Skip to main content
F1000Research logoLink to F1000Research
. 2018 May 30;7:F1000 Faculty Rev-680. [Version 1] doi: 10.12688/f1000research.14581.1

New targets for resolution of airway remodeling in obstructive lung diseases

Ajay P Nayak 1, Deepak A Deshpande 1, Raymond B Penn 1,a
PMCID: PMC5981194  PMID: 29904584

Abstract

Airway remodeling (AR) is a progressive pathological feature of the obstructive lung diseases, including asthma and chronic obstructive pulmonary disease (COPD). The pathology manifests itself in the form of significant, progressive, and (to date) seemingly irreversible changes to distinct respiratory structural compartments. Consequently, AR correlates with disease severity and the gradual decline in pulmonary function associated with asthma and COPD. Although current asthma/COPD drugs manage airway contraction and inflammation, none of these effectively prevent or reverse features of AR. In this review, we provide a brief overview of the features and putative mechanisms affecting AR. We further discuss recently proposed strategies with promise for deterring or treating AR.

Keywords: airway remodeling, asthma, GPCR, smooth muscle

Airway remodeling in obstructive lung diseases

Airway remodeling (AR) can be defined as a progressive pathological reorganization of the cellular and molecular constitution of the airway wall. While the onset and rate of progression of structural changes in the airways have been subjects of immense debate, AR has been associated with each of the asthmatic phenotypes 1. Furthermore, the gradual deleterious transformation in lungs can affect airways of all sizes along the bronchial tree. Although the strategies for reversing airway contraction and mitigating airway inflammation have been mainstays of asthma therapy, AR has been clinically intractable. Consequently, a pressing need exists for defining the fundamental pathways contributing to AR pathology and for empowering both basic and clinical research to address this problem. For a comprehensive understanding of the conceptual and practical challenges in AR research, readers are encouraged to review the official research statement of the American Thoracic Society 2. In this current report, we provide an overview of the limitations of currently approved anti-asthma/chronic obstructive pulmonary disease (COPD) drugs in addressing AR and further describe the therapeutic potential of recently proposed approaches for targeting AR.

AR was first described in 1922 in patients whose death was attributed to asthma. Necropsy specimens from these patients revealed extensive bronchial mucus plugs and thickening of the airway wall 3. Numerous subsequent clinical investigations have revealed that AR encompasses broad structural changes in the airway that includes thickening of the airway wall, airway smooth muscle (ASM) hyperplasia and hypertrophy, edema, subepithelial fibrosis, increased extracellular matrix (ECM) deposition, immune cell and fibroblast accumulation, angiogenesis, altered matrix composition, goblet cell metaplasia, and mucus hypersecretion 2. A consensus has emerged that multiple cell types (including epithelium, ASM, fibroblasts, and immune cells) contribute to the development of AR in asthma and COPD 4, 5 ( Table 1).

Table 1. Contribution by distinct cell types to the overall pathology of airway remodeling in obstructive lung diseases.

Lung cell type Contribution to the pathophysiology of airway remodeling
Epithelial cells Epithelial shedding
Mucus secretion
Subepithelial fibrosis
Goblet cell hyperplasia
Stimulating airway smooth muscle (ASM) proliferation through release
of growth factors
Recruitment of pro-inflammatory cells
Promoting extracellular matrix (ECM) deposition
Promoting angiogenesis
ASM cells Increased ASM mass
ASM migration and invasion of the epithelium
Adoption of synthetic phenotype (for example, secretion of transforming
growth factor-beta, chemokines, and ECM components)
Interaction with immune cells through cell adhesion molecules
Fibroblasts Differentiation into myofibroblasts and secretion of ECM components
Accumulation in subepithelial regions

The role of the airway epithelium in triggering initial responses and sustaining architectural changes in asthmatic lungs is evident 6, 7. In asthma, repetitive damage to the epithelium from exposure to noxious environmental agents and immune modulators promotes shedding of the epithelium. Consequently, the underlying epithelial-mesenchymal trophic unit may be persistently active and in a reparative state, thus promoting chronic and progressive remodeling of the airway 8. Remodeling manifests in the form of thickening of the epithelial layer, loss of cilia, compromised barrier function, mucus hypersecretion, and ECM remodeling of the subepithelial space 6, 7, 917. Moreover, the number of mucus-secreting goblet cells also increases in asthmatics 18, 19. These features collectively contribute to anatomical changes that cause airway narrowing, increased fixed resistance, and mucus plugging of the bronchial lumen.

Physiological ASM function is crucial for maintaining adequate airflow. Changes in both ASM responsiveness and morphology occur with asthma, which affects airway resistance and airflow. A critical feature of AR is an increase in ASM mass that contributes significantly to asthma pathology 20, 21. Furthermore, the increased ASM mass and increased airway wall thickness reduce airway lumen area, resulting in increased dynamic and fixed resistance 2126. Asthmatic ASM can also acquire a synthetic phenotype, which is characterized by increased secretion of ECM, cytokines, and growth factors. Clinical outcomes associated with bronchial thermoplasty intervention (application of controlled radiofrequency energy to the airway wall) suggest that reducing ASM area is sufficient to improve outcomes in asthmatics 27.

Fibroblasts can contribute to AR through increased secretion of ECM 28, 29. Beyond contributing to increased airway wall thickness, ECM components can modulate cellular proliferation and migration. However, the role of fibroblast and ECM components in AR in the context of obstructive lung diseases is not fully understood.

The structural changes may contribute toward a gradual decline in lung function and potentially in loss of pulmonary elasticity, leading to hyperinflation and air trapping in lungs. Moreover, remodeling reduces effectiveness of bronchodilatory treatments 5, 3032. A correlation between AR and disease severity has been established, but the clinical consequences of AR are yet to be fully understood 3338. This lack of knowledge also impacts drug discovery efforts. In the subsequent sections, we review the efficacy of current therapeutics in blunting or reversing AR and discuss novel therapeutic approaches to regulate progression of AR.

Overview of current therapeutics and their limitations

Current management of asthma focuses on reversing ASM contraction and mitigating airway inflammation. None of these approaches directly addresses the progressive pathology that causes remodeling in the lung ( Table 2).

Table 2. Current therapeutic targets for asthma management and their effect on airway remodeling.

Class of
therapeutic
drugs
Target Effect on airway remodeling
β-agonists β 2-AR (beta 2
adrenergic receptor)
Limited effect on airway remodeling 5. Combination therapy with inhaled
corticosteroid limits angiogenesis and fibroblast proliferation 55, 56, 71.
Inhaled
corticosteroids
Glucocorticoid
receptor
Combination therapy with β-agonists limits angiogenesis 71. Mixed anti-
proliferative actions on airway smooth muscle cells and human fibroblasts 55, 56.
Reduced mucin secretion and limited extracellular matrix deposition 6164.
Anti-leukotrienes CysLTR (cysteinyl
leukotriene receptor)
Moderate effect on airway smooth muscle mass, goblet cell metaplasia, and
subepithelial collagen deposition 45, 46, 62. Decreased accumulation of fibroblasts
in lungs 47.

As noted earlier, bronchial thermoplasty has been shown to reduce ASM mass in conducting airways of some, but not all, severe asthmatics undergoing the procedure 27, 39, 40. This procedure has been shown to significantly reduce collagen deposition in the basement membrane. Although bronchial thermoplasty has been shown to improve quality of life for severe asthmatics in the short term, the cost of the procedure, post-procedure exacerbations, and questions regarding long-term efficacy have limited its application 41.

Among pharmacological options, β-agonists are the drug of choice for evoking bronchorelaxation in attempting to reverse an acute asthma attack or for providing bronchoprotection when used in combination with an inhaled corticosteroid as a maintenance therapy. However, there is no compelling evidence that β-agonists deter or reverse AR 5. Signaling through cysteinyl leukotriene receptors (CysLTRs) and muscarinic acetylcholine receptors (mAChRs) has been established to promote outcomes that contribute to AR 4244. Antagonists of both receptors have shown some utility in preventing AR. Treatment with the CysLTR antagonist montelukast reversed ovalbumin-induced AR by decreasing goblet cell metaplasia, ASM mass, and subepithelial collagen deposition 45, 46. In a cohort of mild asthmatics, montelukast treatment showed reduced accumulation of myofibroblasts in the airway wall, suggesting some potential to mitigate AR 47. Similarly, the long-acting mAChR antagonist, tiotropium, has demonstrated a robust ability in preventing AR in rodent (guinea pig and mouse) models of ovalbumin-induced asthma and lipopolysaccharide-induced COPD 4852. Overall, although some evidence suggests that mAChR and CysLTR antagonists may have utility in deterring AR, additional studies in humans are necessary to establish the true effectiveness of these drugs in preventing or reversing AR 5.

Persistent asthma is commonly treated with inhaled corticosteroids either as a monotherapy or in combination with a β-agonist or mAChR antagonist. In epithelial cells, corticosteroids limit the inflammatory response and induce apoptosis 53, 54. In vitro, multiple corticosteroids have been shown to significantly inhibit fibroblast proliferation either alone or in combination with β-agonists 55, 56. Similar anti-proliferative effects have also been reported with corticosteroids in ASM cells stimulated with distinct mitogenic agents 57, 58. However, others have shown that corticosteroids have no effect on ASM proliferation 59, 60. While corticosteroids inhibit growth factor–stimulated proliferation of ASM cells sourced from healthy controls, this effect was lacking on ASM cells from asthmatics 59. In animal models, dexamethasone has been shown to reduce goblet cell metaplasia; however, this treatment showed no effect on ASM mass and subepithelial fibrosis 45. In humans, in conjunction with limiting inflammation and airway hyperresponsiveness, corticosteroid treatment can also reduce mucin secretion and limit ECM deposition and AR 6164. However, others have shown that corticosteroids have a mixed effect on the resolution of subepithelial fibrosis 6570. Collectively, studies to date indicate a need for developing better therapeutic drugs for targeting AR pathology in obstructive lung diseases.

New targets and approaches for airway remodeling

In recent years, basic science research has begun to provide insight into the mechanisms, mediated by multiple cell types, that promote AR and these studies help to inform potential strategies for managing AR. Certain approaches that show promise in mitigating features of AR have recently been proposed ( Table 3).

Table 3. Anti-remodeling effects of novel therapeutic approaches ( in vitro, animal, and human studies).

Class of therapeutic drugs Target Potential effect on airway remodeling (AR)
G protein–coupled receptor
modulators
E-prostanoid receptors Suppression of airway smooth muscle (ASM) proliferation 7274.
Bitter taste receptors (TAS2Rs) Regulation of ASM proliferation 81, 82. Reversal of allergen-
induced AR features, including ASM mass 83. Alteration of
mitochondrial function and induction of autophagy 84.
Biologics Interleukin-5 (IL-5) cytokine Reduced subepithelial fibrosis and extracellular matrix (ECM)
deposition 85, 86.
Immunoglobulin E Reduced thickening of reticular lamina 87.
Mitogen-activated protein
kinase (MAPK) inhibitors
MEK1 (MAPK kinase) Regulation of mucus secretion 88, 89.
p38 Reduced ASM mass and goblet cell metaplasia 90.
c-Jun N-terminal kinases (JNKs) Reduced mucus secretion and expansion of goblet cells 91, 92.
Reduced proliferation of ASM and epithelial cells 93.
Transforming growth factor-
beta-activated kinase 1 (TAK1)
Reduced synthesis of IL-8 in ASM cells and reduced
proliferation 94, 95.
Receptor tyrosine kinase
inhibitors
Epidermal growth factor
receptor
Reduced proliferation of ASM and epithelial cells 9698.
Regulation of mucus secretion 99102.
Reduced ASM thickening and goblet cell metaplasia 103.
Platelet-derived growth factor
receptor
Reduced ASM proliferation 104.
Stem cell growth factor receptor
(c-kit)
Attenuated collagen accumulation in lungs 105.
Non-receptor tyrosine kinase
inhibitors
Spleen tyrosine kinase (Syk) Reduced bronchial edema 106.
Janus kinase (JAK) Reduced expression of Gob-5 107.
Other kinase inhibitors TGF-β receptor type I (T-βRI)
kinase
Diminished collagen deposition and reduced proliferation of
ASM and epithelial cells in lungs 108.
Rho-associated protein kinase
(ROCK)
Curtailed ECM remodeling process 109.
Phosphodiesterase (PDE)
inhibitors
PDEs Marked reduction in subepithelial fibrosis and epithelial layer
thickening 110. Reduced proliferation of ASM 111.

(Other) G protein–coupled receptor ligands

G protein–coupled receptors (GPCRs) play a substantial role in numerous normal physiological functions. Unsurprisingly, they can contribute towards the pathophysiology of various diseases. As noted earlier, GPCR agonists (of the β2-adrenergic receptor) and antagonists (of the mAChRs and CysLTR) are principal drugs in the management of asthma and COPD. In this section, we provide a brief overview of novel targets, the drugs that modulate them, and the potential of such drugs to address AR pathology.

E-prostanoid receptor agonists. The role of prostaglandin E 2 (PGE 2) and E-prostanoid (EP) receptor subtypes in mitigating AR has been a subject of recent research. Early studies demonstrated that autocrine PGE 2, generated as a consequence of cytokine-induced cyclooxygenase-2 induction, significantly suppresses mitogen-induced ASM proliferation in vitro 72. Furthermore, studies of cultured human ASM in our laboratory have demonstrated that exogenous PGE 2 shows relatively superior anti-mitogenic activity in comparison to multiple β-agonists with anti-mitogenic effects corresponding to drug efficacy in activating the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) axis 73, 74. Application of PGE 2 in humans has been hindered by the ability of PGE 2 to signal through multiple receptor subtypes (EP1–4) 7579, which contributes to undesirable side effects. It is now known that the cough response is mediated by PGE 2 activation of the EP3 receptor on vagal sensory nerves 80. Additionally, studies in our lab have shown that EP3 receptor signaling has a pro-mitogenic role in ASM 74. To overcome the heterogeneity of PGE 2 signaling, the development of EP receptor subtype–specific modulators that specifically promote Gs-cAMP-PKA axis activity (via EP2 and EP4 subtypes) has been purposed 84, 112114. Currently, our lab is evaluating various strategies of targeting specific EP receptor subtypes in pre-clinical models of allergen-induced asthma 84.

Bitter taste receptors. Recently, our laboratory showed that bitter taste receptor (TAS2R) agonists can limit proliferation of ASM cells in vitro 81, 82. Mechanistically, TAS2R agonists restrict ASM proliferation by inhibiting (1) the growth factor–activated protein kinase B (Akt) phosphorylation; (2) transcription factors AP-1, STAT3, E2 factor, and NFAT; and (3) genes associated with cell cycle progression.

The anti-mitogenic effects of TAS2R agonists further translate to pre-clinical asthma models as well. In a chronic allergen (ovalbumin or house dust mite) challenge model, treatment with bitter taste compounds (chloroquine and quinine) significantly reversed remodeling features 83. Specifically, treatment with bitter compounds inhibited the expression of calponin, smooth muscle alpha-actin, and smooth muscle myosin heavy chain in lungs. Furthermore, levels of matrix metallopeptidase-8 (MMP-8) (neutrophil collagenase), pro-MMP-9 (gelatinase), and MMP-12 (macrophage metalloelastase) were significantly reduced in lungs following treatment with TAS2R agonists. Finally, allergen-induced expression of pro-fibrotic cytokine transforming growth factor-beta (TGF-β) as well as phospho-mothers against decapentaplegic homolog 2 (pSmad2) and fibronectin in the lung tissue was also curtailed by TAS2R agonists. Collectively, these studies indicate that TAS2R agonists, unlike current GPCR ligands used to treat asthma, address multiple features of asthma pathology, including AR. Advancements in the development of selective ligands for TAS2R subtypes will allow for a refined therapeutic approach in the near future.

Bitter tastants have also been shown to modulate function of ciliated epithelial cells 115. Specifically, the motile cilia on human airway epithelia express TAS2Rs (T2R4, T2R43, T2R38, and T2R46). The organization of TAS2Rs on cilia with the distribution of the signaling machinery along the ciliary shaft and within the attached epithelial cell presents an interesting mechanical apparatus for signal transduction. Stimulation of TAS2Rs with bitter tastants induces transient Ca 2+ flux within the epithelial cells and increases ciliary beat frequency. Functionally, promoting increased ciliary movement could be beneficial in the removal of excess mucus from the airways.

In recent years, the role of mitochondrial dysfunction in disease states, including obstructive lung diseases, has become increasingly clear 116, 117. Specifically, a role for autophagy/mitophagy in regulating mitochondrial function in pathophysiology of obstructive lung diseases is emerging. As noted earlier, our studies show that activation of TAS2Rs can promote anti-mitogenic activities. Further explorations into the mechanisms that underlie the anti-mitogenic effects of TAS2R agonists have uncovered an interesting role for bitter tastants in altering mitochondrial function and inducing autophagy 82. TAS2R agonists can induce changes in mitochondrial membrane potential, increase reactive oxygen species generation, and promote mitochondrial fragmentation. These observations provide insight into the broader therapeutic potential of targeting mitochondrial function and promoting autophagy to restrict cellular proliferation.

Biologics

Asthma pathology is orchestrated by multiple immunologic mediators (cellular and secreted) 118. Consequently, in recent years, therapies that target specific cytokines or immune cells to disrupt immune networks responsible for asthma pathology have gained significant interest. Targeting key cytokines with specific antibodies—biologics, including antibodies targeting interleukin-4 (IL-4), IL-5, and IL-13—can significantly limit recruitment of inflammatory cells to the lungs or blunt their pleiotropic effects. For instance, anti–IL-5 treatment can significantly reduce the number of circulating eosinophils in asthmatics and improve lung function 119121. Anti–IL-5 treatment has been shown to prevent the development of subepithelial fibrosis in a murine model of asthma and reduce incorporation of proteoglycans in the human airway wall 85, 86. However, antibodies targeting other cytokines or their receptors have not been studied in the context of AR 122. Collectively, the data on the effects of biologics on AR are lacking and this is possibly due to the relatively recent development of these drugs. Future longitudinal studies that evaluate biologics in the context of remediation of AR features are needed to address the utility of these drugs as anti-AR agents.

Allergen-specific immunoglobulin E (IgE) isotype antibodies can cross-link on mast cells and basophils, causing degranulation and release of histamine, cytokines, and growth factors 123. Biologics that block the interactions of IgE antibodies to the high-affinity FcεRI receptors on mast cells and antigen presenting cells can curtail the sensitization profile in asthmatics and reduce exacerbations. In severe asthmatics evaluated for 36 months, blocking IgE activity was sufficient to reduce the thickening of the reticular lamina, thereby having an impact on AR 87.

Although biologics have become an increasingly important tool in the management of severe asthma, their application in the clinic has some drawbacks 124126. Application of cytokine-specific antibody therapy is limited by the heterogeneity of asthma phenotypes 127. Non-atopic asthmatics are also not suitable for certain therapy (for example, anti-IgE). Biologics can also cause side effects such as hypersensitivity reactions, although the underlying mechanisms are unclear. Finally, there is also significant cost associated with the use of biologics.

Kinase inhibitors

Kinase enzymes modulate multiple cellular functions by regulating various signaling networks, including those regulating cellular proliferation and growth. Consequently, inhibitors that target various kinases have received increasing attention. Modulators of kinase functions account for one third of drugs in the development pipeline, and the majority of these represent cancer therapeutics 128. In this section, we provide a brief overview of drugs that target distinct kinases. For a more comprehensive discussion of targeting kinases in the context of obstructive lung diseases, the reader is referred to 129.

Mitogen-activated protein kinase inhibitors. Mitogen-activated protein kinases (MAPKs) have been studied extensively for their contribution to inflammatory gene expression and activation of multiple networks that contribute to the pathophysiology of obstructive lung diseases 130. Extracellular signal-regulated kinases (ERK1/2) are particularly interesting given that they are activated in multiple cell types that contribute to asthma and COPD pathology 88, 131, 132. Inhibition of ERK kinase (MAPK1, or MEK1) which is upstream of ERK1/2 can significantly reduce mucin 5AC, oligomeric mucus/gel-forming ( MUC5AC) expression in cultured human bronchial epithelial cells subjected to chronic mechanical stress at the air-liquid interface 88, 89. Other MAPKs, such as p38, c-Jun N-terminal kinase (JNK), and transforming growth factor beta-activated kinase 1 (TAK1), are activated in asthma and COPD 129, and inhibitors of these targets can mitigate various features of AR in both cell and animal asthma models 9095. However, to date, no studies in humans have evaluated these inhibitors. Another limitation of kinase inhibitors is that these compounds are predominantly inhibitors of ATP-competitive and catalytic sites and block all enzymatic activity, including MAPK functions important for normal physiological activity in cells. Given the ubiquitous functions of the MAPK signaling pathway, development of substrate-selective MAPK inhibitors with the goal of targeting specific kinase functions associated with disease, while preserving kinase functions in normal cells, appears necessary to overcome limitations of off-target effects.

Receptor tyrosine kinase inhibitors. Receptor tyrosine kinases (RTKs) occupy a central role in critical signaling networks that promote asthma pathology, including remodeling 133. With inflammation, distinct RTKs and their ligands (for example, epidermal growth factor) are upregulated in human asthmatic airways and show a strong correlation with disease severity 134143. RTKs can stimulate pathophysiological functions in ASM and epithelial cells. Thus, significant interest in advancing tyrosine kinase inhibitors for targeting RTKs has developed.

Activation of epidermal growth factor receptor (EGFR) is essential for mucus secretion and goblet cell metaplasia 144. It is also responsible for sustaining oxidative damage in the epithelial compartment through recruitment of neutrophils in a TGF-β–dependent manner 145148. EGFR inhibitors tyrphostin AG1478 and BIBX1522 have been evaluated in vitro and in animal models of lung inflammation 99101. Collectively, these studies report significant reductions in expression of mucus-associated MUC5AC gene and mucin secretion. More importantly, there is a concomitant reduction in collagen deposition and ASM proliferation 9698. Although these observations are encouraging, some inhibitors of EGFR have failed to produce similar outcomes in clinical studies 149. Activation of platelet-derived growth factor receptor (PDGFR) has been shown to stimulate ASM proliferation in vitro and in vivo 138, 150, 151. Multiple drugs targeting PDGFR can mitigate ASM proliferation in vitro, although animal and human studies that address AR are lacking 104. In severe corticosteroid-dependent asthmatics, treatment with the tyrosine kinase inhibitor mastinib has shown improved outcomes; however, some adverse effects, including skin rash and edema, have also been reported 152. During airway inflammation, multiple cell types (immune and resident) can be stimulated to secrete angiogenic factors, including vascular endothelial growth factor (VEGF) 153159. These angiogenic factors can further stimulate increase in formation of new blood vessels from endothelial cells in the subepithelial mucosa 160162. Although the contribution of neovascularization in AR is unclear, it has been suggested that newly formed vasculature is permeable, thus contributing to edema and limiting airflow 71, 161, 163, 164. An antagonist of VEGFR-1 and VEGFR-2 (SU5416) has been shown to limit inflammatory responses in animals 165, although its impact on AR is unknown and studies in humans are lacking.

Because multiple RTKs contribute to pathology of AR, a novel strategy that targets multiple RTKs has gained momentum. In a pre-clinical murine model of ovalbumin-induced asthma, treatment with nintedanib—a small-molecule inhibitor that targets multiple RTKs (VEGFR, fibroblast growth factor receptor, and PDGFR)—significantly improved indices of remodeling and airway inflammation 166. This approach could be useful in targeting multiple redundant RTK networks that contribute to AR. Finally, inhibitors of non-RTKs, such as stem cell growth factor receptor (c-kit), spleen tyrosine kinase (SYK), the proto-oncogene tyrosine-protein kinase Src, and Janus kinase (JAK), have also been investigated in rodent models of asthma but have not yet progressed to studies in humans 105, 106, 167171.

Other kinase inhibitors. Diverse stimuli (cytokines, viruses, growth factors, free radicals, and so on) can activate the transcription factor nuclear factor-kappa B (NF-κB) in multiple airway cell types. This transcription factor plays a key role in orchestrating immune responses and thus multiple intra- and inter-cell inflammatory signals 129. Although inhibitors that target activation of NF-κB have been shown to suppress certain synthetic functions of ASM 172 and modulate pro-inflammatory outcomes in epithelial cells 173, specific NF-κB inhibitors have not translated into clinical trials for asthma and this is due to their multiple side effects 129. Inhibitors of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI-3K) that regulate cellular lipids and coordinate inflammatory pathways have undergone extensive investigation in asthma and COPD 174, 175. However, data assessing AR indices are lacking. TGF-β plays an important role in cellular proliferation and differentiation and its expression increases in asthmatic airways, especially in the submucosal compartment 176178. TGF-β has also been implicated in AR and can promote proliferation in ASM 179, 180. TGF-β activates TGF-β receptor type I (T-βRI) kinase, which in turn activates Smad-dependent signaling that regulates expression of various genes. Small-molecule inhibitors of (T-βRI) kinase have yielded mixed results in studies assessing their effects on mechanisms mediating AR. T-βRI kinase inhibitors have been shown to diminish collagen deposition in lungs of rats challenged repeatedly with an allergen 108. In vitro, T-βRI inhibitors have demonstrated the ability to limit ASM proliferation, although their use in animal studies failed to inhibit TGF-β–induced increases in ASM mass 108, 181. Clinical application of these inhibitors has been limited due to adverse effects, including cardiotoxicity in clinical trials for cancer therapy 182. Protein kinase C (PKC) is another target of interest given its ability to promote contractile signaling in ASM following activation of Gq-coupled GPCRs. PKC is also relevant to AR because of its role in ASM proliferation 183 and mucus secretion in epithelium 148. Owing to severe toxicity, non-selective inhibitors of PKC have not progressed to clinical trials 129, 184. Selective inhibitors of PKC isoforms have been developed for clinical studies for treatment of cancer, metabolic diseases, and psychiatric disorders, although adverse effects have been problematic and no clinical trials have been conducted for treatment of obstructive lung diseases in humans 129, 184. Finally, Rho-associated protein kinase (ROCK) inhibitors have been shown to mitigate multiple features of asthma, including AR in guinea pig and murine models 109, 185. Although ROCK inhibitors have been approved for certain indications, clinical trials in asthma or COPD are lacking 129 as issues regarding selectivity and toxicity have limited progression of these inhibitors to clinical application in airway diseases 129.

In summary, in vitro studies of pharmacological inhibition of multiple kinases that contribute to dysfunction in ASM and epithelium have yielded promising results 181, 186. However, certain limitations have stalled progression of many drugs for clinical use. Specificity, efficacy, solubility issues, and poor pharmacokinetic profiles plague drug development 187. With chronic inhibitor treatment, compensatory signaling by other kinases may limit drug efficacy; this appears to be the case with p38 isoform inhibitors 129, 188. Inhibition of any widely expressed kinase runs the risk of adverse effects. For example, given that NF-κB is crucial for mounting an immune response to microbial pathogens, blocking its activation could render patients susceptible to life-threatening infections 129. Current challenges in developing effective and safe kinase inhibitors hinge on improving the poor solubility, selectivity, and targeting of the current versions of these drugs 129.

Other small-molecule inhibitors

Phosphodiesterase (PDE) inhibitors have a beneficial effect of promoting ASM relaxation by increasing intracellular cAMP resulting in PKA-mediated ASM relaxation. In murine models of asthma, PDE inhibitors have also been shown to curtail inflammation and reduce AR 110. Inhibition of PDE3 (but not PDE4) has anti-proliferative action on mitogen-activated human ASM cells in vitro, but as with most potential anti-AR drugs, useful in vivo data are lacking 111. More recently, an inhibitor of PDE8 (PF-04957325) has been shown to regulate proliferation of ASM cells by enhancing cAMP accumulation generated specifically from the β2-AR/AC6 pathway 189.

The rapamycin derivative SAR-943 has been shown to limit the mitogen-induced proliferation of human ASM cells (but not human epithelial cells) in vitro and mitigate inflammation and AR in vivo in ovalbumin-challenged mice 190, yet no clinical studies for asthma or COPD have been reported.

Conclusions

The correlation between AR and obstructive lung disease severity suggests a strong pathogenic role of AR in these diseases. Thus, remediation of AR appears critical for improving the severity and progression of these diseases. A growing arsenal of small-molecule inhibitors and biologics in conjunction with non-pharmacological interventions such as bronchial thermoplasty has shown promise in addressing this unmet clinical need. As our understanding of mechanisms underlying AR improves, so will the drug development approaches as well as the phenotyping capabilities that accurately assess AR in humans. These advances will undoubtedly fulfill our need for more refined, efficacious, and safer drugs that enable us to finally control the entire spectrum of asthma pathology.

Abbreviations

AR, airway remodeling; β 2-AR, beta 2 adrenergic receptor; ASM, airway smooth muscle; cAMP, cyclic adenosine monophosphate; c-kit, Stem cell growth factor receptor; COPD, chronic obstructive pulmonary disease; CysLTR, cysteinyl leukotriene receptor; ECM, extracellular matrix; EGFR, epidermal growth factor receptor; EP, E prostanoid; ERK1/2, extracellular signal-regulated kinases 1/2; GPCR, G protein–coupled receptor; FGFR, fibroblast growth factor receptor; ICS, inhaled corticosteroids; IgE, immunoglobulin E; IL, interleukin; JAK, Janus kinase; JNK, c-Jun N-terminal kinase; mAChR, muscarinic acetylcholine receptor; MAPK, mitogen-activated protein kinase; MEK1, mitogen-activated protein kinase kinase 1; MMP, matrix metalloproteinase; MUC5AC: mucin 5AC, oligomeric mucus/gel-forming; NF-κB, nuclear factor-kappa B; PDE, phosphodiesterase; PDGFR, platelet-derived growth factor receptor; PGE 2, prostaglandin E 2; PI-3K, phosphatidylinositol-4,5 bisphosphate 3-kinase; PKA, protein kinase A; PKC, protein kinase C; ROCK, Rho-associated proteinase kinase; RTK, receptor tyrosine kinase; Smad2: Mothers against decapentaplegic homolog 2; Syk, spleen tyrosine kinase; TAK1, Transforming growth factor-β-activated kinase 1; TAS2R, bitter taste receptor; TGF-β, transforming growth factor-beta; T-βRI, transforming growth factor-beta receptor type I kinase; VEGFR; vascular endothelial growth factor receptor

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Omar Tliba, Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, The State University of New Jersey, New Jersey, USA

  • Rennolds Ostrom, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, California, USA

Funding Statement

The work in Dr. Penn’s lab is supported by National Institutes of Health (NIH) grants HL58506, AI110007, HL136209, and HL114471. The work in Dr. Deshpande’s lab is supported by NIH grants AG041265 and AI126492.

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

[version 1; referees: 2 approved]

References

  • 1. James AL, Maxwell PS, Pearce-Pinto G, et al. : The relationship of reticular basement membrane thickness to airway wall remodeling in asthma. Am J Respir Crit Care Med. 2002;166(12 Pt 1):1590–5. 10.1164/rccm.2108069 [DOI] [PubMed] [Google Scholar]
  • 2. Prakash YS, Halayko AJ, Gosens R, et al. : An Official American Thoracic Society Research Statement: Current Challenges Facing Research and Therapeutic Advances in Airway Remodeling. Am J Respir Crit Care Med. 2017;195(2):e4–e19. 10.1164/rccm.201611-2248ST [DOI] [PubMed] [Google Scholar]
  • 3. HUBER HL, KOESSLER KK: THE PATHOLOGY OF BRONCHIAL ASTHMA. Arch Intern Med. 1922;30:689 10.1001/archinte.1922.00110120002001 [DOI] [Google Scholar]
  • 4. Kuwano K, Bosken CH, Paré PD, et al. : Small airways dimensions in asthma and in chronic obstructive pulmonary disease. Am Rev Respir Dis. 1993;148(5):1220–5. 10.1164/ajrccm/148.5.1220 [DOI] [PubMed] [Google Scholar]
  • 5. Halwani R, Al-Muhsen S, Hamid Q: Airway remodeling in asthma. Curr Opin Pharmacol. 2010;10(3):236–45. 10.1016/j.coph.2010.06.004 [DOI] [PubMed] [Google Scholar]
  • 6. Fahy JV: Remodeling of the airway epithelium in asthma. Am J Respir Crit Care Med. 2001;164(10 Pt 2):S46–51. 10.1164/ajrccm.164.supplement_2.2106066 [DOI] [PubMed] [Google Scholar]
  • 7. Davies DE: The bronchial epithelium in chronic and severe asthma. Curr Allergy Asthma Rep. 2001;1(2):127–33. 10.1007/s11882-001-0080-9 [DOI] [PubMed] [Google Scholar]
  • 8. Holgate ST, Arshad HS, Roberts GC, et al. : A new look at the pathogenesis of asthma. Clin Sci (Lond). 2009;118(7):439–50. 10.1042/CS20090474 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Loxham M, Davies DE, Blume C: Epithelial function and dysfunction in asthma. Clin Exp Allergy. 2014;44(11):1299–313. 10.1111/cea.12309 [DOI] [PubMed] [Google Scholar]
  • 10. Lambrecht BN, Hammad H: The airway epithelium in asthma. Nat Med. 2012;18(5):684–92. 10.1038/nm.2737 [DOI] [PubMed] [Google Scholar]
  • 11. Erle DJ, Sheppard D: The cell biology of asthma. J Cell Biol. 2014;205(5):621–31. 10.1083/jcb.201401050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Holgate ST: The airway epithelium is central to the pathogenesis of asthma. Allergol Int. 2008;57(1):1–10. 10.2332/allergolint.R-07-154 [DOI] [PubMed] [Google Scholar]
  • 13. Holgate ST: Pathogenesis of asthma. Clin Exp Allergy. 2008;38(6):872–97. 10.1111/j.1365-2222.2008.02971.x [DOI] [PubMed] [Google Scholar]
  • 14. Sumi Y, Hamid Q: Airway remodeling in asthma. Allergol Int. 2007;56(4):341–8. 10.2332/allergolint.R-07-153 [DOI] [PubMed] [Google Scholar]
  • 15. Hirota JA, Hackett TL, Inman MD, et al. : Modeling asthma in mice: what have we learned about the airway epithelium? Am J Respir Cell Mol Biol. 2011;44(4):431–8. 10.1165/rcmb.2010-0146TR [DOI] [PubMed] [Google Scholar]
  • 16. Bossé Y, Paré PD, Seow CY: Airway wall remodeling in asthma: from the epithelial layer to the adventitia. Curr Allergy Asthma Rep. 2008;8(4):357–66. 10.1007/s11882-008-0056-0 [DOI] [PubMed] [Google Scholar]
  • 17. Proud D, Leigh R: Epithelial cells and airway diseases. Immunol Rev. 2011;242(1):186–204. 10.1111/j.1600-065X.2011.01033.x [DOI] [PubMed] [Google Scholar]
  • 18. Aikawa T, Shimura S, Sasaki H, et al. : Marked goblet cell hyperplasia with mucus accumulation in the airways of patients who died of severe acute asthma attack. Chest. 1992;101(4):916–21. 10.1378/chest.101.4.916 [DOI] [PubMed] [Google Scholar]
  • 19. Kim KC, McCracken K, Lee BC, et al. : Airway goblet cell mucin: its structure and regulation of secretion. Eur Respir J. 1997;10(11):2644–9. 10.1183/09031936.97.10112644 [DOI] [PubMed] [Google Scholar]
  • 20. Pascoe CD, Seow CY, Hackett TL, et al. : Heterogeneity of airway wall dimensions in humans: a critical determinant of lung function in asthmatics and nonasthmatics. Am J Physiol Lung Cell Mol Physiol. 2017;312(3):L425–L431. 10.1152/ajplung.00421.2016 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 21. Prakash YS: Airway smooth muscle in airway reactivity and remodeling: what have we learned? Am J Physiol Lung Cell Mol Physiol. 2013;305(12):L912–33. 10.1152/ajplung.00259.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Gosens R, Grainge C: Bronchoconstriction and airway biology: potential impact and therapeutic opportunities. Chest. 2015;147(3):798–803. 10.1378/chest.14-1142 [DOI] [PubMed] [Google Scholar]
  • 23. Noble PB, Pascoe CD, Lan B, et al. : Airway smooth muscle in asthma: linking contraction and mechanotransduction to disease pathogenesis and remodelling. Pulm Pharmacol Ther. 2014;29(2):96–107. 10.1016/j.pupt.2014.07.005 [DOI] [PubMed] [Google Scholar]
  • 24. Johnson PR: Role of human airway smooth muscle in altered extracellular matrix production in asthma. Clin Exp Pharmacol Physiol. 2001;28(3):233–6. 10.1046/j.1440-1681.2001.03426.x [DOI] [PubMed] [Google Scholar]
  • 25. Johnson PR, Burgess JK: Airway smooth muscle and fibroblasts in the pathogenesis of asthma. Curr Allergy Asthma Rep. 2004;4(2):102–8. 10.1007/s11882-004-0054-9 [DOI] [PubMed] [Google Scholar]
  • 26. Panettieri RA, Jr: Airway smooth muscle: an immunomodulatory cell. J Allergy Clin Immunol. 2002;110(6 Suppl):S269–74. 10.1067/mai.2002.129429 [DOI] [PubMed] [Google Scholar]
  • 27. Chakir J, Haj-Salem I, Gras D, et al. : Effects of Bronchial Thermoplasty on Airway Smooth Muscle and Collagen Deposition in Asthma. Ann Am Thorac Soc. 2015;12(11):1612–8. 10.1513/AnnalsATS.201504-208OC [DOI] [PubMed] [Google Scholar]
  • 28. Pain M, Bermudez O, Lacoste P, et al. : Tissue remodelling in chronic bronchial diseases: from the epithelial to mesenchymal phenotype. Eur Respir Rev. 2014;23(131):118–30. 10.1183/09059180.00004413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Ball SL, Mann DA, Wilson JA, et al. : The Role of the Fibroblast in Inflammatory Upper Airway Conditions. Am J Pathol. 2016;186(2):225–33. 10.1016/j.ajpath.2015.09.020 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 30. Bonacci JV, Stewart AG: Regulation of human airway mesenchymal cell proliferation by glucocorticoids and beta 2-adrenoceptor agonists. Pulm Pharmacol Ther. 2006;19(1):32–8. 10.1016/j.pupt.2005.02.011 [DOI] [PubMed] [Google Scholar]
  • 31. Girodet PO, Ozier A, Bara I, et al. : Airway remodeling in asthma: new mechanisms and potential for pharmacological intervention. Pharmacol Ther. 2011;130(3):325–37. 10.1016/j.pharmthera.2011.02.001 [DOI] [PubMed] [Google Scholar]
  • 32. Hassan M, Jo T, Risse PA, et al. : Airway smooth muscle remodeling is a dynamic process in severe long-standing asthma. J Allergy Clin Immunol. 2010;125(5):1037–1045.e3. 10.1016/j.jaci.2010.02.031 [DOI] [PubMed] [Google Scholar]
  • 33. Mitsunobu F, Tanizaki Y: The use of computed tomography to assess asthma severity. Curr Opin Allergy Clin Immunol. 2005;5(1):85–90. 10.1097/00130832-200502000-00015 [DOI] [PubMed] [Google Scholar]
  • 34. Bai TR: Evidence for airway remodeling in chronic asthma. Curr Opin Allergy Clin Immunol. 2010;10(1):82–6. 10.1097/ACI.0b013e32833363b2 [DOI] [PubMed] [Google Scholar]
  • 35. Yamauchi K: Airway remodeling in asthma and its influence on clinical pathophysiology. Tohoku J Exp Med. 2006;209(2):75–87. 10.1620/tjem.209.75 [DOI] [PubMed] [Google Scholar]
  • 36. Niimi A, Matsumoto H, Takemura M, et al. : Clinical assessment of airway remodeling in asthma: utility of computed tomography. Clin Rev Allergy Immunol. 2004;27(1):45–58. 10.1385/CRIAI:27:1:045 [DOI] [PubMed] [Google Scholar]
  • 37. Aysola R, de Lange EE, Castro M, et al. : Demonstration of the heterogeneous distribution of asthma in the lungs using CT and hyperpolarized helium-3 MRI. J Magn Reson Imaging. 2010;32(6):1379–87. 10.1002/jmri.22388 [DOI] [PubMed] [Google Scholar]
  • 38. Montaudon M, Lederlin M, Reich S, et al. : Bronchial measurements in patients with asthma: comparison of quantitative thin-section CT findings with those in healthy subjects and correlation with pathologic findings. Radiology. 2009;253(3):844–53. 10.1148/radiol.2533090303 [DOI] [PubMed] [Google Scholar]
  • 39. Cox G, Thomson NC, Rubin AS, et al. : Asthma control during the year after bronchial thermoplasty. N Engl J Med. 2007;356(13):1327–37. 10.1056/NEJMoa064707 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 40. Pavord ID, Cox G, Thomson NC, et al. : Safety and efficacy of bronchial thermoplasty in symptomatic, severe asthma. Am J Respir Crit Care Med. 2007;176(12):1185–91. 10.1164/rccm.200704-571OC [DOI] [PubMed] [Google Scholar]
  • 41. Wahidi MM, Kraft M: Bronchial thermoplasty for severe asthma. Am J Respir Crit Care Med. 2012;185(7):709–14. 10.1164/rccm.201105-0883CI [DOI] [PubMed] [Google Scholar]
  • 42. Trian T, Allard B, Dupin I, et al. : House dust mites induce proliferation of severe asthmatic smooth muscle cells via an epithelium-dependent pathway. Am J Respir Crit Care Med. 2015;191(5):538–46. 10.1164/rccm.201409-1582OC [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 43. Grainge CL, Lau LC, Ward JA, et al. : Effect of bronchoconstriction on airway remodeling in asthma. N Engl J Med. 2011;364(21):2006–15. 10.1056/NEJMoa1014350 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 44. Kistemaker LE, Oenema TA, Meurs H, et al. : Regulation of airway inflammation and remodeling by muscarinic receptors: perspectives on anticholinergic therapy in asthma and COPD. Life Sci. 2012;91(21-22):1126–33. 10.1016/j.lfs.2012.02.021 [DOI] [PubMed] [Google Scholar]
  • 45. Henderson WR, Jr, Chiang GK, Tien YT, et al. : Reversal of allergen-induced airway remodeling by CysLT 1 receptor blockade. Am J Respir Crit Care Med. 2006;173(7):718–28. 10.1164/rccm.200501-088OC [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 46. Muz MH, Deveci F, Bulut Y, et al. : The effects of low dose leukotriene receptor antagonist therapy on airway remodeling and cysteinyl leukotriene expression in a mouse asthma model. Exp Mol Med. 2006;38(2):109–18. 10.1038/emm.2006.14 [DOI] [PubMed] [Google Scholar]
  • 47. Kelly MM, Chakir J, Vethanayagam D, et al. : Montelukast treatment attenuates the increase in myofibroblasts following low-dose allergen challenge. Chest. 2006;130(3):741–53. 10.1378/chest.130.3.741 [DOI] [PubMed] [Google Scholar]
  • 48. Gosens R, Bos IS, Zaagsma J, et al. : Protective effects of tiotropium bromide in the progression of airway smooth muscle remodeling. Am J Respir Crit Care Med. 2005;171(10):1096–102. 10.1164/rccm.200409-1249OC [DOI] [PubMed] [Google Scholar]
  • 49. Pera T, Zuidhof A, Valadas J, et al. : Tiotropium inhibits pulmonary inflammation and remodelling in a guinea pig model of COPD. Eur Respir J. 2011;38(4):789–96. 10.1183/09031936.00146610 [DOI] [PubMed] [Google Scholar]
  • 50. Ohta S, Oda N, Yokoe T, et al. : Effect of tiotropium bromide on airway inflammation and remodelling in a mouse model of asthma. Clin Exp Allergy. 2010;40(8):1266–75. 10.1111/j.1365-2222.2010.03478.x [DOI] [PubMed] [Google Scholar]
  • 51. Kang JY, Rhee CK, Kim JS, et al. : Effect of tiotropium bromide on airway remodeling in a chronic asthma model. Ann Allergy Asthma Immunol. 2012;109(1):29–35. 10.1016/j.anai.2012.05.005 [DOI] [PubMed] [Google Scholar]
  • 52. Bos IS, Gosens R, Zuidhof AB, et al. : Inhibition of allergen-induced airway remodelling by tiotropium and budesonide: a comparison. Eur Respir J. 2007;30(4):653–61. 10.1183/09031936.00004907 [DOI] [PubMed] [Google Scholar]
  • 53. Levine SJ, Larivée P, Logun C, et al. : Corticosteroids differentially regulate secretion of IL-6, IL-8, and G-CSF by a human bronchial epithelial cell line. Am J Physiol. 1993;265(4 Pt 1):L360–8. 10.1152/ajplung.1993.265.4.L360 [DOI] [PubMed] [Google Scholar]
  • 54. Dorscheid DR, Wojcik KR, Sun S, et al. : Apoptosis of airway epithelial cells induced by corticosteroids. Am J Respir Crit Care Med. 2001;164(10 Pt 1):1939–47. 10.1164/ajrccm.164.10.2103013 [DOI] [PubMed] [Google Scholar]
  • 55. Sabatini F, Silvestri M, Sale R, et al. : Concentration-dependent effects of mometasone furoate and dexamethasone on foetal lung fibroblast functions involved in airway inflammation and remodeling. Pulm Pharmacol Ther. 2003;16(5):287–97. 10.1016/S1094-5539(03)00068-3 [DOI] [PubMed] [Google Scholar]
  • 56. Descalzi D, Folli C, Nicolini G, et al. : Anti-proliferative and anti-remodelling effect of beclomethasone dipropionate, formoterol and salbutamol alone or in combination in primary human bronchial fibroblasts. Allergy. 2008;63(4):432–7. 10.1111/j.1398-9995.2007.01582.x [DOI] [PubMed] [Google Scholar]
  • 57. Young PG, Skinner SJ, Black PN: Effects of glucocorticoids and beta-adrenoceptor agonists on the proliferation of airway smooth muscle. Eur J Pharmacol. 1995;273(1–2):137–43. 10.1016/0014-2999(94)00679-2 [DOI] [PubMed] [Google Scholar]
  • 58. Stewart AG, Fernandes D, Tomlinson PR: The effect of glucocorticoids on proliferation of human cultured airway smooth muscle. Br J Pharmacol. 1995;116(8):3219–26. 10.1111/j.1476-5381.1995.tb15127.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Damera G, Fogle HW, Lim P, et al. : Vitamin D inhibits growth of human airway smooth muscle cells through growth factor-induced phosphorylation of retinoblastoma protein and checkpoint kinase 1. Br J Pharmacol. 2009;158(6):1429–41. 10.1111/j.1476-5381.2009.00428.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Perry MM, Baker JE, Gibeon DS, et al. : Airway smooth muscle hyperproliferation is regulated by microRNA-221 in severe asthma. Am J Respir Cell Mol Biol. 2014;50(1):7–17. 10.1165/rcmb.2013-0067OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ward C, Reid DW, Orsida BE, et al. : Inter-relationships between airway inflammation, reticular basement membrane thickening and bronchial hyper-reactivity to methacholine in asthma; a systematic bronchoalveolar lavage and airway biopsy analysis. Clin Exp Allergy. 2005;35(12):1565–71. 10.1111/j.1365-2222.2005.02365.x [DOI] [PubMed] [Google Scholar]
  • 62. Bergeron C, Tulic MK, Hamid Q: Airway remodelling in asthma: from benchside to clinical practice. Can Respir J. 2010;17(4):e85–93. 10.1155/2010/318029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Laitinen A, Altraja A, Kämpe M, et al. : Tenascin is increased in airway basement membrane of asthmatics and decreased by an inhaled steroid. Am J Respir Crit Care Med. 1997;156(3 Pt 1):951–8. 10.1164/ajrccm.156.3.9610084 [DOI] [PubMed] [Google Scholar]
  • 64. Laitinen LA, Laitinen A: Inhaled corticosteroid treatment for asthma. Allergy Proc. 1995;16(2):63–6. 10.1002/9780470987377.ch12 [DOI] [PubMed] [Google Scholar]
  • 65. Boulet LP, Turcotte H, Laviolette M, et al. : Airway hyperresponsiveness, inflammation, and subepithelial collagen deposition in recently diagnosed versus long-standing mild asthma. Influence of inhaled corticosteroids. Am J Respir Crit Care Med. 2000;162(4 Pt 1):1308–13. 10.1164/ajrccm.162.4.9910051 [DOI] [PubMed] [Google Scholar]
  • 66. Olivieri D, Chetta A, Del Donno M, et al. : Effect of short-term treatment with low-dose inhaled fluticasone propionate on airway inflammation and remodeling in mild asthma: a placebo-controlled study. Am J Respir Crit Care Med. 1997;155(6):1864–71. 10.1164/ajrccm.155.6.9196087 [DOI] [PubMed] [Google Scholar]
  • 67. Trigg CJ, Manolitsas ND, Wang J, et al. : Placebo-controlled immunopathologic study of four months of inhaled corticosteroids in asthma. Am J Respir Crit Care Med. 1994;150(1):17–22. 10.1164/ajrccm.150.1.8025745 [DOI] [PubMed] [Google Scholar]
  • 68. Hoshino M, Takahashi M, Takai Y, et al. : Inhaled corticosteroids decrease subepithelial collagen deposition by modulation of the balance between matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 expression in asthma. J Allergy Clin Immunol. 1999;104(2 Pt 1):356–63. 10.1016/S0091-6749(99)70379-9 [DOI] [PubMed] [Google Scholar]
  • 69. Sont JK, Willems LN, Bel EH, et al. : Clinical control and histopathologic outcome of asthma when using airway hyperresponsiveness as an additional guide to long-term treatment. The AMPUL Study Group. Am J Respir Crit Care Med. 1999;159(4 Pt 1):1043–51. 10.1164/ajrccm.159.4.9806052 [DOI] [PubMed] [Google Scholar]
  • 70. Chakir J, Shannon J, Molet S, et al. : Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J Allergy Clin Immunol. 2003;111(6):1293–8. 10.1067/mai.2003.1557 [DOI] [PubMed] [Google Scholar]
  • 71. Orsida BE, Li X, Hickey B, et al. : Vascularity in asthmatic airways: relation to inhaled steroid dose. Thorax. 1999;54(4):289–95. 10.1136/thx.54.4.289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Belvisi MG, Saunders M, Yacoub M, et al. : Expression of cyclo-oxygenase-2 in human airway smooth muscle is associated with profound reductions in cell growth. Br J Pharmacol. 1998;125(5):1102–8. 10.1038/sj.bjp.0702104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Misior AM, Yan H, Pascual RM, et al. : Mitogenic effects of cytokines on smooth muscle are critically dependent on protein kinase A and are unmasked by steroids and cyclooxygenase inhibitors. Mol Pharmacol. 2008;73(2):566–74. 10.1124/mol.107.040519 [DOI] [PubMed] [Google Scholar]
  • 74. Yan H, Deshpande DA, Misior AM, et al. : Anti-mitogenic effects of β-agonists and PGE 2 on airway smooth muscle are PKA dependent. FASEB J. 2011;25(1):389–97. 10.1096/fj.10-164798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Kawakami Y, Uchiyama K, Irie T, et al. : Evaluation of aerosols of prostaglandins E 1 and E 2 as bronchodilators. Eur J Clin Pharmacol. 1973;6(2):127–32. 10.1007/BF00562439 [DOI] [PubMed] [Google Scholar]
  • 76. Walters EH, Davies BH: Dual effect of prostaglandin E 2 on normal airways smooth muscle in vivo. Thorax. 1982;37(12):918–22. 10.1136/thx.37.12.918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Costello JF, Dunlop LS, Gardiner PJ: Characteristics of prostaglandin induced cough in man. Br J Clin Pharmacol. 1985;20(4):355–9. 10.1111/j.1365-2125.1985.tb05077.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Coleridge HM, Coleridge JC, Ginzel KH, et al. : Stimulation of 'irritant' receptors and afferent C-fibres in the lungs by prostaglandins. Nature. 1976;264(5585):451–3. 10.1038/264451a0 [DOI] [PubMed] [Google Scholar]
  • 79. Gauvreau GM, Watson RM, O'Byrne PM: Protective effects of inhaled PGE 2 on allergen-induced airway responses and airway inflammation. Am J Respir Crit Care Med. 1999;159(1):31–6. 10.1164/ajrccm.159.1.9804030 [DOI] [PubMed] [Google Scholar]
  • 80. Maher SA, Birrell MA, Belvisi MG: Prostaglandin E 2 mediates cough via the EP 3 receptor: implications for future disease therapy. Am J Respir Crit Care Med. 2009;180(10):923–8. 10.1164/rccm.200903-0388OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sharma P, Panebra A, Pera T, et al. : Antimitogenic effect of bitter taste receptor agonists on airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2016;310(4):L365–76. 10.1152/ajplung.00373.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Pan S, Sharma P, Shah SD, et al. : Bitter taste receptor agonists alter mitochondrial function and induce autophagy in airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2017;313(1):L154–L165. 10.1152/ajplung.00106.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Sharma P, Yi R, Nayak AP, et al. : Bitter Taste Receptor Agonists Mitigate Features of Allergic Asthma in Mice. Sci Rep. 2017;7: 46166. 10.1038/srep46166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Pera T, Penn RB: Bronchoprotection and bronchorelaxation in asthma: New targets, and new ways to target the old ones. Pharmacol Ther. 2016;164:82–96. 10.1016/j.pharmthera.2016.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Flood-Page P, Menzies-Gow A, Phipps S, et al. : Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112(7):1029–36. 10.1172/JCI17974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Blyth DI, Wharton TF, Pedrick MS, et al. : Airway subepithelial fibrosis in a murine model of atopic asthma: suppression by dexamethasone or anti-interleukin-5 antibody. Am J Respir Cell Mol Biol. 2000;23(2):241–6. 10.1165/ajrcmb.23.2.3999 [DOI] [PubMed] [Google Scholar]
  • 87. Riccio AM, Mauri P, De Ferrari L, et al. : Galectin-3: an early predictive biomarker of modulation of airway remodeling in patients with severe asthma treated with omalizumab for 36 months. Clin Transl Allergy. 2017;7:6. 10.1186/s13601-017-0143-1 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 88. Li N, Li Q, Zhou XD, et al. : Chronic mechanical stress induces mucin 5AC expression in human bronchial epithelial cells through ERK dependent pathways. Mol Biol Rep. 2012;39(2):1019–28. 10.1007/s11033-011-0827-x [DOI] [PubMed] [Google Scholar]
  • 89. Shinkai M, López-Boado YS, Rubin BK: Clarithromycin has an immunomodulatory effect on ERK-mediated inflammation induced by Pseudomonas aeruginosa flagellin. J Antimicrob Chemother. 2007;59(6):1096–101. 10.1093/jac/dkm084 [DOI] [PubMed] [Google Scholar]
  • 90. Nath P, Leung SY, Williams A, et al. : Importance of p38 mitogen-activated protein kinase pathway in allergic airway remodelling and bronchial hyperresponsiveness. Eur J Pharmacol. 2006;544(1–3):160–7. 10.1016/j.ejphar.2006.06.031 [DOI] [PubMed] [Google Scholar]
  • 91. Nath P, Eynott P, Leung SY, et al. : Potential role of c-Jun NH 2-terminal kinase in allergic airway inflammation and remodelling: effects of SP600125. Eur J Pharmacol. 2005;506(3):273–83. 10.1016/j.ejphar.2004.11.040 [DOI] [PubMed] [Google Scholar]
  • 92. Wu H, Fang L, Shen QY, et al. : SP600125 promotes resolution of allergic airway inflammation via TLR9 in an OVA-induced murine acute asthma model. Mol Immunol. 2015;67(2 Pt B):311–6. 10.1016/j.molimm.2015.06.016 [DOI] [PubMed] [Google Scholar]
  • 93. Eynott PR, Nath P, Leung SY, et al. : Allergen-induced inflammation and airway epithelial and smooth muscle cell proliferation: role of Jun N-terminal kinase. Br J Pharmacol. 2003;140(8):1373–80. 10.1038/sj.bjp.0705569 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Pera T, Atmaj C, van der Vegt M, et al. : Role for TAK1 in cigarette smoke-induced proinflammatory signaling and IL-8 release by human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2012;303(3):L272–8. 10.1152/ajplung.00291.2011 [DOI] [PubMed] [Google Scholar]
  • 95. Pera T, Sami R, Zaagsma J, et al. : TAK1 plays a major role in growth factor-induced phenotypic modulation of airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2011;301(5):L822–8. 10.1152/ajplung.00017.2011 [DOI] [PubMed] [Google Scholar]
  • 96. Krymskaya VP, Orsini MJ, Eszterhas AJ, et al. : Mechanisms of proliferation synergy by receptor tyrosine kinase and G protein-coupled receptor activation in human airway smooth muscle. Am J Respir Cell Mol Biol. 2000;23(4):546–54. 10.1165/ajrcmb.23.4.4115 [DOI] [PubMed] [Google Scholar]
  • 97. Puddicombe SM, Polosa R, Richter A, et al. : Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 2000;14(10):1362–74. 10.1096/fj.14.10.1362 [DOI] [PubMed] [Google Scholar]
  • 98. Booth BW, Adler KB, Bonner JC, et al. : Interleukin-13 induces proliferation of human airway epithelial cells in vitro via a mechanism mediated by transforming growth factor-alpha. Am J Respir Cell Mol Biol. 2001;25(6):739–43. 10.1165/ajrcmb.25.6.4659 [DOI] [PubMed] [Google Scholar]
  • 99. Hegab AE, Sakamoto T, Nomura A, et al. : Niflumic acid and AG-1478 reduce cigarette smoke-induced mucin synthesis: the role of hCLCA1. Chest. 2007;131(4):1149–56. 10.1378/chest.06-2031 [DOI] [PubMed] [Google Scholar]
  • 100. Takezawa K, Ogawa T, Shimizu S, et al. : Epidermal growth factor receptor inhibitor AG1478 inhibits mucus hypersecretion in airway epithelium. Am J Rhinol Allergy. 2016;30(1):1–6. 10.2500/ajra.2016.30.4263 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 101. Burgel PR, Lazarus SC, Tam DC, et al. : Human eosinophils induce mucin production in airway epithelial cells via epidermal growth factor receptor activation. J Immunol. 2001;167(10):5948–54. 10.4049/jimmunol.167.10.5948 [DOI] [PubMed] [Google Scholar]
  • 102. Kim S, Lewis C, Nadel JA: CCL20/CCR6 feedback exaggerates epidermal growth factor receptor-dependent MUC5AC mucin production in human airway epithelial (NCI-H292) cells. J Immunol. 2011;186(6):3392–400. 10.4049/jimmunol.1003377 [DOI] [PubMed] [Google Scholar]
  • 103. Le Cras TD, Acciani TH, Mushaben EM, et al. : Epithelial EGF receptor signaling mediates airway hyperreactivity and remodeling in a mouse model of chronic asthma. Am J Physiol Lung Cell Mol Physiol. 2011;300(3):L414–21. 10.1152/ajplung.00346.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Bossé Y, Thompson C, Stankova J, et al. : Fibroblast growth factor 2 and transforming growth factor beta1 synergism in human bronchial smooth muscle cell proliferation. Am J Respir Cell Mol Biol. 2006;34(6):746–53. 10.1165/rcmb.2005-0309OC [DOI] [PubMed] [Google Scholar]
  • 105. Berlin AA, Hogaboam CM, Lukacs NW: Inhibition of SCF attenuates peribronchial remodeling in chronic cockroach allergen-induced asthma. Lab Invest. 2006;86(6):557–65. 10.1038/labinvest.3700419 [DOI] [PubMed] [Google Scholar]
  • 106. Yamamoto N, Takeshita K, Shichijo M, et al. : The orally available spleen tyrosine kinase inhibitor 2-[7-(3,4-dimethoxyphenyl)-imidazo[1,2-c]pyrimidin-5-ylamino]nicotinamide dihydrochloride (BAY 61-3606) blocks antigen-induced airway inflammation in rodents. J Pharmacol Exp Ther. 2003;306(3):1174–81. 10.1124/jpet.103.052316 [DOI] [PubMed] [Google Scholar]
  • 107. Matsunaga Y, Inoue H, Fukuyama S, et al. : Effects of a Janus kinase inhibitor, pyridone 6, on airway responses in a murine model of asthma. Biochem Biophys Res Commun. 2011;404(1):261–7. 10.1016/j.bbrc.2010.11.104 [DOI] [PubMed] [Google Scholar]
  • 108. Leung SY, Niimi A, Noble A, et al. : Effect of transforming growth factor-beta receptor I kinase inhibitor 2,4-disubstituted pteridine (SD-208) in chronic allergic airway inflammation and remodeling. J Pharmacol Exp Ther. 2006;319(2):586–94. 10.1124/jpet.106.109314 [DOI] [PubMed] [Google Scholar]
  • 109. Possa SS, Charafeddine HT, Righetti RF, et al. : Rho-kinase inhibition attenuates airway responsiveness, inflammation, matrix remodeling, and oxidative stress activation induced by chronic inflammation. Am J Physiol Lung Cell Mol Physiol. 2012;303(11):L939–52. 10.1152/ajplung.00034.2012 [DOI] [PubMed] [Google Scholar]
  • 110. Kumar RK, Herbert C, Thomas PS, et al. : Inhibition of inflammation and remodeling by roflumilast and dexamethasone in murine chronic asthma. J Pharmacol Exp Ther. 2003;307(1):349–55. 10.1124/jpet.103.053819 [DOI] [PubMed] [Google Scholar]
  • 111. Billington CK, Joseph SK, Swan C, et al. : Modulation of human airway smooth muscle proliferation by type 3 phosphodiesterase inhibition. Am J Physiol. 1999;276(3 Pt 1):L412–9. 10.1152/ajplung.1999.276.3.L412 [DOI] [PubMed] [Google Scholar]
  • 112. Buckley J, Birrell MA, Maher SA, et al. : EP 4 receptor as a new target for bronchodilator therapy. Thorax. 2011;66(12):1029–35. 10.1136/thx.2010.158568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Birrell MA, Maher SA, Dekkak B, et al. : Anti-inflammatory effects of PGE 2 in the lung: role of the EP 4 receptor subtype. Thorax. 2015;70(8):740–7. 10.1136/thoraxjnl-2014-206592 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Tilley SL, Hartney JM, Erikson CJ, et al. : Receptors and pathways mediating the effects of prostaglandin E 2 on airway tone. Am J Physiol Lung Cell Mol Physiol. 2003;284(4):L599–606. 10.1152/ajplung.00324.2002 [DOI] [PubMed] [Google Scholar]
  • 115. Shah AS, Ben-Shahar Y, Moninger TO, et al. : Motile cilia of human airway epithelia are chemosensory. Science. 2009;325(5944):1131–4. 10.1126/science.1173869 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 116. Aravamudan B, Thompson MA, Pabelick CM, et al. : Mitochondria in lung diseases. Expert Rev Respir Med. 2013;7(6):631–46. 10.1586/17476348.2013.834252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Prakash YS, Pabelick CM, Sieck GC: Mitochondrial Dysfunction in Airway Disease. Chest. 2017;152(3):618–26. 10.1016/j.chest.2017.03.020 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 118. Lambrecht BN, Hammad H: The immunology of asthma. Nat Immunol. 2015;16(1):45–56. 10.1038/ni.3049 [DOI] [PubMed] [Google Scholar]
  • 119. Nair P: Anti-interleukin-5 monoclonal antibody to treat severe eosinophilic asthma. N Engl J Med. 2014;371(13):1249–51. 10.1056/NEJMe1408614 [DOI] [PubMed] [Google Scholar]
  • 120. Russell R, Brightling C: Mepolizumab for the reduction of exacerbations in severe eosinophilic asthma. Expert Rev Respir Med. 2016;10(6):607–17. 10.1080/17476348.2016.1176532 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 121. Russell R, Brightling CE: Anti-IL-5 for Severe Asthma: Aiming High to Achieve Success. Chest. 2016;150(4):766–8. 10.1016/j.chest.2016.06.013 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 122. Cho JY: Recent advances in mechanisms and treatments of airway remodeling in asthma: a message from the bench side to the clinic. Korean J Intern Med. 2011;26(4):367–83. 10.3904/kjim.2011.26.4.367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Gould HJ, Sutton BJ: IgE in allergy and asthma today. Nat Rev Immunol. 2008;8(3):205–17. 10.1038/nri2273 [DOI] [PubMed] [Google Scholar]
  • 124. Bousquet J, Chiron R, Humbert M: Biologics in asthma: difficulties and drawbacks. Expert Opin Biol Ther. 2008;8(12):1921–8. 10.1517/14712590802496928 [DOI] [PubMed] [Google Scholar]
  • 125. McCracken JL, Tripple JW, Calhoun WJ: Biologic therapy in the management of asthma. Curr Opin Allergy Clin Immunol. 2016;16(4):375–82. 10.1097/ACI.0000000000000284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Darveaux J, Busse WW: Biologics in asthma--the next step toward personalized treatment. J Allergy Clin Immunol Pract. 2015;3(2):152–60; quiz 161. 10.1016/j.jaip.2014.09.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Godar M, Blanchetot C, de Haard H, et al. : Personalized medicine with biologics for severe type 2 asthma: current status and future prospects. MAbs. 2018;10(1):34–45. 10.1080/19420862.2017.1392425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Wu P, Nielsen TE, Clausen MH: FDA-approved small-molecule kinase inhibitors. Trends Pharmacol Sci. 2015;36(7):422–39. 10.1016/j.tips.2015.04.005 [DOI] [PubMed] [Google Scholar]
  • 129. Barnes PJ: Kinases as Novel Therapeutic Targets in Asthma and Chronic Obstructive Pulmonary Disease. Pharmacol Rev. 2016;68(3):788–815. 10.1124/pr.116.012518 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 130. Khorasanizadeh M, Eskian M, Gelfand EW, et al. : Mitogen-activated protein kinases as therapeutic targets for asthma. Pharmacol Ther. 2017;174:112–26. 10.1016/j.pharmthera.2017.02.024 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 131. Kim DH, Jung WS, Kim ME, et al. : Genistein inhibits pro-inflammatory cytokines in human mast cell activation through the inhibition of the ERK pathway. Int J Mol Med. 2014;34(6):1669–74. 10.3892/ijmm.2014.1956 [DOI] [PubMed] [Google Scholar]
  • 132. Kono Y, Nishiuma T, Okada T, et al. : Sphingosine kinase 1 regulates mucin production via ERK phosphorylation. Pulm Pharmacol Ther. 2010;23(1):36–42. 10.1016/j.pupt.2009.10.005 [DOI] [PubMed] [Google Scholar]
  • 133. Guntur VP, Reinero CR: The potential use of tyrosine kinase inhibitors in severe asthma. Curr Opin Allergy Clin Immunol. 2012;12(1):68–75. 10.1097/ACI.0b013e32834ecb4f [DOI] [PubMed] [Google Scholar]
  • 134. Amishima M, Munakata M, Nasuhara Y, et al. : Expression of epidermal growth factor and epidermal growth factor receptor immunoreactivity in the asthmatic human airway. Am J Respir Crit Care Med. 1998;157(6 Pt 1):1907–12. 10.1164/ajrccm.157.6.9609040 [DOI] [PubMed] [Google Scholar]
  • 135. Polosa R, Puddicombe SM, Krishna MT, et al. : Expression of c-erbB receptors and ligands in the bronchial epithelium of asthmatic subjects. J Allergy Clin Immunol. 2002;109(1):75–81. 10.1067/mai.2002.120274 [DOI] [PubMed] [Google Scholar]
  • 136. Takeyama K, Dabbagh K, Lee HM, et al. : Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci U S A. 1999;96(6):3081–6. 10.1073/pnas.96.6.3081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Takeyama K, Fahy JV, Nadel JA: Relationship of epidermal growth factor receptors to goblet cell production in human bronchi. Am J Respir Crit Care Med. 2001;163(2):511–6. 10.1164/ajrccm.163.2.2001038 [DOI] [PubMed] [Google Scholar]
  • 138. Wu LS, Tan C, Wang LM, et al. : Variant in promoter region of platelet-derived growth factor receptor-alpha (PDGFRalpha) gene is associated with the severity and allergic status of childhood asthma. Int Arch Allergy Immunol. 2006;141(1):37–46. 10.1159/000094180 [DOI] [PubMed] [Google Scholar]
  • 139. Leung TF, Wong GW, Ko FW, et al. : Analysis of growth factors and inflammatory cytokines in exhaled breath condensate from asthmatic children. Int Arch Allergy Immunol. 2005;137(1):66–72. 10.1159/000085106 [DOI] [PubMed] [Google Scholar]
  • 140. Lee CG, Link H, Baluk P, et al. : Vascular endothelial growth factor (VEGF) induces remodeling and enhances TH2-mediated sensitization and inflammation in the lung. Nat Med. 2004;10(10):1095–103. 10.1038/nm1105 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 141. Asai K, Kanazawa H, Kamoi H, et al. : Increased levels of vascular endothelial growth factor in induced sputum in asthmatic patients. Clin Exp Allergy. 2003;33(5):595–9. 10.1046/j.1365-2222.2003.01576.x [DOI] [PubMed] [Google Scholar]
  • 142. Da Silva CA, Blay F, Israel-Biet D, et al. : Effect of glucocorticoids on stem cell factor expression in human asthmatic bronchi. Clin Exp Allergy. 2006;36(3):317–24. 10.1111/j.1365-2222.2006.02431.x [DOI] [PubMed] [Google Scholar]
  • 143. Makowska JS, Cieslak M, Kowalski ML: Stem cell factor and its soluble receptor (c-kit) in serum of asthmatic patients- correlation with disease severity. BMC Pulm Med. 2009;9:27. 10.1186/1471-2466-9-27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Nadel JA, Burgel PR: The role of epidermal growth factor in mucus production. Curr Opin Pharmacol. 2001;1(3):254–8. 10.1016/S1471-4892(01)00045-5 [DOI] [PubMed] [Google Scholar]
  • 145. Hamilton LM, Torres-Lozano C, Puddicombe SM, et al. : The role of the epidermal growth factor receptor in sustaining neutrophil inflammation in severe asthma. Clin Exp Allergy. 2003;33(2):233–40. 10.1046/j.1365-2222.2003.01593.x [DOI] [PubMed] [Google Scholar]
  • 146. Takeyama K, Dabbagh K, Jeong Shim J, et al. : Oxidative stress causes mucin synthesis via transactivation of epidermal growth factor receptor: role of neutrophils. J Immunol. 2000;164(3):1546–52. 10.4049/jimmunol.164.3.1546 [DOI] [PubMed] [Google Scholar]
  • 147. Takeyama K, Jung B, Shim JJ, et al. : Activation of epidermal growth factor receptors is responsible for mucin synthesis induced by cigarette smoke. Am J Physiol Lung Cell Mol Physiol. 2001;280(1):L165–72. 10.1152/ajplung.2001.280.1.L165 [DOI] [PubMed] [Google Scholar]
  • 148. Shao MX, Nadel JA: Neutrophil elastase induces MUC5AC mucin production in human airway epithelial cells via a cascade involving protein kinase C, reactive oxygen species, and TNF-α-converting enzyme. J Immunol. 2005;175(6):4009–16. 10.4049/jimmunol.175.6.4009 [DOI] [PubMed] [Google Scholar]
  • 149. Woodruff PG, Wolff M, Hohlfeld JM, et al. : Safety and efficacy of an inhaled epidermal growth factor receptor inhibitor (BIBW 2948 BS) in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181(5):438–45. 10.1164/rccm.200909-1415OC [DOI] [PubMed] [Google Scholar]
  • 150. Hirst SJ, Barnes PJ, Twort CH: PDGF isoform-induced proliferation and receptor expression in human cultured airway smooth muscle cells. Am J Physiol. 1996;270(3 Pt 1):L415–28. 10.1152/ajplung.1996.270.3.L415 [DOI] [PubMed] [Google Scholar]
  • 151. Hirota JA, Ask K, Farkas L, et al. : In vivo role of platelet-derived growth factor-BB in airway smooth muscle proliferation in mouse lung. Am J Respir Cell Mol Biol. 2011;45(3):566–72. 10.1165/rcmb.2010-0277OC [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 152. Humbert M, de Blay F, Garcia G, et al. : Masitinib, a c-kit/PDGF receptor tyrosine kinase inhibitor, improves disease control in severe corticosteroid-dependent asthmatics. Allergy. 2009;64(8):1194–201. 10.1111/j.1398-9995.2009.02122.x [DOI] [PubMed] [Google Scholar]
  • 153. Chetta A, Zanini A, Foresi A, et al. : Vascular endothelial growth factor up-regulation and bronchial wall remodelling in asthma. Clin Exp Allergy. 2005;35(11):1437–42. 10.1111/j.1365-2222.2005.02360.x [DOI] [PubMed] [Google Scholar]
  • 154. Zanini A, Chetta A, Saetta M, et al. : Chymase-positive mast cells play a role in the vascular component of airway remodeling in asthma. J Allergy Clin Immunol. 2007;120(2):329–33. 10.1016/j.jaci.2007.04.021 [DOI] [PubMed] [Google Scholar]
  • 155. Hoshino M, Nakamura Y, Hamid QA: Gene expression of vascular endothelial growth factor and its receptors and angiogenesis in bronchial asthma. J Allergy Clin Immunol. 2001;107(6):1034–8. 10.1067/mai.2001.115626 [DOI] [PubMed] [Google Scholar]
  • 156. Hoshino M, Takahashi M, Aoike N: Expression of vascular endothelial growth factor, basic fibroblast growth factor, and angiogenin immunoreactivity in asthmatic airways and its relationship to angiogenesis. J Allergy Clin Immunol. 2001;107(2):295–301. 10.1067/mai.2001.111928 [DOI] [PubMed] [Google Scholar]
  • 157. Simcock DE, Kanabar V, Clarke GW, et al. : Induction of angiogenesis by airway smooth muscle from patients with asthma. Am J Respir Crit Care Med. 2008;178(5):460–8. 10.1164/rccm.200707-1046OC [DOI] [PubMed] [Google Scholar]
  • 158. Simcock DE, Kanabar V, Clarke GW, et al. : Proangiogenic activity in bronchoalveolar lavage fluid from patients with asthma. Am J Respir Crit Care Med. 2007;176(2):146–53. 10.1164/rccm.200701-042OC [DOI] [PubMed] [Google Scholar]
  • 159. Keglowich L, Roth M, Philippova M, et al. : Bronchial smooth muscle cells of asthmatics promote angiogenesis through elevated secretion of CXC-chemokines (ENA-78, GRO-α, and IL-8). PLoS One. 2013;8(12):e81494. 10.1371/journal.pone.0081494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Barbato A, Turato G, Baraldo S, et al. : Epithelial damage and angiogenesis in the airways of children with asthma. Am J Respir Crit Care Med. 2006;174(9):975–81. 10.1164/rccm.200602-189OC [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 161. Vrugt B, Wilson S, Bron A, et al. : Bronchial angiogenesis in severe glucocorticoid-dependent asthma. Eur Respir J. 2000;15(6):1014–21. 10.1034/j.1399-3003.2000.01507.x [DOI] [PubMed] [Google Scholar]
  • 162. Tanaka H, Yamada G, Saikai T, et al. : Increased airway vascularity in newly diagnosed asthma using a high-magnification bronchovideoscope. Am J Respir Crit Care Med. 2003;168(12):1495–9. 10.1164/rccm.200306-727OC [DOI] [PubMed] [Google Scholar]
  • 163. Nagy JA, Benjamin L, Zeng H, et al. : Vascular permeability, vascular hyperpermeability and angiogenesis. Angiogenesis. 2008;11(2):109–19. 10.1007/s10456-008-9099-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Li X, Wilson JW: Increased vascularity of the bronchial mucosa in mild asthma. Am J Respir Crit Care Med. 1997;156(1):229–33. 10.1164/ajrccm.156.1.9607066 [DOI] [PubMed] [Google Scholar]
  • 165. Kim YS, Hong SW, Choi JP, et al. : Vascular endothelial growth factor is a key mediator in the development of T cell priming and its polarization to type 1 and type 17 T helper cells in the airways. J Immunol. 2009;183(8):5113–20. 10.4049/jimmunol.0901566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Lee HY, Hur J, Kim IK, et al. : Effect of nintedanib on airway inflammation and remodeling in a murine chronic asthma model. Exp Lung Res. 2017;43(4–5):187–96. 10.1080/01902148.2017.1339141 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 167. Berlin AA, Lukacs NW: Treatment of cockroach allergen asthma model with imatinib attenuates airway responses. Am J Respir Crit Care Med. 2005;171(1):35–9. 10.1164/rccm.200403-385OC [DOI] [PubMed] [Google Scholar]
  • 168. Seow CJ, Chue SC, Wong WS: Piceatannol, a Syk-selective tyrosine kinase inhibitor, attenuated antigen challenge of guinea pig airways in vitro. Eur J Pharmacol. 2002;443(1–3):189–96. 10.1016/S0014-2999(02)01534-0 [DOI] [PubMed] [Google Scholar]
  • 169. Sakai H, Nishimura A, Watanabe Y, et al. : Involvement of Src family kinase activation in angiotensin II-induced hyperresponsiveness of rat bronchial smooth muscle. Peptides. 2010;31(12):2216–21. 10.1016/j.peptides.2010.09.012 [DOI] [PubMed] [Google Scholar]
  • 170. Kudlacz E, Conklyn M, Andresen C, et al. : The JAK-3 inhibitor CP-690550 is a potent anti-inflammatory agent in a murine model of pulmonary eosinophilia. Eur J Pharmacol. 2008;582(1–3):154–61. 10.1016/j.ejphar.2007.12.024 [DOI] [PubMed] [Google Scholar]
  • 171. Ghoreschi K, Jesson MI, Li X, et al. : Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol. 2011;186(7):4234–43. 10.4049/jimmunol.1003668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Catley MC, Sukkar MB, Chung KF, et al. : Validation of the anti-inflammatory properties of small-molecule IkappaB Kinase (IKK)-2 inhibitors by comparison with adenoviral-mediated delivery of dominant-negative IKK1 and IKK2 in human airways smooth muscle. Mol Pharmacol. 2006;70(2):697–705. 10.1124/mol.106.023150 [DOI] [PubMed] [Google Scholar]
  • 173. Newton R, Holden NS, Catley MC, et al. : Repression of inflammatory gene expression in human pulmonary epithelial cells by small-molecule IkappaB kinase inhibitors. J Pharmacol Exp Ther. 2007;321(2):734–42. 10.1124/jpet.106.118125 [DOI] [PubMed] [Google Scholar]
  • 174. Doukas J, Eide L, Stebbins K, et al. : Aerosolized phosphoinositide 3-kinase gamma/delta inhibitor TG100-115 [3-[2,4-diamino-6-(3-hydroxyphenyl)pteridin-7-yl]phenol] as a therapeutic candidate for asthma and chronic obstructive pulmonary disease. J Pharmacol Exp Ther. 2009;328(3):758–65. 10.1124/jpet.108.144311 [DOI] [PubMed] [Google Scholar]
  • 175. Wilson R, Cahn A, Deans A, et al. : Safety, tolerability and pharmacokinetics (PK) of single and repeat nebulised doses of a novel phosphoinositide 3-kinase δ inhibitor (PI3Kδ), GSK2269557, administered to healthy male subjects in a phase I study. Eur Respir J. 2013;42:729 Reference Source [Google Scholar]
  • 176. Massagué J: TGFβ signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616–30. 10.1038/nrm3434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Kokturk N, Tatlicioglu T, Memis L, et al. : Expression of transforming growth factor beta1 in bronchial biopsies in asthma and COPD. J Asthma. 2003;40(8):887–93. 10.1081/JAS-120023580 [DOI] [PubMed] [Google Scholar]
  • 178. Balzar S, Chu HW, Silkoff P, et al. : Increased TGF-beta2 in severe asthma with eosinophilia. J Allergy Clin Immunol. 2005;115(1):110–7. 10.1016/j.jaci.2004.09.034 [DOI] [PubMed] [Google Scholar]
  • 179. Aschner Y, Downey GP: Transforming Growth Factor-β: Master Regulator of the Respiratory System in Health and Disease. Am J Respir Cell Mol Biol. 2016;54(5):647–55. 10.1165/rcmb.2015-0391TR [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 180. Goldsmith AM, Bentley JK, Zhou L, et al. : Transforming growth factor-beta induces airway smooth muscle hypertrophy. Am J Respir Cell Mol Biol. 2006;34(2):247–54. 10.1165/rcmb.2005-0166OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Xie S, Sukkar MB, Issa R, et al. : Mechanisms of induction of airway smooth muscle hyperplasia by transforming growth factor-beta. Am J Physiol Lung Cell Mol Physiol. 2007;293(1):L245–53. 10.1152/ajplung.00068.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Herbertz S, Sawyer JS, Stauber AJ, et al. : Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther. 2015;9:4479–99. 10.2147/DDDT.S86621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Carlin S, Poronnik P, Cook DI, et al. : An antisense of protein kinase C-zeta inhibits proliferation of human airway smooth muscle cells. Am J Respir Cell Mol Biol. 2000;23(4):555–9. 10.1165/ajrcmb.23.4.4197 [DOI] [PubMed] [Google Scholar]
  • 184. Mochly-Rosen D, Das K, Grimes KV: Protein kinase C, an elusive therapeutic target? Nat Rev Drug Discov. 2012;11(12):937–57. 10.1038/nrd3871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Schaafsma D, Bos IS, Zuidhof AB, et al. : The inhaled Rho kinase inhibitor Y-27632 protects against allergen-induced acute bronchoconstriction, airway hyperresponsiveness, and inflammation. Am J Physiol Lung Cell Mol Physiol. 2008;295(1):L214–9. 10.1152/ajplung.00498.2007 [DOI] [PubMed] [Google Scholar]
  • 186. Lee KY, Ito K, Hayashi R, et al. : NF-kappaB and activator protein 1 response elements and the role of histone modifications in IL-1beta-induced TGF-beta1 gene transcription. J Immunol. 2006;176(1):603–15. 10.4049/jimmunol.176.1.603 [DOI] [PubMed] [Google Scholar]
  • 187. Dienstmann R, Rodon J, Serra V, et al. : Picking the point of inhibition: a comparative review of PI3K/AKT/mTOR pathway inhibitors. Mol Cancer Ther. 2014;13(15):1021–31. 10.1158/1535-7163.MCT-13-0639 [DOI] [PubMed] [Google Scholar]
  • 188. Norman P: Investigational p38 inhibitors for the treatment of chronic obstructive pulmonary disease. Expert Opin Investig Drugs. 2015;24(3):383–92. 10.1517/13543784.2015.1006358 [DOI] [PubMed] [Google Scholar]
  • 189. Johnstone TB, Smith KH, Koziol-White CJ, et al. : PDE8 Is Expressed in Human Airway Smooth Muscle and Selectively Regulates cAMP Signaling by β 2-Adrenergic Receptors and Adenylyl Cyclase 6. Am J Respir Cell Mol Biol. 2018;58(4):530–41. 10.1165/rcmb.2017-0294OC [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 190. Fujitani Y, Trifilieff A: In vivo and in vitro effects of SAR 943, a rapamycin analogue, on airway inflammation and remodeling. Am J Respir Crit Care Med. 2003;167(2):193–8. 10.1164/rccm.200205-455OC [DOI] [PubMed] [Google Scholar]

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES