Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2011 May;163(1):68–80. doi: 10.1111/j.1476-5381.2010.01165.x

Does airway smooth muscle express an inflammatory phenotype in asthma?

Gautam Damera 1, Reynold A Panettieri Jr 1,2
PMCID: PMC3085869  PMID: 21175578

Abstract

In addition to hyperresponsiveness in asthma, airway smooth muscle (ASM) also manifests an inflammatory phenotype characterized by augmented expression of mediators that enhance inflammation, contribute to tissue remodelling and augment leucocyte trafficking and activity. Our present review summarizes contemporary understanding of ASM-derived mediators and their paracrine and autocrine actions in airway diseases.

LINKED ARTICLES

This article is part of a themed issue on Respiratory Pharmacology. To view the other articles in this issue visit http://dx.doi.org/10.1111/bph.2011.163.issue-1

Keywords: airway inflammation, airway obstruction, airway remodelling, COPD, cytokines, non-canonical pathways

Introduction

Asthma, a heterogeneous disease, manifests by chronic airway inflammation associated with infiltration of eosinophils and T-helper 2 (Th2) lymphocytes, airway hyperresponsiveness (AHR), remodelling, mucus hypersecretion and, in part, reversible airway obstruction. While imbalance in T-helper 1 (Th1) and Th2 inflammatory responses may underlie asthma pathogenesis, evidence suggests that AHR persists in the absence of airway inflammation implying that structural cells likely play a role in development of the disease. In this context, studies have identified airway smooth muscle (ASM) as a pivotal tissue promoting the asthma diathesis. Similar to other airway structural tissues, ASM expresses a broad class of chemokines/cytokines, eicosanoids and prostaglandins that modulate airway-health and -disease (Table 1). Although much work has been performed in vitro, contemporary studies have identified ASM to express aforementioned molecules in vivo. Previous reviews by us and others have elaborated on the immunomodulatory faculties of ASM (Hershenson et al., 2008; Dekkers et al., 2009; Tliba and Panettieri, 2009). Our focus here encapsulates the evolving role of ASM as a modulator and effector population with an overarching role in airway-inflammation, -remodelling and -irreversible obstruction.

Table 1.

Inflammatory mediators and airway smooth muscle (ASM) function

Mediator Synthesized by ASM Autocrine effects on ASM Modulates ASM function
IL-1 + + +++
IL-4/13 +/− +/− +++
IL-6 +++ +/−
IL-8 +++
IL-9 ++
PGE2 +++ +++ +++
LTD4 +/− +/− +++

+++, substantial effect; +, modest effect; −, little or no effect.

Modulation of inflammation via paracrine and autocrine pathways

Tumour necrosis factor-α (TNF-α) as a key cytokine mediates pathogenic mechanisms of a number of chronic inflammatory diseases, including rheumatoid arthritis, Crohn's disease, ankylosing spondylitis, psoriasis and asthma (Russo and Polosa, 2005). In addition to cross-sectional studies showing enhanced prevalence of TNF-α in the airway of asthmatics, the development of AHR post-TNF-α inhalation in normal individuals reiterates its singular relevance towards airway dysfunction in asthma (Bradding et al., 1994; Thomas and Heywood, 2002). Similarly, murine-isolated tracheal rings incubated with TNF-α show exaggerated responses to contractile agonists, including carbachol, bradykinin and serotonin (Brightling et al., 2008). In airways of individuals with asthma, mast cells are prominent contributors of TNF-α secretions, and ASM bundle-resident mast cell numbers positively correlate with the degree of AHR and with the bronchoconstrictor response to a deep inspiration, leading some to speculate that their juxtaposition facilitates rapid mast cell-derived TNF-α activation and myocyte shortening (Carroll et al., 2002; Slats et al., 2007; Brightling et al., 2008). Overlaying its pathological effects on airway mechanics, localized TNF-α abundance has other disease-specific connotations in asthma. Several in vitro studies have elucidated an ability of TNF-α to elicit airway neutrophilia and eosinophilia, mucus hypersecretion, T-cell activation, expression of cell adhesion molecules (CAMs), and maturation and differentiation of structural tissue (Matera et al., 2010). As demonstrated extensively, TNF-α effects on ASM are elaborate, and the following text elucidates its biological role in the context of inflammation, infection and remodelling.

Together with TNF-α, interleukin (IL)-1β identifies as a key cytokine that modulates inflammatory cascades during exacerbations of asthma and chronic obstructive pulmonary disease (COPD) (Howarth et al., 2005; Hackett et al., 2008). The pleiotropic nature of IL-1β results in the activation of a wide range of cells such as mast cells, T- and B-lymphocytes, epithelial cells and ASM. As a prominent mediator of asthma pathophysiology, IL-1β expression is increased in the airways of asthmatic individuals, and IL-1 receptor antagonist (IL-1Ra) administration reduces AHR induced by allergens in mice (Barnes, 2008). In ASM cells of human origin, IL-1β treatment enhances production of numerous inflammatory mediators including prostaglandin-E2 (PGE2), eotaxin, regulated upon activation normal T-cell expressed and secreted (RANTES), granulocyte-macrophage colony-stimulating factor (GM-CSF), growth-related oncogene-α, epithelial neutrophil-activating protein-78, nerve growth factor, monocyte chemotactic protein (MCP)-1,2,3, IL-8 and IL-11 (Pype et al., 1999; Freund et al., 2002; Jarai et al., 2004; Issa et al., 2006; Tliba et al., 2008; Clarke et al., 2009a). Besides contributing to pro-inflammatory milieu, IL-1β amplifies ASM responses by modulating receptor expression as with IL-5R and protease-activated receptor-2 expression in human myocyte cultures, and ASM-resident IL-1RI, TNF-RI,RII and 5-HT2A receptors in ex vivo cultures of murine tracheal rings, further amplifying its effects on ASM function (Hedges et al., 2000; Cardell et al., 2008). Numerous studies in ASM have identified COX-2 enzyme inducing pivotal IL-1-mediated responses. While IL-1β enhances bradykinin-induced responsiveness in tracheal ring segments, desensitization of the H1R via induction of PGE2 renders bronchial rings less responsive to histamine (Pype et al., 2001). Further, IL-1β synergizes with TNF-α to promote β2AR hyporesponsiveness, a phenomenon that is ablated by selective COX-2 inhibition (Shore et al., 1997). Evidence suggests that glucocorticoids render IL-1β and TNF-α mitogenic to ASM, via suppression of COX-2-dependent protein kinase A (PKA) activity (Misior et al., 2009). As a constituent of IL-1 superfamily, IL-33 levels are also elevated and co-expressed with TNF-α within ASM bundles in endobronchial specimens from severe phenotypes of asthma (Prefontaine et al., 2009). Interestingly, TNF-α-induced IL-33 was found to be resistant to the mainstay anti-inflammatory treatments in cultured ASM, implying a role for IL-33 in severe and refractory asthma. Besides being the effector population, ASM also secrete IL-1β. Exposure of ASM to IgE immune complexes induces IL-5 secretion. The secreted IL-5, in a paracrine manner, enhances eosinophil survival, and, in an autocrine fashion, augments IL-1β expression (Hakonarson et al., 1999). Accordingly, ASM cells simulated with IgE complexes, IL-5 or IL-1β synthesize signalling molecules in the IL-1 axis, including IL-1α, IL-1β, IL-1β-converting enzyme, IL-1 receptor accessory protein, IL-1 receptor, IL-1 receptor-like 1 and IL-18 receptor 1 (Whelan et al., 2004; Hershenson et al., 2008). Among other functions, members of the IL-17 cytokine family also enhance influx of leucocytes and induce pro-inflammatory mediators in airway tissues. While IL-17 increases ASM-derived eotaxin-1 and CXCL-8 levels, IL-1β and TNF-α increase IL-17 receptors isoforms on ASM, further perpetuating inflammation.

Interleukin-4 (IL-4) and interleukin-13 (IL-13)

Although asthma has been genetically linked to polymorphisms in the genes encoding IL-4Rα, IL-13Rα, STAT-6 and BCL6, the role of IL-4 or IL-13 in mediating airway inflammation or dysfunction remains unclear. Recent studies show that the presence of IL-4+ and OX40+ (a protein with implications in Th2 polarization and eosinophilic inflammation) cells among ASM correlates with asthma severity (Siddiqui et al., 2010). As a consequence of IL-4 and TNF-α stimulus, ASM cells express FcεRI that further augments the selective CC (eotaxin-1 and RANTES, but not thymus activation-regulated chemokine) and IL-8, IP-10 chemokine secretion following IgE exposure (Redhu et al., 2009). In vitro studies showed that IL-4 and IL-13 induce AHR in a STAT6-dependent manner and that carbachol-induced hyperresponsiveness was attenuated in STAT6−/− mice. Others showed that anti-IL-4Rα antibodies attenuate acetylcholine-induced responsiveness in IgE-sensitized ASM, supporting the role of IL-4Rα-STAT6 pathway in mediating AHR (Grunstein et al., 2002). In response to IL-4 or IL-13, human ASM cells release eotaxin in vitro that is inhibited by anti-IL-4Rα antibodies and antisense oligonucleotides to STAT6 (Hirst et al., 2002; Peng et al., 2004). In either IL-13−/− mice, IL-4/5/9/13−/− or wild-type mice, IL-13 was required for AHR, ASM hyperplasia and increases in IgE levels (Nath et al., 2007). Interestingly, these reports stand in contrast to those reported in smooth muscle myosin heavy chain creIL-4Rα−/lox mice whose IL-4Rα showed little effect on allergen-induced AHR or expression of other inflammatory biomarkers as compared to wild-type mice (Kirstein et al., 2010). Collectively, the role of IL-4 and IL-13 in allergic asthma remains unclear, and IL-4Rα-independent compensatory mechanisms may mediate AHR.

Interleukin-6 (IL-6)

Disruption of IL-6-mediated responses by IL-6Rα-blocking antibody diminishes ovalbumin (OVA)-induced Th2 inflammation; others showed that Aspergillus fumigatus (Af)-mediated mucus secretion is substantially diminished in IL-6−/− mice, suggesting a role for IL-6 in mediating/resolving allergen-induced AHR and inflammation. Pursuing experimental models of allergen-induced AHR in transgenic mice, investigators show that airway inflammation and bronchial reactivity can be uncoupled with IL-6 overexpression enhancing pro-inflammatory cytokines, TNF-α and IL-1β, but diminishing carbachol-induced responsiveness (Elias et al., 1997). Based on the demonstrated ability of several disease-specific mediators including TNF-α, IL-1β, transforming growth factor β (TGF-β), thymic stromal lymphopoietin, IL-17A, bradykinin, endothelin-1 (ET-1) and sphingosine-1-phosphate (S-1-P) to induce IL-6 secretion in ASM, airway myocytes may directly contribute to IL-6 production in asthma (Ammit et al., 2001; McKay and Sharma, 2002; Iwata et al., 2009; Tliba and Panettieri, 2009; Shan et al., 2010). In a more complex role, TNF-α induces interferon (IFN)-β secretion from ASM, which by its autocrine actions alters TNF-α-mediated IL-6 and RANTES secretions (Tliba et al., 2003). ASM in spatial proximity to epithelium also enhances basal or TNF-α-induced IL-6 and IP-10 secretions (Damera et al., 2009b).

IL-17A, a CD4+ cell-derived cytokine, also selectively augments TNF-α-induced IL-6 and IL-8 expression by altering transcript stability, while showing little effect on IL-1β-mediated responses (Henness et al., 2004). Despite the lack of a canonical IL-6 membrane-bound receptor (mIL-6R) in ASM, IL-6 induces inflammation and vessel expansion by release of eotaxin and vascular endothelial growth factor (VEGF) via the soluble IL-6R (sIL-6Rα) receptor (Ammit et al., 2007). Evolving evidence also shows that conditioned serum from ASM cells treated with cytomix (TNF-α, IL-1β and IFN-γ) promotes an eosinophilopoietic potential on CD34+ bone marrow-derived cells that was ablated in the presence of neutralizing antibodies to IL-5 and GM-CSF (Fanat et al., 2009). As a modulator of eosinophil trafficking, activation and the survival in asthma, ASM cells secrete GM-CSF in response to TNF-α/IL-1 alone or in combination with serum, or mast cell-derived tryptase. In response to histamine, ASM cell secretion of IL-1β-induced GM-CSF is increased while diminishing TNF-α-mediated RANTES output, an effect abrogated by selective antagonism at the H1R (Chhabra et al., 2007). As a facilitator and sustainer of leucocyte trafficking in airways, ASM stimulation with endothelin (ET-1) and TNF-α enhances both GM-CSF and ET-1 secretion by positive feedback loops, a mechanism impaired by bosentan and specific inhibition of either ET(A)R or ET(B)R (Knobloch et al., 2009). Oncostatin M, another member of the IL-6 family, enhances IL-1R1 expression and augments IL-1β-mediated VEGF, MCP-1 and IL-6 secretion, or synergizes with IL-13 and IL-4 to increase eotaxin-1 expression in human ASM (Faffe et al., 2005a,b;). Collectively, these data suggest a dynamic, paracrine crosstalk modulating IL-6 secretion in asthma.

Interleukin-8 (IL-8)

As with COPD, increases in neutrophilic inflammation in subjects with severe asthma correlate with production of CXCL-8 within the lungs. CXCL-8, found in bronchoalveolar lavage (BAL) fluid and in the supernatants of cytokine-stimulated ASM cells, activates CXCR-1 receptors promoting mast cell migration. Emerging evidence shows that enhancement in CXCL-8 secretion in asthma can increase binding of NF-κB, C/EBPβ and RNA Pol II elements to CXCL-8 promoter (John et al., 2009) in ASM cells. Likewise, phenotypic differences in ASM have been suggested to augment IL-8-dependent AHR and to enhance IgE-mediated IL-8, IL-6 and IL-4 secretion from normal ASM (Govindaraju et al., 2006; 2008;). Human ASM cells produce IL-8 when treated exogenously with IL-1β, TNF-α or TGF-β (Chung, 2000). In a scenario reminiscent of COPD, pro-inflammatory stimuli, such as TNF-α and cigarette smoke, synergize to induce IL-8 secretion from ASM (Oltmanns et al., 2005). Bradykinin directly elevates IL-8 secretion via the canonical bradykinin receptor-Gq-ERK cascade. Such effects are augmented by β2-AdR-agonists and/or by cAMP-regulated Epac1, Epac2 and PKA (Pang and Knox, 1998) (Roscioni et al., 2009). Acetylcholine induces M3R-mediated neutrophil chemotactic responses and mucus hypersecretion in airway epithelial cells. Later studies show that M3R agonists augment cigarette smoke-induced IL-8 secretion in airway myocytes (Gosens et al., 2009). These data suggest that anticholinergics such as tiotropium bromide in COPD (Oenema et al., 2010; Wollin and Pieper, 2010) may offer value in IL-8-dependent airway inflammation. In addition to inhibiting AHR, other M3R antagonists such as aclidinium bromide have substantial pharmacological benefits by inhibiting airway eosinophilia in murine models of asthma (Damera et al., 2010). Whether mast cell-derived histamine stimulates IL-8 secretion in ASM remains unknown; however, β-tryptase, another mast cell-derived serine protease, substantially enhances IL-8 via transcriptional and post-transcriptional mechanisms (Mullan et al., 2008).

Interleukin-9 (IL-9)

Overexpression of IL-9 in murine models evokes airway eosinophilia, mast cell hyperplasia, mucus production and AHR, reminiscent of the asthma diathesis. Gene mapping for bronchial hyperresponsiveness in humans suggested IL-9 and IL-9R are candidate genes for asthma. Enhanced expression of ASM IL-9R in biopsies from atopic asthma subjects as compared with that of non-asthmatic subjects has been shown. Early studies demonstrate that, in spite of its minimal effect in directly mediating ASM-derived cytokine secretion, IL-9 augments TNF-α-induced IL-8- or IL-13-induced eotaxin release in isolated human ASM (Baraldo et al., 2003). Further, IL-9 selectively and directly enhances eotaxin-1/CCL11 secretion that can promote airway eosinophilia (Yamasaki et al., 2010).

Prostaglandins and leukotrienes

Activation of ASM COX-2 enzymes generates arachidonic acid metabolites including PGE2. PGE2 stimulates EP1 receptors that mediate Ca2+ mobilization, EP3 receptors that inhibit adenylate cyclase, and EP2 and EP4 receptors that activate adenylate cyclase. Overall, physiological responses to PGE2 were thought to depend on selective receptor expression on target tissues, and recent reports suggest that PGE2 expression is also mediated by receptor sensitivity (Rolin et al., 2006). In murine models of asthma, evaluating pharmacological consequences of prostaglandin depletion with COX inhibitors or chimeric knockouts of COX show augmented airway eosinophilia, enhanced Th2 cytokine profile and AHR. A wide range of stimuli including bradykinin, IL-1β, TNF-α, IFN-γ, ozone, cigarette smoke extract, lipopolysaccharide (LPS), mechanical stretch and cAMP-elevating agents potentiate PGE2 production by ASM cells (Clarke et al., 2009b). ASM-derived PGE2 can modulate a wide variety of outcomes such as mast cell mediator release, epithelial barrier function, neutrophil and eosinophil chemotaxis, lymphocyte-derived IgE production and structural cell mitogenesis. Leptin, an obesity-linked hormone, inhibits platelet-derived growth factor (PDGF)-mediated human ASM proliferation and migration and IL-13-induced eotaxin production via the production of PGE2 (Nair et al., 2008). Environmental pollutants such as ozone induce structural cell-derived IL-6 via early enhancement of PGE2 (Damera et al., 2009b). While cytokine treatment induces β2AR desensitization in murine tracheal rings, mice deficient in EP2 receptor appear resistant to such effects, implying that PGE2-mediated mechanisms serve as modulators of both inflammation and airway β2AR responsiveness. Others postulate a protective role for PGE2 concerning airway inflammation. For instance, mice lacking EP3 receptors show exaggerated airway inflammation and hyperresponsiveness in OVA-induced models of AHR (Kunikata et al., 2005). In individuals with atopic asthma, inhaled PGE2 abrogates early- and late-phase responses to antigen challenge (Gauvreau et al., 1999). Similarly, individuals with aspirin-sensitive airways disease show depleted PGE2 production in a variety of cell types, and inhalation of PGE2 reversed aspirin-induced airflow obstruction (Sestini et al., 1996). In addition to PGE2, other prostaglandins such as prostaglandin-D2 and thromboxane are bronchoconstrictors and could also exaggerate allergen-mediated airway eosinophilia (Allen et al., 2006; Boyce, 2008).

The activation of 5-lipoxygenase (5-LO) enzyme generates bioactive leukotrienes (LTs) from arachidonic acid. Two groups of LTs, the dihydroxy LT, LTB4, and the cysteinyl LTs (Cys-LTs), composed of LTC4, LTD4 and LTE4, act through distinct receptors (Hallstrand and Henderson, 2010; Singh et al., 2010). CysLT production is increased in asthma subjects, particularly during an exacerbation or upon challenge. Indeed, CysLT production is increased in some asthma subjects, particularly during exacerbation or upon provocative challenge (Montuschi et al., 2006). Accordingly, 5-LO inhibition by zileuton or antagonism of CysLT1 receptor by zafirlukast improves FEV1measurements in individuals with asthma (Drazen, 1998). Subsequent studies also show a positive correlation between CysLTs in the exhaled breath condensate and reticular basement membrane thickening in asthma (Lex et al., 2006). In vivo studies investigating OVA sensitization and challenge in 5-LOnull and LTC4Snull mice reported reduced BAL fluid eosinophil numbers, diminished methacholine-induced AHR, and attenuation of total IgE and OVA-specific IgG in serum (Bosse et al., 2009; Mehrotra and Henderson, 2009). Treatment of ASM with atopic serum or IL-1 can activate the 5-LO pathway. Post Af sensitization and challenge, Th2-type inflammation and AHR to methacholine were associated with enhanced LTD4-CysLT1 receptor interactions in ASM tissue (Kim et al., 2006). While CysLT1 receptor enhancement drives IL-13 and LTD4 mediated ASM proliferation, Cys-LT directly or in conjunction with histamine mitigates ASM contractility. Others have shown that enzyme inhibitors 5-LO and LT receptor antagonist (pranlukast), but not an LTB4 receptor antagonist, diminished DNA synthesis in a rat model of allergic asthma (Salmon et al., 1999). The outcomes were further substantiated, when treatment with montelukast substantially inhibited development of ASM cell hyperplasia in another murine model (Henderson et al., 2002). Others showed that LTD4 modulates myocyte hyperplasia and AHR via paracrine mechanisms involving epithelial cell-derived TGF-β1 (Espinosa et al., 2003; Bosse et al., 2008).

Current evidence is compelling to suggest that ASM secretes numerous cytokines/chemokines, eicosanoids and prostaglandins that may alter autocrine and paracrine function of structural and trafficking leucocytes in the airways. The specific mechanisms that induce expression of such molecules, in part, differ from that of other cells. Future studies focusing on the inhibition or the stimulation of such molecules and structural cells may offer new therapeutic targets in the treatment of asthma and COPD.

Toll-like receptor-mediated immunomodulation and ASM

Respiratory infections evoke asthma exacerbations mediated, in part, by the expression and activation of toll-like receptor (TLR) by microbial components that promote airway inflammation. Resolution of infection depends on the specificity of the 11 known TLRs to bind distinct products of microbial-derived molecules and to elicit downstream signalling (Phipps et al., 2007; Drexler and Foxwell, 2010). ASM cells express transcripts for most TLR isoforms, although quantitative disparities exist among TLR isoforms (Chaudhuri et al., 2007). ASM cells express functional TLR-2, TLR-3 and TLR-4 (Sukkar et al., 2006). While in vitro infection of human ASM cells with respiratory viruses induces production of IL-1β, IL-6, IL-8 and IL-11, expression of TLRs is also increased after treatment with TNF-α, IL-1β and IFN-γ. These data suggest that ASM TLR activation may amplify ongoing inflammatory processes (Damera et al., 2009a). Similarly, bacterial endotoxins or autologous proteins with conserved TLR recognition sequences can also enhance synthesis of leucocyte chemotractant mediators including eotaxin-1, CXCL-8 or GM-CSF (Issa et al., 2008). LPS-induced secretion of monocyte chemotractants such as CCL2 (MCP-1) are also conditionally expressed by ASM when co-cultured with peripheral blood mononuclear cells (Morris et al., 2005). Besides mediating TLR-4-mediated induction of IL-6 secretion, LPS alters agonist-mediated constrictor and relaxant responses (Shan et al., 2006). Likewise, tracheal organ culture incubated with LPS or polyinosinic polycytidylic acid [poly(I:C), a TLR-3 agonist] enhances bradykinin- and [des-Arg(9)]-bradykinin-induced contractions (Bachar et al., 2004). Surprisingly, TLR effects in human precision-cut lung slices did not modulate agonist-induced bronchodilation (Cooper et al., 2009).

Besides activating TLR receptors on airway epithelial cells and immune cells, Der p2 (house dust mite) allergen has intrinsic enzymatic activity that enhances epithelial permeability and detachment, potentially exposing the submucosa to allergens (Roche et al., 1997). Recent studies show that Der p2 activates TLR-2/MyD88 in ASM cells that induce secretion of MCP-1 and IL-6 (Chiou and Lin, 2009). Respiratory syncytial virus (RSV) ssRNA are potent activators of TLR-7/8, and RSV infection enhances airway epithelial barrier permeability, potentially leading to enhanced passage of RSV ssRNA to the submucosa (Wang et al., 1998). However, the consequences of TLR-7/8 activation in specific airway tissues remain unclear; some studies show that activation of TLR-7/8 induces pro-inflammatory mediators and enhances mucus hypersecretion, while others demonstrate a substantial reduction in immune cell-mediated increases in airway myocyte mass, inflammation and hyperresponsiveness in murine models.

ASM modulates leucocyte transmigration

As leucocyte transmigration is crucial in mediating immune responses, elucidating molecular mechanisms may offer new therapeutic targets for mitigating airway inflammation. Leucocyte migration and retention, initially evoked by selectins on endothelial cells, subsequently express CAMs presented by ‘primed’ airway structural cells. Studies in vitro and in vivo show that expression of CAMs mediates cell–cell interactions implicated in inflammation and tissue remodelling (Kelly et al., 2007). ICAM-1 expression in ASM cells is mediated by pro-inflammatory mediators including cytokines, bacterial endotoxins and viral proteins, implying a broader role in chronic airway diseases (Tliba et al., 2008). Surface expression of CAMs and their interactions with immune cells play a role in mediating ASM-leucocyte function interactions in asthma.

TNF-α and IL-1β induce ICAM-1 and VCAM-1 in ASM. Additionally, independent of replication, rhinovirus (RV-15) induces ASM-derived IL-5 and IL-1β secretion via interactions with ASM-resident ICAM-1 molecules (Grunstein et al., 2001; Oliver et al., 2006). In ASM cells, TNF-α activation and nuclear translocation of p65 NF-κB are sufficient to induce VCAM-1 but not ICAM-1, implying signalling diversity in CAM expression (Zerfaoui et al., 2008). Reinforcing the role of NF-κB-independent mechanisms in CAM modulation, IL-1-mediated expression of ICAM-1 is attenuated by PGE2, forskolin and short- and long-acting β2-AdR-agonists in a PKA-dependent and NF-κB-independent manner (Kaur et al., 2008). While functionally inconclusive, expression of CAMs could mediate T-cell adherence to airways potentially altering Ach-induced bronchoconstriction and isoprenaline-mediated bronchodilation (β2-AdR) (Hughes et al., 2000; Hakonarson et al., 2001). While toluene diisocyanate (TDI) administration potentiates AHR and airway inflammation including ASM-resident CAM expression in mice, administration of peroxisome proliferator-activated receptor (PPAR)γ agonists or adenovirus carrying PPARγ2 cDNA reversed TDI-mediated induction of cytokine and CAMs, implying a protective role of PPARγ in the pathogenesis of asthma (Lee et al., 2006). Studies using antibodies blocking lymphocyte function-associated antigen 1 (LFA-1) and Very Late Antigen-4 (VLA-4) on activated T cells or antibodies against ICAM-1 and VCAM-1 on ASM cells showed greater attenuation of T-cell adherence to ASM compared to either anti-ICAM or anti-VCAM treatment alone (Duplaa et al., 1997). Addition of anti-CD44 Abs to combination of mAbs against LFA-1, VLA-4, synergistically, reduces the binding of activated T cells to the level observed for resting T cells (Lazaar et al., 1994). Suggestive of a broader role of CAMs in leucocyte trafficking, anti-ICAM-1 or anti-VCAM-1 attenuates eosinophil and neutrophil adherence to ASM. Expanding the role of CAMs, more recent studies suggest that mast cell infiltration of ASM cells in asthma occurs via expression of a heterophilic adhesion molecule, tumour suppressor in lung cancer-1 (TSLC-1) (Yang et al., 2006). Studies by Ramos-Barbón et al. showed that adoptive transfer of CD4+ T cells from sensitized rats induces proliferation and attenuates apoptosis of ASM in naive recipients after successive antigen challenges. Concomitantly, modified CD4+ T cells expressing enhanced green fluorescent protein were localized in juxtaposition to ASM cells conferring that cell–cell interaction participates in airway remodelling. In an expanding role for CAMs in airway inflammation, studies also determined a critical role for a β-galactoside-binding lectin, Galectin-3 (Gal-3), in eosinophil trafficking and recruitment in allergic inflammation (Ramos-Barbón et al., 2005). Comparative studies in transgenic mice revealed that gal3−/− mice showed significantly less AHR, lower Th2 responses after challenge and altered expression of other CAMs when compared to gal3+/+ mice (Zuberi et al., 2004).

ASM phenotype switching

Severe asthma represents a heterogeneous disease, with lung function that is, in part, irreversibly reduced in some (Moore and Peters, 2006; Chanez et al., 2007). Among other features, a prominent feature in such patients includes increases in ASM mass, leading investigators to postulate that ASM growth, hypertrophy or hyperplasia could evoke irreversible airway obstruction. Proliferative responses in ASM are stimulated by epidermal growth factor (EGF), insulin-like growth factors (IGFs), PDGF and fibroblast growth factor-2. Some contractile agents such as histamine, ET-1, substance P, 5-HT, α-thrombin, thromboxane A2 and LTD4 also mediate ASM mitogenesis (Lazaar and Panettieri, 2005; Dekkers et al., 2009). The precise role of cytokines, however, remains ambiguous concerning ASM growth. While IL-1β and IL-6 mediate hyperplasia in guinea pig ASM cells, such effects are less evident in human ASM. Similarly, while TGF-β1 and TNF-α induce proliferation or potentiate the effects of some mitogens, some studies suggest that these cytokines inhibit mitogenesis. More definitive are the effects of IFN (type-1 and -2) and IL-4 that inhibit ASM proliferation by diverse signalling mechanisms (Amrani et al., 2003; Tliba et al., 2003).

Studies have identified signalling events that are both necessary and sufficient to promote ASM growth. Such events serve as critical checkpoints that modulate ASM mitogenesis. Mitogens stimulate their cognate receptors inducing activation of ERK, p21Ras (Ras), Src and PI3K leading to human ASM growth. As with most cell types, expression of d-type cyclins (D1, D2 and D3) is required for mitogen-induced cell cycle progression. In ASM cells, studies showed that cyclin D1 expression may serve as a possible surrogate for cell proliferation. BAL fluid derived from airways of subjects with asthma enhances ERK activation, cyclin D1 protein abundance, DNA synthesis and cell number of cultured human ASM cells (Naureckas et al., 1999). In cultured ASM cells, activation of ERK is required for cyclin D1-mediated DNA synthesis. Likewise, studies also implicate involvement of Ras/Raf proteins in ERK-mediated ASM proliferation. For instance, anti-pan Ras neutralizing antibody attenuates DNA synthesis in human airway myocytes, and overexpression of a dominant-negative form of Ras inhibits PDGF-stimulated ERK activation (Page et al., 1999). Selective overexpression of an activity deficient mutant of Raf-1 substantially diminishes endothelin-mediated ERK activation in rat airway myocytes, suggesting a role for Ras/Raf (Vichi et al., 1999). Besides ERK, pharmacological inhibitors of PI3K also diminish cyclin D1 protein expression and DNA synthesis. Similarly, overexpression of active Rac1 or PI3K initiates transcription at the cyclin D1 promoter, and such mechanisms occur independent of ERK activation and remain insensitive to mitogen-activated ERK kinase inhibitors, suggesting that Rac1- or PI3K-mediated cell cycle progression occurs in parallel. Supportive of these outcomes, overexpression of Src or PI3K alone stimulated cell growth, and inhibition of PI3K abrogated mitogen-induced ASM proliferation. PI3K-dependent downstream events ultimately activate S6K1 that is associated with the efficiency of translation of mRNAs that are involved in mitogenesis and whose inhibition by rapamycin attenuates growth factor-induced DNA synthesis in airway myocytes (Scott et al., 1996). Importantly, prolonged activation of PI3K and S6K1 at 12 h discriminated ASM mitogens from non-mitogenic agonists and mediators that otherwise equally activated ERK1/2 at 1 h (Krymskaya et al., 2000). Activation of receptor tyrosine kinases (RTKs) can also initiate signalling cascades involving Rac and NADPH oxidase to produce reactive oxygen species leading to JAK-2/3-mediated cyclin D1 transcription (Simeone-Penney et al., 2008).

Among mitogenic responses elicited by growth factors and contractile agonists, important disparities exist. Mitogenesis stimulated by growth factors is mediated by RTK activity whereas G-protein coupled receptors (GPCRs) lack intrinsic kinase activity (Ritter and Hall, 2009). Growth induced by RTK activation appears to be Ras- and PI3K-dependent whereas that induced by GPCRs may be more diverse and use an array of small G-proteins and phosphotyrosine scaffolding proteins including Src (Chambard et al., 1987; Kavanaugh et al., 1988). In addition to differences between the proliferative mechanisms of growth factors and those of GPCR agonists, there exists substantial variability in the mitogenic capacity among agonists (Ritter and Hall, 2009). First, some, but not all, GPCR contractile agonists stimulate ASM proliferation, even though most induce comparable levels of PI hydrolysis and cytosolic calcium transients (Krymskaya et al., 2000; Billington et al., 2005; Kong et al., 2006). Second, the mitogenic effects of endothelin and serotonin on vascular smooth muscle (SM), and thrombin on ASM, are coupled to Pertussis toxin-sensitive G-proteins, not Pertussis toxin-insensitive G-proteins that regulate agonist-induced ASM contraction (Kavanaugh et al., 1988; Komuro et al., 1988; Noveral et al., 1992; Panettieri et al., 1995; 1996;). The differences in proliferative responses induced by growth factors and agonists (and among agonists) imply that other regulatory components are also involved.

ASM cells manifest phenotype plasticity, whereby contractile myocytes undergo switching toward a more proliferative phenotype in the presence of mitogens or when grown on distinct extracellular matrix (ECM) proteins (Halayko et al., 2006; Dekkers et al., 2007). Predictably, such alterations correlated with quantitative increases in synthetic pathways for protein and lipids and mitochondrial function with a diminished abundance of contractile proteins. In contrast to specific proteins, namely, smooth muscle myosin heavy chain, SM22, calponin and smooth muscle α-actin that mark pro-contractile phenotype, proliferating ASM show enhanced non-muscle myosin heavy chain, l-caldesmon, vimentin, α/β-protein kinase C and CD44 homing cellular adhesion molecule (Halayko et al., 2008; Hirota et al., 2009). Additionally, dystrophin glycoprotein complex expression and accumulation correlated with a contractile phenotype and diminished in proliferating ASM cells (Sharma et al., 2008). Although investigators have examined the acute effects of growth factors on contraction of SM, few have addressed whether more prolonged exposure that likely occurs in vivo modulates agonist-induced force generation.

Our studies to identify intracellular mediators that modulate regulators of G-protein signalling (RGS) molecule identified this molecule as potential candidate that mediates ASM plasticity. RGS molecules act as canonical modulators of GTPase accelerating (GAP) activity and interact with the G-alpha subunits; RGS expression or depletion may define GPCR-mediated contractile outcomes in varied tissues (Hollinger and Hepler, 2002; Sethakorn et al., 2010). Mice deficient in RGS2 develop a hypertensive phenotype that, in part, is reversed with an angiotensin II inhibitor, suggesting that loss of RGS2 increases angiotensin II-induced vasomotor tone (Heximer et al., 2003). In cardiac muscle, RGS4 plays a critical role in regulating the chronotropic actions of Ach. Lack of RGS4 enhances sensitivity to carbachol-induced bradycardia and evokes arrhythmias (Neubig, 2008). Others report that increases in cardiac muscle expression of RGS4 decreases cardiac inotropy promoting heart failure (Owen et al., 2001). More recent studies show that RGS molecules also have GAP-independent functions that mediate cell proliferation by interacting with components of PI3K. Druey et al. showed that RGS13 inhibits allergic responses by physically interacting with the regulatory p85α-PI3K subunit of phosphatidylinositol-3-OH kinase in mast cells, thus disrupting its association with an Fc∈RI-activated scaffolding complex (Bansal et al., 2008). Similarly, RGS16 binding to the amino-terminal SH2 and inter-SH2 domains of p85α-PI3K inhibits interaction with the EGF receptor-associated adapter protein Gab1, altering proliferation. Among other events, newer studies now show that spatial positioning of downstream signal transduction molecules such as Grb2 and PI3K are critical for optimal RTK-mediated proliferation (Fukumoto et al., 2009). In our studies, we showed that the kinetics of PI3K and S6K1 defined the cooperative mitogenic effects of GPCR and RTK activation in ASM cells. Although the agonist- and growth factor-induced amplitude and sustained activation of PI3K and S6K1 at 4 h predicted the magnitude of ASM mitogenesis, the specific mechanism remains elusive (Krymskaya et al., 2000). We now speculate that the early activation of Src, PI3K and ERK1/2 that subsequently activates S6K1, which occurs at 0.5–2 h, increases expression of RGS proteins that then act to sustain activation of PI3K and S6K1 for 4–8 h, signals necessary to promote ASM cell proliferation through the G0/G1 checkpoint of the cell cycle. As illustrated in Figure 1, expression of distinct RGS isoforms and their interactions with PI3K and G-alpha subunits could define critical signalling events that mediate ASM plasticity in severe asthma (Liang et al., 2009).

Figure 1.

Figure 1

Potential mechanisms involving regulators of G-protein signalling (RGS) proteins as modulators of airway smooth muscle plasticity. Airway smooth muscle (ASM) mitogens including growth factors and distinct contractile agonists mediate expression of RGS isoforms, which interact with (i) p85α subunit of PI3K to sustain AKT activity critical for proliferation, and (ii) Gα subunits of heterodimeric G-protein coupled receptors to attenuate agonist-induced ASM contractile responses.

Extracellular matrix- and matrix metalloproteinase-mediated airway remodelling

Chronic inflammation may promote airway remodelling manifested by increases in ASM mass, in goblet cell/mucous glands and matrix deposition. Surprisingly, some of these remodelling effects are independent of inflammation and often precede the onset of asthma symptoms (Benayoun et al., 2003). ASM cells can play a role in airway remodelling by producing ECM components and matrix modifying enzymes, matrix metalloproteinases (MMPs). These molecules substantially alter proliferation, migration and contractile responses to mediators. In comparison to normal airways, ECM deposition is altered in individuals with asthma with enhanced deposition of collagens I, III and V, fibronectin, tenascin, hyaluronan, versican and laminin and decreased collagen IV and elastin deposition (Freyer et al., 2001; Burgess et al., 2009). Accordingly, isolated ASM from individuals with asthma produces a distinct array of ECM proteins, while ASM cells from healthy subjects manifested enhanced proliferation when cultured on a matrix provided by the asthma-derived ASM cells. Further, altered ECM deposition surrounding ASM cells may have a physiological impact by modulating airway rigidity and attenuating distensibility. ECM composition can alter contractile protein expression such as SM specific α-actin, myosin heavy chain and calponin in airway myocyte cultures. Studies in vitro showed that ECM produced by ASM isolated from individuals with asthma augments IL-13-induced eotaxin release, implying a potential role of ECM components in augmenting eosinophil chemotaxis. ECM can also modulate fibrotic signals by sequestering and influencing the effects of TGF-β, a cytokine whose levels correlate with basement membrane thickness in asthma.

Integral to the remodelling process, ECM components and mediators of inflammation could potentiate the production and activity of matrix modifying MMP enzymes that, in turn, modulate matrix-mediated signalling. Cell and matrix crosstalk occurs via the integrin family of transmembrane receptors, and proteolytic activity of discreet MMPs unmasks integrin-binding sites in the substrate altering cell function (Gueders et al., 2006). While degrading ECM structure, MMPs also facilitate leucocyte migration through the ECM and endothelial cells and affect activation and survival by cleaving cytokines and their cognate receptors (Lagente and Boichot, 2010). Of the 25 mammalian MMPs, ASM-derived collagen modifying MMP-1,19, gelatinases MMP-2,9, stromelysins MMP-3,10, metalloelastase MMP-12 and membrane-bound MMP-14 are regulated transcriptionally and coordinate expression of endogenous tissue inhibitors of metalloproteinases (TIMPs) (McKay and Sharma, 2002; Elshaw et al., 2004; Gueders et al., 2010).

Transcriptional enhancement of MMPs appears to be selective to growth factors in MMP-2 and -9, LTs in MMP-1, endoproteases in MMP-2, and mechanical stress in MMP-1, -2, -3 and -14 (Rajah et al., 1996; 1999; Foda et al., 1999; Hirst, 2003; Xie et al., 2005). Allergen exposure of mice enhances MMP-19, while MMP-19 gene deletion evokes tenascin-C accumulation in peribronchial areas. Accordingly, Th2-associated eosinophilic inflammation and AHR appear to be mediated in a MMP-19-dependent manner where tenascin could mitigate Th2 inflammation in allergic asthma (Gueders et al., 2010). Enhancement of MMP-2 by thrombin or overexpression of MMP-14 also mediate ASM migration (Hasaneen et al., 2005; Henderson et al., 2007). Th2 cytokines including IL-4 and IL-13 induce MMP-1 that, in turn, alters collagen type I matrix and airway contractility (Ohta et al., 2008). MMP-1 activation degrades insulin-like growth factor binding protein, releasing IGF and facilitating ASM proliferation and migration. Others show that mitogens including PDGF, TGF-β or combination up-regulate MMP-1 and MMP-3, and that silencing MMP-3 production decreases migration of ASM cells (Ito et al., 2009). Epithelium is a prominent source of mitogens, cytokines and MMP; disruption of epithelial integrity could enhance MMP secretion affecting ASM migration and growth (Malavia et al., 2009). Similar paracrine interactions exist between mast cells and ASM whereby activation of primary human mast cells by IgE receptor cross-linking stimulates contraction of ASM via expression of MMP-1 and MMP-2 (Margulis et al., 2009). Concomitant and proportional secretion of TIMP-1 inhibits MMP-1, -2, -3 and -9 activity and TIMP-2 selectively complexes and attenuates MMP-2 proteolytic activity, and disruption in these ratios could modulate airway obstruction in asthma (Foda et al., 1999; Johnson, 2001; Elshaw et al., 2004).

Conclusion

ASM plays a pivotal role in modulating bronchomotor tone and serves as an important therapeutic target for β-agonists in the promotion of bronchodilation. Contemporary thought identifies ASM as an immunomodulatory cell that may orchestrate and perpetuate airway inflammation. The molecular mechanisms that regulate secretion of chemokines and cytokines from structural cells appear to differ from that of trafficking leucocytes, and further studies to identify the critical pathways mediating such processes may identify novel therapeutic targets in the treatment of asthma and COPD.

Acknowledgments

The authors acknowledge Mary McNichol for editorial assistance in preparation of this manuscript.

Glossary

Abbreviations

5-LO

5-lipoxygenase

Af

Aspergillus fumigatus

AHR

airway hyperresponsiveness

ASM

airway smooth muscle

CAM

cell adhesion molecule

COPD

chronic obstructive pulmonary disease

EGF

epidermal growth factor

GM-CSF

granulocyte-macrophage colony-stimulating factor

GPCR

G-protein coupled receptor

IL

interleukin

LPS

lipopolysaccharide

LT

leukotriene

MCP

monocyte chemotactic protein

MMP

matrix metalloproteinase

PGE2

prostaglandin-E2

PDGF

platelet-derived growth factor

PPAR

peroxisome proliferator-activated receptor

RANTES

regulated upon activation normal T-cell expressed and secreted

RGS

regulator of G-protein signalling

RTK

receptor with intrinsic tyrosine kinase

TDI

toluene diisocyanate

TGF-β

transforming growth factor β

Th1

T-helper 1

Th2

T-helper 2

TIMP

tissue inhibitor of metalloproteinases

TNF-α

tumour necrosis factor-α

VEGF

vascular endothelial growth factor

Conflicts of interest

None.

Supporting Information

Teaching Materials; Fig 1 as PowerPoint slide.

bph0163-0068-SD1.pptx (150.2KB, pptx)

References

  1. Allen IC, Hartney JM, Coffman TM, Penn RB, Wess J, Koller BH. Thromboxane A2 induces airway constriction through an M3 muscarinic acetylcholine receptor-dependent mechanism. Am J Physiol Lung Cell Mol Physiol. 2006;290:L526–L533. doi: 10.1152/ajplung.00340.2005. [DOI] [PubMed] [Google Scholar]
  2. Ammit AJ, Hastie AT, Edsall LC, Hoffman RK, Amrani Y, Krymskaya VP, et al. Sphingosine 1-phosphate modulates human airway smooth muscle cell functions that promote inflammation and airway remodeling in asthma. FASEB J. 2001;15:1212–1214. doi: 10.1096/fj.00-0742fje. [DOI] [PubMed] [Google Scholar]
  3. Amrani Y, Tliba O, Choubey D, Huang CD, Krymskaya VP, Eszterhas A, et al. IFN-gamma inhibits human airway smooth muscle cell proliferation by modulating the E2F-1/Rb pathway. Am J Physiol Lung Cell Mol Physiol. 2003;284:L1063–L1071. doi: 10.1152/ajplung.00363.2002. [DOI] [PubMed] [Google Scholar]
  4. Ammit AJ, Moir LM, Oliver BG, Hughes JM, Alkhouri H, Ge Q, et al. Effect of IL-6 trans-signaling on the pro-remodeling phenotype of airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2007;292:L199–L206. doi: 10.1152/ajplung.00230.2006. [DOI] [PubMed] [Google Scholar]
  5. Bachar O, Adner M, Uddman R, Cardell LO. Toll-like receptor stimulation induces airway hyper-responsiveness to bradykinin, an effect mediated by JNK and NF-kappa B signaling pathways. Eur J Immunol. 2004;34:1196–1207. doi: 10.1002/eji.200324569. [DOI] [PubMed] [Google Scholar]
  6. Bansal G, Xie Z, Rao S, Nocka KH, Druey KM. Suppression of immunoglobulin E-mediated allergic responses by regulator of G protein signaling 13. Nat Immunol. 2008;9:73–80. doi: 10.1038/ni1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baraldo S, Faffe DS, Moore PE, Whitehead T, McKenna M, Silverman ES, et al. Interleukin-9 influences chemokine release in airway smooth muscle: role of ERK. Am J Physiol Lung Cell Mol Physiol. 2003;284:L1093–L1102. doi: 10.1152/ajplung.00300.2002. [DOI] [PubMed] [Google Scholar]
  8. Barnes PJ. The cytokine network in asthma and chronic obstructive pulmonary disease. J Clin Invest. 2008;118:3546–3556. doi: 10.1172/JCI36130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Benayoun L, Druilhe A, Dombret MC, Aubier M, Pretolani M. Airway structural alterations selectively associated with severe asthma. Am J Respir Crit Care Med. 2003;167:1360–1368. doi: 10.1164/rccm.200209-1030OC. [DOI] [PubMed] [Google Scholar]
  10. Billington CK, Kong KC, Bhattacharyya R, Wedegaertner PB, Panettieri RA, Jr, Chan TO, et al. Cooperative regulation of p70S6 kinase by receptor tyrosine kinases and G protein-coupled receptors augments airway smooth muscle growth. Biochemistry. 2005;44:14595–14605. doi: 10.1021/bi0510734. [DOI] [PubMed] [Google Scholar]
  11. Bosse Y, Thompson C, McMahon S, Dubois CM, Stankova J, Rola-Pleszczynski M. Leukotriene D4-induced, epithelial cell-derived transforming growth factor beta1 in human bronchial smooth muscle cell proliferation. Clin Exp Allergy. 2008;38:113–121. doi: 10.1111/j.1365-2222.2007.02873.x. [DOI] [PubMed] [Google Scholar]
  12. Bosse Y, Stankova J, Rola-Pleszczynski M. Cysteinyl-leukotrienes in asthmatic airway smooth muscle cell hyperplasia. Ann Allergy Asthma Immunol. 2009;102:16–21. doi: 10.1016/S1081-1206(10)60102-0. [DOI] [PubMed] [Google Scholar]
  13. Boyce JA. Eicosanoids in asthma, allergic inflammation, and host defense. Curr Mol Med. 2008;8:335–349. doi: 10.2174/156652408785160989. [DOI] [PubMed] [Google Scholar]
  14. Bradding P, Roberts JA, Britten KM, Montefort S, Djukanovic R, Mueller R, et al. Interleukin-4, -5, and -6 and tumor necrosis factor-alpha in normal and asthmatic airways: evidence for the human mast cell as a source of these cytokines. Am J Respir Cell Mol Biol. 1994;10:471–480. doi: 10.1165/ajrcmb.10.5.8179909. [DOI] [PubMed] [Google Scholar]
  15. Brightling C, Berry M, Amrani Y. Targeting TNF-alpha: a novel therapeutic approach for asthma. J Allergy Clin Immunol. 2008;121:5–10. doi: 10.1016/j.jaci.2007.10.028. quiz 11–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Burgess JK, Ceresa C, Johnson SR, Kanabar V, Moir LM, Nguyen TT, et al. Tissue and matrix influences on airway smooth muscle function. Pulm Pharmacol Ther. 2009;22:379–387. doi: 10.1016/j.pupt.2008.12.007. [DOI] [PubMed] [Google Scholar]
  17. Carroll NG, Mutavdzic S, James AL. Distribution and degranulation of airway mast cells in normal and asthmatic subjects. Eur Respir J. 2002;19:879–885. doi: 10.1183/09031936.02.00275802. [DOI] [PubMed] [Google Scholar]
  18. Cardell LO, Uddman R, Zhang Y, Adner M. Interleukin-1beta up-regulates tumor necrosis factor receptors in the mouse airways. Pulm Pharmacol Ther. 2008;21:675–681. doi: 10.1016/j.pupt.2008.04.002. [DOI] [PubMed] [Google Scholar]
  19. Chambard JC, Paris S, L'Allemain G, Pouyssegur J. Two growth factor signalling pathways in fibroblasts distinguished by pertussis toxin. Nature. 1987;326:800–803. doi: 10.1038/326800a0. [DOI] [PubMed] [Google Scholar]
  20. Chanez P, Wenzel SE, Anderson GP, Anto JM, Bel EH, Boulet LP, et al. Severe asthma in adults: what are the important questions. J Allergy Clin Immunol. 2007;119:1337–1348. doi: 10.1016/j.jaci.2006.11.702. [DOI] [PubMed] [Google Scholar]
  21. Chaudhuri N, Whyte MK, Sabroe I. Reducing the toll of inflammatory lung disease. Chest. 2007;131:1550–1556. doi: 10.1378/chest.06-2869. [DOI] [PubMed] [Google Scholar]
  22. Chhabra J, Li YZ, Alkhouri H, Blake AE, Ge Q, Armour CL, et al. Histamine and tryptase modulate asthmatic airway smooth muscle GM-CSF and RANTES release. Eur Respir J. 2007;29:861–870. doi: 10.1183/09031936.00106306. [DOI] [PubMed] [Google Scholar]
  23. 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;220:311–318. doi: 10.1002/jcp.21764. [DOI] [PubMed] [Google Scholar]
  24. Chung KF. Airway smooth muscle cells: contributing to and regulating airway mucosal inflammation. Eur Respir J. 2000;15:961–968. doi: 10.1034/j.1399-3003.2000.15e26.x. [DOI] [PubMed] [Google Scholar]
  25. Clarke D, Damera G, Sukkar MB, Tliba O. Transcriptional regulation of cytokine function in airway smooth muscle cells. Pulm Pharmacol Ther. 2009a;22:436–445. doi: 10.1016/j.pupt.2009.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Clarke DL, Dakshinamurti S, Larsson AK, Ward JE, Yamasaki A. Lipid metabolites as regulators of airway smooth muscle function. Pulm Pharmacol Ther. 2009b;22:426–435. doi: 10.1016/j.pupt.2008.12.003. [DOI] [PubMed] [Google Scholar]
  27. Cooper PR, Lamb R, Day ND, Branigan PJ, Kajekar R, San Mateo L, et al. 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–L537. doi: 10.1152/ajplung.00133.2009. [DOI] [PubMed] [Google Scholar]
  28. Damera G, Tliba O, Panettieri RA., Jr Airway smooth muscle as an immunomodulatory cell. Pulm Pharmacol Ther. 2009a;22:353–359. doi: 10.1016/j.pupt.2008.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Damera G, Zhao H, Wang M, Smith M, Kirby C, Jester WF, et al. Ozone modulates IL-6 secretion in human airway epithelial and smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2009b;296:L674–L683. doi: 10.1152/ajplung.90585.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Damera G, Jiang M, Zhao H, Fogle HW, Jester WF, Freire J, et al. Aclidinium bromide abrogates allergen-induced hyperresponsiveness and reduces eosinophilia in murine model of airway inflammation. Eur J Pharmacol. 2010 doi: 10.1016/j.ejphar.2010.09.043. [DOI] [PubMed] [Google Scholar]
  31. Dekkers BG, Schaafsma D, Nelemans SA, Zaagsma J, Meurs H. Extracellular matrix proteins differentially regulate airway smooth muscle phenotype and function. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1405–L1413. doi: 10.1152/ajplung.00331.2006. [DOI] [PubMed] [Google Scholar]
  32. Dekkers BG, Maarsingh H, Meurs H, Gosens R. Airway structural components drive airway smooth muscle remodeling in asthma. Proc Am Thorac Soc. 2009;6:683–692. doi: 10.1513/pats.200907-056DP. [DOI] [PubMed] [Google Scholar]
  33. Drazen J. Clinical pharmacology of leukotriene receptor antagonists and 5-lipoxygenase inhibitors. Am J Respir Crit Care Med. 1998;157(6 Pt 2):S233–S237. discussion S247–S248. [PubMed] [Google Scholar]
  34. Drexler SK, Foxwell BM. The role of toll-like receptors in chronic inflammation. Int J Biochem Cell Biol. 2010;42:506–518. doi: 10.1016/j.biocel.2009.10.009. [DOI] [PubMed] [Google Scholar]
  35. Duplaa C, Couffinhal T, Dufourcq P, Llanas B, Moreau C, Bonnet J. The integrin very late antigen-4 is expressed in human smooth muscle cell. Involvement of alpha 4 and vascular cell adhesion molecule-1 during smooth muscle cell differentiation. Circ Res. 1997;80:159–169. doi: 10.1161/01.res.80.2.159. [DOI] [PubMed] [Google Scholar]
  36. Elias JA, Wu Y, Zheng T, Panettieri R. Cytokine- and virus-stimulated airway smooth muscle cells produce IL-11 and other IL-6-type cytokines. Am J Physiol. 1997;273(3 Pt 1):L648–L655. doi: 10.1152/ajplung.1997.273.3.L648. [DOI] [PubMed] [Google Scholar]
  37. Elshaw SR, Henderson N, Knox AJ, Watson SA, Buttle DJ, Johnson SR. Matrix metalloproteinase expression and activity in human airway smooth muscle cells. Br J Pharmacol. 2004;142:1318–1324. doi: 10.1038/sj.bjp.0705883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Espinosa K, Bosse Y, Stankova J, Rola-Pleszczynski M. CysLT1 receptor upregulation by TGF-beta and IL-13 is associated with bronchial smooth muscle cell proliferation in response to LTD4. J Allergy Clin Immunol. 2003;111:1032–1040. doi: 10.1067/mai.2003.1451. [DOI] [PubMed] [Google Scholar]
  39. Faffe DS, Flynt L, Mellema M, Moore PE, Silverman ES, Subramaniam V, et al. Oncostatin M causes eotaxin-1 release from airway smooth muscle: synergy with IL-4 and IL-13. J Allergy Clin Immunol. 2005a;115:514–520. doi: 10.1016/j.jaci.2004.11.033. [DOI] [PubMed] [Google Scholar]
  40. Faffe DS, Flynt L, Mellema M, Whitehead TR, Bourgeois K, Panettieri RA, et al. Oncostatin M causes VEGF release from human airway smooth muscle: synergy with IL-1beta. Am J Physiol Lung Cell Mol Physiol. 2005b;288:L1040–L1048. doi: 10.1152/ajplung.00333.2004. [DOI] [PubMed] [Google Scholar]
  41. Fanat AI, Thomson JV, Radford K, Nair P, Sehmi R. Human airway smooth muscle promotes eosinophil differentiation. Clin Exp Allergy. 2009;39:1009–1017. doi: 10.1111/j.1365-2222.2009.03246.x. [DOI] [PubMed] [Google Scholar]
  42. Foda HD, George S, Rollo E, Drews M, Conner C, Cao J, et al. Regulation of gelatinases in human airway smooth muscle cells: mechanism of progelatinase A activation. Am J Physiol. 1999;277(1 Pt 1):L174–L182. doi: 10.1152/ajplung.1999.277.1.L174. [DOI] [PubMed] [Google Scholar]
  43. Freund V, Pons F, Joly V, Mathieu E, Martinet N, Frossard N. Upregulation of nerve growth factor expression by human airway smooth muscle cells in inflammatory conditions. Eur Respir J. 2002;20:458–463. doi: 10.1183/09031936.02.00269202. [DOI] [PubMed] [Google Scholar]
  44. Freyer AM, Johnson SR, Hall IP. Effects of growth factors and extracellular matrix on survival of human airway smooth muscle cells. Am J Respir Cell Mol Biol. 2001;25:569–576. doi: 10.1165/ajrcmb.25.5.4605. [DOI] [PubMed] [Google Scholar]
  45. Fukumoto T, Kubota Y, Kitanaka A, Yamaoka G, Ohara-Waki F, Imataki O, et al. Gab1 transduces PI3K-mediated erythropoietin signals to the Erk pathway and regulates erythropoietin-dependent proliferation and survival of erythroid cells. Cell Signal. 2009;21:1775–1783. doi: 10.1016/j.cellsig.2009.07.013. [DOI] [PubMed] [Google Scholar]
  46. Gauvreau GM, Watson RM, O'Byrne PM. Protective effects of inhaled PGE2 on allergen-induced airway responses and airway inflammation. Am J Respir Crit Care Med. 1999;159:31–36. doi: 10.1164/ajrccm.159.1.9804030. [DOI] [PubMed] [Google Scholar]
  47. Gosens R, Rieks D, Meurs H, Ninaber DK, Rabe KF, Nanninga J, et al. Muscarinic M3 receptor stimulation increases cigarette smoke-induced IL-8 secretion by human airway smooth muscle cells. Eur Respir J. 2009;34:1436–1443. doi: 10.1183/09031936.00045209. [DOI] [PubMed] [Google Scholar]
  48. Govindaraju V, Michoud MC, Al-Chalabi M, Ferraro P, Powell WS, Martin JG. Interleukin-8: novel roles in human airway smooth muscle cell contraction and migration. Am J Physiol Cell Physiol. 2006;291:C957–C965. doi: 10.1152/ajpcell.00451.2005. [DOI] [PubMed] [Google Scholar]
  49. Govindaraju V, Michoud MC, Ferraro P, Arkinson J, Safka K, Valderrama-Carvajal H, et al. The effects of interleukin-8 on airway smooth muscle contraction in cystic fibrosis. Respir Res. 2008;9:76. doi: 10.1186/1465-9921-9-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. 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. doi: 10.1067/mai.2001.120276. [DOI] [PubMed] [Google Scholar]
  51. Grunstein MM, Hakonarson H, Leiter J, Chen M, Whelan R, Grunstein JS, et al. IL-13-dependent autocrine signaling mediates altered responsiveness of IgE-sensitized airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2002;282:L520–L528. doi: 10.1152/ajplung.00343.2001. [DOI] [PubMed] [Google Scholar]
  52. Gueders MM, Foidart JM, Noel A, Cataldo DD. Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs in the respiratory tract: potential implications in asthma and other lung diseases. Eur J Pharmacol. 2006;533:133–144. doi: 10.1016/j.ejphar.2005.12.082. [DOI] [PubMed] [Google Scholar]
  53. Gueders MM, Hirst SJ, Quesada-Calvo F, Paulissen G, Hacha J, Gilles C, et al. Matrix metalloproteinase-19 deficiency promotes tenascin-C accumulation and allergen-induced airway inflammation. Am J Respir Cell Mol Biol. 2010;43:286–295. doi: 10.1165/rcmb.2008-0426OC. [DOI] [PubMed] [Google Scholar]
  54. Hackett TL, Holloway R, Holgate ST, Warner JA. Dynamics of pro-inflammatory and anti-inflammatory cytokine release during acute inflammation in chronic obstructive pulmonary disease: an ex vivo study. Respir Res. 2008;9:47. doi: 10.1186/1465-9921-9-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hakonarson H, Maskeri N, Carter C, Chuang S, Grunstein MM. Autocrine interaction between IL-5 and IL-1beta mediates altered responsiveness of atopic asthmatic sensitized airway smooth muscle. J Clin Invest. 1999;104:657–667. doi: 10.1172/JCI7137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hakonarson H, Kim C, Whelan R, Campbell D, Grunstein MM. Bi-directional activation between human airway smooth muscle cells and T lymphocytes: role in induction of altered airway responsiveness. J Immunol. 2001;166:293–303. doi: 10.4049/jimmunol.166.1.293. [DOI] [PubMed] [Google Scholar]
  57. Halayko AJ, Tran T, Ji SY, Yamasaki A, Gosens R. Airway smooth muscle phenotype and function: interactions with current asthma therapies. Curr Drug Targets. 2006;7:525–540. doi: 10.2174/138945006776818728. [DOI] [PubMed] [Google Scholar]
  58. Halayko AJ, Tran T, Gosens R. Phenotype and functional plasticity of airway smooth muscle: role of caveolae and caveolins. Proc Am Thorac Soc. 2008;5:80–88. doi: 10.1513/pats.200705-057VS. [DOI] [PubMed] [Google Scholar]
  59. Hallstrand TS, Henderson WR., Jr An update on the role of leukotrienes in asthma. Curr Opin Allergy Clin Immunol. 2010;10:60–66. doi: 10.1097/ACI.0b013e32833489c3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Hasaneen NA, Zucker S, Cao J, Chiarelli C, Panettieri RA, Foda HD. Cyclic mechanical strain-induced proliferation and migration of human airway smooth muscle cells: role of EMMPRIN and MMPs. FASEB J. 2005;19:1507–1509. doi: 10.1096/fj.04-3350fje. [DOI] [PubMed] [Google Scholar]
  61. Hedges JC, Singer CA, Gerthoffer WT. Mitogen-activated protein kinases regulate cytokine gene expression in human airway myocytes. Am J Respir Cell Mol Biol. 2000;23:86–94. doi: 10.1165/ajrcmb.23.1.4014. [DOI] [PubMed] [Google Scholar]
  62. Henderson WR, Jr, Tang LO, Chu SJ, Tsao SM, Chiang GK, Jones F, et al. A role for cysteinyl leukotrienes in airway remodeling in a mouse asthma model. Am J Respir Crit Care Med. 2002;165:108–116. doi: 10.1164/ajrccm.165.1.2105051. [DOI] [PubMed] [Google Scholar]
  63. Henderson N, Markwick LJ, Elshaw SR, Freyer AM, Knox AJ, Johnson SR. Collagen I and thrombin activate MMP-2 by MMP-14-dependent and -independent pathways: implications for airway smooth muscle migration. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1030–L1038. doi: 10.1152/ajplung.00317.2006. [DOI] [PubMed] [Google Scholar]
  64. Henness S, Johnson CK, Ge Q, Armour CL, Hughes JM, Ammit AJ. IL-17A augments TNF-alpha-induced IL-6 expression in airway smooth muscle by enhancing mRNA stability. J Allergy Clin Immunol. 2004;114:958–964. doi: 10.1016/j.jaci.2004.06.023. [DOI] [PubMed] [Google Scholar]
  65. Hershenson MB, Brown M, Camoretti-Mercado B, Solway J. Airway smooth muscle in asthma. Annu Rev Pathol. 2008;3:523–555. doi: 10.1146/annurev.pathmechdis.1.110304.100213. [DOI] [PubMed] [Google Scholar]
  66. Heximer SP, Knutsen RH, Sun X, Kaltenbronn KM, Rhee MH, Peng N, et al. Hypertension and prolonged vasoconstrictor signaling in RGS2-deficient mice. J Clin Invest. 2003;111:445–452. doi: 10.1172/JCI15598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hirota JA, Nguyen TT, Schaafsma D, Sharma P, Tran T. Airway smooth muscle in asthma: phenotype plasticity and function. Pulm Pharmacol Ther. 2009;22:370–378. doi: 10.1016/j.pupt.2008.12.004. [DOI] [PubMed] [Google Scholar]
  68. Hirst SJ. Regulation of airway smooth muscle cell immunomodulatory function: role in asthma. Respir Physiol Neurobiol. 2003;137:309–326. doi: 10.1016/s1569-9048(03)00155-1. [DOI] [PubMed] [Google Scholar]
  69. Hirst SJ, Hallsworth MP, Peng Q, Lee TH. Selective induction of eotaxin release by interleukin-13 or interleukin-4 in human airway smooth muscle cells is synergistic with interleukin-1beta and is mediated by the interleukin-4 receptor alpha-chain. Am J Respir Crit Care Med. 2002;165:1161–1171. doi: 10.1164/ajrccm.165.8.2107158. [DOI] [PubMed] [Google Scholar]
  70. Hollinger S, Hepler JR. Cellular regulation of RGS proteins: modulators and integrators of G protein signaling. Pharmacol Rev. 2002;54:527–559. doi: 10.1124/pr.54.3.527. [DOI] [PubMed] [Google Scholar]
  71. Howarth PH, Babu KS, Arshad HS, Lau L, Buckley M, McConnell W, et al. Tumour necrosis factor (TNFalpha) as a novel therapeutic target in symptomatic corticosteroid dependent asthma. Thorax. 2005;60:1012–1018. doi: 10.1136/thx.2005.045260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hughes JM, Arthur CA, Baracho S, Carlin SM, Hawker KM, Johnson PR, et al. Human eosinophil-airway smooth muscle cell interactions. Mediators Inflamm. 2000;9:93–99. doi: 10.1080/096293500411550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Issa R, Xie S, Lee KY, Stanbridge RD, Bhavsar P, Sukkar MB, et al. GRO-alpha regulation in airway smooth muscle by IL-1beta and TNF-alpha: role of NF-kappaB and MAP kinases. Am J Physiol Lung Cell Mol Physiol. 2006;291:L66–L74. doi: 10.1152/ajplung.00384.2005. [DOI] [PubMed] [Google Scholar]
  74. Issa R, Sorrentino R, Sukkar MB, Sriskandan S, Chung KF, Mitchell JA. Differential regulation of CCL-11/eotaxin-1 and CXCL-8/IL-8 by gram-positive and gram-negative bacteria in human airway smooth muscle cells. Respir Res. 2008;9:30. doi: 10.1186/1465-9921-9-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Ito I, Fixman ED, Asai K, Yoshida M, Gounni AS, Martin JG, et al. Platelet-derived growth factor and transforming growth factor-beta modulate the expression of matrix metalloproteinases and migratory function of human airway smooth muscle cells. Clin Exp Allergy. 2009;39:1370–1380. doi: 10.1111/j.1365-2222.2009.03293.x. [DOI] [PubMed] [Google Scholar]
  76. Iwata S, Ito S, Iwaki M, Kondo M, Sashio T, Takeda N, et al. Regulation of endothelin-1-induced interleukin-6 production by Ca2+ influx in human airway smooth muscle cells. Eur J Pharmacol. 2009;605:15–22. doi: 10.1016/j.ejphar.2008.12.045. [DOI] [PubMed] [Google Scholar]
  77. Jarai G, Sukkar M, Garrett S, Duroudier N, Westwick J, Adcock I, et al. Effects of interleukin-1beta, interleukin-13 and transforming growth factor-beta on gene expression in human airway smooth muscle using gene microarrays. Eur J Pharmacol. 2004;497:255–265. doi: 10.1016/j.ejphar.2004.06.055. [DOI] [PubMed] [Google Scholar]
  78. John AE, Zhu YM, Brightling CE, Pang L, Knox AJ. Human airway smooth muscle cells from asthmatic individuals have CXCL8 hypersecretion due to increased NF-kappa B p65, C/EBP beta, and RNA polymerase II binding to the CXCL8 promoter. J Immunol. 2009;183:4682–4692. doi: 10.4049/jimmunol.0803832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Johnson PR. Role of human airway smooth muscle in altered extracellular matrix production in asthma. Clin Exp Pharmacol Physiol. 2001;28:233–236. doi: 10.1046/j.1440-1681.2001.03426.x. [DOI] [PubMed] [Google Scholar]
  80. Kaur M, Holden NS, Wilson SM, Sukkar MB, Chung KF, Barnes PJ, et al. Effect of beta2-adrenoceptor agonists and other cAMP-elevating agents on inflammatory gene expression in human ASM cells: a role for protein kinase A. Am J Physiol Lung Cell Mol Physiol. 2008;295:L505–L514. doi: 10.1152/ajplung.00046.2008. [DOI] [PubMed] [Google Scholar]
  81. Kavanaugh WM, Williams LT, Ives HE, Coughlin SR. Serotonin-induced deoxyribonucleic acid synthesis in vascular smooth muscle cells involves a novel, pertussis toxin-sensitive pathway. Mol Endocrinol. 1988;2:599–605. doi: 10.1210/mend-2-7-599. [DOI] [PubMed] [Google Scholar]
  82. Kelly M, Hwang JM, Kubes P. Modulating leukocyte recruitment in inflammation. J Allergy Clin Immunol. 2007;120:3–10. doi: 10.1016/j.jaci.2007.05.017. [DOI] [PubMed] [Google Scholar]
  83. Kim DC, Hsu FI, Barrett NA, Friend DS, Grenningloh R, Ho IC, et al. Cysteinyl leukotrienes regulate Th2 cell-dependent pulmonary inflammation. J Immunol. 2006;176:4440–4448. doi: 10.4049/jimmunol.176.7.4440. [DOI] [PubMed] [Google Scholar]
  84. Kirstein F, Horsnell WG, Kuperman DA, Huang X, Erle DJ, Lopata AL, et al. Expression of IL-4 receptor alpha on smooth muscle cells is not necessary for development of experimental allergic asthma. J Allergy Clin Immunol. 2010;126:347–354. doi: 10.1016/j.jaci.2010.04.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Knobloch J, Peters H, Jungck D, Muller K, Strauch J, Koch A. TNFalpha-induced GM-CSF release from human airway smooth muscle cells depends on activation of an ET-1 autoregulatory positive feedback mechanism. Thorax. 2009;64:1044–1052. doi: 10.1136/thx.2008.111047. [DOI] [PubMed] [Google Scholar]
  86. Komuro I, Kurihara H, Sugiyama T, Yoshizumi M, Takaku F, Yazaki Y. Endothelin stimulates c-fos and c-myc expression and proliferation of vascular smooth muscle cells. FEBS Lett. 1988;238:249–252. doi: 10.1016/0014-5793(88)80489-7. [DOI] [PubMed] [Google Scholar]
  87. Kong KC, Billington CK, Gandhi U, Panettieri RA, Jr, Penn RB. Cooperative mitogenic signaling by G protein-coupled receptors and growth factors is dependent on G(q/11) FASEB J. 2006;20:1558–1560. doi: 10.1096/fj.05-5622fje. [DOI] [PubMed] [Google Scholar]
  88. Krymskaya VP, Orsini MJ, Eszterhas AJ, Brodbeck KC, Benovic JL, Panettieri RA, 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;23:546–554. doi: 10.1165/ajrcmb.23.4.4115. [DOI] [PubMed] [Google Scholar]
  89. Kunikata T, Yamane H, Segi E, Matsuoka T, Sugimoto Y, Tanaka S, et al. Suppression of allergic inflammation by the prostaglandin E receptor subtype EP3. Nat Immunol. 2005;6:524–531. doi: 10.1038/ni1188. [DOI] [PubMed] [Google Scholar]
  90. Lagente V, Boichot E. Role of matrix metalloproteinases in the inflammatory process of respiratory diseases. J Mol Cell Cardiol. 2010;48:440–444. doi: 10.1016/j.yjmcc.2009.09.017. [DOI] [PubMed] [Google Scholar]
  91. Lazaar AL, Panettieri RA., Jr Airway smooth muscle: a modulator of airway remodeling in asthma. J Allergy Clin Immunol. 2005;116:488–495. doi: 10.1016/j.jaci.2005.06.030. quiz 496. [DOI] [PubMed] [Google Scholar]
  92. Lazaar AL, Albelda SM, Pilewski JM, Brennan B, Pure E, Panettieri RA., Jr T lymphocytes adhere to airway smooth muscle cells via integrins and CD44 and induce smooth muscle cell DNA synthesis. J Exp Med. 1994;180:807–816. doi: 10.1084/jem.180.3.807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lee KS, Park SJ, Kim SR, Min KH, Jin SM, Lee HK, et al. Modulation of airway remodeling and airway inflammation by peroxisome proliferator-activated receptor gamma in a murine model of toluene diisocyanate-induced asthma. J Immunol. 2006;177:5248–5257. doi: 10.4049/jimmunol.177.8.5248. [DOI] [PubMed] [Google Scholar]
  94. Lex C, Zacharasiewicz A, Payne DN, Wilson NM, Nicholson AG, Kharitonov SA, et al. Exhaled breath condensate cysteinyl leukotrienes and airway remodeling in childhood asthma: a pilot study. Respir Res. 2006;7:63. doi: 10.1186/1465-9921-7-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Liang G, Bansal G, Xie Z, Druey KM. RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase signaling. J Biol Chem. 2009;284:21719–21727. doi: 10.1074/jbc.M109.028407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Malavia NK, Raub CB, Mahon SB, Brenner M, Panettieri RA, Jr, George SC. Airway epithelium stimulates smooth muscle proliferation. Am J Respir Cell Mol Biol. 2009;41:297–304. doi: 10.1165/rcmb.2008-0358OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Margulis A, Nocka KH, Brennan AM, Deng B, Fleming M, Goldman SJ, et al. Mast cell-dependent contraction of human airway smooth muscle cell-containing collagen gels: influence of cytokines, matrix metalloproteases, and serine proteases. J Immunol. 2009;183:1739–1750. doi: 10.4049/jimmunol.0803951. [DOI] [PubMed] [Google Scholar]
  98. 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;23:121–128. doi: 10.1016/j.pupt.2009.10.007. [DOI] [PubMed] [Google Scholar]
  99. McKay S, Sharma HS. Autocrine regulation of asthmatic airway inflammation: role of airway smooth muscle. Respir Res. 2002;3:11. doi: 10.1186/rr160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Mehrotra AK, Henderson WR., Jr The role of leukotrienes in airway remodeling. Curr Mol Med. 2009;9:383–391. doi: 10.2174/156652409787847209. [DOI] [PubMed] [Google Scholar]
  101. Misior AM, Deshpande DA, Loza MJ, Pascual RM, Hipp JD, Penn RB. Glucocorticoid- and protein kinase A-dependent transcriptome regulation in airway smooth muscle. Am J Respir Cell Mol Biol. 2009;41:24–39. doi: 10.1165/rcmb.2008-0266OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Montuschi P, Mondino C, Koch P, Barnes PJ, Ciabattoni G. Effects of a leukotriene receptor antagonist on exhaled leukotriene E4 and prostanoids in children with asthma. J Allergy Clin Immunol. 2006;118:347–353. doi: 10.1016/j.jaci.2006.04.010. [DOI] [PubMed] [Google Scholar]
  103. Moore WC, Peters SP. Severe asthma: an overview. J Allergy Clin Immunol. 2006;117:487–494. doi: 10.1016/j.jaci.2006.01.033. quiz 495. [DOI] [PubMed] [Google Scholar]
  104. Morris GE, Whyte MK, Martin GF, Jose PJ, Dower SK, Sabroe I. Agonists of toll-like receptors 2 and 4 activate airway smooth muscle via mononuclear leukocytes. Am J Respir Crit Care Med. 2005;171:814–822. doi: 10.1164/rccm.200403-406OC. [DOI] [PubMed] [Google Scholar]
  105. Mullan CS, Riley M, Clarke D, Tatler A, Sutcliffe A, Knox AJ, et al. Beta-tryptase regulates IL-8 expression in airway smooth muscle cells by a PAR-2-independent mechanism. Am J Respir Cell Mol Biol. 2008;38:600–608. doi: 10.1165/rcmb.2007-0082OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. 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;39:475–481. doi: 10.1165/rcmb.2007-0091OC. [DOI] [PubMed] [Google Scholar]
  107. Nath P, Leung SY, Williams AS, Noble A, Xie S, McKenzie AN, et al. Complete inhibition of allergic airway inflammation and remodelling in quadruple IL-4/5/9/13-mice. Clin Exp Allergy. 2007;37:1427–1435. doi: 10.1111/j.1365-2222.2007.02789.x. [DOI] [PubMed] [Google Scholar]
  108. Naureckas ET, Ndukwu IM, Halayko AJ, Maxwell C, Hershenson MB, Solway J. Bronchoalveolar lavage fluid from asthmatic subjects is mitogenic for human airway smooth muscle. Am J Respir Crit Care Med. 1999;160:2062–2066. doi: 10.1164/ajrccm.160.6.9903131. [DOI] [PubMed] [Google Scholar]
  109. Neubig RR. And the winner is . . . RGS4. Circ Res. 2008;103:444–446. doi: 10.1161/CIRCRESAHA.108.183384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Noveral JP, Rosenberg SM, Anbar RA, Pawlowski NA, Grunstein MM. Role of endothelin-1 in regulating proliferation of cultured rabbit airway smooth muscle cells. Am J Physiol. 1992;263(3 Pt 1):L317–L324. doi: 10.1152/ajplung.1992.263.3.L317. [DOI] [PubMed] [Google Scholar]
  111. Oenema TA, Kolahian S, Nanninga JE, Rieks D, Hiemstra PS, Zuyderduyn S, et al. Pro-inflammatory mechanisms of muscarinic receptor stimulation in airway smooth muscle. Respir Res. 2010;11:130. doi: 10.1186/1465-9921-11-130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Ohta Y, Hayashi M, Kanemaru T, Abe K, Ito Y, Oike M. Dual modulation of airway smooth muscle contraction by Th2 cytokines via matrix metalloproteinase-1 production. J Immunol. 2008;180:4191–4199. doi: 10.4049/jimmunol.180.6.4191. [DOI] [PubMed] [Google Scholar]
  113. Oliver BG, Johnston SL, Baraket M, Burgess JK, King NJ, Roth M, et al. Increased proinflammatory responses from asthmatic human airway smooth muscle cells in response to rhinovirus infection. Respir Res. 2006;7:71. doi: 10.1186/1465-9921-7-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Oltmanns U, Chung KF, Walters M, John M, Mitchell JA. Cigarette smoke induces IL-8, but inhibits eotaxin and RANTES release from airway smooth muscle. Respir Res. 2005;6:74. doi: 10.1186/1465-9921-6-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Owen VJ, Burton PB, Mullen AJ, Birks EJ, Barton P, Yacoub MH. Expression of RGS3, RGS4 and Gi alpha 2 in acutely failing donor hearts and end-stage heart failure. Eur Heart J. 2001;22:1015–1020. doi: 10.1053/euhj.2000.2578. [DOI] [PubMed] [Google Scholar]
  116. Page K, Li J, Hershenson MB. Platelet-derived growth factor stimulation of mitogen-activated protein kinases and cyclin D1 promoter activity in cultured airway smooth-muscle cells. Role Ras Am J Respir Cell Mol Biol. 1999;20:1294–1302. doi: 10.1165/ajrcmb.20.6.3597. [DOI] [PubMed] [Google Scholar]
  117. Panettieri RA, Jr, Hall IP, Maki CS, Murray RK. alpha-Thrombin increases cytosolic calcium and induces human airway smooth muscle cell proliferation. Am J Respir Cell Mol Biol. 1995;13:205–216. doi: 10.1165/ajrcmb.13.2.7626288. [DOI] [PubMed] [Google Scholar]
  118. Panettieri RA, Jr, Goldie RG, Rigby PJ, Eszterhas AJ, Hay DW. Endothelin-1-induced potentiation of human airway smooth muscle proliferation: an ETA receptor-mediated phenomenon. Br J Pharmacol. 1996;118:191–197. doi: 10.1111/j.1476-5381.1996.tb15385.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Pang L, Knox AJ. Bradykinin stimulates IL-8 production in cultured human airway smooth muscle cells: role of cyclooxygenase products. J Immunol. 1998;161:2509–2515. [PubMed] [Google Scholar]
  120. Peng Q, Matsuda T, Hirst SJ. Signaling pathways regulating interleukin-13-stimulated chemokine release from airway smooth muscle. Am J Respir Crit Care Med. 2004;169:596–603. doi: 10.1164/rccm.200307-888OC. [DOI] [PubMed] [Google Scholar]
  121. Phipps S, Lam CE, Foster PS, Matthaei KI. The contribution of toll-like receptors to the pathogenesis of asthma. Immunol Cell Biol. 2007;85:463–470. doi: 10.1038/sj.icb.7100104. [DOI] [PubMed] [Google Scholar]
  122. Prefontaine D, Lajoie-Kadoch S, Foley S, Audusseau S, Olivenstein R, Halayko AJ, et al. Increased expression of IL-33 in severe asthma: evidence of expression by airway smooth muscle cells. J Immunol. 2009;183:5094–5103. doi: 10.4049/jimmunol.0802387. [DOI] [PubMed] [Google Scholar]
  123. Pype JL, Dupont LJ, Menten P, Van Coillie E, Opdenakker G, Van Damme J, et al. Expression of monocyte chemotactic protein (MCP)-1, MCP-2, and MCP-3 by human airway smooth-muscle cells. Modulation by corticosteroids and T-helper 2 cytokines. Am J Respir Cell Mol Biol. 1999;21:528–536. doi: 10.1165/ajrcmb.21.4.3660. [DOI] [PubMed] [Google Scholar]
  124. Pype JL, Xu H, Schuermans M, Dupont LJ, Wuyts W, Mak JC, et al. Mechanisms of interleukin 1beta-induced human airway smooth muscle hyporesponsiveness to histamine. Involvement of p38 MAPK NF-kappaB. Am J Respir Crit Care Med. 2001;163:1010–1017. doi: 10.1164/ajrccm.163.4.9911091. [DOI] [PubMed] [Google Scholar]
  125. Rajah R, Nunn SE, Herrick DJ, Grunstein MM, Cohen P. Leukotriene D4 induces MMP-1, which functions as an IGFBP protease in human airway smooth muscle cells. Am J Physiol. 1996;271(6 Pt 1):L1014–L1022. doi: 10.1152/ajplung.1996.271.6.L1014. [DOI] [PubMed] [Google Scholar]
  126. Rajah R, Nachajon RV, Collins MH, Hakonarson H, Grunstein MM, Cohen P. Elevated levels of the IGF-binding protein protease MMP-1 in asthmatic airway smooth muscle. Am J Respir Cell Mol Biol. 1999;20:199–208. doi: 10.1165/ajrcmb.20.2.3148. [DOI] [PubMed] [Google Scholar]
  127. Ramos-Barbón D, Presley JF, Hamid QA, Fixman ED, Martin JG. Antigen-specific CD4+ T cells drive airway smooth muscle remodeling in experimental asthma. J Clin Invest. 2005;115:1580–1589. doi: 10.1172/JCI19711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Redhu NS, Saleh A, Shan L, Gerthoffer WT, Kung SK, Halayko AJ, et al. Proinflammatory and Th2 cytokines regulate the high affinity IgE receptor (FcepsilonRI) and IgE-dependant activation of human airway smooth muscle cells. Plos One. 2009;4:e6153. doi: 10.1371/journal.pone.0006153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Ritter SL, Hall RA. Fine-tuning of GPCR activity by receptor-interacting proteins. Nat Rev Mol Cell Biol. 2009;10:819–830. doi: 10.1038/nrm2803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Roche N, Chinet TC, Huchon GJ. Allergic and nonallergic interactions between house dust mite allergens and airway mucosa. Eur Respir J. 1997;10:719–726. [PubMed] [Google Scholar]
  131. Rolin S, Masereel B, Dogne JM. Prostanoids as pharmacological targets in COPD and asthma. Eur J Pharmacol. 2006;533:89–100. doi: 10.1016/j.ejphar.2005.12.058. [DOI] [PubMed] [Google Scholar]
  132. Roscioni SS, Kistemaker LE, Menzen MH, Elzinga CR, Gosens R, Halayko AJ, et al. PKA and Epac cooperate to augment bradykinin-induced interleukin-8 release from human airway smooth muscle cells. Respir Res. 2009;10:88. doi: 10.1186/1465-9921-10-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Russo C, Polosa R. TNF-alpha as a promising therapeutic target in chronic asthma: a lesson from rheumatoid arthritis. Clin Sci (Lond) 2005;109:135–142. doi: 10.1042/CS20050038. [DOI] [PubMed] [Google Scholar]
  134. Salmon M, Walsh DA, Huang TJ, Barnes PJ, Leonard TB, Hay DW, et al. Involvement of cysteinyl leukotrienes in airway smooth muscle cell DNA synthesis after repeated allergen exposure in sensitized Brown Norway rats. Br J Pharmacol. 1999;127:1151–1158. doi: 10.1038/sj.bjp.0702669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Scott PH, Belham CM, al-Hafidh J, Chilvers ER, Peacock AJ, Gould GW, et al. A regulatory role for cAMP in phosphatidylinositol 3-kinase/p70 ribosomal S6 kinase-mediated DNA synthesis in platelet-derived-growth-factor-stimulated bovine airway smooth-muscle cells. Biochem J. 1996;318:965–971. doi: 10.1042/bj3180965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Sestini P, Armetti L, Gambaro G, Pieroni MG, Refini RM, Sala A, et al. Inhaled PGE2 prevents aspirin-induced bronchoconstriction and urinary LTE4 excretion in aspirin-sensitive asthma. Am J Respir Crit Care Med. 1996;153:572–575. doi: 10.1164/ajrccm.153.2.8564100. [DOI] [PubMed] [Google Scholar]
  137. Sethakorn N, Yau DM, Dulin NO. Non-canonical functions of RGS proteins. Cell Signal. 2010;22:1274–1281. doi: 10.1016/j.cellsig.2010.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Shan X, Hu A, Veler H, Fatma S, Grunstein JS, Chuang S, et al. Regulation of Toll-like receptor 4-induced proasthmatic changes in airway smooth muscle function by opposing actions of ERK1/2 and p38 MAPK signaling. Am J Physiol Lung Cell Mol Physiol. 2006;291:L324–L333. doi: 10.1152/ajplung.00056.2006. [DOI] [PubMed] [Google Scholar]
  139. 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;184:7134–7143. doi: 10.4049/jimmunol.0902515. [DOI] [PubMed] [Google Scholar]
  140. Sharma P, Tran T, Stelmack GL, McNeill K, Gosens R, Mutawe MM, et al. Expression of the dystrophin-glycoprotein complex is a marker for human airway smooth muscle phenotype maturation. Am J Physiol Lung Cell Mol Physiol. 2008;294:L57–L68. doi: 10.1152/ajplung.00378.2007. [DOI] [PubMed] [Google Scholar]
  141. Shore SA, Laporte J, Hall IP, Hardy E, Panettieri RA., Jr Effect of IL-1 beta on responses of cultured human airway smooth muscle cells to bronchodilator agonists. Am J Respir Cell Mol Biol. 1997;16:702–712. doi: 10.1165/ajrcmb.16.6.9191472. [DOI] [PubMed] [Google Scholar]
  142. Siddiqui S, Mistry V, Doe C, Stinson S, Foster M, Brightling C. Airway wall expression of OX40/OX40L and interleukin-4 in asthma. Chest. 2010;137:797–804. doi: 10.1378/chest.09-1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Simeone-Penney MC, Severgnini M, Rozo L, Takahashi S, Cochran BH, Simon AR. PDGF-induced human airway smooth muscle cell proliferation requires STAT3 and the small GTPase Rac1. Am J Physiol Lung Cell Mol Physiol. 2008;294:L698–L704. doi: 10.1152/ajplung.00529.2007. [DOI] [PubMed] [Google Scholar]
  144. Singh RK, Gupta S, Dastidar S, Ray A. Cysteinyl leukotrienes and their receptors: molecular and functional characteristics. Pharmacology. 2010;85:336–349. doi: 10.1159/000312669. [DOI] [PubMed] [Google Scholar]
  145. Slats AM, Janssen K, van Schadewijk A, van der Plas DT, Schot R, van den Aardweg JG, et al. Bronchial inflammation and airway responses to deep inspiration in asthma and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;176:121–128. doi: 10.1164/rccm.200612-1814OC. [DOI] [PubMed] [Google Scholar]
  146. Sukkar MB, Xie S, Khorasani NM, Kon OM, Stanbridge R, Issa R, et al. Toll-like receptor 2, 3, and 4 expression and function in human airway smooth muscle. J Allergy Clin Immunol. 2006;118:641–648. doi: 10.1016/j.jaci.2006.05.013. [DOI] [PubMed] [Google Scholar]
  147. Thomas PS, Heywood G. Effects of inhaled tumour necrosis factor alpha in subjects with mild asthma. Thorax. 2002;57:774–778. doi: 10.1136/thorax.57.9.774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Tliba O, Panettieri RA., Jr Noncontractile functions of airway smooth muscle cells in asthma. Annu Rev Physiol. 2009;71:509–535. doi: 10.1146/annurev.physiol.010908.163227. [DOI] [PubMed] [Google Scholar]
  149. Tliba O, Tliba S, Da Huang C, Hoffman RK, DeLong P, Panettieri RA, et al. Tumor necrosis factor alpha modulates airway smooth muscle function via the autocrine action of interferon beta. J Biol Chem. 2003;278:50615–50623. doi: 10.1074/jbc.M303680200. [DOI] [PubMed] [Google Scholar]
  150. Tliba O, Amrani Y, Panettieri RA., Jr Is airway smooth muscle the ‘missing link’ modulating airway inflammation in asthma. Chest. 2008;133:236–242. doi: 10.1378/chest.07-0262. [DOI] [PubMed] [Google Scholar]
  151. Vichi P, Whelchel A, Knot H, Nelson M, Kolch W, Posada J. Endothelin-stimulated ERK activation in airway smooth-muscle cells requires calcium influx and Raf activation. Am J Respir Cell Mol Biol. 1999;20:99–105. doi: 10.1165/ajrcmb.20.1.3210. [DOI] [PubMed] [Google Scholar]
  152. Wang SZ, Xu H, Wraith A, Bowden JJ, Alpers JH, Forsyth KD. Neutrophils induce damage to respiratory epithelial cells infected with respiratory syncytial virus. Eur Respir J. 1998;12:612–618. doi: 10.1183/09031936.98.12030612. [DOI] [PubMed] [Google Scholar]
  153. Whelan R, Kim C, Chen M, Leiter J, Grunstein MM, Hakonarson H. Role and regulation of interleukin-1 molecules in pro-asthmatic sensitised airway smooth muscle. Eur Respir J. 2004;24:559–567. doi: 10.1183/09031936.04.00133803. [DOI] [PubMed] [Google Scholar]
  154. Wollin L, Pieper MP. Tiotropium bromide exerts anti-inflammatory activity in a cigarette smoke mouse model of COPD. Pulm Pharmacol Ther. 2010;23:345–354. doi: 10.1016/j.pupt.2010.03.008. [DOI] [PubMed] [Google Scholar]
  155. Xie S, Issa R, Sukkar MB, Oltmanns U, Bhavsar PK, Papi A, et al. Induction and regulation of matrix metalloproteinase-12 in human airway smooth muscle cells. Respir Res. 2005;6:148. doi: 10.1186/1465-9921-6-148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Yamasaki A, Saleh A, Koussih L, Muro S, Halayko AJ, Gounni AS. IL-9 induces CCL11 expression via STAT3 signalling in human airway smooth muscle cells. Plos One. 2010;5:e9178. doi: 10.1371/journal.pone.0009178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Yang W, Kaur D, Okayama Y, Ito A, Wardlaw AJ, Brightling CE, et al. Human lung mast cells adhere to human airway smooth muscle, in part, via tumor suppressor in lung cancer-1. J Immunol. 2006;176:1238–1243. doi: 10.4049/jimmunol.176.2.1238. [DOI] [PubMed] [Google Scholar]
  158. Zerfaoui M, Suzuki Y, Naura AS, Hans CP, Nichols C, Boulares AH. Nuclear translocation of p65 NF-kappaB is sufficient for VCAM-1, but not ICAM-1, expression in TNF-stimulated smooth muscle cells: differential requirement for PARP-1 expression and interaction. Cell Signal. 2008;20:186–194. doi: 10.1016/j.cellsig.2007.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Zuberi RI, Hsu DK, Kalayci O, Chen HY, Sheldon HK, Yu L, et al. Critical role for galectin-3 in airway inflammation and bronchial hyperresponsiveness in a murine model of asthma. Am J Pathol. 2004;165:2045–2053. doi: 10.1016/S0002-9440(10)63255-5. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

bph0163-0068-SD1.pptx (150.2KB, pptx)

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES