51
|
Shifren A, Witt C, Christie C, Castro M. Mechanisms of remodeling in asthmatic airways. J Allergy (Cairo) 2012; 2012:316049. [PMID: 22315625 PMCID: PMC3270414 DOI: 10.1155/2012/316049] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 09/07/2011] [Accepted: 10/10/2011] [Indexed: 01/09/2023] Open
Abstract
Asthma is a chronic inflammatory airway disorder characterized by airway hyperresponsiveness and reversible airflow obstruction. Subgroups of asthma patients develop airflow obstruction that is irreversible or only partially reversible and experience an accelerated rate of lung function decline. The structural changes in the airways of these patients are referred to as airway remodeling. All elements of the airway wall are involved, and remodeled airway wall thickness is substantially increased compared to normal control airways. Airway remodeling is thought to contribute to the subphenotypes of irreversible airflow obstruction and airway hyperresponsiveness, and it has been associated with increased disease severity. Reversal of remodeling is therefore of paramount therapeutic importance, and mechanisms responsible for airway remodeling are feasible therapeutic targets for asthma treatment. This paper will focus on our current understanding of the mechanisms of airway remodeling in asthma and potential targets for future intervention.
Collapse
Affiliation(s)
- Adrian Shifren
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chad Witt
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chandrika Christie
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mario Castro
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
52
|
Niimi K, Ge Q, Moir LM, Ammit AJ, Trian T, Burgess JK, Black JL, Oliver BGG. β2-Agonists upregulate PDE4 mRNA but not protein or activity in human airway smooth muscle cells from asthmatic and nonasthmatic volunteers. Am J Physiol Lung Cell Mol Physiol 2011; 302:L334-42. [PMID: 22101762 DOI: 10.1152/ajplung.00163.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
β(2)-Adrenergic receptor (β2AR) agonists induce airway relaxation via cAMP. Phosphodiesterase (PDE)s degrade and regulate cAMP, and in airway smooth muscle (ASM) cells PDE4D degrades cAMP. Long-acting β(2)-agonists are now contraindicated as monotherapy for asthma, and increased PDE4D has been speculated to contribute to this phenomenon. In this study we investigated the expression of PDE4D in asthmatic and nonasthmatic ASM cells and its regulation by formoterol and budesonide. Primary ASM cells from people with or without asthma were stimulated with transforming growth factor (TGF)-β(1), formoterol, and/or budesonide. PDE4D mRNA was assessed by real-time PCR, or PCR to assess splice variant production. PDE4D protein was assessed by Western blotting, and we investigated the effect of formoterol on cAMP production and PDE activity. Interleukin (IL)-6 was assessed using ELISA. PDE4D mRNA was dose dependently upregulated by formoterol, with a single splice variant, PDE4D5, present. Formoterol did not induce PDE4D protein at time points between 3 to 72 h, whereas it did induce and increase IL-6 secretion. We pretreated cells with actinomycin D and a proteasome inhibitor, MG132, and found no evidence of alterations in mRNA, protein expression, or degradation of PDE4D. Finally PDE activity was not altered by formoterol. This study shows, for the first time, that PDE4D5 is predominantly expressed in human ASM cells from people with and without asthma and that formoterol does not upregulate PDE4D protein production. This leads us to speculate that continual therapy with β2AR agonists is unlikely to cause PDE4-mediated tachyphylaxis.
Collapse
Affiliation(s)
- Kyoko Niimi
- Cell Biology Group, Woolcock Institute of Medical Research, School of Medical Sciences, The Univ. of Sydney, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Study of gastric fluid induced cytokine and chemokine expression in airway smooth muscle cells and airway remodeling. Cytokine 2011; 56:726-31. [PMID: 21996013 DOI: 10.1016/j.cyto.2011.09.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/11/2011] [Accepted: 09/26/2011] [Indexed: 11/20/2022]
Abstract
Asthma is a chronic airway inflammatory disease. Chronic aspiration by gastric fluid in gastroesophageal reflux disease (GERD) is considered a primary inflammatory factor exacerbating or predisposing patients to asthma. Airway smooth muscle cells (SMCs) are considered an important component in airway remodeling. To investigate the role of gastric fluid in airway SMC inflammation and airway remodeling, we examined gastric fluid-induced cytokine and chemokine profiles, airway SMC migration and matrix metalloproteinase expression in rat primary rat airway SMCs. The T helper cell type 2 (Th2) cytokines interleukin 4, interleukin 6 and tumor necrosis factor 2 (TNF-α) and the chemokines, lipopolysaccharide-induced CXC chemokine (LIX/CXCL5), cytokine-induced neutrophil chemoattractant 2 (CINC-2), CINC-3, fractalkine, ciliary neurotrophic factor (CNTF), and vascular endothelial growth factor were induced by gastric fluid in primary cultured rat airway SMCs. Migration of rat airway SMCs was enhanced by gastric fluid and conditioned medium. The migration of rat airway SMCs enhanced by gastric fluid was associated with actin polymerization and activation of focal adhesion kinase. Matrix metalloproteinase 2 expressions in airway SMCs was enhanced by gastric fluid and conditioned medium. The results suggest potential mechanisms by which gastric fluid aspiration might influence SMC-mediated airway remodeling.
Collapse
|
54
|
Zhang Y, Zhang G, Clarke PA, Huang JTJ, Takahashi E, Muirhead D, Steenwyk RC, Lin Z. Simultaneous and high-throughput quantitation of urinary tetranor PGDM and tetranor PGEM by online SPE-LC-MS/MS as inflammatory biomarkers. JOURNAL OF MASS SPECTROMETRY : JMS 2011; 46:705-711. [PMID: 21706677 DOI: 10.1002/jms.1941] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Quantitation of urinary tetranor PGDM or tetranor PGEM (tPGDM and tPGEM) in the past was performed separately using off-line SPE LC-MS/MS methods. The manual SPE procedure is generally time-consuming and cost-ineffective. In addition, simultaneous quantitation of tPGDM and tPGEM is favorable yet very challenging because of the similar chemical structures and identical MRM transitions. This work describes the development and validation of a high-throughput online SPE-LC-MS/MS method, allowing simultaneous and high-throughput measurement of tPGDM and tPGEM in human urine. The reportable range of the assay was 0.2-40 ng/ml for tPGDM and 0.5-100 ng/ml for tPGEM. Intra- and inter-assay precision and accuracy determined using quality control samples were all within acceptable ranges (% CV and % Bias < 15%). Tetranor PGDM was stable under all tested conditions while tPGEM was stable at 4 °C and after three F/T cycles but not stable at room temperature for 24 h (recovery below 80%). The assay was applied to measure urinary tPGDM and tPGEM among healthy volunteers, smokers and COPD patients. Significantly higher urinary levels of both tPGDM and tPGEM were observed in COPD patients than those of non-smoking healthy volunteers. These results demonstrated that the high-throughput online SPE-LC-MS/MS assay provides sensitive, reproducible and accurate measurement of urinary tPGDM and tPGEM as biomarkers for assessing inflammatory diseases such as COPD.
Collapse
|
55
|
Baarsma HA, Meurs H, Halayko AJ, Menzen MH, Schmidt M, Kerstjens HAM, Gosens R. Glycogen synthase kinase-3 regulates cigarette smoke extract- and IL-1β-induced cytokine secretion by airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2011; 300:L910-9. [PMID: 21421749 DOI: 10.1152/ajplung.00232.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a constitutively active kinase that regulates multiple signaling proteins and transcription factors involved in inflammation. Its role in inflammatory lung diseases, including chronic obstructive pulmonary disease (COPD), is largely unknown. We investigated the role of GSK-3 in the secretion of chemokines and growth factors by human airway smooth muscle cells after exposure to cigarette smoke extract (CSE) or interleukin-1β (IL-1β), important factors involved in the development of COPD. Cultured human airway smooth muscle cells were exposed to increasing concentrations of CSE (1-15%) and IL-1β (0.01-1.0 ng/ml), which induced the secretion of VEGF-A and IL-8, whereas eotaxin secretion was induced by IL-1β only. Inhibition of GSK-3 by the selective inhibitor SB216763 or CHIR/CT99021 attenuated the cytokine and growth factor release induced by CSE and/or IL-1β, without affecting the basal release. Secretion of the cytokines by airway smooth muscle partially depends on NF-κB signaling, and GSK-3 has been implicated in regulating multiple steps in activating the NF-κB signaling pathway. IL-1β treatment induced degradation of the NF-κB inhibitory protein Iκ-Bα followed by nuclear translocation and DNA binding of p65 NF-κB, which were unaffected by inhibition of GSK-3. However, induction of NF-κB-dependent transcriptional activity by IL-1β and CSE was largely reduced upon GSK-3 inhibition by SB216763. Collectively, we demonstrate that CSE and IL-1β activate airway smooth muscle cells to secrete the proinflammatory cytokines IL-8, eotaxin, and VEGF-A. Furthermore, we show that GSK-3 regulates the release of these cytokines induced by CSE and IL-1β by promoting NF-κB-dependent gene transcription.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
56
|
TH17 cytokines induce human airway smooth muscle cell migration. J Allergy Clin Immunol 2011; 127:1046-53.e1-2. [PMID: 21345484 DOI: 10.1016/j.jaci.2010.12.1117] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2010] [Revised: 11/10/2010] [Accepted: 12/16/2010] [Indexed: 01/21/2023]
Abstract
BACKGROUND Migration of airway smooth muscle cells (ASMCs) might contribute to increased airway smooth muscle mass in asthma. T(H)17 cells and T(H)17-associated cytokines are involved in the pathogenesis of asthma and might also contribute to airway remodeling. OBJECTIVE We sought to explore the possibility that migration of ASMCs might contribute to airway remodeling through the action of T(H)17-related cytokines. METHODS The effect of exogenous T(H)17 cytokines on ex vivo human ASMC migration was investigated by using a chemotaxis assay. The involvement of signaling pathways, including p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 1/2 MAPK, nuclear factor κB, and phosphoinositide 3-kinase, was also examined. RESULTS We demonstrated that IL-17A, IL-17F, and IL-22 promote migration in a dose-dependent manner. We further demonstrated that ASMCs express receptors for IL-17RA, IL-17RC, and IL-22R1. Using mAbs directed against these receptors, we confirmed that T(H)17-associated cytokine-induced migration was dependent on selective receptor activation. Moreover, IL-17A and IL-17F exert their effects through signaling pathways that are distinct from those used by IL-22. The p38 MAPK inhibitor BIRB0796 inhibited the migration induced by IL-17A and IL-17F. PS1145, an inhibitor of nuclear factor κB, abolished the IL-22-induced migration. CONCLUSION These data raise the possibility that T(H)17-associated cytokines promote human ASMC migration in vivo and suggest an important new mechanism for the promotion of airway remodeling in asthma.
Collapse
|
57
|
Singer CA. T-bet is induced by interferon-γ to mediate chemokine secretion and migration in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2011; 300:L633-41. [PMID: 21239533 DOI: 10.1152/ajplung.00163.2010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
An inappropriate balance between T-helper (Th)1 and Th2 cytokine production underlies inflammatory changes that result in airway disease. Expression of the T-box transcription factor T-bet regulates differentiation of Th cells and production of Th1 cytokines, particularly IFNγ. T-bet-deficient mice develop airway hyperreactivity, undergo airway remodeling, and exhibit defects in IFNγ production while overproducing Th2 cytokines. T-bet is also reduced in the airways of asthmatic patients, suggesting loss of T-bet expression or activity promotes development of inflammatory airway disease. We present novel data demonstrating T-bet expression is induced in human airway smooth muscle cells (ASMC) by IFNγ. This IFNγ-stimulated expression of T-bet is dependent on signaling through JAK2 and signal transducers and activators of transcription 1 (STAT1) and activates T-bet-dependent DNA binding activity. Expression of T-bet stimulates IFNγ-stimulated IFNγ expression, secretion, and promoter activity, while inhibiting IFNγ-stimulated release of chemokines including monocyte chemoattractant protein (MCP)-1/CCL2, regulated on activation normal T-expressed and secreted (RANTES)/CCL5, and eotaxin/CCL11. This is accompanied by changes in expression of the chemokine receptors CCR3 and IL12Rβ2 and TNFα. T-bet expression also reduces chemotactic migration of ASMC in response to serum and PDGF, which contributes to airway hyperplasia. These results are the first to identify T-bet expression and activity in a structural cell of the lung and may provide new insights into therapeutic targets for inflammatory airway disease.
Collapse
Affiliation(s)
- Cherie A Singer
- Univ. of Nevada School of Medicine, Dept. of Pharmacology CMM MS-0573, Reno, NV 89557-0046, USA.
| |
Collapse
|
58
|
Lee BY, Cho S, Shin DH, Kim H. Genome-wide association study of copy number variations associated with pulmonary function measures in Korea Associated Resource (KARE) cohorts. Genomics 2010; 97:101-5. [PMID: 21059387 DOI: 10.1016/j.ygeno.2010.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/29/2010] [Accepted: 11/02/2010] [Indexed: 01/13/2023]
Abstract
Copy number variation (CNV) is an attractive emerging approach to study the association with various diseases. We performed a CNV-based genome-wide association study of pulmonary function measures (FEV(1), FVC, and FEV(1)/FVC) in KARE cohorts. Affymetrix Genome-wide Human SNP Array 5.0 was used to measure genome-wide variation and CNV segmentation was performed using Golden Helix SVS 7.0. Single and multivariate regressions were used for the association study using the R statistical package and the Dabatase for Annotation, Visualization and Integrated (DAVID v6.7b) tool for the functional annotation. We identified significantly associated 1260 CNVs with pulmonary function measures of FEV(1) and FVC. Functional gene classification and annotation analysis found 5 highly enriched clusters, the BPI/LBP/Plunc superfamily, myosin, serpin peptidase inhibitor, protein tyrosine phosphatase, and olfactory receptors. According to the functional annotation, gene-based CNVs are likely to be involved in the pathogenesis and inflammatory responsiveness of pulmonary diseases.
Collapse
Affiliation(s)
- Bo-Young Lee
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, San 56-1, Sillim-dong, Gwanak-gu, Seoul 151-742, Republic of Korea.
| | | | | | | |
Collapse
|
59
|
Jude JA, Solway J, Panettieri RA, Walseth TF, Kannan MS. Differential induction of CD38 expression by TNF-{alpha} in asthmatic airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2010; 299:L879-90. [PMID: 20693316 DOI: 10.1152/ajplung.00021.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ADP-ribosyl cyclase activity of CD38, a membrane protein expressed in human airway smooth muscle (ASM) cells, generates cyclic ADP-ribose (cADPR), a Ca²(+)-mobilizing agent. cADPR-mediated Ca²(+) responses to agonists are augmented in human ASM cells by TNF-α. CD38-deficient mice fail to develop airway hyperresponsiveness following intranasal TNF-α or IL-13 challenge, suggesting a role in asthma. The role of CD38 in human asthma remains unknown. We hypothesized that CD38 expression will be elevated in ASM cells from asthmatic donors (ASMA cells). CD38 mRNA and ADP-ribosyl cyclase activity were measured in cells maintained in growth-arrested conditions and exposed to vehicle or TNF-α (10-40 ng/ml). TNF-α-induced induction of CD38 expression was greater in ASMA than in ASM cells from nonasthmatic donors (ASMNA). In four of the six donors, basal and TNF-α-induced ERK and p38 MAPK activation were higher in ASMA than ASMNA cells. JNK MAPK activation was lower in ASMA than ASMNA cells. Nuclear NF-κB (p50 subunit) and phosphorylated c-Jun were comparable in cells from both groups, although nuclear c-Fos (part of the AP-1 complex) levels were lower in ASMA than ASMNA cells. NF-κB or AP-1 binding to their consensus sequences was comparable in ASMNA and ASMA cells, as are the decay kinetics of CD38 mRNA. The findings suggest that the differential induction of CD38 by TNF-α in ASMA cells is due to increased transcriptional regulation involving ERK and p38 MAPK activation and is independent of changes in NF-κB or AP-1 activation. The findings suggest a potential role for CD38 in the pathophysiology of asthma.
Collapse
Affiliation(s)
- Joseph A Jude
- Dept. of Veterinary and Biomedical Sciences, Univ. of Minnesota, St. Paul, 55108, USA
| | | | | | | | | |
Collapse
|
60
|
Xia YC, Schuliga M, Shepherd M, Powell M, Harris T, Langenbach SY, Tan PS, Gerthoffer WT, Hogarth PM, Stewart AG, Mackay GA. Functional expression of IgG-Fc receptors in human airway smooth muscle cells. Am J Respir Cell Mol Biol 2010; 44:665-72. [PMID: 20595464 DOI: 10.1165/rcmb.2009-0371oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
IgE-Fc receptors and IgG-Fc receptors are expressed on hematopoietic cells, but some evidence suggests that these receptors are also found on nonhematopoietic cells, including human airway smooth muscle (hASM) cells. Our study characterizes the expression of IgE-Fc receptors (FcεRI/CD23) and IgG-Fc receptors (FcγRs-I, -II, and -III) in cultured hASM cells by flow cytometry and Western blotting, and the functional activity of receptors was determined through quantification of cell proliferation and released cytokines. Expression of Fc receptor-linked intracellular signaling proteins and phosphorylation of the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase 1/2 and p38(MAPK) in hASM cells was examined by Western blotting. Expression of FcεRI and CD23 was not detectable in hASM cells. However, FcγRI and FcγRII were shown to be expressed on these cells. Specific antibodies, validated using transfected cell lines, revealed that the inhibitory IgG receptor, FcγRIIb, was the most abundant Fc receptor subtype expressed. Although cross-linking FcγR with heat-aggregated γ globulin (HAGG) did not induce detectable cell stimulation, pretreating hASM cells with HAGG significantly inhibited IL-1α-induced increases in cytokine levels and basic fibroblast growth factor-induced cell proliferation. This inhibitory effect of HAGG was abrogated by preincubation of cells with an anti-FcγRIIb antigen-binding fragment (Fab). Expression of proteins involved in the canonical FcγRIIb inhibitory signaling pathway was established in hASM cells. Pretreatment of hASM cells with HAGG significantly inhibited IL-1α- and basic fibroblast growth factor-induced extracellular signal-regulated kinase 1/2 and p38(MAPK) phosphorylation. This study identifies functional expression of FcγRIIb in hASM cells, with the potential to suppress their remodeling and immunomodulatory roles.
Collapse
Affiliation(s)
- YuXiu C Xia
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Kirstein F, Horsnell WGC, Kuperman DA, Huang X, Erle DJ, Lopata AL, Brombacher F. Expression of IL-4 receptor alpha on smooth muscle cells is not necessary for development of experimental allergic asthma. J Allergy Clin Immunol 2010; 126:347-54. [PMID: 20579713 DOI: 10.1016/j.jaci.2010.04.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Revised: 03/25/2010] [Accepted: 04/14/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND Airflow in the lungs of patients with allergic asthma is impaired by excessive mucus production and airway smooth muscle contractions. Elevated levels of the cytokines IL-4 and IL-13 are associated with this pathology. In vitro studies have suggested that IL-4 receptor alpha (IL-4Ralpha) signaling on smooth muscle cells is critical for airway inflammation and airway hyperresponsiveness. OBJECTIVE To define the contribution of IL-4 and IL-13 to the onset of asthmatic pathology, the role of their key receptor IL-4Ralpha in smooth muscle cells was examined in vivo. METHODS By using transgenic smooth muscle myosin heavy chain(cre)IL-4Ralpha(-/lox) mice deficient in IL-4Ralpha in smooth muscle cells, in vivo effects of impaired IL-4Ralpha signaling in smooth muscle cells on the outcome of asthmatic disease were investigated for the first time. Allergic asthma was introduced in mice by repeated sensitization with ovalbumin/aluminum hydroxide on days 0, 7, and 14, followed by intranasal allergen challenge on days 21 to 23. Mice were investigated for the presence of airway hyperresponsiveness, airway inflammation, allergen-specific antibody production, T(h)2-type cytokine responses, and lung pathology. RESULTS Airway hyperresponsiveness, airway inflammation, mucus production, T(h)2 cytokine production, and specific antibody responses were unaffected in smooth muscle myosin heavy chain(cre)IL-4Ralpha(-/lox) mice compared with control animals. CONCLUSION The impairment of IL-4Ralpha on smooth muscle cells had no effect on major etiologic markers of allergic asthma. These findings suggest that IL-4Ralpha responsiveness in airway smooth muscle cells during the early phase of allergic asthma is not, as suggested, necessary for the outcome of the disease.
Collapse
Affiliation(s)
- Frank Kirstein
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town and International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | | | | | | | | | | | | |
Collapse
|
62
|
Abstract
Proteomic approaches have already been successfully implemented in areas such as cancer research. Surprisingly, only a few proteomics analyses have been published reporting on the protein profiles associated with asthma. Although proteomics has its limitations and experimental challenges, it can successfully contribute to the understanding of a complex disease such as asthma. We have reviewed the current literature that has reported the use of proteomic techniques to identify proteins that may contribute to altered lung function in asthma. Only a few of these studies have used proteomic techniques on human tissues associated with asthma, while most research has been performed with animal models of asthma. Proteomic applications have been used as a complimentary technique to verify the suspected candidate proteins involved in asthma. In addition, novel proteins have been identified as potential therapeutic targets. Future collaboration between the different scientific disciplines using proteomic studies of animal models of asthma and confirmation of these findings in human tissues will significantly contribute to the understanding of the etiology of asthma and lead to the development of new therapeutic strategies for this highly prevalent disease.
Collapse
Affiliation(s)
- Annette Osei-Kumah
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, University of Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
63
|
Kajiwara K, Morishima H, Akiyama K, Yanagihara Y. Expression and function of the inducible costimulator ligand B7-H2 in human airway smooth muscle cells. Allergol Int 2009; 58:573-83. [PMID: 19776675 DOI: 10.2332/allergolint.09-oa-0113] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 05/24/2009] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND B7-H2 is a ligand for the inducible costimulator (ICOS). The aim of this study was to examine the expression and function of B7-H2 in human airway smooth muscle (ASM) cells and compare them with those of CD40 or OX40 ligand (OX40L). METHODS Expression of B7-H2, CD40 and OX40L in ASM cells and their respective counterparts in T cells was analyzed by RT-PCR or flow cytometry. The modulating effect of polyinosinic-polycytidylic acid (poly I:C) on expression of B7-H2, CD40 and OX40L was also examined. The function of these three molecules was evaluated by virtue of adhesion of anti-CD3-activated T cells, IL-6 and IL-8 production and DNA synthesis. RESULTS ASM cells constitutively expressed B7-H2, CD40 and OX40L that mediated adhesion of activated T cells expressing ICOS, CD40L and OX40. ASM cells responded to poly I:C with upregulated expression of B7-H2, CD40 and OX40L and displayed enhanced adhesion of activated T cells. Functional analysis performed on untreated ASM cells showed that engagement of B7-H2 with ICOS-Ig clearly induced DNA synthesis, whereas that of CD40 or OX40L with trimeric CD40L or OX40-Ig greatly increased IL-6 and IL-8 production. These responses were enhanced in poly I:C-treated ASM cells. CONCLUSIONS The data demonstrate that ASM cells express functionally active B7-H2, CD40 and OX40L and suggest that B7-H2-dependent signaling may play an active role in a proliferative response rather than in cytokine and chemokine production. In addition, the modulation of B7-H2, CD40 and OX40L expression and function by poly I:C may have important implications for the function of virus-infected ASM cells.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/genetics
- CD3 Complex/immunology
- CD40 Antigens/biosynthesis
- CD40 Antigens/genetics
- Cell Adhesion/immunology
- Cell Separation
- Cells, Cultured
- Flow Cytometry
- Humans
- Inducible T-Cell Co-Stimulator Ligand
- Inducible T-Cell Co-Stimulator Protein
- Interleukin-6/metabolism
- Interleukin-8/metabolism
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- OX40 Ligand/biosynthesis
- OX40 Ligand/genetics
- Respiratory System/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Keiichi Kajiwara
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | | | | | | |
Collapse
|
64
|
Tliba O, Panettieri RA. Noncontractile functions of airway smooth muscle cells in asthma. Annu Rev Physiol 2009; 71:509-35. [PMID: 18851708 DOI: 10.1146/annurev.physiol.010908.163227] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although pivotal in regulating bronchomotor tone in asthma, airway smooth muscle (ASM) also modulates airway inflammation and undergoes hypertrophy and hyperplasia, contributing to airway remodeling in asthma. ASM myocytes secrete or express a wide array of immunomodulatory mediators in response to extracellular stimuli, and in chronic severe asthma, increases in ASM mass may render the airway irreversibly obstructed. Although the mechanisms by which ASM secretes cytokines and chemokines are the same as those regulating immune cells, there exist unique ASM signaling pathways that may provide novel therapeutic targets. This review provides an overview of our current understanding of the proliferative as well as the synthetic properties of ASM.
Collapse
Affiliation(s)
- Omar Tliba
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
65
|
John AE, Zhu YM, Brightling CE, Pang L, Knox AJ. Human airway smooth muscle cells from asthmatic individuals have CXCL8 hypersecretion due to increased NF-kappa B p65, C/EBP beta, and RNA polymerase II binding to the CXCL8 promoter. THE JOURNAL OF IMMUNOLOGY 2009; 183:4682-92. [PMID: 19734226 DOI: 10.4049/jimmunol.0803832] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
CXCL8 is a neutrophil and mast cell chemoattractant that is involved in regulating inflammatory cell influx in asthma. Here, we investigated the transcriptional mechanism involved in CXCL8 induction by TNF-alpha in cultured human airway smooth muscle (HASM) cells and compared these in cells from nonasthmatic and asthmatic individuals. Transfection studies with mutated CXCL8 promoter constructs identified NF-kappaB, activating protein-1, and CAAT/enhancer binding protein (C/EBP)beta as key transcription factors, and binding of these three transcription factors to the CXCL8 promoter after TNF-alpha stimulation was confirmed by chromatin immunoprecipitation analysis. Cells derived from asthmatic individuals produced significantly higher levels of CXCL8 than nonasthmatic cells both basally and following 24 h of stimulation with TNF-alpha (p < 0.001). Furthermore, chromatin immunoprecipitation studies detected increased binding of NF-kappaB p65 and RNA polymerase II to the CXCL8 promoter of asthmatic HASM cells both in the presence and absence of TNF-alpha stimulation. This was not due to either an increased activation or phosphorylation of NF-kappaB per se or to an increase in its translocation to the nucleus. Increased binding of C/EBPbeta to the CXCL8 promoter of unstimulated cells was also detected in the asthmatic HASM cells. Collectively these studies show that HASM cells from asthmatic individuals have increased CXCL8 production due to the presence of a transcription complex on the CXCL8 promoter, which contains NF-kappaB, C/EBPbeta, and RNA polymerase II. This is the first description of an abnormality in transcription factor binding altering chemokine expression in airway structural cells in asthma.
Collapse
Affiliation(s)
- Alison E John
- Centre for Respiratory Research and Nottingham Respiratory Biomedical Research Unit, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | | |
Collapse
|
66
|
Yang CM, Lee IT, Lin CC, Yang YL, Luo SF, Kou YR, Hsiao LD. Cigarette smoke extract induces COX-2 expression via a PKCalpha/c-Src/EGFR, PDGFR/PI3K/Akt/NF-kappaB pathway and p300 in tracheal smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2009; 297:L892-902. [PMID: 19717552 DOI: 10.1152/ajplung.00151.2009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Exposure to cigarette smoke extract (CSE) leads to airway or lung inflammation, which may be mediated through cyclooxygenase-2 (COX-2) expression and its product prostaglandin E2 (PGE2) synthesis. The aim of this study was to investigate the molecular mechanisms underlying CSE-induced COX-2 expression in human tracheal smooth muscle cells (HTSMCs). Here, we describe that COX-2 induction is dependent on PKCalpha/c-Src/EGFR, PDGFR/PI3K/Akt/NF-kappaB signaling in HTSMCs. CSE stimulated the phosphorylation of c-Src, EGFR, PDGFR, and Akt, which were inhibited by pretreatment with the inhibitor of PKCalpha (Gö6976 or Gö6983), c-Src (PP1), EGFR (AG1478), PDGFR (AG1296), or PI3K (LY294002). Moreover, CSE induced a significant increase in COX-2 expression, which was reduced by pretreatment with these inhibitors or transfection with siRNA of PKCalpha, Src, or Akt. Furthermore, CSE-stimulated NF-kappaB p65 phosphorylation and translocation were also attenuated by pretreatment with Gö6976, PP1, AG1478, AG1296, or LY294002. CSE-induced COX-2 expression was also mediated through the recruitment of p300 associated with NF-kappaB in HTSMCs, revealed by coimmunoprecipitation and Western blot analysis. In addition, pretreatment with the inhibitors of NF-kappaB (helenalin) and p300 (garcinol) or transfection with p65 siRNA and p300 siRNA markedly inhibited CSE-regulated COX-2 expression. However, CSE-induced PGE2 generation was reduced by pretreatment with the inhibitor of COX-2 (NS-398). These results demonstrated that in HTSMCs, CSE-induced COX-2-dependent PGE2 generation was mediated through PKCalpha/c-Src/EGFR, PDGFR/PI3K/Akt leading to the recruitment of p300 with NF-kappaB complex.
Collapse
Affiliation(s)
- Chuen-Mao Yang
- Department of Pharmacology, Chang Gung University, Chang Gung Memorial Hospital, 259 Wen-Hwa 1st Road, Kwei-San, Tao-Yuan, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
67
|
Ho SC, Lee KY, Chan YF, Kuo LW, Ito K, Adcock IM, Chen BC, Sheu JR, Lin CH, Kuo HP. Neutrophil Elastase Represses IL-8/CXCL8 Synthesis in Human Airway Smooth Muscle Cells through Induction of NF-κB Repressing Factor. THE JOURNAL OF IMMUNOLOGY 2009; 183:411-20. [DOI: 10.4049/jimmunol.0803729] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
68
|
Saleh A, Shan L, Halayko AJ, Kung S, Gounni AS. Critical role for STAT3 in IL-17A-mediated CCL11 expression in human airway smooth muscle cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:3357-65. [PMID: 19265112 DOI: 10.4049/jimmunol.0801882] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IL-17A has been shown to be expressed at higher levels in respiratory secretions from asthmatics and to correlate with airway hyperresponsiveness. Although these studies raise the possibility that IL-17A may influence allergic disease, the mechanism remains unknown. We previously demonstrated that IL-17A mediates CC chemokine (CCL11) production from human airway smooth muscle (ASM) cells. In this study, we demonstrate that STAT3 activation is critical in IL-17A-mediated CCL11 expression in ASM cells. IL-17A mediated a rapid phosphorylation of STAT3 but not STAT6 or STAT5 in ASM cells. Interestingly, transient transfection with wild-type or mutated CCL11 promoter constructs showed that IL-17A-mediated CCL11 expression relies on the STAT6 binding site. However, STAT3 but not STAT6 in vivo binding to the CCL11 promoter was detected following IL-17A stimulation of ASM cells. Overexpression of DN STAT3 (STAT3beta) abolishes IL-17A-induced CCL11 promoter activity. This effect was not observed with STAT6 DN or the STAT3 mutant at Ser(727). Interestingly, disruption of STAT3 activity with the SH2 domain binding peptide, but not with control peptide, results in a significant reduction of IL-17A-mediated STAT3 phosphorylation and CCL11 promoter activity. IL-17A-mediated CCL11 promoter activity and mRNA were significantly diminished in STAT3- but not STAT6-silenced ASM cells. Finally, IL-17A-induced STAT3 phosphorylation was sensitive to pharmacological inhibitors of JAK2 and ERK1/2. Taken together, our data provide the first evidence of IL-17A-mediated gene expression via STAT3 in ASM cells. Collectively, our results raise the possibility that the IL-17A/STAT3 signaling pathway may play a crucial role in airway inflammatory responses.
Collapse
Affiliation(s)
- Ali Saleh
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | |
Collapse
|
69
|
Lavoie TL, Dowell ML, Lakser OJ, Gerthoffer WT, Fredberg JJ, Seow CY, Mitchell RW, Solway J. Disrupting actin-myosin-actin connectivity in airway smooth muscle as a treatment for asthma? PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2009; 6:295-300. [PMID: 19387033 PMCID: PMC2677405 DOI: 10.1513/pats.200808-078rm] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2008] [Accepted: 01/27/2009] [Indexed: 11/20/2022]
Abstract
Breathing is known to functionally antagonize bronchoconstriction caused by airway muscle contraction. During breathing, tidal lung inflation generates force fluctuations that are transmitted to the contracted airway muscle. In vitro, experimental application of force fluctuations to contracted airway smooth muscle strips causes them to relengthen. Such force fluctuation-induced relengthening (FFIR) likely represents the mechanism by which breathing antagonizes bronchoconstriction. Thus, understanding the mechanisms that regulate FFIR of contracted airway muscle could suggest novel therapeutic interventions to increase FFIR, and so to enhance the beneficial effects of breathing in suppressing bronchoconstriction. Here we propose that the connectivity between actin filaments in contracting airway myocytes is a key determinant of FFIR, and suggest that disrupting actin-myosin-actin connectivity by interfering with actin polymerization or with myosin polymerization merits further evaluation as a potential novel approach for preventing prolonged bronchoconstriction in asthma.
Collapse
Affiliation(s)
- Tera L Lavoie
- Department of Medicine, University of Chicago, MC6026, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Clarke D, Damera G, Sukkar MB, Tliba O. Transcriptional regulation of cytokine function in airway smooth muscle cells. Pulm Pharmacol Ther 2009; 22:436-45. [PMID: 19393330 DOI: 10.1016/j.pupt.2009.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 01/28/2009] [Accepted: 04/14/2009] [Indexed: 01/01/2023]
Abstract
The immuno-modulatory properties of airway smooth muscle have become of increasing importance in our understanding of the mechanisms underlying chronic inflammation and structural remodeling of the airway wall in asthma and chronic obstructive pulmonary disease (COPD). ASM cells respond to many cytokines, growth factors and lipid mediators to produce a wide array of immuno-modulatory molecules which may in turn orchestrate and perpetuate the disease process in asthma and COPD. Despite numerous studies of the cellular effects of cytokines on cultured ASM, few have identified intracellular signaling pathways by which cytokines modulate or induce these cellular responses. In this review we provide an overview of the transcriptional mechanisms as well as intracellular signaling pathways regulating cytokine functions in ASM cells. The recent discovery of toll-like receptors in ASM cells represents a significant development in our understanding of the immuno-modulatory capabilities of ASM cells. Thus, we also review emerging evidence of the inflammatory response to toll-like receptor activation in ASM cells.
Collapse
Affiliation(s)
- Deborah Clarke
- Respiratory Pharmacology, National Heart & Lung Institute, Imperial College London, London, UK
| | | | | | | |
Collapse
|
71
|
Ceresa CC, Knox AJ, Johnson SR. Use of a three-dimensional cell culture model to study airway smooth muscle-mast cell interactions in airway remodeling. Am J Physiol Lung Cell Mol Physiol 2009; 296:L1059-66. [PMID: 19346431 DOI: 10.1152/ajplung.90445.2008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased airway smooth muscle (ASM) mass and infiltration by mast cells are key features of airway remodeling in asthma. We describe a model to investigate the relationship between ASM, the extracellular matrix, mast cells, and airway remodeling. ASM cells were cultured in a three-dimensional (3-D) collagen I gel (3-D culture) alone or with mast cells. Immunocytochemistry and Western blotting of ASM in 3-D cultures revealed a spindle-shaped morphology and significantly lower alpha-smooth muscle actin and vimentin expression than in ASM cultured in monolayers on collagen type I or plastic (2-D culture). In 3-D cultures, basal ASM proliferation, examined by Ki67 immunocytochemistry, was reduced to 33 +/- 7% (P < 0.05) of that in 2-D cultures. The presence of mast cells in cocultures increased ASM proliferation by 1.8-fold (P < 0.05). Gelatin zymography revealed more active matrix metalloproteinase (MMP)-2 in 3-D than in 2-D culture supernatants over 7 days. Functional MMP activity was examined by gel contraction. The spontaneous gel contraction over 7 days was significantly inhibited by the MMP inhibitor ilomastat. Mast cell coculture enhanced ASM gel contraction by 22 +/- 16% (not significant). Our model shows that ASM has different morphology, with lower contractile protein expression and basal proliferation in 3-D culture. Compared with standard techniques, ASM synthetic function, as shown by MMP production and activity, is sustained over longer periods. The presence of mast cells in the 3-D model enhanced ASM proliferation and MMP production. Airway remodeling in asthma may be more accurately modeled by our system than by standard culture systems.
Collapse
Affiliation(s)
- Claudia C Ceresa
- Divisions of Therapeutics and Molecular Medicine, University of Nottingham, Nottingham, United Kingdom
| | | | | |
Collapse
|
72
|
Shin JH, Shim JW, Kim DS, Shim JY. TGF-beta effects on airway smooth muscle cell proliferation, VEGF release and signal transduction pathways. Respirology 2009; 14:347-53. [PMID: 19192227 DOI: 10.1111/j.1440-1843.2008.01469.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Airway smooth muscle (ASM) cell hyperplasia is a key feature of airway remodelling. Mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) are key components in signal transduction associated with cell proliferation; MAPK consists of the extracellular signal-regulated kinase (ERK), p38MAPK and c-Jun NH(2)-terminal kinase (JNK). The effect of transforming growth factor (TGF)-beta on the proliferation of ASM cells, the release of vascular endothelial growth factor (VEGF) by ASM cells and relevant signal transduction pathways were investigated. METHODS ASM cells were growth-arrested for 48 h then stimulated with platelet-derived growth factor (PDGF), TGF-beta and dexamethasone. ASM cells were also treated with specific inhibitors of MAPK (PD98059), PI3K (wortmannin) and JNK (SP600125). Cell proliferation and VEGF concentrations were measured. RESULTS TGF-beta neither augmented ASM cell proliferation nor showed a synergistic effect on PDGF-mediated ASM cell proliferation. Dexamethasone did not suppress ASM cell proliferation. VEGF release was augmented by TGF-beta stimulation in a time-dependent manner, and was further enhanced by co-stimulation with PDGF and TGF-beta. Dexamethasone suppressed VEGF release significantly. TGF-beta enhanced PI3K phosphorylation, while PDGF augmented both ERK and PI3K phosphorylation. Wortmannin inhibited both TGF-beta- and PDGF-stimulated VEGF release. CONCLUSIONS TGF-beta may facilitate airway remodelling by promoting VEGF release through the PI3K pathway, rather than via ASM cell proliferation.
Collapse
Affiliation(s)
- Joo Hwa Shin
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
73
|
Dekkers BGJ, Schaafsma D, Tran T, Zaagsma J, Meurs H. Insulin-induced laminin expression promotes a hypercontractile airway smooth muscle phenotype. Am J Respir Cell Mol Biol 2009; 41:494-504. [PMID: 19213874 DOI: 10.1165/rcmb.2008-0251oc] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway smooth muscle (ASM) plays a key role in the development of airway hyperresponsiveness and remodeling in asthma, which may involve maturation of ASM cells to a hypercontractile phenotype. In vitro studies have indicated that long-term exposure of bovine tracheal smooth muscle (BTSM) to insulin induces a functional hypercontractile, hypoproliferative phenotype. Similarly, the extracellular matrix protein laminin has been found to be involved in both the induction and maintenance of a contractile ASM phenotype. Using BTSM, we now investigated the role of laminins in the insulin-induced hypercontractile, hypoproliferative ASM phenotype. The results demonstrate that insulin-induced hypercontractility after 8 days of tissue culture was fully prevented by combined treatment of BTSM-strips with the laminin competing peptides Tyr-Ile-Gly-Ser-Arg (YIGSR) and Arg-Gly-Asp-Ser (RGDS). YIGSR also prevented insulin-induced increases in sm-myosin expression and abrogated the suppressive effects of prolonged insulin treatment on platelet-derived growth factor-induced DNA synthesis in cultured cells. In addition, insulin time-dependently increased laminin alpha2, beta1, and gamma1 chain protein, but not mRNA abundance in BTSM strips. Moreover, as previously found for contractile protein accumulation, signaling through PI3-kinase- and Rho kinase-dependent pathways was required for the insulin-induced increase in laminin abundance and contractility. Collectively, our results indicate a critical role for beta1-containing laminins, likely laminin-211, in the induction of a hypercontractile, hypoproliferative ASM phenotype by prolonged insulin exposure. Increased laminin production by ASM could be involved in the increased ASM contractility and contractile protein expression in asthma. Moreover, the results may be of interest for the use of inhaled insulin administrations by diabetics.
Collapse
Affiliation(s)
- Bart G J Dekkers
- Department of Molecular Pharmacology, University centre for Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
74
|
Damera G, Zhao H, Wang M, Smith M, Kirby C, Jester WF, Lawson JA, Panettieri RA. Ozone modulates IL-6 secretion in human airway epithelial and smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2009; 296:L674-83. [PMID: 19201813 DOI: 10.1152/ajplung.90585.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although ozone enhances leukocyte function and recruitment in airways, the direct effect of ozone in modulating structural cell-derived inflammatory mediators remains unknown. Using a coculture model comprised of differentiated human airway epithelial cells (NHBE) and smooth muscle cells (ASM), we postulate that ozone regulates IL-6 secretion in basal and cytokine-primed structural cells. Air-liquid interface (ALI) cultures of NHBE cells underwent differentiation as determined by mucin secretion, transepithelial electrical resistance (TEER), and ultrastructure parameters. Whereas TNF enhanced basal secretion of IL-6 (57 +/- 3%), ozone exposure at 0.6 ppm for 6 h augmented IL-6 levels in basal (41 +/- 3%) and TNF- (50 +/- 5%) primed cocultures compared with that derived from NHBE or ASM monolayers alone. Levels of PGE(2), 6-keto-PGF(1alpha), PGF(2alpha), and thromboxane B(2) (TxB(2)) levels in basal and TNF-primed cocultures revealed that ozone selectively enhanced PGE(2) production in TNF- (6 +/- 3-fold) primed cocultures, with little effect (P > 0.05) on diluent-treated cultures. In accordance with ozone-induced increases in PGE(2) levels, cyclooxygenase inhibition with indomethacin partially abolished IL-6 secretion. Surprisingly, indomethacin had little effect on constitutive secretion of IL-6 in cocultures, whereas indomethacin completely restored ozone-mediated TEER reduction in TNF-primed cocultures. Collectively, our data for the first time suggest a dual role of ozone in modulating IL-6 secretion and TEER outcomes in a PGE(2)-dependent (in presence of TNF stimulus) and -independent manner (in absence of cytokine stimulus).
Collapse
Affiliation(s)
- Gautam Damera
- University of Pennsylvania, Philadelphia, PA 19104-3403, USA
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Burgess JK. The role of the extracellular matrix and specific growth factors in the regulation of inflammation and remodelling in asthma. Pharmacol Ther 2009; 122:19-29. [PMID: 19141302 DOI: 10.1016/j.pharmthera.2008.12.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 12/17/2008] [Indexed: 12/12/2022]
Abstract
Asthma is a disease characterised by persistent inflammation and structural changes in the airways, referred to as airway remodelling. The mechanisms underlying these processes may be interdependent or they may be separate processes that are driven by common factors. The levels of a variety of growth factors (including transforming growth factor beta, granulocyte macrophage colony stimulating factor, and vascular endothelial growth factor) are known to be changed in the asthmatic airway. These and other growth factors can contribute to the development and persistence of inflammation and remodelling. One of the prominent features of the structural changes of the airways is the increased deposition and alterations in the composition of the extracellular matrix proteins. These proteins include fibronectin, many different collagen types and hyaluronan. There is a dynamic relationship between the extracellular matrix proteins and the airway mesenchymal cells such that the changes in the extracellular matrix proteins can also contribute to the persistence of inflammation and the airway remodelling. This review aims to summarise the role growth factors and extracellular matrix proteins play in the regulation of inflammation and airway remodelling in the asthmatic airway.
Collapse
Affiliation(s)
- Janette K Burgess
- Discipline of Pharmacology, The University of Sydney, Woolcock Institute of Medical Research and the Cooperative Research Centre for Asthma and Airways, Sydney, NSW Australia.
| |
Collapse
|
76
|
Hirota JA, Nguyen TTB, Schaafsma D, Sharma P, Tran T. Airway smooth muscle in asthma: phenotype plasticity and function. Pulm Pharmacol Ther 2008; 22:370-8. [PMID: 19114115 DOI: 10.1016/j.pupt.2008.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 11/14/2008] [Accepted: 12/10/2008] [Indexed: 10/24/2022]
Abstract
Clinical asthma is characterized by reversible airway obstruction which is commonly due to an exaggerated airway narrowing referred to as airway hyperresponsiveness (AHR). Although debate exists on the complex etiology of AHR, it is clear that airway smooth muscle (ASM) mediated airway narrowing is a major contributor to airway dysfunction. More importantly, it is now appreciated that smooth muscle is far from being a simple cell with only contractile ability properties. Rather, it is more versatile with the capacity to exhibit numerous cellular functions as it adapts to the microenvironment to which it is exposed. The emerging ability of individual smooth muscle cells to undergo changes in their phenotype (phenotype plasticity) and function (functional plasticity) in response to physiological and pathological cues is an important and active area of research. This article provides a brief review of the current knowledge and emerging concepts in the field of ASM phenotype and function both under healthy and asthmatic conditions.
Collapse
Affiliation(s)
- Jeremy A Hirota
- Firestone Institute for Respiratory Health, McMaster University, Ontario, Canada
| | | | | | | | | |
Collapse
|
77
|
Damera G, Tliba O, Panettieri RA. Airway smooth muscle as an immunomodulatory cell. Pulm Pharmacol Ther 2008; 22:353-9. [PMID: 19121407 DOI: 10.1016/j.pupt.2008.12.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 12/09/2008] [Accepted: 12/10/2008] [Indexed: 10/21/2022]
Abstract
Although pivotal in regulating bronchomotor tone in asthma, airway smooth muscle (ASM) also modulates airway inflammation in asthma. ASM myocytes secrete or express a wide array of immunomodulatory mediators in response to extracellular stimuli, and in chronic severe asthma, increases in ASM mass may also render the airway irreversibly obstructed. Although the mechanisms by which ASM secretes cytokines and chemokines are shared with those regulating immune cells, there exist unique ASM signaling pathways that may provide novel therapeutic targets. This review provides an overview of our current understanding of the proliferative as well as synthetic properties of ASM.
Collapse
Affiliation(s)
- Gautam Damera
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, University of Pennsylvania, Philadelphia, PA 19104-3403, USA
| | | | | |
Collapse
|
78
|
Kaur M, Holden NS, Wilson SM, Sukkar MB, Chung KF, Barnes PJ, Newton R, Giembycz MA. Effect of beta2-adrenoceptor agonists and other cAMP-elevating agents on inflammatory gene expression in human ASM cells: a role for protein kinase A. Am J Physiol Lung Cell Mol Physiol 2008; 295:L505-14. [PMID: 18586957 DOI: 10.1152/ajplung.00046.2008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In diseases such as asthma, airway smooth muscle (ASM) cells play a synthetic role by secreting inflammatory mediators such as granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-6, or IL-8 and by expressing surface adhesion molecules, including ICAM-1. In the present study, PGE(2), forskolin, and short-acting (salbutamol) and long-acting (salmeterol and formoterol) beta(2)-adrenoceptor agonists reduced the expression of ICAM-1 and the release of GM-CSF evoked by IL-1beta in ASM cells. IL-1beta-induced IL-8 release was also repressed by PGE(2) and forskolin, whereas the beta(2)-adrenoceptor agonists were ineffective. In each case, repression of these inflammatory indexes was prevented by adenoviral overexpression of PKIalpha, a highly selective PKA inhibitor. These data indicate a PKA-dependent mechanism of repression and suggest that agents that elevate intracellular cAMP, and thereby activate PKA, may have a widespread anti-inflammatory effect in ASM cells. Since ICAM-1 and GM-CSF are highly NF-kappaB-dependent genes, we used an adenoviral-delivered NF-kappaB-dependent luciferase reporter to examine the effects of forskolin and the beta(2)-adrenoceptor agonists on NF-kappaB activation. There was no effect on luciferase activity measured in the presence of forskolin or beta(2)-adrenoceptor agonists. This finding is consistent with the observation that IL-1beta-induced expression of IL-6, a known NF-kappaB-dependent gene in ASM, was also unaffected by beta(2)-adrenoceptor agonists, forskolin, PGE(2), 8-bromo-cAMP, or rolipram. Collectively, these results indicate that repression of IL-1beta-induced ICAM-1 expression and GM-CSF release by cAMP-elevating agents, including beta(2)-adrenoceptor agonists, may not occur through a generic effect on NF-kappaB.
Collapse
Affiliation(s)
- Manminder Kaur
- Dept. of Cell Biology & Anatomy, Faculty of Medicine, Univ. of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada T2N 4N1
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Joubert P, Lajoie-Kadoch S, Welman M, Dragon S, Létuvée S, Tolloczko B, Halayko AJ, Gounni AS, Maghni K, Hamid Q. Expression and regulation of CCR1 by airway smooth muscle cells in asthma. THE JOURNAL OF IMMUNOLOGY 2008; 180:1268-75. [PMID: 18178867 DOI: 10.4049/jimmunol.180.2.1268] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
C-C chemokines such as CCL11, CCL5, and CCL3 are central mediators in the pathogenesis of asthma. They are mainly associated with the recruitment and the activation of specific inflammatory cells, such as eosinophils, lymphocytes, and neutrophils. It has recently been shown that they can also activate structural cells, such as airway smooth muscle and epithelial cells. The aims of this study were to examine the expression of the CCL3 receptor, CCR1, on human airway smooth muscle cells (ASMC) and to document the regulation of this receptor by cytokines involved in asthma pathogenesis. We first demonstrated that CCR1 mRNA is increased in the airways of asthmatic vs control subjects and showed for the first time that ASMC express CCR1 mRNA and protein, both in vitro and in vivo. Calcium mobilization by CCR1 ligands confirmed its functionality on ASMC. Stimulation of ASMC with TNF-alpha and, to a lesser extent, IFN-gamma resulted in an up-regulation of CCR1 expression, which was totally suppressed by both dexamethasone or mithramycin. Taken together, our data suggest that CCR1 might be involved in the pathogenesis of asthma, through the activation of ASMC by its ligands.
Collapse
Affiliation(s)
- Philippe Joubert
- Meakins-Christie Laboratories, McGill University, and Hôpital du Sacré-Coeur de Montréal, Research Center, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Tliba O, Amrani Y, Panettieri RA. Is airway smooth muscle the "missing link" modulating airway inflammation in asthma? Chest 2008; 133:236-42. [PMID: 18187748 DOI: 10.1378/chest.07-0262] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Airway smooth muscle (ASM) plays a central role in regulating bronchomotor tone in patients with asthma. New evidence, however, suggests that ASM may also orchestrate and perpetuate airway inflammation by promoting the recruitment, activation, and trafficking of inflammatory cells in the airways. This review addresses the immunomodulatory function of ASM and highlights how such function may have therapeutic implications in the management of asthma.
Collapse
Affiliation(s)
- Omar Tliba
- Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, 125 South 31st St, Philadelphia, PA 19104-3403, USA.
| | | | | |
Collapse
|
81
|
de Boer WI, Alagappan VKT, Sharma HS. Molecular mechanisms in chronic obstructive pulmonary disease: potential targets for therapy. Cell Biochem Biophys 2008; 47:131-48. [PMID: 17406066 DOI: 10.1385/cbb:47:1:131] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease associated with progressive airflow obstruction. Tobacco smoking is the main risk factor worldwide. In contrast to asthma, antiinflammatory therapies are rather ineffective in improving chronic symptoms and reducing inflammation, lung function decline, and airway remodeling. Specific drugs that are directed against the remodeling and chronic inflammation, thereby preventing lung tissue damage and progressive lung function decline, must be developed. Experimental models and expression studies suggest that anti-vascular endothelial growth factor (VEGF) receptor strategies may be of use in patients with emphysema, whereas anti-HER1-directed strategies may be more useful in patients with pulmonary mucus hypersecretion, as seen in chronic bronchitis and asthma. Growth factors and cytokines including VEGF, fibroblast growth factors, transforming growth factor-beta, tumor necrosis factor-alpha, CXCL1, CXCL8, and CCL2, and signal transduction proteins such as mitogen-activated protein kinase p38 and nuclear factor-kappaB, seem to be important pathogenetic molecules in COPD. Specific antagonists for these proteins may be effective for different inflammatory diseases. However, their efficacy for COPD therapy has not yet been demonstrated. Finally, other drugs such as retinoic acids may provide restoration of lung tissue structure. Such approaches, however, must await the first results of growth factor or cytokine antagonist therapy in chronic lung diseases.
Collapse
|
82
|
Role of the extracellular signal-regulated kinase 1/2 signaling pathway in regulating the secretion of bronchial smooth muscle cells in a rat model of chronic asthma. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200801010-00014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
83
|
Scichilone N, Battaglia S, La Sala A, Bellia V. Clinical implications of airway hyperresponsiveness in COPD. Int J Chron Obstruct Pulmon Dis 2007; 1:49-60. [PMID: 18046902 PMCID: PMC2706603 DOI: 10.2147/copd.2006.1.1.49] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
COPD represents one of the leading causes of mortality in the general population. This study aimed at evaluating the relationship between airway hyperresponsiveness (AHR) and COPD and its relevance for clinical practice. We performed a MEDLINE search that yielded a total of 1919 articles. Eligible studies were defined as articles that addressed specific aspects of AHR in COPD, such as prevalence, pathogenesis, or prognosis. AHR appears to be present in at least one out of two individuals with COPD. The occurrence of AHR in COPD is influenced by multiple mechanisms, among which impairment of factors that oppose airway narrowing plays an important role. The main determinants of AHR are reduction in lung function and smoking status. We envision a dual role of AHR: in suspected COPD, specific determinants of AHR, such as reactivity and the plateau response, may help the physician to discriminate COPD from asthma; in definite COPD, AHR may be relevant for the prognosis. Indeed, AHR is an independent predictor of mortality in COPD patients. Smoking cessation has been shown to reduce AHR. Further studies are needed to elucidate whether this functional change is associated with improvement in lung function and respiratory symptoms.
Collapse
Affiliation(s)
- Nicola Scichilone
- Istituto di Medicina Generale e Pneumologia, Cattedra di Malattie dell'Apparato Respiratorio, Università di Palermo, Palermo, Italy.
| | | | | | | |
Collapse
|
84
|
Mullan CS, Riley M, Clarke D, Tatler A, Sutcliffe A, Knox AJ, Pang L. Beta-tryptase regulates IL-8 expression in airway smooth muscle cells by a PAR-2-independent mechanism. Am J Respir Cell Mol Biol 2007; 38:600-8. [PMID: 18079491 DOI: 10.1165/rcmb.2007-0082oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mast cells are central in the development of several allergic diseases and contain a number of pre-formed mediators. beta-tryptase, the most abundant mast cell product, is increasingly recognized as a key inflammatory mediator, as it causes the release of cytokines, particularly the chemokine IL-8, from both inflammatory and structural cells. The molecular mechanisms, however, remain largely unknown. In this study we sought to investigate whether beta-tryptase could induce IL-8 expression in human airway smooth muscle (ASM) cells and to explore the molecular mechanisms involved. We found that purified human beta-tryptase stimulated IL-8 production in a time- and concentration-dependent manner, which was inhibited by protease inhibitors and mimicked by recombinant human beta-tryptase, but not by the protease-activated receptor-2 (PAR-2) agonist SLIGKV-NH(2), consistent with the low-level expression of PAR-2 protein in these cells. beta-tryptase also up-regulated IL-8 mRNA expression, as analyzed by RT-PCR and real-time PCR, which was abolished by the transcription inhibitor actinomycin D. Reporter gene assay showed that beta-tryptase-induced IL-8 transcription was mediated by the transcription factors activator protein-1, CCAAT/enhancer binding protein, and NF-kappaB, and chromatin immunoprecipitation assay demonstrated that beta-tryptase induced in vivo binding of these transcription factors to the IL-8 gene promoter. Furthermore, beta-tryptase stabilized IL-8 mRNA, suggesting additional post-transcriptional regulation. Collectively these findings show that beta-tryptase up-regulates IL-8 expression in ASM cells through a PAR-2-independent proteolytic mechanism and coordinated transcriptional and post-transcriptional regulation, which may be of particular importance in understanding the role and the mechanisms of action of beta-tryptase in regulating chemokine expression in mast cell-related disorders.
Collapse
Affiliation(s)
- Charlotte S Mullan
- Division of Respiratory Medicine, Clinical Sciences Building, City Hospital, University of Nottingham, Hucknall Road, Nottingham NG5 1PB, UK
| | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
Airway remodeling can be defined as changes in the composition, content, and organization of the cellular and molecular constituents of the airway wall. Airway remodeling is a characteristic feature of asthma, and has important functional implications. These structural changes include epithelial detachment, subepithelial fibrosis, increased airway smooth muscle (ASM) mass, decreased distance between epithelium and ASM cells, goblet cell hyperplasia, mucus gland hyperplasia, proliferation of blood vessels and airway edema and changes in the cartilage. Each can contribute to airway hyperreactivity (AHR), and may eventually lead to irreversible airflow obstruction with disease progression. Structural changes can be observed from early onset of the disease and thus remodeling is thought to be characteristic of asthma. Some aspects of airway remodeling can be explained as a consequence of TH2 inflammation, although it has also been suggested that the exaggerated inflammation and remodeling seen in asthmatic airways is the consequence of abnormal injury and repair responses stemming from the susceptibility of bronchial epithelia to components of the inhaled environment. According to this view, remodeling occurs by way of a noninflammatory mechanism, where inflammation of airways and altered structure and function of the airways are parallel and interacting factors. Airway remodeling in established asthma is poorly responsive to current therapies, such as inhalation of corticosteroids and administration of beta(2)-agonists, antileukotrienes, and theophylline.
Collapse
Affiliation(s)
- Yuki Sumi
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada
| | | |
Collapse
|
86
|
Lu D, Xie S, Sukkar MB, Lu X, Scully MF, Chung KF. Inhibition of Airway Smooth Muscle Adhesion and Migration by the Disintegrin Domain of ADAM-15. Am J Respir Cell Mol Biol 2007; 37:494-500. [PMID: 17575078 DOI: 10.1165/rcmb.2006-0364oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Disintegrin and metalloprotease proteins (ADAMs) are membrane-anchored glycoproteins involved in cell adhesion, cell fusion, protein ecto-domain shedding, and intracellular signaling. We examined whether the disintegrin domain of ADAM-15 (named ddADAM-15) containing an Asp-Gly-Asp (RGD) integrin-binding motif could interfere with airway smooth muscle cell (ASMC) adhesion and migration. Recombinant ddADAM-15 adhered to human ASMCs with saturation kinetics, and was beta(1)-integrin dependent. ddADAM-15 inhibited the binding of fibrinogen but not of fibronectin to ASMCs. ddADAM-15 also inhibited platelet-derived growth factor (PDGF)-induced ASMC migration, and this was reversed by an anti-beta(1)-integrin antibody. PDGF induced the activation of phosphoinositol-3-kinase (PI3K) and p38 mitogen-activated protein kinase (MAPK), and selective inhibitors of these kinases inhibited PDGF-induced ASMC migration. ddADAM-15 did not inhibit PDGF-induced activation of PI3K or p38, thereby excluding these kinase pathways as a mechanism by which ddADAM-15 inhibits ASMC migration. ADAM-15 mRNA and protein were expressed under basal conditions, and both gene and protein expression were inhibited by PDGF. In summary, ddADAM-15 inhibits ASMC adhesion and migration through the beta(1)-integrin, without modulating signaling pathways involved in ASMC migratory responses.
Collapse
Affiliation(s)
- Dong Lu
- Molecular Immunology Section, National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | | | | |
Collapse
|
87
|
Bloemen K, Verstraelen S, Van Den Heuvel R, Witters H, Nelissen I, Schoeters G. The allergic cascade: review of the most important molecules in the asthmatic lung. Immunol Lett 2007; 113:6-18. [PMID: 17765979 DOI: 10.1016/j.imlet.2007.07.010] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Revised: 07/17/2007] [Accepted: 07/22/2007] [Indexed: 12/17/2022]
Abstract
Asthma is the most common chronic inflammatory disorder of the airways among children. It is a complex clinical disease characterized by airway obstruction, airway inflammation and airway hyperresponsiveness to a variety of stimuli. The development of allergic asthma exists of three phases, namely the induction phase, the early-phase asthmatic reaction (EAR) and the late-phase asthmatic reaction (LAR). Each phase is characterized by the production and interplay of various cell-derived mediators. In the induction phase, T helper cytokines are important in the development of asthma. Most important mediators in the EAR are preformed mediators, newly synthesized lipid mediators and cytokines that are produced by mast cells. During the LAR, inflammatory molecules are produced by various cell types, such as eosinophils, neutrophils, T cells, macrophages, dendritic cells, and structural cells. Chronical inflammation leads to structural changes of the airway architecture. In this review, the most important mediators involved in the induction phase, the early-phase and late-phase asthmatic reaction are discussed.
Collapse
Affiliation(s)
- Karolien Bloemen
- Centre of Expertise in Environmental Toxicology, Flemish Institute for Technological Research (VITO), Boeretang 200, B-2400 Mol, Belgium.
| | | | | | | | | | | |
Collapse
|
88
|
Billington CK, Le Jeune IR, Young KW, Hall IP. A major functional role for phosphodiesterase 4D5 in human airway smooth muscle cells. Am J Respir Cell Mol Biol 2007; 38:1-7. [PMID: 17673687 DOI: 10.1165/rcmb.2007-0171oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Relaxation of airway smooth muscle is dependent predominantly upon elevation of cell cAMP content. Although the processes involved in elevation of cell cAMP content are reasonably well established, the mechanisms governing subsequent control of cAMP turnover are less clear. Breakdown of cAMP is solely regulated by phosphodiesterase (PDE) isoenzymes. We have previously reported that PDE4 family members are likely to be important in this process, and that expression of PDE4D variants is actively regulated at the transcriptional level. Here, we demonstrate a key role for PDE4D5 in the control of beta(2)-adrenoceptor (beta(2)AR)-stimulated cAMP activity in human airway smooth muscle cells using splice variant-specific small interfering RNA knockdown. Furthermore, we show, using an Epac (exchange protein directly activated by cAMP)-based, cAMP-sensitive fluorescent probe, that these intracellular cAMP gradients are controlled both temporally and dynamically by PDE4D5. Elevation of cAMP within the cytoplasm after beta(2)AR stimulation is rapid and shows no distinct spatial compartmentalization in these cells. These data suggest that PDE4D5, despite being a minor component of the tissue PDE pool, is the key physiological regulator of beta(2)AR-induced cAMP turnover within human airway smooth muscle.
Collapse
MESH Headings
- Cells, Cultured
- Cyclic AMP/genetics
- Cyclic AMP/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 3
- Cyclic Nucleotide Phosphodiesterases, Type 4
- Gene Expression
- Humans
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Muscle Relaxation/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/enzymology
- Phosphoric Diester Hydrolases/genetics
- Phosphoric Diester Hydrolases/metabolism
- RNA, Small Interfering/genetics
- Receptors, Adrenergic, beta-2/biosynthesis
- Receptors, Adrenergic, beta-2/genetics
- Trachea/cytology
- Trachea/enzymology
- Transcription, Genetic/physiology
Collapse
Affiliation(s)
- Charlotte K Billington
- Division of Therapeutics and Molecular Medicine, University of Nottingham, Queens Medical Centre, Nottingham, UK
| | | | | | | |
Collapse
|
89
|
Bao Z, Lim S, Liao W, Lin Y, Thiemermann C, Leung BP, Wong WSF. Glycogen synthase kinase-3beta inhibition attenuates asthma in mice. Am J Respir Crit Care Med 2007; 176:431-8. [PMID: 17556716 DOI: 10.1164/rccm.200609-1292oc] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Persistent activation of nuclear factor-kappaB has been associated with the development of asthma. Glycogen synthase kinase-3beta is known to regulate the activity of nuclear factor-kappaB. OBJECTIVES We hypothesized that inhibition of glycogen synthase kinase-3beta may have anti-inflammatory effects in allergic asthma. METHODS BALB/c mice sensitized and challenged with ovalbumin developed airway inflammation. Bronchoalveolar lavage fluid was assessed for total and differential cell counts, and for cytokine and chemokine levels. Lung tissues were examined for cell infiltration and mucus hypersecretion, and for the expression of inflammatory biomarkers. Serum immunoglobulin E levels were determined by enzyme-linked immunosorbant assay. Airway hyperresponsiveness was monitored by direct airway resistance analysis. MEASUREMENTS AND MAIN RESULTS Intravenous administration of 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), a selective glycogen synthase kinase-3beta inhibitor, significantly inhibited ovalbumin-induced increases in total cell counts, eosinophil counts, and IL-5, IL-13, and eotaxin levels recovered in bronchoalveolar lavage fluid in a dose-dependent manner. TDZD-8 substantially reduced the serum levels of ovalbumin-specific IgE. Histologic studies showed that TDZD-8 dramatically inhibited ovalbumin-induced lung tissue eosinophilia and airway mucus production. TDZD-8 also markedly suppressed ovalbumin-induced mRNA expression of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, Muc5ac, and three members of the chitinase family (acidic mammalian chitinase, Ym1, and Ym2). In addition, TDZD-8 significantly reduced ovalbumin-induced airway hyperresponsiveness to inhaled methacholine. Western blot analysis of whole lung lysates revealed that TDZD-8 markedly attenuated the phosphorylation of the nuclear factor-kappaB subunit p65 from ovalbumin-challenged mice. CONCLUSIONS Our findings suggest that inhibition of glycogen synthase kinase-3beta may provide a novel means for the treatment of allergic airway inflammation.
Collapse
Affiliation(s)
- Zhang Bao
- Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore
| | | | | | | | | | | | | |
Collapse
|
90
|
An S, Bai T, Bates J, Black J, Brown R, Brusasco V, Chitano P, Deng L, Dowell M, Eidelman D, Fabry B, Fairbank N, Ford L, Fredberg J, Gerthoffer W, Gilbert S, Gosens R, Gunst S, Halayko A, Ingram R, Irvin C, James A, Janssen L, King G, Knight D, Lauzon A, Lakser O, Ludwig M, Lutchen K, Maksym G, Martin J, Mauad T, McParland B, Mijailovich S, Mitchell H, Mitchell R, Mitzner W, Murphy T, Paré P, Pellegrino R, Sanderson M, Schellenberg R, Seow C, Silveira P, Smith P, Solway J, Stephens N, Sterk P, Stewart A, Tang D, Tepper R, Tran T, Wang L. Airway smooth muscle dynamics: a common pathway of airway obstruction in asthma. Eur Respir J 2007; 29:834-60. [PMID: 17470619 PMCID: PMC2527453 DOI: 10.1183/09031936.00112606] [Citation(s) in RCA: 284] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Excessive airway obstruction is the cause of symptoms and abnormal lung function in asthma. As airway smooth muscle (ASM) is the effecter controlling airway calibre, it is suspected that dysfunction of ASM contributes to the pathophysiology of asthma. However, the precise role of ASM in the series of events leading to asthmatic symptoms is not clear. It is not certain whether, in asthma, there is a change in the intrinsic properties of ASM, a change in the structure and mechanical properties of the noncontractile components of the airway wall, or a change in the interdependence of the airway wall with the surrounding lung parenchyma. All these potential changes could result from acute or chronic airway inflammation and associated tissue repair and remodelling. Anti-inflammatory therapy, however, does not "cure" asthma, and airway hyperresponsiveness can persist in asthmatics, even in the absence of airway inflammation. This is perhaps because the therapy does not directly address a fundamental abnormality of asthma, that of exaggerated airway narrowing due to excessive shortening of ASM. In the present study, a central role for airway smooth muscle in the pathogenesis of airway hyperresponsiveness in asthma is explored.
Collapse
Affiliation(s)
- S.S. An
- Division of Physiology, Dept of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health
| | - T.R. Bai
- James Hogg iCAPTURE Centre, University of British Columbia, Vancouver
| | - J.H.T. Bates
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, VT
| | - J.L. Black
- Dept of Pharmacology, University of Sydney, Sydney
| | - R.H. Brown
- Dept of Anesthesiology and Critical Care medicine, Johns Hopkins Medical Institutions, Baltimore, MD
| | - V. Brusasco
- Dept of Internal Medicine, University of Genoa, Genoa
| | - P. Chitano
- Dept of Paediatrics, Duke University Medical Center, Durham, NC
| | - L. Deng
- Program in Molecular and Integrative Physiological Sciences, Dept of Environmental Health, Harvard School of Public Health
- Bioengineering College, Chongqing University, Chongqing, China
| | - M. Dowell
- Section of Pulmonary and Critical Care Medicine
| | - D.H. Eidelman
- Meakins-Christie Laboratories, Dept of Medicine, McGill University, Montreal
| | - B. Fabry
- Center for Medical Physics and Technology, Erlangen, Germany
| | - N.J. Fairbank
- School of Biomedical Engineering, Dalhousie University, Halifax
| | | | - J.J. Fredberg
- Program in Molecular and Integrative Physiological Sciences, Dept of Environmental Health, Harvard School of Public Health
| | - W.T. Gerthoffer
- Dept of Pharmacology, University of Nevada School of Medicine, Reno, NV
| | | | - R. Gosens
- Dept of Physiology, University of Manitoba, Winnipeg
| | - S.J. Gunst
- Dept of Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - A.J. Halayko
- Dept of Physiology, University of Manitoba, Winnipeg
| | - R.H. Ingram
- Dept of Medicine, Emory University School of Medicine, Atlanta, GA
| | - C.G. Irvin
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, VT
| | - A.L. James
- West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands
| | - L.J. Janssen
- Dept of Medicine, McMaster University, Hamilton, Canada
| | - G.G. King
- Woolcock Institute of Medical Research, Camperdown
| | - D.A. Knight
- James Hogg iCAPTURE Centre, University of British Columbia, Vancouver
| | - A.M. Lauzon
- Meakins-Christie Laboratories, Dept of Medicine, McGill University, Montreal
| | - O.J. Lakser
- Section of Paediatric Pulmonary Medicine, University of Chicago, Chicago, IL
| | - M.S. Ludwig
- Meakins-Christie Laboratories, Dept of Medicine, McGill University, Montreal
| | - K.R. Lutchen
- Dept of Biomedical Engineering, Boston University, Boston
| | - G.N. Maksym
- School of Biomedical Engineering, Dalhousie University, Halifax
| | - J.G. Martin
- Meakins-Christie Laboratories, Dept of Medicine, McGill University, Montreal
| | - T. Mauad
- Dept of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | | | - S.M. Mijailovich
- Program in Molecular and Integrative Physiological Sciences, Dept of Environmental Health, Harvard School of Public Health
| | - H.W. Mitchell
- Discipline of Physiology, School of Biomedical, Biomolecular and Chemical Sciences, University of Western Australia, Perth
| | | | - W. Mitzner
- Division of Physiology, Dept of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health
| | - T.M. Murphy
- Dept of Paediatrics, Duke University Medical Center, Durham, NC
| | - P.D. Paré
- James Hogg iCAPTURE Centre, University of British Columbia, Vancouver
| | - R. Pellegrino
- Dept of Respiratory Physiopathology, S. Croce e Carle Hospital, Cuneo, Italy
| | - M.J. Sanderson
- Dept of Physiology, University of Massachusetts Medical School, Worcester, MA
| | - R.R. Schellenberg
- James Hogg iCAPTURE Centre, University of British Columbia, Vancouver
| | - C.Y. Seow
- James Hogg iCAPTURE Centre, University of British Columbia, Vancouver
| | - P.S.P. Silveira
- Dept of Pathology, Sao Paulo University Medical School, Sao Paulo, Brazil
| | - P.G. Smith
- Dept of Paediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - J. Solway
- Section of Pulmonary and Critical Care Medicine
| | - N.L. Stephens
- Dept of Physiology, University of Manitoba, Winnipeg
| | - P.J. Sterk
- Dept of Pulmonology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - A.G. Stewart
- Dept of Pharmacology, University of Melbourne, Parkville, Australia
| | - D.D. Tang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY, USA
| | - R.S. Tepper
- Dept of Paediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - T. Tran
- Dept of Physiology, University of Manitoba, Winnipeg
| | - L. Wang
- Dept of Paediatrics, Duke University Medical Center, Durham, NC
| |
Collapse
|
91
|
Ye YM, Kang YM, Kim SH, Kim CW, Kim HR, Hong CS, Park CS, Kim HM, Nahm DH, Park HS. Relationship between neurokinin 2 receptor gene polymorphisms and serum vascular endothelial growth factor levels in patients with toluene diisocyanate-induced asthma. Clin Exp Allergy 2007; 36:1153-60. [PMID: 16961715 DOI: 10.1111/j.1365-2222.2006.02547.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Among the various pathogenic mechanisms of toluene diisocyanate (TDI)-induced asthma, a contribution from neurogenic inflammation has been suggested. OBJECTIVE To evaluate neurokinin 2 receptor (NK2R) gene polymorphisms in association with the clinical phenotype of TDI-induced asthma, 70 TDI-induced occupational asthma (TDI-OA)patients, 59 asymptomatic exposed controls (AEC), and 93 unexposed healthy controls (NC) were enrolled in the study. METHODS Two single-nucleotide polymorphisms (SNPs) of NK2R, 7853G>A (Gly231Glu) and 11 424G>A (Arg375His), were genotyped using a single base extension method. The levels of PC20 methacholine, specific IgE and IgG to TDI-human serum albumin conjugate, and serum vascular endothelial growth factor (VEGF), matrix metalloproteinase-9, and TGF-beta1 were compared according to the NK2R genotypes of the subjects with TDI-OA and AEC. RESULTS No significant differences in allele, genotype, or haplotype frequencies of these two SNPs were noted among the three groups (P>0.05, respectively). Moreover, subjects with the NK2R 7853GG genotype had higher serum VEGF levels than those with GA or AA among the TDI-exposed workers (P=0.040). CONCLUSION The NK2R 7853GG genotype may contribute to increased serum VEGF levels, which result in airway inflammation after TDI exposure.
Collapse
Affiliation(s)
- Y-M Ye
- Department of Allergy & Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Dragon S, Rahman MS, Yang J, Unruh H, Halayko AJ, Gounni AS. IL-17 enhances IL-1beta-mediated CXCL-8 release from human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2006; 292:L1023-9. [PMID: 17189320 DOI: 10.1152/ajplung.00306.2006] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies into the pathogenesis of airway disorders such as asthma have revealed a dynamic role for airway smooth muscle cells in the perpetuation of airway inflammation via secretion of cytokines and chemokines. In this study, we evaluated whether IL-17 could enhance IL-1beta-mediated CXCL-8 release from human airway smooth muscle cells (HASMC) and investigated the upstream and downstream signaling events regulating the induction of CXCL-8. CXCL-8 mRNA and protein induction were assessed by real-time RT-PCR and ELISA from primary HASMC cultures. HASMC transfected with site-mutated activator protein (AP)-1/NF-kappaB CXCL-8 promoter constructs were treated with selective p38, MEK1/2, and phosphatidylinositol 3-kinase (PI3K) inhibitors to determine the importance of MAPK and PI3K signaling pathways as well as AP-1 and NF-kappaB promoter binding sites. We demonstrate IL-17 induced and synergized with IL-1beta to upregulate CXCL-8 mRNA and protein levels. Erk1/2 and p38 modulated IL-17 and IL-1beta CXCL-8 promoter activity; however, IL-1beta also activated the PI3K pathway. The synergistic response mediating CXCL-8 promoter activity was dependent on both MAPK and PI3K signal transduction pathways and required the cooperation of AP-1 and NF-kappaB cis-acting elements upstream of the CXCL-8 gene. Collectively, our observations indicate MAPK and PI3K pathways regulate the synergy of IL-17 and IL-1beta to enhance CXCL-8 promoter activity, mRNA induction, and protein synthesis in HASMC via the cooperative activation of AP-1 and NF-kappaB trans-acting elements.
Collapse
Affiliation(s)
- Stéphane Dragon
- Department of Immunology, University of Manitoba, Manitoba, Canada
| | | | | | | | | | | |
Collapse
|
93
|
Chiou YL, Shieh JJ, Lin CY. Blocking of Akt/NF-kappaB signaling by pentoxifylline inhibits platelet-derived growth factor-stimulated proliferation in Brown Norway rat airway smooth muscle cells. Pediatr Res 2006; 60:657-62. [PMID: 17065572 DOI: 10.1203/01.pdr.0000246105.56278.98] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The proliferation of airway smooth muscle cells (ASMC) can cause airway hyperresponsiveness (AHR). It has been reported that platelet-derived growth factor (PDGF) can stimulate the proliferation of ASMC through phosphatidylinositol 3-kinase (PI3 K) signaling pathway, which can activate Akt protein. Activated-Akt can activate downstream signal protein [p70S6 K, nuclear factor (NF)-kappaB, and extracellular signal regulated kinase (ERK)], increasing the cyclin D1 level and suppressing the transcription of p27Kip1 to enable cell cycle entry. This investigation demonstrated that pentoxifylline (PTX) inhibited the PDGF-stimulated proliferation of ASMC by suppressing activation of the Akt/NF-kappaB pathway. ASMC were treated with PTX for 48 h, which attenuated the PDGF-stimulated proliferation of ASMC. PTX and wortmannin, a PI3 K inhibitor, not only inhibited the PDGF-activated phosphorylation of Akt but also suppressed p70S6 K expression and IkappaBalpha degradation, inhibiting nuclear translocation and the DNA binding activity of NF-kappaB. However, PTX did not influence the phosphorylation of ERK1/2. The suppression of p70S6 K by rapamycin did not influence cyclin D1 expression in PDGF-stimulated cells. These data reveal that the down-regulation of the Akt/NF-kappaB signaling pathway by PTX inhibited the proliferation of ASMC. PTX may provide information on the pathogenesis of asthma.
Collapse
Affiliation(s)
- Ya-Ling Chiou
- Institute of Immunology and Microbiology, National Yang-Ming University, Taipei 112, Taiwan
| | | | | |
Collapse
|
94
|
Sabroe I, Parker LC, Dockrell DH, Davies DE, Dower SK, Whyte MKB. Targeting the networks that underpin contiguous immunity in asthma and chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2006; 175:306-11. [PMID: 17138954 DOI: 10.1164/rccm.200606-777pp] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Recent advances in the field of innate immunity have driven an important reappraisal of the role of these processes in airway disease. Various strands of evidence indicate that resident cells, such as macrophages and epithelial cells, have central importance in the initiation of inflammation. Macrophage activation has the potential to regulate not just typical aspects of innate immunity but also, via a variety of intricate cell-cell networks, adaptive responses and responses characterized by Th2-type cytokine production. In turn, such adaptive immune processes modify the phenotype and function of the innate immune system. Cooperative responses between monocytic cells and tissue cells are likely to be crucial to the generation of effective inflammatory responses, and a realization of the importance of these networks is providing a new way of identifying antiinflammatory therapies. Importantly, the repeated cycles of allergic and nonallergic inflammation that comprise chronic human airway disease are not necessarily well described by current terminology, and we propose and describe a concept of contiguous immunity, in which continual bidirectional cross-talk between innate and adaptive immunity describes disease processes more accurately.
Collapse
Affiliation(s)
- Ian Sabroe
- Academic Unit of Respiratory Medicine, School of Medicine and Biomedical Sciences, University of Sheffield, L Floor, Royal Hallamshire Hospital, Sheffield, UK.
| | | | | | | | | | | |
Collapse
|
95
|
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. [PMID: 16968549 PMCID: PMC1586013 DOI: 10.1186/1465-9921-7-117] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 09/12/2006] [Indexed: 01/08/2023] Open
Abstract
Background Airway smooth muscle (ASM) contraction underlies acute bronchospasm in asthma. ASM cells can switch between a synthetic-proliferative phenotype and a contractile phenotype. While the effects of extracellular matrix (ECM) components on modulation of ASM cells to a synthetic phenotype have been reported, the role of ECM components on maturation of ASM cells to a contractile phenotype in adult lung is unclear. As both changes in ECM components and accumulation of contractile ASM are features of airway wall remodelling in asthma, we examined the role of the ECM protein, laminin, in the maturation of contractile phenotype in human ASM cells. Methods Human ASM cells were made senescence-resistant by stable expression of human telomerase reverse transcriptase. Maturation to a contractile phenotype was induced by 7-day serum deprivation, as assessed by immunoblotting for desmin and calponin. The role of laminin on ASM maturation was investigated by comparing the effects of exogenous laminin coated on culture plates, and of soluble laminin peptide competitors. Endogenous expression of laminin chains during ASM maturation was also measured. Results Myocyte binding to endogenously expressed laminin was required for ASM phenotype maturation, as laminin competing peptides (YIGSR or GRGDSP) significantly reduced desmin and calponin protein accumulation that otherwise occurs with prolonged serum deprivation. Coating of plastic cell culture dishes with different purified laminin preparations was not sufficient to further promote accumulation of desmin or calponin during 7-day serum deprivation. Expression of α2, β1 and γ1 laminin chains by ASM cells was specifically up-regulated during myocyte maturation, suggesting a key role for laminin-2 in the development of the contractile phenotype. Conclusion While earlier reports suggest exogenously applied laminin slows the spontaneous modulation of ASM to a synthetic phenotype, we show for the first time that endogenously expressed laminin is required for ASM maturation to the contractile phenotype. As endogenously expressed laminin chains α2, β1 and γ1 are uniquely increased during myocyte maturation, these laminin chains may be key in this process. Thus, human ASM maturation appears to involve regulated endogenous expression of a select set of laminin chains that are essential for accumulation of contractile phenotype myocytes.
Collapse
Affiliation(s)
- Thai Tran
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
| | - Karol D McNeill
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
| | - William T Gerthoffer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV, USA
| | - Helmut Unruh
- Section of Thoracic Surgery, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew J Halayko
- Departments of Physiology and Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, MB, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, MB, Canada
- Section of Respiratory Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
96
|
Abstract
Airway remodeling in asthma has been recognized as structural changes of airways such as smooth muscle hypertrophy (an increase in size of airway smooth muscle cells) and hyperplasia (an increase in the number of airway smooth muscle cells), thickening and fibrosis of sub-epithelial basement membrane, hypertrophy of bronchial glands, goblet cell hyperplasia, and thickening of airway epithelium. In these pathological changes, airway smooth muscle remodeling has been recognized as one of the most important factors related to in vitro and in vitro airway responsiveness and the severity of asthma. Both hypertrophy and hyperplasia have been shown in asthmatic airways by morphometrical analyses, although there is a wide variation in the contribution of each mechanism in each patient. Such changes could also be recognized as a phenotypic modulation of airway smooth muscle. On the background of airway smooth muscle remodeling, the existence of several contributing factors, such as inflammatory mediators, growth factors, cytokines, extra-cellular matrix proteins, and genetic factors have been suggested. On the other hand, recent studies revealed that airway smooth muscle could also be a source of inflammatory mediators promoting airway inflammation. In this article, the recent understanding in the mechanisms of airway smooth muscle remodeling in asthma, its relations to airway inflammation and airway physiology, and possible usefulness of early intervention with inhaled glucocorticoids have been discussed.
Collapse
Affiliation(s)
- Mitsuru Munakata
- Department of Pulmonary Medicine, School of Medicine, Fukushima Medical University, Fukushima City, Fukushima, Japan.
| |
Collapse
|
97
|
Abstract
There has been a recent increase in the prevalence of asthma worldwide; however, the 5-10% of patients with severe disease account for a substantial proportion of the health costs. Although most asthma cases can be satisfactorily managed with a combination of anti-inflammatory drugs and bronchodilators, patients who remain symptomatic despite maximum combination treatment represent a heterogeneous group consisting of those who are under-treated or non-adherent with their prescribed medication. After excluding under-treatment and poor compliance, corticosteroid refractory asthma can be identified as a subphenotype characterised by a heightened neutrophilic airway inflammatory response in the presence or absence of eosinophils, with evidence of increased tissue injury and remodelling. Although a wide range of environmental factors such as allergens, smoking, air pollution, infection, hormones, and specific drugs can contribute to this phenotype, other features associated with changes in the airway inflammatory response should be taken into account. Aberrant communication between an injured airway epithelium and underlying mesenchyme contributes to disease chronicity and refractoriness to corticosteroids. The importance of identifying underlying causative factors and the recent introduction of novel therapeutic approaches, including the targeting of immunoglobulin E and tumour necrosis factor alpha with biological agents, emphasise the need for careful phenotyping of patients with severe disease to target improved management of the individual patient's needs.
Collapse
Affiliation(s)
- Stephen T Holgate
- AIR Division, Level D Centre Block, Southampton General Hospital, Southampton, UK.
| | | |
Collapse
|
98
|
Ammit AJ, Moir LM, Oliver BG, Hughes JM, Alkhouri H, Ge Q, Burgess JK, Black JL, Roth M. Effect of IL-6 trans-signaling on the pro-remodeling phenotype of airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2006; 292:L199-206. [PMID: 16936245 DOI: 10.1152/ajplung.00230.2006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Increased levels of IL-6 are documented in asthma, but its contribution to the pathology is unknown. Asthma is characterized by airway wall thickening due to increased extracellular matrix deposition, inflammation, angiogenesis, and airway smooth muscle (ASM) mass. IL-6 binds to a specific membrane-bound receptor, IL-6 receptor-alpha (mIL-6Ralpha), and subsequently to the signaling protein gp130. Alternatively, IL-6 can bind to soluble IL-6 recpetor-alpha (sIL-6Ralpha) to stimulate membrane receptor-deficient cells, a process called trans-signaling. We discovered that primary human ASM cells do not express mIL-6Ralpha and, therefore, investigated the effect of IL-6 trans-signaling on the pro-remodeling phenotype of ASM. ASM required sIL-6Ralpha to activate signal transducer and activator 3, with no differences observed between cells from asthmatic subjects compared with controls. Further analysis revealed that IL-6 alone or with sIL-6Ralpha did not induce release of matrix-stimulating factors (including connective tissue growth factor, fibronectin, or integrins) and had no effect on mast cell adhesion to ASM or ASM proliferation. However, in the presence of sIL-6Ralpha, IL-6 increased eotaxin and VEGF release and may thereby contribute to local inflammation and vessel expansion in airway walls of asthmatic subjects. As levels of sIL-6Ralpha are increased in asthma, this demonstration of IL-6 trans-signaling in ASM has relevance to the development of airway remodeling.
Collapse
Affiliation(s)
- Alaina J Ammit
- Faculty of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Lazaar AL, Panettieri RA. Airway smooth muscle as a regulator of immune responses and bronchomotor tone. Clin Chest Med 2006; 27:53-69, vi. [PMID: 16543052 DOI: 10.1016/j.ccm.2005.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The traditional view of airway smooth muscle (ASM) in asthma, as 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. This article also discusses how altered synthetic function contributes to airway remodeling.
Collapse
Affiliation(s)
- Aili L Lazaar
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Medical Center, BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA.
| | | |
Collapse
|
100
|
Tang MLK, Wilson JW, Stewart AG, Royce SG. Airway remodelling in asthma: current understanding and implications for future therapies. Pharmacol Ther 2006; 112:474-88. [PMID: 16759709 DOI: 10.1016/j.pharmthera.2006.05.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Accepted: 05/01/2006] [Indexed: 02/08/2023]
Abstract
Airway remodelling refers to the structural changes that occur in the airway wall in asthma. These include epithelial hyperplasia and metaplasia, subepithelial fibrosis, muscle cell hyperplasia and angiogenesis. These structural changes result in thickening of the airway wall, airway hyperresponsiveness (AHR), and a progressive irreversible loss of lung function. The precise sequence of events that take place during the remodelling process and the mechanisms regulating these changes remain poorly understood. It is thought that airway remodelling is initiated and promoted by repeated episodes of allergic inflammation that damage the surface epithelium of the airway. However, other mechanisms are also likely to contribute to this process. Moreover, the interrelationship between airway remodelling, inflammation and AHR has not been clearly defined. Currently, there are no effective treatments that halt or reverse the changes of airway remodelling and its effects on lung function. Glucocorticoids have been unable to eliminate the progression of remodelling changes and there is limited evidence of a beneficial effect from other available therapies. The search for novel therapies that can directly target individual components of the remodelling process should be made a priority. In this review, we describe the current understanding of the airway remodelling process and the mechanisms regulating its development. The impact of currently available asthma therapies on airway remodelling is also discussed.
Collapse
Affiliation(s)
- Mimi L K Tang
- Department of Immunology, Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne 3052, Australia.
| | | | | | | |
Collapse
|