Skip to main content
Erschienen in: Journal of Translational Medicine 1/2018

Open Access 01.12.2018 | Review

Roles of airway smooth muscle dysfunction in chronic obstructive pulmonary disease

verfasst von: Furong Yan, Hongzhi Gao, Hong Zhao, Madhav Bhatia, Yiming Zeng

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2018

Abstract

The airway smooth muscle (ASM) plays an indispensable role in airway structure and function. Dysfunction in ASM plays a central role in the pathogenesis of chronic obstructive pulmonary disease (COPD) and contributes to alterations of contractility, inflammatory response, immunoreaction, phenotype, quantity, and size of airways. ASM makes a key contribution in COPD by various mechanisms including altered contractility and relaxation induce by [Ca2+]i, cell proliferation and hypertrophy, production and modulation of extracellular cytokines, and release of pro-and-anti-inflammatory mediators. Multiple dysfunctions of ASM contribute to modulating airway responses to stimuli, remodeling, and fibrosis, as well as influence the compliance of lungs. The present review highlights regulatory roles of multiple factors in the development of ASM dysfunction in COPD, aims to understand the regulatory mechanism by which ASM dysfunctions are initiated, and explores the clinical significance of ASM on alterations of airway structure and function in COPD and development of novel therapeutic strategies for COPD.
Abkürzungen
ASM
airway smooth muscle
COPD
chronic obstructive pulmonary disease
[Ca2+]i
intracellular free calcium concentration
CSE
cigarette smoke extract
EBP-α
enhancer-binding protein-α
IFN-γ
interferon-γ
IL
interleukin
IP3
inositol tris-phosphate
MMPs
matrix metalloproteinases
NOX4
nicotinamide adenine dinucleotide phosphate oxidase 4
PLC
phospholipase C
TGF-β
transforming growth factor-β
TNF-α
tumor necrosis factor-α
TRP
transient receptor potential

Background

Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality of patients with lung diseases, characterized by persistent airflow obstruction, with an enhanced inflammatory response in lungs and airways. COPD was the third leading cause of death in China with more than 0.9 million deaths in 2013 [1]. Overwhelming majority of patients had varying degrees of airway remodeling and narrowing. The number of airway smooth muscle (ASM) of COPD patients with the GOLD standard in grade 3 and 4 increased by nearly 50% [2], which was negatively correlated with lung function in COPD patients. A variety of cellular mediators and pathways contribute to the pathogenesis of COPD, evoking a large number of airway and lung dysfunctions [3]. Cigarette smoking is the major cause for COPD, while the pollution exposure such as biomass cooking, heating, and exhaust gas, are also very important factors [4]. In addition, environmental alterations, genetic abnormalities, abnormal lung development and accelerated aging also contribute to the development of COPD [3]. The limitation of airflow as the principal feature of COPD is progressive and not completely reversible [5] and is caused by airway remodeling, loss of small airways, and emphysema [6]. Of those, the airway remodeling and inflammation are considered as the major factors resulting in irreversible airflow limitation [2]. The airway remodeling in COPD includes mucosal hyperproduction, epithelial hyperplasia and metaplasia, increased basement membrane thickness, and connective tissue over-deposition, as well as increased mass of ASM (Fig. 1).
Smooth muscle cells are a crucial component of airway for the contractile function and contributions to the production of inflammatory factors, proteases and growth factors [7]. The altered contractile function and mass of ASM leads to airway inflammation, hyperresponsiveness, and remodeling [8], which are key defining features of COPD. Recent studies demonstrated that changes in other ASM functions like oxidant/antioxidant imbalance, inflammatory secretion and metabolic disorder also contribute significantly to COPD pathophysiology. For example, the nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) expression was up-regulated in ASM during COPD, and strongly related with smoking [9]. ASM in COPD showed imbalance and accumulation in some glycolytic products like lactate, glutamine, fatty acids and amino acids [10]. Moreover, ASM also increases the release of inflammatory mediators to play an important role in many aspects of COPD pathogenesis. The present review aims to briefly overview how the abnormal contractility of ASM contributes to the airway remodeling in COPD, ASM-dominated airway inflammation occurs, ASM is involved in the development of local and systemic immune response to challenges, and the ASM mass and phenotype changes during COPD. Altered ASM plays an important role in the pathogenesis of COPD and contributes to the severity of the disease.

ASM contractility

Airway smooth muscle plays a vital role in the regulation of bronchomotor tone and in the control of the airway caliber, by which COPD patients may have an exaggerated progression of ASM contractility. A variety of regulatory mechanisms are involved in the ASM contraction, leading to the occurrence of airway hyperresponsiveness. Key mechanisms include G-protein coupled receptor-based pathways [11] such as Gq and Gi-dependent signaling, nonselective cation channels especially transient receptor potential channels [12], and store-operated calcium channel [13] (Fig. 2). ASM maintains a balance between the airway hyper-reactivity and bronchodilation when it response to stimuli [14]. The abnormal regulation of contraction and relaxation in ASM leads to the development of disease. For example, the ASM contractile activity altered in lung fibrotic processes, leading to the abnormality of the mechanical properties of airway and contributing to the pathogenesis of COPD.
A number of factors are involved in the dysregulated mechanism of ASM tone in COPD, e.g. ion channels reside in ASM cell plasma membrane, including voltage-gated channels, receptor- and store-depended channels which are consisted of a variety of transient receptor potential (TRP) channels, stretch-activated channels, and Ca2+-dependent K+ channels [15]. Of these, TRPV4, the subtype of TRP channels plays an important role in the development of COPD-related airway activities. The bronchial epithelial cells serve as the primary receptor to be initially stimulated by external challenges and as the barrier to separate ASM from the air. When ASM was exposed to hypotonic airway space liquid and gas, the airway epithelial cell barrier dysfunction occurred in patients with COPD. The activation of TRPV4 in ASM can trigger the ASM contraction, damaged epithelial cells induce the loss of ASM constrictive capability for releasing NO, and the airway can be consistently contracted in COPD patients under hypotonic stimulation [16]. TRPA1, another subtype of TRP channels are a determinant of susceptibility in the development of COPD, mobilize Ca2+ influx in ASM upon activation, and regulate airway contraction via release of neuropeptides, e.g. calcitonin related polypeptide alpha and substance P [17]. G protein-coupled receptors especially Gq-coupled pathways which mainly affects the contractility of ASM could be used as a major drug targets for COPD through elevating Ca2+ levels by activating the phospholipase C-inositol 1,4,5-trisphosphate pathway in ASM. Conversely, Gs-coupled pathway is important for airway dilation, particularly through β2-adrenoceptor action enhancing cAMP [11, 18, 19]. Recent studies suggests that the interactions between taste 2 receptor member 1-bronchoconstrictor and G protein-coupled receptors may contribute to airway contractility and its agonist such as caffeine can influence the actin polymerization of ASM in COPD [20]. Local levels of thromboxane 2 are elevated in the airway of patients with COPD [21]. It indicates that endogenous bronchoconstrictors can be one of the major reasons responsible for the high intension of ASM and the airway hypercontraction in COPD through the coupling between the thromboxane-prostanoid receptor with Gq-coupled pathway and through the PLC/IP3/Ca2+ pathway.

ASM-dominated inflammatory responses

The over-contraction of ASM induced through the activation of multi-kinases also plays a key role in the development of chronic inflammation and airway remodeling in patients with COPD [18]. When the airway is exposed to viruses, bacteria, or other pathogens, a number of airway and lung cells are provoked and over-reacted to resulting in the development of the airway inflammation in COPD. ASM cells are activated by inflammatory signals from airway epithelial cells, leukocytes, and others, and then induce the secondary inflammation by producing inflammatory mediators. A recently study has shown that ASM cells can act as producers of pro-and anti-inflammatory mediators (e.g. cytokines, chemokines, growth factors) to regulate the local immune environment and influence proliferation, migration, and apoptosis of other resident cells [22].
Airway smooth muscle can also act as a source of extracellular matrix proteins, leading to structural changes and remodeling of the airway and alveoli. ASM cells produce monocyte chemoattractant protein-1, C-X-C motif chemokine ligand 10, interleukin (IL)-6, IL-8, IL-1β and eotaxin [2325], through the selective expression of most Toll-like receptors isoforms. In human ASM, the active or inactivated virus could cause the release of IL-5 and IL-1β from human ASM cells, probably through intercellular adhesion molecule-1, since such production is prevented by treatment with the antibody against intercellular adhesion molecule-1 [26]. IL-1β and tumor necrosis factor-alpha (TNFα) can modulate inflammatory responses during the exacerbations of COPD.
In addition, the abnormality of the mitochondrial function occurs within ASM cells from patients with COPD, evidenced by the excessive production of reactive oxygen species. The production of inflammatory responses and mitochondrial dysfunction are the part of cell self-defenses and the secondary inducer of tissue inflammation and injury to activate and drive the innate immune system in the pathogenesis of COPD [27]. β-catenin as a cellular homeostasis regulator can control the cell division and differentiation and take part in the inflammatory processes of chronic airway disease, primarily through the co-action with nuclear factor-κB in human ASM and with the nuclear cofactors CREB-binding protein (CBP) and its homologue p300 [28].

ASM-associated immunoreactions

The tissue forming cells, especially of ASM, play an important role in the immune reaction during the development of COPD by recognizing environmental factors through immune globulin receptors or/and through non-immune systems [29]. ASM may amplify or dampen responses to pathogens by releasing mediators, interacting with recruiting immune cells, and increasing the responsiveness of ASM to stimuli. The immune-regulatory capacity of ASM also includes its response to cytokines such as IL-1β, TNF-α and IFN-γ. During such process, ASM can initiate the over-expression of cell adhesion and co-stimulatory molecules, which attract the recruitment of multiple immune cells into airways and modulate responses to irritants. ASM-produced cytokines can induce the hyperplasia and modulate immune cell function. ASM-origin TNF-α stimulates eosinophilia and neutrophilia in the airway, results in the maturation and differentiation of structural tissues, and leads to the over-expression of cell adhesion molecules and T cell activation [30]. On the other hand, external TNF-α can promote roles of ASM in the infiltration and adhesion of activated immune cells and interaction with and adhesion to inflammatory cells [3133]. ASM can also communicate with airway epithelial cells which can be activated to produce the inflammatory mediators, resulting to the secondary systemic inflammation [34]. ASM can activate immune cells like T lymphocytes and dendritic cells (DCs) by producing thymic stromal lymphopoietin (TSLP), and promote the maturation of T cell phenotype shift [35, 36]. TSLP as a proinflammatory cytokines has been linked to chronic airway diseases. Recent studies have shown that ASM are capable in expressing TSLP in vitro and in vivo, and the enhanced production of it, in turn, creates an inflammatory microenvironment to activating local inflammatory response and aggravating airway remodeling, which is associated with COPD. TSLP-activated DCs in vitro play an important role in promoting the differentiation of Th17 cells with the central memory T cell phenotype [37]. They also induce a unique Th2 cell phenotype that may produces a large amounts of TNF, but little or no IL-10, which different with normal Th2 cells [38].

Change of ASM remodeling

The hypertrophy and hyperplasia of ASM can be negatively correlated with lung function and positively with the severity of airway thickening [2] (Fig. 3). Airrway remodeling is usually accompanied with ASM proliferation. ASM is an important source of matrix metalloproteinases (MMPs). Zinc-dependent proteolytic enzymes produced by ASM play a major regulatory role in matrix turnover, remodeling, and angiogenesis of airways in COPD [6]. Of these, MMPs induce smooth muscle proliferation by release of immobilized growth factors, such as TGF-β, and insulin-like growth factor II. Altered ASM are considered as a distinct metabolic phenotype of COPD. The damaged energy balance and accumulation of the glycolytic products, e.g. lactate, glutamine, fatty acids, and amino acids, increased biosynthesis and redox imbalance in ASM of patients with COPD, and supported ASM proliferation and survival [10]. The cigarette smoke extract (CSE) is the most dangerous environmental factor that promotes the proliferation of ASM in COPD, and it happens in connection with up-regulated expression of calreticulin and down-regulated expression of C/EBPα [39]. Moreover, CSE enhances proliferation of ASM by up-regulation of PCNA and Cyclin E [40]. Calcium influx in ASM by α7 nAChR-PI3K/Akt-TRPC6 pathway contributes to the proliferation process of ASM primarily associated with cigarette smoke [41]. Hypertrophy of ASM is also an important mechanism of airway remodeling. The elevated expression of NOX4 in ASM of small airway may increased the volume of ASM [42]. Up-regulation in orosomucoid-like 3 (ORMDL3) expression can stimulate ASM proliferation, hypertrophy and contractility through enhancing Ca2+ flux induced by increased sarcoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) expression [43]. The ASM remodeling has become an intractable problem in COPD, because of its irreversible changes and multiple mechanism regulation.

Phenotype shift

The ASM phenotype shift is characterized by reversible switching between contractile and proliferative phenotype, of which such shift altered obviously in COPD. It has been demonstrated that ASM cells switch from a contractile to a proliferative phenotype are usually accompanied with reduced KCa1.1 channels and enhanced KCa3.1 channels. Up-regulated KCa3.1 channels caused the increased expression of contractile phenotypic marker proteins and induced ASM migration and proliferation. Blockade of KCa3.1 channels is considered as a therapeutic target in COPD [44]. Long non-coding RNAs (lncRNAs) are associated with ASM phenotype in COPD. The expression of some lncRNAs in healthy ASM cells was increased after stimulation with proliferation inducer [45]. Muscarinic receptors are activated to enhance functional effects of TGF-β1 in ASM, underpin ASM remodeling in COPD [46]. Smoking and local inflammation could directly lead to ASM proliferation through transformation from contractile ASM phenotype into proliferative phenotype in airway remodeling in COPD, which depends on phosphorylation of ERK 1/2 and p38 MAP kinase and downstream mitogenic signaling [47] (Fig. 4).

Regulation of TRP channels in the function of ASM

Adjustment in intracellular calcium concentration ([Ca2+]i) homeostasis directly affects ASM multiple functions. Several members of TRP channels is calcium permeable cation channels and are important for calcium signal transduction in many cell types of respiratory pathophysiology. The TRP channels superfamily can be divided into seven subfamilies based on sequence homology: TRPA (ANKTM1), TRPC (canonical), TRPM (melastatin), TRPML (mucolipidins), TRPN (NompC), TRPV (vanil) and TRPP (polycystin) [48]. Transient receptor potential canonical (TRPC) channels belong to the TRP superfamily and most of them expressed in ASM [49, 50]. They may mediated changes in [Ca2+]i and induced ASM abnormal contraction, proliferation, hypertrophy, as well as secretion of inflammatory mediators [41, 51]. Calcium influx in ASM stimulating by cigarette smoke activates α7 nAChR-PI3K/Akt-TRPC6 pathway and contributes to the proliferation process of ASM [41]. Deletion of TRPC6 has both elevated in airway resistance and arterial pressure. TRPC3 involved in both TNF-α and acetylcholine induced Ca2+ influx in ASM [51] and overexpression of it may contributes to hyperresponsiveness and remodeling in ASM [52]. TRPV4 as an osmolarity sensor is expressed in ASM and may induced Ca2+ influx when stimulated by hypotonic stimulation [53]. In recent years, TRPA1 has also been found to be highly expressed on the membrane of ASM and can be activated by temperature and exogenous irritants, leading to airway inflammation and high reactivity [54]. TRP channels were widely involved in the physiological and pathological changes of various respiratory systems, and have become a new target for the treatment of COPD.

ASM in the development of novel therapeutic approaches for COPD

Airway smooth muscle is the important part in the treatment of airways disorders, for which the targeted therapies are the mainstay in current management of COPD, although controversies are full in the use of those drugs. Potential new targets related to ASM are emerging, despite a long time needed to discover novel classes of COPD drugs. Pathophysiological mechanisms of ASM significantly contribute to the occurrence and progression of COPD. In order to develop therapeutic strategies, key pathological mechanisms and special clinical targets need to be furthermore identified. Current therapies for ASM to treat COPD primarily is bronchodilators through preventing or reversing abnormal shrinkage enhancement of ASM. Most bronchodilators targets in GPCR ligands and work by either promoting relaxant signaling or inhibiting contractile signaling. Airway changes in COPD are also affected by structural modification, which induced by phenotypic shift and increased secretion of cytokines, but currently available therapies for COPD rarely targeted on it. The use of inhibitors of KCa1.1 channels is used to treat COPD by suppressing ASM phenotype shift to proliferative type [45]. Oxidative stress is crucial in the pathogenesis of COPD, but current treatment do not specifically target oxidative stress. Imbalance of oxidative stress responses in ASM have been shown in patients with COPD and contributed to airway inflammatory reaction and influenced ASM functions, probably through mitochondrial dysfunction. Targeting treatment with the mitochondria-targeted antioxidant leads to a reduction of the increased the secretion functions and reduce proliferation of ASM from patients with COPD [27]. Inhibition of NOX4 expression was markedly slowing COPD progress by modulating ROS production in ASM [9]. Sul-121 is a novel compound with anti-oxidative capacity and can effectively inhibit airway inflammation and airway hyperresponsiveness in COPD models by reducing intracellular reactive oxygen species production in ASM [55]. Leptin receptor increased expressed on ASM of COPD and has be found inhibited ASM proliferation, migration and eotaxin production through stimulating ASM to secrete prostaglandin E2 [56]. Retinoic acid can inhibit ASM migration by blocking the PI3K/Akt pathway [57]. There are huge research prospects on the treatment of COPD. The main therapies for ASM to treat COPD have been summarised in Table 1.
Table 1
Therapeutic approaches targeted on ASM
ASM function changes
Type
Action mechanism
References
Contraction and relaxation
Anticholinergics
Inhibit the M2 and M3 subtypes of muscarinic acetylcholine receptors
[59]
β2-adrenoceptor agonists
Stimulation of adenylyl cyclase and subsequent activation of the cAMP-dependent signaling
[60]
Glucocorticoids
Reduce ASM contraction induced by histamine, bradykinin and acetylcholine
[61]
Proliferation and migration
KCa1.1 channels inhibitor
Suppressed phenotype shift to proliferation
[44]
β2-adrenoceptor agonists
Blocked chemoattractants induced ASM migration
[62]
Leukotriene receptor antagonists
Leukotriene B4 markedly induced proliferation of human ASM cells
[63]
Antioxidant, glucocorticoids, ASK1 inhibitor
Inhibited TGF-β-induced ASM cell proliferation
[27, 64, 65]
Leptin
Stimulating ASM to secrete prostaglandin E2
[56]
Secretion
Antioxidant
Inhibited CXCL8 release from ASM
[27]
Reduced intracellular reactive oxygen production in ASM
[55]
Vitamin D
Inhibits secretion of RANTES
[66]

Conclusion

Alterations of ASM morphology and function are considered as the determinant of the airway function and contribute to the severity of COPD. Different factors produced by ASM can induce inflammation, proliferation, apoptosis, and differentiation of ASM per se and also epithelial cells and immune cells. ASM can serve as a receptor cell to be stimulated and activated by systemic inflammation to develop hypertrophy and hyperplasia in COPD. More importantly, ASM can act as an initiator of secondary inflammation to activate other cells. Thus, ASM plays critical and irreplaceable roles in the development of airway inflammation and remodeling during COPD. Current researches on COPD often using stimulant likes cigarette, lipopolysaccharide and protease either individually or jointly to construct models. A low dose of rhinovirus infection in patient with COPD would reproduce the features of COPD exacerbations and be know as a human model of COPD exacerbations [58]. Although these models have provided great help for clinical and scientific research on COPD, there are still some weakness on them for ASM dysfunction researches in COPD because of the complex mechanism. Due to limited understanding of mechanism and lack of appropriate models, the importance of ASM in COPD has not been given sufficient attention. To elucidate the etiology and improve treatment of COPD, further research is needed. As we find out more about the ASM dysfunction in COPD, we will find more options for specific therapeutic method to improve the clinical symptoms and the quality of life in patients with COPD.

Authors’ contributions

All authors contributed to the writing of this review. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Funding

This work was supported by The National Key Research and Development Program of China (2016YFC1304000; 2016YFC1304003).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Zhou M, Wang H, Zhu J, Chen W, Wang L, Liu S, Li Y, Wang L, Liu Y, Yin P, et al. Cause-specific mortality for 240 causes in China during 1990–2013: a systematic subnational analysis for the Global Burden of Disease Study 2013. Lancet. 2016;387:251–72.CrossRef Zhou M, Wang H, Zhu J, Chen W, Wang L, Liu S, Li Y, Wang L, Liu Y, Yin P, et al. Cause-specific mortality for 240 causes in China during 1990–2013: a systematic subnational analysis for the Global Burden of Disease Study 2013. Lancet. 2016;387:251–72.CrossRef
2.
Zurück zum Zitat Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–53.CrossRef Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:2645–53.CrossRef
3.
Zurück zum Zitat Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al. Erratum to “Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report: gOLD executive summary” [Arch Bronconeumol. 2017;53:128-49]. Arch Bronconeumol. 2017;53:411–2.CrossRef Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al. Erratum to “Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report: gOLD executive summary” [Arch Bronconeumol. 2017;53:128-49]. Arch Bronconeumol. 2017;53:411–2.CrossRef
4.
Zurück zum Zitat Gan WQ, FitzGerald JM, Carlsten C, Sadatsafavi M, Brauer M. Associations of ambient air pollution with chronic obstructive pulmonary disease hospitalization and mortality. Am J Respir Crit Care Med. 2013;187:721–7.CrossRef Gan WQ, FitzGerald JM, Carlsten C, Sadatsafavi M, Brauer M. Associations of ambient air pollution with chronic obstructive pulmonary disease hospitalization and mortality. Am J Respir Crit Care Med. 2013;187:721–7.CrossRef
5.
Zurück zum Zitat Woodruff PG, Barr RG, Bleecker E, Christenson SA, Couper D, Curtis JL, Gouskova NA, Hansel NN, Hoffman EA, Kanner RE, et al. Clinical significance of symptoms in smokers with preserved pulmonary function. N Engl J Med. 2016;374:1811–21.CrossRef Woodruff PG, Barr RG, Bleecker E, Christenson SA, Couper D, Curtis JL, Gouskova NA, Hansel NN, Hoffman EA, Kanner RE, et al. Clinical significance of symptoms in smokers with preserved pulmonary function. N Engl J Med. 2016;374:1811–21.CrossRef
6.
Zurück zum Zitat Chen L, Ge Q, Tjin G, Alkhouri H, Deng L, Brandsma CA, Adcock I, Timens W, Postma D, Burgess JK, et al. Effects of cigarette smoke extract on human airway smooth muscle cells in COPD. Eur Respir J. 2014;44:634–46.CrossRef Chen L, Ge Q, Tjin G, Alkhouri H, Deng L, Brandsma CA, Adcock I, Timens W, Postma D, Burgess JK, et al. Effects of cigarette smoke extract on human airway smooth muscle cells in COPD. Eur Respir J. 2014;44:634–46.CrossRef
7.
Zurück zum Zitat Howarth PH, Knox AJ, Amrani Y, Tliba O, Panettieri RA Jr, Johnson M. Synthetic responses in airway smooth muscle. J Allergy Clin Immunol. 2004;114:S32–50.CrossRef Howarth PH, Knox AJ, Amrani Y, Tliba O, Panettieri RA Jr, Johnson M. Synthetic responses in airway smooth muscle. J Allergy Clin Immunol. 2004;114:S32–50.CrossRef
8.
Zurück zum Zitat Hirota N, Martin JG. Mechanisms of airway remodeling. Chest. 2013;144:1026–32.CrossRef Hirota N, Martin JG. Mechanisms of airway remodeling. Chest. 2013;144:1026–32.CrossRef
9.
Zurück zum Zitat Hollins F, Sutcliffe A, Gomez E, Berair R, Russell R, Szyndralewiez C, Saunders R, Brightling C. Airway smooth muscle NOX4 is upregulated and modulates ROS generation in COPD. Respir Res. 2016;17:84.CrossRef Hollins F, Sutcliffe A, Gomez E, Berair R, Russell R, Szyndralewiez C, Saunders R, Brightling C. Airway smooth muscle NOX4 is upregulated and modulates ROS generation in COPD. Respir Res. 2016;17:84.CrossRef
11.
Zurück zum Zitat Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res. 2003;4:2.CrossRef Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res. 2003;4:2.CrossRef
12.
Zurück zum Zitat Gosling M, Poll C, Li S. TRP channels in airway smooth muscle as therapeutic targets. Naunyn Schmiedebergs Arch Pharmacol. 2005;371:277–84.CrossRef Gosling M, Poll C, Li S. TRP channels in airway smooth muscle as therapeutic targets. Naunyn Schmiedebergs Arch Pharmacol. 2005;371:277–84.CrossRef
13.
Zurück zum Zitat Ay B, Prakash YS, Pabelick CM, Sieck GC. Store-operated Ca2+ entry in porcine airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2004;286:L909–17.CrossRef Ay B, Prakash YS, Pabelick CM, Sieck GC. Store-operated Ca2+ entry in porcine airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2004;286:L909–17.CrossRef
14.
Zurück zum Zitat Prakash YS. Airway smooth muscle in airway reactivity and remodeling: what have we learned? Am J Physiol Lung Cell Mol Physiol. 2013;305:L912–33.CrossRef Prakash YS. Airway smooth muscle in airway reactivity and remodeling: what have we learned? Am J Physiol Lung Cell Mol Physiol. 2013;305:L912–33.CrossRef
15.
Zurück zum Zitat Perez-Zoghbi JF, Karner C, Ito S, Shepherd M, Alrashdan Y, Sanderson MJ. Ion channel regulation of intracellular calcium and airway smooth muscle function. Pulm Pharmacol Ther. 2009;22:388–97.CrossRef Perez-Zoghbi JF, Karner C, Ito S, Shepherd M, Alrashdan Y, Sanderson MJ. Ion channel regulation of intracellular calcium and airway smooth muscle function. Pulm Pharmacol Ther. 2009;22:388–97.CrossRef
16.
Zurück zum Zitat Zhu G, Investigators I, Gulsvik A, Bakke P, Ghatta S, Anderson W, Lomas DA, Silverman EK, Pillai SG. Association of TRPV4 gene polymorphisms with chronic obstructive pulmonary disease. Hum Mol Genet. 2009;18:2053–62.CrossRef Zhu G, Investigators I, Gulsvik A, Bakke P, Ghatta S, Anderson W, Lomas DA, Silverman EK, Pillai SG. Association of TRPV4 gene polymorphisms with chronic obstructive pulmonary disease. Hum Mol Genet. 2009;18:2053–62.CrossRef
17.
Zurück zum Zitat Jha A, Sharma P, Anaparti V, Ryu MH, Halayko AJ. A role for transient receptor potential ankyrin 1 cation channel (TRPA1) in airway hyper-responsiveness? Can J Physiol Pharmacol. 2015;93:171–6.CrossRef Jha A, Sharma P, Anaparti V, Ryu MH, Halayko AJ. A role for transient receptor potential ankyrin 1 cation channel (TRPA1) in airway hyper-responsiveness? Can J Physiol Pharmacol. 2015;93:171–6.CrossRef
18.
Zurück zum Zitat Penn RB, Bond RA, Walker JK. GPCRs and arrestins in airways: implications for asthma. Handb Exp Pharmacol. 2014;219:387–403.CrossRef Penn RB, Bond RA, Walker JK. GPCRs and arrestins in airways: implications for asthma. Handb Exp Pharmacol. 2014;219:387–403.CrossRef
19.
Zurück zum Zitat Pera T, Penn RB. Crosstalk between beta-2-adrenoceptor and muscarinic acetylcholine receptors in the airway. Curr Opin Pharmacol. 2014;16:72–81.CrossRef Pera T, Penn RB. Crosstalk between beta-2-adrenoceptor and muscarinic acetylcholine receptors in the airway. Curr Opin Pharmacol. 2014;16:72–81.CrossRef
20.
Zurück zum Zitat Tazzeo T, Bates G, Roman HN, Lauzon AM, Khasnis MD, Eto M, Janssen LJ. Caffeine relaxes smooth muscle through actin depolymerization. Am J Physiol Lung Cell Mol Physiol. 2012;303:L334–42.CrossRef Tazzeo T, Bates G, Roman HN, Lauzon AM, Khasnis MD, Eto M, Janssen LJ. Caffeine relaxes smooth muscle through actin depolymerization. Am J Physiol Lung Cell Mol Physiol. 2012;303:L334–42.CrossRef
21.
Zurück zum Zitat Rolin S, Masereel B, Dogne JM. Prostanoids as pharmacological targets in COPD and asthma. Eur J Pharmacol. 2006;533:89–100.CrossRef Rolin S, Masereel B, Dogne JM. Prostanoids as pharmacological targets in COPD and asthma. Eur J Pharmacol. 2006;533:89–100.CrossRef
22.
Zurück zum Zitat Barnes PJ. Kinases as novel therapeutic targets in asthma and chronic obstructive pulmonary disease. Pharmacol Rev. 2016;68:788–815.CrossRef Barnes PJ. Kinases as novel therapeutic targets in asthma and chronic obstructive pulmonary disease. Pharmacol Rev. 2016;68:788–815.CrossRef
23.
Zurück zum Zitat Sukkar MB, Xie S, Khorasani NM, Kon OM, Stanbridge R, Issa R, Chung KF. Toll-like receptor 2, 3, and 4 expression and function in human airway smooth muscle. J Allergy Clin Immunol. 2006;118:641–8.CrossRef Sukkar MB, Xie S, Khorasani NM, Kon OM, Stanbridge R, Issa R, Chung KF. Toll-like receptor 2, 3, and 4 expression and function in human airway smooth muscle. J Allergy Clin Immunol. 2006;118:641–8.CrossRef
24.
Zurück zum Zitat Chiou YL, Lin CY. Der p2 activates airway smooth muscle cells in a TLR2/MyD88-dependent manner to induce an inflammatory response. J Cell Physiol. 2009;220:311–8.CrossRef Chiou YL, Lin CY. Der p2 activates airway smooth muscle cells in a TLR2/MyD88-dependent manner to induce an inflammatory response. J Cell Physiol. 2009;220:311–8.CrossRef
25.
Zurück zum Zitat Cooper PR, Lamb R, Day ND, Branigan PJ, Kajekar R, San Mateo L, Hornby PJ, Panettieri RA Jr. TLR3 activation stimulates cytokine secretion without altering agonist-induced human small airway contraction or relaxation. Am J Physiol Lung Cell Mol Physiol. 2009;297:L530–7.CrossRef Cooper PR, Lamb R, Day ND, Branigan PJ, Kajekar R, San Mateo L, Hornby PJ, Panettieri RA Jr. TLR3 activation stimulates cytokine secretion without altering agonist-induced human small airway contraction or relaxation. Am J Physiol Lung Cell Mol Physiol. 2009;297:L530–7.CrossRef
26.
Zurück zum Zitat Grunstein MM, Hakonarson H, Whelan R, Yu Z, Grunstein JS, Chuang S. Rhinovirus elicits proasthmatic changes in airway responsiveness independently of viral infection. J Allergy Clin Immunol. 2001;108:997–1004.CrossRef Grunstein MM, Hakonarson H, Whelan R, Yu Z, Grunstein JS, Chuang S. Rhinovirus elicits proasthmatic changes in airway responsiveness independently of viral infection. J Allergy Clin Immunol. 2001;108:997–1004.CrossRef
27.
Zurück zum Zitat Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE, Bao W, Pavlidis S, Barnes PJ, Kanerva J, et al. Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2015;136:769–80.CrossRef Wiegman CH, Michaeloudes C, Haji G, Narang P, Clarke CJ, Russell KE, Bao W, Pavlidis S, Barnes PJ, Kanerva J, et al. Oxidative stress-induced mitochondrial dysfunction drives inflammation and airway smooth muscle remodeling in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2015;136:769–80.CrossRef
28.
Zurück zum Zitat Koopmans T, Eilers R, Menzen M, Halayko A, Gosens R. Beta-catenin directs nuclear factor-kappaB p65 output via CREB-binding protein/p300 in human airway smooth muscle. Front Immunol. 2017;8:1086.CrossRef Koopmans T, Eilers R, Menzen M, Halayko A, Gosens R. Beta-catenin directs nuclear factor-kappaB p65 output via CREB-binding protein/p300 in human airway smooth muscle. Front Immunol. 2017;8:1086.CrossRef
29.
Zurück zum Zitat Roth M, Tamm M. Airway smooth muscle cells respond directly to inhaled environmental factors. Swiss Med Wkly. 2010;140:w13066.PubMed Roth M, Tamm M. Airway smooth muscle cells respond directly to inhaled environmental factors. Swiss Med Wkly. 2010;140:w13066.PubMed
30.
Zurück zum Zitat Matera MG, Calzetta L, Cazzola M. TNF-alpha inhibitors in asthma and COPD: we must not throw the baby out with the bath water. Pulm Pharmacol Ther. 2010;23:121–8.CrossRef Matera MG, Calzetta L, Cazzola M. TNF-alpha inhibitors in asthma and COPD: we must not throw the baby out with the bath water. Pulm Pharmacol Ther. 2010;23:121–8.CrossRef
31.
Zurück zum Zitat Hughes JM, Arthur CA, Baracho S, Carlin SM, Hawker KM, Johnson PR, Armour CL. Human eosinophil-airway smooth muscle cell interactions. Mediators Inflamm. 2000;9:93–9.CrossRef Hughes JM, Arthur CA, Baracho S, Carlin SM, Hawker KM, Johnson PR, Armour CL. Human eosinophil-airway smooth muscle cell interactions. Mediators Inflamm. 2000;9:93–9.CrossRef
32.
Zurück zum Zitat Lee CW, Lin CC, Luo SF, Lee HC, Lee IT, Aird WC, Hwang TL, Yang CM. Tumor necrosis factor-alpha enhances neutrophil adhesiveness: induction of vascular cell adhesion molecule-1 via activation of Akt and CaM kinase II and modifications of histone acetyltransferase and histone deacetylase 4 in human tracheal smooth muscle cells. Mol Pharmacol. 2008;73:1454–64.CrossRef Lee CW, Lin CC, Luo SF, Lee HC, Lee IT, Aird WC, Hwang TL, Yang CM. Tumor necrosis factor-alpha enhances neutrophil adhesiveness: induction of vascular cell adhesion molecule-1 via activation of Akt and CaM kinase II and modifications of histone acetyltransferase and histone deacetylase 4 in human tracheal smooth muscle cells. Mol Pharmacol. 2008;73:1454–64.CrossRef
33.
Zurück zum Zitat Lee CW, Lin WN, Lin CC, Luo SF, Wang JS, Pouyssegur J, Yang CM. Transcriptional regulation of VCAM-1 expression by tumor necrosis factor-alpha in human tracheal smooth muscle cells: involvement of MAPKs, NF-kappaB, p300, and histone acetylation. J Cell Physiol. 2006;207:174–86.CrossRef Lee CW, Lin WN, Lin CC, Luo SF, Wang JS, Pouyssegur J, Yang CM. Transcriptional regulation of VCAM-1 expression by tumor necrosis factor-alpha in human tracheal smooth muscle cells: involvement of MAPKs, NF-kappaB, p300, and histone acetylation. J Cell Physiol. 2006;207:174–86.CrossRef
34.
Zurück zum Zitat Damera G, Zhao H, Wang M, Smith M, Kirby C, Jester WF, Lawson JA, Panettieri RA Jr. Ozone modulates IL-6 secretion in human airway epithelial and smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2009;296:L674–83.CrossRef Damera G, Zhao H, Wang M, Smith M, Kirby C, Jester WF, Lawson JA, Panettieri RA Jr. Ozone modulates IL-6 secretion in human airway epithelial and smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2009;296:L674–83.CrossRef
35.
Zurück zum Zitat Redhu NS, Saleh A, Halayko AJ, Ali AS, Gounni AS. Essential role of NF-kappaB and AP-1 transcription factors in TNF-alpha-induced TSLP expression in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2011;300:L479–85.CrossRef Redhu NS, Saleh A, Halayko AJ, Ali AS, Gounni AS. Essential role of NF-kappaB and AP-1 transcription factors in TNF-alpha-induced TSLP expression in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2011;300:L479–85.CrossRef
36.
Zurück zum Zitat Shan L, Redhu NS, Saleh A, Halayko AJ, Chakir J, Gounni AS. Thymic stromal lymphopoietin receptor-mediated IL-6 and CC/CXC chemokines expression in human airway smooth muscle cells: role of MAPKs (ERK1/2, p38, and JNK) and STAT3 pathways. J Immunol. 2010;184:7134–43.CrossRef Shan L, Redhu NS, Saleh A, Halayko AJ, Chakir J, Gounni AS. Thymic stromal lymphopoietin receptor-mediated IL-6 and CC/CXC chemokines expression in human airway smooth muscle cells: role of MAPKs (ERK1/2, p38, and JNK) and STAT3 pathways. J Immunol. 2010;184:7134–43.CrossRef
37.
Zurück zum Zitat 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.CrossRef 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.CrossRef
38.
Zurück zum Zitat Liu YJ, Soumelis V, Watanabe N, Ito T, Wang YH, Malefyt Rde W, Omori M, Zhou B, Ziegler SF. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007;25:193–219.CrossRef Liu YJ, Soumelis V, Watanabe N, Ito T, Wang YH, Malefyt Rde W, Omori M, Zhou B, Ziegler SF. TSLP: an epithelial cell cytokine that regulates T cell differentiation by conditioning dendritic cell maturation. Annu Rev Immunol. 2007;25:193–219.CrossRef
39.
Zurück zum Zitat Guan P, Cai W, Yu H, Wu Z, Li W, Wu J, Chen J, Feng G. Cigarette smoke extract promotes proliferation of airway smooth muscle cells through suppressing C/EBP-alpha expression. Exp Ther Med. 2017;13:1408–14.CrossRef Guan P, Cai W, Yu H, Wu Z, Li W, Wu J, Chen J, Feng G. Cigarette smoke extract promotes proliferation of airway smooth muscle cells through suppressing C/EBP-alpha expression. Exp Ther Med. 2017;13:1408–14.CrossRef
40.
Zurück zum Zitat Wylam ME, Sathish V, VanOosten SK, Freeman M, Burkholder D, Thompson MA, Pabelick CM, Prakash YS. Mechanisms of cigarette smoke effects on human airway smooth muscle. PLoS ONE. 2015;10:e0128778.CrossRef Wylam ME, Sathish V, VanOosten SK, Freeman M, Burkholder D, Thompson MA, Pabelick CM, Prakash YS. Mechanisms of cigarette smoke effects on human airway smooth muscle. PLoS ONE. 2015;10:e0128778.CrossRef
41.
Zurück zum Zitat Hong W, Peng G, Hao B, Liao B, Zhao Z, Zhou Y, Peng F, Ye X, Huang L, Zheng M, et al. Nicotine-induced airway smooth muscle cell proliferation involves TRPC6-dependent calcium influx via alpha7 nAChR. Cell Physiol Biochem. 2017;43:986–1002.CrossRef Hong W, Peng G, Hao B, Liao B, Zhao Z, Zhou Y, Peng F, Ye X, Huang L, Zheng M, et al. Nicotine-induced airway smooth muscle cell proliferation involves TRPC6-dependent calcium influx via alpha7 nAChR. Cell Physiol Biochem. 2017;43:986–1002.CrossRef
42.
Zurück zum Zitat Liu X, Hao B, Ma A, He J, Liu X, Chen J. The expression of NOX4 in smooth muscles of small airway correlates with the disease severity of COPD. Biomed Res Int. 2016;2016:2891810.PubMedPubMedCentral Liu X, Hao B, Ma A, He J, Liu X, Chen J. The expression of NOX4 in smooth muscles of small airway correlates with the disease severity of COPD. Biomed Res Int. 2016;2016:2891810.PubMedPubMedCentral
43.
Zurück zum Zitat Chen J, Miller M, Unno H, Rosenthal P, Sanderson MJ, Broide DH. Orosomucoid-like 3 (ORMDL3) upregulates airway smooth muscle proliferation, contraction, and Ca(2+) oscillations in asthma. J Allergy Clin Immunol. 2018;142(207–218):e206. Chen J, Miller M, Unno H, Rosenthal P, Sanderson MJ, Broide DH. Orosomucoid-like 3 (ORMDL3) upregulates airway smooth muscle proliferation, contraction, and Ca(2+) oscillations in asthma. J Allergy Clin Immunol. 2018;142(207–218):e206.
44.
Zurück zum Zitat Yu ZH, Wang YX, Song Y, Lu HZ, Hou LN, Cui YY, Chen HZ. Up-regulation of KCa3.1 promotes human airway smooth muscle cell phenotypic modulation. Pharmacol Res. 2013;77:30–8.CrossRef Yu ZH, Wang YX, Song Y, Lu HZ, Hou LN, Cui YY, Chen HZ. Up-regulation of KCa3.1 promotes human airway smooth muscle cell phenotypic modulation. Pharmacol Res. 2013;77:30–8.CrossRef
45.
Zurück zum Zitat Perry MM, Tsitsiou E, Austin PJ, Lindsay MA, Gibeon DS, Adcock IM, Chung KF. Role of non-coding RNAs in maintaining primary airway smooth muscle cells. Respir Res. 2014;15:58.CrossRef Perry MM, Tsitsiou E, Austin PJ, Lindsay MA, Gibeon DS, Adcock IM, Chung KF. Role of non-coding RNAs in maintaining primary airway smooth muscle cells. Respir Res. 2014;15:58.CrossRef
46.
Zurück zum Zitat Oenema TA, Smit M, Smedinga L, Racke K, Halayko AJ, Meurs H, Gosens R. Muscarinic receptor stimulation augments TGF-beta1-induced contractile protein expression by airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2012;303:L589–97.CrossRef Oenema TA, Smit M, Smedinga L, Racke K, Halayko AJ, Meurs H, Gosens R. Muscarinic receptor stimulation augments TGF-beta1-induced contractile protein expression by airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2012;303:L589–97.CrossRef
47.
Zurück zum Zitat Pera T, Gosens R, Lesterhuis AH, Sami R, van der Toorn M, Zaagsma J, Meurs H. Cigarette smoke and lipopolysaccharide induce a proliferative airway smooth muscle phenotype. Respir Res. 2010;11:48.CrossRef Pera T, Gosens R, Lesterhuis AH, Sami R, van der Toorn M, Zaagsma J, Meurs H. Cigarette smoke and lipopolysaccharide induce a proliferative airway smooth muscle phenotype. Respir Res. 2010;11:48.CrossRef
48.
Zurück zum Zitat Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol. 2011;12:218.CrossRef Nilius B, Owsianik G. The transient receptor potential family of ion channels. Genome Biol. 2011;12:218.CrossRef
49.
Zurück zum Zitat Zhang X, Zhao Z, Ma L, Guo Y, Li X, Zhao L, Tian C, Tang X, Cheng D, Chen Z, Zhang L. The effects of transient receptor potential channel (TRPC) on airway smooth muscle cell isolated from asthma model mice. J Cell Biochem. 2018;119:6033–44.CrossRef Zhang X, Zhao Z, Ma L, Guo Y, Li X, Zhao L, Tian C, Tang X, Cheng D, Chen Z, Zhang L. The effects of transient receptor potential channel (TRPC) on airway smooth muscle cell isolated from asthma model mice. J Cell Biochem. 2018;119:6033–44.CrossRef
50.
Zurück zum Zitat Corteling RL, Li S, Giddings J, Westwick J, Poll C, Hall IP. Expression of transient receptor potential C6 and related transient receptor potential family members in human airway smooth muscle and lung tissue. Am J Respir Cell Mol Biol. 2004;30:145–54.CrossRef Corteling RL, Li S, Giddings J, Westwick J, Poll C, Hall IP. Expression of transient receptor potential C6 and related transient receptor potential family members in human airway smooth muscle and lung tissue. Am J Respir Cell Mol Biol. 2004;30:145–54.CrossRef
51.
Zurück zum Zitat White TA, Xue A, Chini EN, Thompson M, Sieck GC, Wylam ME. Role of transient receptor potential C3 in TNF-alpha-enhanced calcium influx in human airway myocytes. Am J Respir Cell Mol Biol. 2006;35:243–51.CrossRef White TA, Xue A, Chini EN, Thompson M, Sieck GC, Wylam ME. Role of transient receptor potential C3 in TNF-alpha-enhanced calcium influx in human airway myocytes. Am J Respir Cell Mol Biol. 2006;35:243–51.CrossRef
52.
Zurück zum Zitat Xu BM, Zhang JH, Wang JL, Xiao JH. TRPC3 overexpression and intervention in airway smooth muscle of ovalbumin-induced hyperresponsiveness and remodeling. Cell Biol Int. 2018;42:1021–9.CrossRef Xu BM, Zhang JH, Wang JL, Xiao JH. TRPC3 overexpression and intervention in airway smooth muscle of ovalbumin-induced hyperresponsiveness and remodeling. Cell Biol Int. 2018;42:1021–9.CrossRef
53.
Zurück zum Zitat Jia Y, Wang X, Varty L, Rizzo CA, Yang R, Correll CC, Phelps PT, Egan RW, Hey JA. Functional TRPV4 channels are expressed in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2004;287:L272–8.CrossRef Jia Y, Wang X, Varty L, Rizzo CA, Yang R, Correll CC, Phelps PT, Egan RW, Hey JA. Functional TRPV4 channels are expressed in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2004;287:L272–8.CrossRef
54.
Zurück zum Zitat Nassini R, Pedretti P, Moretto N, Fusi C, Carnini C, Facchinetti F, Viscomi AR, Pisano AR, Stokesberry S, Brunmark C, et al. Transient receptor potential ankyrin 1 channel localized to non-neuronal airway cells promotes non-neurogenic inflammation. PLoS ONE. 2012;7:e42454.CrossRef Nassini R, Pedretti P, Moretto N, Fusi C, Carnini C, Facchinetti F, Viscomi AR, Pisano AR, Stokesberry S, Brunmark C, et al. Transient receptor potential ankyrin 1 channel localized to non-neuronal airway cells promotes non-neurogenic inflammation. PLoS ONE. 2012;7:e42454.CrossRef
55.
Zurück zum Zitat Han B, Poppinga WJ, Zuo H, Zuidhof AB, Bos IS, Smit M, Vogelaar P, Krenning G, Henning RH, Maarsingh H, et al. The novel compound Sul-121 inhibits airway inflammation and hyperresponsiveness in experimental models of chronic obstructive pulmonary disease. Sci Rep. 2016;6:26928.CrossRef Han B, Poppinga WJ, Zuo H, Zuidhof AB, Bos IS, Smit M, Vogelaar P, Krenning G, Henning RH, Maarsingh H, et al. The novel compound Sul-121 inhibits airway inflammation and hyperresponsiveness in experimental models of chronic obstructive pulmonary disease. Sci Rep. 2016;6:26928.CrossRef
56.
Zurück zum Zitat Nair P, Radford K, Fanat A, Janssen LJ, Peters-Golden M, Cox PG. The effects of leptin on airway smooth muscle responses. Am J Respir Cell Mol Biol. 2008;39:475–81.CrossRef Nair P, Radford K, Fanat A, Janssen LJ, Peters-Golden M, Cox PG. The effects of leptin on airway smooth muscle responses. Am J Respir Cell Mol Biol. 2008;39:475–81.CrossRef
57.
Zurück zum Zitat Day RM, Lee YH, Park AM, Suzuki YJ. Retinoic acid inhibits airway smooth muscle cell migration. Am J Respir Cell Mol Biol. 2006;34:695–703.CrossRef Day RM, Lee YH, Park AM, Suzuki YJ. Retinoic acid inhibits airway smooth muscle cell migration. Am J Respir Cell Mol Biol. 2006;34:695–703.CrossRef
58.
Zurück zum Zitat Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, Aniscenko J, Laza-Stanca V, Edwards MR, Slater L, et al. Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011;183:734–42.CrossRef Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, Aniscenko J, Laza-Stanca V, Edwards MR, Slater L, et al. Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011;183:734–42.CrossRef
59.
Zurück zum Zitat Hansel TT, Barnes PJ. Tiotropium bromide: a novel once-daily anticholinergic bronchodilator for the treatment of COPD. Drugs Today (Barc). 2002;38:585–600.CrossRef Hansel TT, Barnes PJ. Tiotropium bromide: a novel once-daily anticholinergic bronchodilator for the treatment of COPD. Drugs Today (Barc). 2002;38:585–600.CrossRef
60.
Zurück zum Zitat Giembycz MA, Newton R. Beyond the dogma: novel beta2-adrenoceptor signalling in the airways. Eur Respir J. 2006;27:1286–306.CrossRef Giembycz MA, Newton R. Beyond the dogma: novel beta2-adrenoceptor signalling in the airways. Eur Respir J. 2006;27:1286–306.CrossRef
61.
Zurück zum Zitat Sun HW, Miao CY, Liu L, Zhou J, Su DF, Wang YX, Jiang CL. Rapid inhibitory effect of glucocorticoids on airway smooth muscle contractions in guinea pigs. Steroids. 2006;71:154–9.CrossRef Sun HW, Miao CY, Liu L, Zhou J, Su DF, Wang YX, Jiang CL. Rapid inhibitory effect of glucocorticoids on airway smooth muscle contractions in guinea pigs. Steroids. 2006;71:154–9.CrossRef
62.
Zurück zum Zitat Carlin SM, Roth M, Black JL. Urokinase potentiates PDGF-induced chemotaxis of human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2003;284:L1020–6.CrossRef Carlin SM, Roth M, Black JL. Urokinase potentiates PDGF-induced chemotaxis of human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2003;284:L1020–6.CrossRef
63.
Zurück zum Zitat 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):e51–3. 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):e51–3.
64.
Zurück zum Zitat Xie S, Sukkar MB, Issa R, Khorasani NM, Chung KF. Mechanisms of induction of airway smooth muscle hyperplasia by transforming growth factor-beta. Am J Physiol Lung Cell Mol Physiol. 2007;293:L245–53.CrossRef Xie S, Sukkar MB, Issa R, Khorasani NM, Chung KF. Mechanisms of induction of airway smooth muscle hyperplasia by transforming growth factor-beta. Am J Physiol Lung Cell Mol Physiol. 2007;293:L245–53.CrossRef
65.
Zurück zum Zitat Eapen MS, Kota A, Vindin H, McAlinden KD, Xenaki D, Oliver BG, Deshpande DA, Sohal SS, Sharma P. Apoptosis signal-regulating kinase 1 (ASK1) inhibition attenuates human airway smooth muscle growth and migration in chronic obstructive pulmonary disease (COPD). Clin Sci (Lond). 2018. https://doi.org/10.1042/CS20180398.CrossRef Eapen MS, Kota A, Vindin H, McAlinden KD, Xenaki D, Oliver BG, Deshpande DA, Sohal SS, Sharma P. Apoptosis signal-regulating kinase 1 (ASK1) inhibition attenuates human airway smooth muscle growth and migration in chronic obstructive pulmonary disease (COPD). Clin Sci (Lond). 2018. https://​doi.​org/​10.​1042/​CS20180398.CrossRef
66.
Zurück zum Zitat Banerjee A, Damera G, Bhandare R, Gu S, Lopez-Boado Y, Panettieri R Jr, Tliba O. Vitamin D and glucocorticoids differentially modulate chemokine expression in human airway smooth muscle cells. Br J Pharmacol. 2008;155:84–92.CrossRef Banerjee A, Damera G, Bhandare R, Gu S, Lopez-Boado Y, Panettieri R Jr, Tliba O. Vitamin D and glucocorticoids differentially modulate chemokine expression in human airway smooth muscle cells. Br J Pharmacol. 2008;155:84–92.CrossRef
Metadaten
Titel
Roles of airway smooth muscle dysfunction in chronic obstructive pulmonary disease
verfasst von
Furong Yan
Hongzhi Gao
Hong Zhao
Madhav Bhatia
Yiming Zeng
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2018
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1635-z

Weitere Artikel der Ausgabe 1/2018

Journal of Translational Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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