1
|
Liu Y, Li J, Chen R, Shi F, Xiong Y. Airway epithelial cells promote in vitro airway smooth muscle cell proliferation by activating the Wnt/β-catenin pathway. Respir Physiol Neurobiol 2024; 331:104368. [PMID: 39536926 DOI: 10.1016/j.resp.2024.104368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/02/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Asthma is a common chronic inflammatory airway disease, imposing a substantial health and economic burden on society and individuals. Current treatments primarily focus on symptom relief and lung function improvement, often failing to address the underlying pathology. Thus, exploring new therapeutic approaches is crucial. Airway smooth muscle cells (ASMCs) play a key role in regulating airway tone and airflow, while abnormal ASMCs proliferation contributes to airway remodeling in asthma. Airway epithelial cells (AECs), serving as the first barrier against pathogens and allergens, also have critical immune functions. This study focuses on the interaction between AECs and ASMCs, as AECs are more accessible for drug delivery due to their location at the airway surface. Investigating this relationship could facilitate novel interventions targeting AECs to inhibit pathological ASMCs activity. In our experiment, we isolated ASMCs and AECs from healthy mice and found that AECs significantly promoted ASMCs proliferation in co-culture. RNA sequencing revealed that this process might be linked to the activation of the canonical Wnt signaling pathway in ASMCs. By using Wnt pathway inhibitors (endo-IWR1) and siRNA to disrupt Wnt receptors, we reversed this phenotype. This finding suggests that AECs may promote ASMCs proliferation by activating the Wnt pathway in ASMCs. The Wnt/β-catenin pathway appears to play an important role in ASMCs proliferation, indicating that future pathological studies should consider the potential involvement of the Wnt pathway in airway remodeling.
Collapse
Affiliation(s)
- Yilun Liu
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518038, China
| | - Jiana Li
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518038, China
| | - Rongchang Chen
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518109, China
| | - Fei Shi
- Emergency Department, Shenzhen People's Hospital, Shenzhen, Guangdong Province 518106, China.
| | - Yi Xiong
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518038, China.
| |
Collapse
|
2
|
Varricchi G, Ferri S, Pepys J, Poto R, Spadaro G, Nappi E, Paoletti G, Virchow JC, Heffler E, Canonica WG. Biologics and airway remodeling in severe asthma. Allergy 2022; 77:3538-3552. [PMID: 35950646 PMCID: PMC10087445 DOI: 10.1111/all.15473] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Asthma is a chronic inflammatory airway disease resulting in airflow obstruction, which in part can become irreversible to conventional therapies, defining the concept of airway remodeling. The introduction of biologics in severe asthma has led in some patients to the complete normalization of previously considered irreversible airflow obstruction. This highlights the need to distinguish a "fixed" airflow obstruction due to structural changes unresponsive to current therapies, from a "reversible" one as demonstrated by lung function normalization during biological therapies not previously obtained even with high-dose systemic glucocorticoids. The mechanisms by which exposure to environmental factors initiates the inflammatory responses that trigger airway remodeling are still incompletely understood. Alarmins represent epithelial-derived cytokines that initiate immunologic events leading to inflammatory airway remodeling. Biological therapies can improve airflow obstruction by addressing these airway inflammatory changes. In addition, biologics might prevent and possibly even revert "fixed" remodeling due to structural changes. Hence, it appears clinically important to separate the therapeutic effects (early and late) of biologics as a new paradigm to evaluate the effects of these drugs and future treatments on airway remodeling in severe asthma.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Sebastian Ferri
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy
| | - Jack Pepys
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization (WAO) Center of Excellence, Naples, Italy
| | - Emanuele Nappi
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Giovanni Paoletti
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Enrico Heffler
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Walter G Canonica
- Personalized Medicine Asthma and Allergy Unit - IRCCS Humanitas Research Hospital, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
3
|
Glucocorticoid Insensitivity in Asthma: The Unique Role for Airway Smooth Muscle Cells. Int J Mol Sci 2022; 23:ijms23168966. [PMID: 36012240 PMCID: PMC9408965 DOI: 10.3390/ijms23168966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Although most patients with asthma symptoms are well controlled by inhaled glucocorticoids (GCs), a subgroup of patients suffering from severe asthma respond poorly to GC therapy. Such GC insensitivity (GCI) represents a profound challenge in managing patients with asthma. Even though GCI in patients with severe asthma has been investigated by several groups using immune cells (peripheral blood mononuclear cells and alveolar macrophages), uncertainty exists regarding the underlying molecular mechanisms in non-immune cells, such as airway smooth cells (ASM) cells. In asthma, ASM cells are among the targets of GC therapy and have emerged as key contributors not only to bronchoconstriction but also to airway inflammation and remodeling, as implied by experimental and clinical evidence. We here summarize the current understanding of the actions/signaling of GCs in asthma, and specifically, GC receptor (GR) “site-specific phosphorylation” and its role in regulating GC actions. We also review some common pitfalls associated with studies investigating GCI and the inflammatory mediators linked to asthma severity. Finally, we discuss and contrast potential molecular mechanisms underlying the impairment of GC actions in immune cells versus non-immune cells such as ASM cells.
Collapse
|
4
|
Lin TY, Chang PJ, Lo CY, Lo YL, Yu CT, Lin SM, Kuo CHS, Lin HC. Interaction Between CD34 + Fibrocytes and Airway Smooth Muscle Promotes IL-8 Production and Akt/PRAS40/mTOR Signaling in Asthma. Front Med (Lausanne) 2022; 9:823994. [PMID: 35547213 PMCID: PMC9081978 DOI: 10.3389/fmed.2022.823994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background The circulating progenitor cells of fibroblasts (fibrocytes) have been shown to infiltrate the airway smooth muscle compartment of asthma patients; however, the pathological significance of this discovery has yet to be elucidated. This study established a co-culture model of airway smooth muscle cells (ASMCs) and fibrocytes from asthmatic or normal subjects to evaluate innate cytokine production, corticosteroid responses, and signaling in ASMCs. Methods CD34+ fibrocytes were purified from peripheral blood of asthmatic (Global Initiative for Asthma treatment step 4–5) and normal subjects and cultured for 5∼7 days. In a transwell plate, ASMCs were co-cultured with fibrocytes at a ratio of 2:1, ASMCs were cultured alone (control condition), and fibrocytes were cultured alone for 48 h. Measurements were obtained of interleukin-8 (IL-8), IL-6, IL-17, thymic stromal lymphopoietin, and IL-33 levels in the supernatant and IL-33 levels in the cell lysate of the co-culture. Screening for intracellular signaling in the ASMCs after stimulation was performed using condition medium from the patients’ co-culture (PtCM) or IL-8. mRNA and western blot analysis were used to analyze AKT/mTOR signaling in ASMCs stimulated via treatment with PtCM or IL-8. Results Compared with ASMCs cultured alone, IL-8 levels in the supernatant and IL-33 levels in the ASMCs lysate were significantly higher in samples co-cultured from asthmatics, but not in those co-cultured from normal subjects. Corticosteroid-induced suppression of IL-8 production was less pronounced in ASMCs co-cultured with fibrocytes from asthma patients than in ASMCs co-cultured from normal subjects. ASMCs stimulated using PtCM and IL-8 presented elevating activated AKT substrate PRAS40. Treatment with IL-8 and PtCM increased mRNA expression of mTOR and P70S6 kinases in ASMCs. Treatment with IL-8 and PtCM also significantly increased phosphorylation of AKT and mTOR subtract S6 ribosomal protein in ASMCs. Conclusion The interaction between ASMCs and fibrocytes from asthmatic patients was shown to increase IL-8 and IL-33 production and promote AKT/mTOR signaling in ASMCs. IL-8 production in the co-culture from asthmatic patients was less affected by corticosteroid than was that in the co-culture from normal subjects. Our results elucidate the novel role of fibrocytes and ASMCs in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Ting-Yu Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Jui Chang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Lun Lo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Teng Yu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-His Scott Kuo
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Chyuan Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
5
|
Liu D, Xu W, Tang Y, Cao J, Chen R, Wu D, Chen H, Su B, Xu J. Nebulization of risedronate alleviates airway obstruction and inflammation of chronic obstructive pulmonary diseases via suppressing prenylation-dependent RAS/ERK/NF-κB and RhoA/ROCK1/MLCP signaling. Respir Res 2022; 23:380. [PMID: 36575527 PMCID: PMC9795678 DOI: 10.1186/s12931-022-02274-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a progressive disorder that causes airway obstruction and lung inflammation. The first-line treatment of COPD is the bronchodilators of β2-agonists and antimuscarinic drugs, which can help control the airway obstruction, but the long-term use might render the drug tolerance. Bisphosphonates are widely used in osteoclast-mediated bone diseases treatment for decades. For drug repurposing, can delivery of a third generation of nitrogen-containing bisphosphonate, risedronate (RIS) ameliorate the progression of COPD? METHODS COPD rats or mice models have been established through cigarette-smoking and elastase injection, and then the animals are received RIS treatment via nebulization. Lung deposition of RIS was primarily assessed by high-performance liquid chromatography (HPLC). The respiratory parameters of airway obstruction in COPD rats and mice were documented using plethysmography method and resistance-compliance system. RESULTS High lung deposition and bioavailability of RIS was monitored with 88.8% of RIS input dose. We found that RIS could rescue the lung function decline of airspace enlargement and mean linear intercept in the COPD lung. RIS could curb the airway obstruction by suppressing 60% of the respiratory resistance and elevating the airway's dynamic compliance, tidal volume and mid-expiratory flow. As an inhibitor of farnesyl diphosphate synthase (FDPS), RIS suppresses FDPS-mediated RAS and RhoA prenylation to obstruct its membrane localization in airway smooth muscle cells (ASMCs), leading to the inhibition of downstream ERK-MLCK and ROCK1-MLCP pathway to cause ASMCs relaxation. Additionally, RIS nebulization impeded pro-inflammatory cell accumulation, particularly macrophages infiltration in alveolar parenchyma. The NF-κB, tumor necrosis factor-alpha, IL-1β, IL-8, and IL-6 declined in microphages following RIS nebulization. Surprisingly, nebulization of RIS could overcome the tolerance of β2-agonists in COPD-rats by increasing the expression of β2 receptors. CONCLUSIONS Nebulization of RIS could alleviate airway obstruction and lung inflammation in COPD, providing a novel strategy for treating COPD patients, even those with β2-agonists tolerance.
Collapse
Affiliation(s)
- Di Liu
- grid.24516.340000000123704535Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China ,grid.24516.340000000123704535Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Wen Xu
- grid.24516.340000000123704535Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yuan Tang
- grid.24516.340000000123704535Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China ,grid.252957.e0000 0001 1484 5512Basic Medical College, Bengbu Medical College, Bengbu, People’s Republic of China
| | - Jingxue Cao
- grid.24516.340000000123704535Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China ,grid.24516.340000000123704535Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Ran Chen
- grid.24516.340000000123704535Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Dingwei Wu
- Zhejiang Xianju Pharmaceutical Co., Ltd, Xianju, People’s Republic of China
| | - Hongpeng Chen
- Zhejiang Xianju Pharmaceutical Co., Ltd, Xianju, People’s Republic of China
| | - Bo Su
- grid.24516.340000000123704535Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China ,grid.252957.e0000 0001 1484 5512School of Life Sciences, Bengbu Medical College, Bengbu, People’s Republic of China
| | - Jinfu Xu
- grid.24516.340000000123704535Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
6
|
Kadowaki M, Sato K, Kamio H, Kumagai M, Sato R, Nyui T, Umeda Y, Waseda Y, Anzai M, Aoki-Saito H, Koga Y, Hisada T, Tomura H, Okajima F, Ishizuka T. Metal-Stimulated Interleukin-6 Production Through a Proton-Sensing Receptor, Ovarian Cancer G Protein-Coupled Receptor 1, in Human Bronchial Smooth Muscle Cells: A Response Inhibited by Dexamethasone. J Inflamm Res 2021; 14:7021-7034. [PMID: 34955648 PMCID: PMC8694576 DOI: 10.2147/jir.s326964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Human bronchial smooth muscle cells (BSMCs) contribute to airway obstruction and hyperresponsiveness in patients with bronchial asthma. BSMCs also generate cytokines and matricellular proteins in response to extracellular acidification through the ovarian cancer G protein-coupled receptor 1 (OGR1). Cobalt (Co) and nickel (Ni) are occupational agents, which cause occupational asthma. We examined the effects of Co and Ni on interleukin-6 (IL-6) secretion by human BSMCs because these metals may act as ligands of OGR1. Methods Human BSMCs were incubated in Dulbecco's Modified Eagle Medium (DMEM) containing 0.1% bovine serum albumin (BSA) (0.1% BSA-DMEM) for 16 hours and stimulated for the indicated time by exchanging the medium with 0.1% BSA-DMEM containing any of the metals or pH-adjusted 0.1% BSA-DMEM. IL-6 mRNA expression was quantified via reverse transcription polymerase chain reaction (RT-PCR) using the real-time TaqMan technology. IL-6 was measured using an enzyme-linked immunosorbent assay. Dexamethasone (DEX) was added 30 minutes before each stimulation. To knock down the expression of OGR1 in BSMCs, small interfering RNA (siRNA) targeting OGR1 (OGR1-siRNA) was transfected to the cells and non-targeting siRNA (NT-siRNA) was used as a control. Results Co and Ni both significantly increased IL-6 secretion in human BSMCs at 300 μM. This significant increase in IL-6 mRNA expression was observed 5 hours after stimulation. BSMCs transfected with OGR1-siRNA produced less IL-6 than BSMCs transfected with NT-siRNA in response to either Co or Ni stimulation. DEX inhibited Co- and Ni-stimulated IL-6 secretion by human BSMCs as well as pH 6.3-stimulated IL-6 secretion in a dose-dependent manner. DEX did not decrease phosphorylation of ERK1/2, p38 MAP kinase, and NF-κB p65 induced by either Co or Ni stimulation. Conclusion Co and Ni induce secretion of IL-6 in human BSMCs through activation of OGR1. Co- and Ni-stimulated IL-6 secretion is inhibited by DEX.
Collapse
Affiliation(s)
- Maiko Kadowaki
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebeshi, 371-8512, Japan
| | - Hisashi Kamio
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| | - Makoto Kumagai
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| | - Rikishi Sato
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| | - Takafumi Nyui
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| | - Yukihiro Umeda
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Yuko Waseda
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Masaki Anzai
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Haruka Aoki-Saito
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, Maebeshi, 371-8511, Japan
| | - Yasuhiko Koga
- Department of Respiratory Medicine, Gunma University Graduate School of Medicine, Maebeshi, 371-8511, Japan
| | - Takeshi Hisada
- Gunma University Graduate School of Health Sciences, Maebeshi, 371-8514, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Division of Life Science, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Fumikazu Okajima
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| |
Collapse
|
7
|
D'Annunzio G, Gobbo F, Avallone G, Bacci B, Sabattini S, Sarli G. Airway Remodeling in Feline Lungs. Top Companion Anim Med 2021; 46:100587. [PMID: 34624551 DOI: 10.1016/j.tcam.2021.100587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022]
Abstract
Airway remodeling encompass structural changes that occur as the result of chronic injury and lead to persistently altered airway structure and function. Although this process is known in several human respiratory conditions such as asthma and chronic obstructive pulmonary disease (COPD), airway remodeling is poorly characterized in the feline counterpart. In this study, we describe the spontaneous pulmonary changes in 3 cats paralleling the airway remodeling reported in humans. We observed airway smooth muscle cells (ASMCs) hyperplasia (peribronchial and interstitial), airway subepithelial and interstitial fibrosis, and vascular remodeling by increased number of vessels in the bronchial submucosa. The hyperplastic ASMCs co-expressed α-SMA, vimentin and desmin suggesting that vimentin, which is not normally expressed by ASMCs, may play a role in airway thickening, and remodeling. ASMCs had strong cytoplasmic expression of TGFβ-1, which is known to contribute to tissue remodeling in asthma and in various bronchial and interstitial lung diseases, suggesting its involvement in the pathogenesis of ASMCs hyperplasia. Our findings provide histologic evidence of airway remodeling in cats. Further studies on larger caseloads are needed to support our conclusions on the value of this feline condition as an animal model for nonspecific airway remodeling in humans.
Collapse
Affiliation(s)
- Giulia D'Annunzio
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy.
| | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Giancarlo Avallone
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Barbara Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| |
Collapse
|
8
|
Role of Airway Smooth Muscle in Inflammation Related to Asthma and COPD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:139-172. [PMID: 33788192 DOI: 10.1007/978-3-030-63046-1_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Airway smooth muscle contributes to both contractility and inflammation in the pathophysiology of asthma and COPD. Airway smooth muscle cells can change the degree of a variety of functions, including contraction, proliferation, migration, and the secretion of inflammatory mediators (phenotype plasticity). Airflow limitation, airway hyperresponsiveness, β2-adrenergic desensitization, and airway remodeling, which are fundamental characteristic features of these diseases, are caused by phenotype changes in airway smooth muscle cells. Alterations between contractile and hyper-contractile, synthetic/proliferative phenotypes result from Ca2+ dynamics and Ca2+ sensitization. Modulation of Ca2+ dynamics through the large-conductance Ca2+-activated K+ channel/L-type voltage-dependent Ca2+ channel linkage and of Ca2+ sensitization through the RhoA/Rho-kinase pathway contributes not only to alterations in the contractile phenotype involved in airflow limitation, airway hyperresponsiveness, and β2-adrenergic desensitization but also to alteration of the synthetic/proliferative phenotype involved in airway remodeling. These Ca2+ signal pathways are also associated with synergistic effects due to allosteric modulation between β2-adrenergic agonists and muscarinic antagonists. Therefore, airway smooth muscle may be a target tissue in the therapy for these diseases. Moreover, the phenotype changing in airway smooth muscle cells with focuses on Ca2+ signaling may provide novel strategies for research and development of effective remedies against both bronchoconstriction and inflammation.
Collapse
|
9
|
van den Bosch WB, James AL, Tiddens HA. Structure and function of small airways in asthma patients revisited. Eur Respir Rev 2021; 30:200186. [PMID: 33472958 PMCID: PMC9488985 DOI: 10.1183/16000617.0186-2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
Small airways (<2 mm in diameter) are probably involved across almost all asthma severities and they show proportionally more structural and functional abnormalities with increasing asthma severity. The structural and functional alterations of the epithelium, extracellular matrix and airway smooth muscle in small airways of people with asthma have been described over many years using in vitro studies, animal models or imaging and modelling methods. The purpose of this review was to provide an overview of these observations and to outline several potential pathophysiological mechanisms regarding the role of small airways in asthma.
Collapse
Affiliation(s)
- Wytse B. van den Bosch
- Dept of Paediatric Pulmonology and Allergology, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Dept of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alan L. James
- Dept of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Harm A.W.M. Tiddens
- Dept of Paediatric Pulmonology and Allergology, Erasmus MC – Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Dept of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Chung E, Ojiaku CA, Cao G, Parikh V, Deeney B, Xu S, Wang S, Panettieri RA, Koziol-White C. Dexamethasone rescues TGF-β1-mediated β 2-adrenergic receptor dysfunction and attenuates phosphodiesterase 4D expression in human airway smooth muscle cells. Respir Res 2020; 21:256. [PMID: 33032603 PMCID: PMC7545943 DOI: 10.1186/s12931-020-01522-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/23/2020] [Indexed: 01/05/2023] Open
Abstract
Glucocorticoids (GCs) and β2-adrenergic receptor (β2AR) agonists improve asthma outcomes in most patients. GCs also modulate gene expression in human airway smooth muscle (HASM), thereby attenuating airway inflammation and airway hyperresponsiveness that define asthma. Our previous studies showed that the pro-fibrotic cytokine, transforming growth factor- β1 (TGF-β1) increases phosphodiesterase 4D (PDE4D) expression that attenuates agonist-induced levels of intracellular cAMP. Decreased cAMP levels then diminishes β2 agonist-induced airway relaxation. In the current study, we investigated whether glucocorticoids reverse TGF-β1-effects on β2-agonist-induced bronchodilation and modulate pde4d gene expression in HASM. Dexamethasone (DEX) reversed TGF-β1 effects on cAMP levels induced by isoproterenol (ISO). TGF-β1 also attenuated G protein-dependent responses to cholera toxin (CTX), a Gαs stimulator downstream from the β2AR receptor. Previously, we demonstrated that TGF-β1 treatment increased β2AR phosphorylation to induce hyporesponsiveness to a β2 agonist. Our current data shows that expression of grk2/3, kinases associated with attenuation of β2AR function, are not altered with TGF-β1 stimulation. Interestingly, DEX also attenuated TGF-β1-induced pde4d gene expression. These data suggest that steroids may be an effective therapy for treatment of asthma patients whose disease is primarily driven by elevated TGF-β1 levels.
Collapse
Affiliation(s)
- Elena Chung
- Department of Pharmacology and Toxicology, School of Pharmacy, EOHSI, Rutgers University, Piscataway, NJ, USA
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Christie A Ojiaku
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Vishal Parikh
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Brian Deeney
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Shengjie Xu
- Department of Pharmacology and Toxicology, School of Pharmacy, EOHSI, Rutgers University, Piscataway, NJ, USA
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Serena Wang
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
12
|
Gazzola M, Flamand N, Bossé Y. [Extracellular molecules controlling the contraction of airway smooth muscle and their potential contribution to bronchial hyperresponsiveness]. Rev Mal Respir 2020; 37:462-473. [PMID: 32487422 DOI: 10.1016/j.rmr.2020.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION A significant portion of symptoms in some lung diseases results from an excessive constriction of airways due to the contraction of smooth muscle and bronchial hyperresponsiveness. A better understanding of the extracellular molecules that control smooth muscle contractility is necessary to identify the underlying causes of the problem. STATE OF KNOWLEDGE Almost a hundred molecules, some of which newly identified, influence the contractility of airway smooth muscle. While some molecules activate the contraction, others activate the relaxation, thus acting directly as bronchoconstrictors and bronchodilators, respectively. Other molecules do not affect contraction directly but rather influence it indirectly by modifying the effect of bronchoconstrictors and bronchodilators. These are called bronchomodulators. Some of these bronchomodulators increase the contractile effect of bronchoconstrictors and could thus contribute to bronchial hyperresponsiveness. PROSPECTS Considering the high number of molecules potentially involved, as well as the level of functional overlap between some of them, identifying the extracellular molecules responsible for excessive airway constriction in a patient is a major contemporary challenge.
Collapse
Affiliation(s)
| | | | - Y Bossé
- Université Laval, Québec, Canada.
| |
Collapse
|
13
|
Regulation of Airway Smooth Muscle Contraction in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:381-422. [PMID: 31183836 DOI: 10.1007/978-981-13-5895-1_16] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Airway smooth muscle (ASM) extends from the trachea throughout the bronchial tree to the terminal bronchioles. In utero, spontaneous phasic contraction of fetal ASM is critical for normal lung development by regulating intraluminal fluid movement, ASM differentiation, and release of key growth factors. In contrast, phasic contraction appears to be absent in the adult lung, and regulation of tonic contraction and airflow is under neuronal and humoral control. Accumulating evidence suggests that changes in ASM responsiveness contribute to the pathophysiology of lung diseases with lifelong health impacts.Functional assessments of fetal and adult ASM and airways have defined pharmacological responses and signaling pathways that drive airway contraction and relaxation. Studies using precision-cut lung slices, in which contraction of intrapulmonary airways and ASM calcium signaling can be assessed simultaneously in situ, have been particularly informative. These combined approaches have defined the relative importance of calcium entry into ASM and calcium release from intracellular stores as drivers of spontaneous phasic contraction in utero and excitation-contraction coupling.Increased contractility of ASM in asthma contributes to airway hyperresponsiveness. Studies using animal models and human ASM and airways have characterized inflammatory and other mechanisms underlying increased reactivity to contractile agonists and reduced bronchodilator efficacy of β2-adrenoceptor agonists in severe diseases. Novel bronchodilators and the application of bronchial thermoplasty to ablate increased ASM within asthmatic airways have the potential to overcome limitations of current therapies. These approaches may directly limit excessive airway contraction to improve outcomes for difficult-to-control asthma and other chronic lung diseases.
Collapse
|
14
|
TRPC channels mediated calcium entry is required for proliferation of human airway smooth muscle cells induced by nicotine-nAChR. Biochimie 2019; 158:139-148. [DOI: 10.1016/j.biochi.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/09/2018] [Indexed: 01/06/2023]
|
15
|
Kadowaki M, Yamada H, Sato K, Shigemi H, Umeda Y, Morikawa M, Waseda Y, Anzai M, Kamide Y, Aoki-Saito H, Hisada T, Okajima F, Ishizuka T. Extracellular acidification-induced CXCL8 production through a proton-sensing receptor OGR1 in human airway smooth muscle cells: a response inhibited by dexamethasone. JOURNAL OF INFLAMMATION-LONDON 2019; 16:4. [PMID: 30828266 PMCID: PMC6381743 DOI: 10.1186/s12950-019-0207-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/27/2019] [Indexed: 12/13/2022]
Abstract
Background Human airway smooth muscle cells (ASMCs) contribute to bronchial contraction and airway hyperresponsiveness in patients with bronchial asthma. They also generate cytokines, chemokines, and matricellular proteins. Ovarian cancer G protein-coupled receptor 1 (OGR1) senses extracellular protons and mediates the production of interleukin-6 (IL-6) and connective tissue growth factor (CTGF) in ASMCs. Methods ASMCs were stimulated for the indicated time by pH 6.3 or pH 7.4-adjusted Dulbecco’s Modified Eagle Medium (DMEM) containing 0.1% bovine serum albumin (BSA) (0.1% BSA-DMEM). As a control stimulant, pH 7.4-adjusted 0.1% BSA-DMEM containing 10 ng/mL tumor necrosis factor-α (TNF-α) was used. Interleukin-8/C-X-C motif chemokine ligand 8 (CXCL8) mRNA expression in ASMCs was quantified by RT-PCR using real-time TaqMan technology. CXCL8 secreted from ASMCs was measured by enzyme-linked immunosorbent assay (ELISA). Phosphorylation at serine 536 of NF-κB p65 and binding of p65 to oligonucleotide containing an NF-κB consensus binding site were analyzed by Western blotting and an ELISA-based kit. Results Acidic pH induced a significant increase of CXCL8 mRNA expression and CXCL8 protein secretion in ASMCs. ASMCs transfected with small interfering RNA (siRNA) targeted for OGR1 produced less CXCL8 compared with those transfected with non-targeting siRNA. Protein kinase C (PKC) inhibitor, MEK1/2 inhibitor, and the inhibitor of IκB phosphorylation reduced acidic pH-stimulated CXCL8 production in ASMCs. Dexamethasone also inhibited acidic pH-stimulated CXCL8 production of ASMCs in a dose-dependent manner. Dexamethasone did not affect either phosphorylation or binding to the consensus DNA site of NF-κB p65. Conclusions CXCL8 released from ASMCs by extracellular acidification may play a pivotal role in airway accumulation of neutrophils. Glucocorticoids inhibit acidic pH-stimulated CXCL8 production independent of serine 536 phosphorylation and the binding to DNA of NF-κB p65, although NF-κB activity is essential for CXCL8 production in ASMCs.
Collapse
Affiliation(s)
- Maiko Kadowaki
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Hidenori Yamada
- 2Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebeshi, 371-8511 Japan
| | - Koichi Sato
- 3Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebeshi, 371-8511 Japan
| | - Hiroko Shigemi
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Yukihiro Umeda
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Miwa Morikawa
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Yuko Waseda
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Masaki Anzai
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| | - Yosuke Kamide
- 2Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebeshi, 371-8511 Japan
| | - Haruka Aoki-Saito
- 2Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebeshi, 371-8511 Japan
| | - Takeshi Hisada
- 2Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebeshi, 371-8511 Japan
| | - Fumikazu Okajima
- 4Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, 2-3-1 Kobata, Aomori, 030-0943 Japan
| | - Tamotsu Ishizuka
- 1Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka-Shimoaizuki, Eiheiji, Fukui, 910-1193 Japan
| |
Collapse
|
16
|
Koziol-White CJ, Panettieri RA. Modulation of Bronchomotor Tone Pathways in Airway Smooth Muscle Function and Bronchomotor Tone in Asthma. Clin Chest Med 2018; 40:51-57. [PMID: 30691716 DOI: 10.1016/j.ccm.2018.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Airway smooth muscle is the primary cell mediating bronchomotor tone. The milieu created in the asthmatic lung modulates airway smooth muscle contractility and relaxation. Experimental findings suggest intrinsic abnormalities in airway smooth muscle derived from patients with asthma in comparison with airway smooth muscle from those without asthma. These changes to excitation-contraction pathways may underlie airway hyperresponsiveness and increased airway resistance associated with asthma.
Collapse
Affiliation(s)
- Cynthia J Koziol-White
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers Institute for Translational Medicine and Science, Rutgers University, State University of New Jersey, 89 French Street, Suite 4268, New Brunswick, NJ 08901, USA.
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, State University of New Jersey, 89 French Street, Room 4210, New Brunswick, NJ 08901, USA
| |
Collapse
|
17
|
Kaczmarek KA, Clifford RL, Knox AJ. Epigenetic Changes in Airway Smooth Muscle as a Driver of Airway Inflammation and Remodeling in Asthma. Chest 2018; 155:816-824. [PMID: 30414795 DOI: 10.1016/j.chest.2018.10.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/10/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Epigenetic changes are heritable changes in gene expression, without changing the DNA sequence. Epigenetic processes provide a critical link between environmental insults to the airway and functional changes that determine how airway cells respond to future stimuli. There are three primary epigenetic processes: histone modifications, DNA modification, and noncoding RNAs. Airway smooth muscle has several important roles in the development and maintenance of the pathologic processes occurring in asthma, including inflammation, remodeling, and contraction/hyperresponsiveness. In this review, we describe the evidence for the role of epigenetic changes in driving these processes in airway smooth muscle cells in asthma, with a particular focus on histone modifications. We also discuss how existing therapies may target some of these changes and how epigenetic processes provide targets for the development of novel asthma therapeutics. Epigenetic marks may also provide a biomarker to assess phenotype and treatment responses.
Collapse
Affiliation(s)
- Klaudia A Kaczmarek
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node
| | - Rachel L Clifford
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node
| | - Alan J Knox
- Division of Respiratory Medicine, Nottingham University Hospitals NHS Trust (City Hospital Campus); and the Nottingham NIHR Biomedical Research Centre, Nottingham MRC Molecular Pathology Node.
| |
Collapse
|
18
|
Yan F, Gao H, Zhao H, Bhatia M, Zeng Y. Roles of airway smooth muscle dysfunction in chronic obstructive pulmonary disease. J Transl Med 2018; 16:262. [PMID: 30257694 PMCID: PMC6158847 DOI: 10.1186/s12967-018-1635-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/16/2018] [Indexed: 12/24/2022] Open
Abstract
The airway smooth muscle (ASM) plays an indispensable role in airway structure and function. Dysfunction in ASM plays a central role in the pathogenesis of chronic obstructive pulmonary disease (COPD) and contributes to alterations of contractility, inflammatory response, immunoreaction, phenotype, quantity, and size of airways. ASM makes a key contribution in COPD by various mechanisms including altered contractility and relaxation induce by [Ca2+]i, cell proliferation and hypertrophy, production and modulation of extracellular cytokines, and release of pro-and-anti-inflammatory mediators. Multiple dysfunctions of ASM contribute to modulating airway responses to stimuli, remodeling, and fibrosis, as well as influence the compliance of lungs. The present review highlights regulatory roles of multiple factors in the development of ASM dysfunction in COPD, aims to understand the regulatory mechanism by which ASM dysfunctions are initiated, and explores the clinical significance of ASM on alterations of airway structure and function in COPD and development of novel therapeutic strategies for COPD.
Collapse
Affiliation(s)
- Furong Yan
- Center for Molecular Diagnosis and Therapy, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hongzhi Gao
- Center for Molecular Diagnosis and Therapy, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hong Zhao
- Center for Molecular Diagnosis and Therapy, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
19
|
Huo Y, Xu J, Guan L, Wu W, Guo B, Yang Y, Lin L, Ou Y, Jiang F, Zhou L, Chen R. Methacholine induces extracellular matrix production by human airway smooth muscle cells through β-catenin signaling. Respir Physiol Neurobiol 2018; 254:55-63. [PMID: 29715518 DOI: 10.1016/j.resp.2018.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 04/21/2018] [Accepted: 04/28/2018] [Indexed: 11/28/2022]
Abstract
Altered extracellular matrix (ECM) production by airway smooth muscle cells (ASMCs) is an important feature of airway remodeling. Muscarinic receptor agonists contribute to ECM production in vivo, but the mechanisms involved remain unclear. This study attempted to investigate the role of methacholine in promoting ECM production by human ASMCs (HASMCs) and the underlying mechanism. We found that methacholine induced the expression of collagen I protein and multiple ECM genes. β-catenin signaling was activated in this process upon GSK3β phosphorylation, leading to upregulation of total and active β-catenin. Silencing β-catenin by specific small interfering RNA (siRNA) or with the β-catenin inhibitor, PKF115-584, decreased collagen I expression. Conversely, overexpression of active β-catenin by adenoviruses carrying the S33Y-β-catenin mutant increased the methacholine-induced collagen I expression. Furthermore, methacholine induced TGF-β expression in HASMCs, while pan-TGF-β-neutralizing antibody only partially decreased collagen I expression. These findings suggest that methacholine induced ECM production through β-catenin signaling and partially through TGF-β.
Collapse
Affiliation(s)
- Yating Huo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Jiawen Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Lili Guan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Weiliang Wu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Bingpeng Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Yuqiong Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Lin Lin
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Yonger Ou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Fangfang Jiang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Luqian Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| | - Rongchang Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Rd., Guangzhou 510120, China.
| |
Collapse
|
20
|
Li X, Michaeloudes C, Zhang Y, Wiegman CH, Adcock IM, Lian Q, Mak JCW, Bhavsar PK, Chung KF. Mesenchymal stem cells alleviate oxidative stress-induced mitochondrial dysfunction in the airways. J Allergy Clin Immunol 2018; 141:1634-1645.e5. [PMID: 28911970 DOI: 10.1016/j.jaci.2017.08.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 07/07/2017] [Accepted: 08/23/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Oxidative stress-induced mitochondrial dysfunction can contribute to inflammation and remodeling in patients with chronic obstructive pulmonary disease (COPD). Mesenchymal stem cells protect against lung damage in animal models of COPD. It is unknown whether these effects occur through attenuating mitochondrial dysfunction in airway cells. OBJECTIVE We sought to examine the effect of induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) on oxidative stress-induce mitochondrial dysfunction in human airway smooth muscle cells (ASMCs) in vitro and in mouse lungs in vivo. METHODS ASMCs were cocultured with iPSC-MSCs in the presence of cigarette smoke medium (CSM), and mitochondrial reactive oxygen species (ROS) levels, mitochondrial membrane potential (ΔΨm), and apoptosis were measured. Conditioned medium from iPSC-MSCs and transwell cocultures were used to detect any paracrine effects. The effect of systemic injection of iPSC-MSCs on airway inflammation and hyperresponsiveness in ozone-exposed mice was also investigated. RESULTS Coculture of iPSC-MSCs with ASMCs attenuated CSM-induced mitochondrial ROS, apoptosis, and ΔΨm loss in ASMCs. iPSC-MSC-conditioned medium or transwell cocultures with iPSC-MSCs reduced CSM-induced mitochondrial ROS but not ΔΨm or apoptosis in ASMCs. Mitochondrial transfer from iPSC-MSCs to ASMCs was observed after direct coculture and was enhanced by CSM. iPSC-MSCs attenuated ozone-induced mitochondrial dysfunction, airway hyperresponsiveness, and inflammation in mouse lungs. CONCLUSION iPSC-MSCs offered protection against oxidative stress-induced mitochondrial dysfunction in human ASMCs and in mouse lungs while reducing airway inflammation and hyperresponsiveness. These effects are, at least in part, dependent on cell-cell contact, which allows for mitochondrial transfer, and paracrine regulation. Therefore iPSC-MSCs show promise as a therapy for oxidative stress-dependent lung diseases, such as COPD.
Collapse
Affiliation(s)
- Xiang Li
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Department of Medicine, University of Hong Kong, Hong Kong, China
| | | | - Yuelin Zhang
- Department of Medicine, University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Coen H Wiegman
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Qizhou Lian
- Department of Medicine, University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, University of Hong Kong, Hong Kong, China.
| | - Judith C W Mak
- Department of Medicine, University of Hong Kong, Hong Kong, China; Department of Pharmacology & Pharmacy, University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, University of Hong Kong, Hong Kong, China.
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Liu G, Cooley MA, Nair PM, Donovan C, Hsu AC, Jarnicki AG, Haw TJ, Hansbro NG, Ge Q, Brown AC, Tay H, Foster PS, Wark PA, Horvat JC, Bourke JE, Grainge CL, Argraves WS, Oliver BG, Knight DA, Burgess JK, Hansbro PM. Airway remodelling and inflammation in asthma are dependent on the extracellular matrix protein fibulin-1c. J Pathol 2017; 243:510-523. [PMID: 28862768 DOI: 10.1002/path.4979] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/08/2023]
Abstract
Asthma is a chronic inflammatory disease of the airways. It is characterized by allergic airway inflammation, airway remodelling, and airway hyperresponsiveness (AHR). Asthma patients, in particular those with chronic or severe asthma, have airway remodelling that is associated with the accumulation of extracellular matrix (ECM) proteins, such as collagens. Fibulin-1 (Fbln1) is an important ECM protein that stabilizes collagen and other ECM proteins. The level of Fbln1c, one of the four Fbln1 variants, which predominates in both humans and mice, is increased in the serum and airways fluids in asthma but its function is unclear. We show that the level of Fbln1c was increased in the lungs of mice with house dust mite (HDM)-induced chronic allergic airway disease (AAD). Genetic deletion of Fbln1c and therapeutic inhibition of Fbln1c in mice with chronic AAD reduced airway collagen deposition, and protected against AHR. Fbln1c-deficient (Fbln1c-/- ) mice had reduced mucin (MUC) 5 AC levels, but not MUC5B levels, in the airways as compared with wild-type (WT) mice. Fbln1c interacted with fibronectin and periostin that was linked to collagen deposition around the small airways. Fbln1c-/- mice with AAD also had reduced numbers of α-smooth muscle actin-positive cells around the airways and reduced airway contractility as compared with WT mice. After HDM challenge, these mice also had fewer airway inflammatory cells, reduced interleukin (IL)-5, IL-13, IL-33, tumour necrosis factor (TNF) and CXCL1 levels in the lungs, and reduced IL-5, IL-33 and TNF levels in lung-draining lymph nodes. Therapeutic targeting of Fbln1c reduced the numbers of GATA3-positive Th2 cells in the lymph nodes and lungs after chronic HDM challenge. Treatment also reduced the secretion of IL-5 and IL-13 from co-cultured dendritic cells and T cells restimulated with HDM extract. Human epithelial cells cultured with Fbln1c peptide produced more CXCL1 mRNA than medium-treated controls. Our data show that Fbln1c may be a therapeutic target in chronic asthma. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Marion A Cooley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrew G Jarnicki
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Qi Ge
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Hock Tay
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - W Scott Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| | - Janette K Burgess
- Woolcock Institute of Medical Research, Discipline of Pharmacology, University of Sydney, Sydney, New South Wales, Australia.,University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen Research Institute of Asthma and COPD, Groningen, The Netherlands
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
22
|
Wang W, Qu X, Dang X, Shang D, Yang L, Li Y, Xu D, Martin JG, Hamid Q, Liu J, Chang Y. Human β-defensin-3 induces IL-8 release and apoptosis in airway smooth muscle cells. Clin Exp Allergy 2017; 47:1138-1149. [PMID: 28437599 DOI: 10.1111/cea.12943] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Human airway smooth muscle cells (ASMCs) may have a pro-inflammatory role through the release of inflammatory mediators. Increasing evidence indicates that human β-defensins (HBDs) are related to pathogenesis of asthma. OBJECTIVES To examine the plasma level of HBD-1, HBD-2 and HBD-3 in asthmatic patients and the expression of their mouse orthologues in the lung tissue of a mouse model of chronic severe asthma. Further to investigate the effect of HBD-3 on the release of the pro-inflammatory cytokine IL-8 and to explore the mechanisms. METHODS The plasma levels of HBD-1, HBD-2 and HBD-3 from 34 healthy controls and 25 asthmatic patients were determined by ELISA. The expression of mouse β-defensins MBD-1, MBD-3 and MBD-14 in the lung tissue of asthmatic mice was detected by Western blot. The ASMCs were cultured with HBD-3 for 24 hour, and then the supernatant level of IL-8 was evaluated by ELISA and the cell viability was examined by WST-1 assay. The signalling pathway was investigated with blocking antibodies or pharmacological inhibitors. RESULTS The plasma levels of HBD-1 and HBD-3 were elevated in asthmatic patients, and the expression of MBD-14, the mouse orthologue for HBD-3, was increased in asthmatic mice. HBD-3-induced IL-8 production in a CCR6 receptor-specific manner and was dependent on multiple signalling pathways. Moreover, HBD-3-induced cell apoptosis concurrently, which was dependent on the ERK1/2 MAPK pathway. Mitochondrial ROS regulated both HBD-3-induced IL-8 production and cell apoptosis. CONCLUSIONS AND CLINICAL RELEVANCE These observations provide clear evidence of an important new mechanism for the promotion of airway inflammation and tissue remodelling with potential relevance for the treatment of asthma.
Collapse
Affiliation(s)
- W Wang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - X Qu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - X Dang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - D Shang
- Department of Respiration, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - L Yang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Li
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - D Xu
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - J G Martin
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Q Hamid
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, Quebec, Canada.,College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - J Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Y Chang
- Center for Translational Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Prakash YS. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1113-L1140. [PMID: 27742732 DOI: 10.1152/ajplung.00370.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Airway structure and function are key aspects of normal lung development, growth, and aging, as well as of lung responses to the environment and the pathophysiology of important diseases such as asthma, chronic obstructive pulmonary disease, and fibrosis. In this regard, the contributions of airway smooth muscle (ASM) are both functional, in the context of airway contractility and relaxation, as well as synthetic, involving production and modulation of extracellular components, modulation of the local immune environment, cellular contribution to airway structure, and, finally, interactions with other airway cell types such as epithelium, fibroblasts, and nerves. These ASM contributions are now found to be critical in airway hyperresponsiveness and remodeling that occur in lung diseases. This review emphasizes established and recent discoveries that underline the central role of ASM and sets the stage for future research toward understanding how ASM plays a central role by being both upstream and downstream in the many interactive processes that determine airway structure and function in health and disease.
Collapse
Affiliation(s)
- Y S Prakash
- Departments of Anesthesiology, and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
24
|
Koga Y, Hisada T, Ishizuka T, Utsugi M, Ono A, Yatomi M, Kamide Y, Aoki-Saito H, Tsurumaki H, Dobashi K, Yamada M. CREB regulates TNF-α-induced GM-CSF secretion via p38 MAPK in human lung fibroblasts. Allergol Int 2016; 65:406-413. [PMID: 27118435 DOI: 10.1016/j.alit.2016.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/13/2016] [Accepted: 03/15/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that mediates eosinophilic differentiation, migration and survival, causing respiratory tract inflammation. GM-CSF is also known to be secreted from respiratory tract structural cells. However, the mechanisms of GM-CSF secretion have not been well established. METHODS Human fetal lung fibroblasts and human primary asthmatic lung fibroblasts were used for the study of tumor necrosis factor alpha (TNF-α)-induced GM-CSF secretion. GM-CSF secretion and mRNA expression were measured by enzyme-linked immunosorbent assay and quantitative real-time reverse transcription polymerase chain reaction, respectively. Knockdown of cAMP response element-binding protein (CREB) in fibroblasts was carried out by using specific small interfering RNAs of CREB. RESULTS Among respiratory tract structural cells, pulmonary fibroblasts exhibited increased GM-CSF secretion and mRNA expression after stimulation with TNF-α in a concentration-dependent manner. Moreover, a p38 mitogen-activated protein kinase (MAPK) inhibitor controlled TNF-α-induced GM-CSF secretion, and roflumilast and rolipram, inhibitors of phosphodiesterase-4, suppressed TNF-α-induced GM-CSF secretion. Consistent with this, forskolin also completely blocked GM-CSF secretion, and similar results were observed in response to cAMP treatment, suggesting that cAMP signaling suppressed TNF-α-induced GM-CSF secretion in human lung fibroblasts. Furthermore, CREB was phosphorylated through p38 MAPK but not cAMP signaling after TNF-α stimulation, and GM-CSF secretion was inhibited by CREB knockdown. Finally, these effects were also demonstrated in human primary lung fibroblasts in a patient with asthma. CONCLUSIONS CREB signaled independent of cAMP signaling and was phosphorylated by p38 MAPK following TNF-α stimulation, playing a critical role in GM-CSF secretion in human lung fibroblasts.
Collapse
Affiliation(s)
- Yasuhiko Koga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan.
| | - Takeshi Hisada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Mitsuyoshi Utsugi
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan; Department of Respiratory Medicine, Kiryu Kosei General Hospital, Gunma, Japan
| | - Akihiro Ono
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masakiyo Yatomi
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yosuke Kamide
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan; Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, Kanagawa, Japan
| | - Haruka Aoki-Saito
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroaki Tsurumaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kunio Dobashi
- Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
25
|
Airway remodeling associated with cough hypersensitivity as a consequence of persistent cough: An experimental study. Respir Investig 2016; 54:419-427. [PMID: 27886853 DOI: 10.1016/j.resinv.2016.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 05/31/2016] [Accepted: 06/27/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Chronic cough involves airway remodeling associated with cough reflex hypersensitivity. Whether cough itself induces these features remains unknown. METHODS Guinea pigs were assigned to receive treatment with citric acid (CA), saline (SA), or CA+dextromethorphan (DEX). All animals were exposed to 0.5M CA on days 1 and 22. On days 4-20, the CA and CA+DEX groups were exposed to CA, and the SA group to saline thrice weekly, during which the CA+DEX group was administered DEX pretreatment to inhibit cough. The number of coughs was counted during each 10-min CA or SA exposure. Terbutaline premedication was started to prevent bronchoconstriction. Bronchoalveolar lavage and pathology were examined on day 25. Average cough number for 10 CA exposures was examined as "cough index" in the CA group, which was divided into frequent (cough index>5) and infrequent (<5) cough subgroups for lavage and pathology analysis. RESULTS The number of coughs significantly increased in the CA group from day 13 onwards. In the CA+DEX and SA groups, the number of coughs did not differ between days 1 and 22, while average number of coughs during days 4-20 was significantly lower than at days 1 and 22. Bronchoalveolar cell profiles were similar among the four groups. The smooth muscle area of small airways was significantly greater in the frequent-cough subgroup than in the other groups (in which it was similar), and highly correlated with cough index in CA group. CONCLUSION Repeated cough induces airway smooth muscle remodeling associated with cough reflex hypersensitivity.
Collapse
|
26
|
Aravamudan B, Thompson MA, Pabelick CM, Prakash YS. Mechanisms of BDNF regulation in asthmatic airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2016; 311:L270-9. [PMID: 27317689 DOI: 10.1152/ajplung.00414.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/09/2016] [Indexed: 12/17/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a neurotrophin produced by airway smooth muscle (ASM), enhances inflammation effects on airway contractility, supporting the idea that locally produced growth factors influence airway diseases such as asthma. We endeavored to dissect intrinsic mechanisms regulating endogenous, as well as inflammation (TNF-α)-induced BDNF secretion in ASM of nonasthmatic vs. asthmatic humans. We focused on specific Ca(2+) regulation- and inflammation-related signaling cascades and quantified BDNF secretion. We find that TNF-α enhances BDNF release by ASM cells, via several mechanisms relevant to asthma, including transient receptor potential channels TRPC3 and TRPC6 (but not TRPC1), ERK 1/2, PI3K, PLC, and PKC cascades, Rho kinase, and transcription factors cAMP response element binding protein and nuclear factor of activated T cells. Basal BDNF expression and secretion are elevated in asthmatic ASM and increase further with TNF-α exposure, involving many of these regulatory mechanisms. We conclude that airway BDNF secretion is regulated at multiple levels, providing a basis for autocrine effects of BDNF under conditions of inflammation and disease, with potential downstream influences on contractility and remodeling.
Collapse
Affiliation(s)
| | | | - Christina M Pabelick
- Departments of Anesthesiology, Mayo Clinic, Rochester, Minnesota; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- Departments of Anesthesiology, Mayo Clinic, Rochester, Minnesota; and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
LIN XIAOLING, YANG CHENG, HUANG LINJIE, CHEN MING, SHI JIANTING, OUYANG LIHUA, TANG TIANTIAN, ZHANG WEI, LI YIQUN, LIANG RUIYUN, JIANG SHANPING. Upregulation of TRPM7 augments cell proliferation and interleukin-8 release in airway smooth muscle cells of rats exposed to cigarette smoke. Mol Med Rep 2016; 13:4995-5004. [PMID: 27108806 PMCID: PMC4878570 DOI: 10.3892/mmr.2016.5161] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 03/10/2016] [Indexed: 12/26/2022] Open
Abstract
Proliferation and synthetic function (i.e. the capacity to release numerous chemokines and cytokines) of airway smooth muscle cells (ASMCs) are important in airway remodeling induced by cigarette smoke exposure. However, the molecular mechanism has not been clarified. Transient receptor potential cation channel subfamily M member 7 (TRPM7) is expressed ubiquitously and is crucial for the cellular physiological function of many cell types. The present study aimed to detect the expression of TRPM7 in ASMCs from smoke‑exposed rats and determine the importance of TRPM7 in proliferation and interleukin‑8 (IL‑8) release. ASMCs were isolated and cultured from smoke‑exposed rats. Expression levels of TRPM7 were determined by reverse transcription‑polymerase chain reaction, western blot analysis and immunofluorescence. TRPM7 was silenced with TRPM7‑short hairpin RNA lentivirus vector. DNA synthesis, cell number and IL‑8 release of ASMCs induced by cigarette smoke extract (CSE) and tumor necrosis factor‑α (TNF‑α) were assessed using [3H]-thymidine incorporation assay, hemocytometer and enzyme‑linked immunosorbent assay, respectively. It was determined that mRNA and protein expression levels of TRPM7 were increased in ASMCs from smoke‑exposed rats. Stimulation with CSE or TNF‑α elevated DNA synthesis, cell number and IL‑8 release were more marked in ASMCs from smoke‑exposed rats. Silencing of TRPM7 reduced DNA synthesis, cell number and IL‑8 release induced by CSE or TNF‑α in ASMCs from smoke-exposed rats. In conclusion, expression of TRPM7 increased significantly in ASMCs from smoke‑exposed rats and the upregulation of TRPM7 led to augmented cell proliferation and IL-8 release in ASMCs from rats exposed to cigarette smoke.
Collapse
Affiliation(s)
- XIAOLING LIN
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - CHENG YANG
- Department of Respiratory Medicine, Meizhou People's Hospital, Meizhou Affiliated Hospital of Sun Yat-Sen University, Meizhou, Guangdong 514031, P.R. China
| | - LINJIE HUANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - MING CHEN
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - JIANTING SHI
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - LIHUA OUYANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - TIANTIAN TANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - WEI ZHANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - YIQUN LI
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - RUIYUN LIANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - SHANPING JIANG
- Department of Respiratory Medicine, Sun Yat-Sen Memorial Hospital, Institute of Respiratory Disease, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
28
|
Su X, Ren Y, Yu N, Kong L, Kang J. Thymoquinone inhibits inflammation, neoangiogenesis and vascular remodeling in asthma mice. Int Immunopharmacol 2016; 38:70-80. [PMID: 27240137 DOI: 10.1016/j.intimp.2016.05.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023]
Abstract
Asthma is a chronic obstructive disease which is characterized by recurring airway inflammation, reversible airway obstruction, airway hyper responsiveness and vascular remodeling. Thymoquinone (TQ), an active ingredient isolated from Nigella sativa, was reported to exhibit anti-inflammation and anti-proliferation of in various cancer cells as well as epithelial cells. The aim of this study was to evaluate the effect of TQ on the inflammation, neoangiogenesis and vascular remodeling induced by Ovalbumin (OVA) in asthma mice in vivo and the anti-angiogenesis effects of TQ in VEGF-induced human umbilical vein endothelial cells (HUVECs) in vitro. Our results revealed that TQ inhibited the production of inflammatory factors interleukin-4/-5 (IL-4/-5) by enzyme-linked immunesorbent assay (ELISA). Immunohistochemistry analysis showed that the increase of platelet endothelial cell adhesion molecule-1, which is also known as CD31 and α-smooth muscle actinalpha (α-SMA) expression in asthma mice challenged by OVA was suppressed by TQ. Moreover, TQ suppressed the activation of VEGFR2-PI3K-Akt pathway and up-regulated the expression of Slit glycoprotein-2 (Slit-2) both in vivo and in vitro with the inhibition of tube information in HUVEC cells. Meanwhile immunofluorescence analysis showed that Slit-2 and Roundabout-4 (Robo-4) were co-expressing after TQ treatment in OVA-challenged asthma mice. Our study demonstrates that TQ attenuated the inflammatory reaction by antagonizing IL-4/-5 while the anti-neoangiogenesis effect of TQ is mediated by inhibition of vascular endothelial growth factor (VEGF) expression through VEGFR2/PI3K/Akt signaling pathway, which supports a potential role for TQ in ameliorating asthma.
Collapse
Affiliation(s)
- Xinming Su
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| | - Yuan Ren
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Na Yu
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Lingfei Kong
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Jian Kang
- Department of Respiratory Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
29
|
Martin N, Reid PT. The potential role of phosphodiesterase inhibitors in the management of asthma. ACTA ACUST UNITED AC 2016; 5:207-17. [PMID: 16696590 DOI: 10.2165/00151829-200605030-00006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Asthma is a chronic inflammatory condition characterised by reversible airflow obstruction and airway hyperreactivity. The course of the illness may be punctuated by exacerbations resulting in deterioration in quality of life and, in some cases, days lost from school or work. That asthma is common and increasingly prevalent magnifies the importance of any potential economic costs, and promoting asthma control represents an important public health agenda. While lifestyle changes represent a valuable contribution in some patients, the majority of asthmatic patients require therapeutic intervention. The recognition of the role of inflammation in the pathogenesis of asthma has led to an emphasis on regular anti-inflammatory therapy, of which inhaled corticosteroid treatment remains the most superior. In selected patients, further improvements in asthma control may be gained by the addition of regular inhaled long-acting beta(2)-adrenoceptor agonists or oral leukotriene receptor antagonists to inhaled corticosteroid therapy. However, a significant minority of patients with asthma remain poorly controlled despite appropriate treatment, suggesting that additional corticosteroid nonresponsive inflammatory pathways may be operative. Furthermore, some patients with asthma display an accelerated decline in lung function, suggesting that active airway re-modeling is occurring. Such observations have focused attention on the potential to develop new therapies which complement existing treatments by targeting additional inflammatory pathways. The central role of phosphodiesterase (PDE), and in particular the PDE4 enzyme, in the regulation of key inflammatory cells believed to be important in asthma - including eosinophils, lymphocytes, neutrophils and airway smooth muscle - suggests that drugs designed to target this enzyme will have the potential to deliver both bronchodilation and modulate the asthmatic inflammatory response. In vivo studies on individual inflammatory cells suggest that the effects are likely to be favorable in asthma, and animal study models have provided proof of concept; however, first-generation PDE inhibitors have been poorly tolerated due to adverse effects. The development of second-generation agents such as cilomilast and roflumilast heralds a further opportunity to test the potential of these agents, although to date only a limited amount of data from human studies has been published, making it difficult to draw firm conclusions.
Collapse
Affiliation(s)
- Neil Martin
- Respiratory Medicine Unit, Western General Hospital, Edinburgh, Scotland
| | | |
Collapse
|
30
|
Yang CH, Tsao CF, Ko WS, Chiou YL. The Oligo Fucoidan Inhibits Platelet-Derived Growth Factor-Stimulated Proliferation of Airway Smooth Muscle Cells. Mar Drugs 2016; 14:15. [PMID: 26761017 PMCID: PMC4728512 DOI: 10.3390/md14010015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/23/2015] [Accepted: 01/04/2016] [Indexed: 12/14/2022] Open
Abstract
In the pathogenesis of asthma, the proliferation of airway smooth muscle cells (ASMCs) is a key factor in airway remodeling and causes airway narrowing. In addition, ASMCs are also the effector cells of airway inflammation. Fucoidan extracted from marine brown algae polysaccharides has antiviral, antioxidant, antimicrobial, anticlotting, and anticancer properties; however, its effectiveness for asthma has not been elucidated thus far. Platelet-derived growth factor (PDGF)-treated primary ASMCs were cultured with or without oligo-fucoidan (100, 500, or 1000 µg/mL) to evaluate its effects on cell proliferation, cell cycle, apoptosis, and Akt, ERK1/2 signaling pathway. We found that PDGF (40 ng/mL) increased the proliferation of ASMCs by 2.5-fold after 48 h (p < 0.05). Oligo-fucoidan reduced the proliferation of PDGF-stimulated ASMCs by 75%-99% after 48 h (p < 0.05) and induced G₁/G₀ cell cycle arrest, but did not induce apoptosis. Further, oligo-fucoidan supplementation reduced PDGF-stimulated extracellular signal-regulated kinase (ERK1/2), Akt, and nuclear factor (NF)-κB phosphorylation. Taken together, oligo-fucoidan supplementation might reduce proliferation of PDGF-treated ASMCs through the suppression of ERK1/2 and Akt phosphorylation and NF-κB activation. The results provide basis for future animal experiments and human trials.
Collapse
Affiliation(s)
- Chao-Huei Yang
- Department of Internal Medicine, Kuang-Tien General Hospital, No. 117, Shatian Road Shalu District, Taichung City 433, Taiwan.
| | - Chiung-Fang Tsao
- Department of Biotechnology, Hungkuang University, 34 Chung-Chie Rd, Sha Lu, Taichung 443, Taiwan.
| | - Wang-Sheng Ko
- Department of Internal Medicine, Kuang-Tien General Hospital, No. 117, Shatian Road Shalu District, Taichung City 433, Taiwan.
- Institute of BioMedical Nutrition, Hungkuang University, 34 Chung-Chie Rd, Sha Lu, Taichung 443, Taiwan.
| | - Ya-Ling Chiou
- Institute of BioMedical Nutrition, Hungkuang University, 34 Chung-Chie Rd, Sha Lu, Taichung 443, Taiwan.
- Department of Nursing, Hungkuang University, 34 Chung-Chie Rd, Sha Lu, Taichung 443, Taiwan.
| |
Collapse
|
31
|
Malykhin FТ, Kostornaya IV. [Morphological changes in the respiratory organs in chronic obstructive pulmonary disease]. Arkh Patol 2016; 78:42-50. [PMID: 27077144 DOI: 10.17116/patol201678142-50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The basis for airway remoldeling in patients with chronic obstructive pulmonary disease (COPD) is tissue changes contributing to thickening of the walls of the airway and its obstruction. As the disease becomes severer, there are increases in mucosal metaplasia, submucosal hypertrophy, peribronchial fibrosis, and airway smooth muscle mass. Drug therapy for COPD does not virtually lead to regression of airway obstruction, except when eosinophilia is present.
Collapse
Affiliation(s)
- F Т Malykhin
- Stavropol State Medical University Ministry of Health of Russia, Stavropol, Russia
| | - I V Kostornaya
- Stavropol State Medical University Ministry of Health of Russia, Stavropol, Russia
| |
Collapse
|
32
|
Kaphalia L, Kalita M, Kaphalia BS, Calhoun WJ. Effects of acute ethanol exposure on cytokine production by primary airway smooth muscle cells. Toxicol Appl Pharmacol 2015; 292:85-93. [PMID: 26721307 DOI: 10.1016/j.taap.2015.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 12/08/2015] [Accepted: 12/17/2015] [Indexed: 01/17/2023]
Abstract
Both chronic and binge alcohol abuse can be significant risk factors for inflammatory lung diseases such as acute respiratory distress syndrome and chronic obstructive pulmonary disease. However, metabolic basis of alcohol-related lung disease is not well defined, and may include key metabolites of ethanol [EtOH] in addition to EtOH itself. Therefore, we investigated the effects of EtOH, acetaldehyde [ACE], and fatty acid ethyl esters [FAEEs] on oxidative stress, endoplasmic reticulum (ER) stress, AMP-activated protein kinase (AMPK) signaling and nuclear translocation of phosphorylated (p)-NF-κB p65 in primary human airway smooth muscle (HASM) cells stimulated to produce cytokines using LPS exposure. Both FAEEs and ACE induced evidence of cellular oxidative stress and ER stress, and increased p-NF-κB in nuclear extracts. EtOH and its metabolites decreased p-AMPKα activation, and induced expression of fatty acid synthase, and decreased expression of sirtuin 1. In general, EtOH decreased secretion of IP-10, IL-6, eotaxin, GCSF, and MCP-1. However, FAEEs and ACE increased these cytokines, suggesting that both FAEEs and ACE as compared to EtOH itself are proinflammatory. A direct effect of EtOH could be consistent with blunted immune response. Collectively, these two features of EtOH exposure, coupled with the known inhibition of innate immune response in our model might explain some clinical manifestations of EtOH exposure in the lung.
Collapse
Affiliation(s)
- Lata Kaphalia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Mridul Kalita
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Bhupendra S Kaphalia
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - William J Calhoun
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
33
|
Tang DD. Critical role of actin-associated proteins in smooth muscle contraction, cell proliferation, airway hyperresponsiveness and airway remodeling. Respir Res 2015; 16:134. [PMID: 26517982 PMCID: PMC4628321 DOI: 10.1186/s12931-015-0296-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/22/2015] [Indexed: 01/16/2023] Open
Abstract
Asthma is characterized by airway hyperresponsiveness and airway remodeling, which are largely attributed to increased airway smooth muscle contractility and cell proliferation. It is known that both chemical and mechanical stimulation regulates smooth muscle contraction. Recent studies suggest that contractile activation and mechanical stretch induce actin cytoskeletal remodeling in smooth muscle. However, the mechanisms that control actin cytoskeletal reorganization are not completely elucidated. This review summarizes our current understanding regarding how actin-associated proteins may regulate remodeling of the actin cytoskeleton in airway smooth muscle. In particular, there is accumulating evidence to suggest that Abelson tyrosine kinase (Abl) plays a critical role in regulating airway smooth muscle contraction and cell proliferation in vitro, and airway hyperresponsiveness and remodeling in vivo. These studies indicate that Abl may be a novel target for the development of new therapy to treat asthma.
Collapse
Affiliation(s)
- Dale D Tang
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA.
| |
Collapse
|
34
|
Bi M, Guo A, Zhao H, Sun X, Chen Q, Yu L, Shi W, Wang Y, Shen G, Wang X, Zhao Y, Zhang N, Xu M, Qin M, Zhu W. Role of the extracellular signal-regulated kinase 1/2 signaling pathway in the process of thrombin-promoting airway remodeling in ovalbumin-allergic rats. Immunopharmacol Immunotoxicol 2015; 37:26-34. [PMID: 25519468 DOI: 10.3109/08923973.2014.993083] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Although it is recognized that thrombin plays a key role in airway remodeling during chronic asthma. In a previous study, we have proved that thrombin promotes airway remodeling via PAR-1 in OVA-allergic rats, but little is known about intracellular signaling pathway involved in the event. OBJECTIVE In this study, we intend to explore the impact of pERK1/2 signaling pathway on the process of thrombin-induced airway remodeling in OVA-allergic rats. MATERIALS AND METHODS A rat model of chronic asthma was set up by systemic sensitization and repeated challenge to OVA. The doses of thrombin, recombinant hirudin, PAR-1 inhibitor ER-112780-06, and pERK1/2 inhibitor PD98059 varied for different groups. The expression of pERK1/2 was analyzed by western blot and RT-PCR. Secretion of TGF-β1 and IL-6 was detected by ELISA. RESULTS The expression of pERK1/2 was higher in the airway of asthmatic rats than those of normal rats, and was significantly increased by thrombin treatment but decreased by thrombin-inhibitor treatment. Airway remodeling was enhanced by thrombin but weakened by pERK1/2 inhibitor. Expression of growth factors and IL-6 in asthmatic rats was significantly increased by thrombin treatment and decreased by thrombin-inhibitor treatment and pERK1/2 inhibitor treatment. CONCLUSION These results suggest that ERK1/2 signaling pathway may play an important role in the process of thrombin-promoting airway remodeling in OVA-allergic rats, and pERK1/2 inhibitor effectively inhibits the process.
Collapse
|
35
|
Sun XJ, Li XQ, Wang XL, Tan WF, Wang JK. Sevoflurane inhibits nuclear factor-κB activation in lipopolysaccharide-induced acute inflammatory lung injury via toll-like receptor 4 signaling. PLoS One 2015; 10:e0122752. [PMID: 25875290 PMCID: PMC4397052 DOI: 10.1371/journal.pone.0122752] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 02/13/2015] [Indexed: 11/24/2022] Open
Abstract
Background Infection is a common cause of acute lung injury (ALI). This study was aimed to explore whether Toll-like receptors 4 (TLR4) of airway smooth muscle cells (ASMCs) play a role in lipopolysaccharide (LPS)-induced airway hyperresponsiveness and potential mechanisms. Methods In vivo: A sensitizing dose of LPS (50 µg) was administered i.p. to female mice before anesthesia with either 3% sevoflurane or phenobarbital i.p. After stabilization, the mice were challenged with 5 µg of intratracheal LPS to mimic inflammatory attack. The effects of sevoflurane were assessed by measurement of airway responsiveness to methacholine, histological examination, and IL-1, IL-6, TNF-α levels in bronchoalveolar lavage fluid (BALF). Protein and gene expression of TLR4 and NF-κB were also assessed. In vitro: After pre-sensitization of ASMCs and ASM segments for 24h, levels of TLR4 and NF-κB proteins in cultured ASMCs were measured after continuous LPS exposure for 1, 3, 5, 12 and 24h in presence or absence of sevoflurane. Constrictor and relaxant responsiveness of ASM was measured 24 h afterwards. Results The mRNA and protein levels of NF-κB and TLR4 in ASM were increased and maintained at high level after LPS challenge throughout 24h observation period, both in vivo and in vitro. Sevoflurane reduced LPS-induced airway hyperresponsiveness, lung inflammatory cell infiltration and proinflammatory cytokines release in BALF as well as maximal isometric contractile force of ASM segments to acetylcholine, but it increased maximal relaxation response to isoproterenol. Treatment with specific NF-κB inhibitor produced similar protections as sevoflurane, including decreased expressions of TLR4 and NF-κB in cultured ASMCs and improved pharmacodynamic responsiveness of ASM to ACh and isoproterenol. Conclusions This study demonstrates the crucial role of TLR4 activation in ASMCs during ALI in response to LPS. Sevoflurane exerts direct relaxant and anti-inflammatory effects in vivo and in vitro via inhibition of TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Xi Jia Sun
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning, China
| | - Xiao Qian Li
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning, China
| | - Xiao Long Wang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning, China
| | - Wen Fei Tan
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning, China
| | - Jun Ke Wang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning, China
- * E-mail:
| |
Collapse
|
36
|
Chaïr-Yousfi I, Laraba-Djebari F, Hammoudi-Triki D. Androctonus australis hector venom contributes to the interaction between neuropeptides and mast cells in pulmonary hyperresponsiveness. Int Immunopharmacol 2015; 25:19-29. [PMID: 25601496 DOI: 10.1016/j.intimp.2015.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/06/2015] [Accepted: 01/09/2015] [Indexed: 11/28/2022]
Abstract
Lung injury and respiratory distress syndrome are frequent symptoms observed in the most severe cases of scorpion envenomation. The uncontrolled transmigration of leukocyte cells into the lung interstitium and alveolar space and pulmonary edema may be the cause of death. Mast cells can release various inflammatory mediators known to be involved in the development of lung edema following scorpion venom injection. The present study was designed to determine the evidence of neurokinin 1 (NK1) receptor and the involvement of mast cell activation to induce pulmonary edema and to increase vascular permeability after Androctonus australis hector (Aah) venom administration. To this end, mast cells were depleted using compound 48/80 (C48/80). Furthermore, the involvement of tachykinin NK1 receptors expressed on mast cell membranes was elucidated by their blocking with an antagonist. On the other hand, the ability of Aah venom to increase vascular permeability and to induce edema was also assessed by measuring the amount of Evans blue dye (EBD) extravasation in bronchoalveolar lavage (BAL) fluid and in the lungs of mice. Pulmonary edema, as assessed by the levels of EBD extravasation, was completely inhibited in compound 48/80-treated animals. Depletion by stimuli non-immunological C48/80 component markedly reduced induced inflammatory response following the venom administration. The mast cells seem to play an important role in the development of lung injury and the increase of vascular permeability in mice following the subcutaneous administration of Aah scorpion venom through the NK1 receptor.
Collapse
Affiliation(s)
- Imène Chaïr-Yousfi
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Algiers, Algeria
| | - Fatima Laraba-Djebari
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Algiers, Algeria.
| | - Djelila Hammoudi-Triki
- University of Sciences and Technology Houari Boumediene, Faculty of Biological Sciences, Laboratory of Cellular and Molecular Biology, Algiers, Algeria
| |
Collapse
|
37
|
Dileepan M, Jude JA, Rao SP, Walseth TF, Panettieri RA, Subramanian S, Kannan MS. MicroRNA-708 regulates CD38 expression through signaling pathways JNK MAP kinase and PTEN/AKT in human airway smooth muscle cells. Respir Res 2014; 15:107. [PMID: 25175907 PMCID: PMC4156970 DOI: 10.1186/s12931-014-0107-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 08/22/2014] [Indexed: 01/29/2023] Open
Abstract
Background The cell-surface protein CD38 mediates airway smooth muscle (ASM) contractility by generating cyclic ADP-ribose, a calcium-mobilizing molecule. In human ASM cells, TNF-α augments CD38 expression transcriptionally by NF-κB and AP-1 activation and involving MAPK and PI3K signaling. CD38−/− mice develop attenuated airway hyperresponsiveness following allergen or cytokine challenge. The post-transcriptional regulation of CD38 expression in ASM is relatively less understood. In ASM, microRNAs (miRNAs) regulate inflammation, contractility, and hyperproliferation. The 3’ Untranslated Region (3’UTR) of CD38 has multiple miRNA binding sites, including a site for miR-708. MiR-708 is known to regulate PI3K/AKT signaling and hyperproliferation of other cell types. We investigated miR-708 expression, its regulation of CD38 expression and the underlying mechanisms involved in such regulation in human ASM cells. Methods Growth-arrested human ASM cells from asthmatic and non-asthmatic donors were used. MiRNA and mRNA expression were measured by quantitative real-time PCR. CD38 enzymatic activity was measured by a reverse cyclase assay. Total and phosphorylated MAPKs and PI3K/AKT as well as enzymes that regulate their activation were determined by Western blot analysis of cell lysates following miRNA transfection and TNF-α stimulation. Dual luciferase reporter assays were performed to determine whether miR-708 binds directly to CD38 3’UTR to alter gene expression. Results Using target prediction algorithms, we identified several miRNAs with potential CD38 3’UTR target sites and determined miR-708 as a potential candidate for regulation of CD38 expression based on its expression and regulation by TNF-α. TNF-α caused a decrease in miR-708 expression in cells from non-asthmatics while it increased its expression in cells from asthmatics. Dual luciferase reporter assays in NIH-3 T3 cells revealed regulation of expression by direct binding of miR-708 to CD38 3’UTR. In ASM cells, miR-708 decreased CD38 expression by decreasing phosphorylation of JNK MAPK and AKT. These effects were associated with increased expression of MKP-1, a MAP kinase phosphatase and PTEN, a phosphatase that terminates PI3 kinase signaling. Conclusions In human ASM cells, TNF-α-induced CD38 expression is regulated by miR-708 directly binding to 3’UTR and indirectly by regulating JNK MAPK and PI3K/AKT signaling and has the potential to control airway inflammation, ASM contractility and proliferation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mathur S Kannan
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, 1971 Commonwealth Avenue, St, Paul 55108, MN, USA.
| |
Collapse
|
38
|
Wilson SJ, Rigden HM, Ward JA, Laviolette M, Jarjour NN, Djukanović R. The relationship between eosinophilia and airway remodelling in mild asthma. Clin Exp Allergy 2014; 43:1342-50. [PMID: 24261944 DOI: 10.1111/cea.12156] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 05/31/2013] [Accepted: 06/03/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Eosinophilia is a marker of corticosteroid responsiveness and risk of exacerbation in asthma; although it has been linked to submucosal matrix deposition, its relationship with other features of airway remodelling is less clear. OBJECTIVE The aim of this study was to investigate the relationship between airway eosinophilia and airway remodelling. METHODS Bronchial biopsies from subjects (n = 20 in each group) with mild steroid-naïve asthma, with either low (0-0.45 mm(-2)) ) or high submucosal eosinophil (23.43-46.28 mm(-2) ) counts and healthy controls were assessed for in vivo epithelial damage (using epidermal growth factor receptor staining), mucin expression, airway smooth muscle (ASM) hypertrophy and inflammatory cells within ASM. RESULTS The proportion of in vivo damaged epithelium was significantly greater (P = 0.02) in the high-eosinophil (27.37%) than the low-eosinophil (4.14%) group. Mucin expression and goblet cell numbers were similar in the two eosinophil groups; however, MUC-2 expression was increased (P = 0.002) in the high-eosinophil group compared with controls. The proportion of submucosa occupied by ASM was higher in both asthma groups (P = 0.021 and P = 0.046) compared with controls. In the ASM, eosinophil and T-lymphocyte numbers were higher (P < 0.05) in the high-eosinophil group than both the low-eosinophil group and the controls, whereas the numbers of mast cells were increased in the high-eosinophil group (P = 0.01) compared with controls. CONCLUSION Submucosal eosinophilia is a marker (and possibly a cause) of epithelial damage and is related to infiltration of ASM with eosinophils and T lymphocytes, but is unrelated to mucus metaplasia or smooth muscle hypertrophy.
Collapse
Affiliation(s)
- S J Wilson
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | | | |
Collapse
|
39
|
Alagappan VKT, de Boer WI, Misra VK, Mooi WJ, Sharma HS. Angiogenesis and vascular remodeling in chronic airway diseases. Cell Biochem Biophys 2014; 67:219-34. [PMID: 23975597 DOI: 10.1007/s12013-013-9713-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asthma and chronic obstructive pulmonary disease remain a global health problem, with increasing morbidity and mortality. Despite differences in the causal agents, both diseases exhibit various degrees of inflammatory changes, structural alterations of the airways leading to airflow limitation. The existence of transient disease phenotypes which overlap both diseases and which progressively decline the lung function has complicated the search for an effective therapy. Important characteristics of chronic airway diseases include airway and vascular remodeling, of which the molecular mechanisms are complex and poorly understood. Recently, we and others have shown that airway smooth muscle (ASM) cells are not only structural and contractile components of airways, rather they bear capabilities of producing large number of pro-inflammatory and mitogenic factors. Increase in size and number of blood vessels both inside and outside the smooth muscle layer as well as hyperemia of bronchial vasculature are contributing factors in airway wall remodeling in patients with chronic airway diseases, proposing for the ongoing mechanisms like angiogenesis and vascular dilatation. We believe that vascular changes directly add to the airway narrowing and hyper-responsiveness by exudation and transudation of proinflammatory mediators, cytokines and growth factors; facilitating trafficking of inflammatory cells; causing oedema of the airway wall and promoting ASM accumulation. One of the key regulators of angiogenesis, vascular endothelial growth factor in concerted action with other endothelial mitogens play pivotal role in regulating bronchial angiogenesis. In this review article we address recent advances in pulmonary angiogenesis and remodelling that contribute in the pathogenesis of chronic airway diseases.
Collapse
|
40
|
Giembycz MA, Maurice DH. Cyclic nucleotide-based therapeutics for chronic obstructive pulmonary disease. Curr Opin Pharmacol 2014; 16:89-107. [PMID: 24810285 DOI: 10.1016/j.coph.2014.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 12/18/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) defines a group of chronic inflammatory disorders of the airways that are characterised by a progressive and largely irreversible decline in expiratory airflow. Drugs used to treat COPD through actions mediated by cyclic AMP (cAMP) are restricted to long-acting and short-acting β2-adrenoceptor agonists and, in a subset of patients with chronic bronchitis, a phosphodiesterase 4 inhibitor, roflumilast. These agents relax airway smooth muscle and suppress inflammation. At the molecular level, these effects in the airways are mediated by two cAMP effectors, cAMP-dependent protein kinase and exchange proteins activated by cAMP. The pharmacology of newer agents, acting through these systems, is discussed here with an emphasis on their potential to interact and increase therapeutic effectiveness.
Collapse
Affiliation(s)
- Mark A Giembycz
- Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Donald H Maurice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
41
|
Overed-Sayer C, Rapley L, Mustelin T, Clarke DL. Are mast cells instrumental for fibrotic diseases? Front Pharmacol 2014; 4:174. [PMID: 24478701 PMCID: PMC3896884 DOI: 10.3389/fphar.2013.00174] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/20/2013] [Indexed: 01/17/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder of unknown etiology characterized by accumulation of lung fibroblasts and extracellular matrix deposition, ultimately leading to compromised tissue architecture and lung function capacity. IPF has a heterogeneous clinical course; however the median survival after diagnosis is only 3–5 years. The pharmaceutical and biotechnology industry has made many attempts to find effective treatments for IPF, but the disease has so far defied all attempts at therapeutic intervention. Clinical trial failures may arise for many reasons, including disease heterogeneity, lack of readily measurable clinical end points other than overall survival, and, perhaps most of all, a lack of understanding of the underlying molecular mechanisms of the progression of IPF. The precise link between inflammation and fibrosis remains unclear, but it appears that immune cells can promote fibrosis by releasing fibrogenic factors. So far, however, therapeutic approaches targeting macrophages, neutrophils, or lymphocytes have failed to alter disease pathogenesis. A new cell to garner research interest in fibrosis is the mast cell. Increased numbers of mast cells have long been known to be present in pulmonary fibrosis and clinically correlations between mast cells and fibrosis have been reported. More recent data suggests that mast cells may contribute to the fibrotic process by stimulating fibroblasts resident in the lung, thus driving the pathogenesis of the disease. In this review, we will discuss the mast cell and its physiological role in tissue repair and remodeling, as well as its pathological role in fibrotic diseases such as IPF, where the process of tissue repair and remodeling is thought to be dysregulated.
Collapse
Affiliation(s)
| | - Laura Rapley
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| | - Tomas Mustelin
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| | - Deborah L Clarke
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| |
Collapse
|
42
|
Cleary RA, Wang R, Wang T, Tang DD. Role of Abl in airway hyperresponsiveness and airway remodeling. Respir Res 2013; 14:105. [PMID: 24112389 PMCID: PMC3852349 DOI: 10.1186/1465-9921-14-105] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 10/09/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Asthma is a chronic disease that is characterized by airway hyperresponsiveness and airway remodeling. The underlying mechanisms that mediate the pathological processes are not fully understood. Abl is a non-receptor protein tyrosine kinase that has a role in the regulation of smooth muscle contraction and smooth muscle cell proliferation in vitro. The role of Abl in airway hyperresponsiveness and airway remodeling in vivo is largely unknown. METHODS To evaluate the role of Abl in asthma pathology, we assessed the expression of Abl in airway tissues from the ovalbumin sensitized and challenged mouse model, and human asthmatic airway smooth muscle cells. In addition, we generated conditional knockout mice in which Abl expression in smooth muscle was disrupted, and then evaluated the effects of Abl conditional knockout on airway resistance, smooth muscle mass, cell proliferation, IL-13 and CCL2 in the mouse model of asthma. Furthermore, we determined the effects of the Abl pharmacological inhibitors imatinib and GNF-5 on these processes in the animal model of asthma. RESULTS The expression of Abl was upregulated in airway tissues of the animal model of asthma and in airway smooth muscle cells of patients with severe asthma. Conditional knockout of Abl attenuated airway resistance, smooth muscle mass and staining of proliferating cell nuclear antigen in the airway of mice sensitized and challenged with ovalbumin. Interestingly, conditional knockout of Abl did not affect the levels of IL-13 and CCL2 in bronchoalveolar lavage fluid of animals treated with ovalbumin. However, treatment with imatinib and GNF-5 inhibited the ovalbumin-induced increase in IL-13 and CCL2 as well as airway resistance and smooth muscle growth in animals. CONCLUSIONS These results suggest that the altered expression of Abl in airway smooth muscle may play a critical role in the development of airway hyperresponsiveness and airway remodeling in asthma. Our findings support the concept that Abl may be a novel target for the development of new therapy to treat asthma.
Collapse
Affiliation(s)
- Rachel A Cleary
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue MC-8, Albany, NY 12208, USA.
| | | | | | | |
Collapse
|
43
|
Wright DB, Trian T, Siddiqui S, Pascoe CD, Johnson JR, Dekkers BG, Dakshinamurti S, Bagchi R, Burgess JK, Kanabar V, Ojo OO. Phenotype modulation of airway smooth muscle in asthma. Pulm Pharmacol Ther 2013; 26:42-9. [DOI: 10.1016/j.pupt.2012.08.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/11/2012] [Accepted: 08/13/2012] [Indexed: 01/26/2023]
|
44
|
Billington CK, Ojo OO, Penn RB, Ito S. cAMP regulation of airway smooth muscle function. Pulm Pharmacol Ther 2013; 26:112-20. [PMID: 22634112 PMCID: PMC3574867 DOI: 10.1016/j.pupt.2012.05.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/14/2012] [Accepted: 05/17/2012] [Indexed: 12/11/2022]
Abstract
Agonists activating β(2)-adrenoceptors (β(2)ARs) on airway smooth muscle (ASM) are the drug of choice for rescue from acute bronchoconstriction in patients with both asthma and chronic obstructive pulmonary disease (COPD). Moreover, the use of long-acting β-agonists combined with inhaled corticosteroids constitutes an important maintenance therapy for these diseases. β-Agonists are effective bronchodilators due primarily to their ability to antagonize ASM contraction. The presumed cellular mechanism of action involves the generation of intracellular cAMP, which in turn can activate the effector molecules cAMP-dependent protein kinase (PKA) and Epac. Other agents such as prostaglandin E(2) and phosphodiesterase inhibitors that also increase intracellular cAMP levels in ASM, can also antagonize ASM contraction, and inhibit other ASM functions including proliferation and migration. Therefore, β(2)ARs and cAMP are key players in combating the pathophysiology of airway narrowing and remodeling. However, limitations of β-agonist therapy due to drug tachyphylaxis related to β(2)AR desensitization, and recent findings regarding the manner in which β(2)ARs and cAMP signal, have raised new and interesting questions about these well-studied molecules. In this review we discuss current concepts regarding β(2)ARs and cAMP in the regulation of ASM cell functions and their therapeutic roles in asthma and COPD.
Collapse
Affiliation(s)
- Charlotte K Billington
- Division of Therapeutics and Molecular Medicine, The University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | | |
Collapse
|
45
|
Markwick LJ, Clements D, Roberts ME, Ceresa CC, Knox AJ, Johnson SR. CCR3 induced-p42/44 MAPK activation protects against staurosporine induced-DNA fragmentation but not apoptosis in airway smooth muscle cells. Clin Exp Allergy 2012; 42:1040-50. [PMID: 22702503 DOI: 10.1111/j.1365-2222.2012.04019.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chemokine receptors (CCRs) are expressed on airway smooth muscle (ASM) cells. As their ligands are present in the airways in asthma, we hypothesized that ASM CCR activation could promote the increase in ASM mass seen in patients with chronic asthma. OBJECTIVE To determine which CCRs are expressed by ASM cells and their potential functional relevance to the chronic airway changes seen in asthma. METHODS CCR expression in primary ASM cell cultures and airway biopsies from patients with and without asthma was examined by RT-PCR, fluorescence-activated cell sorting and immunohistochemistry. ASM p42/44 MAPK activity, proliferation, migration and apoptosis were examined by western blotting, thymidine incorporation, transwell assay and TUNEL assay respectively. RESULTS CCR3 was the most frequently expressed CCR protein and was present on 79 ± 14% of cells. CX3CR1 and CXCR6 were present on 6% and 11% of cells respectively. CCR3 ligands CCL11 and CCL24 caused rapid activation of p42/44 MAPK but not Akt. CCR3 activation did not affect ASM proliferation, migration or VEGF secretion. DNA fragmentation detected by TUNEL staining could be induced by staurosporine and Fas activation although only Fas activation resulted in caspase 3 cleavage. CCL11 and CCL24 protected ASM cells against DNA fragmentation dependent upon p42/44 MAPK activity only via caspase 3 independent pathways. CCR3 was expressed in the smooth muscle and epithelium in the airways of patients with and without asthma. Smooth muscle cell DNA fragmentation in the airways of patients with stable asthma and controls was very uncommon. CONCLUSIONS AND CLINICAL RELEVANCE CCR3 is strongly expressed by ASM cells in vitro and in vivo. Protection against cell death by CCR3 activation is dependent on p42/44 MAPK but does not affect caspase 3 mediated apoptosis.
Collapse
Affiliation(s)
- L J Markwick
- Division of Therapeutics and Molecular Medicine and Nottingham NIHR Respiratory Biomedical Research Unit, University Hospital Queens Medical Centre, Nottingham, UK
| | | | | | | | | | | |
Collapse
|
46
|
Airway smooth muscle as a target in asthma and the beneficial effects of bronchial thermoplasty. J Allergy (Cairo) 2012; 2012:593784. [PMID: 23024662 PMCID: PMC3457660 DOI: 10.1155/2012/593784] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/01/2012] [Indexed: 11/17/2022] Open
Abstract
Airflow within the airways is determined directly by the lumenal area of that airway. In this paper, we consider several factors which can reduce airway lumenal area, including thickening and/or active constriction of the airway smooth muscle (ASM). The latter cell type can also contribute in part to inflammation, another feature of asthma, through its ability to take on a synthetic/secretory phenotype. The ASM therefore becomes a strategically important target in the treatment of asthma, given these key contributions to the pathophysiology of that disease. Pharmacological approaches have been developed to elicit relaxation of the ASM, but these are not always effective in all patients, nor do they address the long-term structural changes which impinge on the airway lumen. The recent discovery that thermal energy can be used to ablate smooth muscle has led to the development of a novel physical intervention—bronchial thermoplasty—in the treatment of asthma. Here, we review the evolution of this novel approach, consider some of the possible mechanisms that account for its salutary effects, and pose new questions which may lead to even better therapies for asthma.
Collapse
|
47
|
Moretto N, Bertolini S, Iadicicco C, Marchini G, Kaur M, Volpi G, Patacchini R, Singh D, Facchinetti F. Cigarette smoke and its component acrolein augment IL-8/CXCL8 mRNA stability via p38 MAPK/MK2 signaling in human pulmonary cells. Am J Physiol Lung Cell Mol Physiol 2012; 303:L929-38. [PMID: 22983351 DOI: 10.1152/ajplung.00046.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interleukin-8 (IL-8/CXCL8) is an important neutrophil chemoattractant known to be elevated in the airways of cigarette smokers and in patients with chronic obstructive pulmonary disease (COPD). We examined the acute effect of aqueous cigarette smoke extract (CSE) on IL-8 expression in primary human pulmonary cells, in particular in normal human bronchial smooth muscle cells (HBSMCs). IL-8 mRNA levels increased upon CSE exposure in a concentration- and time-dependent manner, and such an effect was accompanied by IL-8 secretion. CSE-evoked elevation of IL-8 mRNA was mimicked by its component acrolein. Both CSE and acrolein induced p38 mitogen-activated protein kinase (MAPK) phosphorylation, accompanied by the phosphorylation of MAPK-activated kinase 2 (MK2), a known downstream substrate of the p38 MAPK, both in HBSMCs and in human airway epithelial cells. Furthermore, pharmacological inhibition of p38 MAPK or MK2 strongly accelerated the decay of IL-8 mRNA levels upon stimulation with CSE or acrolein and subsequent blockade of mRNA neosynthesis with actinomycin D in pulmonary structural cells (HBSMCs and airways epithelial cells) as well as in human alveolar macrophages. Conversely, pharmacological inhibition of ERK1/2 signaling inhibited CSE-induced steady-state levels of IL-8 mRNA without affecting mRNA stability, thus suggesting inhibition at the transcriptional level. In sum, p38 MAPK/MK2 signaling is an important posttranscriptional mechanism underlying upregulation of IL-8 mRNA levels elicited by CSE and acrolein. Given the pivotal role of IL-8 in neutrophil chemotaxis and activation, our results shed light on the mechanisms through which cigarette smoke can initiate inflammation in the lung.
Collapse
Affiliation(s)
- Nadia Moretto
- Dept. of Pharmacology, Chiesi Farmaceutici, Parma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yeganeh B, Mukherjee S, Moir LM, Kumawat K, Kashani HH, Bagchi RA, Baarsma HA, Gosens R, Ghavami S. Novel non-canonical TGF-β signaling networks: emerging roles in airway smooth muscle phenotype and function. Pulm Pharmacol Ther 2012; 26:50-63. [PMID: 22874922 DOI: 10.1016/j.pupt.2012.07.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 12/19/2022]
Abstract
The airway smooth muscle (ASM) plays an important role in the pathophysiology of asthma and chronic obstructive pulmonary disease (COPD). ASM cells express a wide range of receptors involved in contraction, growth, matrix protein production and the secretion of cytokines and chemokines. Transforming growth factor beta (TGF-β) is one of the major players in determining the structural and functional abnormalities of the ASM in asthma and COPD. It is increasingly evident that TGF-β functions as a master switch, controlling a network of intracellular and autocrine signaling loops that effect ASM phenotype and function. In this review, the various elements that participate in non-canonical TGF-β signaling, including MAPK, PI3K, WNT/β-catenin, and Ca(2+), are discussed, focusing on their effect on ASM phenotype and function. In addition, new aspects of ASM biology and their possible association with non-canonical TGF-β signaling will be discussed.
Collapse
Affiliation(s)
- Behzad Yeganeh
- Department of Physiology, Manitoba Institute of Child Health, University of Manitoba, 675 McDermot Ave, Winnipeg, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Malhotra S, Deshmukh SS, Dastidar SG. COX inhibitors for airway inflammation. Expert Opin Ther Targets 2012; 16:195-207. [PMID: 22324934 DOI: 10.1517/14728222.2012.661416] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION The cyclooxygenase (COX) enzyme, which is responsible for the production of prostaglandins (PGs), key mediators of inflammation, may have the potential to become an attractive target for anti-inflammatory therapy. COX catalyzes the conversion of arachidonic acid (AA) into PGs, which play a significant role in disease. PGs are lipid mediators of central importance in the regulation of inflammation and smooth muscle tone. Airway-resident inflammatory cells release PGs: PGD2 and PDF2a amplify smooth muscle contraction and airway inflammation. Following its conversion from membrane phospholipids by phospholipase, AA enters the prostanoid pathway via COX, which catalyzes the conversion of AA to PGH2. PGH2 is then converted to biologically active PGs by cell-specific PG synthases. As COX is the rate limiting step in the PG pathway, the regulation of this enzyme is of critical importance in PG production. AREAS COVERED This review addresses the opportunities and challenges of COX inhibitors as therapeutic targets in airway inflammation. The review covers literature from the past 20 years. EXPERT OPINION Current literature favors COX inhibitors as potential targets for airway diseases. However, from the information available, it is not clear whether the COX enzyme by itself can serve as a target in drug development for asthma and COPD. Therefore, additional research is required to elucidate the mechanisms of action of COX metabolites before it can be considered as a target.
Collapse
Affiliation(s)
- Sanjay Malhotra
- Daiichi Sankyo India Pharma Private Ltd., Department of Chemistry, Haryana, India.
| | | | | |
Collapse
|
50
|
Banerjee A, Panettieri R. Vitamin D modulates airway smooth muscle function in COPD. Curr Opin Pharmacol 2012; 12:266-74. [PMID: 22365730 DOI: 10.1016/j.coph.2012.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 01/24/2012] [Accepted: 01/26/2012] [Indexed: 11/30/2022]
Abstract
COPD is a disease manifested as persistent airflow obstruction with an enhanced inflammatory response in the airways and lungs to noxious particles and gases which evokes symptoms of dyspnea on exertion, cough and mucus production. Airway smooth muscle plays a central role in the COPD diathesis and is implicated in many aspects of COPD pathogenesis. Vitamin D deficiency has been associated with COPD severity and studies suggest a role for Vitamin D as a treatment for COPD. In this review, we describe the effects of 1,25-dihydroxyvitamin D on airway smooth muscle function, including agonist-induced shortening, secretion of inflammatory mediators, and myocyte hypertrophy and hyperplasia.
Collapse
Affiliation(s)
- Audreesh Banerjee
- Department of Medicine, Airways Biology Initiative, Division of Pulmonary, Allergy and Critical Care Medicine, Hospital of University of Pennsylvania, Philadelphia, PA, United States
| | | |
Collapse
|