Skip to main content
Erschienen in: Seminars in Immunopathology 4-6/2024

Open Access 07.03.2024 | Review

Physiological and immunological barriers in the lung

verfasst von: Takahiro Kageyama, Takashi Ito, Shigeru Tanaka, Hiroshi Nakajima

Erschienen in: Seminars in Immunopathology | Ausgabe 4-6/2024

Abstract

The lungs serve as the primary organ for respiration, facilitating the vital exchange of gases with the bloodstream. Given their perpetual exposure to external particulates and pathogens, they possess intricate protective barriers. Cellular adhesion in the lungs is robustly maintained through tight junctions, adherens junctions, and desmosomes. Furthermore, the pulmonary system features a mucociliary clearance mechanism that synthesizes mucus and transports it to the outside. This mucus is enriched with chemical barriers like antimicrobial proteins and immunoglobulin A (IgA). Additionally, a complex immunological network comprising epithelial cells, neural cells, and immune cells plays a pivotal role in pulmonary defense. A comprehensive understanding of these protective systems offers valuable insights into potential pathologies and their therapeutic interventions.
Hinweise
This article is a contribution to the Article Collection on Immunopathology of Barrier Function - Guest Editor: Koji Hase & Hiroshi Ohno.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

In humans, the respiratory tract is divided into the upper airways (nasal cavity, pharynx, and larynx) and the lower airways (trachea, bronchi, bronchioles, and alveoli). The lungs consist of the bronchi, bronchioles, and alveoli. The primary function of the respiratory tract is to efficiently change gas between inhaled air and the bloodstream [1]. The lungs are constantly exposed to various environmental elements, including pathogens, toxins, and allergens, that cause pulmonary infections or inflammation. Therefore, the human body needs to defend itself against countless intruders through the respiratory tract. Thus, the respiratory system has developed highly sophisticated barrier functions through physiological and immunological mechanisms. Airway epithelial cells (AECs) were originally thought to serve only as a physical barrier. However, recent research has elucidated the interplay between AECs and immune cells, revealing that AECs initiate immune responses [2]. This complex network of epithelial and immune cells is involved in the pathogenesis of various diseases [38]. In fact, inhibitors of thymic stromal lymphopoietin (TSLP), a cytokine mainly produced by AECs in the lungs, have emerged as therapeutic agents for asthma [9]. Furthermore, there is growing interest in the tripartite network of epithelium, immune, and neural cells [1014]. In this review, we summarize how the lungs establish sophisticated barrier mechanisms.

Multi-layered lung barriers

The barrier mechanism of the lung is, first, a continuous layer lining the respiratory tract composed of AECs, separating the body from the environment. Second, mucociliary clearance (MCC), consisting of the production of mucus and the coordinated movement of cilia, is the defense mechanism. Third, antimicrobial peptides and proteins act as a chemical barrier. Fourth, various cells, including both immune and non-immune cells, work in a coordinated manner to establish an immunological barrier. By summarizing the function of AECs and innate immune cells as barriers, we will attempt to understand the defense mechanisms in the lung and their involvement in pulmonary diseases.

Physicochemical barriers

Cell adhesion

The respiratory epithelium begins as a pseudostratified columnar epithelium in the nasal cavity, trachea, and bronchi, transitioning into columnar and cuboidal cells in the bronchioles. Finally, it forms a thin single-cell alveolar epithelium in the alveoli. The bronchial and alveolar epitheliums are composed of different cell types (Fig. 1). Bronchial epithelial cells consist of basal cells, ciliated cells, goblet cells, club cells, tuft cells, neuroendocrine cells, and ionocytes [15]. Alveolar cells can be divided into alveolar type 1 epithelial cells (AT1) and alveolar type 2 epithelial cells (AT2) [16].
The effectiveness of the physical barrier is a result of the coordinated interaction between neighboring epithelial cells through cell–cell adhesion complexes. The connection between adjacent airway epithelial cells is facilitated by apical tight junctions (TJs), adherens junctions (AJs), and desmosomes. They selectively regulate paracellular permeability, limit the transport of macromolecules, and maintain barrier integrity [17].
TJs are the uppermost intracellular junctions and play a pivotal role in regulating paracellular permeability. Using TJs, AECs can selectively control the passage of substances, particularly ions, water, and macromolecules. TJs comprise several essential transmembrane proteins of the claudin family, occludin, tricellulin, and junctional adhesion molecules (JAMs). The claudin family mainly regulates tight junction permeability. Additionally, there are critical cytoplasmic proteins associated with TJs, including zonula occludens (ZO)-1, ZO-2, and ZO-3, which bind directly to the transmembrane proteins, including claudins and occludin, on one end and connect to actin cytoskeleton on the other end [18, 19].
AJs are cadherin-catenin adhesion complexes located below TJs. AJs primarily provide mechanical strength by mediating adhesion between neighboring cells. In addition, AJs are involved in the establishment and maintenance of cell polarity in AECs, which is vital for proper tissue organization and function. The major component of AJs is transmembrane protein E-cadherin: its extracellular domain binds homotypically to adjacent cells, while the cytoplasmic domain binds to the catenins, linking to actin cytoskeleton. Through these networks, AJs regulate cell shape and movement and transmit mechanical forces between the cells [20].
Desmosomes are located around the midpoint of epithelial cells, providing robust mechanical stability. Desmosomes consist of transmembrane proteins, including desmogleins and desmocollins, that interact with intracellular proteins, such as desmoplakin, to anchor intermediate filaments. Hemidesmosomes, on the other hand, assist in anchoring the epithelial layer to the basal membrane, contributing to tissue integrity [17].
The robust cell adhesion described above is crucial not only as a barrier function but also for maintaining respiratory homeostasis. On the contrary, the disruption of this critical function is implicated in numerous pathological conditions. It has been observed that E-cadherin and ZO-1 expression is decreased, and allergen permeability is increased in AECs of asthmatic patients [21, 22]. Additionally, cigarette smoke may contribute to the pathogenesis of chronic obstructive pulmonary disease (COPD) by impairing cell adhesion in AECs [23, 24]. However, whether the dysfunction of cell adhesion is the etiology or the result of diseases must be interpreted with caution.

Mucociliary clearance

In addition to the above barrier mechanism based mainly on cell adhesion, there is another defense mechanism mediated by mucus production in the respiratory system (Fig. 2). The airway epithelium, from nasal to bronchioles, is coated with mucus, which is a viscous and gel-like secretion. The characteristic viscous, elastic, and adhesive properties of mucus are mainly attributed to mucins. Although many genes encoding mucins have been reported, MUC5AC and MUC5B are the predominant mucins in the airways [25]. Through its adhesive nature, mucus captures inhaled allergens, pathogens, and harmful substances, preventing these particles from reaching deeper into the lungs. Furthermore, this mucus layer is transported in a coordinated manner towards the oral cavity to facilitate the removal of these captured particles from the body, a process known as mucociliary clearance (MCC). The continuous and synchronized beating of the ciliated cells mediates this movement.
MUC5AC is primarily found in the proximal airways, including the trachea, bronchi, and bronchioles, but not in the distal bronchioles. It is predominantly synthesized by goblet cells. MUC5B is the dominant mucin in the superficial epithelium and glands across the respiratory tract, including the distal airways. Submucosal glands (SMGs), which are beneath the cartilaginous airways of the human lung, primarily secrete MUC5B along with a lesser amount of MUC5AC. These glands are composed of serous cells, secretory cells, myoepithelial cells, and goblet cells. SMGs are essential secretory structures that contribute to airway defense, mucus production, and antimicrobial protection in the lungs. Their dysfunction can have significant implications for lung health and contribute to the pathogenesis of respiratory diseases. In healthy individuals, MUC5B is more prevalent than MUC5AC, and maintaining a proper balance between MUC5AC and MUC5B is crucial for effective MCC. Pathological increases in MUC5AC have been linked to the onset, progression, and heightened exacerbation risk of COPD [26]. Although controversial results have been reported on the increase or decrease of MUC5AC and MUC5B due to various methods of assessing mucin, there is no doubt that the pathophysiology of cystic fibrosis (CF) is related to the viscous mucus secreted from the epithelial surface of the respiratory and intestinal tract [27, 28].
Interestingly, it has been reported that the ratio of MUC5AC to MUC5B is altered in asthmatic patients with increased production of MUC5AC [29]. Furthermore, the absence of MUC5B may result in impaired mucociliary clearance by persistent inflammation [30]. These findings underscore the close relationship between the function of the mucus layer and various pulmonary diseases.
The periciliary layer (PCL), distinct from the overlaying mucus layer, is a thin, watery layer surrounding the cilia. Composed of a low-viscosity fluid containing membrane-associated mucins (MUC1, MUC4, and MUC16) and other molecules, including glycolipids, the PCL allows unimpeded ciliary movement. This feature is critical for propelling the mucus layer and trapped particles out of the respiratory system, highlighting its essential role in effective MCC [31]. The significance of ciliary movement is apparent when considering primary ciliary dyskinesia (PCD), a genetic disorder characterized by chronic respiratory tract infections and abnormal ciliary structure and function. In PCD, these cilia are either structurally abnormal or paralyzed, leading to a buildup of mucus and bacteria, resulting in frequent infections of the lungs, ears, sinuses, and throat. Treatment typically focuses on managing symptoms and preventing lung damage, often requiring regular airway clearance, antibiotics for infections, and careful monitoring [32].

Chemical barriers

In addition to mucins, the mucus contains water, salts, lipids, immunoglobulin A (IgA), and antimicrobial proteins. These antimicrobial proteins, including defensin, lysosome, and lactoferrin, directly neutralize or destroy bacteria, viruses, and fungi by disrupting microbial cell membranes or interfering with essential microbial processes. Beyond their direct antimicrobial functions, they modulate the immune responses, ensuring a balance between effective defense and tissue protection. Moreover, IgA also provides a multifaceted defense, balancing effective protection against pathogens [33]. IgA is an antibody class present predominantly on the surfaces of mucous tissues, including the lungs. It exists in its secretory form, secretory IgA (sIgA), whose polymeric structure, often dimeric, enhances its avidity and functional capacity in mucus. Produced by tissue-resident memory B cells, plasmablasts, and plasma cells within the respiratory mucosa, IgA is transported across epithelial cells into the mucus by the polymeric immunoglobulin receptor (pIgR), predominantly expressed on secretory cells [3436]. Dimeric IgA (dIgA) binds to pIgR on the basolateral surface of airway epithelial cells. Following binding, the IgA-pIgR complex is internalized through endocytosis and transported across the epithelial cell to the apical surface in a process known as transcytosis. During transcytosis, pIgR undergoes proteolytic cleavage, resulting in the formation of the secretory component (SC). Upon release into the mucus layer, SC remains bound to dIgA, thus forming sIgA. Once bound to pathogens, IgA can neutralize them, preventing them from entering cells. Additionally, IgA has anti-inflammatory properties, ensuring that immune responses do not inadvertently damage lung tissue [37].
Associations between sIgA and various respiratory diseases, such as COPD, asthma, and COVID-19, have been observed. Decreased expression of pIgR and degradation of sIgA may lead to loss of sIgA in COPD small airways [36]. On the other hand, patients with severe COPD have increased IgA expression in lung lymphoid follicles [38]. In asthma patients, the expression of pIgR in bronchial epithelial cells is reduced due to IL-4Rα-mediated signaling [39]. Recent findings indicate that IgA autoantibodies targeting pulmonary surfactant B and C in severe COVID-19 patients may contribute to respiratory failure, suggesting that IgA acts not only protectively but also pathologically [40]. Elucidating the precise role of IgA in the airways will be vital to understanding the pathogenesis of many respiratory diseases.

Airway epithelial cell barriers

AECs play a critical role in establishing the physiological barrier in the lungs. Recent advances in analytical technologies have revealed that AECs are a more heterogeneous population and constitute a more complex network than previously assumed [4145]. Here, we summarize the characteristics of subpopulations of AECs.

Basal cells

Basal cells exhibit a columnar or cuboidal shape and are firmly anchored to the basement membrane through specialized structures known as hemidesmosomes. They can be identified based on the expression of p63 and keratin 5 (KRT5). Functionally, they are stem cell–like and can self-renew and differentiate into various epithelial cell subtypes, playing a crucial role in maintaining the integrity of the epithelial barrier and facilitating repair and regeneration after injury.
Recent studies have suggested that basal cell abnormalities may contribute to the development of several respiratory diseases. The comparison of RNA-seq data of basal cells between smokers and nonsmokers showed that COPD risk genes identified in GWAS are upregulated by smoking [46]. Other findings suggest that sensing apoptotic cells by a TAM receptor tyrosine kinase Axl on basal cells is important for tracheal basal cell expansion, cell cycle reentry, and symmetric cell division and is involved in the pathogenesis of COPD [47]. Meanwhile, single-cell RNA sequencing (scRNA-seq) analysis of lungs from IPF patients identified an aberrant basal cell population that co-expressed basal epithelial, mesenchymal, senescence, and developmental markers [48]. Another group used single-cell cloning technology to generate a library of basal stem cells from lungs derived from IPF patients [49]. Among these clones, a clone that transformed normal lung fibroblasts into pathogenic myofibroblasts in vitro was identified. Interestingly, this clone resembled the genetic profile of the abnormal basal cell population identified by scRNA-seq described above [48, 49]. The relationship between basal cells and allergic pathology has also been the focus of interest. Based on scRNA-seq data of airway epithelial cells derived from patients with chronic rhinosinusitis or asthma, two populations of basal cells with different gene expressions have been reported [50, 51]. These two populations correspond to differentiation stages, with the less mature population characterized by high expression of TP63. Furthermore, IL-4 and IL-13 have been shown to regulate basal cell stem function in vitro [50]. In addition, because of their progenitor function, basal cells have attracted attention for their application in regenerative medicine [52, 53].

Ciliated cells

Ciliated cells are abundant in the large and medium airways and are crucial in moving mucus, which traps debris and pathogens, out of the airways. Each ciliated cell has numerous cilia that extend into the mucus. These cilia are anchored to the cytoskeleton and move in a coordinated manner. They are terminally differentiated and can originate from secretory cells or basal cells. Notch signaling pathways regulate the differentiation of ciliated cells. When Notch signaling is inhibited, it promotes the differentiation of ciliated cells [54]. Also, forkhead box protein J1 (FOXJ1) is a master regulator of ciliogenesis. Its expression is essential for the differentiation of ciliated cells and the formation of motile cilia. Of clinical importance, ciliated cells have been reported to be involved in viral infections and asthma [6, 8, 55]. Rhinovirus C (RV-C), the predominant cause of the common cold, is infectious via cadherin-related family member 3 (CDHR3) in the host [56]. The expression of CDHR3 is shown to be mainly restricted to ciliated cells [57]. Moreover, CDHR3 has been identified as a susceptibility gene for asthma, especially in young children [58]. These findings suggest a strong association between rhinovirus infections, CDHR3, and the development of asthma [55, 58].

Club cells

Club cells, formerly known as Clara cells, are distinctive dome-shaped cubical cells found in the small airways and characterized by the expression of secretoglobin family 1A member 1 (Scgb1a1), also known as Clara cell secretory protein (CCSP). Their differentiation depends on the transforming growth factor-b receptor Alk5 (activin receptor-like kinase 5). They secrete a variety of substances, the most notable being Scgb1a1. This protein has anti-inflammatory and immunosuppressive properties by suppressing various pro-inflammatory cytokines [59]. In an ALI (air–liquid interface) culture using airway epithelial cells derived from COPD patients, the supplementation of Scgb1a1 was shown to regulate IL-8 release by cigarette smoke extract [60]. Immunostaining for Scgb1a1 in the airways was reduced in COPD patients and decreased with increasing severity of COPD, suggesting the association between club cells and the pathogenesis of COPD [61]. It has also been reported that Scgb1a1 in bronchoalveolar lavage (BAL) is decreased in asthma patients [62]. Of interest, low mRNA expression levels of Scgb1a1 in airway epithelial cell brushings in asthmatic patients have been shown to correlate with poor clinical outcomes [63]. In addition, club cells possess the ability to differentiate into both ciliated cells and goblet cells. Moreover, in cases where basal cells are injured or lost, club cells can differentiate into basal cells [64]. Thus, because of the high plasticity of lung epithelial cells, it was difficult to identify the origin cell of the tumor. However, using a lineage-tracking mouse model and scRNA-seq, it has been shown that the causative cells of lung adenocarcinoma are club cells and AT2 cells [65].

Goblet cells

Goblet cells are characterized by their densely packed mucin granules and surfactant proteins. Their primary function is to produce and secrete mucus. In particular, MUC5AC is mainly synthesized by goblet cells. With ciliated cells, they play a crucial role in facilitating effective MCC. Goblet cells are derived from club cells through the activation of SAM pointed domain containing ETS transcription factor (SPDEF) and forkhead box A3 (FOXA3). In several lung diseases, such as asthma and COPD, there is an increase in the number of goblet cells, a condition known as goblet cell hyperplasia [66, 67]. This can cause coughing and wheezing due to excessive mucus production. In particular, allergic inflammation and goblet cells are closely related, and IL-13 induces goblet cell hyperplasia and metaplasia [68, 69].

Pulmonary neuroendocrine cells

Pulmonary neuroendocrine cells (PNECs) can be found either as individual isolated cells or organized in small clusters known as neuroendocrine bodies (NEBs) throughout the conducting airways, especially near the respiratory tree branch. While not being neurons themselves, PNECs are innervated by both the sympathetic and parasympathetic nervous systems. PNECs can monitor airway oxygen and respiratory status and quickly release various substances, including neurotransmitters such as gamma-aminobutyric acid (GABA), calcitonin gene–related peptide (CGRP), bombesin, and serotonin [70]. These neurotransmitters produced by PNECs have been reported to induce goblet cell hyperplasia and ILC2 activation [14, 71]. Thus, PNECs are considered to be important in the coordination of the epithelial, immune, and nervous systems, facilitating respiratory homeostasis.

Tuft cells

Tuft cells, also known as brush cells, are expressed in various tissues, including the respiratory and gastrointestinal tracts. Their distinctive morphology, characterized by a tuft of microvilli extending into the mucosal lumen, enables them to sense the extracellular environment. They can detect changes in the local chemical composition and transmit signals to nearby cells [72, 73]. While the exact function of tuft cells in the lung is still being elucidated, it has been reported that they can release IL-25 and cysteinyl leukotrienes (CysLTs), which synergistically contribute to type 2 inflammation in the lung [74].

Ionocytes

Recently, pulmonary ionocytes were identified as a rare cell type using scRNA-seq of human bronchial epithelial cells and mouse tracheal epithelial cells [41, 45]. Ionocytes co-express forkhead box l1 (FOXL1), multiple subunits of the vacuolar-type H+-ATPase (V-ATPase), and cystic fibrosis transmembrane conductance regulator (CFTR). One of the most notable characteristics is their high expression of CFTR, which encodes an anion channel critical for maintaining the hydration and pH of the airway surface liquid. Thus, they may play an essential role in MCC by cooperating with ciliated cells and secretory cells. Moreover, because mutations in the CFTR gene are responsible for CF, ionocytes have attracted attention as a therapeutic target for CF [75]. However, it has been recently suggested that the function of CFTR in human airways is mainly carried out by secretory cells, as ionocytes are a small population [76]. On the other hand, the regulatory function of CFTR-mediated chloride differs between cell types, with secretory cells involved in the secretion of chloride and ionocytes in absorption [77]. Analysis using the transgenic ferret models suggests that there are at least three subtypes of ionocytes [78]. Further analysis of ionocytes is expected to clarify the pathogenesis of CF and other airway diseases.

Alveolar cells

The alveolar epithelium consists of two types of epithelial cells: alveolar type1 (AT1) cells and alveolar type 2 (AT2) cells [16]. AT1 cells are highly specialized for gas exchange and barrier function, characterized by their flat, thin, and squamous morphology. This distinctive shape allows AT1 cells to efficiently cover about 95% of the alveolar surface. AT1 cells, together with capillary endothelium, form the alveolar-capillary barrier, otherwise known as the air-blood barrier. This incredibly thin barrier is crucial not only for efficient gas exchange between the bloodstream carbon dioxide and airborne oxygen but also for separating the bloodstream from foreign pathogens.
AT2 cells are cuboidal cells with apical microvilli and lamellar bodies. Their most important physiological role is the synthesis and secretion of pulmonary surfactant. Pulmonary surfactant reduces surface tension within the alveoli, preventing alveolar collapse during exhalation and preserving alveolar structure for efficient breathing. Besides surfactants, AT2 cells produce various cytokines, chemokines, growth factors, and antimicrobial peptides, which play roles in inflammation, immune responses, and tissue repair.
In addition, AT2 cells serve as progenitor cells for alveolar epithelium. When the alveolar epithelium is injured, AT2 cells can undergo self-renewal and differentiate into AT1 cells, contributing to tissue repair and regeneration [79]. Wnt signaling may be involved in maintaining the stem cell properties of AT2 cells [8082]. Furthermore, their regenerative potential is likely to inform the development of novel therapies for COPD [83] and pulmonary fibrosis [84, 85].
Two recent reports of scRNA-seq analyses of human distal airways have identified AT0 cells and respiratory airway secretory (RAS) cells. AT0 cells, originated from AT2 cells, can differentiate into AT1 cells or terminal and respiratory bronchiole secretory cells [43]. In contrast, RAS cells can unidirectional differentiate into AT2 cells through Notch and Wnt signaling [86]. These cells, which are capable of differentiation, may exhibit phenotypic alterations with age or in response to pathological conditions; thereby, their function and pathology need to be analyzed under a broader range of conditions.

Immunological barriers by pattern recognition receptors

The first line of defense in the lungs is not only the physicochemical barriers but also the innate immune system. The innate immune system provides immediate, non-specific responses to threats. Pattern recognition receptors (PRRs) on innate immune cells detect conserved structures on pathogens, namely pathogen-associated molecular patterns (PAMPs). PRRs also recognize damage-associated molecular patterns (DAMPs) from stressed or damaged host cells. Upon recognition, these receptors initiate various immune responses, including inflammation, phagocytosis, and the production of cytokines and chemokines, thereby inducing the adaptive immune system. Common types of PRRs include Toll-like receptors (TLRs), NOD-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)–like receptors (RLRs), and C-type lectin receptors (CLRs). Notably, PRRs are expressed in AECs as well as innate immune cells [87].

TLRs

TLRs, which are membrane-bound receptors, can be found on the cell surface or in intracellular vesicles like endosomes. In humans, there are ten distinct TLRs (TLR1 to TLR10), each recognizing specific PAMPs. TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10 are localized to the plasm membrane, where they can recognize and respond to extracellular pathogens. For instance, TLR4 recognizes lipopolysaccharide (LPS) from gram-negative bacteria, and TLR5 detects bacterial flagellin. On the other hand, TLR3, TLR7, TLR8, and TLR9 are localized in endosomes, where they can detect intracellular nucleic acids. TLR3 senses double-stranded RNA, often associated with viral infections. TLRs are expressed in a wide range of cell types, enabling a coordinated and broad response to potential threats [8890].

NLRs

NLRs are a group of cytoplasmic PRRs containing three domains: a C-terminal leucine-rich repeat (LRR) domain, a nucleotide-binding domain (NBD), and an N-terminal effector domain. The LRR domain is involved in ligand recognition. NBD is highly conserved among NLRs and is crucial in forming NLR complexes or inflammasomes. The N-terminal effector domain varies among different NLR family members and determines the downstream signaling pathway and interactions with other proteins. There are different types of effector domains, including caspase and activation and recruitment domain (CARD), pyrin domain (PYD), baculoviral inhibitor of apoptosis repeat (BIR) domain, and transactivator domain (AD). When a ligand binds to the LRR domain, it induces a conformational change in the NLR. This change leads to the exposure or activation of the NBD, initiating the formation of protein complexes called inflammasomes. The formation of inflammasomes results in the activation of caspase-1 and the cleavage of inactive precursor forms of IL-1β and IL-18 into their active forms [91].

RLRs

RLRs, which are a class of cytoplasmic PRRs, consist of three members: RIG-I, melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP-2) [92]. RLRs contain DExD/H box RNA helicase domains that are central to their RNA sensing function. RIG-I recognizes short double-stranded RNA (dsRNA) with 5′-triphosphates, often found in RNA viruses, while MDA5 detects longer dsRNA. When RIG-I or MDA5 binds to RNA ligands, they undergo conformational changes that expose their N-terminal CARD, initiating downstream signaling through interactions with adaptor proteins like mitochondrial antiviral signaling (MAVS). This signaling cascade leads to the production of antiviral cytokines and the activation of immune responses. On the contrary, LGP-2 lacks the CARD and cannot directly initiate antiviral signaling but acts as a regulatory protein that modulates the signaling of RIG-I and MDA5 in a context-dependent manner.

CLRs

CLRs are numerous and include both membrane-bound and soluble receptors. They recognize carbohydrate structures, primarily on pathogens, through their carbohydrate recognition domains (CRDs). Membrane-bound CLRs are mainly found on myeloid cells, including dendritic cells and macrophages. Soluble CLRs, like mannose-binding lectin (MBL), circulate in the extracellular environment and can activate the complement system. Upon ligand binding, CLRs lead to immune responses like phagocytosis and cytokine production [89, 93]. Some CLRs, such as Dectin-1, sense specific pathogens like fungi [94].

Interactions of epithelial and immune cells in the lung barrier

In recent years, deeper analysis of cellular networks, such as crosstalk between epithelial and immune cells, has become feasible. A contributing factor to this advancement is the progress in transcriptome technologies at the single-cell level, exemplified by scRNA-seq. Furthermore, initially, single-cell transcriptomes had difficulties in data integration and interpretation due to problems such as batch effect, but these problems are being overcome due to large datasets, powerful computational resources, and advances in learning algorithms.
Single-cell transcriptomics offers significant advantages, including the identification of rare cell populations like ionocytes and the estimation of cell differentiation processes through trajectory analysis [41, 45]. Furthermore, these techniques have enabled the inference of intercellular networks by utilizing information on ligand-receptor interactions [43, 86]. These advancements are being applied to a variety of respiratory diseases.
Specifically, scRNA-seq conducted on nasopharyngeal and bronchial samples collected from patients with moderate to severe COVID-19 has been reported [95]. In these patients, secretory cells exhibited significantly higher expression of chemokines such as CXCL1, CXCL3, CXCL6, and CXCL17 compared to controls, suggesting an enhancement in the mobilization of neutrophils, T cells, and mast cells. Furthermore, in severe cases compared to moderate, there was a stronger interaction between epithelial and immune cells, with immune cells, including inflammatory macrophages, being more activated. Thus, this interaction may contribute to the exacerbation of infection. Importantly, infected epithelial cells showed upregulation of the SARS-CoV-2 entry receptor ACE2, which was associated with interferon signaling in immune cells. These findings suggest that intercellular links are important not only in the severity of infection but also in the establishment of infection.
Utilizing a human model of localized asthma exacerbation by bronchoscopic segmental allergen challenge, a comprehensive analysis of the lower airway mucosa of allergic asthmatics and allergic non-asthmatics using scRNA-seq has been conducted [96]. In response to allergens, asthmatic airway epithelium was highly dynamically upregulating genes involved in matrix degradation, mucus metaplasia, and glycolysis while failing to induce the injury-repair and antioxidant pathways observed in controls. The study revealed a Th2 cell-mononuclear phagocyte-basal cell interactome unique to asthmatics, driven by interactions between TNF family members and type 2 cytokines from Th2 cells. This pathogenic cellular network in asthmatics may override protective injury-repair responses and drive asthma pathobiology.
In addition to the single-cell transcriptome, the advancement of three-dimensional (3D) culture systems, represented by organoids, experimental models that closely recapitulate human physiology, allow further analysis in a coordinated manner [43, 86, 97]. Fetal lung organoids derived from fetal lung bud tips provide significant insights into lung development. In humans, SOX2 and SOX9 have been identified as progenitor markers, and their maintenance requires EGF, FGF, and WNT signaling, as well as the inhibition of BMP and TGF-β [98]. The interaction between epithelium and immune cells was also shown to play an important role in lung development using fetal lung organoids [99]. In immunohistochemistry of fetal lungs, immune cells were found surrounding lung progenitor cells. Therefore, the expression of cytokine receptors in progenitor cells was evaluated, and candidates for interacting cytokines between epithelial and immune cells were identified. Among various cytokines, the supplementation of IL-1β in fetal lung organoids resulted in decreased expression of SOX9 and increased expression of TP63. These findings suggest that myeloid cells, widespread throughout the lungs, produce IL-1β during early lung development, which induces epithelial stem cell differentiation into mature basal cells.
Besides organoids, precision-cut lung slices (PCLS) have emerged in respiratory research as a 3D culture method [100]. PCLS are live tissue preparations that encompass all resident cell types, including smooth muscle cells, epithelial cells, and fibroblasts. These cells maintain intercellular interactions and cell–matrix relationships within the complex structure of the lung, making them suitable for analysis of cellular networks through single-cell transcriptomics.
Recently, the utilization of human PCLS (hPCLS), single-cell transcriptome analysis, and deep learning-based query-to-reference mapping has been reported as a powerful research platform for elucidating the pathogenesis of lung fibrosis and facilitating drug development [101]. In this study, fibrosis was induced in hPCLS from nonfibrotic human lung tissue by adding a pro-fibrotic cytokine mix, and the scRNA-seq data obtained from the fibrosis-induced hPCLS were merged with single-cell data from a cohort of pulmonary fibrosis patients. Furthermore, single-cell architectural surgery (scArches), which is a deep learning strategy for mapping single-cell datasets to a reference atlas, was employed to map the obtained data to the Human Lung Cell Atlas, thereby validating an ex vivo model of fibrosis. Additionally, analyses of cell morphology and intercellular networks were conducted through micro-CT staging of hPCLS and patient tissues. The pathways of fibrogenesis and healing processes were evaluated by administering antifibrotic drugs to fibrosis-induced hPCLS. As demonstrated by this cutting-edge research, the analysis of cellular networks in lung physiology and pathogenesis is expected to accelerate with the advancements in transcriptome data analysis, human-like experimental systems, and artificial intelligence technology.

Immunological barriers by myeloid cells

The lungs contain numerous immune cells that maintain respiratory homeostasis. However, the immune system is not solely composed of immune cells but also interacts with non-immune cells such as AECs and neural cells. This section summarizes the subtypes of innate immune cells in the lung, along with their characteristics and contribution to the defense mechanisms.

Alveolar macrophages

Alveolar macrophages (AMs) are the most abundant immune cells in the airway lumen and are crucial components of the respiratory innate immune systems [102]. They express the high levels of CD11c, Siglec-F, and CD169 and lack CD11b [103]. GM-CSF is required for the differentiation and maturation of AMs. Recently, it has been reported that AT2 cell–derived GM-CSF plays a nonredundant and critical role in establishing the postnatal AM population and maintaining AMs in adult lungs, emphasizing a critical link between epithelial and immune cells [104]. AMs have the phagocytic function to clear inhaled pathogens, dead cells, and foreign airborne particles, maintaining the health of the alveolar environment. In addition, they can also produce pro-inflammatory cytokines via PRRs and induce adaptive immune responses. Concerning these pro-inflammatory properties, paralysis of immune function due to poor phagocytosis of AMs has been reported following severe infections. Increased SIRP1a expression on AMs during the early stages of paralysis directly impairs phagocytosis, and SIRP1a acts as a tyrosine kinase receptor, triggering the induction of an immunosuppressive microenvironment [105].
On the other hand, they can produce anti-inflammatory molecules, such as IL-10 and TGF-β, to prevent excessive inflammation and tissue damage. A subset of AMs, forming connexin 43 (CX43)–containing gap junction channels with the alveolar epithelium, communicates immunosuppressive signals to reduce lipopolysaccharide-induced lung inflammation [106]. Besides immune regulation, they contribute to the homeostasis of pulmonary surfactants [102]. Pulmonary alveolar proteinosis (PAP) is a disease characterized by the abnormal accumulation of surfactants in the alveoli, leading to respiratory failure. Autoimmune PAP, which constitutes the majority of PAP, is initiated by the presence of anti-GM-CSF autoantibodies in patients’ sera [107]. These antibodies result in the functional impairment of AMs, which is the underlying etiology of PAP [108]. Notably, it has been reported that the administration of GM-CSF can effectively ameliorate respiratory conditions in patients with PAP [109].

Interstitial macrophages

Interstitial macrophages (IMs) are a distinct population of macrophages located in the lung interstitium or parenchyma. They express high levels of CD11b but lack Siglec-F, which distinguishes them from AMs [103]. While they also have phagocytic functions, IMs are more involved in modulating immune responses and tissue repair. In addition, IMs have been shown to produce IL-10 spontaneously [110].

Eosinophils

Eosinophils are involved in immune responses to parasitic, bacterial, and viral infections, as well as in the maintenance of homeostasis. In response to stimuli, they release granule proteins, including cytotoxic major basic protein (MBP), eosinophil cationic protein (ECP), eosinophil peroxidase (EPX), Galectin-10, lipid mediators, and many cytokines. These granule proteins help eliminate parasites, bacteria, and viruses, while excessive release may cause damage to surrounding tissue and cells. For example, MBP promotes histamine release from mast cells and triggers a cascade of type 2 inflammation. Furthermore, eosinophils could secrete type 2 cytokines, such as IL-4, IL-9, and IL-13. Thus, eosinophils are hallmark cells in allergic asthma and other allergic lung diseases, contributing to bronchoconstriction, mucus production, and inflammation [111, 112]. Eosinophil-targeted treatments are crucial for managing eosinophilic asthma and similar conditions. Corticosteroids are commonly used, either inhaled or systemic, to counter eosinophilic inflammation. In addition, biologics have been developed to reduce eosinophils [113]: mepolizumab and reslizumab target IL-5, suppressing eosinophil development and activity, whereas benralizumab specifically binds to the IL-5 receptor to kill eosinophils by antibody-dependent cell-mediated cytotoxicity. Dupilumab inhibits IL-4 and IL-13, both essential for eosinophils to accumulate in inflamed tissues.
Moreover, recent findings have indicated that eosinophils are not a homogenous population [114]. In the murine lung, resident eosinophils (rEos), characterized by Siglec-FintCD62L+CD101lo, are present at a steady state. However, an additional subset of eosinophils as inflammatory eosinophils (iEos), characterized by Siglec-FhiCD62LCD101hi, emerges alongside rEos during allergic inflammation caused by allergen inhalation. Importantly, iEos and rEos possess different functional profiles: rEos can inhibit allergen-loaded dendritic cells’ maturation and pro-Th2 function, whereas iEos promotes allergic inflammation [115]. Identifying and understanding specific eosinophil subtypes can lead to the development of more targeted therapies, minimizing side effects and improving treatment efficacy.

Neutrophils

Neutrophils constitute a pivotal component of the innate immune system, eliminating pathogens, primarily bacteria and fungi. Neutrophils recognize invading pathogens and execute their antimicrobial defense functions through phagocytosis and the release of diverse cytokines and chemokines, thereby orchestrating the recruitment of inflammatory immune cells [116]. Moreover, neutrophils form neutrophil extracellular traps (NETs), which are composed of DNA, histones, and antimicrobial proteins. NETs serve as a physical barrier to trap and kill pathogens. After the initial response, neutrophils undergo apoptosis and are also removed by macrophages to limit excessive inflammation. Thus, while neutrophils play an indispensable role in immune defense, they can be implicated in the pathogenesis of various diseases. For instance, type 2–low asthma is associated with pronounced neutrophil infiltration [117]. Furthermore, their dysregulated responses can contribute to the development of COPD and acute respiratory distress syndrome (ARDS) [118]. Therefore, the appropriate regulation of neutrophil activity is essential for maintaining lung homeostasis.

Dendritic cells

Dendritic cells (DCs) play a crucial role in the immune system by capturing inhaled pathogens and allergens and subsequently migrating to lymph nodes, where they present intracellularly processed antigens to antigen-specific T cells. These DCs encompass diverse subsets that exhibit specialized functions based on their anatomical localization and their roles in pathogen recognition, thereby contributing to various immune responses. While the categorization of DC subsets in the lung remains a subject of debate, they can generally be classified into two major categories: plasmacytoid DCs (pDCs) and conventional DCs (cDCs), with the latter further subdivided into cDC1 and cDC2 [119]. pDCs primarily serve as producers of type 1 interferons, primarily upon TLR7 and TLR9 stimulation, which are vital in combating viral infections. However, the pathogenicity of pDCs deserves attention, as type 1 interferons have also been implicated in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus and psoriasis [120, 121].
Distinct markers differentiate cDC1 from cDC2, with the former expressing CD103 and the latter expressing CD11b. During viral infections, cDC1s assume a critical role in the induction of effector CD8+ T cells within the lung by facilitating cross-presentation via MHC class I molecules. Conversely, cDC2s are pivotal in generating central memory CD8+ T cells. In the context of asthma pathogenesis, cDC1s contribute to the suppression of Th2 and Th17 immune responses through IL-12 production, while cDC2s promote Th2 and Th17 immune responses in response to challenges with house dust mites (HDM) via receptor engagement, such as Dectin-1 [122]. In summary, dendritic cells in the lung encompass various subsets that perform specialized functions, impacting immune responses to pathogens/allergens and their involvement in disease pathogenesis, thereby highlighting their significance in pulmonary immunology [123, 124].

Immunological barriers by innate lymphoid cells

Innate lymphoid cells (ILCs) are a group of innate immune cells that are primarily involved in defending the body against infections, particularly at mucosal surfaces like the lungs, gut, and skin. ILCs lack specific antigen-specific receptors like T cell receptors (TCRs) or B cell receptors (BCRs), leading to their ability to respond quickly to stimuli, particularly cytokines. ILCs can be classified into three distinct functional groups, namely ILC1s, ILC2s, and ILC3, similar to the classification of CD4+ T cells, with each corresponding to Th1, Th2, and Th17 cells, respectively [125].

ILC1s

ILC1s are characterized by the expression of T-box transcription factor 21 and the production of IFN-γ, a key cytokine involved in the defense against intracellular pathogens. They share some functional similarities with Th1 cells, which also produce IFN-γ. In mice infected with the H1N1 influenza virus, ILC1s are activated and respond quickly to release IFN-γ and TNF-α [126]. Additionally, during the Sendai virus infection, ILC1s were identified as the primary source of IFN-γ production in the early phase [127]. Although the roles of ILC1s in the lung are not fully understood, the increased percentage of ILC1s in the blood of COPD patients suggests that ILC1s are involved in the pathogenesis of COPD [128].

ILC2s

The master regulator of ILC2s is a transcription factor called GATA-binding protein-3 (GATA3), similar to Th2 cells. Neuropilin-1 (NRP1), which is induced postnatally and sustained by lung-derived transforming growth factor beta-1 (TGF-β1), has been reported to be a tissue-specific marker of lung ILC2s. ILC2s promote pulmonary inflammation by secreting type 2 cytokines, including IL-5, IL-13, and IL-4, in response to epithelial-derived alarming cytokines such as IL-33, TSLP, and IL-25 (Fig. 3) [129]. Many reports indicate that ILC2s have a pathogenic role in allergic inflammation [130]. In a mouse model of asthma, Th2 cells and ILC2s are the main sources of IL-5 and IL-13 production in the lung. In addition to allergic inflammation, ILC2s are also involved in tissue repair through amphiregulin production. Recently, the relationship between ILC2s and neural cells is an emerging area of research that has revealed intriguing connections between the immune and nervous systems [10]. Neuropeptides, which are signaling molecules produced by neurons, can influence the activity of ILC2s. For instance, vasoactive intestinal peptide (VIP) can directly activate ILC2s, leading to their production of IL-5. Then, IL-5 released from ILC2s stimulates neurons to produce more VIP, establishing a positive feedback loop [11]. Moreover, neuromedin U (NMU), produced by mucosal neurons, has been shown to activate ILC2s [12]. Upon binding to its receptor (neuromedin U receptor 1) on ILC2s, NMU could stimulate ILC2s to produce cytokines. This relationship between NMU and ILC2s has been reported not only in allergic airway inflammation but also in respiratory syncytial virus (RSV) infection. In response to RSV, pulmonary neurons secrete NMU in a TLR4- and TLR7-dependent manner, activating ILC2s via NMU and thus exacerbating airway inflammation [13]. In addition, it has been reported that calcitonin gene–related peptide (CGRP) produced by pulmonary neuroendocrine cells (PNECs) promotes cytokine production from ILC2s [14].

ILC3s

ILC3s are characterized by the expression of retinoic acid receptor-related orphan receptor γt (RORγt) and release IL-17, IL-22, and GM-CSF in response to IL-23 and IL-1b. They are the predominant ILC subpopulation in human lungs, whereas ILC2s are the most abundant in murine lungs. Through the production of IL-22 and IL-17, ILC3s are involved in defense against various pathogens. In addition, they are the main producers of IL-22 in the lung and are essential for maintaining epithelial homeostasis and tissue repair. On the other hand, it has been reported that ILC3s act pathogenically in obesity-related asthma, suggesting diverse roles for ILC3s in the lungs [131, 132].

Concluding remarks

The importance of epithelium as a barrier mechanism in the lungs has been well established. Their central role was thought to be a physical mechanism through cell adhesion and a protective mechanism through mucus production. Indeed, it has been demonstrated that dysfunction of these barrier mechanisms is involved in many pathological conditions, suggesting their importance. Recently, the role of AECs as initiators of immune responses has been recognized. Furthermore, the close relationship between epithelium, neurons, and immune cells is becoming apparent. Further studies are needed to determine the physiological role of this crosstalk and its involvement in the pathogenesis of various lung diseases.

Declarations

Conflict of interest

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
2.
Zurück zum Zitat Hallstrand TS, Hackett TL, Altemeier WA, Matute-Bello G, Hansbro PM, Knight DA (2014) Airway epithelial regulation of pulmonary immune homeostasis and inflammation. Clin Immunol 151(1):1–15PubMedCrossRef Hallstrand TS, Hackett TL, Altemeier WA, Matute-Bello G, Hansbro PM, Knight DA (2014) Airway epithelial regulation of pulmonary immune homeostasis and inflammation. Clin Immunol 151(1):1–15PubMedCrossRef
3.
Zurück zum Zitat Saku A, Hirose K, Ito T, Iwata A, Sato T et al (2019) Fucosyltransferase 2 induces lung epithelial fucosylation and exacerbates house dust mite-induced airway inflammation. J Allergy Clin Immunol 144(3):698-709.e9PubMedCrossRef Saku A, Hirose K, Ito T, Iwata A, Sato T et al (2019) Fucosyltransferase 2 induces lung epithelial fucosylation and exacerbates house dust mite-induced airway inflammation. J Allergy Clin Immunol 144(3):698-709.e9PubMedCrossRef
4.
Zurück zum Zitat Nishimura N, Yokota M, Kurihara S, Iwata A, Kageyama T et al (2022) Airway epithelial STAT3 inhibits allergic inflammation via upregulation of stearoyl-CoA desaturase 1. Allergol Int 71(4):520–527PubMedCrossRef Nishimura N, Yokota M, Kurihara S, Iwata A, Kageyama T et al (2022) Airway epithelial STAT3 inhibits allergic inflammation via upregulation of stearoyl-CoA desaturase 1. Allergol Int 71(4):520–527PubMedCrossRef
5.
Zurück zum Zitat Ordovas-Montanes J, Dwyer DF, Nyquist SK, Buchheit KM, Vukovic M et al (2018) Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 560(7720):649–654PubMedPubMedCentralCrossRef Ordovas-Montanes J, Dwyer DF, Nyquist SK, Buchheit KM, Vukovic M et al (2018) Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 560(7720):649–654PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Ravanetti L, Dijkhuis A, Dekker T, Sabogal Pineros YS, Ravi A et al (2019) IL-33 drives influenza-induced asthma exacerbations by halting innate and adaptive antiviral immunity. J Allergy Clin Immunol 143(4):1355-1370.e16PubMedCrossRef Ravanetti L, Dijkhuis A, Dekker T, Sabogal Pineros YS, Ravi A et al (2019) IL-33 drives influenza-induced asthma exacerbations by halting innate and adaptive antiviral immunity. J Allergy Clin Immunol 143(4):1355-1370.e16PubMedCrossRef
7.
Zurück zum Zitat Veerati PC, Troy NM, Reid AT, Li NF, Nichol KS et al (2020) Airway epithelial cell immunity is delayed during rhinovirus infection in asthma and COPD. Front Immunol 11:974PubMedPubMedCentralCrossRef Veerati PC, Troy NM, Reid AT, Li NF, Nichol KS et al (2020) Airway epithelial cell immunity is delayed during rhinovirus infection in asthma and COPD. Front Immunol 11:974PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Doni Jayavelu N, Altman MC, Benson B, Dufort MJ, Vanderwall ER et al (2023) Type 2 inflammation reduces SARS-CoV-2 replication in the airway epithelium in allergic asthma through functional alteration of ciliated epithelial cells. J Allergy Clin Immunol 152(1):56–67PubMedCrossRef Doni Jayavelu N, Altman MC, Benson B, Dufort MJ, Vanderwall ER et al (2023) Type 2 inflammation reduces SARS-CoV-2 replication in the airway epithelium in allergic asthma through functional alteration of ciliated epithelial cells. J Allergy Clin Immunol 152(1):56–67PubMedCrossRef
9.
Zurück zum Zitat Corren J, Parnes JR, Wang L, Mo M, Roseti SL et al (2017) Tezepelumab in adults with uncontrolled asthma. N Engl J Med 377(10):936–946PubMedCrossRef Corren J, Parnes JR, Wang L, Mo M, Roseti SL et al (2017) Tezepelumab in adults with uncontrolled asthma. N Engl J Med 377(10):936–946PubMedCrossRef
10.
Zurück zum Zitat Yin Z, Zhou Y, Turnquist HR, Liu Q (2022) Neuro-epithelial-ILC2 crosstalk in barrier tissues. Trends Immunol 43(11):901–916PubMedCrossRef Yin Z, Zhou Y, Turnquist HR, Liu Q (2022) Neuro-epithelial-ILC2 crosstalk in barrier tissues. Trends Immunol 43(11):901–916PubMedCrossRef
11.
Zurück zum Zitat Talbot S, Abdulnour R-EE, Burkett PR, Lee S, Cronin SJF et al (2015) Silencing nociceptor neurons reduces allergic airway inflammation. Neuron 87(2):341–354PubMedPubMedCentralCrossRef Talbot S, Abdulnour R-EE, Burkett PR, Lee S, Cronin SJF et al (2015) Silencing nociceptor neurons reduces allergic airway inflammation. Neuron 87(2):341–354PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour R-EE, Nyman J et al (2017) The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549(7672):351–356PubMedPubMedCentralCrossRef Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour R-EE, Nyman J et al (2017) The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549(7672):351–356PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Liu W, Wang S, Wang J, Zheng R, Wang D et al (2023) Neuromedin U induces pulmonary ILC2 activation via the NMUR1 pathway during acute respiratory syncytial virus infection. Am J Respir Cell Mol Biol 68(3):256–266PubMedCrossRef Liu W, Wang S, Wang J, Zheng R, Wang D et al (2023) Neuromedin U induces pulmonary ILC2 activation via the NMUR1 pathway during acute respiratory syncytial virus infection. Am J Respir Cell Mol Biol 68(3):256–266PubMedCrossRef
14.
Zurück zum Zitat Sui P, Wiesner DL, Xu J, Zhang Y, Lee J et al (2018) Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 360:eaan8546 Sui P, Wiesner DL, Xu J, Zhang Y, Lee J et al (2018) Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 360:eaan8546
16.
Zurück zum Zitat Guillot L, Nathan N, Tabary O, Thouvenin G, Le Rouzic P et al (2013) Alveolar epithelial cells: master regulators of lung homeostasis. Int J Biochem Cell Biol 45(11):2568–2573PubMedCrossRef Guillot L, Nathan N, Tabary O, Thouvenin G, Le Rouzic P et al (2013) Alveolar epithelial cells: master regulators of lung homeostasis. Int J Biochem Cell Biol 45(11):2568–2573PubMedCrossRef
18.
Zurück zum Zitat Wittekindt OH (2017) Tight junctions in pulmonary epithelia during lung inflammation. Pflugers Arch 469(1):135–147PubMedCrossRef Wittekindt OH (2017) Tight junctions in pulmonary epithelia during lung inflammation. Pflugers Arch 469(1):135–147PubMedCrossRef
19.
Zurück zum Zitat Koval M (2013) Claudin heterogeneity and control of lung tight junctions. Annu Rev Physiol 75:551–567PubMedCrossRef Koval M (2013) Claudin heterogeneity and control of lung tight junctions. Annu Rev Physiol 75:551–567PubMedCrossRef
20.
Zurück zum Zitat Georas SN, Rezaee F (2014) Epithelial barrier function: at the front line of asthma immunology and allergic airway inflammation. J Allergy Clin Immunol 134(3):509–520PubMedPubMedCentralCrossRef Georas SN, Rezaee F (2014) Epithelial barrier function: at the front line of asthma immunology and allergic airway inflammation. J Allergy Clin Immunol 134(3):509–520PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Xiao C, Puddicombe SM, Field S, Haywood J, Broughton-Head V et al (2011) Defective epithelial barrier function in asthma. J Allergy Clin Immunol 128(3):549–556.e1-12 Xiao C, Puddicombe SM, Field S, Haywood J, Broughton-Head V et al (2011) Defective epithelial barrier function in asthma. J Allergy Clin Immunol 128(3):549–556.e1-12
22.
Zurück zum Zitat de Boer WI, Sharma HS, Baelemans SMI, Hoogsteden HC, Lambrecht BN, Braunstahl GJ (2008) Altered expression of epithelial junctional proteins in atopic asthma: possible role in inflammation. Can J Physiol Pharmacol 86(3):105–112PubMedCrossRef de Boer WI, Sharma HS, Baelemans SMI, Hoogsteden HC, Lambrecht BN, Braunstahl GJ (2008) Altered expression of epithelial junctional proteins in atopic asthma: possible role in inflammation. Can J Physiol Pharmacol 86(3):105–112PubMedCrossRef
23.
Zurück zum Zitat Nishida K, Brune KA, Putcha N, Mandke P, O’Neal WK, et al. (2017) Cigarette smoke disrupts monolayer integrity by altering epithelial cell-cell adhesion and cortical tension. Am J Physiol Lung Cell Mol. Physiol. 313(3):L581–91 Nishida K, Brune KA, Putcha N, Mandke P, O’Neal WK, et al. (2017) Cigarette smoke disrupts monolayer integrity by altering epithelial cell-cell adhesion and cortical tension. Am J Physiol Lung Cell Mol. Physiol. 313(3):L581–91
24.
Zurück zum Zitat Rusznak C, Mills PR, Devalia JL, Sapsford RJ, Davies RJ, Lozewicz S (2000) Effect of cigarette smoke on the permeability and IL-1beta and sICAM-1 release from cultured human bronchial epithelial cells of never-smokers, smokers, and patients with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 23(4):530–536PubMedCrossRef Rusznak C, Mills PR, Devalia JL, Sapsford RJ, Davies RJ, Lozewicz S (2000) Effect of cigarette smoke on the permeability and IL-1beta and sICAM-1 release from cultured human bronchial epithelial cells of never-smokers, smokers, and patients with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 23(4):530–536PubMedCrossRef
25.
Zurück zum Zitat Thornton DJ, Rousseau K, McGuckin MA (2008) Structure and function of the polymeric mucins in airways mucus. Annu Rev Physiol 70:459–486PubMedCrossRef Thornton DJ, Rousseau K, McGuckin MA (2008) Structure and function of the polymeric mucins in airways mucus. Annu Rev Physiol 70:459–486PubMedCrossRef
26.
Zurück zum Zitat Radicioni G, Ceppe A, Ford AA, Alexis NE, Barr RG et al (2021) Airway mucin MUC5AC and MUC5B concentrations and the initiation and progression of chronic obstructive pulmonary disease: an analysis of the SPIROMICS cohort. Lancet Respir Med 9(11):1241–1254PubMedPubMedCentralCrossRef Radicioni G, Ceppe A, Ford AA, Alexis NE, Barr RG et al (2021) Airway mucin MUC5AC and MUC5B concentrations and the initiation and progression of chronic obstructive pulmonary disease: an analysis of the SPIROMICS cohort. Lancet Respir Med 9(11):1241–1254PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Henderson AG, Ehre C, Button B, Abdullah LH, Cai L-H et al (2014) Cystic fibrosis airway secretions exhibit mucin hyperconcentration and increased osmotic pressure. J Clin Invest 124(7):3047–3060PubMedPubMedCentralCrossRef Henderson AG, Ehre C, Button B, Abdullah LH, Cai L-H et al (2014) Cystic fibrosis airway secretions exhibit mucin hyperconcentration and increased osmotic pressure. J Clin Invest 124(7):3047–3060PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Henke MO, John G, Germann M, Lindemann H, Rubin BK (2007) MUC5AC and MUC5B mucins increase in cystic fibrosis airway secretions during pulmonary exacerbation. Am J Respir Crit Care Med 175(8):816–821PubMedCrossRef Henke MO, John G, Germann M, Lindemann H, Rubin BK (2007) MUC5AC and MUC5B mucins increase in cystic fibrosis airway secretions during pulmonary exacerbation. Am J Respir Crit Care Med 175(8):816–821PubMedCrossRef
29.
Zurück zum Zitat Lachowicz-Scroggins ME, Yuan S, Kerr SC, Dunican EM, Yu M et al (2016) Abnormalities in MUC5AC and MUC5B protein in airway mucus in asthma. Am J Respir Crit Care Med 194(10):1296–1299PubMedPubMedCentralCrossRef Lachowicz-Scroggins ME, Yuan S, Kerr SC, Dunican EM, Yu M et al (2016) Abnormalities in MUC5AC and MUC5B protein in airway mucus in asthma. Am J Respir Crit Care Med 194(10):1296–1299PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Roy MG, Livraghi-Butrico A, Fletcher AA, McElwee MM, Evans SE et al (2014) Muc5b is required for airway defence. Nature 505(7483):412–416PubMedCrossRef Roy MG, Livraghi-Butrico A, Fletcher AA, McElwee MM, Evans SE et al (2014) Muc5b is required for airway defence. Nature 505(7483):412–416PubMedCrossRef
31.
32.
Zurück zum Zitat Singh G, Acharya S, Shukla S, Jain D (2023) Muco-obstructive lung disease: a systematic review. Cureus 15(10):e46866PubMedPubMedCentral Singh G, Acharya S, Shukla S, Jain D (2023) Muco-obstructive lung disease: a systematic review. Cureus 15(10):e46866PubMedPubMedCentral
33.
Zurück zum Zitat Renegar KB, Small PA Jr, Boykins LG, Wright PF (2004) Role of IgA versus IgG in the control of influenza viral infection in the murine respiratory tract. J Immunol 173(3):1978–1986PubMedCrossRef Renegar KB, Small PA Jr, Boykins LG, Wright PF (2004) Role of IgA versus IgG in the control of influenza viral infection in the murine respiratory tract. J Immunol 173(3):1978–1986PubMedCrossRef
34.
Zurück zum Zitat Oh JE, Song E, Moriyama M, Wong P, Zhang S et al (2021) Intranasal priming induces local lung-resident B cell populations that secrete protective mucosal antiviral IgA. Sci Immunol 6(66):eabj5129PubMedPubMedCentralCrossRef Oh JE, Song E, Moriyama M, Wong P, Zhang S et al (2021) Intranasal priming induces local lung-resident B cell populations that secrete protective mucosal antiviral IgA. Sci Immunol 6(66):eabj5129PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Bohländer F (2023) A new hope? Possibilities of therapeutic IgA antibodies in the treatment of inflammatory lung diseases. Front Immunol 14:1127339PubMedPubMedCentralCrossRef Bohländer F (2023) A new hope? Possibilities of therapeutic IgA antibodies in the treatment of inflammatory lung diseases. Front Immunol 14:1127339PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Blackburn JB, Schaff JA, Gutor S, Du R-H, Nichols D et al (2022) Secretory cells are the primary source of pIgR in small airways. Am J Respir Cell Mol Biol 67(3):334–345PubMedPubMedCentralCrossRef Blackburn JB, Schaff JA, Gutor S, Du R-H, Nichols D et al (2022) Secretory cells are the primary source of pIgR in small airways. Am J Respir Cell Mol Biol 67(3):334–345PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Diebel LN, Amin PB, Liberati DM (2010) Sequence of immunoglobulin isotype exposure modulates inflammatory response to bacteria and lipopolysaccharide in vitro. Surg Infect 11(2):145–150CrossRef Diebel LN, Amin PB, Liberati DM (2010) Sequence of immunoglobulin isotype exposure modulates inflammatory response to bacteria and lipopolysaccharide in vitro. Surg Infect 11(2):145–150CrossRef
38.
Zurück zum Zitat Ladjemi MZ, Martin C, Lecocq M, Detry B, Nana FA et al (2019) Increased IgA expression in lung lymphoid follicles in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med 199(5):592–602PubMedCrossRef Ladjemi MZ, Martin C, Lecocq M, Detry B, Nana FA et al (2019) Increased IgA expression in lung lymphoid follicles in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med 199(5):592–602PubMedCrossRef
39.
Zurück zum Zitat Ladjemi MZ, Gras D, Dupasquier S, Detry B, Lecocq M, et al. (2018) Bronchial epithelial IgA secretion is impaired in asthma. Role of IL-4/IL-13. Am J Respir Crit Care Med 197(11):1396–1409 Ladjemi MZ, Gras D, Dupasquier S, Detry B, Lecocq M, et al. (2018) Bronchial epithelial IgA secretion is impaired in asthma. Role of IL-4/IL-13. Am J Respir Crit Care Med 197(11):1396–1409
40.
Zurück zum Zitat Sinnberg T, Lichtensteiger C, Ali OH, Pop OT, Jochum A-K et al (2023) Pulmonary surfactant proteins are inhibited by immunoglobulin A autoantibodies in severe COVID-19. Am J Respir Crit Care Med 207(1):38–49PubMedCrossRef Sinnberg T, Lichtensteiger C, Ali OH, Pop OT, Jochum A-K et al (2023) Pulmonary surfactant proteins are inhibited by immunoglobulin A autoantibodies in severe COVID-19. Am J Respir Crit Care Med 207(1):38–49PubMedCrossRef
41.
Zurück zum Zitat Plasschaert LW, Žilionis R, Choo-Wing R, Savova V, Knehr J et al (2018) A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560(7718):377–381PubMedPubMedCentralCrossRef Plasschaert LW, Žilionis R, Choo-Wing R, Savova V, Knehr J et al (2018) A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560(7718):377–381PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Kathiriya JJ, Brumwell AN, Jackson JR, Tang X, Chapman HA (2020) Distinct airway epithelial stem cells hide among club cells but mobilize to promote alveolar regeneration. Cell Stem Cell 26(3):346-358.e4PubMedPubMedCentralCrossRef Kathiriya JJ, Brumwell AN, Jackson JR, Tang X, Chapman HA (2020) Distinct airway epithelial stem cells hide among club cells but mobilize to promote alveolar regeneration. Cell Stem Cell 26(3):346-358.e4PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Kadur Lakshminarasimha Murthy P, Sontake V, Tata A, Kobayashi Y, Macadlo L et al (2022) Human distal lung maps and lineage hierarchies reveal a bipotent progenitor. Nature 604(7904):111–119PubMedPubMedCentralCrossRef Kadur Lakshminarasimha Murthy P, Sontake V, Tata A, Kobayashi Y, Macadlo L et al (2022) Human distal lung maps and lineage hierarchies reveal a bipotent progenitor. Nature 604(7904):111–119PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Habermann AC, Gutierrez AJ, Bui LT, Yahn SL, Winters NI et al (2020) Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci Adv. 6(28):eaba1972PubMedPubMedCentralCrossRef Habermann AC, Gutierrez AJ, Bui LT, Yahn SL, Winters NI et al (2020) Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci Adv. 6(28):eaba1972PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B et al (2018) A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560(7718):319–324PubMedPubMedCentralCrossRef Montoro DT, Haber AL, Biton M, Vinarsky V, Lin B et al (2018) A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560(7718):319–324PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Ryan DM, Vincent TL, Salit J, Walters MS, Agosto-Perez F et al (2014) Smoking dysregulates the human airway basal cell transcriptome at COPD risk locus 19q13.2. PLoS One. 9(2):e88051PubMedPubMedCentralCrossRef Ryan DM, Vincent TL, Salit J, Walters MS, Agosto-Perez F et al (2014) Smoking dysregulates the human airway basal cell transcriptome at COPD risk locus 19q13.2. PLoS One. 9(2):e88051PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Fujino N, Brand OJ, Morgan DJ, Fujimori T, Grabiec AM et al (2019) Sensing of apoptotic cells through Axl causes lung basal cell proliferation in inflammatory diseases. J Exp Med 216(9):2184–2201PubMedPubMedCentralCrossRef Fujino N, Brand OJ, Morgan DJ, Fujimori T, Grabiec AM et al (2019) Sensing of apoptotic cells through Axl causes lung basal cell proliferation in inflammatory diseases. J Exp Med 216(9):2184–2201PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N et al (2020) Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci Adv. 6(28):eaba1983PubMedPubMedCentralCrossRef Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N et al (2020) Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci Adv. 6(28):eaba1983PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Wang S, Rao W, Hoffman A, Lin J, Li J et al (2023) Cloning a profibrotic stem cell variant in idiopathic pulmonary fibrosis. Sci. Transl. Med. 15(693):eab9528CrossRef Wang S, Rao W, Hoffman A, Lin J, Li J et al (2023) Cloning a profibrotic stem cell variant in idiopathic pulmonary fibrosis. Sci. Transl. Med. 15(693):eab9528CrossRef
50.
Zurück zum Zitat Wang X, Hallen NR, Lee M, Samuchiwal S, Ye Q et al (2023) Type 2 inflammation drives an airway basal stem cell program through insulin receptor substrate signaling. J Allergy Clin Immunol 151(6):1536–1549PubMedPubMedCentralCrossRef Wang X, Hallen NR, Lee M, Samuchiwal S, Ye Q et al (2023) Type 2 inflammation drives an airway basal stem cell program through insulin receptor substrate signaling. J Allergy Clin Immunol 151(6):1536–1549PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K et al (2019) A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med 25(7):1153–1163PubMedCrossRef Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K et al (2019) A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med 25(7):1153–1163PubMedCrossRef
52.
Zurück zum Zitat Ma Q, Ma Y, Dai X, Ren T, Fu Y et al (2018) Regeneration of functional alveoli by adult human SOX9+ airway basal cell transplantation. Protein Cell 9(3):267–282PubMedPubMedCentralCrossRef Ma Q, Ma Y, Dai X, Ren T, Fu Y et al (2018) Regeneration of functional alveoli by adult human SOX9+ airway basal cell transplantation. Protein Cell 9(3):267–282PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Vaidyanathan S, Salahudeen AA, Sellers ZM, Bravo DT, Choi SS et al (2020) High-efficiency, selection-free gene repair in airway stem cells from cystic fibrosis patients rescues CFTR function in differentiated epithelia. Cell Stem Cell 26(2):161-171.e4PubMedCrossRef Vaidyanathan S, Salahudeen AA, Sellers ZM, Bravo DT, Choi SS et al (2020) High-efficiency, selection-free gene repair in airway stem cells from cystic fibrosis patients rescues CFTR function in differentiated epithelia. Cell Stem Cell 26(2):161-171.e4PubMedCrossRef
54.
Zurück zum Zitat Tsao P-N, Vasconcelos M, Izvolsky KI, Qian J, Lu J, Cardoso WV (2009) Notch signaling controls the balance of ciliated and secretory cell fates in developing airways. Development 136(13):2297–2307PubMedPubMedCentralCrossRef Tsao P-N, Vasconcelos M, Izvolsky KI, Qian J, Lu J, Cardoso WV (2009) Notch signaling controls the balance of ciliated and secretory cell fates in developing airways. Development 136(13):2297–2307PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Everman JL, Sajuthi S, Saef B, Rios C, Stoner AM et al (2019) Functional genomics of CDHR3 confirms its role in HRV-C infection and childhood asthma exacerbations. J Allergy Clin Immunol 144(4):962–971PubMedPubMedCentralCrossRef Everman JL, Sajuthi S, Saef B, Rios C, Stoner AM et al (2019) Functional genomics of CDHR3 confirms its role in HRV-C infection and childhood asthma exacerbations. J Allergy Clin Immunol 144(4):962–971PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Bochkov YA, Watters K, Ashraf S, Griggs TF, Devries MK et al (2015) Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U S A 112(17):5485–5490PubMedPubMedCentralCrossRef Bochkov YA, Watters K, Ashraf S, Griggs TF, Devries MK et al (2015) Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U S A 112(17):5485–5490PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Griggs TF, Bochkov YA, Basnet S, Pasic TR, Brockman-Schneider RA et al (2017) Rhinovirus C targets ciliated airway epithelial cells. Respir Res 18(1):84PubMedPubMedCentralCrossRef Griggs TF, Bochkov YA, Basnet S, Pasic TR, Brockman-Schneider RA et al (2017) Rhinovirus C targets ciliated airway epithelial cells. Respir Res 18(1):84PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Basnet S, Bochkov YA, Brockman-Schneider RA, Kuipers I, Aesif SW et al (2019) CDHR3 asthma-risk genotype affects susceptibility of airway epithelium to rhinovirus C infections. Am J Respir Cell Mol Biol 61(4):450–458PubMedPubMedCentralCrossRef Basnet S, Bochkov YA, Brockman-Schneider RA, Kuipers I, Aesif SW et al (2019) CDHR3 asthma-risk genotype affects susceptibility of airway epithelium to rhinovirus C infections. Am J Respir Cell Mol Biol 61(4):450–458PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Tokita E, Tanabe T, Asano K, Suzaki H, Rubin BK (2014) Club cell 10-kDa protein attenuates airway mucus hypersecretion and inflammation. Eur Respir J 44(4):1002–1010PubMedCrossRef Tokita E, Tanabe T, Asano K, Suzaki H, Rubin BK (2014) Club cell 10-kDa protein attenuates airway mucus hypersecretion and inflammation. Eur Respir J 44(4):1002–1010PubMedCrossRef
60.
Zurück zum Zitat Gamez AS, Gras D, Petit A, Knabe L, Molinari N et al (2015) Supplementing defect in club cell secretory protein attenuates airway inflammation in COPD. Chest 147(6):1467–1476PubMedCrossRef Gamez AS, Gras D, Petit A, Knabe L, Molinari N et al (2015) Supplementing defect in club cell secretory protein attenuates airway inflammation in COPD. Chest 147(6):1467–1476PubMedCrossRef
61.
Zurück zum Zitat Laucho-Contreras ME, Polverino F, Gupta K, Taylor KL, Kelly E et al (2015) Protective role for club cell secretory protein-16 (CC16) in the development of COPD. Eur Respir J 45(6):1544–1556PubMedPubMedCentralCrossRef Laucho-Contreras ME, Polverino F, Gupta K, Taylor KL, Kelly E et al (2015) Protective role for club cell secretory protein-16 (CC16) in the development of COPD. Eur Respir J 45(6):1544–1556PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Guerra S, Vasquez MM, Spangenberg A, Halonen M, Martin RJ (2016) Club cell secretory protein in serum and bronchoalveolar lavage of patients with asthma. J Allergy Clin Immunol 138(3):932-934.e1PubMedPubMedCentralCrossRef Guerra S, Vasquez MM, Spangenberg A, Halonen M, Martin RJ (2016) Club cell secretory protein in serum and bronchoalveolar lavage of patients with asthma. J Allergy Clin Immunol 138(3):932-934.e1PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Kraft M, Ledford J, Gozdz J, Li X, Francisco D et al (2018) Club cell protein-16 modifies airway inflammation in asthma and is associated with significant clinical asthma outcomes. Eur Respir J 52(Suppl. 62):PA5496 Kraft M, Ledford J, Gozdz J, Li X, Francisco D et al (2018) Club cell protein-16 modifies airway inflammation in asthma and is associated with significant clinical asthma outcomes. Eur Respir J 52(Suppl. 62):PA5496
64.
Zurück zum Zitat Zheng D, Yin L, Chen J (2014) Evidence for Scgb1a1(+) cells in the generation of p63(+) cells in the damaged lung parenchyma. Am J Respir Cell Mol Biol 50(3):595–604PubMedCrossRef Zheng D, Yin L, Chen J (2014) Evidence for Scgb1a1(+) cells in the generation of p63(+) cells in the damaged lung parenchyma. Am J Respir Cell Mol Biol 50(3):595–604PubMedCrossRef
65.
Zurück zum Zitat Chen Y, Toth R, Chocarro S, Weichenhan D, Hey J et al (2022) Club cells employ regeneration mechanisms during lung tumorigenesis. Nat Commun 13(1):4557PubMedPubMedCentralCrossRef Chen Y, Toth R, Chocarro S, Weichenhan D, Hey J et al (2022) Club cells employ regeneration mechanisms during lung tumorigenesis. Nat Commun 13(1):4557PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Ordoñez CL, Khashayar R, Wong HH, Ferrando R, Wu R et al (2001) Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am J Respir Crit Care Med 163(2):517–523PubMedCrossRef Ordoñez CL, Khashayar R, Wong HH, Ferrando R, Wu R et al (2001) Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am J Respir Crit Care Med 163(2):517–523PubMedCrossRef
67.
Zurück zum Zitat Kim V, Oros M, Durra H, Kelsen S, Aksoy M et al (2015) Chronic bronchitis and current smoking are associated with more goblet cells in moderate to severe COPD and smokers without airflow obstruction. PLoS ONE 10(2):e0116108PubMedPubMedCentralCrossRef Kim V, Oros M, Durra H, Kelsen S, Aksoy M et al (2015) Chronic bronchitis and current smoking are associated with more goblet cells in moderate to severe COPD and smokers without airflow obstruction. PLoS ONE 10(2):e0116108PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP et al (1999) Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest 103(6):779–788PubMedPubMedCentralCrossRef Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP et al (1999) Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest 103(6):779–788PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Pezzulo AA, Tudas RA, Stewart CG, Buonfiglio LGV, Lindsay BD et al (2019) HSP90 inhibitor geldanamycin reverts IL-13- and IL-17-induced airway goblet cell metaplasia. J Clin Invest 129(2):744–758PubMedPubMedCentralCrossRef Pezzulo AA, Tudas RA, Stewart CG, Buonfiglio LGV, Lindsay BD et al (2019) HSP90 inhibitor geldanamycin reverts IL-13- and IL-17-induced airway goblet cell metaplasia. J Clin Invest 129(2):744–758PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Xu J, Yu H, Sun X (2020) Less is more: rare pulmonary neuroendocrine cells function as critical sensors in lung. Dev Cell 55(2):123–132PubMedCrossRef Xu J, Yu H, Sun X (2020) Less is more: rare pulmonary neuroendocrine cells function as critical sensors in lung. Dev Cell 55(2):123–132PubMedCrossRef
71.
Zurück zum Zitat Wallrapp A, Burkett PR, Riesenfeld SJ, Kim S-J, Christian E et al (2019) Calcitonin gene-related peptide negatively regulates alarmin-driven type 2 innate lymphoid cell responses. Immunity 51(4):709-723.e6PubMedPubMedCentralCrossRef Wallrapp A, Burkett PR, Riesenfeld SJ, Kim S-J, Christian E et al (2019) Calcitonin gene-related peptide negatively regulates alarmin-driven type 2 innate lymphoid cell responses. Immunity 51(4):709-723.e6PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Kotas ME, Moore CM, Gurrola JG 2nd, Pletcher SD, Goldberg AN et al (2022) IL-13- programmed airway tuft cells produce PGE2, which promotes CFTR-dependent mucociliary function. JCI Insight 7(13):e159832 Kotas ME, Moore CM, Gurrola JG 2nd, Pletcher SD, Goldberg AN et al (2022) IL-13- programmed airway tuft cells produce PGE2, which promotes CFTR-dependent mucociliary function. JCI Insight 7(13):e159832
73.
Zurück zum Zitat Bankova LG, Dwyer DF, Yoshimoto E, Ualiyeva S, McGinty JW et al (2018) The cysteinyl leukotriene 3 receptor regulates expansion of IL-25-producing airway brush cells leading to type 2 inflammation. Sci Immunol 3(28):eaat9453 Bankova LG, Dwyer DF, Yoshimoto E, Ualiyeva S, McGinty JW et al (2018) The cysteinyl leukotriene 3 receptor regulates expansion of IL-25-producing airway brush cells leading to type 2 inflammation. Sci Immunol 3(28):eaat9453
74.
Zurück zum Zitat Ualiyeva S, Lemire E, Aviles EC, Wong C, Boyd AA et al (2021) Tuft cell-produced cysteinyl leukotrienes and IL-25 synergistically initiate lung type 2 inflammation. Sci Immunol. 6(66):eabj0474PubMedPubMedCentralCrossRef Ualiyeva S, Lemire E, Aviles EC, Wong C, Boyd AA et al (2021) Tuft cell-produced cysteinyl leukotrienes and IL-25 synergistically initiate lung type 2 inflammation. Sci Immunol. 6(66):eabj0474PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Allan KM, Farrow N, Donnelley M, Jaffe A, Waters SA (2021) Treatment of cystic fibrosis: from gene- to cell-based therapies. Front Pharmacol 12:639475PubMedPubMedCentralCrossRef Allan KM, Farrow N, Donnelley M, Jaffe A, Waters SA (2021) Treatment of cystic fibrosis: from gene- to cell-based therapies. Front Pharmacol 12:639475PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Okuda K, Dang H, Kobayashi Y, Carraro G, Nakano S et al (2021) Secretory cells dominate airway CFTR expression and function in human airway superficial epithelia. Am J Respir Crit Care Med 203(10):1275–1289PubMedPubMedCentralCrossRef Okuda K, Dang H, Kobayashi Y, Carraro G, Nakano S et al (2021) Secretory cells dominate airway CFTR expression and function in human airway superficial epithelia. Am J Respir Crit Care Med 203(10):1275–1289PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Lei L, Traore S, Romano Ibarra GS, Karp PH, Rehman T et al (2023) CFTR-rich ionocytes mediate chloride absorption across airway epithelia. J Clin Invest 133(20):e171268 Lei L, Traore S, Romano Ibarra GS, Karp PH, Rehman T et al (2023) CFTR-rich ionocytes mediate chloride absorption across airway epithelia. J Clin Invest 133(20):e171268
78.
Zurück zum Zitat Yuan F, Gasser GN, Lemire E, Montoro DT, Jagadeesh K et al (2023) Transgenic ferret models define pulmonary ionocyte diversity and function. Nature 621(7980):857–867PubMedPubMedCentralCrossRef Yuan F, Gasser GN, Lemire E, Montoro DT, Jagadeesh K et al (2023) Transgenic ferret models define pulmonary ionocyte diversity and function. Nature 621(7980):857–867PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR et al (2013) Type 2 alveolar cells are stem cells in adult lung. J Clin Invest 123(7):3025–3036PubMedPubMedCentralCrossRef Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR et al (2013) Type 2 alveolar cells are stem cells in adult lung. J Clin Invest 123(7):3025–3036PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Nabhan AN, Webster JD, Adams JJ, Blazer L, Everrett C et al (2023) Targeted alveolar regeneration with Frizzled-specific agonists. Cell 186(14):2995-3012.e15PubMedCrossRef Nabhan AN, Webster JD, Adams JJ, Blazer L, Everrett C et al (2023) Targeted alveolar regeneration with Frizzled-specific agonists. Cell 186(14):2995-3012.e15PubMedCrossRef
81.
Zurück zum Zitat Zacharias WJ, Frank DB, Zepp JA, Morley MP, Alkhaleel FA et al (2018) Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555(7695):251–255PubMedPubMedCentralCrossRef Zacharias WJ, Frank DB, Zepp JA, Morley MP, Alkhaleel FA et al (2018) Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555(7695):251–255PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ (2018) Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 359(6380):1118–1123PubMedPubMedCentralCrossRef Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ (2018) Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 359(6380):1118–1123PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Cheng Z, Bartel S, Nijnatten JLV, Bults R, Brandsma C et al (2022) Transcriptome analysis indicates more AT2-to-AT1 transition in COPD. Eur Respir J 60(Suppl. 66):2503 Cheng Z, Bartel S, Nijnatten JLV, Bults R, Brandsma C et al (2022) Transcriptome analysis indicates more AT2-to-AT1 transition in COPD. Eur Respir J 60(Suppl. 66):2503
84.
Zurück zum Zitat Liang J, Huang G, Liu X, Taghavifar F, Liu N et al (2022) The ZIP8/SIRT1 axis regulates alveolar progenitor cell renewal in aging and idiopathic pulmonary fibrosis. J Clin Invest 132(11):e157338 Liang J, Huang G, Liu X, Taghavifar F, Liu N et al (2022) The ZIP8/SIRT1 axis regulates alveolar progenitor cell renewal in aging and idiopathic pulmonary fibrosis. J Clin Invest 132(11):e157338
85.
Zurück zum Zitat Enomoto Y, Katsura H, Fujimura T, Ogata A, Baba S et al (2023) Autocrine TGF-β-positive feedback in profibrotic AT2-lineage cells plays a crucial role in non-inflammatory lung fibrogenesis. Nat Commun 14(1):4956PubMedPubMedCentralCrossRef Enomoto Y, Katsura H, Fujimura T, Ogata A, Baba S et al (2023) Autocrine TGF-β-positive feedback in profibrotic AT2-lineage cells plays a crucial role in non-inflammatory lung fibrogenesis. Nat Commun 14(1):4956PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Basil MC, Cardenas-Diaz FL, Kathiriya JJ, Morley MP, Carl J et al (2022) Human distal airways contain a multipotent secretory cell that can regenerate alveoli. Nature 604(7904):120–126PubMedPubMedCentralCrossRef Basil MC, Cardenas-Diaz FL, Kathiriya JJ, Morley MP, Carl J et al (2022) Human distal airways contain a multipotent secretory cell that can regenerate alveoli. Nature 604(7904):120–126PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Leiva-Juárez MM, Kolls JK, Evans SE (2018) Lung epithelial cells: therapeutically inducible effectors of antimicrobial defense. Mucosal Immunol 11(1):21–34PubMedCrossRef Leiva-Juárez MM, Kolls JK, Evans SE (2018) Lung epithelial cells: therapeutically inducible effectors of antimicrobial defense. Mucosal Immunol 11(1):21–34PubMedCrossRef
89.
Zurück zum Zitat Takeuchi O, Akira S (2010) Pattern recognition receptors and inflammation. Cell 140(6):805–820PubMedCrossRef Takeuchi O, Akira S (2010) Pattern recognition receptors and inflammation. Cell 140(6):805–820PubMedCrossRef
90.
91.
Zurück zum Zitat Chou W-C, Jha S, Linhoff MW, Ting JP-Y (2023) The NLR gene family: from discovery to present day. Nat Rev Immunol 23(10):635–654PubMedCrossRef Chou W-C, Jha S, Linhoff MW, Ting JP-Y (2023) The NLR gene family: from discovery to present day. Nat Rev Immunol 23(10):635–654PubMedCrossRef
93.
Zurück zum Zitat Bermejo-Jambrina M, Eder J, Helgers LC, Hertoghs N, Nijmeijer BM et al (2018) C-type lectin receptors in antiviral immunity and viral escape. Front Immunol 9:590PubMedPubMedCentralCrossRef Bermejo-Jambrina M, Eder J, Helgers LC, Hertoghs N, Nijmeijer BM et al (2018) C-type lectin receptors in antiviral immunity and viral escape. Front Immunol 9:590PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Mata-Martínez P, Bergón-Gutiérrez M, Del Fresno C (2022) Dectin-1 signaling update: new perspectives for trained immunity. Front Immunol 13:812148PubMedPubMedCentralCrossRef Mata-Martínez P, Bergón-Gutiérrez M, Del Fresno C (2022) Dectin-1 signaling update: new perspectives for trained immunity. Front Immunol 13:812148PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D et al (2020) COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat Biotechnol 38(8):970–979PubMedCrossRef Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D et al (2020) COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat Biotechnol 38(8):970–979PubMedCrossRef
96.
Zurück zum Zitat Alladina J, Smith NP, Kooistra T, Slowikowski K, Kernin IJ et al (2023) A human model of asthma exacerbation reveals transcriptional programs and cell circuits specific to allergic asthma. Sci Immunol. 8(83):eabq6352PubMedPubMedCentralCrossRef Alladina J, Smith NP, Kooistra T, Slowikowski K, Kernin IJ et al (2023) A human model of asthma exacerbation reveals transcriptional programs and cell circuits specific to allergic asthma. Sci Immunol. 8(83):eabq6352PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Matkovic Leko I, Schneider RT, Thimraj TA, Schrode N, Beitler D et al (2023) A distal lung organoid model to study interstitial lung disease, viral infection and human lung development. Nat Protoc 18(7):2283–2312PubMedCrossRef Matkovic Leko I, Schneider RT, Thimraj TA, Schrode N, Beitler D et al (2023) A distal lung organoid model to study interstitial lung disease, viral infection and human lung development. Nat Protoc 18(7):2283–2312PubMedCrossRef
98.
Zurück zum Zitat Nikolić MZ, Caritg O, Jeng Q, Johnson J-A, Sun D et al (2017) Human embryonic lung epithelial tips are multipotent progenitors that can be expanded in vitro as long-term self- renewing organoids. Elife 6:e26575 Nikolić MZ, Caritg O, Jeng Q, Johnson J-A, Sun D et al (2017) Human embryonic lung epithelial tips are multipotent progenitors that can be expanded in vitro as long-term self- renewing organoids. Elife 6:e26575
99.
Zurück zum Zitat Barnes JL, Yoshida M, He P, Worlock KB, Lindeboom RGH et al (2023) Early human lung immune cell development and its role in epithelial cell fate. Sci Immunol. 8(90):eadf9988PubMedPubMedCentralCrossRef Barnes JL, Yoshida M, He P, Worlock KB, Lindeboom RGH et al (2023) Early human lung immune cell development and its role in epithelial cell fate. Sci Immunol. 8(90):eadf9988PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Lam M, Lamanna E, Organ L, Donovan C, Bourke JE (2023) Perspectives on precision cut lung slices-powerful tools for investigation of mechanisms and therapeutic targets in lung diseases. Front Pharmacol 14:1162889PubMedPubMedCentralCrossRef Lam M, Lamanna E, Organ L, Donovan C, Bourke JE (2023) Perspectives on precision cut lung slices-powerful tools for investigation of mechanisms and therapeutic targets in lung diseases. Front Pharmacol 14:1162889PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Lang NJ, Gote-Schniering J, Porras-Gonzalez D, Yang L, De Sadeleer LJ et al (2023) Ex vivo tissue perturbations coupled to single-cell RNA-seq reveal multilineage cell circuit dynamics in human lung fibrogenesis. Sci. Transl. Med. 15(725):eadh0908PubMedCrossRef Lang NJ, Gote-Schniering J, Porras-Gonzalez D, Yang L, De Sadeleer LJ et al (2023) Ex vivo tissue perturbations coupled to single-cell RNA-seq reveal multilineage cell circuit dynamics in human lung fibrogenesis. Sci. Transl. Med. 15(725):eadh0908PubMedCrossRef
102.
Zurück zum Zitat Bissonnette EY, Lauzon-Joset J-F, Debley JS, Ziegler SF (2020) Cross-talk between alveolar macrophages and lung epithelial cells is essential to maintain lung homeostasis. Front Immunol 11:583042PubMedPubMedCentralCrossRef Bissonnette EY, Lauzon-Joset J-F, Debley JS, Ziegler SF (2020) Cross-talk between alveolar macrophages and lung epithelial cells is essential to maintain lung homeostasis. Front Immunol 11:583042PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Rodriguez-Rodriguez L, Gillet L, Machiels B (2023) Shaping of the alveolar landscape by respiratory infections and long-term consequences for lung immunity. Front Immunol 14:1149015PubMedPubMedCentralCrossRef Rodriguez-Rodriguez L, Gillet L, Machiels B (2023) Shaping of the alveolar landscape by respiratory infections and long-term consequences for lung immunity. Front Immunol 14:1149015PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Gschwend J, Sherman SPM, Ridder F, Feng X, Liang H-E et al (2021) Alveolar macrophages rely on GM-CSF from alveolar epithelial type 2 cells before and after birth. J Exp Med 218(10):e20210745 Gschwend J, Sherman SPM, Ridder F, Feng X, Liang H-E et al (2021) Alveolar macrophages rely on GM-CSF from alveolar epithelial type 2 cells before and after birth. J Exp Med 218(10):e20210745
105.
Zurück zum Zitat Roquilly A, Jacqueline C, Davieau M, Mollé A, Sadek A et al (2020) Alveolar macrophages are epigenetically altered after inflammation, leading to long-term lung immunoparalysis. Nat Immunol 21(6):636–648PubMedCrossRef Roquilly A, Jacqueline C, Davieau M, Mollé A, Sadek A et al (2020) Alveolar macrophages are epigenetically altered after inflammation, leading to long-term lung immunoparalysis. Nat Immunol 21(6):636–648PubMedCrossRef
106.
Zurück zum Zitat Westphalen K, Gusarova GA, Islam MN, Subramanian M, Cohen TS et al (2014) Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity. Nature 506(7489):503–506PubMedPubMedCentralCrossRef Westphalen K, Gusarova GA, Islam MN, Subramanian M, Cohen TS et al (2014) Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity. Nature 506(7489):503–506PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Kitamura T, Tanaka N, Watanabe J, Uchida KS et al (1999) Idiopathic pulmonary alveolar proteinosis as an autoimmune disease with neutralizing antibody against granulocyte/macrophage colony-stimulating factor. J Exp Med 190(6):875–880PubMedPubMedCentralCrossRef Kitamura T, Tanaka N, Watanabe J, Uchida KS et al (1999) Idiopathic pulmonary alveolar proteinosis as an autoimmune disease with neutralizing antibody against granulocyte/macrophage colony-stimulating factor. J Exp Med 190(6):875–880PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Sakagami T, Uchida K, Suzuki T, Carey BC, Wood RE et al (2009) Human GM-CSF autoantibodies and reproduction of pulmonary alveolar proteinosis. N Engl J Med 361(27):2679–2681PubMedPubMedCentralCrossRef Sakagami T, Uchida K, Suzuki T, Carey BC, Wood RE et al (2009) Human GM-CSF autoantibodies and reproduction of pulmonary alveolar proteinosis. N Engl J Med 361(27):2679–2681PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Tazawa R, Ueda T, Abe M, Tatsumi K, Eda R et al (2019) Inhaled GM-CSF for pulmonary alveolar proteinosis. N Engl J Med 381(10):923–932PubMedCrossRef Tazawa R, Ueda T, Abe M, Tatsumi K, Eda R et al (2019) Inhaled GM-CSF for pulmonary alveolar proteinosis. N Engl J Med 381(10):923–932PubMedCrossRef
110.
Zurück zum Zitat Sabatel C, Radermecker C, Fievez L, Paulissen G, Chakarov S et al (2017) Exposure to bacterial CpG DNA protects from airway allergic inflammation by expanding regulatory lung interstitial macrophages. Immunity 46(3):457–473PubMedCrossRef Sabatel C, Radermecker C, Fievez L, Paulissen G, Chakarov S et al (2017) Exposure to bacterial CpG DNA protects from airway allergic inflammation by expanding regulatory lung interstitial macrophages. Immunity 46(3):457–473PubMedCrossRef
111.
Zurück zum Zitat Van Hulst G, Batugedara HM, Jorssen J, Louis R, Bureau F, Desmet CJ (2020) Eosinophil diversity in asthma. Biochem Pharmacol 179:113963PubMedCrossRef Van Hulst G, Batugedara HM, Jorssen J, Louis R, Bureau F, Desmet CJ (2020) Eosinophil diversity in asthma. Biochem Pharmacol 179:113963PubMedCrossRef
112.
113.
Zurück zum Zitat Pelaia C, Crimi C, Vatrella A, Tinello C, Terracciano R, Pelaia G (2020) Molecular targets for biological therapies of severe asthma. Front Immunol 11:603312PubMedPubMedCentralCrossRef Pelaia C, Crimi C, Vatrella A, Tinello C, Terracciano R, Pelaia G (2020) Molecular targets for biological therapies of severe asthma. Front Immunol 11:603312PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Andreev D, Liu M, Kachler K, Llerins Perez M, Kirchner P et al (2021) Regulatory eosinophils induce the resolution of experimental arthritis and appear in remission state of human rheumatoid arthritis. Ann Rheum Dis 80(4):451–468PubMedCrossRef Andreev D, Liu M, Kachler K, Llerins Perez M, Kirchner P et al (2021) Regulatory eosinophils induce the resolution of experimental arthritis and appear in remission state of human rheumatoid arthritis. Ann Rheum Dis 80(4):451–468PubMedCrossRef
115.
Zurück zum Zitat Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA et al (2016) Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 126(9):3279–3295PubMedPubMedCentralCrossRef Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA et al (2016) Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 126(9):3279–3295PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Kumar V (2020) Pulmonary innate immune response determines the outcome of inflammation during pneumonia and sepsis-associated acute lung injury. Front Immunol 11:1722PubMedPubMedCentralCrossRef Kumar V (2020) Pulmonary innate immune response determines the outcome of inflammation during pneumonia and sepsis-associated acute lung injury. Front Immunol 11:1722PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Crisford H, Sapey E, Rogers GB, Taylor S, Nagakumar P et al (2021) Neutrophils in asthma: the good, the bad and the bacteria. Thorax 76(8):835–844PubMedCrossRef Crisford H, Sapey E, Rogers GB, Taylor S, Nagakumar P et al (2021) Neutrophils in asthma: the good, the bad and the bacteria. Thorax 76(8):835–844PubMedCrossRef
118.
Zurück zum Zitat Kwok AJ, Allcock A, Ferreira RC, Cano-Gamez E, Smee M et al (2023) Neutrophils and emergency granulopoiesis drive immune suppression and an extreme response endotype during sepsis. Nat Immunol 24(5):767–779PubMedCrossRef Kwok AJ, Allcock A, Ferreira RC, Cano-Gamez E, Smee M et al (2023) Neutrophils and emergency granulopoiesis drive immune suppression and an extreme response endotype during sepsis. Nat Immunol 24(5):767–779PubMedCrossRef
120.
Zurück zum Zitat Soni C, Perez OA, Voss WN, Pucella JN, Serpas L et al (2020) Plasmacytoid dendritic cells and type I interferon promote extrafollicular B cell responses to extracellular self-DNA. Immunity 52(6):1022-1038.e7PubMedPubMedCentralCrossRef Soni C, Perez OA, Voss WN, Pucella JN, Serpas L et al (2020) Plasmacytoid dendritic cells and type I interferon promote extrafollicular B cell responses to extracellular self-DNA. Immunity 52(6):1022-1038.e7PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M et al (2005) Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 202(1):135–143PubMedPubMedCentralCrossRef Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M et al (2005) Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 202(1):135–143PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Ito T, Hirose K, Norimoto A, Tamachi T, Yokota M et al (2017) Dectin-1 plays an important role in house dust mite-induced allergic airway inflammation through the activation of CD11b+ dendritic cells. J Immunol 198(1):61–70PubMedCrossRef Ito T, Hirose K, Norimoto A, Tamachi T, Yokota M et al (2017) Dectin-1 plays an important role in house dust mite-induced allergic airway inflammation through the activation of CD11b+ dendritic cells. J Immunol 198(1):61–70PubMedCrossRef
123.
Zurück zum Zitat Mansouri S, Gogoi H, Pipkin M, Machuca TN, Emtiazjoo AM et al (2021) In vivo reprogramming of pathogenic lung TNFR2+ cDC2s by IFNβ inhibits HDM-induced asthma. Sci Immunol 6(61):eabi8472 Mansouri S, Gogoi H, Pipkin M, Machuca TN, Emtiazjoo AM et al (2021) In vivo reprogramming of pathogenic lung TNFR2+ cDC2s by IFNβ inhibits HDM-induced asthma. Sci Immunol 6(61):eabi8472
124.
Zurück zum Zitat Liu X, Netto KG, Sokulsky LA, Zhou L, Xu H et al (2022) Single-cell RNA transcriptomic analysis identifies Creb5 and CD11b-DCs as regulator of asthma exacerbations. Mucosal Immunol 15(6):1363–1374PubMedPubMedCentralCrossRef Liu X, Netto KG, Sokulsky LA, Zhou L, Xu H et al (2022) Single-cell RNA transcriptomic analysis identifies Creb5 and CD11b-DCs as regulator of asthma exacerbations. Mucosal Immunol 15(6):1363–1374PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P (2018) Influenza-activated ILC1s contribute to antiviral immunity partially influenced by differential GITR expression. Front Immunol 9:505PubMedPubMedCentralCrossRef Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P (2018) Influenza-activated ILC1s contribute to antiviral immunity partially influenced by differential GITR expression. Front Immunol 9:505PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Weizman O-E, Adams NM, Schuster IS, Krishna C, Pritykin Y et al (2017) ILC1 Confer early host protection at initial sites of viral infection. Cell 171(4):795-808.e12PubMedPubMedCentralCrossRef Weizman O-E, Adams NM, Schuster IS, Krishna C, Pritykin Y et al (2017) ILC1 Confer early host protection at initial sites of viral infection. Cell 171(4):795-808.e12PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Silver JS, Kearley J, Copenhaver AM, Sanden C, Mori M et al (2016) Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs. Nat Immunol 17(6):626–635PubMedPubMedCentralCrossRef Silver JS, Kearley J, Copenhaver AM, Sanden C, Mori M et al (2016) Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs. Nat Immunol 17(6):626–635PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Kabata H, Moro K, Koyasu S (2018) The group 2 innate lymphoid cell (ILC2) regulatory network and its underlying mechanisms. Immunol Rev 286(1):37–52PubMedCrossRef Kabata H, Moro K, Koyasu S (2018) The group 2 innate lymphoid cell (ILC2) regulatory network and its underlying mechanisms. Immunol Rev 286(1):37–52PubMedCrossRef
130.
Zurück zum Zitat Iwata A, Toda Y, Furuya H, Nakajima H (2023) Group 2 innate lymphoid cells in human asthma. Allergol Int 72(2):194–200PubMedCrossRef Iwata A, Toda Y, Furuya H, Nakajima H (2023) Group 2 innate lymphoid cells in human asthma. Allergol Int 72(2):194–200PubMedCrossRef
131.
Zurück zum Zitat Kim HY, Lee HJ, Chang Y-J, Pichavant M, Shore SA et al (2014) Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med 20(1):54–61PubMedCrossRef Kim HY, Lee HJ, Chang Y-J, Pichavant M, Shore SA et al (2014) Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med 20(1):54–61PubMedCrossRef
132.
Zurück zum Zitat Hekking P-P, Loza MJ, Pavlidis S, de Meulder B, Lefaudeux D et al (2018) Pathway discovery using transcriptomic profiles in adult-onset severe asthma. J Allergy Clin Immunol 141(4):1280–1290PubMedCrossRef Hekking P-P, Loza MJ, Pavlidis S, de Meulder B, Lefaudeux D et al (2018) Pathway discovery using transcriptomic profiles in adult-onset severe asthma. J Allergy Clin Immunol 141(4):1280–1290PubMedCrossRef
Metadaten
Titel
Physiological and immunological barriers in the lung
verfasst von
Takahiro Kageyama
Takashi Ito
Shigeru Tanaka
Hiroshi Nakajima
Publikationsdatum
07.03.2024
Verlag
Springer Berlin Heidelberg
Erschienen in
Seminars in Immunopathology / Ausgabe 4-6/2024
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-024-01003-y

Weitere Artikel der Ausgabe 4-6/2024

Seminars in Immunopathology 4-6/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.