Next Article in Journal
Abnormal Uterine Bleeding in Perimenopausal Women: The Role of Hysteroscopy and Its Impact on Quality of Life and Sexuality
Next Article in Special Issue
Special Issue “Diagnosis, Treatment, and Management of COPD and Asthma”
Previous Article in Journal
Outcomes of Kidney Transplantation in Patients with Autosomal Dominant Polycystic Kidney Disease: Our Experience Based on 35-Years Follow-Up
Previous Article in Special Issue
The Role of Digital Tools in the Timely Diagnosis and Prevention of Acute Exacerbations of COPD: A Comprehensive Review of the Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophils and Asthma

Department of Multidisciplinary Internal Medicine, Division of Respiratory Medicine and Rheumatology, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(5), 1175; https://doi.org/10.3390/diagnostics12051175
Submission received: 27 April 2022 / Revised: 4 May 2022 / Accepted: 5 May 2022 / Published: 8 May 2022
(This article belongs to the Special Issue Diagnosis, Treatment, and Management of COPD and Asthma)

Abstract

:
Although eosinophilic inflammation is characteristic of asthma pathogenesis, neutrophilic inflammation is also marked, and eosinophils and neutrophils can coexist in some cases. Based on the proportion of sputum cell differentiation, asthma is classified into eosinophilic asthma, neutrophilic asthma, neutrophilic and eosinophilic asthma, and paucigranulocytic asthma. Classification by bronchoalveolar lavage is also performed. Eosinophilic asthma accounts for most severe asthma cases, but neutrophilic asthma or a mixture of the two types can also present a severe phenotype. Biomarkers for the diagnosis of neutrophilic asthma include sputum neutrophils, blood neutrophils, chitinase-3-like protein, and hydrogen sulfide in sputum and serum. Thymic stromal lymphoprotein (TSLP)/T-helper 17 pathways, bacterial colonization/microbiome, neutrophil extracellular traps, and activation of nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 pathways are involved in the pathophysiology of neutrophilic asthma and coexistence of obesity, gastroesophageal reflux disease, and habitual cigarette smoking have been associated with its pathogenesis. Thus, targeting neutrophilic asthma is important. Smoking cessation, neutrophil-targeting treatments, and biologics have been tested as treatments for severe asthma, but most clinical studies have not focused on neutrophilic asthma. Phosphodiesterase inhibitors, anti-TSLP antibodies, azithromycin, and anti-cholinergic agents are promising drugs for neutrophilic asthma. However, clinical research targeting neutrophilic inflammation is required to elucidate the optimal treatment.

1. Introduction

Asthma is a common chronic airway disease that affects about 350 million people worldwide and varies in prevalence from country to country. In Japan, the prevalence is 9–10% and the number of patients with asthma was 1,177,000 in 2014 [1,2]. Diagnosis of asthma is based on a history or current symptoms, such as chest tightness, wheezing, dyspnea, and cough, together with variable expiratory airway limitation assessed by peak expiratory flow or spirometry. Chronic airway inflammation is an important feature of asthma and is characterized by the presence of eosinophils, basophils, mast cells, neutrophils, T helper 2 (Th2) cells, type 2 innate lymphoid cells (ILC2), CD8+ T cells, B cells, and dendritic cells [3,4,5]. In the Japanese Guidelines for Adult Asthma, a diagnosis is based on: (I) repetitive symptoms, such as paroxysmal dyspnea, wheezing, chest tightness, and cough; (II) reversible airflow limitation; (III) airway hyper-responsiveness; (IV) airway inflammation; (V) an atopic state; and (VI) exclusion of other cardiopulmonary disease [2].
Asthma is a heterogenous airway disease, and since the 2000s, cluster analyses have identified several phenotypes [6,7,8]. The common phenotypes are allergic asthma; non-allergic asthma; adult-onset (late-onset) asthma; asthma with persistent airflow limitation, and asthma with obesity [9]. The Severe Asthma Research Program (SARP) identified five phenotypes in patients with severe and non-severe asthma [10]. Kuo et al. found three transcriptome-associated clusters (TACs) in patients with asthma. TAC1 is characterized by immune receptors and a sputum eosinophil increase, TAC2 is characterized by interferon-, tumor necrosis factor-, and inflammasome-associated genes and a sputum neutrophil increase, and TAC3 is characterized by genes associated with metabolic pathways, ubiquitination, and mitochondrial function, with no sputum increase [11].
Neutrophils are the most abundant cells in peripheral blood and are stored in pulmonary capillary beds [12]. These cells play important roles in the innate immune system by killing microbes, phagocytosis, granule release, and formation of neutrophil extracellular traps (NETs). The role of neutrophils in asthma has been studied, but there is much debate about the presence of neutrophilic asthma [13,14,15,16]. Since glucocorticoids enhance the survival of neutrophils, which constitutively express glucocorticoid receptor β (GRβ) [17,18], the elevation of neutrophil levels in the asthmatic airway is thought to be a consequence of corticosteroid treatment. However, neutrophils are also observed in steroid-naïve patients with asthma [19,20,21,22] and several studies have found evidence that neutrophilic inflammation is associated with severe asthma and with asthma exacerbation [23,24]. A cluster analysis has shown that sputum neutrophil counts were associated with more severe phenotypes [25]. Recently, Minchem et al. reviewed the pathology of chronic lung diseases, including asthma [26]. They described the heterogeneity of neutrophils and their interactions with several immune and structural cells, identifying anti-inflammatory, pro-resolving, and pro-repair functions via direct cell-to-cell communication as well as via soluble mediators [26]. Neutrophils also connect with other cells via exosomes and extracellular vesicles [27]. In chronic lung diseases, an overabundance of neutrophils may exacerbate inflammation and remodeling [26]. Therefore, neutrophilic inflammation is involved in the heterogeneity of asthma, and neutrophil-targeted treatment may be important for severe asthma. The pathogenesis, definition, and biomarkers of neutrophilic asthma and potential therapy for neutrophilic asthma are discussed in this review.

2. Definition of Neutrophilic Asthma

The phenotype of asthma is generally categorized by the cell profile of induced sputum. In a healthy person, this profile has 0.4 ± 0.9% eosinophils and 37.5 ± 20.5% neutrophils, with means plus 2SD and 90th percentiles of 2.2% and 1.1% for eosinophils, and 77.7% and 64.4% for neutrophils, respectively [28]. Eosinophilic asthma is defined as an increase in eosinophils to above 2% or 3% and neutrophilic asthma as an increase in neutrophils to above 60% or 76% in induced sputum [29]. Paucigranulocytic asthma is defined as neutrophils < 76% and eosinophils < 3%, and conversely, mixed granulocytic asthma is defined as neutrophils > 76% and eosinophils > 3% [30]. However, there is still no clear definition of neutrophilic asthma [13]. In children, neutrophil-predominant severe asthma is defined using a cut-off of ≥5% neutrophils in bronchial lavage fluid [31]. Alternative methods, such as nasal wash or nasal lavage, have also been used to evaluate neutrophilic asthma or non-eosinophilic asthma [32].

3. Association of Eosinophils and Neutrophils

Coexistence of neutrophils and eosinophils occurs in severe asthma [10,33,34], and recent studies have shown that patients with asthma with a mixture of neutrophilic and eosinophilic inflammation had accelerated decline of respiratory function [35,36,37]. In studies of the coexistence mechanism, Nagata et al. found that activation of neutrophils may induce migration of eosinophils through the basement membrane via interleukin-8 (IL-8) [38], and that leukotriene B4 (LTB4)-activated neutrophils which induced eosinophil migration and Toll-like receptor 4 (TLR4) expression on neutrophils may be involved in this mechanism [36,39]. Theophylline attenuates trans-basement membrane migration of eosinophils in vitro by suppressing superoxide anion generation [40]. Lavinskinene et al. showed that the sputum neutrophil counts after bronchial allergen challenge were related to peripheral blood neutrophil chemotaxis in patients with asthma [41].

4. Pathogenesis of Asthma

4.1. TSLP

Thymic stromal lymphopoietin (TSLP) is secreted from a variety of cells, including basophils, mast cells, and airway epithelial cells [42]. In the human airway, airway epithelial cells secrete TSLP by recognition of allergens, viruses, pollutants and cigarette smoke, bacteria, and other external stimuli by pattern recognition receptors (PRPs) [43]. TSLP triggers allergic/eosinophilic and non-allergic/eosinophilic inflammation [44,45], and is also involved in neutrophilic inflammation in asthma. TSLP and TLR3 ligands promote conversion of naïve T cells to Th17 cells [46] and subsequently induce neutrophil recruitment via IL-8 and GM-CSF from airway epithelial cells [47]. TSLP polymorphism may also be related to allergic disease and eosinophilia in patients with asthma [48].

4.2. IL-17

IL-17 is a key cytokine in neutrophilic asthma. IL-17 and IL-17A are produced by Th17 cells and ILC3 cells, and may stimulate epithelial cells and fibroblasts and induce neutrophil activation and migration via IL-6, IL-8, and tumor necrosis factor-α (TNF-α). IL-17 induces glucocorticoid receptor (GR) β on epithelial cells in patients with asthma [49]. This may be related to glucocorticoid insensitivity in neutrophilic asthma. IL-17 induces eotaxin expression in human airway smooth muscle (HASM) cells [50], which may be linked to mixed neutrophilic and eosinophilic inflammation in asthma. IL-17 is increased in bronchial biopsy in severe asthma [51] and in sputum from patients with moderate-to-severe asthma [52]. Bulles et al. showed that the IL17 mRNA level correlated with the IL8 mRNA level and with CD3 gamma cell and neutrophil counts, which suggested a link between IL-17 and neutrophilic inflammation [52]. IL-17 also enhances IL-1β-mediated IL-8 release from HASM cells [53], and the IL-17/Th17 axis is involved in microbiomes in the development of asthma [54].

4.3. Bacterial Colonization and Microbiome in the Airway in Neutrophilic Asthma

The intestinal and respiratory microbiomes are both thought to be associated with the pathogenesis of asthma [55]. In patients with neutrophilic asthma, 50% of patients have bacterial infection based on bronchoalveolar lavage [56], and at the time of asthma exacerbation, 87.8% of patients have bacteria in sputum, with neutrophils > 65% [13]. Recent studies have shown that bacterial microbiome profiles in the airway were associated with neutrophil inflammation in asthma [57,58,59] and that the Th17/IL-17 axis was involved in this process [60,61]. Microbiome-derived cluster analysis of sputum in severe asthma showed two distinct phenotypes: cluster 1 had less-severe asthma and commensal bacterial profile, and higher bacterial richness and diversity; cluster 2 had more severe asthma with a reduced commensal bacterial profile, clear deficiency of several bacterial species, and neutrophilic inflammation [57]. The intestinal microbiome has also been linked to the development of asthma, but its relationship with neutrophilic inflammation in asthma is unclear [62].

4.4. Obesity

Obesity increases the risk of asthma development [63,64,65,66], worsens asthma control and severity [8,67], increases hospitalization [68], and reduces responses to inhaled corticosteroids (ICS) alone or in conjunction with a long-acting β2 agonist (LABA) [68,69,70]. In cluster analyses, obesity-related asthma has been grouped into non-Th2 asthma, with later onset, female preponderance, and severe symptoms [7,8,10]. Obesity is associated with inflammatory adipokines including leptin, resistin, lipocain 2, IL-6, TNF-α, IL-1β, and IFN-γ [71,72,73,74,75]. These mediators induce airway inflammation. In a mouse obese asthma model, ILC3 stimulated by IL-1β, IL-6, or IL-23 produced IL-17A [76]. IL-17A alone or in combination with TNF-α has been shown to induce IL-8 production from epithelial cells [77], and cigarette smoke can also enhance IL-17A-induced IL-8 and IL-6 production [78,79,80,81]. IL-6 and IL-8 recruit and activate neutrophils in an asthmatic airway [41,81]. In obese patients with asthma, IL-17 is associated with steroid resistance by dysregulation of GRα and GRβ [82], while in human bronchial epithelial cells, IL-17A induces glucocorticoid insensitivity [83]. Insulin resistance and vitamin D deficiency related to obesity may aggravate airway remodeling and hyper-responsiveness by enhancing leptin, transforming growth factor (TGF)-β1, IL-1β, and IL-6 expression [84,85,86,87], which might then promote neutrophilic inflammation.

4.5. NETs and NETosis

Neutrophil extracellular traps (NETs) were first described by Brinkmann et al. [88]. Neutrophils stimulated by bacteria or inflammatory mediators, such as IL-8, platelet activating factor, and lipopolysaccharide (LPS), release NETs that include neutrophil elastase, cathepsin G, myeloperoxidase, defensins, lactoferrin, histones, pentraxin 3, reactive oxygen species (ROS), and DNA to captivate and kill bacteria [89]. NETosis is an active form of neutrophil death related to NETs formation [88]. Several studies have related NETs to the pathogenesis of autoimmune disease, cancer, and atherosclerosis [90,91]; dysregulation of NETs may also result in asthma pathobiology, although the mechanisms associated with NETs are not fully understood. In a mouse model, allergen exposure with endotoxin induced NETosis [92]. In severe neutrophilic asthma, Krishnamoorthy et al. determined that cytoplasts and neutrophils positively correlated with IL-17 levels in the bronchoalveolar fluid [92]. The sputum extracellular DNA (eDNA) level has been correlated with expressions of IL-8, IL-1β, and NLRP3 [93], and Lachowicz-Scroggins et al. found that high extracellular DNA (eDNA) in sputum was associated with poor asthma control, mucus hypersecretion, and oral steroid use in patients with asthma [94]. The same group also showed that the eDNA level was correlated with neutrophil inflammation, NET components, caspase-1 activity, and IL-1β. In vitro, epithelial damage caused by NETs has been prevented by DNase [94]. These studies indicate that NETs and eDNA are related to severe neutrophilic asthma.

4.6. NLRP3 Inflammasome and Asthma

Nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing (NLRP) inflammasomes are a critical component of the innate immune system and they play an important role in activation of inflammation. NLRP3, an NLR family PRP, responds to pathogen-associated molecule patterns (PAMPs) or danger (damage)-associated molecular patterns (DAMPs). Activation of NLRP3 inflammasomes is mediated by two signals: the priming signal triggered by PAMPs, DAMPs, IL-1β, and TNF-α; the second (activation) signal mediated by extracellular ATP, RNA viruses, particulate matter, ionic flux, ROS, mitochondrial dysfunction, and lysosomal damage. Upon activation of NLRP3 inflammasomes, IL-1β and IL-18 are secreted [95,96]. Dysregulation of NLRP3 inflammasome activation is related to Alzheimer’s disease [97], Parkinson’s disease [98], diabetes mellitus, atherosclerosis [99], and pulmonary inflammatory disorders, including lung fibrosis [100], acute exacerbation of interstitial pneumonia [101], sarcoidosis [102], asbestosis, and silicosis [103]. Since human lungs are exposed to many endogenous and exogenous noxious stimuli, including viruses, bacteria, cigarette smoke, and particulate matter, the innate immune response in the airway via NLRP3 inflammasomes is important. However, excess or persistent activation of NLRP3 inflammasomes by allergens or irritants has been shown to induce persistent inflammation and tissue damage in the airway of patients with asthma [104,105]. In these patients, the sputum NLRP3 level was increased and was correlated with neutrophilic airway inflammation [106,107]. NLRP3 expression has also been shown to be increased in obese patients with asthma [108]. Kim et al. found that a high-fat diet induced airway hyper-reactivity and increased NLRP3, IL17A, and IL1B mRNA in an obese mouse model [76], suggesting that obesity-induced airway hypersensitivity is mediated by NLRP3 inflammasomes that are activated by fatty acids or cholesterol crystals from macrophages in adipose tissue or in the lungs [76]. In other experimental models, NLPR3 and apoptosis-associated speck-like protein containing CARD (ASC)-deficient mice exhibited reduced airway inflammation [109]. Ovalbumin (OVA) mouse models with alum [110], LPS, Aspergillus fumigatus [111], Chlamydia muridarum, or Haemophilus influenzae infection also have been shown to have increased NLRP3 [106]. In this latter model, neutrophil depletion suppressed IL-1β-induced airway hyper-responsiveness.

4.7. S100A8/A9, HMGB-1, RAGE, and TLR4

The S100A8/A9 complex belongs to the Ca2+-binding S100 protein family and is a DAMP protein complex expressed in neutrophils, monocytes, and macrophages [112,113]. High mobility group box 1 (HMGB-1), which is also a DAMP protein, a non-histone, chromatin-associated nuclear protein is released from necrotic, inflammatory, macrophage, dendritic, natural killer, and resident cells (epithelial cells, smooth muscle cells, and fibroblasts) [114,115,116,117]. TNF-α, IL-1β, and IFN-γ induce HMBG-1 release from activated macrophages [118,119]. HMBG-1 and S100A8/S100A9 bind to two types of receptors: the receptor for advanced glycation end products (RAGE) and TLR-4. RAGE is expressed on lung [120], skeletal muscle, heart, liver, kidney [121], lung epithelial, and immune cells [122,123,124,125,126]. Perkins et al. showed that knockout of RAGE abolished type 2 cytokine-induced airway inflammation and mucus hyperplasia in a mouse model [127]. Oczypok et al. reported that RAGE induced asthma/allergic airway inflammation by promoting IL-33 expression, and that ILC2 accumulation was critical in the pathogenesis of asthma in a mouse model [128].
TLR4 is also expressed on B cells [129], T cells [130], monocytes, macrophages [131], and neutrophils [132]. S100A8/A9 and HMGB-1 might be involved in the pathobiology of remodeling in asthma by promoting inflammation and tissue repair in the airway [117]. In a mouse model, blocking HMGB-1 and TLR-4 attenuated disonoyl phthalate-induced asthma [133]. HMGB-1 is increased in OVA-induced asthma [134]. In patients with asthma, the sputum HMGB-1 level is increased and inversely correlated with the percentage predicted forced expiratory volume in 1 s (%FEV1) and FEV1/forced vital capacity (FVC) ratio. The HMGB-1 level is also associated with the severity of asthma and neutrophils in sputum [135,136]. An endogenous form of RAGE (esRAGE), which is a decoy receptor for AGE, was elevated in sputum from a patient with asthma; however, the esRAGE level was not associated with asthma severity [135], in contrast to the RAGE level [136]. Since HMGB-1 stimulates TNF-α, IL-6, and IL-8 production from monocytes [137,138], it might be a key player in inducing neutrophilic asthma. Recent studies have shown that a soluble form of RAGE prevents Th17-mediated neutrophilic asthma by blocking HMBG1/RAGE signaling in a mouse model [139]. In patients with neutrophilic asthma, decreased sRAGE was associated with asthma severity [140], and a recent study showed that sRAGE was associated with low eosinophil count and IgE in children with asthma [141]. RAGE has been linked to cigarette-smoke-induced neutrophilic inflammation and airway hyper-responsiveness in a mouse model, but TLR4, another receptor for HMGB-1 and S100A8/A, was not involved [142]. Furthermore, AGER (which encodes RAGE) expression, rather than TLR4 expression, was significantly correlated with the sputum neutrophil count and airway hyper-responsiveness in patients with chronic obstructive pulmonary disease (COPD) [142]. Therefore, HMGB-1 and sRAGE might be biomarkers for neutrophilic asthma.

4.8. House Dust Mites and Neutrophilic Asthma

House dust mites (HDMs) are the most important allergen for the development and worsening of allergic asthma, with 90% of cases of pediatric asthma sensitized to HDMs. Many studies of allergic and eosinophilic asthma have been conducted using a mouse model sensitized to HDMs, and several recent studies have described neutrophilic or mixed-granulocytic asthma models. Menson et al. reported a novel BALB/c female mouse model using Mycobacterium tuberculosis extract, complete Freund’s adjuvant, and HDM, in which the bronchial alveolar lavage fluid (BALF) contained 80% neutrophils and 10% eosinophils [143]. Mack et al. described an old (9 months) C57BL/6 female mouse model sensitized to HDMs that showed elevated neutrophils in BALF as compared with young (3 months) mice, as a model of adult-onset asthma [144]. Sadamatsu et al. found that a high-fat diet induced elevated neutrophils in BALF in an HDM-sensitized mouse model [145]. Neutrophil counts in the sputum of patients with chronic neutrophilic asthma have been shown to be correlated with the serum HDM-specific IgG levels, and these patients have HDM-derived enolase in their serum [146]. In the same study, HDM-derived enolase was shown to induce epithelial barrier disintegration and neutrophilic inflammation in a mouse model [146]. Blockade of leukotriene B4 receptor 1 (BLT1)/BLT2 by antagonists can reduce neutrophil infiltration based on findings in an HDM- and LPS-induced mouse asthma model [147]. IL-1β was found to be required to promote neutrophilic inflammation in an HDM-sensitized and viral-exacerbated model, using double-stranded RNA to mimic rhinovirus [148]. In contrast, Patel et al. found neutrophil depletion in an HDM allergic airway disease mouse, with this depletion enhancing Th2 inflammation by inducing G-colony stimulating factor-induced ILC2 activation and cytokine production [149].

4.9. Electric, Heat-Not-Burn Cigarettes, and Combustible Cigarettes

Almost one-quarter of patients with adult asthma are thought to have smoking habits. Several studies have also shown that the efficacy of ICS is reduced in patients with asthma who are exposed to smoking [150,151,152]. Passive smoking in a family increases the use of drugs for pediatric asthma [153]. E-cigarette or electric cigarette (vapor) exposure induces neutrophil protease, matrix metalloproteinase-2 (MMP-2), and MMP-9 in healthy subjects [154]. Schweitzer et al. showed that e-cigarette use was independently associated with asthma in adolescents [155]. A study from Korea also showed an association of e-cigarette use with asthma diagnosis and absence from school due to asthma [156]. E-cigarette liquid has been shown to induce IL-6 production from human epithelial cells and addition of nicotine further increased IL-6 production [157], while electronic nicotine delivery systems using aerosols also induced IL-6 and IL-8 secretion [158].
A 2015 internet survey showed that the use of heat-not-burn (HNB) cigarettes among patients with asthma was 0.0% in Japan [159]. The first HNB cigarette, IQOS, was released in 2014 in Japan, and the harmfulness of HNB cigarettes to asthma remains uncertain. However, HNB cigarettes contain nicotine and many other toxins [160,161], as well as particulate matter [162], and thus, may worsen asthma control by inducing neutrophilic inflammation. Further studies are needed to examine how HNB cigarettes affect asthma pathogenesis and neutrophilic inflammation [80]. In patients with mild asthma, combustible cigarette smoking increases neutrophil counts, and IL-17A, IL-6, and IL-8 levels [80]. Exposure of human epithelial cells to cigarette smoke extracts, IL-17A, and aeroallergens has been shown to induce IL-6 and IL-8 production, which may be associated with the neutrophil accumulation in asthmatic airways [80]. In a rat model, the late asthmatic response to OVA increased with cigarette smoke (CS) exposure as compared with no exposure. ICS decreased eosinophil and lymphocyte accumulation with and without CS exposure but did not decrease neutrophil accumulation with CS exposure [163]. Quitting smoking and avoiding environmental smoking can resolve neutrophil inflammation in patients with asthma who smoke. A combination of pharmacotherapy using bupropion and varenicline with counseling was most effective for smoking cessation [164]. Smoking cessation-support therapy using a smartphone application has recently been covered by insurance in Japan [165].

4.10. Air Pollution

Relationships of air pollution with asthma development or exacerbation have been reported for several years. Examples of outdoor or indoor pollution include diesel exhaust, foreign workplace matter, ozone, nitrogen dioxide, sulfur dioxide, second-hand smoke, heating sources, cooking smoke, and molds [166,167,168]. These pollutants induce asthma exacerbation through oxidative stress and damage, airway remodeling, inflammatory pathways, immunological responses, and enhancement of airway sensitivity [166,168]. Particulate matter induces Th2 and Th17 inflammation in allergic conditions and this induces eosinophilic and neutrophilic inflammation in asthma [169,170,171,172]. In an in vivo study, ozone exposure induced IL-8 secretion from epithelial cells [173], which was related to neutrophil accumulation in the airway after exposure to ozone in patients with asthma [174].

4.11. Gastroesophageal Reflux Disease

Gastroesophageal reflux disease (GERD) is a common comorbidity in asthma, and the severity of asthma is increased when complicated with GERD [175]. In the SARP study, a subgroup of patients with asthma defined as having a low pH in exhaled breath condensate had a high body mass index (BMI) and high neutrophilic airway inflammation, and had GERD as a complication [176]. GERD is often accompanied by mixed eosinophilic and neutrophilic inflammation (reviewed in [177]). Simpson et al. found that patients with neutrophilic asthma had a high prevalence of rhinosinusitis and symptoms of GERD as compared with patients with eosinophilic asthma [178]. The mechanism through which GERD induces or enhances airway inflammation in asthma has not been determined, but GERD is associated with obesity [179], which may lead to neutrophilic inflammation, as mentioned above. The triangle of inflammation, obesity, and GERD with sleep disordered breathing syndrome is important in children with asthma [180].
Figure 1 shows the pathology of neutrophilic asthma (Figure 1).

5. Biomarkers of Neutrophilic Asthma

Non-type 2 subtypes of asthma, including neutrophilic and paucigranulocytic asthma, are difficult to diagnose because of a lack of appropriate biomarkers. However, recent studies have suggested promising diagnostic biomarkers for neutrophilic asthma (Table 1).

5.1. YKL40

Chitinase-3-like protein (YKL-40) is a human glycoprotein that is released from several cell types, including neutrophils, macrophages, and epithelial cells. YKL-40 is involved in the pathogenesis of many diseases, including rheumatoid arthritis [192], multiple sclerosis [193], chronic obstructive lung disease [194,195], Alzheimer’s disease [196], and asthma [181,197]. Serum YKL-40 levels are related to asthma severity, while lung YKL-40 levels are correlated with airway remodeling [181,182]. In the multicenter BIOAIR study, the serum YKL-40 level was negatively correlated with lung function (FEV1% predicted, FVC, and FEV1/FVC), but not with fraction of exhaled nitric oxide or blood and sputum eosinophil and neutrophil counts [182]. Cluster analyses have shown that high serum YKL-40 levels were associated with neutrophilic asthma and paucigranulocytic asthma [183] and that patients with high serum YKL-40 had severe airflow obstruction and near fatal or frequent exacerbation [183]. The serum YKL-40 level has been shown to be positively correlated with blood neutrophils, IL-6, and sputum IL-1β [119], while the sputum YKL-40 level has been shown to be strongly correlated with neutrophilic asthma and sputum myeloperoxidase, and was associated with sputum IL-8 and soluble IL-6 receptor levels [187]. Therefore, serum and sputum YKL-40 levels are useful biomarkers for neutrophilic asthma.

5.2. Hydrogen Sulfide

Nitric oxide is a biomarker of type 2 inflammation and carbon monoxide is a partial biomarker of asthma severity [198,199]. Hydrogen sulfide (H2S) is the third biomarker in breath, and sputum H2S is a novel biomarker of neutrophilic asthma. Sputum H2S levels are correlated with neutrophils in sputum and airflow limitation [184,185,186], and the sputum-to-serum H2S ratio predicts the risk of asthma exacerbation [186]. Therefore, sputum H2S is a diagnostic marker for neutrophilic asthma and a predictor of exacerbation when combined with serum H2S. These biomarkers are also elevated in asthma-COPD overlap [200].

5.3. Myeloperoxidase

Myeloperoxidase (MPO) is a marker of neutrophil activation. Serum MPO has been shown to be elevated in ANCA-associated vasculitis, including microscopic polyangiitis and eosinophilic granulomatous polyangiitis, while sputum MPO has been shown to correlate positively with sputum YKL-40 levels [187] and sputum neutrophils [23]. Thus, sputum MPO is a useful biomarker for neutrophilic asthma, whereas elevation of serum MPO is thought to be a marker for small vessel vasculitis.

5.4. Blood Neutrophil Count

The peripheral blood neutrophil count is not appropriate as a surrogate marker for neutrophilic asthma defined based on sputum cell differentiation [201,202,203]. However, neutrophilia has been shown to be associated with chronic airway obstruction [189] and an annual decline in FEV1 [188]. The sputum neutrophil count after bronchial allergen challenge has been shown to be related to peripheral blood neutrophil chemotaxis in patients with asthma [41].

5.5. MicroRNA

Several studies have shown that microRNAs (miRNAs) are biomarkers for asthma. Panganiban et al. found upregulation of miRNA-1248 in patients with asthma [204] and also showed that miRNAs in serum could be used to phenotype asthma [205]. Huang et al. revealed that miR-199a-5p in sputum and plasma was increased in neutrophilic asthma [190] and showed that levels of miRNA-199a-5p secreted from human LPS-stimulated peripheral neutrophils were inversely correlated with FEV1 [190]. A genome-wide analysis of miRNAs in sputum from patients with asthma showed that hsa-miR-223-3p was expressed in neutrophils and was associated with neutrophil counts in response to ozone exposure [206]. Maes et al. showed that miR-223-3p, miR-142-3p, and miR-629-3p were upregulated in severe, neutrophilic asthma [191]. Therefore, several miRNAs are biomarkers for diagnosis of neutrophilic asthma, and they are also considered to be therapeutic targets [207,208].

6. Airway Remodeling in Neutrophilic Asthma

Airway remodeling in asthma is caused by chronic airway inflammation and is a characteristic feature of chronic asthma. The pathological changes in airway remodeling involve mucous metaplasia, thickening of the reticular basement membrane, increases of goblet cells and mucus hypersecretion, shedding of epithelial cells, submucosal infiltration of inflammatory cells, extracellular matrix deposition, airway smooth muscle (ASM) cell hyperplasia, and hypertrophy. Neutrophilic asthma and airway remodeling are not fully understood, but several studies have shown that inflammatory mediators, such as LTB4, IL-6, IL-8, and TNF-α, which are related to neutrophilic inflammation, were elevated in an asthmatic airway. Several of these mediators and cytokines have also been shown to be elevated in neutrophilic asthma, of which LTB4, IL-8, TNF-α, IL-17, and IL-6 may be related to airway remodeling. Figure 2 shows neutrophilic inflammation-associated airway remodeling in asthma (Figure 2).

6.1. Leukotriene B4

In severe asthma, leukotriene B4 (LTB4) is increased in sputum, BALF, exhaled breath condensate, urine, and arterial blood [209]. LTB4 is a chemoattractant mediator of neutrophils [210] and has been found to recruit eosinophils in a guinea pig model [211,212]. The relationship between LTB4 and airway remodeling has not been fully studied, but BLT1 and BLT2 are expressed on HASM cells. LTB4 has been shown to induce HASM cell migration and proliferation in vitro [213]. Therefore, LTB4 might be involved in airway remodeling in asthma.

6.2. IL-8

IL-8 is increased in sputum or BALF from patients with severe asthma and is inversely correlated with %predicted FEV1 and sputum neutrophil counts [23,24,59,214,215,216]. A recent study showed that IL-8 in BALF was the only cytokine that distinguished controlled from uncontrolled asthma among 48 evaluated cytokines [216]. IL-8 has been shown to induce HASM cell proliferation and migration [217,218,219], to stimulate mucin secretion [220], and to upregulate MUC5A and MUC5B in goblet cells [221]. YKL-40 has been shown to induce IL-8 in bronchial epithelial cells and to cause HASM cell proliferation and migration [222]. Therefore, IL-8 might be related to severe neutrophilic asthma and airway remodeling in asthma.

6.3. TNF-α

TNF-α is a proinflammatory cytokine related to neutrophilic asthma. In vitro, TNF-α induced airway smooth muscle migration and proliferation [223], extracellular matrix deposition, subepithelial fibrosis, and inflammatory cytokine secretion [224,225]. In a mouse model, TNF-α was involved in glucocorticoid insensitivity in neutrophilic inflammation in asthma, which may induce chronic inflammation and lead to airway remodeling [226]. In vitro, miR874, which may be associated with the development of asthma, has been shown to inhibit TNF-α-induced IL-6, IL-8, collagen I, and collagen III production in ASM cells [224].

6.4. IL-17A

IL-17A is an independent risk factor for severe asthma and is involved in obesity-associated asthma and CS-related airway neutrophilia [82,163,227]. In a mouse model, IL-17A induced type V collagen expression, TGFB1 mRNA expression, and SMAD3 activation in airway epithelial cells [228]. In vitro, MUC5A and MUC5B expressions have been induced by IL-17A via IL-6 and NF-κB in epithelial cells [229,230,231]. IL-17A has also been shown to be involved in the epithelia mesenchymal transition via expression of TGF-β1 in airway epithelial cells [232]. In a mouse model, IL-17 was involved in airway smooth muscle hyperplasia mediated by fibroblast growth factor 2 from airway epithelial cells, and neutrophil elastase played an important role in this model [233,234]. In other mouse models using OVA and LPS for exacerbation, anti-IL-17A antibody decreased extracellular matrix deposition [235] and vascular remodeling [234]. Therefore, IL-17A comodulated with TGF-β1 is involved in airway remodeling in asthma and is related to neutrophils [236].

6.5. Other Inflammatory Mediators and Cytokines

IL-1β has been shown to induce neutrophilic asthma and IL-33 expression in a mouse model of asthma with viral infection exacerbation [148], and was a key cytokine in induction of airway smooth muscle hypersensitivity [237]. IL-1β alone or with TNF superfamily members has been observed to cause airway neutrophilic inflammation and remodeling in an adult animal model [238,239]. Oncostatin M (OSM) is released from neutrophils and induces epithelial barrier dysfunction [240]. In severe asthma, there are increases in OSM in sputum and in OSM-positive neutrophils in biopsy specimens [241]. OSM is also increased in patients with asthma with fixed airway obstruction [242]. Furthermore, MMP9 and elastase may be involved in airway remodeling in asthma [243,244,245].

7. Treatment

Treatment with an ICS is a key approach for asthma, but corticosteroids are not effective in neutrophilic asthma [246,247]. Treatment of asthma related to neutrophilic inflammation can be categorized into non-pharmacological approaches, nonspecific treatment for neutrophil inflammation, treatment specific to neutrophils and neutrophil mediators, and biologics (Table 2).

7.1. Non-Pharmacological Approach

Smoking cessation may be the best way to reduce neutrophilic inflammation in neutrophilic asthma patients who smoke. In a clinical trial, smoking cessation in young adults with asthma improved asthma control, but with persistent eosinophil counts and little neutrophil reduction [248]. In this trial, 17% of the subjects had neutrophilic asthma. Another clinical trial showed improvements in lung function and sputum neutrophil counts [151]. Weight loss by diet, exercise, diet with exercise, or surgical intervention also improved asthma control, quality of life, lung function, and airway hyper-responsiveness [249,263,264,265,266]. Thus, smoking cessation and weight loss are good approaches for patients with severe asthma, regardless of the inflammatory phenotype.

7.2. Nonspecific Treatment for Neutrophilic Inflammation

7.2.1. Macrolides

Macrolides have various functions, in addition to their actions as antibiotics [267]. The effectiveness of clarithromycin has been shown in chronic stable asthma with Mycoplasma pneumoniae or Chlamydia pneumoniae mRNA in the airway [268]. The AMAZES study showed the effectiveness of azithromycin for persistent uncontrolled asthma [269]. In this study, 43% of the cases were eosinophilic, 11% neutrophilic, 30% paucigranulocytic, and 4% mixed, based on sputum phenotyping. A subset analysis in the AMAZES study showed that azithromycin was similarly effective for severe asthma in the cases with an eosinophilic sputum phenotype [269]. The effect of azithromycin was correlated with the abundance of Haemophiles influenzae colonization as assessed by quantitative polymerase chain reaction [270]. In the AMAZES study, sputum TNFR1 and TNFR2 were increased in neutrophilic asthma and azithromycin reduced sputum TNFR2 in non-eosinophilic asthma, which may be related to the therapeutic mechanism [271]. The AZISAST study showed a reduced rate of severe exacerbation by azithromycin in non-eosinophilic severe asthma [272]; in a study in severe neutrophilic asthma, 8-week administration of this drug improved quality of life and reduced airway IL-8 and neutrophils [250]. Therefore, long term macrolide treatment is a promising therapy in severe asthma, particularly for the neutrophil-dominant phenotype.

7.2.2. Phosphodiesterase Inhibitors

Roflumilast is an oral phosphodiesterase (PDE) inhibitor that has therapeutic effects on COPD [273] and asthma-COPD overlap [274]. Several studies have shown the efficacy of roflumilast alone [275,276] or in combination with a leukotriene receptor antagonist in moderate-to-severe asthma [277]. Roflumilast attenuates both eosinophilic and neutrophilic inflammation induced by allergens [251,278]. Inhaled PDE inhibitors have also been examined in patients with asthma (reviewed in [279]): CH6001 showed inhibition of the late asthmatic response induced by allergen exposure [280] and RPL554 (a PDE3 and PDE4 inhibitor) increased FEV1 in patients with asthma and reduced neutrophils and total cells in sputum from healthy individuals [281]. Studies of PDE inhibitors focusing on neutrophilic asthma are needed, but roflumilast and inhaled PDE4 inhibitors may be promising for neutrophilic asthma [282].

7.2.3. Anticholinergics

Anticholinergics have been used for treatment of COPD and asthma. Long-acting muscarinic antagonist (LAMAs) and short-acting muscarinic antagonists are both available for treatment of asthma. LAMAs decreased eosinophils in sensitized mice [283,284], and in an obstructive airway disease model in rat, tiotropium decreased neutrophil counts, IL-1β and IL-6 in bronchoalveolar lavage [285]. In an in vitro study in human epithelial cells, tiotropium reduced IL-8 production induced by IL-17A [286] or LPS [287]. In clinical studies, tiotropium has been shown to be effective as an add-on therapy to ICS [288] or ICS/LABA [289] in uncontrolled asthma, and Iwamoto et al. found that anti-cholinergics were effective in non-eosinophilic asthma [290]. Tiotropium has been shown to be effective, independent of type 2 inflammation in adults [252,291,292] and in children and adolescents [253]. However, the efficacy of ICS or tiotropium was similar to that of a placebo in patients with mild persistent asthma, including 73% with low eosinophilic asthma [293].

7.3. Specific Therapy for Neutrophils and Neutrophil Mediators

7.3.1. CXCR2 Antagonists

CXCR2 is a receptor for IL-8 that is expressed on neutrophils. A CXCR2 inhibitor, SCH527123, reduced sputum neutrophils and exacerbation in severe asthma cases in a 4-week clinical trial [254]. Another CXCR2 antagonist, AZD5069, reduced neutrophils in bronchial mucosa, sputum, and blood, but failed to reduce severe exacerbation [294,295].

7.3.2. 5-Lipoxygenase-Activating Protein Inhibitors and 5-Lipoxygenase Inhibitors

Five-lipoxygenase-activating protein (FLAP) and 5-lipoxygenase (5-LO) are required for synthesis of LTB4. GSK2190915 is a FLAP inhibitor that has been evaluated for patients with asthma in several studies [255,296,297]. In one study focused on neutrophilic asthma, a FLAP inhibitor suppressed sputum LTB4 and urine LTE4 levels, but failed to reduce neutrophil counts in sputum and had no clinical effects on FEV1, PEF, and ACQ scores [296]. Zileuton is a 5-LO inhibitor that has also been evaluated in patients with asthma [256,298] and has been shown to be effective in moderate-to-severe asthma based on improved PEF and asthma symptoms [256]. A recent retrospective study showed no associations among Th2-high or Th2-low phenotypes and a poor response rate to zileuton in association with severe asthma and obesity [298].

7.4. Biologics

Several biological agents are currently available for patients with severe asthma. There are six FDA-approved monoclonal antibodies (mAbs): omalizumab, which is anti-IgE antibody; mepolizumab and reslizumab, which are anti-IL-5 antibodies; benralizumab, which is an anti-IL-5 receptor α antibody; dupilumab, which is an anti-IL-4 receptor α antibody; and tezepelumab, which is an anti-TSLP antibody. These biologics exhibited clinical benefits for allergic/Th2-high asthma [299].

7.4.1. Targeting TSLP

Tezepelumab, a humanized mAb for TSLP, has been tested in a phase 2 clinical trial in patients with moderate-to-severe asthma [300] and in a phase 3 clinical trial in patients with severe asthma [257]. Tezepelumab reduced the rate of exacerbation and improved FEV1, ACQ, and AQLQ scores, regardless of type 2 inflammation. Therefore, tezepelumab may be effective for severe neutrophilic asthma. Biphasic antibodies for TSLP/IL-13 (zweimab and doppelmab) have recently been developed [301] and may also be evaluated for treatment of severe asthma with type 2, non-type 2, or neutrophilic inflammation.

7.4.2. Targeting TNF-α

Blocking of TNF-α by infliximab and golimumab, which are anti-TNF-α mAbs, and etanercept, which is a recombinant TNF-α receptor, has been examined as treatment for moderate and severe asthma [258,259,302,303,304]. In patients with severe and uncontrolled asthma under treatment with high-dose ICS and LABAs, golimumab did not improve FEV1 or the rate of exacerbation [258]. Etanercept, in several clinical trails, has been shown to improve airway hyper-responsiveness (AHR); FEV1, AQLQ, and ACQ scores; and asthma symptoms; as well as to reduce sputum macrophages and CRP levels in several clinicals trials [302,303,304]. However, a large, randomized clinical study of etanercept for moderate-to-severe asthma showed no efficacy for ACQ, AQLQ, FEV1, exacerbation rate, or AHR [259].

7.4.3. Targeting IL-17

Anti-IL-17 antibody has been shown to decrease airway hyper-responsiveness and airway inflammation in a mouse model of obesity, alone [305] or in combination with a Rho-kinase inhibitor [306]. Secukinumab, an mAb targeting IL-17A, was tested in a randomized clinical trial in patients with severe asthma treated with high doses of ICS alone or in combination with a LABA. In this trial, responders (defined as patients with a 5% increase in predicted FEV1) showed increased nasal epithelial neutrophilic inflammation and had decreased markers of IgE-driven systemic inflammation based on a nasal brushing pathway analysis of differentially regulated genes [307]. A randomized, double-blind, placebo-controlled study of brodalumab, a monoclonal antibody targeting IL-17 receptor A, showed no treatment effect in subjects with moderate-to-severe asthma [260]. A bispecific antibody targeting IL-13 and IL-17 showed clinical safety with no deaths or serious adverse events in a phase I study [308].

7.4.4. Targeting IL-23

As mentioned above, IL-17 is involved in neutrophilic inflammation in asthma and IL-23, an IL-12 family cytokine, is important for maintenance and recruitment of Th17 cells [309]. However, risankizumab, an IL23p19 mAb, failed to show efficacy for worsening of asthma as compared with a placebo in a phase I, randomized, double-blind, placebo-controlled study in adults with severe asthma, with no significant changes in sputum cell differentials [261].

7.4.5. Targeting IL-6

Tocilizumab, an anti-IL-6 receptor mAb, had effects on CRP, IL-6, and soluble IL-6 receptor, but did not improve allergen-induced bronchoconstriction in 11 patients with mild asthma [262].

7.5. Other Potential Therapy for Neutrophilic Asthma

Peroxisome proliferator-activated receptor-gamma agonists have been tested in a murine model of neutrophilic asthma [310]. Statins are also candidate drugs for patients with obesity and asthma [311,312]. Inhibitors of protein kinases, p38 MAPK, and phosphoinositide 3-kinase (PI3K δ and γ) have been examined for COPD or asthma [312,313,314,315]. These inhibitors might be effective in neutrophilic asthma because the PI3K pathway is involved in neutrophil migration and degranulation [316,317]. Glucagon-like peptide-1 receptor (GLP-1R) agonists inhibit aeroallergen-induced activation of ILC2 and neutrophilic airway inflammation in obese mice [318]. Fore et al. found that patients with asthma who received GLP-1R agonists had less exacerbation than those treated with sulfonylureas or insulin [319]. Some of these drugs have been tested for asthma or COPD, but not specifically for neutrophilic asthma.

8. Conclusions

Asthma is a heterogenous syndrome that includes neutrophilic asthma as one phenotype. There is still uncertainty about this phenotype, but many studies have shown the importance of neutrophils in asthma. There is no clear definition of neutrophilic asthma, but sputum and peripheral blood neutrophils, YKL-40, H2S, MPA, and miRNAs may be useful biomarkers for this condition. Identification of new biomarkers or combinations of biomarkers will be important for future diagnosis of neutrophilic asthma. Neutrophilic inflammation is involved in airway remodeling in patients with asthma, including those with obesity and GERD. Non-pharmacological and pharmacological therapy, including targeting of neutrophils and nonspecific treatment, may be useful for neutrophilic asthma, but most treatments have yet to be tested in patients with this condition. Further studies, focused on non-type 2 cases and neutrophilic inflammation, are needed to develop treatment for severe neutrophilic asthma.

Author Contributions

Conceptualization, A.Y.; writing—original draft preparation, A.Y.; writing—review and editing, A.Y., R.O. and T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

Figures are created with BioRender.com assessed on 27 April 2022 and on 4 May 2022.

Conflicts of Interest

A.Y. received speaker honorariums from Asahi Kasei Pharma, AstraZeneca, Boehringer Ingelheim, Chugai, Glaxo Smith Kline, Janssen, Kyorin, Mitsubishi Tanabe Pharma, Novartis, Ono, and Sanofi, outside of the submitted work. R.O. received speaker honorariums from Chugai, Ono, Sanofi, Jansen, and Nippon Shinyaku, outside of the submitted work. T.H. received speaker honorariums from Asahi Kasei Pharma, AstraZeneca, Glaxo Smith Kline, Janssen, Nippon Shinyaku, and Sanofi, outside of the submitted work.

References

  1. Fukutomi, Y.; Nakamura, H.; Kobayashi, F.; Taniguchi, M.; Konno, S.; Nishimura, M.; Kawagishi, Y.; Watanabe, J.; Komase, Y.; Akamatsu, Y.; et al. Nationwide cross-sectional population-based study on the prevalences of asthma and asthma symptoms among Japanese adults. Int. Arch. Allergy Immunol. 2010, 153, 280–287. [Google Scholar] [CrossRef]
  2. Nakamura, Y.; Tamaoki, J.; Nagase, H.; Yamaguchi, M.; Horiguchi, T.; Hozawa, S.; Ichinose, M.; Iwanaga, T.; Kondo, R.; Nagata, M.; et al. Japanese guidelines for adult asthma 2020. Allergol. Int. 2020, 69, 519–548. [Google Scholar] [CrossRef]
  3. Lambrecht, B.N.; Hammad, H. The immunology of asthma. Nat. Immunol. 2015, 16, 45–56. [Google Scholar] [CrossRef]
  4. Robinson, D.; Humbert, M.; Buhl, R.; Cruz, A.A.; Inoue, H.; Korom, S.; Hanania, N.A.; Nair, P. Revisiting Type 2-high and Type 2-low airway inflammation in asthma: Current knowledge and therapeutic implications. Clin. Exp. Allergy 2017, 47, 161–175. [Google Scholar] [CrossRef]
  5. Hinks, T.S.C.; Hoyle, R.D.; Gelfand, E.W. CD8+ Tc2 cells: Underappreciated contributors to severe asthma. Eur. Respir. Rev. 2019, 28, 190092. [Google Scholar] [CrossRef] [Green Version]
  6. Bel, E.H. Clinical phenotypes of asthma. Curr. Opin. Pulm. Med. 2004, 10, 44–50. [Google Scholar] [CrossRef]
  7. Wenzel, S.E. Asthma phenotypes: The evolution from clinical to molecular approaches. Nat. Med. 2012, 18, 716–725. [Google Scholar] [CrossRef]
  8. Haldar, P.; Pavord, I.D.; Shaw, D.E.; Berry, M.A.; Thomas, M.; Brightling, C.E.; Wardlaw, A.J.; Green, R.H. Cluster analysis and clinical asthma phenotypes. Am. J. Respir. Crit. Care Med. 2008, 178, 218–224. [Google Scholar] [CrossRef] [Green Version]
  9. Grobal Initiative for Asthma. Global Strategy for Asthma Management and Prevention 2021. Available online: www.ginasthma.org (accessed on 21 September 2021).
  10. Moore, W.C.; Meyers, D.A.; Wenzel, S.E.; Teague, W.G.; Li, H.; Li, X.; D’Agostino, R., Jr.; Castro, M.; Curran-Everett, D.; Fitzpatrick, A.M.; et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 2010, 181, 315–323. [Google Scholar] [CrossRef]
  11. Kuo, C.S.; Pavlidis, S.; Loza, M.; Baribaud, F.; Rowe, A.; Pandis, I.; Sousa, A.; Corfield, J.; Djukanovic, R.; Lutter, R.; et al. T-helper cell type 2 (Th2) and non-Th2 molecular phenotypes of asthma using sputum transcriptomics in U-BIOPRED. Eur. Respir. J. 2017, 49, 1602135. [Google Scholar] [CrossRef]
  12. Hogg, J.C. Neutrophil kinetics and lung injury. Physiol. Rev. 1987, 67, 1249–1295. [Google Scholar] [CrossRef]
  13. Nair, P.; Surette, M.G.; Virchow, J.C. Neutrophilic asthma: Misconception or misnomer? Lancet Respir. Med. 2021, 9, 441–443. [Google Scholar] [CrossRef]
  14. Nair, P.; Prabhavalkar, K.S. Neutrophilic Asthma and Potentially Related Target Therapies. Curr. Drug Targets 2020, 21, 374–388. [Google Scholar] [CrossRef]
  15. Nabe, T. Steroid-Resistant Asthma and Neutrophils. Biol. Pharm. Bull. 2020, 43, 31–35. [Google Scholar] [CrossRef] [Green Version]
  16. Crisford, H.; Sapey, E.; Rogers, G.B.; Taylor, S.; Nagakumar, P.; Lokwani, R.; Simpson, J.L. Neutrophils in asthma: The good, the bad and the bacteria. Thorax 2021, 76, 835–844. [Google Scholar] [CrossRef]
  17. Strickland, I.; Kisich, K.; Hauk, P.J.; Vottero, A.; Chrousos, G.P.; Klemm, D.J.; Leung, D.Y. High constitutive glucocorticoid receptor beta in human neutrophils enables them to reduce their spontaneous rate of cell death in response to corticosteroids. J. Exp. Med. 2001, 193, 585–593. [Google Scholar] [CrossRef] [Green Version]
  18. Saffar, A.S.; Ashdown, H.; Gounni, A.S. The molecular mechanisms of glucocorticoids-mediated neutrophil survival. Curr. Drug Targets 2011, 12, 556–562. [Google Scholar] [CrossRef] [Green Version]
  19. Shimoda, T.; Obase, Y.; Nagasaka, Y.; Nakano, H.; Kishikawa, R.; Iwanaga, T. Airway inflammation phenotype prediction in asthma patients using lung sound analysis with fractional exhaled nitric oxide. Allergol. Int. 2017, 66, 581–585. [Google Scholar] [CrossRef]
  20. Berry, M.; Morgan, A.; Shaw, D.E.; Parker, D.; Green, R.; Brightling, C.; Bradding, P.; Wardlaw, A.J.; Pavord, I.D. Pathological features and inhaled corticosteroid response of eosinophilic and non-eosinophilic asthma. Thorax 2007, 62, 1043–1049. [Google Scholar] [CrossRef] [Green Version]
  21. Toyran, M.; Bakirtas, A.; Dogruman-Al, F.; Turktas, I. Airway inflammation and bronchial hyperreactivity in steroid naive children with intermittent and mild persistent asthma. Pediatr. Pulmonol. 2014, 49, 140–147. [Google Scholar] [CrossRef]
  22. Louis, R.; Sele, J.; Henket, M.; Cataldo, D.; Bettiol, J.; Seiden, L.; Bartsch, P. Sputum eosinophil count in a large population of patients with mild to moderate steroid-naive asthma: Distribution and relationship with methacholine bronchial hyperresponsiveness. Allergy 2002, 57, 907–912. [Google Scholar] [CrossRef] [Green Version]
  23. Jatakanon, A.; Uasuf, C.; Maziak, W.; Lim, S.; Chung, K.F.; Barnes, P.J. Neutrophilic inflammation in severe persistent asthma. Am. J. Respir. Crit. Care Med. 1999, 160, 1532–1539. [Google Scholar] [CrossRef] [Green Version]
  24. Gibson, P.G.; Simpson, J.L.; Saltos, N. Heterogeneity of airway inflammation in persistent asthma: Evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest 2001, 119, 1329–1336. [Google Scholar] [CrossRef] [Green Version]
  25. Moore, W.C.; Hastie, A.T.; Li, X.; Li, H.; Busse, W.W.; Jarjour, N.N.; Wenzel, S.E.; Peters, S.P.; Meyers, D.A.; Bleecker, E.R.; et al. Sputum neutrophil counts are associated with more severe asthma phenotypes using cluster analysis. J. Allergy Clin. Immunol. 2014, 133, 1557–1563.e5. [Google Scholar] [CrossRef] [Green Version]
  26. Mincham, K.T.; Bruno, N.; Singanayagam, A.; Snelgrove, R.J. Our evolving view of neutrophils in defining the pathology of chronic lung disease. Immunology 2021, 164, 701–721. [Google Scholar] [CrossRef]
  27. Alashkar Alhamwe, B.; Potaczek, D.P.; Miethe, S.; Alhamdan, F.; Hintz, L.; Magomedov, A.; Garn, H. Extracellular Vesicles and Asthma-More Than Just a Co-Existence. Int. J. Mol. Sci. 2021, 22, 4984. [Google Scholar] [CrossRef]
  28. Belda, J.; Leigh, R.; Parameswaran, K.; O’Byrne, P.M.; Sears, M.R.; Hargreave, F.E. Induced sputum cell counts in healthy adults. Am. J. Respir. Crit. Care Med. 2000, 161, 475–478. [Google Scholar] [CrossRef] [Green Version]
  29. Chung, K.F. Asthma phenotyping: A necessity for improved therapeutic precision and new targeted therapies. J. Intern. Med. 2016, 279, 192–204. [Google Scholar] [CrossRef]
  30. Schleich, F.; Brusselle, G.; Louis, R.; Vandenplas, O.; Michils, A.; Pilette, C.; Peche, R.; Manise, M.; Joos, G. Heterogeneity of phenotypes in severe asthmatics. The Belgian Severe Asthma Registry (BSAR). Respir. Med. 2014, 108, 1723–1732. [Google Scholar] [CrossRef] [Green Version]
  31. Grunwell, J.R.; Stephenson, S.T.; Tirouvanziam, R.; Brown, L.A.S.; Brown, M.R.; Fitzpatrick, A.M. Children with Neutrophil-Predominant Severe Asthma Have Proinflammatory Neutrophils With Enhanced Survival and Impaired Clearance. J. Allergy Clin. Immunol. Pract. 2019, 7, 516–525.e6. [Google Scholar] [CrossRef]
  32. Stemmy, E.J.; Benton, A.S.; Lerner, J.; Alcala, S.; Constant, S.L.; Freishtat, R.J. Extracellular cyclophilin levels associate with parameters of asthma in phenotypic clusters. J. Asthma 2011, 48, 986–993. [Google Scholar] [CrossRef] [Green Version]
  33. Kikuchi, S.; Nagata, M.; Kikuchi, I.; Hagiwara, K.; Kanazawa, M. Association between neutrophilic and eosinophilic inflammation in patients with severe persistent asthma. Int. Arch. Allergy Immunol. 2005, 137 (Suppl. S1), 7–11. [Google Scholar] [CrossRef]
  34. Jarjour, N.N.; Erzurum, S.C.; Bleecker, E.R.; Calhoun, W.J.; Castro, M.; Comhair, S.A.; Chung, K.F.; Curran-Everett, D.; Dweik, R.A.; Fain, S.B.; et al. Severe asthma: Lessons learned from the National Heart, Lung, and Blood Institute Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 2012, 185, 356–362. [Google Scholar] [CrossRef] [Green Version]
  35. Hastie, A.T.; Mauger, D.T.; Denlinger, L.C.; Coverstone, A.; Castro, M.; Erzurum, S.; Jarjour, N.; Levy, B.D.; Meyers, D.A.; Moore, W.C.; et al. Baseline sputum eosinophil + neutrophil subgroups’ clinical characteristics and longitudinal trajectories for NHLBI Severe Asthma Research Program (SARP 3) cohort. J. Allergy Clin. Immunol. 2020, 146, 222–226. [Google Scholar] [CrossRef] [Green Version]
  36. Marc-Malovrh, M.; Camlek, L.; Skrgat, S.; Kern, I.; Flezar, M.; Dezman, M.; Korosec, P. Elevated eosinophils, IL5 and IL8 in induced sputum in asthma patients with accelerated FEV1 decline. Respir. Med. 2020, 162, 105875. [Google Scholar] [CrossRef]
  37. Katz, B.; Sofonio, M.; Lyden, P.D.; Mitchell, M.D. Prostaglandin concentrations in cerebrospinal fluid of rabbits under normal and ischemic conditions. Stroke 1988, 19, 349–351. [Google Scholar] [CrossRef] [Green Version]
  38. Kikuchi, I.; Kikuchi, S.; Kobayashi, T.; Hagiwara, K.; Sakamoto, Y.; Kanazawa, M.; Nagata, M. Eosinophil trans-basement membrane migration induced by interleukin-8 and neutrophils. Am. J. Respir. Cell Mol. Biol. 2006, 34, 760–765. [Google Scholar] [CrossRef]
  39. Nishihara, F.; Nakagome, K.; Kobayashi, T.; Noguchi, T.; Araki, R.; Uchida, Y.; Soma, T.; Nagata, M. Trans-basement membrane migration of eosinophils induced by LPS-stimulated neutrophils from human peripheral blood in vitro. ERJ Open Res. 2015, 1, 00003-2015. [Google Scholar] [CrossRef] [Green Version]
  40. Kikuchi, I.; Kikuchi, S.; Kobayashi, T.; Takaku, Y.; Hagiwara, K.; Kanazawa, M.; Nagata, M. Theophylline attenuates the neutrophil-dependent augmentation of eosinophil trans-basement membrane migration. Int. Arch. Allergy Immunol. 2007, 143 (Suppl. S1), 44–49. [Google Scholar] [CrossRef]
  41. Lavinskiene, S.; Bajoriuniene, I.; Malakauskas, K.; Jeroch, J.; Sakalauskas, R. Sputum neutrophil count after bronchial allergen challenge is related to peripheral blood neutrophil chemotaxis in asthma patients. Inflamm. Res. 2014, 63, 951–959. [Google Scholar] [CrossRef]
  42. Gauvreau, G.M.; Sehmi, R.; Ambrose, C.S.; Griffiths, J.M. Thymic stromal lymphopoietin: Its role and potential as a therapeutic target in asthma. Expert Opin. Ther. Targets 2020, 24, 777–792. [Google Scholar] [CrossRef]
  43. Menzies-Gow, A.; Wechsler, M.E.; Brightling, C.E. Unmet need in severe, uncontrolled asthma: Can anti-TSLP therapy with tezepelumab provide a valuable new treatment option? Respir. Res. 2020, 21, 268. [Google Scholar] [CrossRef]
  44. Takai, T. TSLP expression: Cellular sources, triggers, and regulatory mechanisms. Allergol. Int. 2012, 61, 3–17. [Google Scholar] [CrossRef] [Green Version]
  45. Gour, N.; Lajoie, S. Epithelial Cell Regulation of Allergic Diseases. Curr. Allergy Asthma Rep. 2016, 16, 65. [Google Scholar] [CrossRef] [Green Version]
  46. Tanaka, J.; Watanabe, N.; Kido, M.; Saga, K.; Akamatsu, T.; Nishio, A.; Chiba, T. Human TSLP and TLR3 ligands promote differentiation of Th17 cells with a central memory phenotype under Th2-polarizing conditions. Clin. Exp. Allergy 2009, 39, 89–100. [Google Scholar] [CrossRef]
  47. Gao, H.; Ying, S.; Dai, Y. Pathological Roles of Neutrophil-Mediated Inflammation in Asthma and Its Potential for Therapy as a Target. J. Immunol. Res. 2017, 2017, 3743048. [Google Scholar] [CrossRef] [Green Version]
  48. Moorehead, A.; Hanna, R.; Heroux, D.; Neighbour, H.; Sandford, A.; Gauvreau, G.M.; Sommer, D.D.; Denburg, J.A.; Akhabir, L. A thymic stromal lymphopoietin polymorphism may provide protection from asthma by altering gene expression. Clin. Exp. Allergy 2020, 50, 471–478. [Google Scholar] [CrossRef]
  49. Vazquez-Tello, A.; Semlali, A.; Chakir, J.; Martin, J.G.; Leung, D.Y.; Eidelman, D.H.; Hamid, Q. Induction of glucocorticoid receptor-beta expression in epithelial cells of asthmatic airways by T-helper type 17 cytokines. Clin. Exp. Allergy 2010, 40, 1312–1322. [Google Scholar] [CrossRef]
  50. Rahman, M.S.; Yamasaki, A.; Yang, J.; Shan, L.; Halayko, A.J.; Gounni, A.S. IL-17A induces eotaxin-1/CC chemokine ligand 11 expression in human airway smooth muscle cells: Role of MAPK (Erk1/2, JNK, and p38) pathways. J. Immunol. 2006, 177, 4064–4071. [Google Scholar] [CrossRef] [Green Version]
  51. Al-Ramli, W.; Prefontaine, D.; Chouiali, F.; Martin, J.G.; Olivenstein, R.; Lemiere, C.; Hamid, Q. T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma. J. Allergy Clin. Immunol. 2009, 123, 1185–1187. [Google Scholar] [CrossRef]
  52. Bullens, D.M.; Truyen, E.; Coteur, L.; Dilissen, E.; Hellings, P.W.; Dupont, L.J.; Ceuppens, J.L. IL-17 mRNA in sputum of asthmatic patients: Linking T cell driven inflammation and granulocytic influx? Respir. Res. 2006, 7, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Oikawa, T.; Shimamura, M.; Ashino-Fuse, H.; Iwaguchi, T.; Ishizuka, M.; Takeuchi, T. Inhibition of angiogenesis by 15-deoxyspergualin. J. Antibiot. 1991, 44, 1033–1035. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, D.; Tan, Y.; Bajinka, O.; Wang, L.; Tang, Z. Th17/IL-17 Axis Regulated by Airway Microbes Get Involved in the Development of Asthma. Curr. Allergy Asthma Rep. 2020, 20, 11. [Google Scholar] [CrossRef] [PubMed]
  55. Noval Rivas, M.; Crother, T.R.; Arditi, M. The microbiome in asthma. Curr. Opin. Pediatr. 2016, 28, 764–771. [Google Scholar] [CrossRef] [Green Version]
  56. Liu, W.; Liu, S.; Verma, M.; Zafar, I.; Good, J.T.; Rollins, D.; Groshong, S.; Gorska, M.M.; Martin, R.J.; Alam, R. Mechanism of TH2/TH17-predominant and neutrophilic TH2/TH17-low subtypes of asthma. J. Allergy Clin. Immunol. 2017, 139, 1548–1558.e4. [Google Scholar] [CrossRef] [Green Version]
  57. Abdel-Aziz, M.I.; Brinkman, P.; Vijverberg, S.J.H.; Neerincx, A.H.; Riley, J.H.; Bates, S.; Hashimoto, S.; Kermani, N.Z.; Chung, K.F.; Djukanovic, R.; et al. Sputum microbiome profiles identify severe asthma phenotypes of relative stability at 12 to 18 months. J. Allergy Clin. Immunol. 2021, 147, 123–134. [Google Scholar] [CrossRef]
  58. Yang, X.; Li, H.; Ma, Q.; Zhang, Q.; Wang, C. Neutrophilic Asthma Is Associated with Increased Airway Bacterial Burden and Disordered Community Composition. Biomed. Res. Int. 2018, 2018, 9230234. [Google Scholar] [CrossRef] [Green Version]
  59. Green, B.J.; Wiriyachaiporn, S.; Grainge, C.; Rogers, G.B.; Kehagia, V.; Lau, L.; Carroll, M.P.; Bruce, K.D.; Howarth, P.H. Potentially pathogenic airway bacteria and neutrophilic inflammation in treatment resistant severe asthma. PLoS ONE 2014, 9, e100645. [Google Scholar] [CrossRef]
  60. Essilfie, A.T.; Simpson, J.L.; Horvat, J.C.; Preston, J.A.; Dunkley, M.L.; Foster, P.S.; Gibson, P.G.; Hansbro, P.M. Haemophilus influenzae infection drives IL-17-mediated neutrophilic allergic airways disease. PLoS Pathog. 2011, 7, e1002244. [Google Scholar] [CrossRef]
  61. Yang, B.; Liu, R.; Yang, T.; Jiang, X.; Zhang, L.; Wang, L.; Wang, Q.; Luo, Z.; Liu, E.; Fu, Z. Neonatal Streptococcus pneumoniae infection may aggravate adulthood allergic airways disease in association with IL-17A. PLoS ONE 2015, 10, e0123010. [Google Scholar] [CrossRef]
  62. Kozik, A.J.; Huang, Y.J. The microbiome in asthma: Role in pathogenesis, phenotype, and response to treatment. Ann. Allergy Asthma Immunol. 2019, 122, 270–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gilliland, F.D.; Berhane, K.; Islam, T.; McConnell, R.; Gauderman, W.J.; Gilliland, S.S.; Avol, E.; Peters, J.M. Obesity and the risk of newly diagnosed asthma in school-age children. Am. J. Epidemiol. 2003, 158, 406–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mamun, A.A.; Lawlor, D.A.; Alati, R.; O’Callaghan, M.J.; Williams, G.M.; Najman, J.M. Increasing body mass index from age 5 to 14 years predicts asthma among adolescents: Evidence from a birth cohort study. Int. J. Obes. 2007, 31, 578–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Weinmayr, G.; Forastiere, F.; Buchele, G.; Jaensch, A.; Strachan, D.P.; Nagel, G.; Group, I.P.T.S. Overweight/obesity and respiratory and allergic disease in children: International study of asthma and allergies in childhood (ISAAC) phase two. PLoS ONE 2014, 9, e113996. [Google Scholar] [CrossRef]
  66. Ho, W.C.; Lin, Y.S.; Caffrey, J.L.; Lin, M.H.; Hsu, H.T.; Myers, L.; Chen, P.C.; Lin, R.S. Higher body mass index may induce asthma among adolescents with pre-asthmatic symptoms: A prospective cohort study. BMC Public Health 2011, 11, 542. [Google Scholar] [CrossRef] [Green Version]
  67. Schatz, M.; Hsu, J.W.; Zeiger, R.S.; Chen, W.; Dorenbaum, A.; Chipps, B.E.; Haselkorn, T. Phenotypes determined by cluster analysis in severe or difficult-to-treat asthma. J. Allergy Clin. Immunol. 2014, 133, 1549–1556. [Google Scholar] [CrossRef]
  68. Holguin, F.; Bleecker, E.R.; Busse, W.W.; Calhoun, W.J.; Castro, M.; Erzurum, S.C.; Fitzpatrick, A.M.; Gaston, B.; Israel, E.; Jarjour, N.N.; et al. Obesity and asthma: An association modified by age of asthma onset. J. Allergy Clin. Immunol. 2011, 127, 1486–1493.e2. [Google Scholar] [CrossRef]
  69. Peters-Golden, M.; Swern, A.; Bird, S.S.; Hustad, C.M.; Grant, E.; Edelman, J.M. Influence of body mass index on the response to asthma controller agents. Eur. Respir. J. 2006, 27, 495–503. [Google Scholar] [CrossRef] [Green Version]
  70. Boulet, L.P.; Franssen, E. Influence of obesity on response to fluticasone with or without salmeterol in moderate asthma. Respir. Med. 2007, 101, 2240–2247. [Google Scholar] [CrossRef] [Green Version]
  71. Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science 1993, 259, 87–91. [Google Scholar] [CrossRef]
  72. Peters, U.; Dixon, A.E.; Forno, E. Obesity and asthma. J. Allergy Clin. Immunol. 2018, 141, 1169–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Loffreda, S.; Yang, S.Q.; Lin, H.Z.; Karp, C.L.; Brengman, M.L.; Wang, D.J.; Klein, A.S.; Bulkley, G.B.; Bao, C.; Noble, P.W.; et al. Leptin regulates proinflammatory immune responses. FASEB J. 1998, 12, 57–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Komakula, S.; Khatri, S.; Mermis, J.; Savill, S.; Haque, S.; Rojas, M.; Brown, L.; Teague, G.W.; Holguin, F. Body mass index is associated with reduced exhaled nitric oxide and higher exhaled 8-isoprostanes in asthmatics. Respir. Res. 2007, 8, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Miethe, S.; Guarino, M.; Alhamdan, F.; Simon, H.U.; Renz, H.; Dufour, J.F.; Potaczek, D.P.; Garn, H. Effects of obesity on asthma: Immunometabolic links. Pol. Arch. Intern. Med. 2018, 128, 469–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kim, H.Y.; Lee, H.J.; Chang, Y.J.; Pichavant, M.; Shore, S.A.; Fitzgerald, K.A.; Iwakura, Y.; Israel, E.; Bolger, K.; Faul, J.; et al. Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat. Med. 2014, 20, 54–61. [Google Scholar] [CrossRef] [Green Version]
  77. Honda, K.; Wada, H.; Nakamura, M.; Nakamoto, K.; Inui, T.; Sada, M.; Koide, T.; Takata, S.; Yokoyama, T.; Saraya, T.; et al. IL-17A synergistically stimulates TNF-alpha-induced IL-8 production in human airway epithelial cells: A potential role in amplifying airway inflammation. Exp. Lung Res. 2016, 42, 205–216. [Google Scholar] [CrossRef]
  78. Prause, O.; Laan, M.; Lotvall, J.; Linden, A. Pharmacological modulation of interleukin-17-induced GCP-2-, GRO-alpha- and interleukin-8 release in human bronchial epithelial cells. Eur. J. Pharmacol. 2003, 462, 193–198. [Google Scholar] [CrossRef]
  79. Lee, K.H.; Lee, C.H.; Woo, J.; Jeong, J.; Jang, A.H.; Yoo, C.G. Cigarette Smoke Extract Enhances IL-17A-Induced IL-8 Production via Up-Regulation of IL-17R in Human Bronchial Epithelial Cells. Mol. Cells 2018, 41, 282–289. [Google Scholar] [CrossRef]
  80. Siew, L.Q.C.; Wu, S.Y.; Ying, S.; Corrigan, C.J. Cigarette smoking increases bronchial mucosal IL-17A expression in asthmatics, which acts in concert with environmental aeroallergens to engender neutrophilic inflammation. Clin. Exp. Allergy 2017, 47, 740–750. [Google Scholar] [CrossRef] [Green Version]
  81. Linden, A. Role of interleukin-17 and the neutrophil in asthma. Int. Arch. Allergy Immunol. 2001, 126, 179–184. [Google Scholar] [CrossRef]
  82. Al Heialy, S.; Gaudet, M.; Ramakrishnan, R.K.; Mogas, A.; Salameh, L.; Mahboub, B.; Hamid, Q. Contribution of IL-17 in Steroid Hyporesponsiveness in Obese Asthmatics Through Dysregulation of Glucocorticoid Receptors alpha and beta. Front. Immunol. 2020, 11, 1724. [Google Scholar] [CrossRef] [PubMed]
  83. Zijlstra, G.J.; Ten Hacken, N.H.; Hoffmann, R.F.; van Oosterhout, A.J.; Heijink, I.H. Interleukin-17A induces glucocorticoid insensitivity in human bronchial epithelial cells. Eur. Respir. J. 2012, 39, 439–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Park, Y.H.; Oh, E.Y.; Han, H.; Yang, M.; Park, H.J.; Park, K.H.; Lee, J.H.; Park, J.W. Insulin resistance mediates high-fat diet-induced pulmonary fibrosis and airway hyperresponsiveness through the TGF-beta1 pathway. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef] [Green Version]
  85. Cardet, J.C.; Ash, S.; Kusa, T.; Camargo, C.A., Jr.; Israel, E. Insulin resistance modifies the association between obesity and current asthma in adults. Eur. Respir. J. 2016, 48, 403–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Sanchez Jimenez, J.; Herrero Espinet, F.J.; Mengibar Garrido, J.M.; Roca Antonio, J.; Penos Mayor, S.; Penas Boira Mdel, M.; Roca Comas, A.; Ballester Martinez, A. Asthma and insulin resistance in obese children and adolescents. Pediatr. Allergy Immunol. 2014, 25, 699–705. [Google Scholar] [CrossRef] [PubMed]
  87. Han, H.; Chung, S.I.; Park, H.J.; Oh, E.Y.; Kim, S.R.; Park, K.H.; Lee, J.H.; Park, J.W. Obesity-induced Vitamin D Deficiency Contributes to Lung Fibrosis and Airway Hyperresponsiveness. Am. J. Respir. Cell Mol. Biol. 2021, 64, 357–367. [Google Scholar] [CrossRef]
  88. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  89. Cheng, O.Z.; Palaniyar, N. NET balancing: A problem in inflammatory lung diseases. Front. Immunol. 2013, 4, 1. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, C.L.; Tangsombatvisit, S.; Rosenberg, J.M.; Mandelbaum, G.; Gillespie, E.C.; Gozani, O.P.; Alizadeh, A.A.; Utz, P.J. Specific post-translational histone modifications of neutrophil extracellular traps as immunogens and potential targets of lupus autoantibodies. Arthritis Res. Ther. 2012, 14, R25. [Google Scholar] [CrossRef] [Green Version]
  91. Doring, Y.; Manthey, H.D.; Drechsler, M.; Lievens, D.; Megens, R.T.; Soehnlein, O.; Busch, M.; Manca, M.; Koenen, R.R.; Pelisek, J.; et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation 2012, 125, 1673–1683. [Google Scholar] [CrossRef] [Green Version]
  92. Krishnamoorthy, N.; Douda, D.N.; Bruggemann, T.R.; Ricklefs, I.; Duvall, M.G.; Abdulnour, R.E.; Martinod, K.; Tavares, L.; Wang, X.; Cernadas, M.; et al. Neutrophil cytoplasts induce TH17 differentiation and skew inflammation toward neutrophilia in severe asthma. Sci. Immunol. 2018, 3, eaao4747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Wright, T.K.; Gibson, P.G.; Simpson, J.L.; McDonald, V.M.; Wood, L.G.; Baines, K.J. Neutrophil extracellular traps are associated with inflammation in chronic airway disease. Respirology 2016, 21, 467–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lachowicz-Scroggins, M.E.; Dunican, E.M.; Charbit, A.R.; Raymond, W.; Looney, M.R.; Peters, M.C.; Gordon, E.D.; Woodruff, P.G.; Lefrancais, E.; Phillips, B.R.; et al. Extracellular DNA, Neutrophil Extracellular Traps, and Inflammasome Activation in Severe Asthma. Am. J. Respir. Crit. Care Med. 2019, 199, 1076–1085. [Google Scholar] [CrossRef] [PubMed]
  95. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. He, Y.; Hara, H.; Nunez, G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem. Sci. 2016, 41, 1012–1021. [Google Scholar] [CrossRef] [Green Version]
  97. Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372. [Google Scholar] [CrossRef]
  98. Wang, S.; Yuan, Y.H.; Chen, N.H.; Wang, H.B. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson’s disease. Int. Immunopharmacol. 2019, 67, 458–464. [Google Scholar] [CrossRef]
  99. Strowig, T.; Henao-Mejia, J.; Elinav, E.; Flavell, R. Inflammasomes in health and disease. Nature 2012, 481, 278–286. [Google Scholar] [CrossRef]
  100. Lasithiotaki, I.; Giannarakis, I.; Tsitoura, E.; Samara, K.D.; Margaritopoulos, G.A.; Choulaki, C.; Vasarmidi, E.; Tzanakis, N.; Voloudaki, A.; Sidiropoulos, P.; et al. NLRP3 inflammasome expression in idiopathic pulmonary fibrosis and rheumatoid lung. Eur. Respir. J. 2016, 47, 910–918. [Google Scholar] [CrossRef] [Green Version]
  101. Jager, B.; Seeliger, B.; Terwolbeck, O.; Warnecke, G.; Welte, T.; Muller, M.; Bode, C.; Prasse, A. The NLRP3-Inflammasome-Caspase-1 Pathway Is Upregulated in Idiopathic Pulmonary Fibrosis and Acute Exacerbations and Is Inducible by Apoptotic A549 Cells. Front. Immunol. 2021, 12, 642855. [Google Scholar] [CrossRef]
  102. Huppertz, C.; Jager, B.; Wieczorek, G.; Engelhard, P.; Oliver, S.J.; Bauernfeind, F.G.; Littlewood-Evans, A.; Welte, T.; Hornung, V.; Prasse, A. The NLRP3 inflammasome pathway is activated in sarcoidosis and involved in granuloma formation. Eur. Respir. J. 2020, 55, 1900119. [Google Scholar] [CrossRef] [PubMed]
  103. Dostert, C.; Petrilli, V.; Van Bruggen, R.; Steele, C.; Mossman, B.T.; Tschopp, J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008, 320, 674–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Pinkerton, J.W.; Kim, R.Y.; Robertson, A.A.B.; Hirota, J.A.; Wood, L.G.; Knight, D.A.; Cooper, M.A.; O’Neill, L.A.J.; Horvat, J.C.; Hansbro, P.M. Inflammasomes in the lung. Mol. Immunol. 2017, 86, 44–55. [Google Scholar] [CrossRef]
  105. Lamkanfi, M. Emerging inflammasome effector mechanisms. Nat. Rev. Immunol. 2011, 11, 213–220. [Google Scholar] [CrossRef] [PubMed]
  106. Kim, R.Y.; Pinkerton, J.W.; Essilfie, A.T.; Robertson, A.A.B.; Baines, K.J.; Brown, A.C.; Mayall, J.R.; Ali, M.K.; Starkey, M.R.; Hansbro, N.G.; et al. Role for NLRP3 Inflammasome-mediated, IL-1beta-Dependent Responses in Severe, Steroid-Resistant Asthma. Am. J. Respir. Crit. Care Med. 2017, 196, 283–297. [Google Scholar] [CrossRef]
  107. Rossios, C.; Pavlidis, S.; Hoda, U.; Kuo, C.H.; Wiegman, C.; Russell, K.; Sun, K.; Loza, M.J.; Baribaud, F.; Durham, A.L.; et al. Sputum transcriptomics reveal upregulation of IL-1 receptor family members in patients with severe asthma. J. Allergy Clin. Immunol. 2018, 141, 560–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Wood, L.G.; Li, Q.; Scott, H.A.; Rutting, S.; Berthon, B.S.; Gibson, P.G.; Hansbro, P.M.; Williams, E.; Horvat, J.; Simpson, J.L.; et al. Saturated fatty acids, obesity, and the nucleotide oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in asthmatic patients. J. Allergy Clin. Immunol. 2019, 143, 305–315. [Google Scholar] [CrossRef] [Green Version]
  109. Ritter, M.; Straubinger, K.; Schmidt, S.; Busch, D.H.; Hagner, S.; Garn, H.; Prazeres da Costa, C.; Layland, L.E. Functional relevance of NLRP3 inflammasome-mediated interleukin (IL)-1beta during acute allergic airway inflammation. Clin. Exp. Immunol. 2014, 178, 212–223. [Google Scholar] [CrossRef]
  110. Li, H.; Willingham, S.B.; Ting, J.P.; Re, F. Cutting edge: Inflammasome activation by alum and alum’s adjuvant effect are mediated by NLRP3. J. Immunol. 2008, 181, 17–21. [Google Scholar] [CrossRef] [Green Version]
  111. Sebag, S.C.; Koval, O.M.; Paschke, J.D.; Winters, C.J.; Jaffer, O.A.; Dworski, R.; Sutterwala, F.S.; Anderson, M.E.; Grumbach, I.M. Mitochondrial CaMKII inhibition in airway epithelium protects against allergic asthma. JCI Insight 2017, 2, e88297. [Google Scholar] [CrossRef]
  112. Edgeworth, J.; Gorman, M.; Bennett, R.; Freemont, P.; Hogg, N. Identification of p8,14 as a highly abundant heterodimeric calcium binding protein complex of myeloid cells. J. Biol. Chem. 1991, 266, 7706–7713. [Google Scholar] [CrossRef]
  113. Bhardwaj, R.S.; Zotz, C.; Zwadlo-Klarwasser, G.; Roth, J.; Goebeler, M.; Mahnke, K.; Falk, M.; Meinardus-Hager, G.; Sorg, C. The calcium-binding proteins MRP8 and MRP14 form a membrane-associated heterodimer in a subset of monocytes/macrophages present in acute but absent in chronic inflammatory lesions. Eur. J. Immunol. 1992, 22, 1891–1897. [Google Scholar] [CrossRef] [PubMed]
  114. Hamada, N.; Maeyama, T.; Kawaguchi, T.; Yoshimi, M.; Fukumoto, J.; Yamada, M.; Yamada, S.; Kuwano, K.; Nakanishi, Y. The role of high mobility group box1 in pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2008, 39, 440–447. [Google Scholar] [CrossRef] [PubMed]
  115. Lotze, M.T.; Tracey, K.J. High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nat. Rev. Immunol. 2005, 5, 331–342. [Google Scholar] [CrossRef] [PubMed]
  116. Ellerman, J.E.; Brown, C.K.; de Vera, M.; Zeh, H.J.; Billiar, T.; Rubartelli, A.; Lotze, M.T. Masquerader: High mobility group box-1 and cancer. Clin. Cancer Res. 2007, 13, 2836–2848. [Google Scholar] [CrossRef] [Green Version]
  117. Halayko, A.J.; Ghavami, S. S100A8/A9: A mediator of severe asthma pathogenesis and morbidity? Can. J. Physiol. Pharmacol. 2009, 87, 743–755. [Google Scholar] [CrossRef]
  118. Ogawa, E.N.; Ishizaka, A.; Tasaka, S.; Koh, H.; Ueno, H.; Amaya, F.; Ebina, M.; Yamada, S.; Funakoshi, Y.; Soejima, J.; et al. Contribution of high-mobility group box-1 to the development of ventilator-induced lung injury. Am. J. Respir. Crit. Care Med. 2006, 174, 400–407. [Google Scholar] [CrossRef]
  119. Liu, S.; Stolz, D.B.; Sappington, P.L.; Macias, C.A.; Killeen, M.E.; Tenhunen, J.J.; Delude, R.L.; Fink, M.P. HMGB1 is secreted by immunostimulated enterocytes and contributes to cytomix-induced hyperpermeability of Caco-2 monolayers. Am. J. Physiol. Cell Physiol. 2006, 290, C990–C999. [Google Scholar] [CrossRef] [Green Version]
  120. Buckley, S.T.; Ehrhardt, C. The receptor for advanced glycation end products (RAGE) and the lung. J. Biomed. Biotechnol. 2010, 2010, 917108. [Google Scholar] [CrossRef] [Green Version]
  121. Brett, J.; Schmidt, A.M.; Yan, S.D.; Zou, Y.S.; Weidman, E.; Pinsky, D.; Nowygrod, R.; Neeper, M.; Przysiecki, C.; Shaw, A.; et al. Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am. J. Pathol. 1993, 143, 1699–1712. [Google Scholar]
  122. Akirav, E.M.; Preston-Hurlburt, P.; Garyu, J.; Henegariu, O.; Clynes, R.; Schmidt, A.M.; Herold, K.C. RAGE expression in human T cells: A link between environmental factors and adaptive immune responses. PLoS ONE 2012, 7, e34698. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, Y.; Akirav, E.M.; Chen, W.; Henegariu, O.; Moser, B.; Desai, D.; Shen, J.M.; Webster, J.C.; Andrews, R.C.; Mjalli, A.M.; et al. RAGE ligation affects T cell activation and controls T cell differentiation. J. Immunol. 2008, 181, 4272–4278. [Google Scholar] [CrossRef] [Green Version]
  124. Manfredi, A.A.; Capobianco, A.; Esposito, A.; De Cobelli, F.; Canu, T.; Monno, A.; Raucci, A.; Sanvito, F.; Doglioni, C.; Nawroth, P.P.; et al. Maturing dendritic cells depend on RAGE for in vivo homing to lymph nodes. J. Immunol. 2008, 180, 2270–2275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Moser, B.; Desai, D.D.; Downie, M.P.; Chen, Y.; Yan, S.F.; Herold, K.; Schmidt, A.M.; Clynes, R. Receptor for advanced glycation end products expression on T cells contributes to antigen-specific cellular expansion in vivo. J. Immunol. 2007, 179, 8051–8058. [Google Scholar] [CrossRef] [PubMed]
  126. Narumi, K.; Miyakawa, R.; Ueda, R.; Hashimoto, H.; Yamamoto, Y.; Yoshida, T.; Aoki, K. Proinflammatory Proteins S100A8/S100A9 Activate NK Cells via Interaction with RAGE. J. Immunol. 2015, 194, 5539–5548. [Google Scholar] [CrossRef] [Green Version]
  127. Perkins, T.N.; Oczypok, E.A.; Dutz, R.E.; Donnell, M.L.; Myerburg, M.M.; Oury, T.D. The receptor for advanced glycation end products is a critical mediator of type 2 cytokine signaling in the lungs. J. Allergy Clin. Immunol. 2019, 144, 796–808.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Oczypok, E.A.; Milutinovic, P.S.; Alcorn, J.F.; Khare, A.; Crum, L.T.; Manni, M.L.; Epperly, M.W.; Pawluk, A.M.; Ray, A.; Oury, T.D. Pulmonary receptor for advanced glycation end-products promotes asthma pathogenesis through IL-33 and accumulation of group 2 innate lymphoid cells. J. Allergy Clin. Immunol. 2015, 136, 747–756.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Hay, A.N.; Potter, A.; Kasmark, L.; Zhu, J.; Leeth, C.M. Rapid Communication: TLR4 expressed but with reduced functionality on equine B lymphocytes. J. Anim. Sci. 2019, 97, 2175–2180. [Google Scholar] [CrossRef]
  130. Zhao, S.; Sun, M.; Meng, H.; Ji, H.; Liu, Y.; Zhang, M.; Li, H.; Li, P.; Zhang, Y.; Zhang, Q. TLR4 expression correlated with PD-L1 expression indicates a poor prognosis in patients with peripheral T-cell lymphomas. Cancer Manag. Res. 2019, 11, 4743–4756. [Google Scholar] [CrossRef] [Green Version]
  131. Rossol, M.; Heine, H.; Meusch, U.; Quandt, D.; Klein, C.; Sweet, M.J.; Hauschildt, S. LPS-induced cytokine production in human monocytes and macrophages. Crit. Rev. Immunol. 2011, 31, 379–446. [Google Scholar] [CrossRef]
  132. Alves-Filho, J.C.; Tavares-Murta, B.M.; Barja-Fidalgo, C.; Benjamim, C.F.; Basile-Filho, A.; Arraes, S.M.; Cunha, F.Q. Neutrophil function in severe sepsis. Endocr. Metab. Immune Disord. Drug Targets 2006, 6, 151–158. [Google Scholar] [CrossRef] [PubMed]
  133. Hwang, Y.H.; Lee, Y.; Paik, M.J.; Yee, S.T. Inhibitions of HMGB1 and TLR4 alleviate DINP-induced asthma in mice. Toxicol. Res. 2019, 8, 621–629. [Google Scholar] [CrossRef] [PubMed]
  134. Shang, L.; Wang, L.; Shi, X.; Wang, N.; Zhao, L.; Wang, J.; Liu, C. HMGB1 was negatively regulated by HSF1 and mediated the TLR4/MyD88/NF-kappaB signal pathway in asthma. Life Sci. 2020, 241, 117120. [Google Scholar] [CrossRef] [PubMed]
  135. Watanabe, T.; Asai, K.; Fujimoto, H.; Tanaka, H.; Kanazawa, H.; Hirata, K. Increased levels of HMGB-1 and endogenous secretory RAGE in induced sputum from asthmatic patients. Respir. Med. 2011, 105, 519–525. [Google Scholar] [CrossRef] [Green Version]
  136. Zhou, Y.; Jiang, Y.Q.; Wang, W.X.; Zhou, Z.X.; Wang, Y.G.; Yang, L.; Ji, Y.L. HMGB1 and RAGE levels in induced sputum correlate with asthma severity and neutrophil percentage. Hum. Immunol. 2012, 73, 1171–1174. [Google Scholar] [CrossRef]
  137. Yang, H.; Hreggvidsdottir, H.S.; Palmblad, K.; Wang, H.; Ochani, M.; Li, J.; Lu, B.; Chavan, S.; Rosas-Ballina, M.; Al-Abed, Y.; et al. A critical cysteine is required for HMGB1 binding to Toll-like receptor 4 and activation of macrophage cytokine release. Proc. Natl. Acad. Sci. USA 2010, 107, 11942–11947. [Google Scholar] [CrossRef] [Green Version]
  138. Andersson, U.; Wang, H.; Palmblad, K.; Aveberger, A.C.; Bloom, O.; Erlandsson-Harris, H.; Janson, A.; Kokkola, R.; Zhang, M.; Yang, H.; et al. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J. Exp. Med. 2000, 192, 565–570. [Google Scholar] [CrossRef]
  139. Zhang, F.; Su, X.; Huang, G.; Xin, X.F.; Cao, E.H.; Shi, Y.; Song, Y. sRAGE alleviates neutrophilic asthma by blocking HMGB1/RAGE signalling in airway dendritic cells. Sci. Rep. 2017, 7, 14268. [Google Scholar] [CrossRef] [Green Version]
  140. Lyu, Y.; Zhao, H.; Ye, Y.; Liu, L.; Zhu, S.; Xia, Y.; Zou, F.; Cai, S. Decreased soluble RAGE in neutrophilic asthma is correlated with disease severity and RAGE G82S variants. Mol. Med. Rep. 2018, 17, 4131–4137. [Google Scholar] [CrossRef] [Green Version]
  141. Patregnani, J.T.; Brooks, B.A.; Chorvinsky, E.; Pillai, D.K. High BAL sRAGE is Associated with Low Serum Eosinophils and IgE in Children with Asthma. Children 2020, 7, 110. [Google Scholar] [CrossRef]
  142. Allam, V.; Faiz, A.; Lam, M.; Rathnayake, S.N.H.; Ditz, B.; Pouwels, S.D.; Brandsma, C.A.; Timens, W.; Hiemstra, P.S.; Tew, G.W.; et al. RAGE and TLR4 differentially regulate airway hyperresponsiveness: Implications for COPD. Allergy 2021, 76, 1123–1135. [Google Scholar] [CrossRef] [PubMed]
  143. Menson, K.E.; Mank, M.M.; Reed, L.F.; Walton, C.J.; Van Der Vliet, K.E.; Ather, J.L.; Chapman, D.G.; Smith, B.J.; Rincon, M.; Poynter, M.E. Therapeutic efficacy of IL-17A neutralization with corticosteroid treatment in a model of antigen-driven mixed-granulocytic asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 2020, 319, L693–L709. [Google Scholar] [CrossRef] [PubMed]
  144. Mack, S.; Shin, J.; Ahn, Y.; Castaneda, A.R.; Peake, J.; Fulgar, C.; Zhang, J.; Cho, Y.H.; Pinkerton, K.E. Age-dependent pulmonary reactivity to house dust mite allergen: A model of adult-onset asthma? Am. J. Physiol. Lung Cell. Mol. Physiol. 2019, 316, L757–L763. [Google Scholar] [CrossRef] [PubMed]
  145. Sadamatsu, H.; Takahashi, K.; Tashiro, H.; Kurihara, Y.; Kato, G.; Uchida, M.; Noguchi, Y.; Kurata, K.; Omura, S.; Sunazuka, T.; et al. The Nonantibiotic Macrolide EM900 Attenuates House Dust Mite-Induced Airway Inflammation in a Mouse Model of Obesity-Associated Asthma. Int. Arch. Allergy Immunol. 2020, 181, 665–674. [Google Scholar] [CrossRef] [PubMed]
  146. Lin, J.; Huang, N.; Li, J.; Liu, X.; Xiong, Q.; Hu, C.; Chen, D.; Guan, L.; Chang, K.; Li, D.; et al. Cross-reactive antibodies against dust mite-derived enolase induce neutrophilic airway inflammation. Eur. Respir. J. 2021, 57, 1902375. [Google Scholar] [CrossRef] [PubMed]
  147. Kwak, D.W.; Park, D.; Kim, J.H. Leukotriene B4 receptors play critical roles in house dust mites-induced neutrophilic airway inflammation and IL-17 production. Biochem. Biophys. Res. Commun. 2021, 534, 646–652. [Google Scholar] [CrossRef]
  148. Mahmutovic Persson, I.; Menzel, M.; Ramu, S.; Cerps, S.; Akbarshahi, H.; Uller, L. IL-1beta mediates lung neutrophilia and IL-33 expression in a mouse model of viral-induced asthma exacerbation. Respir. Res. 2018, 19, 16. [Google Scholar] [CrossRef] [Green Version]
  149. Patel, D.F.; Peiro, T.; Bruno, N.; Vuononvirta, J.; Akthar, S.; Puttur, F.; Pyle, C.J.; Suveizdyte, K.; Walker, S.A.; Singanayagam, A.; et al. Neutrophils restrain allergic airway inflammation by limiting ILC2 function and monocyte-dendritic cell antigen presentation. Sci. Immunol. 2019, 4, eaax7006. [Google Scholar] [CrossRef]
  150. Chalmers, G.W.; Macleod, K.J.; Little, S.A.; Thomson, L.J.; McSharry, C.P.; Thomson, N.C. Influence of cigarette smoking on inhaled corticosteroid treatment in mild asthma. Thorax 2002, 57, 226–230. [Google Scholar] [CrossRef] [Green Version]
  151. Chaudhuri, R.; Livingston, E.; McMahon, A.D.; Lafferty, J.; Fraser, I.; Spears, M.; McSharry, C.P.; Thomson, N.C. Effects of smoking cessation on lung function and airway inflammation in smokers with asthma. Am. J. Respir. Crit. Care Med. 2006, 174, 127–133. [Google Scholar] [CrossRef]
  152. Shimoda, T.; Obase, Y.; Kishikawa, R.; Iwanaga, T. Influence of cigarette smoking on airway inflammation and inhaled corticosteroid treatment in patients with asthma. Allergy Asthma Proc. 2016, 37, 50–58. [Google Scholar] [CrossRef] [PubMed]
  153. Yamasaki, A.; Hanaki, K.; Tomita, K.; Watanabe, M.; Hasagawa, Y.; Okazaki, R.; Igishi, T.; Horimukai, K.; Fukutani, K.; Sugimoto, Y.; et al. Environmental tobacco smoke and its effect on the symptoms and medication in children with asthma. Int. J. Environ. Health Res. 2009, 19, 97–108. [Google Scholar] [CrossRef] [PubMed]
  154. Ghosh, A.; Coakley, R.D.; Ghio, A.J.; Muhlebach, M.S.; Esther, C.R., Jr.; Alexis, N.E.; Tarran, R. Chronic E-Cigarette Use Increases Neutrophil Elastase and Matrix Metalloprotease Levels in the Lung. Am. J. Respir. Crit. Care Med. 2019, 200, 1392–1401. [Google Scholar] [CrossRef] [PubMed]
  155. Schweitzer, R.J.; Wills, T.A.; Tam, E.; Pagano, I.; Choi, K. E-cigarette use and asthma in a multiethnic sample of adolescents. Prev. Med. 2017, 105, 226–231. [Google Scholar] [CrossRef] [PubMed]
  156. Choi, K.; Bernat, D. E-Cigarette Use Among Florida Youth With and Without Asthma. Am. J. Prev. Med. 2016, 51, 446–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Wu, Q.; Jiang, D.; Minor, M.; Chu, H.W. Electronic cigarette liquid increases inflammation and virus infection in primary human airway epithelial cells. PLoS ONE 2014, 9, e108342. [Google Scholar] [CrossRef] [Green Version]
  158. Lerner, C.A.; Sundar, I.K.; Yao, H.; Gerloff, J.; Ossip, D.J.; McIntosh, S.; Robinson, R.; Rahman, I. Vapors produced by electronic cigarettes and e-juices with flavorings induce toxicity, oxidative stress, and inflammatory response in lung epithelial cells and in mouse lung. PLoS ONE 2015, 10, e0116732. [Google Scholar] [CrossRef]
  159. Kioi, Y.; Tabuchi, T. Electronic, heat-not-burn, and combustible cigarette use among chronic disease patients in Japan: A cross-sectional study. Tob. Induc. Dis. 2018, 16, 41. [Google Scholar] [CrossRef]
  160. Schaller, J.P.; Keller, D.; Poget, L.; Pratte, P.; Kaelin, E.; McHugh, D.; Cudazzo, G.; Smart, D.; Tricker, A.R.; Gautier, L.; et al. Evaluation of the Tobacco Heating System 2.2. Part 2: Chemical composition, genotoxicity, cytotoxicity, and physical properties of the aerosol. Regul. Toxicol. Pharmacol. 2016, 81 (Suppl. S2), S27–S47. [Google Scholar] [CrossRef] [Green Version]
  161. Smith, M.R.; Clark, B.; Ludicke, F.; Schaller, J.P.; Vanscheeuwijck, P.; Hoeng, J.; Peitsch, M.C. Evaluation of the Tobacco Heating System 2.2. Part 1: Description of the system and the scientific assessment program. Regul. Toxicol. Pharmacol. 2016, 81 (Suppl. S2), S17–S26. [Google Scholar] [CrossRef] [Green Version]
  162. Protano, C.; Manigrasso, M.; Cammalleri, V.; Biondi Zoccai, G.; Frati, G.; Avino, P.; Vitali, M. Impact of Electronic Alternatives to Tobacco Cigarettes on Indoor Air Particular Matter Levels. Int. J. Environ. Res. Public Health 2020, 17, 2947. [Google Scholar] [CrossRef] [PubMed]
  163. Belvisi, M.G.; Baker, K.; Malloy, N.; Raemdonck, K.; Dekkak, B.; Pieper, M.; Nials, A.T.; Birrell, M.A. Modelling the asthma phenotype: Impact of cigarette smoke exposure. Respir. Res. 2018, 19, 89. [Google Scholar] [CrossRef] [PubMed]
  164. Perret, J.L.; Bonevski, B.; McDonald, C.F.; Abramson, M.J. Smoking cessation strategies for patients with asthma: Improving patient outcomes. J. Asthma Allergy 2016, 9, 117–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Masaki, K.; Tateno, H.; Kameyama, N.; Morino, E.; Watanabe, R.; Sekine, K.; Ono, T.; Satake, K.; Suzuki, S.; Nomura, A.; et al. Impact of a Novel Smartphone App (CureApp Smoking Cessation) on Nicotine Dependence: Prospective Single-Arm Interventional Pilot Study. JMIR Mhealth Uhealth 2019, 7, e12694. [Google Scholar] [CrossRef] [PubMed]
  166. Guarnieri, M.; Balmes, J.R. Outdoor air pollution and asthma. Lancet 2014, 383, 1581–1592. [Google Scholar] [CrossRef] [Green Version]
  167. Tiotiu, A.I.; Novakova, P.; Nedeva, D.; Chong-Neto, H.J.; Novakova, S.; Steiropoulos, P.; Kowal, K. Impact of Air Pollution on Asthma Outcomes. Int. J. Environ. Res. Public Health 2020, 17, 6212. [Google Scholar] [CrossRef] [PubMed]
  168. Gowers, A.M.; Cullinan, P.; Ayres, J.G.; Anderson, H.R.; Strachan, D.P.; Holgate, S.T.; Mills, I.C.; Maynard, R.L. Does outdoor air pollution induce new cases of asthma? Biological plausibility and evidence; a review. Respirology 2012, 17, 887–898. [Google Scholar] [CrossRef] [Green Version]
  169. Wu, J.Z.; Ge, D.D.; Zhou, L.F.; Hou, L.Y.; Zhou, Y.; Li, Q.Y. Effects of particulate matter on allergic respiratory diseases. Chronic Dis. Transl. Med. 2018, 4, 95–102. [Google Scholar] [CrossRef]
  170. Wang, P.; Thevenot, P.; Saravia, J.; Ahlert, T.; Cormier, S.A. Radical-containing particles activate dendritic cells and enhance Th17 inflammation in a mouse model of asthma. Am. J. Respir. Cell Mol. Biol. 2011, 45, 977–983. [Google Scholar] [CrossRef] [Green Version]
  171. van Voorhis, M.; Knopp, S.; Julliard, W.; Fechner, J.H.; Zhang, X.; Schauer, J.J.; Mezrich, J.D. Exposure to atmospheric particulate matter enhances Th17 polarization through the aryl hydrocarbon receptor. PLoS ONE 2013, 8, e82545. [Google Scholar] [CrossRef] [Green Version]
  172. Brandt, E.B.; Kovacic, M.B.; Lee, G.B.; Gibson, A.M.; Acciani, T.H.; Le Cras, T.D.; Ryan, P.H.; Budelsky, A.L.; Khurana Hershey, G.K. Diesel exhaust particle induction of IL-17A contributes to severe asthma. J. Allergy Clin. Immunol. 2013, 132, 1194–1204.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Chang, M.M.; Wu, R.; Plopper, C.G.; Hyde, D.M. IL-8 is one of the major chemokines produced by monkey airway epithelium after ozone-induced injury. Am. J. Physiol. 1998, 275, L524–L532. [Google Scholar] [CrossRef] [PubMed]
  174. Hiltermann, T.J.; Stolk, J.; Hiemstra, P.S.; Fokkens, P.H.; Rombout, P.J.; Sont, J.K.; Sterk, P.J.; Dijkman, J.H. Effect of ozone exposure on maximal airway narrowing in non-asthmatic and asthmatic subjects. Clin. Sci. 1995, 89, 619–624. [Google Scholar] [CrossRef] [PubMed]
  175. Havemann, B.D.; Henderson, C.A.; El-Serag, H.B. The association between gastro-oesophageal reflux disease and asthma: A systematic review. Gut 2007, 56, 1654–1664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Liu, L.; Teague, W.G.; Erzurum, S.; Fitzpatrick, A.; Mantri, S.; Dweik, R.A.; Bleecker, E.R.; Meyers, D.; Busse, W.W.; Calhoun, W.J.; et al. Determinants of exhaled breath condensate pH in a large population with asthma. Chest 2011, 139, 328–336. [Google Scholar] [CrossRef] [Green Version]
  177. Paoletti, G.; Melone, G.; Ferri, S.; Puggioni, F.; Baiardini, I.; Racca, F.; Canonica, G.W.; Heffler, E.; Malipiero, G. Gastroesophageal reflux and asthma: When, how, and why. Curr. Opin. Allergy Clin. Immunol. 2021, 21, 52–58. [Google Scholar] [CrossRef]
  178. Simpson, J.L.; Baines, K.J.; Ryan, N.; Gibson, P.G. Neutrophilic asthma is characterised by increased rhinosinusitis with sleep disturbance and GERD. Asian Pac. J. Allergy Immunol. 2014, 32, 66–74. [Google Scholar] [CrossRef]
  179. Icitovic, N.; Onyebeke, L.C.; Wallenstein, S.; Dasaro, C.R.; Harrison, D.; Jiang, J.; Kaplan, J.R.; Lucchini, R.G.; Luft, B.J.; Moline, J.M.; et al. The association between body mass index and gastroesophageal reflux disease in the World Trade Center Health Program General Responder Cohort. Am. J. Ind. Med. 2016, 59, 761–766. [Google Scholar] [CrossRef]
  180. Gupta, S.; Lodha, R.; Kabra, S.K. Asthma, GERD and Obesity: Triangle of Inflammation. Indian J. Pediatr. 2018, 85, 887–892. [Google Scholar] [CrossRef]
  181. Chupp, G.L.; Lee, C.G.; Jarjour, N.; Shim, Y.M.; Holm, C.T.; He, S.; Dziura, J.D.; Reed, J.; Coyle, A.J.; Kiener, P.; et al. A chitinase-like protein in the lung and circulation of patients with severe asthma. N. Engl. J. Med. 2007, 357, 2016–2027. [Google Scholar] [CrossRef]
  182. James, A.J.; Reinius, L.E.; Verhoek, M.; Gomes, A.; Kupczyk, M.; Hammar, U.; Ono, J.; Ohta, S.; Izuhara, K.; Bel, E.; et al. Increased YKL-40 and Chitotriosidase in Asthma and Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2016, 193, 131–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Liu, L.; Zhang, X.; Liu, Y.; Zhang, L.; Zheng, J.; Wang, J.; Hansbro, P.M.; Wang, L.; Wang, G.; Hsu, A.C. Chitinase-like protein YKL-40 correlates with inflammatory phenotypes, anti-asthma responsiveness and future exacerbations. Respir. Res. 2019, 20, 95. [Google Scholar] [CrossRef] [PubMed]
  184. Suzuki, Y.; Saito, J.; Munakata, M.; Shibata, Y. Hydrogen sulfide as a novel biomarker of asthma and chronic obstructive pulmonary disease. Allergol. Int. 2021, 70, 181–189. [Google Scholar] [CrossRef] [PubMed]
  185. Saito, J.; Zhang, Q.; Hui, C.; Macedo, P.; Gibeon, D.; Menzies-Gow, A.; Bhavsar, P.K.; Chung, K.F. Sputum hydrogen sulfide as a novel biomarker of obstructive neutrophilic asthma. J. Allergy Clin. Immunol. 2013, 131, 232–234.e3. [Google Scholar] [CrossRef]
  186. Suzuki, Y.; Saito, J.; Kikuchi, M.; Uematsu, M.; Fukuhara, A.; Sato, S.; Munakata, M. Sputum-to-serum hydrogen sulphide ratio as a novel biomarker of predicting future risks of asthma exacerbation. Clin. Exp. Allergy 2018, 48, 1155–1163. [Google Scholar] [CrossRef] [Green Version]
  187. Hinks, T.S.C.; Brown, T.; Lau, L.C.K.; Rupani, H.; Barber, C.; Elliott, S.; Ward, J.A.; Ono, J.; Ohta, S.; Izuhara, K.; et al. Multidimensional endotyping in patients with severe asthma reveals inflammatory heterogeneity in matrix metalloproteinases and chitinase 3-like protein 1. J. Allergy Clin. Immunol. 2016, 138, 61–75. [Google Scholar] [CrossRef] [Green Version]
  188. Backman, H.; Lindberg, A.; Hedman, L.; Stridsman, C.; Jansson, S.A.; Sandstrom, T.; Lundback, B.; Ronmark, E. FEV1 decline in relation to blood eosinophils and neutrophils in a population-based asthma cohort. World Allergy Organ. J. 2020, 13, 100110. [Google Scholar] [CrossRef]
  189. Backman, H.; Jansson, S.A.; Stridsman, C.; Muellerova, H.; Wurst, K.; Hedman, L.; Lindberg, A.; Ronmark, E. Chronic airway obstruction in a population-based adult asthma cohort: Prevalence, incidence and prognostic factors. Respir. Med. 2018, 138, 115–122. [Google Scholar] [CrossRef]
  190. Huang, Y.; Zhang, S.; Fang, X.; Qin, L.; Fan, Y.; Ding, D.; Liu, X.; Xie, M. Plasma miR-199a-5p is increased in neutrophilic phenotype asthma patients and negatively correlated with pulmonary function. PLoS ONE 2018, 13, e0193502. [Google Scholar] [CrossRef]
  191. Maes, T.; Cobos, F.A.; Schleich, F.; Sorbello, V.; Henket, M.; De Preter, K.; Bracke, K.R.; Conickx, G.; Mesnil, C.; Vandesompele, J.; et al. Asthma inflammatory phenotypes show differential microRNA expression in sputum. J. Allergy Clin. Immunol. 2016, 137, 1433–1446. [Google Scholar] [CrossRef] [Green Version]
  192. Jafari-Nakhjavani, M.R.; Ghorbanihaghjo, A.; Bagherzadeh-Nobari, B.; Malek-Mahdavi, A.; Rashtchizadeh, N. Serum YKL-40 levels and disease characteristics in patients with rheumatoid arthritis. Casp. J. Intern. Med. 2019, 10, 92–97. [Google Scholar] [CrossRef]
  193. Malmestrom, C.; Axelsson, M.; Lycke, J.; Zetterberg, H.; Blennow, K.; Olsson, B. CSF levels of YKL-40 are increased in MS and replaces with immunosuppressive treatment. J. Neuroimmunol. 2014, 269, 87–89. [Google Scholar] [CrossRef] [PubMed]
  194. Przysucha, N.; Gorska, K.; Krenke, R. Chitinases and Chitinase-Like Proteins in Obstructive Lung Diseases—Current Concepts and Potential Applications. Int. J. Chronic Obstr. Pulm. Dis. 2020, 15, 885–899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Tong, X.; Wang, D.; Liu, S.; Ma, Y.; Li, Z.; Tian, P.; Fan, H. The YKL-40 protein is a potential biomarker for COPD: A meta-analysis and systematic review. Int. J. Chronic Obstr. Pulm. Dis. 2018, 13, 409–418. [Google Scholar] [CrossRef] [PubMed]
  196. Olsson, B.; Lautner, R.; Andreasson, U.; Ohrfelt, A.; Portelius, E.; Bjerke, M.; Holtta, M.; Rosen, C.; Olsson, C.; Strobel, G.; et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: A systematic review and meta-analysis. Lancet Neurol. 2016, 15, 673–684. [Google Scholar] [CrossRef]
  197. Harrison, L.I.; Schuppan, D.; Rohlfing, S.R.; Hansen, A.R.; Hansen, C.S.; Funk, M.L.; Collins, S.H.; Ober, R.E. Determination of flumequine and a hydroxy metabolite in biological fluids by high-pressure liquid chromatographic, fluorometric, and microbiological methods. Antimicrob. Agents Chemother. 1984, 25, 301–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Harnan, S.E.; Essat, M.; Gomersall, T.; Tappenden, P.; Pavord, I.; Everard, M.; Lawson, R. Exhaled nitric oxide in the diagnosis of asthma in adults: A systematic review. Clin. Exp. Allergy 2017, 47, 410–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Zhang, J.; Yao, X.; Yu, R.; Bai, J.; Sun, Y.; Huang, M.; Adcock, I.M.; Barnes, P.J. Exhaled carbon monoxide in asthmatics: A meta-analysis. Respir. Res. 2010, 11, 50. [Google Scholar] [CrossRef]
  200. Gao, J.; Iwamoto, H.; Koskela, J.; Alenius, H.; Hattori, N.; Kohno, N.; Laitinen, T.; Mazur, W.; Pulkkinen, V. Characterization of sputum biomarkers for asthma-COPD overlap syndrome. Int. J. Chronic Obstr. Pulm. Dis. 2016, 11, 2457–2465. [Google Scholar] [CrossRef] [Green Version]
  201. Zhang, X.Y.; Simpson, J.L.; Powell, H.; Yang, I.A.; Upham, J.W.; Reynolds, P.N.; Hodge, S.; James, A.L.; Jenkins, C.; Peters, M.J.; et al. Full blood count parameters for the detection of asthma inflammatory phenotypes. Clin. Exp. Allergy 2014, 44, 1137–1145. [Google Scholar] [CrossRef] [Green Version]
  202. Hastie, A.T.; Moore, W.C.; Li, H.; Rector, B.M.; Ortega, V.E.; Pascual, R.M.; Peters, S.P.; Meyers, D.A.; Bleecker, E.R.; National Heart, L.; et al. Biomarker surrogates do not accurately predict sputum eosinophil and neutrophil percentages in asthmatic subjects. J. Allergy Clin. Immunol. 2013, 132, 72–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Hartjes, F.J.; Vonk, J.M.; Faiz, A.; Hiemstra, P.S.; Lapperre, T.S.; Kerstjens, H.A.M.; Postma, D.S.; van den Berge, M.; Groningen and Leiden Universities Corticosteroids in Obstructive Lung Disease (GLUCOLD) Study Group. Predictive value of eosinophils and neutrophils on clinical effects of ICS in COPD. Respirology 2018, 23, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  204. Panganiban, R.P.; Pinkerton, M.H.; Maru, S.Y.; Jefferson, S.J.; Roff, A.N.; Ishmael, F.T. Differential microRNA epression in asthma and the role of miR-1248 in regulation of IL-5. Am. J. Clin. Exp. Immunol. 2012, 1, 154–165. [Google Scholar]
  205. Milger, K.; Gotschke, J.; Krause, L.; Nathan, P.; Alessandrini, F.; Tufman, A.; Fischer, R.; Bartel, S.; Theis, F.J.; Behr, J.; et al. Identification of a plasma miRNA biomarker signature for allergic asthma: A translational approach. Allergy 2017, 72, 1962–1971. [Google Scholar] [CrossRef] [PubMed]
  206. Gomez, J.L.; Chen, A.; Diaz, M.P.; Zirn, N.; Gupta, A.; Britto, C.; Sauler, M.; Yan, X.; Stewart, E.; Santerian, K.; et al. A Network of Sputum MicroRNAs Is Associated with Neutrophilic Airway Inflammation in Asthma. Am. J. Respir. Crit. Care Med. 2020, 202, 51–64. [Google Scholar] [CrossRef] [PubMed]
  207. Canas, J.A.; Rodrigo-Munoz, J.M.; Sastre, B.; Gil-Martinez, M.; Redondo, N.; Del Pozo, V. MicroRNAs as Potential Regulators of Immune Response Networks in Asthma and Chronic Obstructive Pulmonary Disease. Front. Immunol. 2020, 11, 608666. [Google Scholar] [CrossRef]
  208. Specjalski, K.; Niedoszytko, M. MicroRNAs: Future biomarkers and targets of therapy in asthma? Curr. Opin. Pulm. Med. 2020, 26, 285–292. [Google Scholar] [CrossRef]
  209. Gelfand, E.W. Importance of the leukotriene B4-BLT1 and LTB4-BLT2 pathways in asthma. Semin. Immunol. 2017, 33, 44–51. [Google Scholar] [CrossRef]
  210. Ford-Hutchinson, A.W.; Bray, M.A.; Doig, M.V.; Shipley, M.E.; Smith, M.J. Leukotriene B, a potent chemokinetic and aggregating substance released from polymorphonuclear leukocytes. Nature 1980, 286, 264–265. [Google Scholar] [CrossRef]
  211. Teixeira, M.M.; Lindsay, M.A.; Giembycz, M.A.; Hellewell, P.G. Role of arachidonic acid in leukotriene B(4)-induced guinea-pig eosinophil homotypic aggregation. Eur. J. Pharmacol. 1999, 384, 183–190. [Google Scholar] [CrossRef]
  212. Ng, C.F.; Sun, F.F.; Taylor, B.M.; Wolin, M.S.; Wong, P.Y. Functional properties of guinea pig eosinophil leukotriene B4 receptor. J. Immunol. 1991, 147, 3096–3103. [Google Scholar] [PubMed]
  213. Watanabe, S.; Yamasaki, A.; Hashimoto, K.; Shigeoka, Y.; Chikumi, H.; Hasegawa, Y.; Sumikawa, T.; Takata, M.; Okazaki, R.; Watanabe, M.; et al. Expression of functional leukotriene B4 receptors on human airway smooth muscle cells. J. Allergy Clin. Immunol. 2009, 124, 59–65.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Lamblin, C.; Gosset, P.; Tillie-Leblond, I.; Saulnier, F.; Marquette, C.H.; Wallaert, B.; Tonnel, A.B. Bronchial neutrophilia in patients with noninfectious status asthmaticus. Am. J. Respir. Crit. Care Med. 1998, 157, 394–402. [Google Scholar] [CrossRef]
  215. Wood, L.G.; Baines, K.J.; Fu, J.; Scott, H.A.; Gibson, P.G. The neutrophilic inflammatory phenotype is associated with systemic inflammation in asthma. Chest 2012, 142, 86–93. [Google Scholar] [CrossRef]
  216. Hosoki, K.; Ying, S.; Corrigan, C.; Qi, H.; Kurosky, A.; Jennings, K.; Sun, Q.; Boldogh, I.; Sur, S. Analysis of a Panel of 48 Cytokines in BAL Fluids Specifically Identifies IL-8 Levels as the Only Cytokine that Distinguishes Controlled Asthma from Uncontrolled Asthma, and Correlates Inversely with FEV1. PLoS ONE 2015, 10, e0126035. [Google Scholar] [CrossRef]
  217. Kuo, P.L.; Hsu, Y.L.; Huang, M.S.; Chiang, S.L.; Ko, Y.C. Bronchial epithelium-derived IL-8 and RANTES increased bronchial smooth muscle cell migration and proliferation by Kruppel-like factor 5 in areca nut-mediated airway remodeling. Toxicol. Sci. 2011, 121, 177–190. [Google Scholar] [CrossRef] [Green Version]
  218. Govindaraju, V.; Michoud, M.C.; Al-Chalabi, M.; Ferraro, P.; Powell, W.S.; Martin, J.G. Interleukin-8: Novel roles in human airway smooth muscle cell contraction and migration. Am. J. Physiol. Cell Physiol. 2006, 291, C957–C965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Halwani, R.; Al-Abri, J.; Beland, M.; Al-Jahdali, H.; Halayko, A.J.; Lee, T.H.; Al-Muhsen, S.; Hamid, Q. CC and CXC chemokines induce airway smooth muscle proliferation and survival. J. Immunol. 2011, 186, 4156–4163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Tamaoki, J.; Nakata, J.; Tagaya, E.; Konno, K. Effects of roxithromycin and erythromycin on interleukin 8-induced neutrophil recruitment and goblet cell secretion in guinea pig tracheas. Antimicrob. Agents Chemother. 1996, 40, 1726–1728. [Google Scholar] [CrossRef] [Green Version]
  221. Smirnova, M.G.; Birchall, J.P.; Pearson, J.P. In vitro study of IL-8 and goblet cells: Possible role of IL-8 in the aetiology of otitis media with effusion. Acta Oto-Laryngol. 2002, 122, 146–152. [Google Scholar] [CrossRef]
  222. Tang, H.; Sun, Y.; Shi, Z.; Huang, H.; Fang, Z.; Chen, J.; Xiu, Q.; Li, B. YKL-40 induces IL-8 expression from bronchial epithelium via MAPK (JNK and ERK) and NF-kappaB pathways, causing bronchial smooth muscle proliferation and migration. J. Immunol. 2013, 190, 438–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Li, X.; Zou, F.; Lu, Y.; Fan, X.; Wu, Y.; Feng, X.; Sun, X.; Liu, Y. Notch1 contributes to TNF-alpha-induced proliferation and migration of airway smooth muscle cells through regulation of the Hes1/PTEN axis. Int. Immunopharmacol. 2020, 88, 106911. [Google Scholar] [CrossRef] [PubMed]
  224. Sun, M.; Huang, Y.; Li, F.; Li, H.; Zhang, B.; Jin, L. MicroRNA-874 inhibits TNF-alpha-induced remodeling in human fetal airway smooth muscle cells by targeting STAT3. Respir. Physiol. Neurobiol. 2018, 251, 34–40. [Google Scholar] [CrossRef] [PubMed]
  225. Cho, J.Y.; Pham, A.; Rosenthal, P.; Miller, M.; Doherty, T.; Broide, D.H. Chronic OVA allergen challenged TNF p55/p75 receptor deficient mice have reduced airway remodeling. Int. Immunopharmacol. 2011, 11, 1038–1044. [Google Scholar] [CrossRef] [Green Version]
  226. Dejager, L.; Dendoncker, K.; Eggermont, M.; Souffriau, J.; Van Hauwermeiren, F.; Willart, M.; Van Wonterghem, E.; Naessens, T.; Ballegeer, M.; Vandevyver, S.; et al. Neutralizing TNFalpha restores glucocorticoid sensitivity in a mouse model of neutrophilic airway inflammation. Mucosal Immunol. 2015, 8, 1212–1225. [Google Scholar] [CrossRef] [Green Version]
  227. Agache, I.; Ciobanu, C.; Agache, C.; Anghel, M. Increased serum IL-17 is an independent risk factor for severe asthma. Respir. Med. 2010, 104, 1131–1137. [Google Scholar] [CrossRef] [Green Version]
  228. Vittal, R.; Fan, L.; Greenspan, D.S.; Mickler, E.A.; Gopalakrishnan, B.; Gu, H.; Benson, H.L.; Zhang, C.; Burlingham, W.; Cummings, O.W.; et al. IL-17 induces type V collagen overexpression and EMT via TGF-beta-dependent pathways in obliterative bronchiolitis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013, 304, L401–L414. [Google Scholar] [CrossRef] [Green Version]
  229. Chen, Y.; Thai, P.; Zhao, Y.H.; Ho, Y.S.; DeSouza, M.M.; Wu, R. Stimulation of airway mucin gene expression by interleukin (IL)-17 through IL-6 paracrine/autocrine loop. J. Biol. Chem. 2003, 278, 17036–17043. [Google Scholar] [CrossRef] [Green Version]
  230. Fujisawa, T.; Chang, M.M.; Velichko, S.; Thai, P.; Hung, L.Y.; Huang, F.; Phuong, N.; Chen, Y.; Wu, R. NF-kappaB mediates IL-1beta- and IL-17A-induced MUC5B expression in airway epithelial cells. Am. J. Respir. Cell Mol. Biol. 2011, 45, 246–252. [Google Scholar] [CrossRef] [Green Version]
  231. Fujisawa, T.; Velichko, S.; Thai, P.; Hung, L.Y.; Huang, F.; Wu, R. Regulation of airway MUC5AC expression by IL-1beta and IL-17A; the NF-kappaB paradigm. J. Immunol. 2009, 183, 6236–6243. [Google Scholar] [CrossRef] [Green Version]
  232. Wang, T.; Liu, Y.; Zou, J.F.; Cheng, Z.S. Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via the TGF-beta1 mediated Smad2/3 and ERK1/2 activation. PLoS ONE 2017, 12, e0183972. [Google Scholar] [CrossRef]
  233. Ogawa, H.; Azuma, M.; Tsunematsu, T.; Morimoto, Y.; Kondo, M.; Tezuka, T.; Nishioka, Y.; Tsuneyama, K. Neutrophils induce smooth muscle hyperplasia via neutrophil elastase-induced FGF-2 in a mouse model of asthma with mixed inflammation. Clin. Exp. Allergy 2018, 48, 1715–1725. [Google Scholar] [CrossRef] [PubMed]
  234. Camargo, L.D.N.; Dos Santos, T.M.; de Andrade, F.C.P.; Fukuzaki, S.; Dos Santos Lopes, F.; de Arruda Martins, M.; Prado, C.M.; Leick, E.A.; Righetti, R.F.; Tiberio, I. Bronchial Vascular Remodeling Is Attenuated by Anti-IL-17 in Asthmatic Responses Exacerbated by LPS. Front. Pharmacol. 2020, 11, 1269. [Google Scholar] [CrossRef]
  235. Camargo, L.D.N.; Righetti, R.F.; Aristoteles, L.; Dos Santos, T.M.; de Souza, F.C.R.; Fukuzaki, S.; Cruz, M.M.; Alonso-Vale, M.I.C.; Saraiva-Romanholo, B.M.; Prado, C.M.; et al. Effects of Anti-IL-17 on Inflammation, Remodeling, and Oxidative Stress in an Experimental Model of Asthma Exacerbated by LPS. Front. Immunol. 2017, 8, 1835. [Google Scholar] [CrossRef]
  236. Ramakrishnan, R.K.; Al Heialy, S.; Hamid, Q. Role of IL-17 in asthma pathogenesis and its implications for the clinic. Expert Rev. Respir. Med. 2019, 13, 1057–1068. [Google Scholar] [CrossRef] [PubMed]
  237. Liao, Z.; Xiao, H.T.; Zhang, Y.; Tong, R.S.; Zhang, L.J.; Bian, Y.; He, X. IL-1beta: A key modulator in asthmatic airway smooth muscle hyper-reactivity. Expert Rev. Respir. Med. 2015, 9, 429–436. [Google Scholar] [CrossRef] [PubMed]
  238. Lappalainen, U.; Whitsett, J.A.; Wert, S.E.; Tichelaar, J.W.; Bry, K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am. J. Respir. Cell Mol. Biol. 2005, 32, 311–318. [Google Scholar] [CrossRef]
  239. Mehta, A.K.; Doherty, T.; Broide, D.; Croft, M. Tumor necrosis factor family member LIGHT acts with IL-1beta and TGF-beta to promote airway remodeling during rhinovirus infection. Allergy 2018, 73, 1415–1424. [Google Scholar] [CrossRef]
  240. Pothoven, K.L.; Norton, J.E.; Hulse, K.E.; Suh, L.A.; Carter, R.G.; Rocci, E.; Harris, K.E.; Shintani-Smith, S.; Conley, D.B.; Chandra, R.K.; et al. Oncostatin M promotes mucosal epithelial barrier dysfunction, and its expression is increased in patients with eosinophilic mucosal disease. J. Allergy Clin. Immunol. 2015, 136, 737–746.e4. [Google Scholar] [CrossRef] [Green Version]
  241. Pothoven, K.L.; Norton, J.E.; Suh, L.A.; Carter, R.G.; Harris, K.E.; Biyasheva, A.; Welch, K.; Shintani-Smith, S.; Conley, D.B.; Liu, M.C.; et al. Neutrophils are a major source of the epithelial barrier disrupting cytokine oncostatin M in patients with mucosal airways disease. J. Allergy Clin. Immunol. 2017, 139, 1966–1978.e9. [Google Scholar] [CrossRef]
  242. Simpson, J.L.; Baines, K.J.; Boyle, M.J.; Scott, R.J.; Gibson, P.G. Oncostatin M (OSM) is increased in asthma with incompletely reversible airflow obstruction. Exp. Lung Res. 2009, 35, 781–794. [Google Scholar] [CrossRef] [PubMed]
  243. Ventura, I.; Vega, A.; Chacon, P.; Chamorro, C.; Aroca, R.; Gomez, E.; Bellido, V.; Puente, Y.; Blanca, M.; Monteseirin, J. Neutrophils from allergic asthmatic patients produce and release metalloproteinase-9 upon direct exposure to allergens. Allergy 2014, 69, 898–905. [Google Scholar] [CrossRef] [PubMed]
  244. Cundall, M.; Sun, Y.; Miranda, C.; Trudeau, J.B.; Barnes, S.; Wenzel, S.E. Neutrophil-derived matrix metalloproteinase-9 is increased in severe asthma and poorly inhibited by glucocorticoids. J. Allergy Clin. Immunol. 2003, 112, 1064–1071. [Google Scholar] [CrossRef] [PubMed]
  245. Nadel, J.A. Role of enzymes from inflammatory cells on airway submucosal gland secretion. Respiration 1991, 58 (Suppl. S1), 3–5. [Google Scholar] [CrossRef]
  246. McGrath, K.W.; Icitovic, N.; Boushey, H.A.; Lazarus, S.C.; Sutherland, E.R.; Chinchilli, V.M.; Fahy, J.V.; Asthma Clinical Research Network of the National Heart, Lung, and Blood Institute. A large subgroup of mild-to-moderate asthma is persistently noneosinophilic. Am. J. Respir. Crit. Care Med. 2012, 185, 612–619. [Google Scholar] [CrossRef]
  247. Barnes, P.J. Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap syndromes. J. Allergy Clin. Immunol. 2015, 136, 531–545. [Google Scholar] [CrossRef]
  248. Westergaard, C.G.; Porsbjerg, C.; Backer, V. The effect of smoking cessation on airway inflammation in young asthma patients. Clin. Exp. Allergy 2014, 44, 353–361. [Google Scholar] [CrossRef]
  249. Pakhale, S.; Baron, J.; Dent, R.; Vandemheen, K.; Aaron, S.D. Effects of weight loss on airway responsiveness in obese adults with asthma: Does weight loss lead to reversibility of asthma? Chest 2015, 147, 1582–1590. [Google Scholar] [CrossRef]
  250. Simpson, J.L.; Powell, H.; Boyle, M.J.; Scott, R.J.; Gibson, P.G. Clarithromycin targets neutrophilic airway inflammation in refractory asthma. Am. J. Respir. Crit. Care Med. 2008, 177, 148–155. [Google Scholar] [CrossRef] [Green Version]
  251. Bardin, P.; Kanniess, F.; Gauvreau, G.; Bredenbroker, D.; Rabe, K.F. Roflumilast for asthma: Efficacy findings in mechanism of action studies. Pulm. Pharmacol. Ther. 2015, 35, S4–S10. [Google Scholar] [CrossRef]
  252. Casale, T.B.; Aalbers, R.; Bleecker, E.R.; Meltzer, E.O.; Zaremba-Pechmann, L.; de la Hoz, A.; Kerstjens, H.A.M. Tiotropium Respimat(R) add-on therapy to inhaled corticosteroids in patients with symptomatic asthma improves clinical outcomes regardless of baseline characteristics. Respir. Med. 2019, 158, 97–109. [Google Scholar] [CrossRef] [PubMed]
  253. Szefler, S.J.; Vogelberg, C.; Bernstein, J.A.; Goldstein, S.; Mansfield, L.; Zaremba-Pechmann, L.; Engel, M.; Hamelmann, E. Tiotropium Is Efficacious in 6- to 17-Year-Olds with Asthma, Independent of T2 Phenotype. J. Allergy Clin. Immunol. Pract. 2019, 7, 2286–2295.e4. [Google Scholar] [CrossRef] [PubMed]
  254. Nair, P.; Gaga, M.; Zervas, E.; Alagha, K.; Hargreave, F.E.; O’Byrne, P.M.; Stryszak, P.; Gann, L.; Sadeh, J.; Chanez, P.; et al. Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: A randomized, placebo-controlled clinical trial. Clin. Exp. Allergy 2012, 42, 1097–1103. [Google Scholar] [CrossRef] [PubMed]
  255. Follows, R.M.; Snowise, N.G.; Ho, S.Y.; Ambery, C.L.; Smart, K.; McQuade, B.A. Efficacy, safety and tolerability of GSK2190915, a 5-lipoxygenase activating protein inhibitor, in adults and adolescents with persistent asthma: A randomised dose-ranging study. Respir. Res. 2013, 14, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. O’Connor, B.J.; Lofdahl, C.G.; Balter, M.; Szczeklik, A.; Boulet, L.P.; Cairns, C.B. Zileuton added to low-dose inhaled beclomethasone for the treatment of moderate to severe persistent asthma. Respir. Med. 2007, 101, 1088–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Menzies-Gow, A.; Corren, J.; Bourdin, A.; Chupp, G.; Israel, E.; Wechsler, M.E.; Brightling, C.E.; Griffiths, J.M.; Hellqvist, A.; Bowen, K.; et al. Tezepelumab in Adults and Adolescents with Severe, Uncontrolled Asthma. N. Engl. J. Med. 2021, 384, 1800–1809. [Google Scholar] [CrossRef]
  258. Wenzel, S.E.; Barnes, P.J.; Bleecker, E.R.; Bousquet, J.; Busse, W.; Dahlen, S.E.; Holgate, S.T.; Meyers, D.A.; Rabe, K.F.; Antczak, A.; et al. A randomized, double-blind, placebo-controlled study of tumor necrosis factor-alpha blockade in severe persistent asthma. Am. J. Respir. Crit. Care Med. 2009, 179, 549–558. [Google Scholar] [CrossRef]
  259. Holgate, S.T.; Noonan, M.; Chanez, P.; Busse, W.; Dupont, L.; Pavord, I.; Hakulinen, A.; Paolozzi, L.; Wajdula, J.; Zang, C.; et al. Efficacy and safety of etanercept in moderate-to-severe asthma: A randomised, controlled trial. Eur. Respir. J. 2011, 37, 1352–1359. [Google Scholar] [CrossRef]
  260. Busse, W.W.; Holgate, S.; Kerwin, E.; Chon, Y.; Feng, J.; Lin, J.; Lin, S.L. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am. J. Respir. Crit. Care Med. 2013, 188, 1294–1302. [Google Scholar] [CrossRef]
  261. Brightling, C.E.; Nair, P.; Louis, R.; Singh, D. Risankizumab in severe asthma: A Phase IIa, placebo-controlled study. Eur. Respir. J. 2020, 56, 3699. [Google Scholar]
  262. Revez, J.A.; Bain, L.M.; Watson, R.M.; Towers, M.; Collins, T.; Killian, K.J.; O’Byrne, P.M.; Gauvreau, G.M.; Upham, J.W.; Ferreira, M.A. Effects of interleukin-6 receptor blockade on allergen-induced airway responses in mild asthmatics. Clin. Transl. Immunol. 2019, 8, e1044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Scott, H.A.; Gibson, P.G.; Garg, M.L.; Pretto, J.J.; Morgan, P.J.; Callister, R.; Wood, L.G. Dietary restriction and exercise improve airway inflammation and clinical outcomes in overweight and obese asthma: A randomized trial. Clin. Exp. Allergy 2013, 43, 36–49. [Google Scholar] [CrossRef] [PubMed]
  264. Boulet, L.P.; Turcotte, H.; Martin, J.; Poirier, P. Effect of bariatric surgery on airway response and lung function in obese subjects with asthma. Respir. Med. 2012, 106, 651–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Freitas, P.D.; Ferreira, P.G.; Silva, A.G.; Stelmach, R.; Carvalho-Pinto, R.M.; Fernandes, F.L.; Mancini, M.C.; Sato, M.N.; Martins, M.A.; Carvalho, C.R. The Role of Exercise in a Weight-Loss Program on Clinical Control in Obese Adults with Asthma. A Randomized Controlled Trial. Am. J. Respir. Crit. Care Med. 2017, 195, 32–42. [Google Scholar] [CrossRef]
  266. da Silva, P.L.; de Mello, M.T.; Cheik, N.C.; Sanches, P.L.; Correia, F.A.; de Piano, A.; Corgosinho, F.C.; Campos, R.M.; do Nascimento, C.M.; Oyama, L.M.; et al. Interdisciplinary therapy improves biomarkers profile and lung function in asthmatic obese adolescents. Pediatr. Pulmonol. 2012, 47, 8–17. [Google Scholar] [CrossRef]
  267. Crosbie, P.A.; Woodhead, M.A. Long-term macrolide therapy in chronic inflammatory airway diseases. Eur. Respir. J. 2009, 33, 171–181. [Google Scholar] [CrossRef]
  268. Kraft, M.; Cassell, G.H.; Pak, J.; Martin, R.J. Mycoplasma pneumoniae and Chlamydia pneumoniae in asthma: Effect of clarithromycin. Chest 2002, 121, 1782–1788. [Google Scholar] [CrossRef]
  269. Gibson, P.G.; Yang, I.A.; Upham, J.W.; Reynolds, P.N.; Hodge, S.; James, A.L.; Jenkins, C.; Peters, M.J.; Marks, G.B.; Baraket, M.; et al. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): A randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 659–668. [Google Scholar] [CrossRef] [Green Version]
  270. Taylor, S.L.; Ivey, K.L.; Gibson, P.G.; Simpson, J.L.; Rogers, G.B.; Group, A.S.R. Airway abundance of Haemophilus influenzae predicts response to azithromycin in adults with persistent uncontrolled asthma. Eur. Respir. J. 2020, 56, 2000194. [Google Scholar] [CrossRef]
  271. Niessen, N.M.; Gibson, P.G.; Baines, K.J.; Barker, D.; Yang, I.A.; Upham, J.W.; Reynolds, P.N.; Hodge, S.; James, A.L.; Jenkins, C.; et al. Sputum TNF markers are increased in neutrophilic and severe asthma and are reduced by azithromycin treatment. Allergy 2021, 76, 2090–2101. [Google Scholar] [CrossRef]
  272. Brusselle, G.G.; Vanderstichele, C.; Jordens, P.; Deman, R.; Slabbynck, H.; Ringoet, V.; Verleden, G.; Demedts, I.K.; Verhamme, K.; Delporte, A.; et al. Azithromycin for prevention of exacerbations in severe asthma (AZISAST): A multicentre randomised double-blind placebo-controlled trial. Thorax 2013, 68, 322–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Calverley, P.M.; Rabe, K.F.; Goehring, U.M.; Kristiansen, S.; Fabbri, L.M.; Martinez, F.J.; The M2-124 and M2-125 Study Groups. Roflumilast in symptomatic chronic obstructive pulmonary disease: Two randomised clinical trials. Lancet 2009, 374, 685–694. [Google Scholar] [CrossRef]
  274. Zhang, X.; Chen, Y.; Fan, L.; Ye, J.; Fan, J.; Xu, X.; You, D.; Liu, S.; Chen, X.; Luo, P. Pharmacological mechanism of roflumilast in the treatment of asthma-COPD overlap. Drug Des. Dev. Ther. 2018, 12, 2371–2379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Timmer, W.; Leclerc, V.; Birraux, G.; Neuhauser, M.; Hatzelmann, A.; Bethke, T.; Wurst, W. The new phosphodiesterase 4 inhibitor roflumilast is efficacious in exercise-induced asthma and leads to suppression of LPS-stimulated TNF-alpha ex vivo. J. Clin. Pharmacol. 2002, 42, 297–303. [Google Scholar] [CrossRef]
  276. Bousquet, J.; Aubier, M.; Sastre, J.; Izquierdo, J.L.; Adler, L.M.; Hofbauer, P.; Rost, K.D.; Harnest, U.; Kroemer, B.; Albrecht, A.; et al. Comparison of roflumilast, an oral anti-inflammatory, with beclomethasone dipropionate in the treatment of persistent asthma. Allergy 2006, 61, 72–78. [Google Scholar] [CrossRef]
  277. Bateman, E.D.; Goehring, U.M.; Richard, F.; Watz, H. Roflumilast combined with montelukast versus montelukast alone as add-on treatment in patients with moderate-to-severe asthma. J. Allergy Clin. Immunol. 2016, 138, 142–149.e8. [Google Scholar] [CrossRef] [Green Version]
  278. Gauvreau, G.M.; Boulet, L.P.; Schmid-Wirlitsch, C.; Cote, J.; Duong, M.; Killian, K.J.; Milot, J.; Deschesnes, F.; Strinich, T.; Watson, R.M.; et al. Roflumilast attenuates allergen-induced inflammation in mild asthmatic subjects. Respir. Res. 2011, 12, 140. [Google Scholar] [CrossRef]
  279. Phillips, J.E. Inhaled Phosphodiesterase 4 (PDE4) Inhibitors for Inflammatory Respiratory Diseases. Front. Pharmacol. 2020, 11, 259. [Google Scholar] [CrossRef] [Green Version]
  280. Singh, D.; Leaker, B.; Boyce, M.; Nandeuil, M.A.; Collarini, S.; Mariotti, F.; Santoro, D.; Barnes, P.J. A novel inhaled phosphodiesterase 4 inhibitor (CHF6001) reduces the allergen challenge response in asthmatic patients. Pulm. Pharmacol. Ther. 2016, 40, 1–6. [Google Scholar] [CrossRef]
  281. Franciosi, L.G.; Diamant, Z.; Banner, K.H.; Zuiker, R.; Morelli, N.; Kamerling, I.M.; de Kam, M.L.; Burggraaf, J.; Cohen, A.F.; Cazzola, M.; et al. Efficacy and safety of RPL554, a dual PDE3 and PDE4 inhibitor, in healthy volunteers and in patients with asthma or chronic obstructive pulmonary disease: Findings from four clinical trials. Lancet Respir. Med. 2013, 1, 714–727. [Google Scholar] [CrossRef]
  282. Luo, J.; Yang, L.; Yang, J.; Yang, D.; Liu, B.C.; Liu, D.; Liang, B.M.; Liu, C.T. Efficacy and safety of phosphodiesterase 4 inhibitors in patients with asthma: A systematic review and meta-analysis. Respirology 2018, 23, 467–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Damera, G.; Jiang, M.; Zhao, H.; Fogle, H.W.; Jester, W.F.; Freire, J.; Panettieri, R.A., Jr. Aclidinium bromide abrogates allergen-induced hyperresponsiveness and reduces eosinophilia in murine model of airway inflammation. Eur. J. Pharmacol. 2010, 649, 349–353. [Google Scholar] [CrossRef] [PubMed]
  284. Ohta, S.; Oda, N.; Yokoe, T.; Tanaka, A.; Yamamoto, Y.; Watanabe, Y.; Minoguchi, K.; Ohnishi, T.; Hirose, T.; Nagase, H.; et al. Effect of tiotropium bromide on airway inflammation and remodelling in a mouse model of asthma. Clin. Exp. Allergy 2010, 40, 1266–1275. [Google Scholar] [CrossRef] [PubMed]
  285. Toumpanakis, D.; Loverdos, K.; Tzouda, V.; Vassilakopoulou, V.; Litsiou, E.; Magkou, C.; Karavana, V.; Pieper, M.; Vassilakopoulos, T. Tiotropium bromide exerts anti-inflammatory effects during resistive breathing, an experimental model of severe airway obstruction. Int. J. Chronic Obstr. Pulm. Dis. 2017, 12, 2207–2220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Anzalone, G.; Gagliardo, R.; Bucchieri, F.; Albano, G.D.; Siena, L.; Montalbano, A.M.; Bonanno, A.; Riccobono, L.; Pieper, M.P.; Gjomarkaj, M.; et al. IL-17A induces chromatin remodeling promoting IL-8 release in bronchial epithelial cells: Effect of Tiotropium. Life Sci. 2016, 152, 107–116. [Google Scholar] [CrossRef]
  287. Suzaki, I.; Asano, K.; Shikama, Y.; Hamasaki, T.; Kanei, A.; Suzaki, H. Suppression of IL-8 production from airway cells by tiotropium bromide in vitro. Int. J. Chronic Obstr. Pulm. Dis. 2011, 6, 439–448. [Google Scholar] [CrossRef] [Green Version]
  288. Peters, S.P.; Kunselman, S.J.; Icitovic, N.; Moore, W.C.; Pascual, R.; Ameredes, B.T.; Boushey, H.A.; Calhoun, W.J.; Castro, M.; Cherniack, R.M.; et al. Tiotropium bromide step-up therapy for adults with uncontrolled asthma. N. Engl. J. Med. 2010, 363, 1715–1726. [Google Scholar] [CrossRef] [Green Version]
  289. Kerstjens, H.A.; Engel, M.; Dahl, R.; Paggiaro, P.; Beck, E.; Vandewalker, M.; Sigmund, R.; Seibold, W.; Moroni-Zentgraf, P.; Bateman, E.D. Tiotropium in asthma poorly controlled with standard combination therapy. N. Engl. J. Med. 2012, 367, 1198–1207. [Google Scholar] [CrossRef] [Green Version]
  290. Iwamoto, H.; Yokoyama, A.; Shiota, N.; Shoda, H.; Haruta, Y.; Hattori, N.; Kohno, N. Tiotropium bromide is effective for severe asthma with noneosinophilic phenotype. Eur. Respir. J. 2008, 31, 1379–1380. [Google Scholar] [CrossRef]
  291. Kerstjens, H.A.; Moroni-Zentgraf, P.; Tashkin, D.P.; Dahl, R.; Paggiaro, P.; Vandewalker, M.; Schmidt, H.; Engel, M.; Bateman, E.D. Tiotropium improves lung function, exacerbation rate, and asthma control, independent of baseline characteristics including age, degree of airway obstruction, and allergic status. Respir. Med. 2016, 117, 198–206. [Google Scholar] [CrossRef] [Green Version]
  292. Casale, T.B.; Bateman, E.D.; Vandewalker, M.; Virchow, J.C.; Schmidt, H.; Engel, M.; Moroni-Zentgraf, P.; Kerstjens, H.A.M. Tiotropium Respimat Add-on Is Efficacious in Symptomatic Asthma, Independent of T2 Phenotype. J. Allergy Clin. Immunol. Pract. 2018, 6, 923–935.e9. [Google Scholar] [CrossRef] [PubMed]
  293. Lazarus, S.C.; Krishnan, J.A.; King, T.S.; Lang, J.E.; Blake, K.V.; Covar, R.; Lugogo, N.; Wenzel, S.; Chinchilli, V.M.; Mauger, D.T.; et al. Mometasone or Tiotropium in Mild Asthma with a Low Sputum Eosinophil Level. N. Engl. J. Med. 2019, 380, 2009–2019. [Google Scholar] [CrossRef] [PubMed]
  294. O’Byrne, P.M.; Metev, H.; Puu, M.; Richter, K.; Keen, C.; Uddin, M.; Larsson, B.; Cullberg, M.; Nair, P. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: A randomised, double-blind, placebo-controlled trial. Lancet Respir. Med. 2016, 4, 797–806. [Google Scholar] [CrossRef]
  295. Watz, H.; Uddin, M.; Pedersen, F.; Kirsten, A.; Goldmann, T.; Stellmacher, F.; Groth, E.; Larsson, B.; Bottcher, G.; Malmgren, A.; et al. Effects of the CXCR2 antagonist AZD5069 on lung neutrophil recruitment in asthma. Pulm. Pharmacol. Ther. 2017, 45, 121–123. [Google Scholar] [CrossRef] [PubMed]
  296. Chaudhuri, R.; Norris, V.; Kelly, K.; Zhu, C.Q.; Ambery, C.; Lafferty, J.; Cameron, E.; Thomson, N.C. Effects of a FLAP inhibitor, GSK2190915, in asthmatics with high sputum neutrophils. Pulm. Pharmacol. Ther. 2014, 27, 62–69. [Google Scholar] [CrossRef]
  297. Snowise, N.G.; Clements, D.; Ho, S.Y.; Follows, R.M. Addition of a 5-lipoxygenase-activating protein inhibitor to an inhaled corticosteroid (ICS) or an ICS/long-acting beta-2-agonist combination in subjects with asthma. Curr. Med. Res. Opin. 2013, 29, 1663–1674. [Google Scholar] [CrossRef]
  298. Thalanayar Muthukrishnan, P.; Nouraie, M.; Parikh, A.; Holguin, F. Zileuton use and phenotypic features in asthma. Pulm. Pharmacol. Ther. 2020, 60, 101872. [Google Scholar] [CrossRef]
  299. Chiu, C.J.; Huang, M.T. Asthma in the Precision Medicine Era: Biologics and Probiotics. Int. J. Mol. Sci. 2021, 22, 4528. [Google Scholar] [CrossRef]
  300. Corren, J.; Parnes, J.R.; Wang, L.; Mo, M.; Roseti, S.L.; Griffiths, J.M.; van der Merwe, R. Tezepelumab in Adults with Uncontrolled Asthma. N. Engl. J. Med. 2017, 377, 936–946. [Google Scholar] [CrossRef]
  301. Venkataramani, S.; Low, S.; Weigle, B.; Dutcher, D.; Jerath, K.; Menzenski, M.; Frego, L.; Truncali, K.; Gupta, P.; Kroe-Barrett, R.; et al. Design and characterization of Zweimab and Doppelmab, high affinity dual antagonistic anti-TSLP/IL13 bispecific antibodies. Biochem. Biophys. Res. Commun. 2018, 504, 19–24. [Google Scholar] [CrossRef]
  302. Howarth, P.H.; Babu, K.S.; Arshad, H.S.; Lau, L.; Buckley, M.; McConnell, W.; Beckett, P.; Al Ali, M.; Chauhan, A.; Wilson, S.J.; et al. Tumour necrosis factor (TNFalpha) as a novel therapeutic target in symptomatic corticosteroid dependent asthma. Thorax 2005, 60, 1012–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Berry, M.A.; Hargadon, B.; Shelley, M.; Parker, D.; Shaw, D.E.; Green, R.H.; Bradding, P.; Brightling, C.E.; Wardlaw, A.J.; Pavord, I.D. Evidence of a role of tumor necrosis factor alpha in refractory asthma. N. Engl. J. Med. 2006, 354, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Morjaria, J.B.; Chauhan, A.J.; Babu, K.S.; Polosa, R.; Davies, D.E.; Holgate, S.T. The role of a soluble TNFalpha receptor fusion protein (etanercept) in corticosteroid refractory asthma: A double blind, randomised, placebo controlled trial. Thorax 2008, 63, 584–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Liang, L.; Hur, J.; Kang, J.Y.; Rhee, C.K.; Kim, Y.K.; Lee, S.Y. Effect of the anti-IL-17 antibody on allergic inflammation in an obesity-related asthma model. Korean J. Intern. Med. 2018, 33, 1210–1223. [Google Scholar] [CrossRef] [Green Version]
  306. Dos Santos, T.M.; Righetti, R.F.; Camargo, L.D.N.; Saraiva-Romanholo, B.M.; Aristoteles, L.; de Souza, F.C.R.; Fukuzaki, S.; Alonso-Vale, M.I.C.; Cruz, M.M.; Prado, C.M.; et al. Effect of Anti-IL17 Antibody Treatment Alone and in Combination With Rho-Kinase Inhibitor in a Murine Model of Asthma. Front. Physiol. 2018, 9, 1183. [Google Scholar] [CrossRef]
  307. Khokhlovich, E.; Grant, S.; Kazani, S.; Strieter, R.; Thornton-Wells, T.; Laramie, J.; Morgan, T.; Kennedy, S. The biological pathways underlying response to anti-IL-17A (AIN457; secukinumab) therapy differ across severe asthmatic patients. Eur. Respir. J. 2017, 50, OA2897. [Google Scholar]
  308. Staton, T.L.; Peng, K.; Owen, R.; Choy, D.F.; Cabanski, C.R.; Fong, A.; Brunstein, F.; Alatsis, K.R.; Chen, H. A phase I, randomized, observer-blinded, single and multiple ascending-dose study to investigate the safety, pharmacokinetics, and immunogenicity of BITS7201A, a bispecific antibody targeting IL-13 and IL-17, in healthy volunteers. BMC Pulm. Med. 2019, 19, 5. [Google Scholar] [CrossRef]
  309. Langrish, C.L.; Chen, Y.; Blumenschein, W.M.; Mattson, J.; Basham, B.; Sedgwick, J.D.; McClanahan, T.; Kastelein, R.A.; Cua, D.J. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med. 2005, 201, 233–240. [Google Scholar] [CrossRef] [Green Version]
  310. Zhao, Y.; Huang, Y.; He, J.; Li, C.; Deng, W.; Ran, X.; Wang, D. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, attenuates airway inflammation by inhibiting the proliferation of effector T cells in a murine model of neutrophilic asthma. Immunol. Lett. 2014, 157, 9–15. [Google Scholar] [CrossRef]
  311. Han, W.; Li, J.; Tang, H.; Sun, L. Treatment of obese asthma in a mouse model by simvastatin is associated with improving dyslipidemia and decreasing leptin level. Biochem. Biophys. Res. Commun. 2017, 484, 396–402. [Google Scholar] [CrossRef]
  312. Lee, H.Y.; Lee, E.G.; Hur, J.; Rhee, C.K.; Kim, Y.K.; Lee, S.Y.; Kang, J.Y. Pravastatin alleviates allergic airway inflammation in obesity-related asthma mouse model. Exp. Lung Res. 2019, 45, 275–287. [Google Scholar] [CrossRef] [PubMed]
  313. Norman, P. Evaluation of WO2013136076: Two crystalline forms of the phosphatidylinositol 3-kinase-delta inhibitor RV-1729. Expert Opin. Ther. Pat. 2014, 24, 471–475. [Google Scholar] [CrossRef] [PubMed]
  314. Leaker, B.R.; Barnes, P.J.; O’Connor, B.J.; Ali, F.Y.; Tam, P.; Neville, J.; Mackenzie, L.F.; MacRury, T. The effects of the novel SHIP1 activator AQX-1125 on allergen-induced responses in mild-to-moderate asthma. Clin. Exp. Allergy 2014, 44, 1146–1153. [Google Scholar] [CrossRef] [PubMed]
  315. Cahn, A.; Hamblin, J.N.; Begg, M.; Wilson, R.; Dunsire, L.; Sriskantharajah, S.; Montembault, M.; Leemereise, C.N.; Galinanes-Garcia, L.; Watz, H.; et al. Safety, pharmacokinetics and dose-response characteristics of GSK2269557, an inhaled PI3Kdelta inhibitor under development for the treatment of COPD. Pulm. Pharmacol. Ther. 2017, 46, 69–77. [Google Scholar] [CrossRef]
  316. Winkler, D.G.; Faia, K.L.; DiNitto, J.P.; Ali, J.A.; White, K.F.; Brophy, E.E.; Pink, M.M.; Proctor, J.L.; Lussier, J.; Martin, C.M.; et al. PI3K-delta and PI3K-gamma inhibition by IPI-145 abrogates immune responses and suppresses activity in autoimmune and inflammatory disease models. Chem. Biol. 2013, 20, 1364–1374. [Google Scholar] [CrossRef] [Green Version]
  317. Kampe, M.; Lampinen, M.; Stolt, I.; Janson, C.; Stalenheim, G.; Carlson, M. PI3-kinase regulates eosinophil and neutrophil degranulation in patients with allergic rhinitis and allergic asthma irrespective of allergen challenge model. Inflammation 2012, 35, 230–239. [Google Scholar] [CrossRef]
  318. Toki, S.; Newcomb, D.C.; Printz, R.L.; Cahill, K.N.; Boyd, K.L.; Niswender, K.D.; Peebles, R.S., Jr. Glucagon-like peptide-1 receptor agonist inhibits aeroallergen-induced activation of ILC2 and neutrophilic airway inflammation in obese mice. Allergy 2021, 76, 3433–3445. [Google Scholar] [CrossRef]
  319. Foer, D.; Beeler, P.E.; Cui, J.; Karlson, E.W.; Bates, D.W.; Cahill, K.N. Asthma Exacerbations in Patients with Type 2 Diabetes and Asthma on Glucagon-like Peptide-1 Receptor Agonists. Am. J. Respir. Crit. Care Med. 2021, 203, 831–840. [Google Scholar] [CrossRef]
Figure 1. Pathogenesis of neutrophilic asthma. Several cells, including airway epithelial cells, macrophages, T helper (Th) cells, innate helper 3 cells (ILC3), airway smooth muscle cells (ASMCs), and neutrophils play important roles in the pathogenesis of neutrophilic asthma. Airway epithelial cells, stimulated by air pollution, cigarette smoke, bacterial colonization, virus, and allergens, secrete TSLP, IL-33, and IL-25. TSLP secreted from epithelial cells and inflammatory cells converts Th0 to Th17 cells and subsequently induced neutrophil recruitment via IL-8 and GM-CSF, induced by IL-17 from airway epithelial cells. The IL-17/Th17 axis is involved in bacterial colonization and microbiome associated neutrophilic inflammation in asthma. Obesity and GERD are related to severe, neutrophilic asthma and the IL-17/Th17 axis is involved in these conditions. Neutrophil extracellular trap (NETs) formation, damage-associated molecular patterns (DAMPs), and NLPR3 inflammasome are also involved in the pathogenesis of neutrophil asthma.
Figure 1. Pathogenesis of neutrophilic asthma. Several cells, including airway epithelial cells, macrophages, T helper (Th) cells, innate helper 3 cells (ILC3), airway smooth muscle cells (ASMCs), and neutrophils play important roles in the pathogenesis of neutrophilic asthma. Airway epithelial cells, stimulated by air pollution, cigarette smoke, bacterial colonization, virus, and allergens, secrete TSLP, IL-33, and IL-25. TSLP secreted from epithelial cells and inflammatory cells converts Th0 to Th17 cells and subsequently induced neutrophil recruitment via IL-8 and GM-CSF, induced by IL-17 from airway epithelial cells. The IL-17/Th17 axis is involved in bacterial colonization and microbiome associated neutrophilic inflammation in asthma. Obesity and GERD are related to severe, neutrophilic asthma and the IL-17/Th17 axis is involved in these conditions. Neutrophil extracellular trap (NETs) formation, damage-associated molecular patterns (DAMPs), and NLPR3 inflammasome are also involved in the pathogenesis of neutrophil asthma.
Diagnostics 12 01175 g001
Figure 2. Airway remodeling in asthma related to neutrophilic inflammation. Airway remodeling in asthma is a characteristic feature of chronic asthma. LTB4, IL-8, LTB4, and TNF-α are elevated in an asthmatic airway and are related to airway remodeling. LTB4, IL-8, and TNF-α induce airway smooth muscle cell proliferation and migration. IL-8 and IL-17 upregulate MUC5A and MUC5B expression in epithelial cells. Abbreviations: IL, interleukin; LTB4, leukotriene B4, TNF-α; Tumor necrosis factor α, BLT1/2: leukotriene B4 receptor 1/2, IL-17R: IL-17 receptor, TNFR: TNF receptor, ASMCs: airway smooth muscle cells.
Figure 2. Airway remodeling in asthma related to neutrophilic inflammation. Airway remodeling in asthma is a characteristic feature of chronic asthma. LTB4, IL-8, LTB4, and TNF-α are elevated in an asthmatic airway and are related to airway remodeling. LTB4, IL-8, and TNF-α induce airway smooth muscle cell proliferation and migration. IL-8 and IL-17 upregulate MUC5A and MUC5B expression in epithelial cells. Abbreviations: IL, interleukin; LTB4, leukotriene B4, TNF-α; Tumor necrosis factor α, BLT1/2: leukotriene B4 receptor 1/2, IL-17R: IL-17 receptor, TNFR: TNF receptor, ASMCs: airway smooth muscle cells.
Diagnostics 12 01175 g002
Table 1. Possible biomarkers for neutrophilic asthma.
Table 1. Possible biomarkers for neutrophilic asthma.
BiomarkerSampleDefinitionSignificanceRefs.
YKL-40Serum, sputumNot established, but serum YKL-40 > 60.94 ng/mL showed impaired lung function and require corticosteroidYKL-40 is released from neutrophil and epithelial cells, YKL-40 is released from neutrophils and epithelial cells
Serum YKL-40 correlates with sputum neutrophil counts
[181,182,183]
Hydrogen sulfide (H2S)Serum, exhaled breath, sputumNot establishedSputum H2S correlates with the degree of airflow limitation
Serum/sputum H2S predicts asthma exacerbation
[184,185,186]
MPOSputumNot establishedSputum MPO correlates with sputum YKL-40 and neutrophils[23,187]
NeutrophilSerum, sputumSputum > 60% or 76%Associated with chronic airway obstruction, annual decline of FEV1[188,189]
MicroRNASputum, serum, and plasmaNot establishedmiR-199a-5p, miR142-3p, miR233-3p, and miR629-3p are increased in neutrophilic asthmamiR299a -5p is negatively correlated with FEV1[190,191]
Table 2. Summary of treatment for asthma related to neutrophilic inflammation.
Table 2. Summary of treatment for asthma related to neutrophilic inflammation.
Non-Pharmacological Approach
ApproachPatient PopulationOutcomesRef.
Smoking cessationYoung patients with asthma (19–40 years old), steroid-free, 17% neutrophilic asthmaImproved asthma control and flung function[248]
Weight loss18–75-year-old, obese patients with asthma (BMI > 35 kg/m2)Improved asthma control, QOL, lung function, and AHR[249]
Nonspecific treatment for neutrophilic asthma
TherapyPatient populationOutcomesRef.
Macrolide (azithromycin, clarithromycin)Non-eosinophilic or neutrophilic severe asthma (18–75-year-old patients)Reduced asthma exacerbation, QOL, and lung function[250]
PDE inhibitorPatients 18–70 years of age, moderate-to-severe asthmaImproved lung function and asthma control[251]
TiotropiumAdult symptomatic patients with asthma despite treatment with medium-dose ICSImproved lung function and asthma control, reduced risk of severe exacerbation, independent of type 2 inflammation[252]
Tiotropium6–17-year-old patients, symptomatic severe asthmaImproved lung function and ACQ, reduced risk of exacerbation, independent of type 2 inflammation[253]
Specific treatment for neutrophil and mediators
SCH527123/CXCR2Severe asthma and sputum neutrophil >40%Fewer mild exacerbations and a trend towards improvement in the ACQ, but not statistically significant[254]
GSK2090915/FLAPPersistent asthma treated with SABA onlyImproved symptom score and reduced SABA use[255]
Zileuton/5-LOModerate-to-severe asthma treated with low dose ICSImproved PEF and symptoms[256]
Biologics
Tezepelumab/TSLPModerate-to-severe asthmaReduced rate of exacerbation, improved lung function, ACQ, and AQLQ, regardless of type 2 inflammation[257]
Golimumab/TNF-αUncontrolled asthma with high-dose ICS/LABANo improvement in FEV1 and exacerbation[258]
Etanercept/TNF-αModerate-to-severe persistent asthmaNo improvement in FEV1 and ACQ, exacerbation, AHR, AQLQ[259]
Brodalumab/IL-17 receptorInadequately controlled moderate-to-severe asthma treated with high-dose ICS ± LABANo treatment differences were observed[260]
Risankinumab/IL-23Adult patients with severe asthmaNo improvement in asthma exacerbation[261]
Tocilizumab/IL-6Mild asthmaNo improvement in allergen-induced bronchoconstriction[262]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yamasaki, A.; Okazaki, R.; Harada, T. Neutrophils and Asthma. Diagnostics 2022, 12, 1175. https://doi.org/10.3390/diagnostics12051175

AMA Style

Yamasaki A, Okazaki R, Harada T. Neutrophils and Asthma. Diagnostics. 2022; 12(5):1175. https://doi.org/10.3390/diagnostics12051175

Chicago/Turabian Style

Yamasaki, Akira, Ryota Okazaki, and Tomoya Harada. 2022. "Neutrophils and Asthma" Diagnostics 12, no. 5: 1175. https://doi.org/10.3390/diagnostics12051175

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop