilities in the airways is the identification of the mRNA transcription and protein production. However,
G-protein coupled bitter- and sweet-taste receptors (T2R recent evidence shows that they are endowed with exten-
and T1R, respectively) in respiratory epithelia [reviewed sive adaptive abilities, which underlie an equally exten-
in 24]. Extraoral taste receptors have been detected in hu- sive functional complexity [8].
man bronchial epithelial cells and specialized solitary si-
nonasal chemosensory cells in the upper respiratory tract Neutrophil Lifespan and Margination in the Lung
[25, 26]. Bitter taste receptors are activated by bacterial Vasculature
quorum-sensing molecules, whereas sweet receptors re- The human blood neutrophil lifespan was previously
spond to sugars. For example, the bitter taste receptor estimated to be shorter than a day; however, recent stud-
T2R38 is activated by homoserine lactones from the ies suggested an upper limit closer to 5 days, even in the

Innate Immunity of the Lung J Innate Immun 3


DOI: 10.1159/000487057
absence of inflammation [32]. This topic remains contro- ing and they are concomitant with de novo protein syn-
versial and requires further investigation. In addition to thesis, granule exocytosis, and later induction of apopto-
circulating freely in blood, a large fraction of neutrophils sis [40–42]. While typical surface receptors are altered in
is tethered to the lining of the liver, spleen, bone marrow, neutrophils during chronic lung inflammation, further
and lung vasculature and is referred to as the “marginat- studies suggested a more profound modulation of their
ed” pool [33]. Within the lung alone, this pool constitutes metabolism and function. In particular, recent studies
the most prominent reservoir of neutrophils in the sys- showed that upon recruitment into CF airways neutro-
temic circulation (∼40% of total body neutrophils) [34]. phils undergo significant activation of both cAMP re-
Key protective mechanisms have been attributed to the sponse element-binding protein (CREB) and mechanistic
lung marginated pool because it enables rapid neutrophil (formerly mammalian) target of rapamycin (mTOR)
recruitment inside of the tissue following injury and/or pathways, linked to surface upregulation of functional re-
infection. Additionally, the lung vasculature has been in- ceptors (e.g., the receptor for advanced glycation end
volved in neutrophil “de-priming” via its ability to filter products) and metabolite transporters (e.g., glucose and
and inactivate primed cells before they are rereleased in amino acid transporters Glut1 and ASCT2) [43–45].
the blood stream [35]. However, this mechanism of neu- Since CF airways are enriched in inflammatory mediators
trophil retention is impaired in acute respiratory distress and nutrients such as glucose and amino acids, these re-
syndrome patients, resulting in the exposure of periph- sults suggest the ability of neutrophils to adapt to specific
eral organs to primed systemic neutrophils, leading to se- microenvironments through coordinated stress respons-
vere complications [36]. es and metabolic reprogramming [reviewed in 46, 47].

Neutrophil Adaption to the Pulmonary Regulatory Activities of Lung Neutrophils


Microenvironment A hallmark of neutrophil activity within inflamed air-
As the first circulating immune subset recruited into ways is the extracellular release of proteases and oxidases
the lung upon infection or sterile inflammation, neutro- (e.g., neutrophil elastase and myeloperoxidase) following
phils orchestrate both pro- and anti-inflammatory ac- granule mobilization to the cell surface. This process, well
tions therein, as required to efficiently kill pathogens and recognized in airway diseases such as CF and COPD, in-
resolve inflammation. In fulfilling these tasks, neutro- duces an upregulation of characteristic markers for sec-
phils undergo profound phenotypic and functional ondary and primary granules (CD66b and CD63, respec-
changes, delineating distinct fates. Indeed, various neu- tively) on the surface of airway neutrophils and results in
trophil fates can be observed in blood and tissue under high oxidative and proteolytic activities. The latter was
both homeostatic and pathological conditions such as in- shown to be responsible for the cleavage as key receptors
fection, autoimmunity, and cancer [37]. Within the air- involved in neutrophil antibacterial activities such as
ways, recruited neutrophils display an activated pheno- CXCR1, CD16, and CD14 [43, 46–48]. Further studies
type (CD16high CD62Ldim CD11bhigh) under both healthy revealed a role for airway neutrophils in regulation of the
and inflamed conditions [38, 39]. When crossing into the adaptive immune system, further emphasizing the multi-
airway lumen, neutrophils further modulate the surface dimensional importance of neutrophil plasticity [37]. For
expression of chemokine and TLR, which together medi- example, a strong immunosuppressive function was
ate core inflammatory signaling. These changes in neu- identified in CF airway neutrophils, which can downreg-
trophil phenotypes include a lower expression level of re- ulate T-cell activity through the release and activation of
ceptors for the CXCL8 (IL8) chemokine, CXCR1 and arginase I, concomitant with granule exocytosis [49]. Ac-
CXCR2, than in blood. By contrast, a large fraction of tivated neutrophils may also impact T cells positively
neutrophils isolated from the bronchoalveolar lavage flu- within CF airways but also within the lymphatic compart-
id of patients suffering from chronic airway inflamma- ment by displaying antigen-presenting cell capabilities
tion upregulate CCR1, CCR2, CCR3, CCR5, CXCR3, and (e.g., expression of CD80, CD86, and MHC II). Interest-
CXCR4, as observed in CF, chronic obstructive pulmo- ingly, CXCR4 expression, usually characteristic of imma-
nary disease (COPD), and asthma, as well as TLR2, TLR4, ture or senescent neutrophils, is strongly upregulated on
TLR5, and TLR9, as observed in CF and non CF-bronchi- activated airway neutrophils and could relate to their ac-
ectasis, when compared to airway neutrophils from quired ability to egress from inflammatory tissues into
healthy subjects. These phenotypic changes are linked to lymphatic vessels [40, 43, 50]. Neutrophil transit toward
neutrophil respiratory burst activity and/or bacterial kill- lymph nodes has been associated with T-cell proliferation

4 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
and therefore participates in positive regulation of the concept of “neutrophil heterogeneity” originated by Gal-
adaptive immune response by neutrophils [50, 51]. In lin et al. [63] in 1984 has now emerged in full bloom, en-
some cases, lymphatic neutrophils could be used a “Tro- compassing the formation of distinct subsets in both
jan horses” by intracellular pathogens, which can use blood and peripheral tissues and raising particular inter-
them as a vehicle to spread throughout the body [52, 53]. est in the functional characterization of these subsets for
novel neutrophil-targeted therapies for CF, COPD, neu-
Neutrophil Extracellular Traps trophilic asthma, and other chronic airway diseases.
Until recently, neutrophils have been primarily recog-
nized as professional killers critical to the initial immune
response that leads to pathogen clearance, using both in- Macrophages
tracellular killing by phagocytosis and extracellular kill-
ing by exocytosis of granule components. In the past de- Macrophages, first discovered by Ilya Metchnikoff, be-
cade, a third effector mechanism used by neutrophils has long to the mononuclear phagocyte system and represent
been described, which consists of the release of extracel- potent antimicrobial innate immune cells that are found
lular complexes or “traps” formed by decondensed nucle- in all tissues in the human body. Macrophages in the pul-
ar DNA (e.g., after histone citrullination), histones, and monary compartment are classified and termed accord-
cationic effectors such as neutrophil elastase and myelo- ing to their anatomical location in the lung as alveolar or
peroxidase. The deployment of neutrophil extracellular interstitial macrophages [64]. Since interstitial macro-
traps (NET), termed “NETosis,” is believed to enable im- phages are less defined and more heterogeneous depend-
mobilization and possibly killing of pathogens, while pre- ing on the pulmonary subcompartment, the species stud-
cipitating neutrophil death [54, 55]. While NET have ied, and the disease model investigated, we will focus here
been associated with numerous pathologies including au- on AM.
toimmune disorders and infectious diseases related to
bacteria, fungi, and viruses, their role in acute and chron- AM Functions
ic inflammation has not yet been fully elucidated [8, 54]. AM are 15–50 μm in diameter, they are mainly located
In the context of airway diseases, studies focusing on NET in the alveolar space, and they represent the predominant
have shown both beneficial and harmful effects of these phagocytic and antigen-presenting cell in the human re-
structures [reviewed in 56]. The ability of NET to spread spiratory tract [65]. Under homeostatic/healthy condi-
out and trap microbes could enable an increased killing tions, AM are the most abundant cellular fraction with-
efficiency and reduction of the pathogen burden. Mitigat- in bronchoalveolar lavage fluids, while under acute or
ing these positive effects of NET are the facts that patho- chronic inflammatory conditions other leukocyte popu-
gens such as Staphylococcus aureus were shown to devel- lations, prototypically neutrophils (e.g., in acute infec-
oped NET evasion strategies, and that the extracellular tions, CF or acute respiratory distress syndrome) and
presence of host DNA, histones, neutrophil elastase, and lymphocytes (e.g., in sarcoidosis and allergic alveolitis)
myeloperoxidase can cause direct or indirect cell toxicity accumulate and shift this balance. Distinct from other tis-
and subsequent lung injury [57–59], as well as airway ob- sue macrophage subsets, AM are endowed with a remark-
struction via an increase in mucus viscosity [60–62]. able phenotypic, metabolic, and functional plasticity [65–
67]. Metabolically, AM exhibit a high basal glucose con-
Neutrophil Plasticity and Heterogeneity sumption and respiratory rate but a low respiratory burst
Taken together, numerous studies in the past decade activity. Phenotypically, they directly reflect the alveolar
have highlighted the adaptability of neutrophils in acute host-environment interface zone and contain granules of
and chronic immune responses, contradicting the con- exogenous material, as exemplified in chronic smokers in
ventional view that they are preprogrammed, poorly whom AM accumulate in the bronchoalveolar lavage flu-
adaptable cells, incapable of de novo protein synthesis, id, are larger in diameter and activated, and stain dark on
with a limited lifespan and primarily relying on oxidative cytospin preparations. By taking up inhaled environmen-
and proteolytic killing to carry out their function. The tal particles, pollutants, allergens, and airborne microbes,
identification of novel neutrophil functions and regula- AM serve as airway scavengers to keep the respiratory
tory mechanisms highlights their role in balancing pro- system clean and homeostatic. Another function related
and anti-inflammatory signaling in order to promote a to their potent phagocytic capacity is the maintenance of
swift return to homeostasis and limit tissue damage. The surfactant homeostasis by engulfing and catabolizing lo-

Innate Immunity of the Lung J Innate Immun 5


DOI: 10.1159/000487057
cal surrounding surfactant proteins, a functionality that on the respective tissue compartment and disease model
is impaired in a severe disorder called pulmonary alveolar analyzed. For a more detailed review of macrophage acti-
proteinosis, characterized by a dysfunction of the granu- vation, plasticity, and M1/M2 polarization, we refer read-
locyte/monocyte cell stimulating factor pathway and re- ers to recent reviews in that field [76–78]. Overall, the
lated AM functions [68, 69]. By sensing bronchoalveolar complex multifunctionality of AM, interacting as antigen-
PAMP and DAMP, AM integrate various microbial- and presenting cells with T cells, clearing apoptotic and ne-
host-derived signals and respond via the concerted re- crotic cells through efferocytosis, and killing pathogens
lease of various pro- or anti-inflammatory mediators and releasing both pro- and anti-inflammatory factors,
(mainly cytokines such as IL-1-β, IL-6, IL-10, or tumor makes a “good-versus-bad-cop” assignment for these cells
necrosis factor [TNF]-α) into the alveolar space. Recent for the majority of lung diseases challenging. In vivo mac-
studies further highlight that AM can have immunoregu- rophage depletion studies revealed different outcomes de-
latory roles. In murine asthma model systems, pulmo- pending on the lung disease model and the time point of
nary macrophages were found to generate regulatory T intervention used [64, 65, 79, 80]. Idiopathic pulmonary
cells to induce airway tolerance [70]. AM were further fibrosis (IPF) is a prototypic example of a complex chron-
found to attach to the alveolar wall (becoming “sessile”) ic lung disease in which the role of pulmonary macro-
and communicate with the alveolar epithelium to induce phages remains controversial. In this context, AM seem to
an immunosuppressive/-modulatory signal in order to play a dual role in lung fibrosis given their release of pro-
dampen lung inflammation [71]. fibrotic factors (such as TGF-β1 and PDGF), thereby driv-
ing disease progression and, on the other hand, their abil-
AM Ontogeny ity to release proteases (MMP) that can digest the extracel-
Ontologically, tissue macrophages either derive from lular matrix and thus exert antifibrotic/fibrinolytic
circulating/recruited monocytes or are established dur- activities. For a more thorough discussion of macrophage
ing embryogenesis (yolk sac and fetal liver), independent- phenotypes and functions in IPF, we refer readers to a re-
ly of monocytes, as supported by recent murine studies, cent review in that field [79].
and they populate different organs as long-lived tissue-
resident macrophages [64, 72]. However, the precise ori-
gin of AM across species remains a matter of intense on- Innate Lymphoid Cells
going investigations. Previous studies have shown that
AM develop from fetal monocytes that differentiate into ILC are a recently identified group of heterogeneous
long-lived mononuclear cells in the first week of life, me- innate immune cells belonging to the lymphoid lineage
diated through the granulocyte/monocyte cell-stimulat- but lacking antigen-specific receptors [81]. Mechanisti-
ing factor [73]. In murine fate-mapping studies, yolk sac- cally, ILC are involved in protective antimicrobial re-
derived erythro-myeloid progenitors have been identi- sponses, particularly at mucosal-barrier organs, but they
fied as the origin of several tissue-resident macrophage also play pathogenic roles in inflammation, autoimmuni-
populations, including AM [74]. The airway environ- ty, allergy, and fibrosis within tissues [81, 82]. ILC are dis-
ment/niche seems to be essential as well to shaping the tinguished from canonical T and B cells by their ontogeny/
unique phenotype and functional characteristics of AM development and the expression of a distinct set of cellular
in vivo [75]. For a more detailed and comprehensive re- markers [81]. Of note, ILC do not express RAG (recombi-
view of the ontogeny of AM, we refer readers to a recent nation-activating gene) genes, implying that ILC, in con-
dedicated review [64]. trast to T and B cells, can be activated directly [83]. ILC
typically express IL-2Rα (CD25), IL-7Rα (CD127), and IL-
AM Polarization 7Rγ (CD132) [84]. Depending on their ability to synthe-
Macrophages have a remarkable functional and pheno- size and release cytokines and their transcription factor
typic diversity and can be dichotomized into M1 (IFN-γ/ profile, ILC are divided into 3 distinct subsets.
classically activated) and M2 (IL-4/alternatively activated)
polarization phenotypes with different roles in cancer, in- Type I ILC
fection, allergy, and fibrosis. Despite compelling evidence ILC1, which include NK cells, are defined by analogy
in vitro, the in vivo relevance of this classification has been to Th1 cells according to their ability to release IFN-γ and
challenged and is continuously refined given the high TNF, and expression of the T-box transcription factor T-
plasticity and heterogeneity of macrophages depending bet or eomesodermin (Eomes) [85]. Across tissues, ILC1

6 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
cells are found mostly in the liver, the thymus, the uterus, cules (e.g., ICAM and VCAM) and chemokines (CXCL13,
skin, secondary lymphoid tissue, the spleen, the gut, and CCL19, and CCL21) required for the formation of lym-
the lung. ILC1 have been demonstrated to accumulate in phoid follicles [93]. Previous studies established that B-
response to viral and bacterial infections in mice [86]. cell-dependent inducible bronchus associated lymphoid
ILC1 were increased in lung specimen from patients with tissue formation is essential for emphysema development
severe COPD, while type 2 ILC (ILC2) were decreased in the cigarette smoke-induced COPD mouse model [94].
[87]. Based on these and the above data, it is tempting to specu-
late that LTi cells play a key role in the development of
Type 2 ILC COPD immunopathogenesis. However, while mechanis-
ILC2, the counterpart of Th2 cells, produce IL-4, IL-5, tic and functional contributions of ILC in animal models
IL-9, and IL-13 in response to IL-25, IL-33 and thymic of asthma and COPD have been studied to a certain ex-
stromal lymphopoietin and express high levels of the Th2 tent, there is very limited evidence for a potential role in
signature transcription factor GATA-3, which is neces- human disease conditions so far. With regards to lung fi-
sary for their functional maturation and maintenance brosis, the potential role of ILC is discussed in a compre-
[88, 89]. ILC2 are localized mostly in the healthy skin, hensive review [95]. In brief, given that IL-17A plays a key
adipose tissue, and lung of mice and humans and have role in pulmonary inflammation and fibrosis in COPD,
been mainly involved in tissue inflammation, remodel- IPF, and CF [96–98], and given that ILC3 are an essential
ing, and fibrosis. Interestingly, ILC2 are the main ILC source of IL-17 at mucosal sites ILC3s are suggested to
subset found in the murine lung, although they represent function as early orchestrators of lung tissue remodeling
only 0.4–1% of total lung cells. Using single-cell RNA se- and fibrogenesis. Very recently, a mast cell/ILC2/Th9
quencing of lung-resident ILC, a recent study demon- pathway has been shown to drive pulmonary inflamma-
strated that in the context of allergic lung inflammation tion in murine infection models of CF-like lung disease
the alarmins/cytokines IL-25 and IL-33 can induce acti- [99], although the precise role and functional relevance of
vation and expansion of lung-resident ILC2 [88]. In vivo this pathway and ILC overall in human CF lung disease
activation by IL-25 induced a high expression of neuro- remains to be defined by further translational studies.
medin U receptor 1 in ILC2. Interestingly, the combined
treatment with IL-25 and neuromedin U synergistically
promoted allergic inflammation. In line with previous Mucosal-Associated Invariant T Cells
studies showing increased numbers of ILC2 in the blood
of asthma patients [90], this study suggests that ILC2 MAIT are a group of innate-like T lymphocytes that
could represent a potentially predictive biomarker for pa- which are highly abundant in human blood (∼1–10%
tients with asthma. Interestingly, the deletion of IL-33 [100]) and in mucosal tissues such as the intestine and
prevented cigarette smoke-primed exacerbations in re- lungs [101–104]. These cells are characterized by their ex-
sponse to viral infection in mice and cigarette smoke de- pression of the invariant T-cell receptor (TCR) α chain,
creased the expression of the IL-33 receptor ST2 on ILC2 TRAV1–2 joined with TRAJ33 and a limited range of
[91]. Therefore, the IL-33/ILC2 axis may offer a potential TCRβ chains, and abundant expression of CD161 and
therapeutic target for controlling chronic lung diseases CD218 (IL18Rα) [102, 105, 106]. Recently, studies have
like asthma and COPD. shown that MAIT cells can recognize vitamin B meta-
bolic byproducts produced by a range of microorganisms
Type 3 ILC presented by the ubiquitously expressed class I MHC-re-
Type 3 ILC contain natural cytotoxicity receptor-posi- lated protein MR1 [107–110]. Activated MAIT cells have
tive (NCR+) ILC3 and NCR– lymphoid tissue inducer been shown to secrete high levels of IFN-γ, TNF-α, IL-17,
(LTi) cells, which depend on the transcription factor and cytotoxic/cytolytic perforin and granzymes A, B, and
RORγt and produce either IL-22 (NCR+ ILC3, a.k.a., K [111–116].
ILC22 cells) or both IL-17 and IL-22 (NCR– LTi, a.k.a.,
ILC17 cells) in response to IL-23 [82, 92]. LTi cells were MAIT Cell Frequency and Susceptibility to
reported to contribute to the development of secondary Corticosteroid Treatment
lymphoid tissue, wherein they activate stromal cells The frequency of MAIT cells in normal human lungs
through the lymphotoxin-α1β2-mediated LTβR signaling ranges from 2 to 20% of all T cells [104, 117]. Given their
pathway. This triggers the expression of adhesion mole- abundance in blood, many studies have examined blood

Innate Immunity of the Lung J Innate Immun 7


DOI: 10.1159/000487057
MAIT cells in various respiratory diseases. There were no tant in controlling bacterial infections, and deficiencies
change in MAIT cell frequency in the blood of inhaled in these cells occur in patients with active bacterial infec-
corticosteroid (ICS)-naive COPD patients relative to tions.
healthy donors; however, their frequency was significant-
ly reduced in ICS-treated COPD patients’ blood and Lung MAIT Cells in the Context of Bacterial Infection
bronchial biopsies [104]. The reduced MAIT cell fre- Identification of mechanisms propagated by MAIT
quency in blood was more pronounced in moderate-to- cells in pulmonary immunity has been hindered by the
severe-COPD patients, and it was associated with elevat- low abundance of these cells in germ-free mice. However,
ed serum C-reactive protein levels and a reduced lung following the development of iVα19 TCRα [130] trans-
function [118]. Consistent with a role of ICS in modulat- genic and B6-MAITCAST [131] mice, and the commer-
ing MAIT cell numbers, these were also found to be sig- cial availability of murine MR1 tetramers, recent studies
nificantly reduced in the blood and lung tissue of ICS- have characterized the role of these cells in models of pul-
treated asthma patients relative to normal individuals monary inflammation and infection. Utilizing iVα19-
[119, 120], a result that was especially prominent in pa- transgenic MR1-sufficient or iVα19-transgenic MR1-de-
tients with severe asthma [120]. Together, these studies ficient mice to study the role of MAIT cells in anti-bacte-
suggest that MAIT cells are susceptible to ICS treatment rial immunity, Le Bourhis et al. [123] observed that the
and that their deficiency may be associated with a more former had more activated MAIT cells and a lower bacte-
severe respiratory pathology. rial burden relative to the latter. In a model of pulmonary
bacterial infection, one study provided evidence for a key
MAIT Cells in Lung Pathologies role of MAIT cells in protecting against Francisella tula-
Experimental evidence suggested that MAIT cells can rensis (LVS) pulmonary infection [132], based on the ob-
be activated upon epithelial cell infection by multiple servation of an MR1-dependent recruitment and/or ex-
bacterial strains [111]. Indeed, an important role of these pansion of MAIT cells in LVS-infected mice, and a lower
cells in antibacterial immunity is supported by studies bacterial burden in the lungs of MR1-sufficient relative to
showing a significant inverse correlation between pe- MR1-deficient mice. In vitro coculture studies utilizing
ripheral blood MAIT cell numbers and CF disease sever- MAIT cells and LVS-infected macrophages indicated that
ity and an inverse association with P. aeruginosa infec- MAIT cell-derived IL-12p40, TNF, and IFN-γ were indis-
tion and the frequency of pulmonary exacerbations pensable in controlling intracellular bacterial growth in
[121]. Similarly, blood MAIT cells were also observed to macrophages. Finally, this study showed a delayed re-
be decreased in patients with tuberculosis (TB) and non- cruitment of effector CD4+ helper and CD8+ cytotoxic T
tuberculous mycobacterial infection [122, 123], and cells in MR1-deficient mice relative to their MR1-suffi-
their deficiency was correlated with disease severity cient counterparts. In a subsequent report, this group
[122]. In vitro stimulation of freshly isolated MAIT cells identified MAIT cell-derived granulocyte/monocyte cell
using phorbol ester, ionomycin, IL-15, anti-CD28 anti- stimulating factor to be required for differentiation of
bodies, and/or mycobacterial lysates suggested a severe CCR2+ monocytes into dendritic cells and the subse-
functional deficiency in TB patient-derived relative to quent recruitment of effector helper and cytotoxic T cells
healthy donor-derived MAIT cells, as assessed by the in- for efficient bacterial clearance [133]. Collectively, these
duction of IFN-γ and IL-17 production [122, 124]. Var- studies suggest that MAIT cells elaborate a protective ef-
ious studies have shown an important, nonredundant fect against bacterial infection via multiple mechanisms,
role for IL-12, IL-18, and IL-2 signaling in MAIT cell including macrophage activation and monocyte-to-den-
activation [114, 116, 125–127], and a significant reduc- dritic cell differentiation, and bridging of innate and
tion of IL-12 and/or IL-2 receptors in in vitro-stimulated adaptive immunity.
blood MAIT cells from TB patients [124, 127]. Finally, 2
reports suggested that functionally deficient MAIT cells Lung MAIT Cells in the Context of Viral Infection
from the blood of TB patients and HIV/TB coinfected Given the identification of bacterial and fungal vita-
patients have elevated levels of cell surface PD-1 expres- min B metabolites as MR1-presented antigens [107–110],
sion relative to healthy controls [128, 129], suggesting MAIT cells were thought not to play a major role in viral
that immune checkpoint pathways may functionally infection. However, a recent study found higher MAIT
regulate these cells during infection. Collectively, these cell numbers in H7N9 influenza patients who recovered
studies suggest that MAIT cells are functionally impor- from infection relative to those who succumbed to the

8 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
infection [126]. Utilizing an in vitro coculture system of Chitinases in Lung Pathologies
influenza-infected airway epithelial cells (A549) with hu- Elevated levels of chitinase activity has been observed
man peripheral blood mononuclear cells, an MR1-inde- in lysosomal storage disease such as Gaucher disease, as
pendent, CD14+ cell- and IL-18-dependent activation of well as in inflammatory lung diseases such as COPD,
MAIT cells was observed, featuring increased intracellu- asthma, sarcoidosis, and CF [141–144]. Acidic mam­
lar IFN-γ and granzyme B levels [126]. These results were malian chitinase (AMCase/Chit2), 1 of 2 true chitinases
later confirmed in another study in which MAIT cells present in mammals, was shown to mediate type 2 in-
were found to be activated in an MR1-independent but flammation and pathology in a mouse model of asthma
IL-18-, IL-12-, and IL-15-dependent manner in response [144]. A similar role was observed for chitotriosidase
to various viral infections, including dengue, hepatitis C, (Chit1) during fungal lung infection, where cleavage of
and influenza viruses [134]. Together these studies sug- fungal chitin by chitotriosidase mediates pathological re-
gest that MAIT cells may play an important role in host sponses [145]. In both asthma and fungal infection mod-
responses to viral infections. els, better outcomes and survival were observed in mice
lacking AMCase and Chit1 [144, 145]. To put this into a
human perspective, a substantial portion of the popula-
Novel Mediators: CLP tion (3–20%) lacks true chitinase activity due to a 24-bp
mutation in the Chit1 gene, the major contributor of chi-
In recent years, several novel mediators have been tinase activity in humans, suggesting a nonessential but
identified that participate in innate immunity in the nevertheless potentially harmful role during pathological
lungs. These include chitinase and CLP, a conserved challenges [146–148].
group of proteins that belong to the 18-glycosyl hydrolase
family [135]. CLP are well conserved but with high diver- CLP in Lung Pathologies
gence in mammals (represented by YKL-39 and YKL-40 Chil1 (BRP-39 in mice and YKL 40 in humans) is one
in humans vs. BRP-39, Ym1, and Ym2 in mice), suggest- of the most prevalent CLP, with a high expression in leu-
ing important but potentially variable roles in hosts [135]. kocytes including macrophages and neutrophils suggest-
The divergence in CLP among mammals has been attrib- ing immune-specific roles. Indeed, studies focusing on
uted to the difference in microbial threats faced by indi- bacterial lung infection indicated an important role for
vidual species [135]. Chil1 in inflammasome regulation in vivo and in vitro
[149, 150]. The absence of BRP-39 in mice during lung
Chitin, Chitinases, and CLP infection with gram-positive Streptococcus or gram-neg-
Although mammals do not synthesize chitins, the ative Pseudomonas leads to exaggerated inflammasome
presence of chitinases and CLP suggests a potential role activation. BRP-39 limits macrophage pyroptosis during
in the digestion of chitin-containing food and/or protec- bacterial infection to give an advantage to the host by lim-
tion against chitin-containing pathogens. This hypothe- iting bacterial growth and exuberant inflammation lead-
sis was based in part on epidemiological evidence show- ing to lung injury, thereby improving survival [149, 150].
ing increased expression of these proteins in populations BRP-39 has also been shown to bind IL-13 receptor α,
exposed to higher levels of chitin-containing pathogens relying on the protein TMEM to exert its effect [151, 152].
and food products [136, 137]. However, later studies cast Other examples of CLP in mice include Ym1 and Ym2
doubts on this hypothesis, indicating a limited correla- [135]. These 2 proteins are highly expressed in the lung,
tion between Chit1 deficiency and exposure to chitin- and their expression is increased during the induction of
containing parasites and food [138–140]. CLP have a high type 2 inflammation such as in nematode infection and
binding capacity to chitin but lack enzymatic activity to the HDM allergen model of asthma [153, 154]. Ym1 over-
cleave chitin, which suggests a limited direct role in the expression leads to an increase in lung neutrophilia, and
protection against chitin-containing pathogens or in the blocking Ym1 using monoclonal antibody results in a de-
digestion of chitin-containing food. In keeping with their creased neutrophil accumulation upon HDM challenge
conserved presence, recent advances have shown that in mouse lungs. γδ T cells are the major target of CLP,
CLP as well as chitinases play important roles in immune- responding to the overexpression of CLP with an in-
related pathophysiology. creased production of IL-17A [153]. Overall, chitinases
and CLP are associated with many inflammatory diseas-
es, and their causal role in various diseases has been es-

Innate Immunity of the Lung J Innate Immun 9


DOI: 10.1159/000487057
tablished using mouse models. These proteins might pro- severe tissue injury by degrading extracellular matrix
vide novel diagnostic and therapeutic targets to under- proteins, such as elastin, thereby remodeling the fragile
stand, treat, and monitor therapeutic efficacy in various pulmonary architecture. Temporally, innate immune ef-
inflammatory and infectious diseases of the respiratory fector responses are mostly protective in the initial phase
tract and beyond. of infection and inflammation, but they can become
harmful and induce autoinflammatory complications if
they fail to resolve, as exemplified in chronic lung dis-
Summary and Translational Implications eases such as CF, IPF, and COPD [42]. From a cellular
perspective, pulmonary innate immune cells can be fur-
The multidimensional nature of pulmonary innate ther subdivided into 2 simplified categories: first, granu-
immunity can be viewed from 3 vantage points: (1) cel- locytes (neutrophils, eosinophils, and basophils) that are
lular [155] versus noncellular components, (2) protective short-lived and bear a higher acute pathogenic potential
versus harmful mechanisms, and (3) translational disease by rapidly releasing their toxic and tissue-damaging com-
relevance for biomarker development and therapeutic ponents (proteases and oxidants) upon local activation in
targeting. airways [8, 46], and, second, mononuclear cells (mono-
cytes, alveolar/interstitial macrophages, and dendritic
Cellular versus Noncellular Components cells) that are generally longer lived and more controlled
Specific to the pulmonary compartment is the muco- in terms of enzyme and mediator release, mainly serving
ciliary escalator as a physical innate host defense barrier as antigen-presenting cells and scavengers of apoptotic
in conjunction with airway epithelial cell-derived innate bodies/debris and invading microbes. Short-lived innate
effector proteins (prototypically antimicrobial proteins, immune cells play a major role in the early phase of acute
such as defensins and collectins). Among innate immune respiratory conditions, such as neutrophils in lung infec-
cells, AM represent a potent phagocytic immune cell sen- tions or acute respiratory distress syndrome, whereas
tinel in the lower airway compartment equipped with a long-lived innate immune cells, such as macrophages and
broad cellular armamentarium to protect the airspace ILC, orchestrate chronic outcomes of tissue inflamma-
from microbial and nonmicrobial (dust and cigarette tion and remodeling in chronic pulmonary conditions
smoke) airborne exposures and serving as a rheostat like lung fibrosis/IPF and COPD. Notably, different mac-
maintaining alveolar homeostasis. While AM dominate rophage subtypes have been proposed to differentially af-
in the lower/alveolar space, neutrophils are often found fect pulmonary disease outcomes in asthma, COPD, IPF,
to accumulate in the more proximal/bronchial airway and CF.
compartments. Lymphocytes, in turn, are mainly found
in the lung tissue/parenchyma [10], while they are scarce Translational Disease Relevance for Biomarker
in the bronchoalveolar space, which could be due in part Development and Therapeutic Targeting
to suppressive neutrophil-T-cell interactions [49]. Newly Therapeutic approaches targeting innate immune
identified innate immune cell types, such as myeloid-de- cells and particularly innate immune cell-derived prote-
rived suppressor cells, MAIT cells, and ILC add to the ases appear reasonable given the excess of these enzymes
emerging complexity and layering of innate host defense in the pulmonary microenvironment (particularly in CF
shields in the pulmonary mucosal environment. While and COPD). However, clinical studies have been so far
our understanding of the regulatory and pathophysiolog- limited either by safety (for small molecule approaches
ical roles of these subsets is increasing in murine model targeting MMP), or by efficacy (for supplementation of
systems, their relevance for human pathologies remains antiproteases, such as α-1 antitrypsin) [156, 157]. Thera-
poorly defined. peutic strategies targeting rarer innate immune cell sub-
populations, such as ILC or MAIT cells, are hampered
Protective versus Harmful Activities by: (1) the incomplete understanding of their protective
The activities of innate immune cells and proteins versus harmful potential in human disease and (2) the
mainly depend on the spatiotemporal context. For exam- current lack of knowledge of how to target these cell types
ple, intracellular enzymes such as elastase or matrix me- specifically without affecting other conventional lym-
talloproteases provide protection by degrading phagocy- phocyte subsets. Recently, novel innate immune media-
tosed microbial proteins, yet the very same enzymes liber- tors have emerged as potential biomarkers and/or drug
ated into the extracellular microenvironment can cause targets for respiratory diseases. For example, alarmins/

10 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
DAMP (such as S100 proteins, ATP, or HMGB1) play an (upper vs. lower airways, bronchoalveolar space vs. lung
inducing role in lung immunity by binding to PRR like parenchyma), mucosal barrier interactions with mi-
TLR4 and RAGE. Targeting these innate inflammation- crobes, and spatiotemporal dynamics of infiltrating (e.g.,
amplifying loops while leaving bacterial sensing intact is neutrophils) and resident (e.g., AM or ILC) immune cells.
an exciting challenge to tackle in future drug develop- The key challenge for biomarker development and thera-
ment approaches. Examples for current pharmacothera- peutic success will be to define beneficial versus harmful
peutic strategies targeting innate immune components innate immune subsets across species and disease condi-
in the respiratory space include: (1) monoclonal antibod- tions and to identify ways to selectively and safely target
ies against innate immune cytokines (such as thymic these subsets or their released mediators in order to pre-
stromal lymphopoietin [158, 159] or IL-33/ST2), (2) vent, attenuate, or resolve pulmonary pathologies.
small molecule inhibitors targeting innate immune path-
ways or innate immune cell recruitment (such as CXCR2
antagonists [160–162] to dampen neutrophil recruit- Acknowledgements
ment), and (3) even newer technologies such as siRNA This work was supported by funds from the DFG CRC/SFB685
knockdown, microRNA, mRNA supplementation, or to D.H. (University of Tübingen, Germany).
CRISPR/Cas9. A common challenge for these different
therapeutic modalities is delivery into the lower airways,
crossing the mucus barrier, and efficient uptake by pul- Disclosure Statement
monary target cells. D.H. works for a pharmaceutical company (Roche Pharma Re-
Collectively, the innate immunity of the lung is multi- search and Early Development, Roche Innovation Center, Basel,
faceted and multilayered with anatomical complexity Switzerland).

References
  1 Hiemstra PS, McCray PB Jr, Bals R: The in- 10 Regamey N, Tsartsali L, Hilliard TN, Fuchs O, 16 Page K, Ledford JR, Zhou P, Dienger K, Wills-
nate immune function of airway epithelial Tan HL, Zhu J, Qiu YS, Alton EW, Jeffery PK, Karp M: Mucosal sensitization to German
cells in inflammatory lung disease. Eur Respir Bush A, Davies JC: Distinct patterns of in- cockroach involves protease-activated recep-
J 2015;45:1150–1162. flammation in the airway lumen and bronchi- tor-2. Respir Res 2010;11:62.
 2 Hasenberg M, Stegemann-Koniszewski S, al mucosa of children with cystic fibrosis. 17 Post S, Heijink IH, Petersen AH, de Bruin
Gunzer M: Cellular immune reactions in the Thorax 2012;67:164–170. HG, van Oosterhout AJ, Nawijn MC: Prote-
lung. Immunol Rev 2013;251:189–214. 11 Pabst R: Is BALT a major component of the ase-activated receptor-2 activation contrib-
  3 Chaudhuri N, Sabroe I: Basic science of the human lung immune system? Immunol To- utes to house dust mite-induced IgE respons-
innate immune system and the lung. Paediatr day 1992;13:119–122. es in mice. PLoS One 2014;9:e91206.
Respir Rev 2008;9:236–242. 12 Tengroth L, Millrud CR, Kvarnhammar AM, 18 Kool M, Willart MA, van Nimwegen M, Ber-
  4 Sabroe I, Lloyd CM, Whyte MK, Dower SK, Kumlien Georen S, Latif L, Cardell LO: Func- gen I, Pouliot P, Virchow JC, Rogers N, Oso-
Williams TJ, Pease JE: Chemokines, innate tional effects of toll-like receptor (TLR)3, 7, 9, rio F, Reis e Sousa C, Hammad H, Lambrecht
and adaptive immunity, and respiratory dis- RIG-I and MDA-5 stimulation in nasal epi- BN: An unexpected role for uric acid as an
ease. Eur Respir J 2002;19:350–355. thelial cells. PLoS One 2014;9:e98239. inducer of T helper 2 cell immunity to inhaled
 5 Panina-Bordignon P, D’Ambrosio D: Che- 13 Kim HJ, Kim CH, Kim MJ, Ryu JH, Seong SY, antigens and inflammatory mediator of aller-
mokines and their receptors in asthma and Kim S, Lim SJ, Holtzman MJ, Yoon JH: The gic asthma. Immunity 2011;34:527–540.
chronic obstructive pulmonary disease. Curr induction of pattern-recognition receptor ex- 19 Lambrecht BN, Hammad H: The airway epi-
Opin Pulm Med 2003;9:104–110. pression against influenza a virus through thelium in asthma. Nat Med 2012; 18: 684–
  6 Owen C: Chemokine receptors in airway dis- Duox2-derived reactive oxygen species in na- 692.
ease: which receptors to target? Pulm Phar- sal mucosa. Am J Respir Cell Mol Biol 2015; 20 Bogefors J, Rydberg C, Uddman R, Fransson
macol Ther 2001;14:193–202. 53:525–535. M, Mansson A, Benson M, Adner M, Cardell
  7 Gerard C, Rollins BJ: Chemokines and dis- 14 Kim JH, Kim YS, Cho GS, Kim NH, Gong CH, LO: Nod1, Nod2 and Nalp3 receptors, new
ease. Nat Immunol 2001;2:108–115. Lee BJ, Jang YJ: Human rhinovirus-induced potential targets in treatment of allergic rhini-
  8 Kruger P, Saffarzadeh M, Weber AN, Rieber proinflammatory cytokine and interferon-be- tis? Allergy 2010;65:1222–1226.
N, Radsak M, von Bernuth H, Benarafa C, ta responses in nasal epithelial cells from 21 Tabeling C, Scheer H, Schonrock SM, Runge
Roos D, Skokowa J, Hartl D: Neutrophils: be- chronic rhinosinusitis patients. Allergy Asth- F, Gutbier B, Lienau J, Hamelmann E, Opitz
tween host defence, immune modulation, and ma Immunol Res 2015;7:489–496. B, Suttorp N, Mayer K, Behrens GM,
tissue injury. Plos Pathog 2015;11:e1004651. 15 Nathan AT, Peterson EA, Chakir J, Wills- Tschernig T, Witzenrath M: Nucleotide
  9 Tang AC, Turvey SE, Alves MP, Regamey N, Karp M: Innate immune responses of airway oligomerization domain 1 ligation suppressed
Tummler B, Hartl D: Current concepts: host- epithelium to house dust mite are mediated murine allergen-specific T-cell proliferation
pathogen interactions in cystic fibrosis airways through beta-glucan-dependent pathways. J and airway hyperresponsiveness. Am J Respir
disease. Eur Respir Rev 2014;23:320–332. Allergy Clin Immunol 2009;123:612–618. Cell Mol Biol 2014;50:903–911.

Innate Immunity of the Lung J Innate Immun 11


DOI: 10.1159/000487057
22 Hirota JA, Gold MJ, Hiebert PR, Parkinson in vitro with radioactive diisopropylfluoro- shifts in nutrient transporter expression. J Im-
LG, Wee T, Smith D, Hansbro PM, Carlsten phosphate (DFP). J Clin Invest 1960;39:1481– munol 2013;190:6043–6050.
C, VanEeden S, Sin DD, McNagny KM, 1486. 46 Ralhan A, Laval J, Lelis F, Ballbach M, Grund
Knight DA: The nucleotide-binding domain, 34 Zhang P, Summer WR, Bagby GJ, Nelson S: C, Hector A, Hartl D: Current concepts and
leucine-rich repeat protein 3 inflammasome/ Innate immunity and pulmonary host de- controversies in innate immunity of cystic fi-
IL-1 receptor I axis mediates innate, but not fense. Immunol Rev 2000;173:39–51. brosis lung disease. J Innate Immun 2016; 8:
adaptive, immune responses after exposure to 35 Summers C, Chilvers ER, Peters AM: Math- 531–540.
particulate matter under 10 μm. Am J Respir ematical modeling supports the presence of 47 Laval J, Ralhan A, Hartl D: Neutrophils in cys-
Cell Mol Biol 2015;52:96–105. neutrophil depriming in vivo. Physiol Rep tic fibrosis. Biol Chem 2016;397:485–496.
23 Unger BL, Ganesan S, Comstock AT, Faris 2014;2:e00241. 48 Hartl D, Latzin P, Hordijk P, Marcos V, Ru-
AN, Hershenson MB, Sajjan US: Nod-like re- 36 Summers C, Singh NR, White JF, Mackenzie dolph C, Woischnik M, Krauss-Etschmann S,
ceptor X-1 is required for rhinovirus-induced IM, Johnston A, Solanki C, Balan KK, Peters Koller B, Reinhardt D, Roscher AA, Roos D,
barrier dysfunction in airway epithelial cells. AM, Chilvers ER: Pulmonary retention of Griese M: Cleavage of CXCR1 on neutrophils
J Virol 2014;88:3705–3718. primed neutrophils: a novel protective host disables bacterial killing in cystic fibrosis lung
24 Lee RJ, Cohen NA: Bitter and sweet taste recep- response, which is impaired in the acute respi- disease. Nat Med 2007;13:1423–1430.
tors in the respiratory epithelium in health and ratory distress syndrome. Thorax 2014; 69: 49 Ingersoll SA, Laval J, Forrest OA, Preininger
disease. J Mol Med (Berl) 2014;92:1235–1244. 623–629. M, Brown MR, Arafat D, Gibson G, Tangpri-
25 Barham HP, Cooper SE, Anderson CB, Tiz­ 37 Scapini P, Marini O, Tecchio C, Cassatella cha V, Tirouvanziam R: Mature cystic fibrosis
zano M, Kingdom TT, Finger TE, Kinnamon MA: Human neutrophils in the saga of cellu- airway neutrophils suppress T cell function:
SC, Ramakrishnan VR: Solitary chemosenso- lar heterogeneity: insights and open ques- evidence for a role of arginase 1 but not pro-
ry cells and bitter taste receptor signaling in tions. Immunol Rev 2016;273:48–60. grammed death-ligand 1. J Immunol 2015;
human sinonasal mucosa. Int Forum Allergy 38 Williams AE, Chambers RC: The mercurial 194:5520–5528.
Rhinol 2013;3:450–457. nature of neutrophils: still an enigma in 50 Hampton HR, Bailey J, Tomura M, Brink R,
26 Shah AS, Ben-Shahar Y, Moninger TO, Kline ARDS? Am J Physiol Lung Cell Mol Physiol Chtanova T: Microbe-dependent lymphatic
JN, Welsh MJ: Motile cilia of human airway 2014;306:L217–L230. migration of neutrophils modulates lympho-
epithelia are chemosensory. Science 2009; 39 Fortunati E, Kazemier KM, Grutters JC, cyte proliferation in lymph nodes. Nat Com-
325:1131–1134. Koenderman L, Van den Bosch VJ: Human mun 2015;6:7139.
27 Lee RJ, Cohen NA: The emerging role of the neutrophils switch to an activated phenotype 51 Duffy D, Perrin H, Abadie V, Benhabiles N,
bitter taste receptor T2R38 in upper respira- after homing to the lung irrespective of in- Boissonnas A, Liard C, Descours B, Reboul-
tory infection and chronic rhinosinusitis. Am flammatory disease. Clin Exp Immunol 2009; leau D, Bonduelle O, Verrier B, Van Rooijen
J Rhinol Allergy 2013;27:283–286. 155:559–566. N, Combadiere C, Combadiere B: Neutro-
28 Lee RJ, Kofonow JM, Rosen PL, Siebert AP, 40 Hartl D, Krauss-Etschmann S, Koller B, phils transport antigen from the dermis to the
Chen B, Doghramji L, Xiong G, Adappa ND, Hordijk PL, Kuijpers TW, Hoffmann F, Hec- bone marrow, initiating a source of memory
Palmer JN, Kennedy DW, Kreindler JL, Mar- tor A, Eber E, Marcos V, Bittmann I, Eickel- CD8+ T cells. Immunity 2012;37:917–929.
golskee RF, Cohen NA: Bitter and sweet taste berg O, Griese M, Roos D: Infiltrated neutro- 52 Abadie V, Badell E, Douillard P, Ensergueix
receptors regulate human upper respiratory phils acquire novel chemokine receptor ex- D, Leenen PJ, Tanguy M, Fiette L, Saeland S,
innate immunity. J Clin Invest 2014; 124: pression and chemokine responsiveness in Gicquel B, Winter N: Neutrophils rapidly mi-
1393–1405. chronic inflammatory lung diseases. J Immu- grate via lymphatics after Mycobacterium bo-
29 Mirkovic B, Murray MA, Lavelle GM, Molloy nol 2008;181:8053–8067. vis BCG intradermal vaccination and shuttle
K, Azim AA, Gunaratnam C, Healy F, Slattery 41 Koller B, Kappler M, Latzin P, Gaggar A, live bacilli to the draining lymph nodes. Blood
D, McNally P, Hatch J, Wolfgang M, Tunney Schreiner M, Takyar S, Kormann M, Kabesch 2005;106:1843–1850.
MM, Muhlebach MS, Devery R, Greene CM, M, Roos D, Griese M, Hartl D: TLR expres- 53 Coombes JL, Charsar BA, Han SJ, Halkias J,
McElvaney NG: The role of short-chain fatty sion on neutrophils at the pulmonary site of Chan SW, Koshy AA, Striepen B, Robey EA:
acids, produced by anaerobic bacteria, in the infection: TLR1/TLR2-mediated up-regula- Motile invaded neutrophils in the small intes-
cystic fibrosis airway. Am J Respir Crit Care tion of TLR5 expression in cystic fibrosis lung tine of Toxoplasma gondii-infected mice
Med 2015;192:1314–1324. disease. J Immunol 2008;181:2753–2763. reveal a potential mechanism for para-
30 Chirkova T, Lin S, Oomens AG, Gaston KA, 42 Koller B, Bals R, Roos D, Korting HC, Griese site spread. Proc Natl Acad Sci USA 2013;
Boyoglu-Barnum S, Meng J, Stobart CC, Cot- M, Hartl D: Innate immune receptors on neu- 110:E1913–E1922.
ton CU, Hartert TV, Moore ML, Ziady AG, trophils and their role in chronic lung disease. 54 Brinkmann V, Zychlinsky A: Beneficial sui-
Anderson LJ: CX3CR1 is an important sur- Eur J Clin Invest 2009;39:535–547. cide: why neutrophils die to make nets. Nat
face molecule for respiratory syncytial virus 43 Tirouvanziam R, Gernez Y, Conrad CK, Moss Rev Microbiol 2007;5:577–582.
infection in human airway epithelial cells. J RB, Schrijver I, Dunn CE, Davies ZA, Herzen- 55 Brinkmann V, Reichard U, Goosmann C, Fau­
Gen Virol 2015;96:2543–2556. berg LA: Profound functional and signaling ler B, Uhlemann Y, Weiss DS, Weinrauch Y,
31 Kotha PL, Sharma P, Kolawole AO, Yan R, changes in viable inflammatory neutrophils Zychlinsky A: Neutrophil extracellular traps
Alghamri MS, Brockman TL, Gomez-Cam- homing to cystic fibrosis airways. Proc Natl kill bacteria. Science 2004;303:1532–1535.
bronero J, Excoffon KJ: Adenovirus entry Acad Sci USA 2008;105:4335–4339. 56 Cortjens B, van Woensel JB, Bem RA: Neutro-
from the apical surface of polarized epithelia 44 Makam M, Diaz D, Laval J, Gernez Y, Conrad phil extracellular traps in respiratory disease:
is facilitated by the host innate immune re- CK, Dunn CE, Davies ZA, Moss RB, Herzen- guided anti-microbial traps or toxic webs?
sponse. PLoS Pathog 2015;11:e1004696. berg LA, Herzenberg LA, Tirouvanziam R: Paediatr Respir Rev 2017;21:54–61.
32 Pillay J, den Braber I, Vrisekoop N, Kwast LM, Activation of critical, host-induced, metabol- 57 Cheng OZ, Palaniyar N: Net balancing: a
de Boer RJ, Borghans JA, Tesselaar K, Koen- ic and stress pathways marks neutrophil entry problem in inflammatory lung diseases. Front
derman L: In vivo labeling with 2H2O reveals into cystic fibrosis lungs. Proc Natl Acad Sci Immunol 2013;4:1.
a human neutrophil lifespan of 5.4 days. USA 2009;106:5779–5783. 58 Yipp BG, Kubes P: Netosis: how vital is it?
Blood 2010;116:625–627. 45 Laval J, Touhami J, Herzenberg LA, Conrad Blood 2013;122:2784–2794.
33 Mauer AM, Athens JW, Ashenbrucker H, C, Taylor N, Battini JL, Sitbon M, Tirouvan- 59 Sorensen OE, Borregaard N: Neutrophil ex-
Cartwright GE, Wintrobe MM: Leukokinetic ziam R: Metabolic adaptation of neutrophils tracellular traps: the dark side of neutrophils.
studies. 2. A method for labeling granulocytes in cystic fibrosis airways involves distinct J Clin Invest 2016;126:1612–1620.

12 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
60 Grabcanovic-Musija F, Obermayer A, Stoiber Rodewald HR: Tissue-resident macrophages kens W, Erjefalt JS, Spits H, Ros XR: IL-1beta,
W, Krautgartner WD, Steinbacher P, Winter- originate from yolk-sac-derived erythro-my- IL-4 and IL-12 control the fate of group 2 in-
berg N, Bathke AC, Klappacher M, Studnicka eloid progenitors. Nature 2015;518:547–551. nate lymphoid cells in human airway inflam-
M: Neutrophil extracellular trap (NET) for- 75 Guth AM, Janssen WJ, Bosio CM, Crouch EC, mation in the lungs. Nat Immunol 2016; 17:
mation characterises stable and exacerbated Henson PM, Dow SW: Lung environment de- 636–645.
COPD and correlates with airflow limitation. termines unique phenotype of alveolar mac- 88 Wallrapp A, Riesenfeld SJ, Burkett PR, Ab-
Respir Res 2015;16:59. rophages. Am J Physiol Lung Cell Mol Physi- dulnour RE, Nyman J, Dionne D, Hofree M,
61 Wright TK, Gibson PG, Simpson JL, McDon- ol 2009;296:L936–L946. Cuoco MS, Rodman C, Farouq D, Haas BJ,
ald VM, Wood LG, Baines KJ: Neutrophil ex- 76 Sica A, Mantovani A: Macrophage plasticity Tickle TL, Trombetta JJ, Baral P, Klose CSN,
tracellular traps are associated with inflam- and polarization: in vivo veritas. J Clin Invest Mahlakoiv T, Artis D, Rozenblatt-Rosen O,
mation in chronic airway disease. Respirology 2012;122:787–795. Chiu IM, Levy BD, Kowalczyk MS, Regev A,
2016;21:467–475. 77 Mantovani A, Biswas SK, Galdiero MR, Sica Kuchroo VK: The neuropeptide NMU ampli-
62 Cortjens B, de Boer OJ, de Jong R, Antonis AF, A, Locati M: Macrophage plasticity and polar- fies ILC2-driven allergic lung inflammation.
Sabogal Pineros YS, Lutter R, van Woensel JB, ization in tissue repair and remodelling. J Nature 2017;549:351–356.
Bem RA: Neutrophil extracellular traps cause Pathol 2013;229:176–185. 89 Bernink JH, Germar K, Spits H: The role of
airway obstruction during respiratory syncy- 78 Murray PJ, Allen JE, Biswas SK, Fisher EA, ILC2 in pathology of type 2 inflammatory dis-
tial virus disease. J Pathol 2016;238:401–411. Gilroy DW, Goerdt S, Gordon S, Hamilton eases. Curr Opin Immunol 2014;31:115–120.
63 Gallin JI: Human neutrophil heterogeneity JA, Ivashkiv LB, Lawrence T, Locati M, Man- 90 Jia Y, Fang X, Zhu X, Bai C, Zhu L, Jin M,
exists, but is it meaningful? Blood 1984; 63: tovani A, Martinez FO, Mege JL, Mosser DM, Wang X, Hu M, Tang R, Chen Z: IL-13+ type
977–983. Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica 2 innate lymphoid cells correlate with asthma
64 Garbi N, Lambrecht BN: Location, function, A, Suttles J, Udalova I, van Ginderachter JA, control status and treatment response. Am J
and ontogeny of pulmonary macrophages Vogel SN, Wynn TA: Macrophage activation Respir Cell Mol Biol 2016;55:675–683.
during the steady state. Pflugers Arch 2017; and polarization: nomenclature and experi- 91 Kearley J, Silver JS, Sanden C, Liu Z, Berlin
469:561–572. mental guidelines. Immunity 2014;41:14–20. AA, White N, Mori M, Pham TH, Ward CK,
65 Hussell T, Bell TJ: Alveolar macrophages: 79 Kolahian S, Fernandez IE, Eickelberg O, Hartl Criner GJ, Marchetti N, Mustelin T, Erjefalt
plasticity in a tissue-specific context. Nat Rev D: Immune mechanisms in pulmonary fibro- JS, Kolbeck R, Humbles AA: Cigarette smoke
Immunol 2014;14:81–93. sis. Am J Respir Cell Mol Biol 2016;55:309–322. silences innate lymphoid cell function and fa-
66 Fels AO, Cohn ZA: The alveolar macrophage. 80 Bruscia EM, Bonfield TL: Cystic fibrosis lung cilitates an exacerbated type I interleukin-
J Appl Physiol (1985) 1986;60:353–369. immunity: the role of the macrophage. J In- 33-dependent response to infection. Immu-
67 du Bois RM: The alveolar macrophage. Tho- nate Immun 2016;8:550–563. nity 2015;42:566–579.
rax 1985;40:321–327. 81 Spits H, Cupedo T: Innate lymphoid cells: 92 Gronke K, Kofoed-Nielsen M, Diefenbach A:
68 Trapnell BC, Carey BC, Uchida K, Suzuki T: emerging insights in development, lineage re- Innate lymphoid cells, precursors and plastic-
Pulmonary alveolar proteinosis, a primary lationships, and function. Annu Rev Immu- ity. Immunol Lett 2016;179:9–18.
immunodeficiency of impaired GM-CSF nol 2012;30:647–675. 93 Mebius RE: Organogenesis of lymphoid tis-
stimulation of macrophages. Curr Opin Im- 82 Sonnenberg GF, Artis D: Innate lymphoid sues. Nat Rev Immunol 2003;3:292–303.
munol 2009;21:514–521. cells in the initiation, regulation and resolu- 94 John-Schuster G, Hager K, Conlon TM, Irm-
69 Trapnell BC, Whitsett JA, Nakata K: Pulmo- tion of inflammation. Nat Med 2015;21:698– ler M, Beckers J, Eickelberg O, Yildirim AO:
nary alveolar proteinosis. N Engl J Med 2003; 708. Cigarette smoke-induced iBALT mediates
349:2527–2539. 83 Spits H, Artis D, Colonna M, Diefenbach A, macrophage activation in a B cell-dependent
70 Soroosh P, Doherty TA, Duan W, Mehta AK, Di Santo JP, Eberl G, Koyasu S, Locksley RM, manner in COPD. Am J Physiol Lung Cell
Choi H, Adams YF, Mikulski Z, Khorram N, McKenzie AN, Mebius RE, Powrie F, Vivier Mol Physiol 2014;307:L692–L706.
Rosenthal P, Broide DH, Croft M: Lung-resi- E: Innate lymphoid cells: a proposal for uni- 95 Hams E, Bermingham R, Fallon PG: Macro-
dent tissue macrophages generate Foxp3+ form nomenclature. Nat Rev Immunol 2013; phage and innate lymphoid cell interplay in the
regulatory T cells and promote airway toler- 13:145–149. genesis of fibrosis. Front Immunol 2015;6:597.
ance. J Exp Med 2013;210:775–788. 84 Guo L, Junttila IS, Paul WE: Cytokine-in- 96 Tan HL, Rosenthal M: IL-17 in lung disease:
71 Westphalen K, Gusarova GA, Islam MN, Sub- duced cytokine production by conventional friend or foe? Thorax 2013;68:788–790.
ramanian M, Cohen TS, Prince AS, Bhat- and innate lymphoid cells. Trends Immunol 97 Chan YR, Chen K, Duncan SR, Lathrop KL,
tacharya J: Sessile alveolar macrophages com- 2012;33:598–606. Latoche JD, Logar AJ, Pociask DA, Wahlberg
municate with alveolar epithelium to modu- 85 Klose CSN, Flach M, Mohle L, Rogell L, Hoy- BJ, Ray P, Ray A, Pilewski JM, Kolls JK: Pa-
late immunity. Nature 2014;506:503–506. ler T, Ebert K, Fabiunke C, Pfeifer D, Sexl V, tients with cystic fibrosis have inducible IL-
72 van de Laar L, Saelens W, De Prijck S, Martens Fonseca-Pereira D, Domingues RG, Veiga- 17+IL-22+ memory cells in lung draining
L, Scott CL, Van Isterdael G, Hoffmann E, Fernandes H, Arnold SJ, Busslinger M, Dunay lymph nodes. J Allergy Clin Immunol 2013;
Beyaert R, Saeys Y, Lambrecht BN, Guilliams IR, Tanriver Y, Diefenbach A: Differentiation 131:1117–1129, 1129.e1111–1129.e1115.
M: Yolk sac macrophages, fetal liver, and of type 1 ILCs from a common progenitor to 98 Tan HL, Regamey N, Brown S, Bush A, Lloyd
adult monocytes can colonize an empty niche all helper-like innate lymphoid cell lineages. CM, Davies JC: The Th17 pathway in cystic
and develop into functional tissue-resident Cell 2014;157:340–356. fibrosis lung disease. Am J Respir Crit Care
macrophages. Immunity 2016;44:755–768. 86 Silver JS, Kearley J, Copenhaver AM, Sanden Med 2011;184:252–258.
73 Guilliams M, De Kleer I, Henri S, Post S, Van- C, Mori M, Yu L, Pritchard GH, Berlin AA, 99 Moretti S, Renga G, Oikonomou V, Galosi C,
houtte L, De Prijck S, Deswarte K, Malissen B, Hunter CA, Bowler R, Erjefalt JS, Kolbeck R, Pariano M, Iannitti RG, Borghi M, Puccetti
Hammad H, Lambrecht BN: Alveolar macro- Humbles AA: Inflammatory triggers associ- M, De Zuani M, Pucillo CE, Paolicelli G, Zel-
phages develop from fetal monocytes that dif- ated with exacerbations of COPD orchestrate ante T, Renauld JC, Bereshchenko O, Sporto-
ferentiate into long-lived cells in the first week plasticity of group 2 innate lymphoid cells in letti P, Lucidi V, Russo MC, Colombo C, Fis-
of life via GM-CSF. J Exp Med 2013; 210: the lungs. Nat Immunol 2016;17:626–635. carelli E, Lass-Florl C, Majo F, Ricciotti G, El-
1977–1992. 87 Bal SM, Bernink JH, Nagasawa M, Groot J, lemunter H, Ratclif L, Talesa VN, Napolioni
74 Gomez Perdiguero E, Klapproth K, Schulz C, Shikhagaie MM, Golebski K, van Drunen V, Romani L: A mast cell-ILC2-Th9 pathway
Busch K, Azzoni E, Crozet L, Garner H, CM, Lutter R, Jonkers RE, Hombrink P, promotes lung inflammation in cystic fibro-
Trouillet C, de Bruijn MF, Geissmann F, Bruchard M, Villaudy J, Munneke JM, Fok- sis. Nat Commun 2017;8:14017.

Innate Immunity of the Lung J Innate Immun 13


DOI: 10.1159/000487057
100 Martin E, Treiner E, Duban L, Guerri L, Kostenko L, Reantragoon R, Williamson associated with airflow limitation in patients
Laude H, Toly C, Premel V, Devys A, Moura NA, Purcell AW, Dudek NL, McConville with asthma. Allergol Int 2017;66:302–309.
IC, Tilloy F, Cherif S, Vera G, Latour S, Sou- MJ, O’Hair RA, Khairallah GN, Godfrey DI, 121 Smith DJ, Hill GR, Bell SC, Reid DW: Re-
dais C, Lantz O: Stepwise development of Fairlie DP, Rossjohn J, McCluskey J: MR1 duced mucosal associated invariant T-cells
MAIT cells in mouse and human. PLoS Biol presents microbial vitamin B metabolites to are associated with increased disease severi-
2009;7:e54. MAIT cells. Nature 2012;491:717–723. ty and Pseudomonas aeruginosa infection in
101 Dusseaux M, Martin E, Serriari N, Peguillet 111 Gold MC, Cerri S, Smyk-Pearson S, Cansler cystic fibrosis. PLoS One 2014;9:e109891.
I, Premel V, Louis D, Milder M, Le Bourhis ME, Vogt TM, Delepine J, Winata E, 122 Kwon YS, Cho YN, Kim MJ, Jin HM, Jung
L, Soudais C, Treiner E, Lantz O: Human Swarbrick GM, Chua WJ, Yu YY, Lantz O, HJ, Kang JH, Park KJ, Kim TJ, Kee HJ, Kim
MAIT cells are xenobiotic-resistant, tissue- Cook MS, Null MD, Jacoby DB, Harriff MJ, N, Kee SJ, Park YW: Mucosal-associated in-
targeted, CD161hi IL-17-secreting T cells. Lewinsohn DA, Hansen TH, Lewinsohn variant T cells are numerically and function-
Blood 2011;117:1250–1259. DM: Human mucosal associated invariant T ally deficient in patients with mycobacterial
102 Treiner E, Duban L, Bahram S, Radosav- cells detect bacterially infected cells. PLoS infection and reflect disease activity. Tuber-
ljevic M, Wanner V, Tilloy F, Affaticati P, Biol 2010;8:e1000407. culosis (Edinb) 2015;95:267–274.
Gilfillan S, Lantz O: Selection of evolution- 112 Gibbs A, Leeansyah E, Introini A, Paquin- 123 Le Bourhis L, Martin E, Peguillet I, Guihot
arily conserved mucosal-associated invari- Proulx D, Hasselrot K, Andersson E, Bro- A, Froux N, Core M, Levy E, Dusseaux M,
ant T cells by MR1. Nature 2003; 422: 164– liden K, Sandberg JK, Tjernlund A: MAIT Meyssonnier V, Premel V, Ngo C, Riteau B,
169. cells reside in the female genital mucosa and Duban L, Robert D, Huang S, Rottman M,
103 Leeansyah E, Loh L, Nixon DF, Sandberg JK: are biased towards IL-17 and IL-22 produc- Soudais C, Lantz O: Antimicrobial activity
Acquisition of innate-like microbial reactiv- tion in response to bacterial stimulation. of mucosal-associated invariant T cells. Nat
ity in mucosal tissues during human fetal Mucosal Immunol 2017;10:35–45. Immunol 2010;11:701–708.
MAIT-cell development. Nat Commun 113 Gracey E, Qaiyum Z, Almaghlouth I, Law- 124 Jiang J, Yang B, An H, Wang X, Liu Y, Cao
2014;5:3143. son D, Karki S, Avvaru N, Zhang Z, Yao Y, Z, Zhai F, Wang R, Cao Y, Cheng X: Muco-
104 Hinks TS, Wallington JC, Williams AP, Dju- Ranganathan V, Baglaenko Y, Inman RD: sal-associated invariant T cells from patients
kanovic R, Staples KJ, Wilkinson TM: Ste- IL-7 primes IL-17 in mucosal-associated in- with tuberculosis exhibit impaired immune
roid-induced deficiency of mucosal-associ- variant T (MAIT) cells, which contribute to response. J Infect 2016;72:338–352.
ated invariant T cells in the chronic obstruc- the Th17-axis in ankylosing spondylitis. 125 Ussher JE, Bilton M, Attwod E, Shadwell J,
tive pulmonary disease lung: implications Ann Rheum Dis 2016;75:2124–2132. Richardson R, de Lara C, Mettke E, Kurioka
for nontypeable Haemophilus influenzae in- 114 Kurioka A, Ussher JE, Cosgrove C, Clough A, Hansen TH, Klenerman P, Willberg CB:
fection. Am J Respir Crit Care Med 2016; C, Fergusson JR, Smith K, Kang YH, Walker CD161++ CD8+ T cells, including the
194:1208–1218. LJ, Hansen TH, Willberg CB, Klenerman P: MAIT cell subset, are specifically activated
105 Porcelli S, Yockey CE, Brenner MB, Balk SP: MAIT cells are licensed through granzyme by IL-12+IL-18 in a TCR-independent man-
Analysis of T cell antigen receptor (TCR) ex- exchange to kill bacterially sensitized tar- ner. Eur J Immunol 2014;44:195–203.
pression by human peripheral blood CD4– gets. Mucosal Immunol 2015;8:429–440. 126 Loh L, Wang Z, Sant S, Koutsakos M, Jegas-
8-alpha/beta T cells demonstrates preferen- 115 Leeansyah E, Svard J, Dias J, Buggert M, kanda S, Corbett AJ, Liu L, Fairlie DP, Crowe
tial use of several V beta genes and an invari- Nystrom J, Quigley MF, Moll M, Sonner- J, Rossjohn J, Xu J, Doherty PC, McCluskey
ant TCR alpha chain. J Exp Med 1993; 178: borg A, Nowak P, Sandberg JK: Arming of J, Kedzierska K: Human mucosal-associated
1–16. MAIT cell cytolytic antimicrobial activity is invariant t cells contribute to antiviral influ-
106 Tilloy F, Treiner E, Park SH, Garcia C, Lem- induced by IL-7 and defective in HIV-1 in- enza immunity via IL-18-dependent activa-
onnier F, de la Salle H, Bendelac A, Bonnev- fection. PLoS Pathog 2015;11:e1005072. tion. Proc Natl Acad Sci USA 2016; 113:
ille M, Lantz O: An invariant T cell receptor 116 Sattler A, Dang-Heine C, Reinke P, Babel N: 10133–10138.
alpha chain defines a novel TAP-indepen- IL-15 dependent induction of IL-18 secre- 127 Jiang J, Chen X, An H, Yang B, Zhang F,
dent major histocompatibility complex class tion as a feedback mechanism controlling Cheng X: Enhanced immune response of
Ib-restricted alpha/beta T cell subpopula- human MAIT-cell effector functions. Eur J MAIT cells in tuberculous pleural effusions
tion in mammals. J Exp Med 1999; 189: Immunol 2015;45:2286–2298. depends on cytokine signaling. Sci Rep 2016;
1907–1921. 117 Gold MC, Napier RJ, Lewinsohn DM: MR1- 6:32320.
107 Mak JY, Xu W, Reid RC, Corbett AJ, Mee- restricted mucosal associated invariant T 128 Jiang J, Wang X, An H, Yang B, Cao Z, Liu Y,
han BS, Wang H, Chen Z, Rossjohn J, Mc- (MAIT) cells in the immune response to my- Su J, Zhai F, Wang R, Zhang G, Cheng X: Mu-
Cluskey J, Liu L, Fairlie DP: Stabilizing cobacterium tuberculosis. Immunol Rev cosal-associated invariant T-cell function is
short-lived Schiff base derivatives of 5-ami- 2015;264:154–166. modulated by programmed death-1 signaling
nouracils that activate mucosal-associated 118 Kwon YS, Jin HM, Cho YN, Kim MJ, Kang in patients with active tuberculosis. Am J
invariant T cells. Nat Commun 2017; 8: JH, Jung HJ, Park KJ, Kee HJ, Kee SJ, Park Respir Crit Care Med 2014;190:329–339.
14599. YW: Mucosal-associated invariant T cell de- 129 Saeidi A, Tien Tien VL, Al-Batran R, Al-
108 McWilliam HE, Birkinshaw RW, Villadan- ficiency in chronic obstructive pulmonary Darraji HA, Tan HY, Yong YK, Ponnampa-
gos JA, McCluskey J, Rossjohn J: MR1 pre- disease. COPD 2016;13:196–202. lavanar S, Barathan M, Rukumani DV, An-
sentation of vitamin B-based metabolite li- 119 Hinks TS, Zhou X, Staples KJ, Dimitrov BD, sari AW, Velu V, Kamarulzaman A, Larsson
gands. Curr Opin Immunol 2015;34:28–34. Manta A, Petrossian T, Lum PY, Smith CG, M, Shankar EM: Attrition of TCR Val-
109 Patel O, Kjer-Nielsen L, Le Nours J, Eckle SB, Ward JA, Howarth PH, Walls AF, Gadola pha7.2+ CD161++ MAIT cells in HIV-tu-
Birkinshaw R, Beddoe T, Corbett AJ, Liu L, SD, Djukanovic R: Innate and adaptive T berculosis co-infection is associated with el-
Miles JJ, Meehan B, Reantragoon R, Sando- cells in asthmatic patients: relationship to se- evated levels of PD-1 expression. PLoS One
val-Romero ML, Sullivan LC, Brooks AG, verity and disease mechanisms. J Allergy 2015;10:e0124659.
Chen Z, Fairlie DP, McCluskey J, Rossjohn Clin Immunol 2015;136:323–333. 130 Kawachi I, Maldonado J, Strader C, Gilfillan
J: Recognition of vitamin B metabolites by 120 Ishimori A, Harada N, Chiba A, Harada S, S: MR1-restricted V alpha 19i mucosal-asso-
mucosal-associated invariant T cells. Nat Matsuno K, Makino F, Ito J, Ohta S, Ono J, ciated invariant T cells are innate T cells in
Commun 2013;4:2142. Atsuta R, Izuhara K, Takahashi K, Miyake S: the gut lamina propria that provide a rapid
110 Kjer-Nielsen L, Patel O, Corbett AJ, Le Circulating activated innate lymphoid cells and diverse cytokine response. J Immunol
Nours J, Meehan B, Liu L, Bhati M, Chen Z, and mucosal-associated invariant T cells are 2006;176:1618–1627.

14 J Innate Immun Hartl et al.


DOI: 10.1159/000487057
131 Cui Y, Franciszkiewicz K, Mburu YK, Mon- 141 Hector A, Chotirmall SH, Lavelle GM, 151 Lee CM, He CH, Nour AM, Zhou Y, Ma B,
dot S, Le Bourhis L, Premel V, Martin E, Mirkovic B, Horan D, Eichler L, Mezger M, Park JW, Kim KH, Dela Cruz C, Sharma L,
Kachaner A, Duban L, Ingersoll MA, Rabot Singh A, Ralhan A, Berenbrinker S, Mack I, Nasr ML, Modis Y, Lee CG, Elias JA: Il-
S, Jaubert J, De Villartay JP, Soudais C, Lantz Ensenauer R, Riethmuller J, Graepler-Main- 13Ralpha2 uses TMEM219 in chitinase
O: Mucosal-associated invariant T cell-rich ka U, Murray MA, Griese M, McElvaney 3-like-1-induced signalling and effector re-
congenic mouse strain allows functional NG, Hartl D: Chitinase activation in patients sponses. Nat Commun 2016;7:12752.
evaluation. J Clin Invest 2015; 125: 4171– with fungus-associated cystic fibrosis lung 152 He CH, Lee CG, Dela Cruz CS, Lee CM,
4185. disease. J Allergy Clin Immunol 2016; 138: Zhou Y, Ahangari F, Ma B, Herzog EL,
132 Meierovics A, Yankelevich WJ, Cowley SC: 1183–1189.e4. Rosenberg SA, Li Y, Nour AM, Parikh CR,
MAIT cells are critical for optimal mucosal 142 Letuve S, Kozhich A, Humbles A, Brewah Y, Schmidt I, Modis Y, Cantley L, Elias JA: Chi-
immune responses during in vivo pulmo- Dombret MC, Grandsaigne M, Adle H, Kol- tinase 3-like 1 regulates cellular and tissue
nary bacterial infection. Proc Natl Acad Sci beck R, Aubier M, Coyle AJ, Pretolani M: responses via IL-13 receptor alpha2. Cell
USA 2013;110:E3119–E3128. Lung chitinolytic activity and chitotriosi- Rep 2013;4:830–841.
133 Meierovics AI, Cowley SC: MAIT cells pro- dase are elevated in chronic obstructive pul- 153 Sutherland TE, Logan N, Ruckerl D, Hum-
mote inflammatory monocyte differentia- monary disease and contribute to lung in- bles AA, Allan SM, Papayannopoulos V,
tion into dendritic cells during pulmonary flammation. Am J Pathol 2010;176:638–649. Stockinger B, Maizels RM, Allen JE: Chitin-
intracellular infection. J Exp Med 2016;213: 143 Gavala ML, Kelly EA, Esnault S, Kukreja S, ase-like proteins promote IL-17-mediated
2793–2809. Evans MD, Bertics PJ, Chupp GL, Jarjour neutrophilia in a tradeoff between nematode
134 van Wilgenburg B, Scherwitzl I, Hutchinson NN: Segmental allergen challenge enhances killing and host damage. Nat Immunol 2014;
EC, Leng T, Kurioka A, Kulicke C, de Lara chitinase activity and levels of CCL18 in 15:1116–1125.
C, Cole S, Vasanawathana S, Limpitikul W, mild atopic asthma. Clin Exp Allergy 2013; 154 Nair MG, Gallagher IJ, Taylor MD, Loke P,
Malasit P, Young D, Denney L, consortium 43:187–197. Coulson PS, Wilson RA, Maizels RM, Allen
S-H, Moore MD, Fabris P, Giordani MT, Oo 144 Zhu Z, Zheng T, Homer RJ, Kim YK, Chen JE: Chitinase and fizz family members are a
YH, Laidlaw SM, Dustin LB, Ho LP, Thomp- NY, Cohn L, Hamid Q, Elias JA: Acidic generalized feature of nematode infection
son FM, Ramamurthy N, Mongkolsapaya J, mammalian chitinase in asthmatic Th2 in- with selective upregulation of Ym1 and
Willberg CB, Screaton GR, Klenerman P: flammation and IL-13 pathway activation. Fizz1 by antigen-presenting cells. Infect Im-
MAIT cells are activated during human viral Science 2004;304:1678–1682. mun 2005;73:385–394.
infections. Nat Commun 2016;7:11653. 145 Wiesner DL, Specht CA, Lee CK, Smith KD, 155 Gasteiger G, D’Osualdo A, Schubert DA,
135 Bussink AP, Speijer D, Aerts JM, Boot RG: Mukaremera L, Lee ST, Lee CG, Elias JA, Weber A, Bruscia EM, Hartl D: Cellular in-
Evolution of mammalian chitinase(-like) Nielsen JN, Boulware DR, Bohjanen PR, Jen- nate immunity: an old game with new play-
members of family 18 glycosyl hydrolases. kins MK, Levitz SM, Nielsen K: Chitin rec- ers. J Innate Immun 2017;9:111–125.
Genetics 2007;177:959–970. ognition via chitotriosidase promotes 156 Gaggar A, Hector A, Bratcher PE, Mall MA,
136 Musumeci M, Malaguarnera L, Simpore J, pathologic type-2 helper T cell responses to Griese M, Hartl D: The role of matrix metal-
Barone R, Whalen M, Musumeci S: Chitot- cryptococcal infection. Plos Pathog 2015; loproteinases in cystic fibrosis lung disease.
riosidase activity in colostrum from African 11:e1004701. Eur Respir J 2011;38:721–727.
and Caucasian women. Clin Chem Lab Med 146 Kanneganti M, Kamba A, Mizoguchi E: Role 157 Griese M, Kappler M, Gaggar A, Hartl D: In-
2005;43:198–201. of chitotriosidase (chitinase 1) under normal hibition of airway proteases in cystic fibrosis
137 Malaguarnera L, Simpore J, Prodi DA, An- and disease conditions. J Epithel Biol Phar- lung disease. Eur Respir J 2008;32:783–795.
gius A, Sassu A, Persico I, Barone R, Musu- macol 2012;5:1–9. 158 Bel EH: Moving upstream: anti-TSLP in per-
meci S: A 24-bp duplication in exon 10 of 147 Piras I, Melis A, Ghiani ME, Falchi A, Lu- sistent uncontrolled asthma. N Engl J Med
human chitotriosidase gene from the sub- iselli D, Moral P, Varesi L, Calo CM, Vona 2017;377:989–991.
Saharan to the Mediterranean area: role of G: Human CHIT1 gene distribution: new 159 Corren J, Parnes JR, Wang L, Mo M, Roseti
parasitic diseases and environmental condi- data from Mediterranean and European SL, Griffiths JM, van der Merwe R: Tezepe-
tions. Genes Immun 2003;4:570–574. populations. J Hum Genet 2007;52:110–116. lumab in adults with uncontrolled asthma.
138 Manno N, Sherratt S, Boaretto F, Coico FM, 148 Boot RG, Renkema GH, Verhoek M, Strij- N Engl J Med 2017;377:936–946.
Camus CE, Campos CJ, Musumeci S, Bat- land A, Bliek J, de Meulemeester TM, Man- 160 De Soyza A, Pavord I, Elborn JS, Smith D,
tisti A, Quinnell RJ, Leon JM, Vazza G, nens MM, Aerts JM: The human chitotriosi- Wray H, Puu M, Larsson B, Stockley R: A
Mostacciuolo ML, Paoletti MG, Falcone FH: dase gene: nature of inherited enzyme defi- randomised, placebo-controlled study of the
High prevalence of chitotriosidase deficien- ciency. J Biol Chem 1998;273:25680–25685. CXCR2 antagonist AZD5069 in bronchiec-
cy in Peruvian Amerindians exposed to chi- 149 Marion CR, Wang J, Sharma L, Losier A, Lui tasis. Eur Respir J 2015;46:1021–1032.
tin-bearing food and enteroparasites. Car- W, Andrews N, Elias JA, Kazmierczak BI, 161 Moss RB, Mistry SJ, Konstan MW, Pilewski
bohydr Polym 2014;113:607–614. Roy CR, Dela Cruz CS: Chitinase 3-like 1 JM, Kerem E, Tal-Singer R, Lazaar AL, In-
139 Hall AJ, Quinnell RJ, Raiko A, Lagog M, Siba (Chil1) regulates survival and macrophage- vestigators CF: Safety and early treatment ef-
P, Morroll S, Falcone FH: Chitotriosidase mediated interleukin-1beta and tumor ne- fects of the CXCR2 antagonist SB-656933 in
deficiency is not associated with human crosis factor alpha during Pseudomonas ae- patients with cystic fibrosis. J Cyst Fibros
hookworm infection in a Papua New Guin- ruginosa pneumonia. Infect Immun 2016; 2013;12:241–248.
ean population. Infect Genet Evol 2007; 7: 84:2094–2104. 162 Nair P, Gaga M, Zervas E, Alagha K, Harg-
743–747. 150 Dela Cruz CS, Liu W, He CH, Jacoby A, Gor- reave FE, O’Byrne PM, Stryszak P, Gann L,
140 Lee P, Waalen J, Crain K, Smargon A, Beu- nitzky A, Ma B, Flavell R, Lee CG, Elias JA: Sadeh J, Chanez P, Study I: Safety and effi-
tler E: Human chitotriosidase polymor- Chitinase 3-like-1 promotes Streptococcus cacy of a CXCR2 antagonist in patients with
phisms G354R and A442V associated with pneumoniae killing and augments host tol- severe asthma and sputum neutrophils: a
reduced enzyme activity. Blood Cells Mol erance to lung antibacterial responses. Cell randomized, placebo-controlled clinical tri-
Dis 2007;39:353–360. Host Microbe 2012;12:34–46. al. Clin Exp Allergy 2012;42:1097–1103.

Innate Immunity of the Lung J Innate Immun 15


DOI: 10.1159/000487057

You might also like