Airway Structural Components Drive Airway Smooth ... - ATS Journals

89 downloads 0 Views 445KB Size Report
Jul 2, 2009 - Bart G. J. Dekkers1, Harm Maarsingh1, Herman Meurs1, and Reinoud ...... Black JL. Airway smooth muscle cell proliferation is increased in.
Airway Structural Components Drive Airway Smooth Muscle Remodeling in Asthma Bart G. J. Dekkers1, Harm Maarsingh1, Herman Meurs1, and Reinoud Gosens1 1

Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands

Chronic asthma is an inflammatory airways disease characterized by pathological changes in the airway smooth muscle (ASM) bundle that contribute to airway obstruction and hyperresponsiveness. Remodeling of the ASM is associated with an increased smooth muscle mass, involving components of cellular hypertrophy and hyperplasia, and changes in the phenotype of the muscle that facilitate proliferative, synthetic and contractile functions. These changes are considered major contributing factors to the pathophysiology of asthma, because of their role in exaggerated airway narrowing. The mechanisms that regulate changes in ASM mass and phenotype are incompletely understood, but likely involve the regulatory role of mediators and growth factors secreted from inflammatory cells on ASM cell proliferation and phenotype. An alternative hypothesis is that cellular and structural components that together constitute the airway wall, such as the airway epithelium, airway nerves, and the extracellular matrix, interact with the ASM bundle to facilitate changes in smooth muscle phenotype and function that drive remodeling under inflammatory conditions. This review discusses the mechanisms by which structural components of the airway wall communicate with the ASM bundle to regulate remodeling and discusses these mechanisms in the context of the pathophysiology of asthma. Keywords: airway remodeling; arginase; acetylcholine; anticholinergics; extracellular matrix

Asthma is a chronic disease of the airways, which is characterized by persistent airway inflammation, reversible airways obstruction, airway remodeling, and airway hyperresponsiveness (1). Airway hyperresponsiveness is defined by an exaggerated narrowing of the airways to a variety of chemical, physical, and pharmacological stimuli (2). Acute, variable airway hyperresponsiveness has been considered to reflect increased airway smooth muscle contraction associated with airway inflammation and is related to asthma activity and severity, whereas chronic persistent airway hyperresponsiveness may reflect airway remodeling (3). Airway remodeling is characterized by changes in the structure of the airway wall, which include shedding of the epithelium, goblet cell hyperplasia, increased blood vessel number and area, increased and changed deposition of extracellular matrix (ECM), and increased airway smooth muscle (ASM) mass (4–6). Airway remodeling, notably the abnormalities in the ASM that encircles the airways and regulates lumen diameter, may contribute to the pathogenesis and pathophysiology of asthma.

(Received in original form July 2, 2009; accepted in final form September 8, 2009) Supported by a Veni grant (916.86.036) from the Dutch Organisation for Scientific Research (NWO) (R.G.), a grant from the Netherlands Asthma Foundation (NAF 3.2.03.36) (B.G.J.D.), and a grant from Schering-Plough Research Institute (Oss, the Netherlands) (H.M.). Correspondence and requests for reprints should be addressed to Reinoud Gosens, Ph.D., Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1 9713 AV, Groningen, The Netherlands. E-mail: [email protected] Proc Am Thorac Soc Vol 6. pp 683–692, 2009 DOI: 10.1513/pats.200907-056DP Internet address: www.atsjournals.org

Several studies have indicated that differences in contractile responses exist between ASM cells derived from people with asthma and cells derived from healthy subjects, which could at least in part explain hyperresponsiveness in asthma. Thus, asthmatic ASM cells contract with greater velocity and maximum shortening capacity compared with healthy ASM (7). These changes may be explained by increases in the expression of smooth muscle myosin light chain kinase (sm-MLCK), transgelin (sm-22), and myosin heavy chain (sm-MHC) as reported in asthmatic ASM cells and in asthmatic biopsies (7, 8). Moreover, asthmatic ASM appears to express increased levels of the seven–amino acid insert SM-B isoform of smMHC, which shows a twofold greater ATPase activity and shortening velocity compared with the SM-A isoform (8, 9), although this latter finding is at odds with the study by Ma and colleagues who reported no expression of the SM-B isoform in smooth muscle obtained from subjects with asthma (7). The increase in contractile protein expression may be of clinical relevance as expression levels of sm-MHC, sm-a-actin, and desmin correlate with methacholine responsiveness in subjects with asthma (8, 10). Studies using asthmatic and nonasthmatic ASM cultured in collagen gels also showed that maximal histamine-induced condensation of the gel was increased when cells derived from subjects with asthma were used (11). In addition, ASM relaxation may also be changed as relaxation of passively sensitized ASM is slower compared with controls (12). Increased ASM mass is one of the most striking features of airway remodeling in asthma. Mathematical modeling studies on the impact of remodeling on airway narrowing indicated that increased ASM mass is likely to be the most important feature in increased airway narrowing in asthma, when assuming that the capacity of the ASM bundle to produce force is proportional to its mass (13, 14). This idea is underscored by the fact that patients with asthma in which the ASM layer has been reduced by bronchial thermoplasty show improved asthma control (15). Several studies have addressed the underlying pathology causing the increased ASM mass in asthma. Ebina and colleagues (16) examined the ASM layer surrounding the airway lumen in fatal asthma and found two different asthmatic phenotypes, one showing an increased number of ASM cells (hyperplasia) and the other showing an increased ASM cell size (hypertrophy). In subsequent studies, Woodruff and colleagues (17) found evidence for hyperplasia, but not hypertrophy, in the ASM layer of subjects with mild to moderate asthma, whereas Benayoun and colleagues (18) found ASM hypertrophy, but not hyperplasia, in patients with intermittent, mild-to-moderate and severe asthma. The latter group also found a clear correlation between disease severity and the degree of ASM thickening, consistent with a recent study showing that ASM thickening is more significant in fatal asthma as compared with nonfatal asthma (19). The relationship between age and duration of disease and ASM thickening is still subject to debate (18–21). Collectively, these findings suggest that increased ASM mass in asthma may reflect both cellular hyperplasia and hypertrophy, the degree of which depends primarily on asthma severity.

684

The increased ASM mass may be explained by intrinsic changes in the asthmatic ASM cells that facilitate their proliferative and secretory characteristics. Asthmatic ASM produces more proinflammatory, proangiogenic, and proremodeling factors, including eotaxin, vascular endothelial growth factor (VEGF), and connective tissue growth factor (CTGF) (22– 24), and fewer antimitogenic factors, such as PGE2 (25). Accordingly, asthmatic ASM cells in culture proliferate faster compared with healthy control cells (26), which is caused by changes in ECM protein deposition (27) and by enhanced mitochondrial biogenesis and mitochondrial activity that support increased cell growth (28). The exact mechanisms that regulate these responses are still incompletely identified; nonetheless these studies do highlight the importance of the ASM cell as an interactive player in the remodeling process rather than being the passive contractile partner as traditionally proposed.

REMODELING OF THE AIRWAY SMOOTH MUSCLE BUNDLE: MECHANISMS Although the mechanisms that regulate airway wall remodeling have thus far been incompletely identified, there is likely a major role for airway inflammation. Airway inflammation precedes airway remodeling in animal models of asthma (29, 30), and ASM is known to proliferate in response to numerous growth factors and mediators that are released during allergic airway inflammation both in vitro and in vivo (31). Nonetheless, both clinical and animal studies indicate that the relationship between inflammation and remodeling is complex, and still incompletely understood. The presence of airway inflammation in patients with asthma is no guarantee at all for the occurrence of airway remodeling, and there is no clear correlation between the degree of inflammation and the degree of remodeling (18). Also, components of remodeling, including smooth muscle thickening, appear to be present already in young children (32–34) and there is no clear relationship between age or duration of disease and the extent of ASM thickening (19). Furthermore, although airway inflammation can be resolved on allergen avoidance in a murine model, remodeling persists, suggesting that ongoing inflammation is not required to support

PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 6

2009

the maintenance of the remodeled airway wall (35). Collectively, although these studies point to an important, probably indispensable, role for airway inflammation in initiating or regulating the remodeling response, these studies also indicate that additional mechanisms exist in the airway wall that are necessary to direct or maintain the remodeling response. Moreover, these studies suggest that targeting inflammation per se may not be sufficient to reverse existing airway smooth muscle remodeling, a contention supported by studies showing that corticosteroid treatment prevents but does not reverse remodeling in allergen-challenged rats and mice (36, 37). In the next sections, we discuss recent findings that underscore the hypothesis that communication between different structural cells and compartments of the airway wall is central to the development of remodeling and may provide useful alternative drug targets for the treatment of smooth muscle remodeling. These mechanisms include communication between the ASM, the airway epithelium, the airway parasympathetic nervous system, and the ECM.

THE AIRWAY EPITHELIUM The airway epithelium forms the interface between the external environment and the airways (38). In asthma, the epithelial barrier is disrupted, which contributes to airway hyperresponsiveness and inflammation associated with this disease via increased release of proinflammatory cytokines. In addition, (damaged) epithelial cells in asthma release a number of growth factors, including epidermal growth factor (EGF), plateletderived growth factor (PDGF), transforming growth factor-b (TGF-b) and VEGF, as well as acetylcholine (see below), which may contribute to airway remodeling in asthma by inducing ASM growth, ECM deposition, and angiogenesis (38–41) (Figure 1). In this section, we focus on the possible contribution of other epithelial processes, particularly alterations in L-arginine homeostasis, to airway remodeling in asthma. Thus far, the role of L-arginine homeostasis in airway (patho)physiology has mostly been studied in the context of regulating airway (hyper)responsiveness. The epithelium is an important source of the bronchodilator nitric oxide (NO), which is produced by NO synthase (NOS) from the hydrolysis of

Figure 1. Putative role of altered L-arginine homeostasis in the airway epithelium in regulating remodeling of the airway smooth muscle (ASM) in asthma. The bioavailability of L-arginine to nitric oxide synthase (NOS) isoforms is decreased in asthma, leading to a deficiency of bronchodilating NO as well as increased formation of procontractile peroxynitrite (ONOO2) due to uncoupling of NOS. The NO deficiency may also contribute to the increased ASM mass in asthma, because NO is antiproliferative. Increased formation of peroxynitrite could also contribute by causing epithelial damage. Reduced L-arginine bioavailability is caused by at least two mechanisms: (1) inhibition of the cationic amino acid transporter by eosinophilic polycations, such as major basic protein (MBP), and (2) increased consumption of L-arginine by arginase, which is induced in asthma, presumably due to increased release of Th2-cytokines and growth factors. Increased arginase activity may directly contribute to the increased ASM mass via the production of polyamines and L-proline downstream from L-ornithine. See text for further detail.

Dekkers, Maarsingh, Meurs, et al.: Airway Smooth Muscle Remodeling

685

L-arginine (42). Three NOS isozymes have been identified: neuronal (nNOS), endothelial (eNOS), and inducible NOS (iNOS). In the airway epithelium, nNOS and eNOS are constitutively expressed, whereas iNOS is particularly induced by proinflammatory cytokines during the late asthmatic reaction (43, 44). The NO production is regulated by the substrate availability to NOS and alterations in the L-arginine homeostasis contribute to the pathophysiology of (acute) allergic asthma (45). Although levels of exhaled NO are elevated in subjects with asthma due the induction of iNOS (43, 46, 47), it has paradoxically been shown that a deficiency in bronchodilating (epithelium-derived) NO underlies the development of airway hyperresponsiveness in animal models of allergic asthma (48– 54) and in patients with asthma (55, 56). This NO deficiency is caused by a decreased bioavailability of L-arginine to NOS isozymes (49, 57, 58), which also leads to uncoupling of the oxidase and reductase moieties within the iNOS enzyme (50). Uncoupled iNOS not only produces NO but also superoxide, leading to an efficient formation of peroxynitrite (59) (Figure 1), which induces airway hyperresponsiveness, epithelial damage, mucus hypersecretion, and inflammation (60–63). A deficiency of NO could contribute to ASM thickening. It has been shown that NO inhibits mitogen-induced proliferation of cultured human (64–66) and guinea pig (67) ASM cells. Scavenging of superoxide anions, thereby increasing the levels of authentic NO and inhibiting peroxynitrite formation, also decreased mitogen-induced human ASM cell proliferation (66). The downstream mechanisms of NO-mediated inhibition of cell proliferation have been studied in more detail in vascular smooth muscle cells (VSMC) and involve cGMP-dependent repression of cell cycle promoting genes, including cyclin D1, and the induction of cell cycle inhibitors, such as p21Waf1/Cip1 (68). Also in VSMC, NO has been shown to inhibit the 26S proteosome, which regulates the degradation of cell cycle proteins, via S-nitrosylation (69), whereas cGMP inactivates p42/p44 MAPK and activates MAPK phosphatase 1 (68). Moreover, NO attenuates PDGF-induced activation of protein kinase B (PKB) and subsequent VSMC proliferation (70) and reduces embryonic fibroblast proliferation by inhibiting EGF receptor tyrosine kinase activity via S-nitrosylation (71, 72). Taken together, these findings suggest that a deficiency of (epithelium-derived) NO in asthma may also contribute to increased ASM mass in chronic asthma (Figure 1). An important mechanism contributing to the L-arginine limitation to NOS and subsequent NO deficiency in asthma is increased consumption of the amino acid by arginase, yielding L-ornithine and urea (Figure 1). Two arginase isoforms have been identified—the cytosolic arginase I and the mitochondrial arginase II—and both are expressed in the airway epithelium (73). Arginase activity and/or expression of particularly arginase I are increased in a number of animal models of acute allergic asthma (for review see Reference 74) and in human subjects with asthma (75–77). Increased arginase activity, which may involve Th2 cytokines (77, 78), importantly contributes to the development of allergen-induced bronchial obstructive reactions (58), airway hyperresponsiveness in vivo and ex vivo (49, 50, 58, 76, 79), and airway inflammation (58). The functional significance of increased arginase activity in asthma is reinforced by reduced L-arginine levels due to inhibition of cellular L-arginine transport by eosinophil-derived polycations (50, 80, 81) (Figure 1).The release of NO by inhibitory nonadrenergic, noncholinergic (iNANC) neurons is also regulated by endogenous arginase (82). Moreover, the allergen-induced increase in arginase activity attenuates iNANC nerve-mediated NO release and ASM relaxation by limiting the L-arginine bioavailability (49). Thus, in addition to reduced ASM relaxation, alterations

in L-arginine homeostasis in the iNANC nervous system may also contribute to airway remodeling via reduced synthesis of NO. Increased arginase activity could also contribute to the pathophysiology of allergic asthma via increased synthesis of L-ornithine (Figure 1). L-Ornithine is a precursor of the polyamines (putrescine, spermidine, and spermine) and L-proline, which are involved in cell proliferation and collagen synthesis, respectively (74, 81, 83–85). Polyamines induce the expression of genes involved in cell proliferation by promoting histone acetyltransferase activity and chromatin hyperacetylation (86), and polyamine synthesis is initiated by ornithine decarboxylase (ODC), which converts L-ornithine into putrescine (85). Both arginase and ODC are expressed in airway epithelial cells (87) and the expression and activation of both enzymes in the vasculature can be induced by growth factors, leading to increased polyamine levels (87–92). Growth factor–induced activation of ODC has also been observed in the airways (93). Interestingly, animal models demonstrate that arginase activity is increased in chronic asthma, underlying AHR in vivo (76) and ex vivo (94) by limiting the L-arginine availability to NOS. Moreover, elevated levels of polyamines have been detected in lungs of allergen-challenged mice (77) and in serum of patients with asthma (95). These findings suggest that increased arginase activity may also contribute to the increased ASM mass in asthma via increased polyamine production. In support, transfection of VSMC with arginase I leads to elevated polyamine levels and increased cell proliferation (96). Because NO inhibits ODC via S-nitrosylation (97), allergen-induced deficiency of NO may contribute to the elevated polyamine levels in asthma. In conclusion, altered L-arginine homeostasis due to increased arginase activity in the airway epithelium of asthmatics could contribute to airway remodeling via increased production of polyamines and L-proline downstream of L-ornithine as well as by limiting the substrate availability to NOS enzymes, leading to NO deficiency and enhanced peroxynitrite formation (Figure 1).

THE AIRWAY PARASYMPATHETIC NERVOUS SYSTEM The airway parasympathetic system, in which acetylcholine is the primary neurotransmitter, has long been recognized for its role in bronchoconstriction and mucus secretion (98, 99). However, recent studies in guinea pigs and mice have revealed that acetylcholine, by acting on muscarinic receptors, is involved in the regulation of ASM mass and phenotype, suggesting an important role of the airway cholinergic system in regulating responses associated with remodeling (100–102). Indeed, the regulation of neuronal release of acetylcholine appears to be highly facilitated by eosinophilic airway inflammation and acetylcholine appears to have postjunctional effects on ASM that could explain its action as a regulator of airway wall remodeling, notably thickening of the smooth muscle (103, 104) (Figure 2A). The release of acetylcholine from parasympathetic nerves is enhanced in allergic airway inflammation because several mechanisms exist that allow inflammatory mediators to activate the cholinergic system and because allergic airway inflammation facilitates the output of parasympathetic nerve endings (99, 104) (Figure 2A). Afferent sensory nerve fibers, or C-fibers, play an important role in this regard, as they can be triggered by a variety of inflammatory mediators and by nonspecific stimuli, such as cold air. This results in the local release of tachykinins as well as the activation of a cholinergic reflex mechanism that facilitates the output of the vagal nerve both centrally and

686

PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 6

2009

Figure 2. Interactions between the airway epithelium, the airway cholinergic system, and the airway smooth muscle (ASM) regulates remodeling of the ASM bundle. (A) Chronic airway inflammation facilitates acetylcholine release from the airway parasympathetic nerves, directly and via the activation of cholinergic reflex mechanisms, which are enhanced by the presence of damaged or stressed epithelium. (B) As a result, increased acetylcholine release, in combination with growth factors and mediators released during inflammation, coordinates cell responses in ASM associated with remodeling, including smooth muscle–specific gene expression and cell proliferation. The mechanisms responsible for these responses include phosphorylation of downstream signaling intermediates, including p70S6K and GSK-3, resulting from muscarinic M3receptor derived PI3K activation (via bg subunits) and PKC activation, respectively. See text for further detail.

locally in the airway parasympathetic ganglia (105, 106). Cfibers are exposed due to epithelial shedding in asthma and have receptors for histamine, prostanoids, thromboxane A2, bradykinin, serotonin, and tachykinins (107, 108). In conditions of damaged or stressed airway epithelium, the underlying mesenchyme can therefore be activated by this cholinergic reflex pathway, producing not only bronchoconstrictor responses but perhaps also responses associated with remodeling. The importance of this reflex mechanism is suggested by a recent study that showed that the majority of the bronchoconstrictor response to the inhaled thromboxane A2 mimetic U46619 is prevented by vagotomy or by administration of the M3 receptor selective ligand 4-DAMP (109). Furthermore, the airway hyperresponsiveness to inhaled histamine in ovalbumin-sensitized guinea pigs after the early asthmatic reaction is markedly reduced by inhaled ipratropium bromide, indicating increased regulation of the cholinergic reflex in allergic airways disease (110). Such studies indicate that inflammatory mediators use the airway cholinergic system to regulate a major part of their bronchoconstrictor response. In addition, several other mechanisms are at work in allergic airways disease that further induce the output of the vagal nerve. Inflammatory mediators, including tachykinins, prostaglandins, and thromboxane A2, facilitate neurotransmission in nerve endings and in the ganglia through effects on facilitatory prejunctional receptors (105, 106). In addition, the prejunctional muscarinic M2 receptor,

which limits acetylcholine release under physiological conditions, is dysfunctional during allergic airway inflammation (98, 111–113). Allergen-induced M2 dysfunction is regulated by eosinophils that are recruited to airway nerves and secrete the allosteric muscarinic M2 receptor antagonist major basic protein (114). The above-mentioned mechanisms collectively could well explain the increased role of the airway cholinergic system in airway hyperresponsiveness that is associated with loss of epithelial integrity. Because the airway cholinergic system also appears to regulate airway remodeling (100, 101, 115, 116), the same may hold true for this pathological response. Postjunctionally, there indeed are significant functional interactions between acetylcholine and growth factors that support ASM proliferation (Figure 2B). Muscarinic receptor stimulation by itself is not mitogenic to human ASM; however, in combination with growth factors such as PDGF or EGF, muscarinic M3 receptor stimulation augments the proliferative response to those factors, which likely involves the activation of multiple downstream effector pathways (117–119). In agreement with such an interaction, inhaled anticholinergics are effective in reducing ASM mass in guinea pigs that are allergen challenged, during which inflammatory mediators and growth factors are released, but have no effect on ASM mass in salinechallenged control animals (101). Muscarinic receptor stimulation cooperates with growth factor receptors to synergistically phosphorylate p70S6K and GSK-3, particularly in their late-

Dekkers, Maarsingh, Meurs, et al.: Airway Smooth Muscle Remodeling

687

phase phosphorylation (2–4 h after stimulation) (118–121). P70S6K is required for ASM cell proliferation and hypertrophy and is activated on phosphorylation, whereas GSK-3 is an antimitogenic and antihypertrophic kinase that is inhibited on phosphorylation (Figure 2B) (118, 122–125). Both actions of muscarinic receptors, which require the respective activation of PKC and bg subunits as signaling intermediates (118, 120, 121), therefore support ASM growth. Effects of muscarinic receptors on smooth muscle phenotype marker protein expression (e.g., sm-MHC) could also be explained this way, as both p70S6K and GSK-3 regulate the expression of smooth muscle–specific proteins (123, 126). Direct activation of smooth muscle–specific genes by muscarinic receptor stimulation has indeed been demonstrated (127, 128), but the underlying signaling events and interactions with growth factors still need to be assessed in future studies. Interestingly, the study by Fairbank and colleagues (127) showed that amplification of MLCK expression by muscarinic receptor stimulation occurred only in the presence of mechanical strain, which highlights another potentially important mechanism for remodeling that is regulated by mechanical factors. Mechanical strain on ASM will result in ASM cell proliferation (127, 129) and mechanical compression of the airway wall is sufficient to activate EGF receptors present in the airway epithelium (130, 131). These studies raise the real possibility that bronchodilation, for example using anticholinergic agents, reduces airway remodeling at least in part via the reduction of mechanical stress and strain within the airway wall. In conclusion, the above-mentioned studies have provided solid support for the hypothesis that airway inflammation can interact with neuronally derived acetylcholine to facilitate bronchoconstriction and ASM thickening. Nonneuronal acetylcholine, released for example by airway epithelial cells or inflammatory cells, may also contribute to these processes, which needs to be established in future studies (104). Although this process is dependent on airway inflammation, the interactive role of the airway cholinergic system is considerable and appears to play a major role in regulating ASM mass and phenotype (Figure 2).

and function. In the airway wall of subjects with asthma the amount and composition of the ECM is altered compared with healthy subjects. These changes are most eminent beneath the basement membrane and include increased deposition of collagens I, III, and V, fibronectin, tenascin, hyaluronan, versican, biglycan, lumican, and laminin a2/b2, as well as decreased expression of collagen IV, elastin, and decorin (132–137) (Figure 3). Changes in the ECM have also been observed within and surrounding the ASM bundles. In patients with fatal asthma, the total amount of ECM within and surrounding the ASM bundles is increased, which was correlated with severity, but not duration, of asthma (20). This increase involves increased deposition of collagen I, fibronectin, hyaluronan, versican, biglycan, lumican, and elastic fibers (138–140). A recent study, however, showed no changes in fractional area of collagen I or versican in the ASM layer (138). Interestingly, an inverse association between elastin expression and methacholine responsiveness has also been observed (10), suggesting that airway hyperresponsiveness is positively linked to the ECM expression in the ASM layer. ASM cells are a rich source of ECM components, as shown by the production of collagens, fibronectin, laminins, perlecan, elastin, thrombospondin, versican, and decorin by ASM (141– 143), the expression of which is increased in response to profibrotic factors such as TGF-b, CTGF, and VEGF (144) (Figure 3). Interestingly, expression of CTGF in response to TGF-b by asthmatic ASM is increased compared with nonasthmatic ASM (22), identifying this factor as a potentially important contributor to ECM production in asthma. Altered ECM production by asthmatic ASM is also supported by findings showing an increased production of collagen I, perlecan, and fibronectin (23, 27), and a decreased production of laminin a1, chondroitin sulfate, collagen IV, and hyaluronan compared with ASM derived from healthy subjects (27, 145). In addition, increased expression of fibronectin, laminin g1, perlecan, and chondroitin sulfate by nonasthmatic ASM cells was observed after exposure to atopic serum, indicating that plasma leakage may contribute to increased ECM production by ASM in asthma (143). Altered deposition of ECM proteins may alter mechanical properties of ASM as well as the transfer of force between the ASM bundle and surrounding tissue (146). In addition to their role in structural support, ECM proteins also regulate the

THE EXTRACELLULAR MATRIX The ECM is a dynamic structure that surrounds cells and provides the mechanical support required for airway structure

Figure 3. Airway smooth muscle (ASM) and extracellular matrix (ECM) mutually affect each other to support abnormal ASM function as observed in asthma. (A) In the asthmatic airways deposition of ECM proteins is increased, not only beneath the basement membrane but also within and surrounding the ASM bundle. Asthmatic ASM creates an altered ECM environment that facilitates increased contractile, proliferative, and synthetic capabilities. (B) Asthmatic ASM cells deposit increased amounts of fibronectin, which increases ASM synthetic function via a mechanism involving the a5b1 integrin. In addition, increased proliferation of asthmatic ASM has been shown to be dependent on the ECM, potentially also via a mechanism involving the a5b1 integrin. Laminins are critically involved in the expression of smooth muscle contractile proteins via the a7b1 integrin. Expression of this integrin has been shown to be increased by proremodeling factors such as TGF-b.

688

function of the cells embedded therein. ECM proteins have been found to differentially regulate survival, migration, cytokine synthesis, maturation, contractility, and proliferation of ASM cells (31, 147, 148). The alterations in the ECM profile produced by asthmatic ASM cells therefore have the potential to influence behavior and characteristics of the ASM cells (Figure 3). In support of this, studies on the effects of asthmatic ECM showed that culture of both healthy and asthmatic ASM cells on an asthmatic ECM enhanced proliferative responses (27). Similarly, increased eotaxin expression by asthmatic ASM is dependent on the ECM produced by these cells (23). This increased secretory response required interaction of the ASM with its ECM via a5b1 integrins (23), the expression of which can be increased in response to TGF-b in both nonasthmatic and asthmatic ASM (149) (Figure 3B). Preliminary findings from our laboratory also suggest an important role for integrins in ASM remodeling in vivo. Using a guinea pig model of chronic allergic asthma, we found that treatment with the integrin-blocking peptide Arg-Gly-Asp-Ser (RGDS), containing the RGD binding motif present in fibronectin, collagens, and laminins (150, 151), inhibits allergen-induced ASM hyperplasia, increased contractile protein expression, and ASM hypercontractility, without effects on inflammatory responses (152). The normalization of allergen-induced hypercontractility also suggests a role for changes in ECM composition in regulating ASM contractility. Indeed, studies indicated that exogenously applied laminin-111 (laminin-1) maintains contractile ASM phenotype (147, 153), whereas endogenously expressed laminin-211 (laminin-2) has been implicated in ASM maturation (154) and the induction of a hypercontractile ASM phenotype (155). ASM maturation required activation of the lamininbinding integrin a7 (156), the expression of which is increased by TGF-b in both nonasthmatic and asthmatic ASM (157). Collectively, these findings indicate that the ECM is not just an innocent bystander, but a component that can be actively regulated by the ASM, which in turn facilitates the abnormal ASM function as observed in asthma. These changes may be initiated by airway inflammation, but remain present in the absence of persistent inflammation. Intriguingly, these studies point to a dominant role of ASM–ECM interactions in the regulation of ASM remodeling and indicate that the muscle itself is capable of and in part responsible for creating an altered ECM environment that supports and maintains its increased contractile, proliferative, and synthetic characteristics (Figure 3).

CONCLUSIONS From numerous studies it is quite evident that ASM thickening is a prominent pathological feature in asthma that contributes to an important extent to increased airway reactivity in patients. The mechanisms underlying this response remain elusive. Clearly, evidence is accumulating to indicate that the model in which remodeling is due solely to the presence of inflammatory cells that secrete mediators and growth factors promoting cell proliferation and hypertrophy is incomplete. Rather, the ASM layer is part of an active epithelial mesenchymal trophic unit that is activated during tissue injury and repair and driven by both changes in inflammatory cells and damaged epithelium. The damaged and stressed epithelium expresses increased levels of arginase, which reduces the presence of bronchodilatory and antiproliferative NO and promotes the presence of amino acids and polyamines that regulate smooth muscle remodeling. Furthermore, the damaged epithelium allows exposure of afferent sensory nerve endings that, together with the ongoing inflammation of the underlying airway wall, promotes the release of acetylcholine that acts as an important regulator of ASM

PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 6

2009

remodeling via its actions on the postjunctional muscarinic M3 receptor. However, although epithelial cell changes and inflammation most likely play a major regulatory or initiating role, the studies summarized above also indicate that it is incorrect to assume that the underlying mesenchyme, including the ASM, is a passive partner in the remodeling process. The ASM actively participates in the remodeling process by regulating inflammation through the secretion of chemokines and cytokines, by producing force on the airway wall during periods of inflammation that regulates gene expression and kinase phosphorylation via mechanisms of mechanotransduction, and by producing an ECM that supports its multifunctional role with respect to its proliferative, secretory, and contractile capacities. Therefore, these studies call for a model of bidirectional rather than unidirectional communication between components of the airway wall, in which ASM thickening is controlled by several structural components, including the muscle itself. Conflict of Interest Statement: B.G.J.D. does not have a financial relationship with a commercial entity that has an interest in the subject of this manuscript. H. Maarsingh has received honoraria for lectures with Schering Plough Research Institute (up to $1,000). H. Meurs has received funding for research with Schering Plough ($100,001 or more), and Boehringer Ingelheim ($100,001 or more). He has also received funding through a noncommercial entity, Netherlands Asthma Foundation ($100,001 or more). R.G. has received honoraria for lectures with Boehringer Ingelheim (up to $1,000). He has received funding for research with Boehringer Ingelheim ($100,001 or more) and noncommercial entities NWO ($100,001 or more), and Netherlands Asthma Fund ($100,001 or more).

References 1. Bousquet J, Jeffery PK, Busse WW, Johnson M, Vignola AM. Asthma. From bronchoconstriction to airways inflammation and remodeling. Am J Respir Crit Care Med 2000;161:1720–1745. 2. Postma DS, Kerstjens HA. Characteristics of airway hyperresponsiveness in asthma and chronic obstructive pulmonary disease. Am J Respir Crit Care Med 1998;158:S187–S192. 3. Cockcroft DW, Davis BE. Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol 2006;118:551–559. 4. Jeffery PK. Remodeling in asthma and chronic obstructive lung disease. Am J Respir Crit Care Med 2001;164:S28–S38. 5. Lloyd CM, Robinson DS. Allergen-induced airway remodelling. Eur Respir J 2007;29:1020–1032. 6. Postma DS, Timens W. Remodeling in asthma and chronic obstructive pulmonary disease. Proc Am Thorac Soc 2006;3:434–439. 7. Ma X, Cheng Z, Kong H, Wang Y, Unruh H, Stephens NL, Laviolette M. Changes in biophysical and biochemical properties of single bronchial smooth muscle cells from asthmatic subjects. Am J Physiol Lung Cell Mol Physiol 2002;283:L1181–L1189. 8. Leguillette R, Laviolette M, Bergeron C, Zitouni NB, Kogut P, Solway J, Kashmar L, Hamid Q, Lauzon AM. Myosin, transgelin, and myosin light chain kinase: expression and function in asthma. Am J Respir Crit Care Med 2008;179:194–204. 9. Leguillette R, Lauzon AM. Molecular mechanics of smooth muscle contractile proteins in airway hyperresponsiveness and asthma. Proc Am Thorac Soc 2008;5:40–46. 10. Slats AM, Janssen K, van Schadewick A, van der Plas DT, Schot R, van den Aardweg JG, de Jongste JC, Hiemstra PS, Mauad T, Rabe KF, et al. Expression of smooth muscle and extracellular matrix proteins in relation to airway function in asthma. J Allergy Clin Immunol 2008; 121:1196–1202. 11. Matsumoto H, Moir LM, Oliver BG, Burgess JK, Roth M, Black JL, McParland BE. Comparison of gel contraction mediated by airway smooth muscle cells from patients with and without asthma. Thorax 2007;62:848–854. 12. Stephens NL, Fust A, Jiang H, Li W, Ma X. Isotonic relaxation of control and sensitized airway smooth muscle. Can J Physiol Pharmacol 2005;83:941–951. 13. Lambert RK, Wiggs BR, Kuwano K, Hogg JC, Pare PD. Functional significance of increased airway smooth muscle in asthma and COPD. J Appl Physiol 1993;74:2771–2781. 14. Oliver MN, Fabry B, Marinkovic A, Mijailovich SM, Butler JP, Fredberg JJ. Airway hyperresponsiveness, remodeling, and smooth

Dekkers, Maarsingh, Meurs, et al.: Airway Smooth Muscle Remodeling

689

muscle mass: right answer, wrong reason? Am J Respir Cell Mol Biol 2007;37:264–272. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC, Olivenstein R, Pavord ID, McCormack D, Chaudhuri R, et al. Asthma control during the year after bronchial thermoplasty. N Engl J Med 2007;356:1327–1337. Ebina M, Takahashi T, Chiba T, Motomiya M. Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 1993;148:720–726. Woodruff PG, Dolganov GM, Ferrando RE, Donnelly S, Hays SR, Solberg OD, Carter R, Wong HH, Cadbury PS, Fahy JV. Hyperplasia of smooth muscle in mild to moderate asthma without changes in cell size or gene expression. Am J Respir Crit Care Med 2004;169:1001– 1006. 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. James AL, Bai TR, Mauad T, Abramson MJ, Dolhnikoff M, McKay KO, Maxwell PS, Elliot JG, Green FH. Airway smooth muscle thickness in asthma is related to severity but not duration of asthma. Eur Respir J 2009;34:1040–1045. Bai TR, Cooper J, Koelmeyer T, Pare PD, Weir TD. The effect of age and duration of disease on airway structure in fatal asthma. Am J Respir Crit Care Med 2000;162:663–669. Pepe C, Foley S, Shannon J, Lemiere C, Olivenstein R, Ernst P, Ludwig MS, Martin JG, Hamid Q. Differences in airway remodeling between subjects with severe and moderate asthma. J Allergy Clin Immunol 2005;116:544–549. Burgess JK, Johnson PR, Ge Q, Au WW, Poniris MH, McParland BE, King G, Roth M, Black JL. Expression of connective tissue growth factor in asthmatic airway smooth muscle cells. Am J Respir Crit Care Med 2003;167:71–77. Chan V, Burgess JK, Ratoff JC, O’Connor BJ, Greenough A, Lee TH, Hirst SJ. Extracellular matrix regulates enhanced eotaxin expression in asthmatic airway smooth muscle cells. Am J Respir Crit Care Med 2006;174:379–385. Simcock DE, Kanabar V, Clarke GW, Mahn K, Karner C, O’Connor BJ, Lee TH, Hirst SJ. Induction of angiogenesis by airway smooth muscle from patients with asthma. Am J Respir Crit Care Med 2008; 178:460–468. Chambers LS, Black JL, Ge Q, Carlin SM, Au WW, Poniris M, Thompson J, Johnson PR, Burgess JK. PAR-2 activation, PGE2, and COX-2 in human asthmatic and nonasthmatic airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2003;285:L619– L627. Johnson PR, Roth M, Tamm M, Hughes M, Ge Q, King G, Burgess JK, Black JL. Airway smooth muscle cell proliferation is increased in asthma. Am J Respir Crit Care Med 2001;164:474–477. Johnson PR, Burgess JK, Underwood PA, Au W, Poniris MH, Tamm M, Ge Q, Roth M, Black JL. Extracellular matrix proteins modulate asthmatic airway smooth muscle cell proliferation via an autocrine mechanism. J Allergy Clin Immunol 2004;113:690–696. Trian T, Benard G, Begueret H, Rossignol R, Girodet PO, Ghosh D, Ousova O, Vernejoux JM, Marthan R, Tunon-de-Lara JM, et al. Bronchial smooth muscle remodeling involves calcium-dependent enhanced mitochondrial biogenesis in asthma. J Exp Med 2007;204: 3173–3181. Southam DS, Ellis R, Wattie J, Inman MD. Components of airway hyperresponsiveness and their associations with inflammation and remodeling in mice. J Allergy Clin Immunol 2007;119:848–854. Xu KF, Vlahos R, Messina A, Bamford TL, Bertram JF, Stewart AG. Antigen-induced airway inflammation in the Brown Norway rat results in airway smooth muscle hyperplasia. J Appl Physiol 2002; 93:1833–1840. Gosens R, Roscioni SS, Dekkers BG, Pera T, Schmidt M, Schaafsma D, Zaagsma J, Meurs H. Pharmacology of airway smooth muscle proliferation. Eur J Pharmacol 2008;585:385–397. Payne DN, Rogers AV, Adelroth E, Bandi V, Guntupalli KK, Bush A, Jeffery PK. Early thickening of the reticular basement membrane in children with difficult asthma. Am J Respir Crit Care Med 2003;167: 78–82. Regamey N, Ochs M, Hilliard TN, Muhlfeld C, Cornish N, Fleming L, Saglani S, Alton EW, Bush A, Jeffery PK, et al. Increased airway smooth muscle mass in children with asthma, cystic fibrosis, and noncystic fibrosis bronchiectasis. Am J Respir Crit Care Med 2008;177: 837–843.

34. Saglani S, Payne DN, Zhu J, Wang Z, Nicholson AG, Bush A, Jeffery PK. Early detection of airway wall remodeling and eosinophilic inflammation in preschool wheezers. Am J Respir Crit Care Med 2007;176:858–864. 35. Leigh R, Ellis R, Wattie J, Southam DS, De HM, Gauldie J, O‘Byrne PM, Inman MD. Dysfunction and remodeling of the mouse airway persist after resolution of acute allergen-induced airway inflammation. Am J Respir Cell Mol Biol 2002;27:526–535. 36. Henderson WR Jr, Chiang GK, Tien YT, Chi EY. Reversal of allergeninduced airway remodeling by CysLT1 receptor blockade. Am J Respir Crit Care Med 2006;173:718–728. 37. Vanacker NJ, Palmans E, Kips JC, Pauwels RA. Fluticasone inhibits but does not reverse allergen-induced structural airway changes. Am J Respir Crit Care Med 2001;163:674–679. 38. Holgate ST. The airway epithelium is central to the pathogenesis of asthma. Allergol Int 2008;57:1–10. 39. Holgate ST, Davies DE, Puddicombe S, Richter A, Lackie P, Lordan J, Howarth P. Mechanisms of airway epithelial damage: epithelialmesenchymal interactions in the pathogenesis of asthma. Eur Respir J Suppl 2003;44:24s–29s. 40. Holgate ST, Holloway J, Wilson S, Bucchieri F, Puddicombe S, Davies DE. Epithelial-mesenchymal communication in the pathogenesis of chronic asthma. Proc Am Thorac Soc 2004;1:93–98. 41. Holgate ST, Davies DE, Powell RM, Howarth PH, Haitchi HM, Holloway JW. Local genetic and environmental factors in asthma disease pathogenesis: chronicity and persistence mechanisms. Eur Respir J 2007;29:793–803. 42. Moncada S, Palmer RM, Higgs EA. Biosynthesis of nitric oxide from Larginine. A pathway for the regulation of cell function and communication. Biochem Pharmacol 1989;38:1709–1715. 43. Ricciardolo FL, Sterk PJ, Gaston B, Folkerts G. Nitric oxide in health and disease of the respiratory system. Physiol Rev 2004;84:731–765. 44. Yan ZQ, Hanson GK, Skoogh BE, Lotvall JO. Induction of nitric oxide synthase in a model of allergic occupational asthma. Allergy 1995;50: 760–764. 45. Maarsingh H, Zaagsma J, Meurs H. Arginine homeostasis in allergic asthma. Eur J Pharmacol 2008;585:375–384. 46. Kharitonov S, O’Connor BJ, Evans DJ, Barnes PJ. Allergen-induced late asthmatic reactions are associated with elevation of exhaled nitric oxide. Am J Respir Crit Care Med 1995;151:1894–1897. 47. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of asthmatic patients. Lancet 1994;343:133–135. 48. de Boer J, Meurs H, Coers W, Koopal M, Bottone AE, Visser AC, Timens W, Zaagsma J. Deficiency of nitric oxide in allergen-induced airway hyperreactivity to contractile agonists after the early asthmatic reaction: an ex vivo study. Br J Pharmacol 1996;119:1109–1116. 49. Maarsingh H, Leusink J, Bos IST, Zaagsma J, Meurs H. Arginase strongly impairs neuronal nitric oxide-mediated airway smooth muscle relaxation in allergic asthma. Respir Res 2006;7:6. 50. Maarsingh H, Bossenga BE, Bos IST, Volders HH, Zaagsma J, Meurs H. L-Arginine deficiency causes airway hyperresponsiveness after the late asthmatic reaction. Eur Respir J 2009;34:191–199. 51. Mehta S, Drazen JM, Lilly CM. Endogenous nitric oxide and allergic bronchial hyperresponsiveness in guinea pigs. Am J Physiol 1997;273: L656–L662. 52. Samb A, Pretolani M, Dinh-Xuan AT, Ouksel H, Callebert J, Lisdero C, Aubier M, Boczkowski J. Decreased pulmonary and tracheal smooth muscle expression and activity of type 1 nitric oxide synthase (nNOS) after ovalbumin immunization and multiple aerosol challenge in guinea pigs. Am J Respir Crit Care Med 2001;164:149–154. 53. Schuiling M, Meurs H, Zuidhof AB, Venema N, Zaagsma J. Dual action of iNOS-derived nitric oxide in allergen-induced airway hyperreactivity in conscious, unrestrained guinea pigs. Am J Respir Crit Care Med 1998;158:1442–1449. 54. Schuiling M, Zuidhof AB, Bonouvrie MA, Venema N, Zaagsma J, Meurs H. Role of nitric oxide in the development and partial reversal of allergen-induced airway hyperreactivity in conscious, unrestrained guinea-pigs. Br J Pharmacol 1998;123:1450–1456. 55. Ricciardolo FL, Timmers MC, Geppetti P, van Schadewijk A, Brahim JJ, Sont JK, de Gouw HW, Hiemstra PS, van Krieken JH, Sterk PJ. Allergen-induced impairment of bronchoprotective nitric oxide synthesis in asthma. J Allergy Clin Immunol 2001;108:198–204. 56. Ricciardolo FLM, DiMaria GU, Mistretta A, Sapienza MA, Geppetti P. Impairment of bronchoprotection by nitric oxide in severe asthma. Lancet 1997;350:1297–1298.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

690 57. de Boer J, Duyvendak M, Schuurman FE, Pouw FM, Zaagsma J, Meurs H. Role of L-arginine in the deficiency of nitric oxide and airway hyperreactivity after the allergen-induced early asthmatic reaction in guinea-pigs. Br J Pharmacol 1999;128:1114–1120. 58. Maarsingh H, Zuidhof AB, Bos IS, Van Duin M, Boucher JL, Zaagsma J, Meurs H. Arginase inhibition protects against allergic airway obstruction, hyperresponsiveness and inflammation. Am J Respir Crit Care Med 2008;178:565–573. 59. Xia Y, Zweier JL. Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc Natl Acad Sci USA 1997;94:6954–6958. 60. de Boer J, Meurs H, Flendrig L, Koopal M, Zaagsma J. Role of nitric oxide and superoxide in allergen-induced airway hyperreactivity after the late asthmatic reaction in guinea-pigs. Br J Pharmacol 2001;133: 1235–1242. 61. Fischer BM, Voynow JA. Neutrophil elastase induces MUC5AC gene expression in airway epithelium via a pathway involving reactive oxygen species. Am J Respir Cell Mol Biol 2002;26:447–452. 62. Muijsers RB, van Ark I, Folkerts G, Koster AS, van Oosterhout AJ, Postma DS, Nijkamp FP. Apocynin and 1400 W prevents airway hyperresponsiveness during allergic reactions in mice. Br J Pharmacol 2001;134:434–440. 63. Sadeghi-Hashjin G, Folkerts G, Henricks PAJ, Verheyen AKCP, van der Linde HJ, Van Ark I, Coene A, Nijkamp FP. Peroxynitrite induces airway hyperresponsiveness in guinea pigs in vitro and in vivo. Am J Respir Crit Care Med 1996;153:1697–1701. 64. Hamad AM, Johnson SR, Knox AJ. Antiproliferative effects of NO and ANP in cultured human airway smooth muscle. Am J Physiol 1999;277:L910–L918. 65. Hamad AM, Knox AJ. Mechanisms mediating the antiproliferative effects of nitric oxide in cultured human airway smooth muscle cells. FEBS Lett 2001;506:91–96. 66. Patel HJ, Belvisi MG, Donnelly LE, Yacoub MH, Chung KF, Mitchell JA. Constitutive expressions of type I NOS in human airway smooth muscle cells: evidence for an antiproliferative role. FASEB J 1999;13: 1810–1816. 67. Kizawa Y, Ohuchi N, Saito K, Kusama T, Murakami H. Effects of endothelin-1 and nitric oxide on proliferation of cultured guinea pig bronchial smooth muscle cells. Comp Biochem Physiol C Toxicol Pharmacol 2001;128:495–501. 68. Pilz RB, Casteel DE. Regulation of gene expression by cyclic GMP. Circ Res 2003;93:1034–1046. 69. Kapadia MR, Eng JW, Jiang Q, Stoyanovsky DA, Kibbe MR. Nitric oxide regulates the 26S proteasome in vascular smooth muscle cells. Nitric Oxide 2009;20:279–288. 70. Sandirasegarane L, Charles R, Bourbon N, Kester M. NO regulates PDGF-induced activation of PKB but not ERK in A7r5 cells: implications for vascular growth arrest. Am J Physiol Cell Physiol 2000;279:C225–C235. 71. Estrada C, Gomez C, Martin-Nieto J, De Frutos T, Jimenez A, Villalobo A. Nitric oxide reversibly inhibits the epidermal growth factor receptor tyrosine kinase. Biochem J 1997;326:369–376. 72. Murillo-Carretero M, Torroglosa A, Castro C, Villalobo A, Estrada C. S-Nitrosylation of the epidermal growth factor receptor: a regulatory mechanism of receptor tyrosine kinase activity. Free Radic Biol Med 2009;46:471–479. 73. Que LG, Kantrow SP, Jenkinson CP, Piantadosi CA, Huang YC. Induction of arginase isoforms in the lung during hyperoxia. Am J Physiol 1998;275:L96–L102. 74. Maarsingh H, Pera T, Meurs H. Arginase and pulmonary diseases. Naunyn Schmiedebergs Arch Pharmacol 2008;378:171–184. 75. Morris CR, Poljakovic M, Lavrisha L, Machado L, Kuypers FA, Morris SM Jr. Decreased arginine bioavailability and increased serum arginase activity in asthma. Am J Respir Crit Care Med 2004;170: 148–153. 76. North ML, Khanna N, Marsden PA, Grasemann H, Scott JA. Functionally important role for arginase 1 in the airway hyperresponsiveness of asthma. Am J Physiol Lung Cell Mol Physiol 2009;296: L911–L920. 77. Zimmermann N, King NE, Laporte J, Yang M, Mishra A, Pope SM, Muntel EE, Witte DP, Pegg AA, Foster PS, et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest 2003;111:1863–1874. 78. Yang M, Rangasamy D, Matthaei KI, Frew AJ, Zimmmermann N, Mahalingam S, Webb DC, Tremethick DJ, Thompson PJ, Hogan SP, et al. Inhibition of arginase I activity by RNA interference attenuates

PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 6

79.

80.

81.

82.

83.

84.

85. 86.

87.

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

99. 100.

2009

IL-13-induced airways hyperresponsiveness. J Immunol 2006;177: 5595–5603. Meurs H, McKay S, Maarsingh H, Hamer MA, Macic L, Molendijk N, Zaagsma J. Increased arginase activity underlies allergen-induced deficiency of cNOS-derived nitric oxide and airway hyperresponsiveness. Br J Pharmacol 2002;136:391–398. Maarsingh H, de Boer J, Kauffman HF, Zaagsma J, Meurs H. Heparin normalizes allergen-induced nitric oxide deficiency and airway hyperresponsiveness. Br J Pharmacol 2004;142:1293–1299. Meurs H, Maarsingh H, Zaagsma J. Arginase and asthma: novel insights into nitric oxide homeostasis and airway hyperresponsiveness. Trends Pharmacol Sci 2003;24:450–455. Maarsingh H, Tio MA, Zaagsma J, Meurs H. Arginase attenuates inhibitory nonadrenergic noncholinergic nerve-induced nitric oxide generation and airway smooth muscle relaxation. Respir Res 2005;6: 23. Hoet PH, Nemery B. Polyamines in the lung: polyamine uptake and polyamine-linked pathological or toxicological conditions. Am J Physiol Lung Cell Mol Physiol 2000;278:L417–L433. Ricciardolo FL, Zaagsma J, Meurs H. The therapeutic potential of drugs targeting the arginase pathway in asthma. Expert Opin Investig Drugs 2005;14:1221–1231. Wu G, Morris SM. Arginine metabolism: nitric oxide and beyond. Biochem J 1998;336:1–17. Hobbs CA, Gilmour SK. High levels of intracellular polyamines promote histone acetyltransferase activity resulting in chromatin hyperacetylation. J Cell Biochem 2000;77:345–360. Bergeron C, Boulet LP, Page N, Laviolette M, Zimmermann N, Rothenberg ME, Hamid Q. Influence of cigarette smoke on the arginine pathway in asthmatic airways: increased expression of arginase I. J Allergy Clin Immunol 2007;119:391–397. Durante W, Liao L, Peyton KJ, Schafer AI. Thrombin stimulates vascular smooth muscle cell polyamine synthesis by inducing cationic amino acid transporter and ornithine decarboxylase gene expression. Circ Res 1998;83:217–223. Durante W, Liao L, Reyna SV, Peyton KJ, Schafer AI. Transforming growth factor-beta(1) stimulates L-arginine transport and metabolism in vascular smooth muscle cells: role in polyamine and collagen synthesis. Circulation 2001;103:1121–1127. Nelin LD, Chicoine LG, Reber KM, English BK, Young TL, Liu Y. Cytokine-induced endothelial arginase expression is dependent on epidermal growth factor receptor. Am J Respir Cell Mol Biol 2005;33: 394–401. Thyberg J, Fredholm BB. Modulation of arterial smooth-muscle cells from contractile to synthetic phenotype requires induction of ornithine decarboxylase activity and polyamine synthesis. Exp Cell Res 1987;170:153–159. Thyberg J, Fredholm BB. Induction of ornithine decarboxylase activity and putrescine synthesis in arterial smooth-muscle cells stimulated with platelet-derived growth-factor. Exp Cell Res 1987;170:160–169. Maarsingh H, Ten Damme A, Joughi FA, Zaagsma J, Meurs H. Role for upregulation and activation of ornithine decarboxylase in plateletderived growth factor (PDGF)-induced airway smooth muscle hypocontractility. Am J Respir Crit Care Med 2008;177:A488. Maarsingh H, Bos IST, Westerhof-Humblot FJ, Zaagsma J, Meurs H. Increased arginase activity underlies airway hyperresponsiveness in a guinea pig model of chronic allergic asthma. Am J Respir Crit Care Med 2007;175:A522. Kurosawa M, Shimizu Y, Tsukagoshi H, Ueki M. Elevated levels of peripheral-blood, naturally occurring aliphatic polyamines in bronchial asthmatic patients with active symptoms. Allergy 1992;47:638– 643. Wei LH, Wu G, Morris SM Jr, Ignarro LJ. Elevated arginase I expression in rat aortic smooth muscle cells increases cell proliferation. Proc Natl Acad Sci USA 2001;98:9260–9264. Bauer PM, Fukuto JM, Buga GM, Pegg AE, Ignarro LJ. Nitric oxide inhibits ornithine decarboxylase by S-nitrosylation. Biochem Biophys Res Commun 1999;262:355–358. Belmonte KE. Cholinergic pathways in the lungs and anticholinergic therapy for chronic obstructive pulmonary disease. Proc Am Thorac Soc 2005;2:297–304. Racke K, Juergens UR, Matthiesen S. Control by cholinergic mechanisms. Eur J Pharmacol 2006;533:57–68. Bos IS, Gosens R, Zuidhof AB, Schaafsma D, Halayko AJ, Meurs H, Zaagsma J. Inhibition of allergen-induced airway remodelling by tiotropium and budesonide: a comparison. Eur Respir J 2007;30:653–661.

Dekkers, Maarsingh, Meurs, et al.: Airway Smooth Muscle Remodeling

691

101. Gosens R, Bos IS, Zaagsma J, Meurs H. Protective effects of tiotropium bromide in the progression of airway smooth muscle remodeling. Am J Respir Crit Care Med 2005;171:1096–1102. 102. Ohta S, Minoguchi K, Oda N, Chen X, Tanaka A, Tazaki T, Yamamoto Y, Yamamoto M, Watanabe Y, Adachi M. Effects of tiotropium bromide on allergen induced airway remodeling in murine model of chronic asthma. Am J Respir Crit Care Med 2007; 175:A464. 103. Gosens R, Zaagsma J, Grootte BM, Nelemans A, Meurs H. Acetylcholine: a novel regulator of airway smooth muscle remodelling? Eur J Pharmacol 2004;500:193–201. 104. Gosens R, Zaagsma J, Meurs H, Halayko AJ. Muscarinic receptor signaling in the pathophysiology of asthma and COPD. Respir Res 2006;7:73. 105. Myers AC. Transmission in autonomic ganglia. Respir Physiol 2001; 125:99–111. 106. Undem BJ, Myers AC. Cholinergic and noncholinergic parasympathetic control of airway smooth muscle. In: Zaagsma J, Meurs H, Roffel AF, editors. Muscarinic receptors in airways diseases. Basel: Birkha¨user Verlag; 2001. p. 1–24. 107. Spina D, Shah S, Harrison S. Modulation of sensory nerve function in the airways. Trends Pharmacol Sci 1998;19:460–466. 108. Undem BJ, Carr MJ. Pharmacology of airway afferent nerve activity. Respir Res 2001;2:234–244. 109. 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. 110. Santing RE, Pasman Y, Olymulder CG, Roffel AF, Meurs H, Zaagsma J. Contribution of a cholinergic reflex mechanism to allergen-induced bronchial hyperreactivity in permanently instrumented, unrestrained guinea-pigs. Br J Pharmacol 1995;114:414–418. 111. Coulson FR, Fryer AD. Muscarinic acetylcholine receptors and airway diseases. Pharmacol Ther 2003;98:59–69. 112. Fryer AD, Jacoby DB. Muscarinic receptors and control of airway smooth muscle. Am J Respir Crit Care Med 1998;158:S154–S160. 113. ten Berge RE, Santing RE, Hamstra JJ, Roffel AF, Zaagsma J. Dysfunction of muscarinic M2 receptors after the early allergic reaction: possible contribution to bronchial hyperresponsiveness in allergic guinea-pigs. Br J Pharmacol 1995;114:881–887. 114. Jacoby DB, Gleich GJ, Fryer AD. Human eosinophil major basic protein is an endogenous allosteric antagonist at the inhibitory muscarinic M2 receptor. J Clin Invest 1993;91:1314–1318. 115. Haag S, Matthiesen S, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of collagen synthesis in human lung fibroblasts. Eur Respir J 2008;32:555–562. 116. Matthiesen S, Bahulayan A, Kempkens S, Haag S, Fuhrmann M, Stichnote C, Juergens UR, Racke K. Muscarinic receptors mediate stimulation of human lung fibroblast proliferation. Am J Respir Cell Mol Biol 2006;35:621–627. 117. Gosens R, Nelemans SA, Grootte Bromhaar MM, McKay S, Zaagsma J, Meurs H. Muscarinic M3-receptors mediate cholinergic synergism of mitogenesis in airway smooth muscle. Am J Respir Cell Mol Biol 2003;28:257–262. 118. Gosens R, Dueck G, Rector E, Nunes RO, Gerthoffer WT, Unruh H, Zaagsma J, Meurs H, Halayko AJ. Cooperative regulation of GSK-3 by muscarinic and PDGF receptors is associated with airway myocyte proliferation. Am J Physiol Lung Cell Mol Physiol 2007;293:L1348– L1358. 119. Krymskaya VP, Orsini MJ, Eszterhas AJ, Brodbeck KC, Benovic JL, Panettieri RA Jr, Penn RB. 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. 120. Billington CK, Kong KC, Bhattacharyya R, Wedegaertner PB, Panettieri RA Jr, Chan TO, Penn RB. Cooperative regulation of p70S6 kinase by receptor tyrosine kinases and G protein-coupled receptors augments airway smooth muscle growth. Biochemistry 2005;44: 14595–14605. 121. 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. 122. Bentley JK, Deng H, Linn MJ, Lei J, Dokshin GA, Fingar DC, Bitar KN, Henderson WR Jr, Hershenson MB. Airway smooth muscle hyperplasia and hypertrophy correlate with glycogen synthase kinase-

3(beta) phosphorylation in a mouse model of asthma. Am J Physiol Lung Cell Mol Physiol 2009;296:L176–L184. Deng H, Dokshin GA, Lei J, Goldsmith AM, Bitar KN, Fingar DC, Hershenson MB, Bentley JK. Inhibition of glycogen synthase kinase3beta is sufficient for airway smooth muscle hypertrophy. J Biol Chem 2008;283:10198–10207. Halayko AJ, Kartha S, Stelmack GL, McConville J, Tam J, CamorettiMercado B, Forsythe SM, Hershenson MB, Solway J. Phophatidylinositol-3 kinase/mammalian target of rapamycin/p70S6K regulates contractile protein accumulation in airway myocyte differentiation. Am J Respir Cell Mol Biol 2004;31:266–275. Nunes RO, Schmidt M, Dueck G, Baarsma H, Halayko AJ, Kerstjens HA, Meurs H, Gosens R. GSK-3/beta-catenin signaling axis in airway smooth muscle: role in mitogenic signaling. Am J Physiol Lung Cell Mol Physiol 2008;294:L1110–L1118. Goldsmith AM, Bentley JK, Zhou L, Jia Y, Bitar KN, Fingar DC, Hershenson MB. Transforming growth factor-beta induces airway smooth muscle hypertrophy. Am J Respir Cell Mol Biol 2006;34: 247–254. Fairbank NJ, Connolly SC, Mackinnon JD, Wehry K, Deng L, Maksym GN. Airway smooth muscle cell tone amplifies contractile function in the presence of chronic cyclic strain. Am J Physiol Lung Cell Mol Physiol 2008;295:L479–L488. Liu HW, Kassiri K, Voros A, Hiller CT, Wang L, Solway J, Halayko AJ. Gaq-receptor coupled signaling induces RHO-dependent transcription of smooth muscle specific genes in cultured canine airway myocytes. Am J Respir Crit Care Med 2002;165:A670. 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. Tschumperlin DJ, Dai G, Maly IV, Kikuchi T, Laiho LH, McVittie AK, Haley KJ, Lilly CM, So PT, Lauffenburger DA, et al. Mechanotransduction through growth-factor shedding into the extracellular space. Nature 2004;429:83–86. Tschumperlin DJ, Drazen JM. Chronic effects of mechanical force on airways. Annu Rev Physiol 2006;68:563–583. de Kluijver J, Schrumpf JA, Evertse CE, Sont JK, Roughley PJ, Rabe KF, Hiemstra PS, Mauad T, Sterk PJ. Bronchial matrix and inflammation respond to inhaled steroids despite ongoing allergen exposure in asthma. Clin Exp Allergy 2005;35:1361–1369. Laitinen A, Altraja A, Kampe M, Linden M, Virtanen I, Laitinen LA. Tenascin is increased in airway basement membrane of asthmatics and decreased by an inhaled steroid. Am J Respir Crit Care Med 1997; 156:951–958. Laitinen LA, Laitinen A. Inhaled corticosteroid treatment and extracellular matrix in the airways in asthma. Int Arch Allergy Immunol 1995;107:215–216. Roberts CR, Walker DC, Schellenberg RR. Extracellular matrix. Clin Allergy Immunol 2002;16:143–178. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1989;1:520–524. Wilson JW, Li X. The measurement of reticular basement membrane and submucosal collagen in the asthmatic airway. Clin Exp Allergy 1997;27:363–371. Araujo BB, Dolhnikoff M, Silva LF, Elliot J, Lindeman JH, Ferreira DS, Mulder A, Gomes HA, Fernezlian SM, James A, et al. Extracellular matrix components and regulators in the airway smooth muscle in asthma. Eur Respir J 2008;32:61–69. Pini L, Hamid Q, Shannon J, Lemelin L, Olivenstein R, Ernst P, Lemiere C, Martin JG, Ludwig MS. Differences in proteoglycan deposition in the airways of moderate and severe asthmatics. Eur Respir J 2007;29:71–77. Roberts CR, Burke AK. Remodelling of the extracellular matrix in asthma: proteoglycan synthesis and degradation. Can Respir J 1998;5:48–50. Panettieri RA, Tan EM, Ciocca V, Luttmann MA, Leonard TB, Hay DW. Effects of LTD4 on human airway smooth muscle cell proliferation, matrix expression, and contraction In vitro: differential sensitivity to cysteinyl leukotriene receptor antagonists. Am J Respir Cell Mol Biol 1998;19:453–461. Coutts A, Chen G, Stephens N, Hirst S, Douglas D, Eichholtz T, Khalil N. Release of biologically active TGF-beta from airway smooth muscle cells induces autocrine synthesis of collagen. Am J Physiol Lung Cell Mol Physiol 2001;280:L999–L1008. Johnson PR, Black JL, Carlin S, Ge Q, Underwood PA. The production of extracellular matrix proteins by human passively sensitized

123.

124.

125.

126.

127.

128.

129.

130.

131. 132.

133.

134.

135. 136. 137.

138.

139.

140. 141.

142.

143.

692

144.

145.

146.

147.

148.

149.

PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 6 airway smooth-muscle cells in culture: the effect of beclomethasone. Am J Respir Crit Care Med 2000;162:2145–2151. Burgess JK, Ceresa C, Johnson SR, Kanabar V, Moir LM, Nguyen TT, Oliver BG, Schuliga M, Ward J. Tissue and matrix influences on airway smooth muscle function. Pulm Pharmacol Ther 2009;5:379– 387. Klagas I, Goulet S, Karakiulakis G, Zhong J, Baraket M, Black JL, Papakonstantinou E, Roth M. Decreased hyaluronan in airway smooth muscle cells from patients with asthma and COPD. Eur Respir J 2009;34:616–628. An SS, Bai TR, Bates JH, Black JL, Brown RH, Brusasco V, Chitano P, Deng L, Dowell M, Eidelman DH, et al. Airway smooth muscle dynamics: a common pathway of airway obstruction in asthma. Eur Respir J 2007;29:834–860. 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. Fernandes DJ, Bonacci JV, Stewart AG. Extracellular matrix, integrins, and mesenchymal cell function in the airways. Curr Drug Targets 2006;7:567–577. Moir LM, Burgess JK, Black JL. Transforming growth factor beta(1) increases fibronectin deposition through integrin receptor alpha(5)beta(1) on human airway smooth muscle. J Allergy Clin Immunol 2008;121:1034–1039.

2009

150. Aumailley M, Gerl M, Sonnenberg A, Deutzmann R, Timpl R. Identification of the Arg-Gly-Asp sequence in laminin A chain as a latent cellbinding site being exposed in fragment P1. FEBS Lett 1990;262:82–86. 151. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding to integrins. J Biol Chem 2000;275:21785–21788. 152. Dekkers BG, Bos IS, Gosens R, Halayko AJ, Zaagsma J, Meurs H. Inhibition of airway smooth muscle remodeling in an animal model of chronic asthma by the integrin-blocking peptide RGDS. Am J Respir Crit Care Med 2009;179:A5600. 153. Hirst SJ, Twort CH, Lee TH. Differential effects of extracellular matrix proteins on human airway smooth muscle cell proliferation and phenotype. Am J Respir Cell Mol Biol 2000;23:335–344. 154. Tran T, McNeill KD, Gerthoffer WT, Unruh H, Halayko AJ. Endogenous laminin is required for human airway smooth muscle cell maturation. Respir Res 2006;7:117. 155. Dekkers BG, Schaafsma D, Tran T, Zaagsma J, Meurs H. Insulininduced laminin expression promotes a hypercontractile airway smooth muscle phenotype. Am J Respir Cell Mol Biol 2009;41:494–504. 156. Tran T, Ens-Blackie K, Rector ES, Stelmack GL, McNeill KD, Tarone G, Gerthoffer WT, Unruh H, Halayko AJ. Laminin-binding Integrin falphag7 is Required for Contractile Phenotype Expression by Human Airway Myocyte. Am J Respir Cell Mol Biol 2007;37:668–680. 157. Kanabar V, Karner LC, Simcock DE, Mahn K, Nguyen NM, Cousins DJ, Lavender P, Hirst SJ. Integrin gene profiling in airway smooth muscle from asthmatics. Proc Am Thorac Soc 2007;4:A302.