Vous êtes sur la page 1sur 17

Clin Chest Med 27 (2006) 53 69

Airway Smooth Muscle as a Regulator of Immune Responses and Bronchomotor Tone


Aili L. Lazaar, MDT, Reynold A. Panettieri, Jr, MD
Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Medical Center, BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA

Asthma, a chronic disease with a prevalence of 3% to 5% in the United States, affects millions of people worldwide. Complex genetic and environmental factors contribute to the development of asthma, which is characterized by reversible airway obstruction, airway inflammation, and airway smooth muscle (ASM) cell hyperplasia. The traditional view that in asthma ASM is a purely contractile tissue seems to be inadequate. Compelling evidence now suggests that ASM plays an important role in regulating bronchomotor tone, in perpetuating airway inflammation, and in remodeling of the airways. This article reviews three distinct functions of ASM cells: the process of excitation contraction coupling, with a particular focus on the role of cytokines in modulating calcium responses; the processes of smooth muscle cell proliferation and migration; and the synthetic and immunomodulatory function of ASM cells. It also discusses how altered synthetic function contributes to airway remodeling.

Airway smooth muscle shortening and airway hyperresponsiveness Airway smooth muscle bronchomotor tone and asthma Smooth muscle cell shortening regulates airway luminal caliber. Many studies have examined whether

T Corresponding author. E-mail address: Aili.L.Lazaar@gsk.com (A.L. Lazaar).

the nonspecific airway hyperresponsiveness (AHR) characterizing asthma can be attributed to increased ASM force generation as a consequence of altered receptor ligand interactions or altered signal transduction pathways. For example, expression of the bradykinin B2 receptor is increased in ASM exposed to interleukin (IL)-1b, tumor necrosis factor-alpha (TNFa), or transforming growth factor-beta (TGFb), whereas the bradykinin B1 receptor is increased by treatment with IL-4 [1 5]. In contrast, TNFa dramatically decreases muscarinic receptor density but enhances acetylcholine-induced hyperresponsiveness because of altered downstream signal transduction pathways [6,7]. Despite evidence that ASM derived from patients who have asthma may not exhibit a greater increase in isometric force generation than in nonasthmatic persons, exposure to inflammatory mediators clearly enhances force generation of normal tissue to contractile agonists. Early studies demonstrated that passive sensitization of human ASM with asthmatic serum nonspecifically increases smooth muscle cell responsiveness [8 11]. TNFa and IL-1b induce bronchial hyperreactivity in both humans and animals, whereas in vitro cytokines and other mediators, including TNFa, IL-1b, IL-13, IL-5, lysophosphatidic acid, and phospholipase A2 also prime ASM to become hyperresponsive to contractile agonists [12 16]. Leukotrienes (LTs) are potent bronchoconstrictors in normal and asthmatic persons, and ASM cells express both receptors for the cysteinyl leukotrienes, LTC4, LTD4, and LTE4 [17,18]. Expression of the CysLT1 receptor is increased by exposure to interferon (IFN)-g and results in an increase in LTD4-

0272-5231/06/$ see front matter D 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.ccm.2005.10.003

chestmed.theclinics.com

54

lazaar

&

panettieri, jr

mediated force generation in cultured ASM cells [19]. Conversely, TNFa, IL-1b, and IL-13, but not IL-4, reduce b-adrenergic relaxation responses [20]. Together, these studies suggest that proinflammatory mediators promote airway hyperresponsiveness by enhancing ASM contraction or altering ASM relaxation. Modulation of calcium homeostasis in airway smooth muscle cells In human ASM cells, contractile agonists bind Gq- or G0-protein coupled receptors (GPCR) and activate phospholipase C. The subsequent hydrolysis of phosphatidylinositol 4,5 bisphosphate into inositol trisphosphate and diacylglycerol ultimately increases cytosolic calcium [21]. Because most inflammatory agents evoke neither a calcium response or nor phosphoinositide (PI) hydrolysis in human ASM, modu-

lation of agonist-induced increases in intracellular + calcium (Ca2 ) by extracellular stimuli probably i modulates downstream GPCR signaling. TNFa increases the expression as well as the activity of G-proteins in several cell types, including ASM [20]. The effect of cytokines on agonist-evoked calcium responses seems to be stimulus specific, however, because bradykinin-evoked PI accumulation is significantly enhanced by TNFa and IL-1b, whereas IL1b diminishes histamine-induced PI metabolism [1,22,23]. + The level of Ca2 i , in ASM modulates excitation contraction coupling and force generation (Fig. 1) + [24]. Agonist-induced elevations in Ca2 activate i the calcium-/calmodulin-sensitive myosin light chain kinase (MLCK), leading to phosphorylation of the regulatory myosin light chain (MLC20). Phosphorylation of MLC20 initiates cross-bridge cycling between myosin and actin and sustains ATP binding,

Fig. 1. Cytokine effects on excitation contraction coupling in ASM cells. Cytokines binding to their receptors influence intracellular signaling as well as the function and expression of GPCRs and CD38. Alterations in intracellular signaling affect calcium homeostasis and calcium sensitization. cADPR, cyclic ADP ribose; CaM, calmodulin; DAG, diacylglycerol; GEF, guanine exchange factor; GPCR, G-protein coupled receptor; IP3, inositol 3-phosphate; MLC, myosin light chain; MLCK, myosin light chain kinase; PLC, phospholipase C; R, cytokine receptor; RyR, ryanodine receptor; SR, sarcoplasmic reticulum.

asm as an immune-response regulator

55

hydrolysis and ADP release. Dephosphorylation by the MLC phosphatase terminates cross-bridge cycling and relaxes smooth muscle [21]. Evidence now suggests that abnormalities in these two major regulatory + events (increases in Ca2 and MLC phosphorylation) i induce bronchial hyperresponsiveness in asthma [12]. Agonist-evoked calcium responses and calcium sensitization Calcium homeostasis in cultured ASM cells is altered by inflammatory mediators. Cytokines such as TNFa, IL-1b, or IL-13 enhance cytosolic calcium responses in human ASM induced by carbachol, bradykinin, or thrombin [1,15,25]. In contrast, IL-4 suppresses carbachol-induced calcium signals [26,27]. Other stimuli may directly enhance agonist-induced calcium signaling in cultured ASM cells, including eosinophil-derived polycationic proteins or major basic protein, the aldehyde pollutant acrolein, or phospholipase A2 [11,28,29]. More recently, a phenomenon known as calcium sensitization has been described that involves calcium-independent mechanisms for maintaining MLC phosphorylation and smooth muscle contraction [30]. The signaling pathways for calcium sensitization involve inhibition of MLC phosphatase and, thus, inhibition of MLC dephosphorylation, most notably by Rho/Rho kinase and integrin-linked kinase [30]. Ultimately, enhanced MLC phosphorylation increases actin myosin interactions. Aberrant RhoA/Rho kinase activation has been implicated in a variety of disease states [31]. In ASM cells, carbachol- and TNF-induced calcium sensitization is mediated through activation of RhoA/ Rho kinase [32,33]. Additionally, animal models of allergen-induced AHR and airway inflammation demonstrate increased RhoA activation; in turn, antigen-induced AHR is abrogated by pharmacologic inhibition of Rho kinase [34,35]. Allergen sensitization also increases lung expression of Rho and directly impugns Rho kinase in agonist-induced ASM contraction [36]. Thus, pathways that modulate Rho and Rho kinase activation, particularly dysregulated contractile responses, may play an important role in asthma pathogenesis. CD38 Recent studies show that CD38, a membranebound protein expressed on ASM cells, modulates cyclic adenosine diphosphate ribose (cADPR) levels [37]. cADPR, a b-nicotinamide adenine dinucleotide metabolite, contributes to agonist-induced elevation + of Ca2 concentration in ASM cells by activating i

ryanodine receptors [38,39]. Recently, TNFa and IL-13 have been shown to increase expression of CD38 on ASM cells [40,41]. Furthermore, a cADPR antagonist inhibited agonist-induced increases in + Ca2 [40,42]. Finally, agonist-induced increases in I lung resistance are attenuated in CD38-deficient animals [43], suggesting that CD38 expression on ASM plays a significant role in maintaining calcium homeostasis and contributes to AHR. Alterations in the biophysical properties of airway smooth muscle can modulate airway smooth muscle shortening Proinflammatory mediators may also alter the internal resistance to shortening of the muscle, thereby affecting force generation. For example, MLCK content is increased in sensitized airways [44], which may contribute to the enhanced shortening velocity of ASM from asthmatic patients [45]. Alternatively, TNFa and contractile agonists such as acetylcholine can induce reorganization of the actin cytoskeleton, a process mediated by the Rho family of guanosine triphosphatases (GTPase), RhoA and cdc42 [46,47]. Inflammatory mediators and agonists also activate ribosomal S6 kinase, p38MAPK. Inhibition of p38MAPK reduces agonist-induced force generation, possibly through effects on heat shock protein 27, an actin-capping protein [48,49]. A more detailed discussion of the effects of mediators on the actin cytoskeleton has recently been published [50]. Proinflammatory cytokines affect ASM contractility on many levels. Alterations in calcium homeostasis and sensitivity, as well as contractile agonist receptor expression and signal transduction pathways, have profound effects on airway hyperreactivity. The identification of the fundamental signaling pathways promoting ASM excitation contraction coupling will provide new therapeutic targets to modulate ASM function in asthma.

Cellular and molecular mechanisms regulating airway smooth muscle cell growth Early studies of the histopathology of asthma described increases in both size and number of airway smooth muscle cells [51,52] and a correlation between airway hyperreactivity and increased airways resistance. During the past decade, significant advances have been made in identifying the many diverse mitogens and signal transduction pathways that modulate ASM growth (reviewed in [53]). This

56

lazaar

&

panettieri, jr

section examines the critical signaling pathways that regulate ASM growth. Hyperplasia Animal models of allergic airway inflammation have shown clear indications of in vivo ASM cell proliferation, using various markers such as bromodeoxyuridine (BrDU) or proliferating cell nuclear antigen (PCNA) [53]. Studies of bronchial biopsies from patients who had asthma, however, have yielded conflicting results regarding the presence of ASM proliferation in vivo [54 56]. These differences may be caused by sampling error or compartmentalization of the proliferative response [53]. Mediators Many inflammatory mediators are increased in bronchoalveolar lavage (BAL) fluid from airways of patients who have asthma, and such mediators induce ASM mitogenesis in vitro [57]. To date, mitogenic stimuli include growth factors such as epidermal growth factor (EGF), insulin-like growth factors (IGFs), platelet-derived growth factor (PDGF) isoforms BB and AB, and basic fibroblast growth factor; plasma- or inflammatory cell-derived mediators, such as lysosomal hydrolases (b-hexosaminidases and b-glucuronidase), a-thrombin, tryptase, and sphingosine 1-phosphate; and contractile agonists, such as histamine, endothelin-1, substance P, phenylephrine, serotonin, thromboxane A2, and LTD4. TGFb inhibits mitogen-induced ASM cell proliferation [58] but also increases expression of the leukotriene receptor cysLT1R, thereby rendering cells responsive to the proliferative effects of LTD4 [59]. Expression of peroxisome-proliferator activated receptor gamma (PPAR-g) is increased in the smooth muscle of patients who have asthma [55]; PPAR-g ligands inhibit mitogen-induced ASM mitogenesis by preventing cell cycle progression [60]. Although IL-1b, IL-6, and TNFa are also increased in the BAL fluid of asthmatics [61], whether these cytokines stimulate ASM proliferation in vitro remains controversial. IL-1b and IL-6 may induce hyperplasia and hypertrophy of cultured guinea pig or rat ASM cells [62,63], but other studies have shown that IL-1b [64] and IL-6 [65] are nonmitogenic for human ASM cells. The effects of TNFa on ASM proliferation are controversial. McKay and colleagues [65] reported that TNFa had no immediate mitogenic effect on human ASM cells, in contrast to the findings of Stewart and colleagues [66] that the proliferative effect of TNFa on human ASM cells seemed to be biphasic [65,66]. New evidence reveals

that the inhibitory effect of TNFa on ASM proliferation is caused, in part, by autocrine release of IFNb [67] as well as by cytokine-induced production of cyclo-oxygenase 2 dependent prostanoids, such as prostaglandin E2 (PGE2), which inhibit DNA synthesis [64]. Agonist-induced release of PGE2 is significantly lower in smooth muscle derived from asthmatics than in smooth muscle obtained from nonasthmatic persons [68]. Therefore, cytokineinduced proliferative responses in ASM may be enhanced under conditions of cyclo-oxygenase inhibition, in which the expression of growth-inhibitory prostanoids, such as PGE2, is limited [62,64,66]. Extracellular matrix Extracellular matrix (ECM) modulates mitogeninduced ASM cell growth. Collagen types I, III, and V, fibronectin, tenascin, hyaluronan, versican, and laminin are increased in the airways of asthmatics [69]. In vitro, fibronectin and collagen I increase human ASM cell mitogenesis in response to PDGFBB or a-thrombin, whereas laminin inhibits proliferation [70]. In addition, the increase in cell proliferation is accompanied by decreased expression of smooth muscle cell contractile proteins such as a-actin, calponin, and myosin heavy chain, suggesting that matrix may also modulate smooth muscle phenotype [70]. ECM composition also affects ASM cell survival. Fibronectin, laminin, and collagens I and IV were identified as important antiapoptotic elements, whereas elastin promoted ASM apoptosis. Recently, human ASM cells were shown to secrete ECM proteins in response to asthmatic sera [71], identifying ASM as a cellular source for ECM deposition in airways and suggesting a novel mechanism by which ASM cells may modulate autocrine proliferative responses. ASM cells from asthmatic patients secrete significantly more perlecan, collagen I, and chondroitin sulfate and less laminin a1 and collagen IV than ASM cells from nonasthmatic persons [72]. In addition, ECM derived from ASM of asthmatics induces greater cell proliferation than that obtained from nonasthmatics. In contrast, conditioned media from either asthmatic or nonasthmatic ASM promote a similar degree of cell proliferation. Mitogenic signaling pathways ASM mitogens may act through different receptor-operated mechanisms. Growth factors induce ASM cell mitogenesis by activating receptors with intrinsic receptor tyrosine kinase (RTK) activity, whereas contractile agonists released from inflammatory cells mediate their effects by activation of GPCRs.

asm as an immune-response regulator

57

Despite disparate receptor-operated mechanisms, recent evidence suggests that the small GTPase, p21ras, acts as a point of convergence for diverse extracellular signal-stimulated pathways in ASM cells (Fig. 2) [73]. In their active state, p21ras proteins interact with downstream effectors, namely, Raf-1 and phosphatidylinositol 3-kinase (PI3K). By recruiting Raf-1 to the plasma membrane, guanosine triphosphate bound p21ras activates the extracellular signal-regulated kinase (ERK) pathway. p21ras also binds and activates PI3K [74]. Synergy can occur between RTK and GPCRs to promote human ASM mitogenesis and p21ras activation [75]. Although alternative pathways do exist (as discussed later), PI3K and ERK activation seems to be the dominant signal transduction pathways for RTK-, GPCR- or cytokine-stimulated growth of ASM cells. PI3K phosphorylates membrane phosphoinositides, which function as second messengers and activate downstream effector molecules, such as 70-kd ribosomal S6 kinase (p70S6k), Rac1, or Cdc42 to regulate cell cycle protein expression and thus modulate cell cycle traversal [76]. Activation of PI3K is

Fig. 2. Intracellular pathways regulating ASM cell mitogenesis. Ras serves as a point of convergence for growth factor receptor tyrosine kinase (RTK) and G-protein coupled receptor (GPCR) signaling. Ras activates the extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase (PI3K), and ultimately leads to activation of cyclins and cyclin-dependent kinases (CDKs). MEK, mitogen-activated protein kinase/ERK kinase; p70S6K, ribosomal S6 kinase; ROS, reactive oxygen species.

critical for ASM cell cycle progression [77 79]; however, although active PI3K is sufficient to stimulate ASM DNA synthesis, other signaling events are also necessary to promote maximal ASM growth responses [80]. As mentioned previously, Raf-1 activation induces phosphorylation and activation of mitogenactivated protein (MAP) kinase/ERK kinase (MEK1). Activated MEK1 directly activates the 42-kd ERK2 and 44-kd ERK1, collectively referred to as p42/p44 MAP kinases. Inhibition of MEK1 and ERK activity attenuates mitogen-induced DNA synthesis in ASM cells, suggesting that activation of MEK1 and ERK is required for proliferation [81,82]. Studies such as these suggest that the ERK pathway is a key signaling event mediating mitogen-induced ASM proliferation. D-type cyclins (cyclins D1, D2, and D3) are key regulators of G1 progression in mammalian cells; consequently, cyclin D1 has been the most widely studied cyclin in ASM biology. Another critical protein is p27Kip1, a cell cycle inhibitor [83]. A coordinated increase of cyclin D1 expression promotes complexing of unbound p27Kip1 molecules with cyclin D-dependent kinases, thereby facilitating cyclin E cyclin-dependent kinase-2 activation later in the G1 phase (Fig. 3) [83]. Recent studies also have implicated the Rho GTPases, Rac1, Cdc42, and RhoA in cyclin D1 regulation and ASM growth. Over expression of the catalytically active subunit of PI3K was sufficient to activate the cyclin D1 promoter and could be attenuated by inhibitors of Rac1 signaling [84], suggesting that Rac1 may be downstream of PI3K; further study is necessary to confirm this observation. Other studies using overexpression constructs of Cdc42, RhoA, and Rac1 also showed that overexpression of Cdc42 or Rac1, but not of RhoA, induced transcription from the cyclin D1 promoter in an ERK-independent manner [85,86]. Although cyclin D1 expression may be necessary for cell growth, it is not sufficient. Amrani and colleagues [87] demonstrated that IFNg inhibits ASM growth without modulating the growth factor induced increased expression of cyclin D1. In human ASM, corticosteroids reduce mitogen-stimulated increases in cyclin D1 protein and attenuate pRb phosphorylation [88]. Corticosteroid binding to the glucocorticoid receptor results in activation of the transcription factor C/EBPa and subsequent increased expression of cyclin-dependent kinase inhibitor p21WAF/Cip1, thus providing a potential mechanism for the antiproliferative effect of glucocorticoids on ASM cell growth [89 91]. A recent study suggests that ASM cells derived from patients

58

lazaar

&

panettieri, jr

Fig. 3. Activation of cell-cycle progression in ASM cells. Activation of cyclin D1 by growth factors or G-protein coupled receptor (GPCR) agonists leads to phosphorylation of retinoblastoma protein (Rb) and cell cycle progression. E2F, elongation factor 2; RTK, growth factor receptor tyrosine kinase.

who have asthma lack expression of C/EBPa and are therefore resistant to the effects of glucocorticoids on proliferation [92]. Despite evidence suggesting that oxidative stress plays an important part in regulating vascular smooth muscle [93], less is known about the role of reactive oxygen species in ASM cell function. Brar and colleagues [94] demonstrated that ASM cells express several components of the phagocyte NADPH, including gp22phox, gp91phox, and gp67phox. Reactive oxygen species can modulate mitogen-induced ASM cell proliferation [86,95,96]. Antioxidants inhibit mitogen-induced growth [95,96], whereas expression of a constitutively active Rac1 (another component of the NADPH oxidase complex) results in cyclin D1 expression [86]. Decreased expression of gp22phox by antisense oligonucleotides also inhibits ASM cell proliferation [94]. Stat3 also has been shown to be important in PDGF-induced pro-

liferation of human ASM cells. Activation of the JAK2 and Stat3 by PDGF seems to be redox dependent and affects the proliferative response to mitogen, independent of MAP kinase activation [97]. IFNg and IFNb activate Stat1/2, JAK1, and Tyk2 in ASM; these cytokines, however, are potent inhibitors of mitogen-induced proliferation, in part through increased expression of interferon-gamma-inducible protein 16 (IFI 16) [67,87]. Hypertrophy There is renewed attention to the role of increased myocyte size or hypertrophy as a mechanism of increased ASM cell mass in asthma. Some mediators, such as IL-1b, IL-6, TGFb, angiotensin II, and cardiotrophin I, induce cellular hypertrophy in vitro, although the mechanisms remain unclear [62,98,99]. In a primate model of asthma, the airways of mon-

asm as an immune-response regulator

59

keys sensitized to house dust mite antigen display an increase in smooth muscle mass as well as in smooth muscle bundle size [100]. Morphologic studies confirm the presence of ASM cell hypertrophy in some, but not all, patients who have asthma [51]. Increased cell size seems to correlate negatively with postbronchodilator forced expiratory volume in 1 second and distinguishes between severe persistent asthma and milder disease [101]. In contrast, Woodruff and colleagues [56] studied ASM cell gene expression and cell morphology in bronchial biopsies obtained from patients who had mild to moderate asthma. These investigators found no evidence for ASM cell hypertrophy, although they did confirm the presence of ASM hyperplasia. More work is needed in this particular area of ASM cell biology. Smooth muscle migration Hyperplasia and hypertrophy are important processes regulating increased smooth muscle mass in asthma. Analogous to vascular smooth muscle migration in atherosclerosis, however, ASM cell migration probably promotes airway remodeling in chronic asthma. Evidence suggests that proliferating smooth muscle cells migrate along chemotactic gradients. During chronic inflammation, myocyte migration would be promoted by exposure of cells to a variety of cytokines and growth factors, as well as to an altered ECM. Structurally, myofibroblasts display a phenotype intermediate between fibroblasts and smooth muscle cells, express a-smooth muscle actin, and have the ability to secrete matrix proteins such as collagen and hyaluronan. In addition, myofibroblasts secrete chemokines and prolong eosinophil survival [102]. Although myofibroblasts are found in the lamina reticularis, the origin of these cells remains unknown. Possibly the cells exist in small numbers within the lamina reticularis and proliferate locally following stimulation with inflammatory mediators or growth factors. Alternatively, under the influence of similar stimuli, smooth muscle cells might migrate from the periphery of the smooth muscle bundle into the submucosa. A final possibility is that cells are recruited from a circulating pool of fibrocytes, as has been described by Schmidt and colleagues [103]. In this study, CD34+ cells expressing procollagen I and a-smooth muscle actin were increased in the bronchial mucosa following allergen challenge in patients who had asthma. Using a murine model of chronic allergic airway inflammation, the investigators demonstrated that these cells were recruited from a circulating population of CD34+ fibrocytes.

The potential for mesenchymal cells such as myofibroblasts to migrate into the airway wall is well established in patients who have asthma [104,105]. BAL-derived fibroblasts from patients who have mild asthma have enhanced migratory capacity compared with tissue fibroblasts isolated from bronchial biopsies [106]. In vitro, many studies support the capacity of ASM cells to migrate. Increased chemotaxis of ASM cells has been demonstrated to PDGF, TGFb, basic fibroblastic growth factor, and IL-1b. Urokinase has been shown to increase chemotaxis in some, but not all, studies [107,108]. Cysteinyl-leukotrienes alone promote chemokinesis only; however, in combination with PDGF, increased chemotaxis is observed over PDGF alone [109]. Vascular endothelial growth factor (VEGF), a potent inducer of vascular smooth muscle migration, has no effect on ASM cell chemotaxis [110,111]. Similarly, not all mitogens are promigratory; recent data demonstrate that only PDGF, but not EGF or thrombin, induce migration in ASM cells [112]. The ECM also regulates the migratory phenotype, but its role in ASM migration has not been extensively characterized. Parameswaran and colleagues [113] recently demonstrated that ASM cells migrate more on collagen III and V and fibronectin than on collagen I, elastin, or laminin. The signaling pathways regulating ASM cell migration are beginning to be elucidated. Early studies suggested that p38 MAP kinase and heat shock protein 27 are important regulators of growth factor and cytokine-induced migration [114]. Activation of p38 MAP kinase seems to occur, in part, through p21-activated protein kinases [115] and is important for phosphorylation of caldesmon, which acts to regulate actin myosin interactions [116]. Subsequent studies have demonstrated that PI3K also regulates PDGF-induced smooth muscle cell migration [110]. PI3K is activated in part through activation of Src kinase, because inhibition of Src attenuates PI3K activity [112]. Others have shown that ASM migration on collagen I, but not on elastin or laminin, is associated with activation of Src [113]. These observations suggest that PDGF-induced migration optimally requires activation of both Src and p38 MAP kinase.

Synthetic function of airway smooth muscle The previous discussion has focused on the influence of cytokines and growth factors on ASM contractility and proliferation, but ASM cells also express a variety of secreted cytokines, chemokines,

60 Table 1 Airway smooth muscle cell derived factors Cytokines IL-1b IL-6 IL-5 IL-11 TNFa IFNb LIF Chemokines Eotaxin IL-8 MCP-1, 2, 3 GM-CSF RANTES TARC SCF Fractalkine IP-10 Growth factors PDGF TGFb VEGF CTGF IGF Cardiotrophin-1

lazaar

&

panettieri, jr

Cell adhesion molecules Integrins ICAM-1 VCAM-1 CD44

Extracellular matrix Hyaluronan Collagen types I, III, V Laminin Tenascin Fibronectin Perlecan Versican Decorin Thrombospondin Proteoglycan

Matrix metalloproteinase MMP-1 MMP-2 MMP-3 MMP-9 MT-MMP1 TIMP-1 TIMP-2 ADAM33

Other CD40 MHC II PGE2 NOS

Abbreviation: IP-10, interferon-inducible protein 10.

and immunomodulatory proteins; this expression is termed synthetic function (Table 1). The synthetic function of ASM may play a considerable role in modulating airway inflammation and remodeling in asthma.

Extracellular matrix ECM proteins are critical for maintaining the structure and function of the airways. ASM cells, by producing ECM components as well as matrixmodifying enzymes, may contribute to airway remodeling. Alterations in the ECM, in turn, modify the growth and synthetic function of ASM cells. The composition of the ECM is tightly controlled and involves a dynamic process of matrix deposition and degradation. In inflammatory processes such as asthma, this balance is disturbed, resulting in an abnormal amount of matrix deposition and also in an altered composition of matrix components. ASM cells secrete a wide variety of matrix proteins, including fibronectin, collagen, hyaluronan, laminin, and versican. In asthma, there is an increase in hyaluronan, fibronectin, tenascin, versican, laminin, and collagen types I, III, and V [117,118]. In vitro, serum from asthmatic patients increases smooth muscle cell release of fibronectin, laminin, perlecan, and chondroitin sulfate [71]. Treatment of cells with corticosteroids had no effect on the production of matrix proteins by ASM cells [71]. This finding has been borne out in human studies in which inhaled corticosteroids had minimal effect on altering ECM composition in asthmatics [119]. TGFb is secreted from ASM cells and, in turn, induces smooth muscle cell synthesis of hyaluronan and collagen [98,120,121]. The mechanism of this

effect probably involves induction of connective tissue growth factor (CTGF) by TGFb [122]. Recent studies have demonstrated that ASM cells also secrete VEGF, a potent endothelial mitogen [111,123]. In vitro, VEGF increases fibronectin expression by ASM but has no effect on migration or proliferation [111]. Finally, LTD4 and EGF increase expression of versican and fibronectin in ASM cells [124]. Increased matrix deposition seen in asthma is probably caused by increased secretion by mesenchymal cells as well as by an imbalance between matrix-degrading enzymes and inhibitors of these proteases. One class of proteins that has been intensively studied is the matrix metalloproteinase (MMP) family. MMP-1 expression is elevated in the smooth muscle cells of patients who have asthma, and studies have shown that LTD4 increases expression of MMP-1, which acts to degrade insulin-like growth factor binding protein, a growth inhibitor [125]. Progelatinase A (MMP-2) is constitutively released by ASM cells but remains inactive because of high levels of tissue inhibitor of metalloproteinases (TIMP)-2 on the cell membrane [126], although thrombin has been shown to activate MMP-2 [127]. In contrast, TIMP-1 is secreted in large amounts into the conditioned media of ASM cells [126]. Membrane type 1 MMP is also found on ASM [126,127]; this proteinase can activate MMP-2 and has been shown to cleave CD44 from the cell surface and promote cell migration [128]. ASM cells also express pro- and active MMP-3 [127], whereas TNFa induces the release of MMP-9, which can degrade matrix but also plays a critical role in cleaving latent TGF-b to its active form [129]. ADAMs (a disintegrinase and metalloproteinase) are a subfamily of metalloproteinases located on the cell surface. Polymorphisms in the ADAM-33 gene have been asso-

asm as an immune-response regulator

61

ciated with airway hyperresponsiveness and asthma [130]. ADAM-33 is expressed by ASM cells and lung fibroblasts [130], although there does not seem to be a significant difference in ADAM-33 protein in persons who have asthma and controls [131]. The composition of the ECM also influences ASM cell function. For example, PDGF-induced proliferation is enhanced by monomeric collagen I, fibronectin, and vitronectin, whereas other ECM components, such as fibrillar collagen I, collagen III, and tenascin-C, have no effect [132]. Others have shown that ECM proteins secreted by asthmatic ASM differ from nonasthmatic ASM. Cells plated on the ECM derived from asthmatic ASM displayed greater rates of proliferation; this finding was true for both nonasthmatic and asthmatic ASM cells [72]. In a study examining the effect of ECM composition on b2adrenoceptor signaling in human ASM cells, the investigators demonstrated that fibronectin increased, whereas collagen V and laminin decreased, accumulation of the second messenger cyclic AMP when compared with collagens I or IV [133]. This finding suggests that altered ECM might affect responses to bronchodilator therapy in patients who have asthma.

motes airway hyporesponsiveness, suggesting a complex role for IL-6 in modulating local inflammation and regulating airway reactivity [142,143]. Other IL-6 family cytokines, such as leukemia inhibitory factor (LIF) and IL-11 but not oncostatin M, are secreted following exposure of ASM cells to viral particles [144]. T-helper (Th)-2 cytokines IL-4, IL-13, and to some extent IL-9 have profound effects on ASM cell synthetic function through induction of a number of signaling molecules and contractile proteins [145,146]. The Th2 cytokines IL-4 and IL-13 induce eotaxin and, in combination with TNFa, TARC expression [69]. In contrast, IL-4 and -13 have no effect on GM-CSF, RANTES, or IL-8 secretion by ASM [147]. IL-5 and other Th2 cytokines may also play a role in sensitizing ASM cells to the effects of contractile agonists, independent of their effects on airway eosinophilia [146]. More controversial, perhaps, is the potential for ASM cells to secrete IL-5 and IL-13 [13].

Immunomodulatory proteins Cell adhesion molecules (CAMs) mediate leukocyte endothelial cell interactions during the process of cell recruitment and homing [148]. Evidence suggests that CAMs mediate inflammatory cell stromal cell interactions that may contribute to airway inflammation. ASM cells express intracellular adhesion molecule-1 (ICAM-1) and VCAM-1, which are inducible by a wide range of inflammatory mediators, and constitutively express CD44, a hyaluronan receptor [149]. Recent data suggest that ASM cells express variable levels of integrin subunits, with the av, a5, and b1 subunits predominating [150]. b1 subunits are important in mediating enhanced chemokine release by ASM cells adherent to fibronectin or collagen I [151]. CAMs also function as accessory molecules for leukocyte activation [152]. Whether CAMs expressed on smooth muscle serve this function remains controversial, however. Although ASM cells do express major histocompatibility complex (MHC) class II and CD40 following stimulation with IFNg [153,154], and a recent study suggests that coculture of T cells with ASM increases T-cell expression of CD25 [155], the physiologic relevance of these findings remains unknown, because ASM cells are unable to present alloantigen to CD4 T cells [153]. Functionally, however, adhesion of stimulated CD4 T cells induces smooth muscle cell DNA syn+ thesis [149,156] Ligation of CD40 increases Ca2 , i

Chemokine and cytokine release Many chemokines, which act to recruit and activate leukocytes, are found in the BAL fluid and lung tissue of patients who have asthma. ASM cells have been shown to secrete RANTES (regulated on activation, normal T-cell expressed and secreted), eotaxin, IL-8, monocyte chemoattractant protein (MCP)-1, -2, and -3, thymus and activation-regulated chemokine (TARC), and granulocyte macrophage colony-stimulating factor (GM-CSF) in response to TNFa and IL-1b (see Table 1). ASM cells may play a role in promoting both the recruitment and survival of eosinophils by secretion of GM-CSF and IL-5 [134 136]. ASM infiltration by mast cells also has been shown in patients who have asthma [137]. In addition to eotaxin, ASM cells secrete stem cell factor (SCF) [138], which acts to recruit and retain mast cells within the smooth muscle layer. Release of lipid mediators and enzymes by activated mast cells probably promotes AHR and either inhibits (chymase) or promotes (tryptase) ASM cell proliferation [139 141]. IL-6 secretion is inducible by multiple stimuli, including TNFa, IL-1b, and bradykinin, and has autocrine effects on smooth muscle cell hyperplasia [62]. Transgenic expression of IL-6 in the murine lung induces peribronchiolar inflammation but pro-

62

lazaar

&

panettieri, jr

whereas engagement of VCAM-1 on ASM cells augments growth factor induced ASM cell proliferation [154,157]. Enhancement of PDGF-induced proliferation by either collagen I or fibronectin requires a2b1, a4b1, and a5b1 integrins [132]; similarly, blocking a5b1 integrin on ASM cells enhances apoptosis [150]. Investigators have demonstrated that ASM cells express Fas in vivo and in vitro, and crosslinking of Fas also induces smooth muscle cell apoptosis [158]. Adhesion of activated T cells also seems to alter smooth muscle cell contractility in a contactdependent manner [155], potentially because of the autocrine secretion of IL-5 and IL-1b [159]. Others have shown that ASM cells cultured with activated T cells show increased calcium mobilization responses to LTD4 and serotonin [160]. These studies highlight the finding that direct interactions between leukocytes and smooth muscle cells through immune or adhesion receptors contribute to the modulation of the local milieu resulting in smooth muscle cell activation and growth. Increased amounts of exhaled nitric oxide have been detected in patients who have asthma [161]. Nitric oxide seems to have a selective suppressive effect on the Th1 subset of Th cells, suggesting that increased levels of nitric oxide may therefore lead to the predominantly Th2-type response associated with asthma. Nitric oxide synthase (NOS) has been demonstrated in cultured ASM cells, where it results in an inhibition of ASM cell proliferation [162,163], and has been localized to ASM cells in patients who have asthma [164]. In an animal model, expression of neural nitrous oxide synthase is decreased following allergen challenge [165]. ASM cells also secrete PGE2 and, to a lesser extent, other prostanoids following stimulation with proinflammatory cytokines [166]. PGE2, a potent bronchodilator, also has significant immunologic effects. For example, PGE2 can decrease expression of CD23 (FcgRII), and may have a role as a negative regulator of airway inflammation and hyperresponsiveness [167 169]. PGE2 inhibits cytokine-induced secretion of GM-CSF, RANTES, and CAMs in vitro [170 172] and allergen-induced release of PGD2 in patients who have asthma [173]. In contrast, PGE2 increases IL-6 secretion by ASM cells [174], primes dendritic cells toward a Th2-promoting capacity, and synergizes with IL-4 to induce IgE synthesis [175,176]. Receptors for the complement-derived anaphylatoxin peptides C3a and C5a have also been described on ASM cells. C3a stimulates enhanced mast cell degranulation following cell cell contact with ASM

cells [177]. These peptides may play an important role in the pathogenesis of asthma by altering airway hyperresponsiveness rather than airway inflammation [178,179]. Finally, new data suggest that immunostimulatory DNA sequences containing the CpG motif induce Th1-type inflammation, thus suppressing Th2-type responses that are characteristic of asthma (reviewed in Ref. [180]). These DNA sequences bind to Toll-like receptors, part of the innate immune system, and show promise as potential immunotherapy for airway remodeling [180]. ASM cells express several Toll-like receptors, including Toll-like receptor 9, the receptor for CpG (A. Lazaar, unpublished data), suggesting that ASM cells play a role in innate immunity. ASM cells provide a rich source of cytokines and chemokines and, under certain conditions, can express a wide variety of adhesion receptors, costimulatory molecules, and other immunomodulatory proteins (see Table 1). Taken together, these data provide strong support for the potential role of ASM cells in perpetuating airway inflammation and in leukocyte activation.

Summary The biology of ASM is complex and fascinating. The myriad pathways regulating cell growth and proliferation, combined with the expanding understanding of the role of ASM as a modulator of inflammation, provide a rich area of investigation. Future studies will focus on mechanisms regulating acute inflammation and the chronic repair processes resulting in airway remodeling. These studies may provide the basis for the development of new therapeutic agents aimed at modulating the pathophysiological changes associated with asthma.

References
[1] Schmidlin F, Scherrer D, Daeffler L, et al. Interleukin1b induces bradykinin B2 receptor gene expression through a prostanoid cyclic AMP dependent pathway in human bronchial smooth muscle cells. Mol Pharmacol 1998;53:1009 15. [2] Hsu Y, Chiu C, Wang C, et al. Tumour necrosis factor-a enhances bradykinin-induced signal transduction via activation of Ras/Raf/MEK/MAPK in canine tracheal smooth muscle cells. Cell Signal 2001; 13:633 43. [3] Zhang Y, Adner M, Cardell LO. Up-regulation of bradykinin receptors in a murine in-vitro model of

asm as an immune-response regulator chronic airway inflammation. Eur J Pharmacol 2004; 489(1 2):117 26. Bryborn M, Adner M, Cardell LO. Interleukin-4 increases murine airway response to kinins, via upregulation of bradykinin B1-receptors and altered signalling along mitogen-activated protein kinase pathways. Clin Exp Allergy 2004;34(8):1291 8. Kim JH, Jain D, Tliba O, et al. TGF-b potentiates airway smooth muscle responsiveness to bradykinin. Am J Physiol Lung Cell Mol Physiol 2005;289(4): L511 20 [Epub 2005 May 27]. Amrani Y, Martinet N, Bronner C. Potentiation by tumour necrosis factor-a of calcium signals induced by bradykinin and carbachol in human tracheal smooth muscle cells. Br J Pharmacol 1995;114:4 5. Hotta K, Emala CW, Hirshman CA. TNF-a upregulates Gia and Gqa protein expression and function in human airway smooth muscle cells. Am J Physiol 1999;276(20):L405 11. Schmidt D, Ruehlmann E, Branscheid D, et al. Passive sensitization of human airways increases responsiveness to leukotriene C4. Eur Respir J 1999;14: 315 9. Black JL, Marthan R, Armour CL, et al. Sensitization alters contractile responses and calcium influx in human airway smooth muscle. J Allergy Clin Immunol 1989;84:440 7. Marthan R, Crevel H, Guenard H, et al. Responsiveness to histamine in human sensitized airway smooth muscle. Respir Physiol 1992;90:239 50. Roux E, Hyvelin JM, Savineau JP, et al. Calcium signaling in airway smooth muscle cells is altered by in vitro exposure to the aldehyde acrolein. Am J Respir Cell Mol Biol 1998;19:437 44. Amrani Y, Panettieri Jr RA. Modulation of calcium homeostasis as a mechanism for altering smooth muscle responsiveness in asthma. Curr Opin Allergy Clin Immunol 2002;2(1):39 45. Grunstein MM, Hakonarson H, Leiter J, et al. IL-13dependent autocrine signaling mediates altered responsiveness of IgE-sensitized airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2002; 282(3):L520 8. Rizzo CA, Yang R, Greenfeder S, et al. The IL-5 receptor on human bronchus selectively primes for hyperresponsiveness. J Allergy Clin Immunol 2002; 109(3):404 9. Tliba O, Deshpande D, Chen H, et al. IL-13 enhances agonist-evoked calcium signals and contractile responses in airway smooth muscle. Br J Pharmacol 2003;140(7):1159 62. Holgate ST, Peters-Golden M, Panettieri RA, et al. Roles of cysteinyl leukotrienes in airway inflammation, smooth muscle function, and remodeling. J Allergy Clin Immunol 2003;111(1 Suppl):S18 34 [discussion: S34 6]. Lynch KR, ONeill GP, Liu Q, et al. Characterization of the human cysteinyl leukotriene CysLT1 receptor. Nature 1999;399:789 93.

63

[4]

[5]

[6]

[7]

[8]

[9]

[10]

[11]

[12]

[13]

[14]

[15]

[16]

[17]

[18] Heise CE, ODowd BF, Figueroa DJ, et al. Characterization of the human cysteinyl leukotriene 2 receptor. J Biol Chem 2000;275:30531 6. [19] Amrani Y, Moore PE, Hoffman R, et al. Interferon-g modulates cysteinyl leukotriene receptor-1 expression and function in human airway myocytes. Am J Respir Crit Care Med 2001;164:2098 101. [20] Shore SA. Cytokine regulation of beta-adrenergic responses in airway smooth muscle. J Allergy Clin Immunol 2002;110(6 Suppl):S255 60. [21] Somlyo AP, Somlyo AV. Signal transduction and regulation in smooth muscle. Nature 1994;372:231 6. [22] Yang CM, Chien CS, Wang CC, et al. Interleukin-1b enhances bradykinin-induced phosphoinositide hydrolysis and Ca2 + mobilization in canine tracheal smooth-muscle cells: involvement of the Ras/Raf/ mitogen-activated protein kinase (MAPK) kinase (MEK)/MAPK pathway. Biochem J 2001;354:439 46. [23] Pype JL, Xu H, Schuermans M, et al. Mechanisms of interleukin 1b-induced human airway smooth muscle hyporesponsiveness to histamine. Involvement of p38 MAPK NF-kB. Am J Respir Crit Care Med 2001;163:1010 7. [24] Amrani Y, Panettieri Jr RA. Cytokines induce airway smooth muscle cell hyperresponsiveness to contractile agonists. Thorax 1998;53:713 6. [25] Amrani Y, Krymskaya V, Maki C, et al. Mechanisms underlying TNFa effects on agonist-mediated calcium homeostasis in human airway smooth muscle cells. Am J Physiol 1997;273:L1020 8. [26] Madison JM, Ethier MF. Interleukin-4 rapidly inhibits calcium transients in response to carbachol in bovine airway smooth muscle cells. Am J Respir Cell Mol Biol 2001;25:239 44. [27] Ethier MF, Cappelluti E, Madison JM. Mechanisms of interleukin-4 effects on calcium signaling in airway smooth muscle cells. J Pharmacol Exp Ther 2005; 313(1):127 33. [28] Wylam ME, Gungor N, Mitchell RW, et al. Eosinophils, major basic protein, and polycationic peptides augment bovine airway myocyte Ca2 + mobilization. Am J Physiol 1998;274:L997 1005. [29] Takata Y, Nishimura Y, Maeda H, et al. Phospholipase A2 augments contraction and intracellular calcium mobilization through thromboxane A2 in bovine tracheal smooth muscle. Eur Respir J 1999;14: 396 404. [30] Somlyo AP, Somlyo AV. Ca2 + sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003;83(4):1325 58. [31] Wettschureck N, Offermanns S. Rho/Rho-kinase mediated signaling in physiology and pathophysiology. J Mol Med 2002;80(10):629 38. [32] Mehta D, Tang DD, Wu MF, et al. Role of Rho in Ca(2 +)-insensitive contraction and paxillin tyrosine phosphorylation in smooth muscle. Am J Physiol 2000;279:C308 18. [33] Hunter I, Cobban HJ, Vandenabeele P, et al. Tumor

64

lazaar

&

panettieri, jr [47] Hirshman CA, Emala CW. Actin reorganization in airway smooth muscle cells involves Gq and Gi-2 activation of Rho. Am J Physiol 1999;277(3 Pt 1): L653 61. [48] Yamboliev IA, Hedges JC, Mutnick JL, et al. Evidence for modulation of smooth muscle force by the p38 MAP kinase/HSP27 pathway. Am J Physiol Heart Circ Physiol 2000;278(6):H1899 907. [49] Gerthoffer WT, Gunst SJ. Invited review: focal adhesion and small heat shock proteins in the regulation of actin remodeling and contractility in smooth muscle. J Appl Physiol 2001;91(2):963 72. [50] Fernandes DJ, Mitchell RW, Lakser O, et al. Do inflammatory mediators influence the contribution of airway smooth muscle contraction to airway hyperresponsiveness in asthma? J Appl Physiol 2003; 95(2):844 53. [51] Ebina M, Takahashi T, Chiba T, et al. Cellular hypertrophy and hyperplasia of ASM underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis 1993;148:720 6. [52] Jeffery PK. Morphology of the airway wall in asthma and in chronic obstructive pulmonary disease. Am Rev Respir Dis 1991;143:1152 8. [53] Hirst SJ, Martin JG, Bonacci JV, et al. Proliferative aspects of airway smooth muscle. J Allergy Clin Immunol 2004;114(2 Suppl):S2 17. [54] Johnson PR, Roth M, Tamm M, et al. Airway smooth muscle cell proliferation is increased in asthma. Am J Respir Crit Care Med 2001;164:474 7. [55] Benayoun L, Letuve S, Druilhe A, et al. Regulation of peroxisome proliferator-activated receptor gamma expression in human asthmatic airways: relationship with proliferation, apoptosis, and airway remodeling. Am J Respir Crit Care Med 2001;164(8 Pt 1): 1487 94. [56] Woodruff PG, Dolganov GM, Ferrando RE, et al. 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(9): 1001 6. [57] Naureckas ET, Ndukwu IM, Halayko AJ, et al. Bronchoalveolar lavage fluid from asthmatic subjects is mitogenic for human airway smooth muscle. Am J Respir Crit Care Med 1999;160(6):2062 6. [58] Cohen MD, Ciocca V, Panettieri Jr RA. TGF-b1 modulates human airway smooth muscle cell proliferation induced by mitogens. Am J Respir Cell Mol Biol 1997;16:85 90. [59] Espinosa K, Bosse Y, Stankova J, et al. 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(5):1032 40. [60] Ward JE, Gould H, Harris T, et al. PPARg ligands, 15-deoxy-D12,14-prostaglandin J2 and rosiglitazone regulate human cultured airway smooth muscle proliferation through different mechanisms. Br J Pharmacol 2004;141:517 25.

[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

[42]

[43]

[44]

[45]

[46]

necrosis factor-alpha-induced activation of RhoA in airway smooth muscle cells: role in the Ca2 + sensitization of myosin light chain20 phosphorylation. Mol Pharmacol 2003;63(3):714 21. Chiba Y, Sakai H, Misawa M. Augmented acetylcholineinduced translocation of RhoA in bronchial smooth muscle from antigen-induced airway hyperresponsive rats. Br J Pharmacol 2001;133(6):886 90. Henry PJ, Mann TS, Goldie RG. A rho kinase inhibitor, Y-27632 inhibits pulmonary eosinophilia, bronchoconstriction and airways hyperresponsiveness in allergic mice. Pulm Pharmacol Ther 2005; 18:67 74. Schaafsma D, Gosens R, Bos IS, et al. Allergic sensitization enhances the contribution of Rho-kinase to airway smooth muscle contraction. Br J Pharmacol 2004;143:477 84. White TA, Kannan MS, Walseth TF. Intracellular calcium signaling through the cADPR pathway is agonist specific in porcine airway smooth muscle. FASEB J 2003;17(3):482 4. Kannan MS, Fenton AM, Prakash YS, et al. Cyclic ADP-ribose stimulates sarcoplasmic reticulum calcium release in porcine coronary artery smooth muscle. Am J Physiol 1996;270(2 Pt 2):H801 6. Wang YX, Zheng YM, Mei QB, et al. FKBP12.6 and cADPR regulation of Ca2 + release in smooth muscle cells. Am J Physiol 2004;286(3):C538 46. Deshpande DA, Dogan S, Walseth TF, et al. Modulation of calcium signaling by interleukin-13 in human airway smooth muscle: role of CD38/cyclic adenosine diphosphate ribose pathway. Am J Respir Cell Mol Biol 2004;31(1):36 42. Tliba O, Panettieri Jr RA, Tliba S, et al. TNF-a differentially regulates the expression of pro-inflammatory genes in human airway smooth muscle cells by activation of IFN-b-dependent CD38 pathway. Mol Pharmacol 2004;66:322 9. Deshpande DA, Walseth TF, Panettieri RA, et al. CD38/cyclic ADP-ribose-mediated Ca2 + signaling contributes to airway smooth muscle hyper-responsiveness. FASEB J 2003;17(3):452 4. Deshpande DA, White TA, Guedes AG, et al. Altered airway responsiveness in CD38-deficient mice. Am J Respir Cell Mol Biol 2005;32(2):149 56. Ammit AJ, Armour CL, Black JL. Smooth-muscle myosin light-chain kinase content is increased in human sensitized airways. Am J Respir Crit Care Med 2000;161:257 63. Ma X, Cheng Z, Kong H, et al. Changes in biophysical and biochemical properties of single bronchial smooth muscle cells from asthmatic subjects. Am J Physiol Lung Cell Mol Physiol 2002;283(6): L1181 9. Wojciak-Stothard B, Entwistle A, Garg R, et al. Regulation of TNF-alpha-induced reorganization of the actin cytoskeleton and cell-cell junctions by Rho, Rac, and Cdc42 in human endothelial cells. J Cell Physiol 1998;176(1):150 65.

asm as an immune-response regulator [61] Broide DH, Lotz M, Cuomo AJ, et al. Cytokines in symptomatic asthma airways. J Allergy Clin Immunol 1992;89:958 67. [62] De S, Zelazny ET, Souhrada JF, et al. IL-1b and IL-6 induce hyperplasia and hypertrophy of cultured guinea pig airway smooth muscle cells. J Appl Physiol 1995;78:1555 63. [63] Zhai W, Eynott PR, Oltmanns U, et al. Mitogenactivated protein kinase signalling pathways in IL-1 beta-dependent rat airway smooth muscle proliferation. Br J Pharmacol 2004;143(8):1042 9. [64] Belvisi MG, Saunders M, Yacoub M, et al. Expression of cyclo-oxygenase-2 in human airway smooth muscle is associated with profound reductions in cell growth. Br J Pharmacol 1998;125:1102 8. [65] McKay S, Hirst SJ, Bertrand-de Haas M, et al. Tumor necrosis factor-a enhances mRNA expression and secretion of interleukin-6 in cultured human airway smooth muscle cells. Am J Respir Cell Mol Biol 2000;23:103 11. [66] Stewart AG, Tomlinson PR, Fernandes DJ, et al. Tumor necrosis factor a modulates mitogenic responses of human cultured airway smooth muscle. Am J Respir Cell Mol Biol 1995;12:110 9. [67] Tliba O, Tliba S, Huang CD, et al. TNFa modulates airway smooth muscle function via the autocrine action of IFNb. J Biol Chem 2003;278(50):50615 23. [68] Chambers LS, Black JL, Ge Q, et al. PAR-2 activation, PGE2, and COX-2 in human asthmatic and nonasthmatic airway smooth muscle cells. Am J Physiol 2003;285:L619 27. [69] Howarth PH, Knox AJ, Amrani Y, et al. Synthetic responses in airway smooth muscle. J Allergy Clin Immunol 2004;114(2 Suppl):S32 50. [70] Hirst SJ, Twort CHC, 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 44. [71] Johnson PRA, Black JL, Cralin S, et al. The production of extracellular matrix proteins by human passively sensitized airway smooth-muscle cells in culture: the effect of beclomethasone. Am J Respir Crit Care Med 2000;162:2145 51. [72] Johnson PR, Burgess JK, Underwood PA, et al. Extracellular matrix proteins modulate asthmatic airway smooth muscle cell proliferation via an autocrine mechanism. J Allergy Clin Immunol 2004; 113:690 6. [73] Ammit AJ, Kane SA, Panettieri Jr RA. Activation of K-p21ras and N-p21ras, but not H-p21ras, is necessary for mitogen-induced human airway smooth muscle proliferation. Am J Respir Cell Mol Biol 1999;21:719 27. [74] Rodriguez-Viciana P, Warne PH, et al. Phosphatidylinositol-3-OH kinase as a direct target of ras. Nature 1994;370:527 32. [75] Krymskaya VP, Orsini MJ, Eszterhas AJ, et al. Mechanisms of proliferation synergy by receptor tyrosine kinase and G protein-coupled receptor activa-

65

[76] [77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87]

[88]

[89]

tion in human airway smooth muscle. Am J Respir Cell Mol Biol 2000;23:546 54. Cantley LC. The phosphoinositide 3-kinase pathway. Science 2002;296(5573):1655 7. Krymskaya VP, Penn RB, Orsini MJ, et al. Phosphatidylinositol 3-kinase mediates mitogen-induced human airways smooth muscle cell proliferation. Am J Physiol 1999;277(21):L65 78. Scott PH, Belham CM, Al-Hafidh J, 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 71. Walker TR, Moore SM, Lawson MF, et al. Plateletderived growth factor-BB and thrombin activate phosphoinositide 3-kinase and protein kinase B: Role in mediating airway smooth muscle proliferation. Mol Pharmacol 1998;54:1007 15. Krymskaya VP, Ammit AJ, Hoffman RK, et al. Activation of class IA phosphatidylinositol 3-kinase stimulates DNA synthesis in human airway smooth muscle cells. Am J Physiol 2001;280:L1009. Karpova AK, Abe MK, Li J, et al. MEK1 is required for PDGF-induced ERK activation and DNA synthesis in tracheal monocytes. Am J Physiol 1997; 272:L558 65. Orsini MJ, Krymskaya VP, Eszterhas AJ, et al. MAPK superfamily activation in human airway smooth muscle: mitogenesis requires prolonged p42/ p44 activation. Am J Physiol 1999;277(21):L479 88. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev 1999;13:1501 12. Page K, Li J, Wang Y, et al. Regulation of cyclin D(1) expression and DNA synthesis by phosphatidylinositol 3-kinase in airway smooth muscle cells. Am J Respir Cell Mol Biol 2000;23:436 43. Bauerfeld CP, Hershenson MB, Page K. Cdc42, but not RhoA, regulates cyclin D1 expression in bovine tracheal myocytes. Am J Physiol 2001;280:L974 82. Page K, Li J, Hodge JA, et al. Characterization of a Rac1 signaling pathway to cyclin D(1) expression in airway smooth muscle cells. J Biol Chem 1999;274: 22065 71. Amrani Y, Tliba O, Choubey D, et al. IFNg inhibits human airway smooth muscle cell proliferation by modulating the E2F 1/Rb pathway. Am J Physiol 2003;284:L1063 71. Fernandes D, Guida E, Koutsoubos V, et al. Glucocorticoids inhibit proliferation, cyclin D1 expression, and retinoblastoma protein phosphorylation, but not activity of the extracellular-regulated kinases in human cultured airway smooth muscle. Am J Respir Cell Mol Biol 1999;21:77 88. Timchenko N, Wilde M, Nakanishi M, et al. CCAAT/ enhancer-binding protein alpha (C/EBP alpha) inhibits cell proliferation through the p21 (WAF-1/CIP-1/ SDI-1) protein. Genes Dev 1996;10(7):804 15.

66

lazaar

&

panettieri, jr myofibroblasts in asthma. J Immunol 2003;171(1): 380 9. Brewster CEP, Howarth PH, Djukanovic R, et al. Myofibroblasts and subepitheial fibrosis in bronchial asthma. Am J Respir Cell Mol Biol 1990;3:507 11. Gizycki MJ, Adelroth E, Rogers AV, et al. Myofibroblast involvement in the allergen-induced late response in mild atopic asthma. Am J Respir Cell Mol Biol 1997;16:664 73. Larsen K, Tufvesson E, Malmstrom J, et al. Presence of activated mobile fibroblasts in bronchoalveolar lavage from patients with mild asthma. Am J Respir Crit Care Med 2004;170(10):1049 56. Mukhina S, Stepanova V, Traktouev D, et al. The chemotactic action of urokinase on smooth muscle cells is dependent on its kringle domain. Characterization of interactions and contribution to chemotaxis. J Biol Chem 2000;275(22):16450 8. Carlin SM, Roth M, Black JL. Urokinase potentiates PDGF-induced chemotaxis of human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2003;284(6):L1020 6. Parameswaran K, Cox G, Radford K, et al. Cysteinyl leukotrienes promote human airway smooth muscle migration. Am J Respir Crit Care Med 2002;166: 738 42. Irani C, Goncharova EA, Hunter DS, et al. Phosphatidylinositol 3-kinase but not tuberin is required for PDGF-induced cell migration. Am J Physiol 2002; 282:L854 62. Kazi AS, Lotfi S, Goncharova EA, et al. Vascular endothelial growth factor-induced secretion of fibronectin is ERK dependent. Am J Physiol 2004;286: L539 45. Krymskaya VP, Goncharova EA, Ammit AJ, et al. Src is necessary and sufficient for human airway smooth muscle cell proliferation and migration. FASEB J 2005;19:428 30. Parameswaran K, Radford K, Zuo J, et al. Extracellular matrix regulates human airway smooth muscle cell migration. Eur Respir J 2004;24(4): 545 51. Hedges JC, Dechert MA, Yamboliev IA, et al. A role for p38mapk/HSP27 pathway in smooth muscle cell migration. J Biol Chem 1999;274:24211 9. Dechert MA, Holder JM, Gerthoffer WT. p21activated kinase 1 participates in tracheal smooth muscle cell migration by signaling to p38 Mapk. Am J Physiol Cell Physiol 2001;281(1):C123 32. Goncharova EA, Vorotnikov AV, Gracheva EO, et al. Activation of p38 MAP-kinase and caldesmon phosphorylation are essential for urokinase-induced human smooth muscle cell migration. Biol Chem 2002;383(1):115 26. Roberts CR, Burke A. Remodelling of the extracellular matrix in asthma: proteoglycan synthesis and degradation. Can Respir J 1998;5:48 50. Laitinen A, Altraja A, Kampe M, et al. Tenascin is increased in airway basement membrane of asth-

[90] Cha HH, Cram EJ, Wang EC, et al. Glucocorticoids stimulate p21 gene expression by targeting multiple transcriptional elements within a steroid responsive region of the p21waf1/cip1 promoter in rat hepatoma cells. J Biol Chem 1998;273(4):1998 2007. [91] Roth M, Johnson PR, Rudiger JJ, et al. Interaction between glucocorticoids and beta2 agonists on bronchial airway smooth muscle cells through synchronised cellular signalling. Lancet 2002;360(9342): 1293 9. [92] Roth M, Johnson PR, Borger P, et al. Dysfunctional interaction of C/EBPalpha and the glucocorticoid receptor in asthmatic bronchial smooth-muscle cells. N Engl J Med 2004;351:560 74. [93] Irani K. Oxidant signaling in vascular cell growth, death, and survival: a review of the roles of reactive oxygen species in smooth muscle and endothelial cell mitogenic and apoptotic signaling. Circ Res 2000; 87:179 83. [94] Brar SS, Kennedy TP, Sturrock AB, et al. NADPH oxidase promotes NF-kappaB activation and proliferation in human airway smooth muscle. Am J Physiol 2002;282:L782 95. [95] Brar SS, Kennedy TP, Whorton AR, et al. Requirement for reactive oxygen species in serum-induced and platelet-derived growth factor-induced growth of airway smooth muscle. J Biol Chem 1999;274: 20017 26. [96] Pandya HC, Snetkov VA, Twort CHC, et al. Oxygen regulates mitogen-stimulated proliferation of fetal human airway smooth muscle cells. Am J Physiol 2002;283:L1220 30. [97] Simon AR, Takahashi S, Severgnini M, et al. Role of the JAK-STAT pathway in PDGF-stimulated proliferation of human airway smooth muscle cells. Am J Physiol 2002;282:L1296 304. [98] McKay S, de Jongste JC, Saxena PR, et al. Angiotensin II induces hypertrophy of human airway smooth muscle cells: expression of transcription factors and transforming growth factor-b1. Am J Respir Cell Mol Biol 1998;18:823 33. [99] Zheng X, Zhou D, Seow CY, et al. Cardiotrophin-1 alters airway smooth muscle structure and mechanical properties in airway explants. Am J Physiol Lung Cell Mol Physiol 2004;287(6):L1165 71. [100] Tran MU, Weir AJ, Fanucchi MV, et al. Smooth muscle hypertrophy in distal airways of sensitized infant rhesus monkeys exposed to house dust mite allergen. Clin Exp Allergy 2004;34(10):1627 33. [101] Benayoun L, Druilhe A, Dombret MC, et al. Airway structural alterations selectively associated with severe asthma. Am J Respir Crit Care Med 2003;167: 1360 8. [102] Zhang S, Mohammed Q, Burbidge A, et al. Cell cultures from bronchial subepithelial myofibroblasts enhance eosinophil survival in vitro. Eur Respir J 1996;9(9):1839 46. [103] Schmidt M, Sun G, Stacey MA, et al. Identification of circulating fibrocytes as precursors of bronchial

[104]

[105]

[106]

[107]

[108]

[109]

[110]

[111]

[112]

[113]

[114]

[115]

[116]

[117]

[118]

asm as an immune-response regulator matics and decreased by an inhaled steroid. Am J Respir Crit Care Med 1997;156:951 8. Laitinen LA, Laitinen A. Inhaled corticosteroid treatment and extracellular matrix in the airways in asthma. Int Arch Allergy Immunol 1995;107: 215 6. Black PN, Young PG, Skinner SJ. Response of airway smooth muscle cells to TGF-beta 1: effects on growth and synthesis of glycosaminoglycans. Am J Physiol 1996;271:L910 17. Coutts A, Chen G, Stephens N, et al. Release of biologically active TGF-b from airway smooth muscle cells induces autocrine synthesis of collagen. Am J Physiol 2001;280:L999 1008. Xie S, Sukkar MB, Issa R, et al. Regulation of TGF-beta 1-induced connective tissue growth factor expression in airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2005;288(1):L68 76. Knox AJ, Corbett L, Stocks J, et al. Human airway smooth muscle cells secrete vascular endothelial growth factor: up-regulation by bradykinin via a protein kinase C and prostanoid-dependent mechanism. FASEB J 2001;15:2480 8. Potter-Perigo S, Baker C, Tsoi C, et al. Regulation of proteoglycan synthesis by leukotriene d4 and epidermal growth factor in bronchial smooth muscle cells. Am J Respir Cell Mol Biol 2004;30(1):101 8. Rajah R, Nunn S, Herrick D, et al. LTD-4 induces matrix metalloproteinase-1 which functions as an IGFBP protease in airway smooth muscle cells. Am J Physiol 1996;271:L1014 22. Foda HD, George S, Rollo E, et al. Regulation of gelatinases in human airway smooth muscle cells: mechanism of progelatinase A activation. Am J Physiol 1999;277(21):L174 82. Elshaw SR, Henderson N, Knox AJ, et al. Matrix metalloproteinase expression and activity in human airway smooth muscle cells. Br J Pharmacol 2004; 142(8):1318 24. Kajita M, Itoh Y, Chiba T, et al. Membrane-type 1 matrix metalloproteinase cleaves CD44 and promotes cell migration. J Cell Biol 2001;153:893 904. Yu Q, Stamenkovic I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-b and promotes tumor invasion and angiogenesis. Genes Dev 2000;14:163 76. Van Eerdewegh P, Little RD, Dupuis J, et al. Association of the ADAM33 gene with asthma and bronchial hyperresponsiveness. Nature 2002;418(6896): 426 30. Haitchi HM, Powell RM, Shaw TJ, et al. ADAM33 expression in asthmatic airways and human embryonic lungs. Am J Respir Crit Care Med 2005;171(9): 958 65. Nguyen TT, Ward JP, Hirst SJ. beta1-Integrins mediate enhancement of airway smooth muscle proliferation by collagen and fibronectin. Am J Respir Crit Care Med 2005;171(3):217 23. Freyer AM, Billington CK, Penn RB, et al. Extra-

67

[119]

[134]

[120]

[135]

[121]

[136]

[122]

[137]

[123]

[138]

[124]

[139]

[125]

[140]

[126]

[141]

[127]

[142]

[128]

[143]

[129]

[144]

[130]

[145]

[131]

[146]

[132]

[147]

[133]

cellular matrix modulates beta2-adrenergic receptor signaling in human airway smooth muscle cells. Am J Respir Cell Mol Biol 2004;31(4):440 5. Saunders MA, Mitchell JA, Seldon PM, et al. Release of granulocyte-macrophage colony stimulating factor by human cultured airway smooth muscle cells: suppression by dexamethasone. Br J Pharmacol 1997; 120:545 6. Hallsworth MP, Soh CPC, Twort CHC, et al. Cultured human airway smooth muscle cells stimulated by interleukin-1b enhance eosinophil survival. Am J Respir Cell Mol Biol 1998;19:910 9. Hakonarson H, Maskeri N, Carter C, et al. Autocrine interaction between IL-5 and IL-1b mediates altered responsiveness of atopic asthmatic sensitized airway smooth muscle. J Clin Invest 1999;104: 657 67. Brightling E, Bradding P, Symon FA, et al. Mast-cell infiltration of airway smooth muscle in asthma. N Engl J Med 2002;346(22):1699 705. Kassel O, Schmidlin F, Duvernelle C, et al. Human bronchial smooth muscle cells in culture produce stem cell factor. Eur Respir J 1999;13(5):951 4. Lazaar AL, Plotnick MI, Kucich U, et al. Mast cell chymase modifies cell-matrix interactions and inhibits mitogen-induced proliferation of human airway smooth muscle cells. J Immunol 2002;169(2): 1014 20. Tyler CL, Ruoss SJ, Hartmann T, et al. Potent mitogenicity of tryptase for airway smooth muscle cells. Am Rev Respir Dis 1993;147:254. Brown JK, Jones CA, Rooney LA, et al. Tryptases potent mitogenic effects in human airway smooth muscle cells are via nonproteolytic actions. Am J Physiol Lung Cell Mol Physiol 2002;282(2): L197 206. DiCosmo BF, Geba GP, Picarella D, et al. Airway epithelial cell expression of interleukin-6 in transgenic mice. Uncoupling of airway inflammation and bronchial hyperreactivity. J Clin Invest 1994;94: 2028 35. Wang J, Homer RJ, Chen Q, et al. Endogenous and exogenous IL-6 inhibit aeroallergen-induced Th2 inflammation. J Immunol 2000;165:4051 61. Elias JA, Wu Y, Zheng T, et al. Cytokine- and virusstimulated airway smooth muscle cells produce IL-11 and other IL-6-type cytokines. Am J Physiol 1997; 273(17):L648 55. Lee JH, Kaminski N, Dolganov G, et al. Interleukin-13 induces dramatically different transcriptional programs in three human airway cell types. Am J Respir Cell Mol Biol 2001;25(4):474 85. Shore SA. Direct effects of Th2 cytokines on airway smooth muscle. Curr Opin Pharmacol 2004;4(3): 235 40. Hirst SJ, Hallsworth MP, Peng Q, et al. 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

68

lazaar

&

panettieri, jr tive expressions of type I NOS in human airway smooth muscle cells: evidence for an antiproliferative role. FASEB J 1999;13:1810 6. Lee SK, Park HS, Lim HE, et al. Localization of inducible nitric oxide synthase and endothelial constitutive nitric oxide synthase in airway mucosa of toluene diisocyanate-induced asthma. Allergy Asthma Proc 2003;24(4):275 80. Samb A, Pretolani M, Dinh-Xuan AT, et al. 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(1):149 54. Belvisi MG, Saunders MA, Haddad E-B, et al. Induction of cyclo-oxygenase-2 by cytokines in human cultured airway smooth muscle cells: novel inflammatory role of this cell type. Br J Pharmacol 1997;120:910 6. Cernadas M, De Sanctis GT, Krinzman SJ, et al. CD23 and allergic pulmonary inflammation: potential role as an inhibitor. Am J Respir Cell Mol Biol 1999;20:1 8. Dasic G, Juillard P, Graber P, et al. Critical role of CD23 in allergen-induced bronchoconstriction in a murine model of allergic asthma. Eur J Immunol 1999;29:2957 67. Haczku A, Takeda K, Hamelmann E, et al. CD23 exhibits negative regulatory effects on allergic sensitization and airway hyperresponsiveness. Am J Respir Crit Care Med 2000;161:952 60. Panettieri Jr RA, Lazaar AL, Pure E, et al. Activation of cAMP-dependent pathways in human airway smooth muscle cells inhibits TNF-a-induced ICAM-1 and VCAM-1 expression and T lymphocyte adhesion. J Immunol 1995;154:2358 65. Lazzeri N, Belvisi MG, Patel HJ, et al. Effects of prostaglandin E(2) and cAMP elevating drugs on GM-CSF release by cultured human airway smooth muscle cells. Relevance to asthma therapy. Am J Respir Cell Mol Biol 2001;24:44 8. Ammit AJ, Hoffman RK, Amrani Y, et al. TNFainduced secretion of RANTES and IL-6 from human airway smooth muscle cells: modulation by cAMP. Am J Respir Cell Mol Biol 2000;23:794 802. Hartert TV, Dworski RT, Mellen BG, et al. Prostaglandin E2 decreases allergen-stimulated release of prostaglandin D2 in airways of subjects with asthma. Am J Respir Crit Care Med 2000;162:637 40. Ammit AJ, Lazaar AL, Irani C, et al. Tumor necrosis factor-a-induced secretion of RANTES and interleukin-6 from human airway smooth muscle cells: modulation by glucocorticoids and b-agonists. Am J Respir Cell Mol Biol 2002;26:465 74. Kapsenberg ML, Hilkens CM, Wierenga EA, et al. The paradigm of type 1 and type 2 antigen-presenting cells. Implications for atopic allergy. Clin Exp Allergy 1999;29(Suppl 2):33 6. Roper RL, Conrad DH, Brown DM, et al. Prosta-

[148] [149]

[150]

[151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160]

[161]

[162]

[163]

by the interleukin-4 receptor alpha-chain. Am J Respir Crit Care Med 2002;165(8):1161 71. Springer TA. Adhesion receptors of the immune system. Nature 1990;346:425 34. Lazaar AL, Albelda SM, Pilewski JM, et al. 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 16. 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 76. Peng Q, Lai D, Nguyen TT, et al. Multiple beta 1 integrins mediate enhancement of human airway smooth muscle cytokine secretion by fibronectin and type I collagen. J Immunol 2005;174(4):2258 64. van Seventer GA, Shimizu Y, Shaw S. Roles of multiple accessory molecules in T-cell activation. Curr Opin Immunol 1991;3(3):294 303. Lazaar AL, Reitz HE, Panettieri Jr RA, et al. Antigen receptor-stimulated peripheral blood and bronchoalveolar lavage-derived T cells induce MHC class II and ICAM-1 expression on human airway smooth muscle. Am J Respir Cell Mol Biol 1997;16:38 45. Lazaar AL, Amrani Y, Hsu J, et al. CD40-mediated signal transduction in human airway smooth muscle. J Immunol 1998;161:3120 7. Hakonarson H, Kim C, Whelan R, et al. 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. Ramos-Barbon D, Presley JF, et al. Antigen-specific CD4(+) T cells drive airway smooth muscle remodeling in experimental asthma. J Clin Invest 2005; 115(6):1580 9 [Epub 2005 May 2]. Lazaar AL, Krymskaya VP, Das SK. VCAM-1 activates phosphatidylinositol 3-kinase and induces p120(Cbl) phosphorylation in human airway smooth muscle cells. J Immunol 2001;166:155 61. Hamann KJ, Vieira JE, Halayko AJ, et al. Fas crosslinking induces apoptosis in human airway smooth muscle cells. Am J Physiol 2000;278:L618 24. Hakonarson H, Maskeri N, Carter C, et al. Autocrine interaction between IL-5 and IL-1beta mediates altered responsiveness of atopic asthmatic sensitized airway smooth muscle. J Clin Invest 1999;104(5): 657 67. Tolloczko B, Ramos-Barbon D, Martin JG. Activated T-cells increase contractile agonist-induced calcium mobilization in airway smooth muscle cells. Proc Am Thorac Soc 2005;2:A27. Bates CA, Silkoff PE. Exhaled nitric oxide in asthma: from bench to bedside. J Allergy Clin Immunol 2003; 111:256 62. Hamad AM, Johnson SR, Knox AJ. Antiproliferative effects of NO and ANP in cultured human airway smooth muscle. Am J Physiol 1999;277:L910 8. Patel HJ, Belvisi MG, Donnelly LE, et al. Constitu-

[164]

[165]

[166]

[167]

[168]

[169]

[170]

[171]

[172]

[173]

[174]

[175]

[176]

asm as an immune-response regulator glandin E2 promotes IL-4-induced IgE and IgG1 synthesis. J Immunol 1990;145:2644 51. [177] Thangam EB, Venkatesha RT, Zaidi AK, et al. Airway smooth muscle cells enhance C3a-induced mast cell degranulation following cell-cell contact. FASEB J 2005;19(7):798 800. [178] Humbles AA, Lu B, Nilsson CA, et al. A role for the C3a anaphylatoxin receptor in the effector phase of asthma. Nature 2000;406:998 1001.

69

[179] Karp CL, Grupe A, Schadt E, et al. Identification of complement factor 5 as a susceptibility locus for experimental allergic asthma. Nat Immunol 2000;1: 221 6. [180] Jain VV, Kline JN. CpG DNA: immunomodulation and remodelling of the asthmatic airway. Expert Opin Biol Ther 2004;4(9):1533 40.

Vous aimerez peut-être aussi