1
|
Chen H, Liu J, Zhang J, Chen Y, Wang Y, Qiu Y, Hu H, Chai L, Zhang Q, Wang Q, Li M. USP22/BRD4 mediated hedgehog pathway activation contributes to airway remodeling in asthma. Int Immunopharmacol 2025; 153:114538. [PMID: 40132456 DOI: 10.1016/j.intimp.2025.114538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Bromodomain-containing protein 4 (BRD4) is recognized as a member of the bromodomain and extraterminal (BET) family that is involved in the airway inflammation and airway remodeling of asthma. However, the underlying mechanisms of BRD4 in airway smooth muscle cells (ASMCs) proliferation and airway remodeling remain unclear. Primary cultured rat ASMCs and ovalbumin (OVA)-induced rat asthma models were applied to address these issues in the present study. We showed that transforming growth factor β1 (TGF-β1) increased the protein expression of ubiquitin specific peptidase 22 (USP22), which deubiquitinated BRD4 therefore increased its expression, and then resulted in the upregulation of glioma-associated oncogene homolog 1 (GLI1) and osteopontin (OPN) leading to ASMCs proliferation. We further confirmed that induction of TGF-β1 sequentially upregulated USP22, BRD4, GLI1 and OPN leading to airway remodeling in OVA-induced rat asthma models, targeting TGF-β1/USP22/BRD4/GLI1/OPN pathway axis effectively attenuated airway remodeling and asthma development. Our study provides novel sights to understand the role of TGF-β1/USP22/BRD4/GLI1/OPN axis in airway remodeling, and targeting this pathway might have potential value for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Huan Chen
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jin Liu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jia Zhang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuqian Chen
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yan Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuanjie Qiu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Huizhong Hu
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Limin Chai
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qianqian Zhang
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qingting Wang
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Manxiang Li
- Department of Respiratory & Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
2
|
Domingo C, Busse WW, Hanania NA, Ertugrul M, Millette LA, Maio‐Twofoot T, Jaumont X, Palomares O. The Direct and Indirect Role of IgE on Airway Epithelium in Asthma. Allergy 2025; 80:919-931. [PMID: 39963805 PMCID: PMC11969325 DOI: 10.1111/all.16459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/29/2024] [Accepted: 12/17/2024] [Indexed: 04/05/2025]
Abstract
Asthma is a chronic airway inflammatory disorder, affecting over 350 million people worldwide, with allergic asthma being the most common form of the disease. Allergic asthma is characterized by a type 2 (T2) inflammatory response triggered by numerous allergens beginning in the airway epithelium, which acts as a physical barrier to allergens as well as other external irritants including infectious agents, and atmospheric pollutants. T2 inflammation is propagated by several key cell types including T helper 2 (Th2) cells, eosinophils, mast cells, and B cells. Immunoglobulin E (IgE), produced by B cells, is a key molecule in allergic airway disease and plays an important role in T2 inflammation, as well as being central to remodeling processes within the airway epithelium. Blocking IgE with omalizumab has been shown to be efficacious in treating allergic asthma however, the role of IgE on airway epithelial cells is less communicated. Developing a deeper explanation of the complex network of interactions between IgE and the airway epithelium will facilitate an improved understanding of asthma pathophysiology. This review discusses the indirect and direct roles of IgE on airway epithelial cells, with a focus on allergic asthma disease.
Collapse
Affiliation(s)
- Christian Domingo
- Department of Pulmonary Medicine, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT‐CERCA)Universitat Autònoma de BarcelonaSabadellSpain
| | - William W. Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Nicola A. Hanania
- Section of Pulmonary, Critical Care and Sleep MedicineBaylor College of MedicineHoustonTexasUSA
| | | | | | | | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of ChemistryComplutense University of MadridMadridSpain
| |
Collapse
|
3
|
He XY, Han MM, Zhao YC, Tang L, Wang Y, Xing L, Wei N, Wang J, Wang GJ, Zhou F, Jeong JH, Jiang HL. Surface-engineered mesenchymal stem cell for refractory asthma therapy: Reversing airway remodeling. J Control Release 2024; 376:972-984. [PMID: 39476873 DOI: 10.1016/j.jconrel.2024.10.056] [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: 06/25/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/09/2024]
Abstract
In the development of asthma, subepithelial fibrosis and vascular proliferation cause airway remodeling and narrowing, leading to disease deterioration and respiratory failure. In the clinic, the treatment of asthma was aimed at reducing the frequency of acute asthma attacks through inhaled corticosteroids (ICSs). However, ICSs cannot prevent the progression into refractory asthma due to the formation of airway remodeling mainly by subepithelial fibrosis and angiogenesis surrounding the tracheal lumen. Herein, we constructed surface-engineered mesenchymal stem cells (MSCs/PVLA) via the bioconjugation of MSCs and reactive oxygen species-responsive polymeric micelles loaded with vactosertib (VST) and linifanib (LFN) for treating refractory asthma through reversing airway remodeling. MSCs/PVLA migrated to the tracheal lumen due to the inflammation tropism of MSCs, and subsequently released VST and LFN could inhibit the formation of airway remodeling by preventing subepithelial fibrosis and angiogenesis. Meanwhile, MSCs reduced inflammatory cell infiltration and cytokine secretion to regulate the pathological microenvironment. Our results suggested that MSCs/PVLA could serve as a promising candidate to prevent disease exacerbations and treat refractory asthma.
Collapse
Affiliation(s)
- Xing-Yue He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Meng-Meng Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu-Chen Zhao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ning Wei
- Jiangsu Renocell Biotech Co., Ltd., Nanjing 210009, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd., Nanjing 210009, China
| | - Guang-Ji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; College of Pharmacy, Yanbian University, Yanji 133002, China; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
4
|
Liu W, Liu Z, Sun XD, Liu ZQ, Dong YY, Qiu S. Investigating the causal association between heme oxygenase-1 and asthma: A bidirectional two-sample Mendelian randomization analysis in a European population. World Allergy Organ J 2024; 17:100987. [PMID: 39512673 PMCID: PMC11541772 DOI: 10.1016/j.waojou.2024.100987] [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: 01/12/2024] [Revised: 08/29/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Background The association between heme oxygenase-1 (HO-1) and asthma has been a subject of debate in both observational and experimental studies. We aimed to evaluate the potential causal relationship between HO-1 and asthma. Materials and methods A bidirectional two-sample Mendelian randomization (TSMR) study was conducted to examine the causal relationship between HO-1 and asthma. In the forward Mendelian randomization (MR) analyses, HO-1 was considered as the exposure, while asthma as the outcome. Conversely, in the reverse MR analyses, asthma was regarded as the exposure, and HO-1 as the outcome. Data for HO-1 and asthma were obtained from publicly accessible genome-wide association studies (GWAS). These causal relationships were identified through 5 MR methods, namely MR-Egger, weighted median, inverse-variance weighted (IVW), simple mode, and weighted mode. Additionally, sensitivity tests were conducted to assess the robustness of MR study. Finally, additional asthma datasets and childhood asthma were selected to validate the findings. Results In the forward MR analyses, according to the IVW method, genetically predicted HO-1 displays a negative correlation with the risk of asthma (OR 0.947, 95% CI 0.905-0.990). It was not found any SNP overly sensitive or disproportionately responsible for the outcome. No evidence of heterogeneity and pleiotropy between SNPs was observed. Genetically predicted asthma was not associated with HO-1 in reverse MR analyses using the IVW method. The same results were validated in additional asthma datasets and in childhood asthma. Conclusion The results of MR analysis revealed heme oxygenase-1 as a protective factor for asthma.
Collapse
Affiliation(s)
- Wen Liu
- Department of Cadre Health Care, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Zhen Liu
- Department of Cadre Health Care, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Xiao-di Sun
- Department of Cadre Health Care, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Zeng-qiang Liu
- Department of Cadre Health Care, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Yuan-yuan Dong
- Department of Cadre Health Care, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Shi Qiu
- Department of Cardiac Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| |
Collapse
|
5
|
Teegala LR, Gudneppanavar R, Kattuman EES, Snyderman M, AV T, Katari V, Thodeti CK, Paruchuri S. Prostaglandin E 2 attenuates lung fibroblast differentiation via inactivation of yes-associated protein signaling. FASEB J 2023; 37:e23199. [PMID: 37732601 PMCID: PMC11996057 DOI: 10.1096/fj.202300745rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Prostaglandin E2 (PGE2 ) has been implicated in counteracting fibroblast differentiation by TGFβ1 during pulmonary fibrosis. However, the precise mechanism is not well understood. We show here that PGE2 via EP2 R and EP4 R inhibits the expression of mechanosensory molecules Lysyl Oxidase Like 2 (LOXL2), myocardin-related transcription factor A (MRTF-A), ECM proteins, plasminogen activation inhibitor 1 (PAI-1), fibronectin (FN), α-smooth muscle actin (α-SMA), and redox sensor (nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4)) required for TGFβ1-mediated fibroblast differentiation. We further demonstrate that PGE2 inhibits fibrotic signaling via Yes-associated protein (YAP) but does so independently from its actions on SMAD phosphorylation and conserved cylindromatosis (CYLD; deubiquitinase) expression. Mechanistically, PGE2 phosphorylates/inactivates YAP downstream of EP2 R/Gαs and restrains its translocation to the nucleus, thus inhibiting its interaction with TEA domain family members (TEADs) and transcription of fibrotic genes. Importantly, pharmacological or siRNA-mediated inhibition of YAP significantly downregulates TGFβ1-mediated fibrotic gene expression and myofibroblast formation. Notably, YAP expression is upregulated in the lungs of D. farinae-treated wild type (WT) mice relative to saline-treated WT mice. Our results unravel a unique role for PGE2 -YAP interactions in fibroblast differentiation, and that PGE2 /YAP inhibition can be used as a novel therapeutic target in the treatment of pathological conditions associated with myofibroblasts like asthma.
Collapse
Affiliation(s)
| | | | | | | | - Thanusha AV
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH
| | - Venkatesh Katari
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH
| | - Charles K Thodeti
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH
| | - Sailaja Paruchuri
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH
| |
Collapse
|
6
|
Wieczfinska J, Sitarek P, Kowalczyk T, Rieske P, Pawliczak R. Curcumin modulates airway remodelling-contributing genes-the significance of transcription factors. J Cell Mol Med 2021; 26:736-749. [PMID: 34939316 PMCID: PMC8817128 DOI: 10.1111/jcmm.17102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
Bronchial epithelial cells and fibroblasts play an essential role in airway remodelling, due to their protective and secretory functions. There are many studies proving that infection caused by human rhinovirus may contribute to the process of airway remodelling. The beneficial properties of curcumin, the basic ingredient of turmeric, have been proved in many studies. Therefore, the aim of this study was the evaluation of curcumin immunomodulatory properties in development of airway remodelling. Fibroblasts (WI‐38 and HFL1) and epithelial cells (NHBE) were incubated with curcumin. Additionally, remodelling conditions were induced with rhinovirus (HRV). Airway remodelling genes were determined by qPCR and immunoblotting. Moreover, NF‐κB, c‐Myc and STAT3 were silenced to analyse the pathways involved in airway remodelling. Curcumin reduced the expression of the genes analysed, especially MMP‐9, TGF‐β and collagen I. Moreover, curcumin inhibited the HRV‐induced expression of MMP‐9, TGF‐β, collagen I and LTC4S (p < 0.05). NF‐κB, c‐Myc and STAT3 changed their course of expression. Concluding, our study shows that curcumin significantly downregulated gene expression related to the remodelling process, which is dependent on NF‐κB and, partially, on c‐Myc and STAT3. The results suggest that the remodelling process may be limited and possibly prevented, however this issue requires further research.
Collapse
Affiliation(s)
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz, Poland
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Medical University of Lodz, Poland
| | - Rafal Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
7
|
Islam R, Dash D, Singh R. Intranasal curcumin and sodium butyrate modulates airway inflammation and fibrosis via HDAC inhibition in allergic asthma. Cytokine 2021; 149:155720. [PMID: 34634654 DOI: 10.1016/j.cyto.2021.155720] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 01/12/2023]
Abstract
Asthma being an inflammatory disease of the airways lead to structural alterations in lungs which often results in the severity of the disease. Curcumin, diferuloylmethane, is well known for its medicinal properties but its anti-inflammatory potential via Histone deacetylase inhibition (HDACi) has not been revealed yet. Therefore, we have explored here, anti-inflammatory and anti-fibrotic potential of intranasal curcumin via HDAC inhibition and compared its potential with Sodium butyrate (SoB), a known histone deacetylase inhibitor of Class I and II series. Anti-inflammatory potential of SoB, has been investigated in cancer but not been studied in asthma before. MATERIALS AND METHODS In present study, ovalbumin (OVA) was used to sensitize Balb/c mice and later exposed to (1%) OVA aerosol. Curcumin (5 mg/kg) and Sodium butyrate (50 mg/kg) was administered through intranasal route an hour before OVA aerosol challenge. Efficacies of SoB and Curcumin as HDAC inhibitors were evaluated in terms of different inflammatory parameters like, total inflammatory cell count, reactive oxygen species (ROS), histamine release, nitric oxide and serum IgE levels. Inflammatory cell recruitment was analyzed by H&E staining and structural alterations were revealed by Masson's Trichrome staining of lung sections. RESULTS Enhanced Matrix Metalloproteinase-2 and 9 (MMP-2 and MMP-9) activities were observed in bronchoalveolar lavage fluid (BALF) of asthmatic mice by gelatin zymography which was inhibited in both treatment groups. Protein expressions of MMP-9, HDAC 1, H3acK9 and NF-kB p65 were modulated in intranasal curcumin and SoB pretreatment groups. CONCLUSION This is the first report where intranasal curcumin inhibited asthma severity via affecting HDAC 1 (H3acK9) leading to NF-kB suppression in mouse model of allergic asthma.
Collapse
Affiliation(s)
- Ramiya Islam
- Department of Zoology, MMV, Banaras Hindu University, Varanasi 221005, India
| | - D Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rashmi Singh
- Department of Zoology, MMV, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
8
|
Sonnenberg-Riethmacher E, Miehe M, Riethmacher D. Periostin in Allergy and Inflammation. Front Immunol 2021; 12:722170. [PMID: 34512647 PMCID: PMC8429843 DOI: 10.3389/fimmu.2021.722170] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Matricellular proteins are involved in the crosstalk between cells and their environment and thus play an important role in allergic and inflammatory reactions. Periostin, a matricellular protein, has several documented and multi-faceted roles in health and disease. It is differentially expressed, usually upregulated, in allergic conditions, a variety of inflammatory diseases as well as in cancer and contributes to the development and progression of these diseases. Periostin has also been shown to influence tissue remodelling, fibrosis, regeneration and repair. In allergic reactions periostin is involved in type 2 immunity and can be induced by IL-4 and IL-13 in bronchial cells. A variety of different allergic diseases, among them bronchial asthma and atopic dermatitis (AD), have been shown to be connected to periostin expression. Periostin is commonly expressed in fibroblasts and acts on epithelial cells as well as fibroblasts involving integrin and NF-κB signalling. Also direct signalling between periostin and immune cells has been reported. The deposition of periostin in inflamed, often fibrotic, tissues is further fuelling the inflammatory process. There is increasing evidence that periostin is also expressed by epithelial cells in several of the above-mentioned conditions as well as in cancer. Augmented periostin expression has also been associated with chronic inflammation such as in inflammatory bowel disease (IBD). Periostin can be expressed in a variety of different isoforms, whose functions have not been elucidated yet. This review will discuss potential functions of periostin and its different isoforms in allergy and inflammation.
Collapse
Affiliation(s)
- Eva Sonnenberg-Riethmacher
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
- Department of Human Development and Health, School of Medicine, University of Southampton, Southampton, United Kingdom
| | - Michaela Miehe
- Department of Biological and Chemical Engineering – Immunological Biotechnology, Aarhus University, Aarhus, Denmark
| | - Dieter Riethmacher
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
- Department of Human Development and Health, School of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
9
|
Chen W, Luo J, Ye Y, Hoyle R, Liu W, Borst R, Kazani S, Shikatani EA, Erpenbeck VJ, Pavord ID, Klenerman P, Sandham DA, Xue L. The Roles of Type 2 Cytotoxic T Cells in Inflammation, Tissue Remodeling, and Prostaglandin (PG) D 2 Production Are Attenuated by PGD 2 Receptor 2 Antagonism. THE JOURNAL OF IMMUNOLOGY 2021; 206:2714-2724. [PMID: 34011519 PMCID: PMC7610864 DOI: 10.4049/jimmunol.2001245] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/20/2021] [Indexed: 12/13/2022]
Abstract
Multiple proinflammatory effects of Tc2 cells are inhibited by DP2 antagonism. Tissue-remodeling functions of Tc2 cells are attenuated by DP2 antagonism. Autocrine/paracrine PGD2 production in Tc2 cells is reduced by DP2 antagonism.
Human type 2 cytotoxic T (Tc2) cells are enriched in severe eosinophilic asthma and can contribute to airway eosinophilia. PGD2 and its receptor PGD2 receptor 2 (DP2) play important roles in Tc2 cell activation, including migration, cytokine production, and survival. In this study, we revealed novel, to our knowledge, functions of the PGD2/DP2 axis in Tc2 cells to induce tissue-remodeling effects and IgE-independent PGD2 autocrine production. PGD2 upregulated the expression of tissue-remodeling genes in Tc2 cells that enhanced the fibroblast proliferation and protein production required for tissue repair and myofibroblast differentiation. PGD2 stimulated Tc2 cells to produce PGD2 using the routine PGD2 synthesis pathway, which also contributed to TCR-dependent PGD2 production in Tc2 cells. Using fevipiprant, a specific DP2 antagonist, we demonstrated that competitive inhibition of DP2 not only completely blocked the cell migration, adhesion, proinflammatory cytokine production, and survival of Tc2 cells triggered by PGD2 but also attenuated the tissue-remodeling effects and autocrine/paracrine PGD2 production in Tc2 induced by PGD2 and other stimulators. These findings further confirmed the anti-inflammatory effect of fevipiprant and provided a better understanding of the role of Tc2 cells in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Wentao Chen
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jian Luo
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Yuan Ye
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ryan Hoyle
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Wei Liu
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Rowie Borst
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Shamsah Kazani
- Novartis Institutes for BioMedical Research, Cambridge MA
| | | | | | - Ian D Pavord
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Translational Gastroenterology Unit and Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | | | - Luzheng Xue
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
10
|
Wieczfinska J, Sitarek P, Kowalczyk T, Pawliczak R. Leonurus sibiricus root extracts decrease airway remodeling markers expression in fibroblasts. Clin Exp Immunol 2020; 202:28-46. [PMID: 32562256 DOI: 10.1111/cei.13481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
Bronchial asthma is believed to be provoked by the interaction between airway inflammation and remodeling. Airway remodeling is a complex and poorly understood process, and controlling it appears key for halting the progression of asthma and other obstructive lung diseases. Plants synthesize a number of valuable compounds as constitutive products and as secondary metabolites, many of which have curative properties. The aim of this study was to evaluate the anti-remodeling properties of extracts from transformed and transgenic Leonurus sibiricus roots with transformed L. sibiricus roots extract with transcriptional factor AtPAP1 overexpression (AtPAP1). Two fibroblast cell lines, Wistar Institute-38 (WI-38) and human fetal lung fibroblast (HFL1), were incubated with extracts from transformed L. sibiricus roots (TR) and roots with transcriptional factor AtPAP1 over-expression (AtPAP1 TR). Additionally, remodeling conditions were induced in the cultures with rhinovirus 16 (HRV16). The expressions of metalloproteinase 9 (MMP)-9, tissue inhibitor of metalloproteinases 1 (TIMP-1), arginase I and transforming growth factor (TGF)-β were determined by quantitative polymerase chain reaction (qPCR) and immunoblotting methods. AtPAP1 TR decreased arginase I and MMP-9 expression with no effect on TIMP-1 or TGF-β mRNA expression. This extract also inhibited HRV16-induced expression of arginase I, MMP-9 and TGF-β in both cell lines (P < 0·05) Our study shows for the first time to our knowledge, that transformed AtPAP1 TR extract from L. sibiricus root may affect the remodeling process. Its effect can be attributed an increased amount of phenolic acids such as: chlorogenic acid, caffeic acid or ferulic acid and demonstrates the value of biotechnology in medicinal research.
Collapse
Affiliation(s)
- J Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| | - P Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz, Poland
| | - T Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz, Poland
| | - R Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Al-Azzam N, Teegala LR, Pokhrel S, Ghebreigziabher S, Chachkovskyy T, Thodeti S, Gavilanes I, Covington K, Thodeti CK, Paruchuri S. Transient Receptor Potential Vanilloid channel regulates fibroblast differentiation and airway remodeling by modulating redox signals through NADPH Oxidase 4. Sci Rep 2020; 10:9827. [PMID: 32555397 PMCID: PMC7299963 DOI: 10.1038/s41598-020-66617-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/22/2020] [Indexed: 12/31/2022] Open
Abstract
Asthma is characterized by pathological airway remodeling resulting from persistent myofibroblast activation. Although transforming growth factor beta 1 (TGFβ1), mechanical signals, and reactive oxygen species (ROS) are implicated in fibroblast differentiation, their integration is still elusive. We identified that Transient Receptor Potential Vanilloid 4 (TRPV4), a mechanosensitive ion channel mediates lung fibroblast (LF) differentiation and D. farinae-induced airway remodeling via a novel TRPV4-NADPH Oxidase 4 (NOX4) interaction. NOX4-mediated ROS production is essential for TGFβ1-induced LF differentiation via myocardin-related transcription factor-A (MRTF-A) and plasminogen activator inhibitor 1 (PAI-1). Importantly, TRPV4 inhibition prevented TGFβ1-induced NOX4 expression and ROS production. Both TRPV4 and NOX4 are activated by phosphatidylinositol 3-kinase (PI3K) downstream of TGFβ1, and signals from both TRPV4 and Rac are necessary for NOX4 upregulation. Notably, NOX4 expression is higher in fibroblasts derived from asthmatic patients (disease human LF; DHLF) in comparison to non-asthmatics (normal human LF; NHLF). Further, NOX4 expression is up-regulated in the lungs of D.farinae-treated wild type mice (WT) relative to saline-treated WT, which was attenuated in TRPV4 knockout (KO) mice. Our findings suggest that TRPV4 integrates TGFβ1 and ROS signaling through NOX4 and, TRPV4-NOX4 interaction is amenable to target lung remodeling during asthma.
Collapse
Affiliation(s)
- Nosayba Al-Azzam
- Department of Chemistry, University of Akron, Akron, OH, US.,Department of Physiology and Biochemistry, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Sabita Pokhrel
- Department of Chemistry, University of Akron, Akron, OH, US
| | | | | | - Sathwika Thodeti
- Department of Chemistry, University of Akron, Akron, OH, US.,Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, US
| | | | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, US
| | | |
Collapse
|
12
|
Wei B, Sun M, Shang Y, Zhang C, Jiao X. Neurokinin 1 receptor promotes rat airway smooth muscle cell migration in asthmatic airway remodelling by enhancing tubulin expression. J Thorac Dis 2018; 10:4849-4857. [PMID: 30233858 DOI: 10.21037/jtd.2018.07.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Airway remodelling is a major contributor to hyper-responsiveness leading to chronic asthma; however, the underlying mechanisms remain unclear. This study aimed to investigate the effects of a neurokinin 1 receptor (NK1R) antagonist (WIN62577) on the migration of airway smooth muscle cells (ASMCs) and the expression of NK1R and alpha-tubulin in airway remodelling using young rats with asthma. Methods Sprague-Dawley rats were randomly divided into a control group and airway remodelling group. Rats in the model group were stimulated with ovalbumin for 8 weeks. Primary ASMCs were cultured and purified from all rats, and then treated with different doses of WIN62577. The expression of NK1R and α-tubulin in ASMCs was assessed using immunofluorescence, real-time quantitative polymerase chain reaction, and western blotting. Changes in ASMC migration were detected by a transwell chamber assay. Results The transwell assay showed that the number of migrating ASMCs in the asthmatic airway remodelling group was significantly greater than that in the control group (P<0.01), which was inhibited by WIN62577 in a dose-dependent manner, with peak inhibition detected at 10-8 mol/L. The mRNA and protein expression levels of NK1R and α-tubulin were significantly higher in the asthmatic airway remodelling group than in the control group (P<0.05 and P<0.01, respectively), and were significantly decreased after treatment with WIN62577 (P<0.01 and P<0.05, respectively). Conclusions NK1R antagonists may suppress ASMC migration in a rat model of airway remodelling by inhibiting tubulin expression, indicating a new potential target for the treatment and control of chronic asthma.
Collapse
Affiliation(s)
- Bing Wei
- Department of Pediatrics, General Hospital of Shenyang Military Area Command, Shenyang 110016, China
| | - Mingwei Sun
- Department of First Clinical Medicine, China Medical University, Shenyang 110122, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Chao Zhang
- Department of Pediatrics, General Hospital of Shenyang Military Area Command, Shenyang 110016, China
| | - Xuyong Jiao
- Department of Pediatrics, General Hospital of Shenyang Military Area Command, Shenyang 110016, China
| |
Collapse
|
13
|
Royce SG, Nold MF, Bui C, Donovan C, Lam M, Lamanna E, Rudloff I, Bourke JE, Nold-Petry CA. Airway Remodeling and Hyperreactivity in a Model of Bronchopulmonary Dysplasia and Their Modulation by IL-1 Receptor Antagonist. Am J Respir Cell Mol Biol 2017; 55:858-868. [PMID: 27482635 DOI: 10.1165/rcmb.2016-0031oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic disease of extreme prematurity that has serious long-term consequences including increased asthma risk. We earlier identified IL-1 receptor antagonist (IL-1Ra) as a potent inhibitor of murine BPD induced by combining perinatal inflammation (intraperitoneal LPS to pregnant dams) and exposure of pups to hyperoxia (fraction of inspired oxygen = 0.65). In this study, we determined whether airway remodeling and hyperresponsiveness similar to asthma are evident in this model, and whether IL-1Ra is protective. During 28-day exposure to air or hyperoxia, pups received vehicle or 10 mg/kg IL-1Ra by daily subcutaneous injection. Lungs were then prepared for histology and morphometry of alveoli and airways, or for real-time PCR, or inflated with agarose to prepare precision-cut lung slices to visualize ex vivo intrapulmonary airway contraction and relaxation by phase-contrast microscopy. In pups reared under normoxic conditions, IL-1Ra treatment did not affect alveolar or airway structure or airway responses. Pups reared in hyperoxia developed a severe BPD-like lung disease, with fewer, larger alveoli, increased subepithelial collagen, and increased expression of α-smooth muscle actin and cyclin D1. After hyperoxia, methacholine elicited contraction with similar potency but with an increased maximum reduction in lumen area (air, 44%; hyperoxia, 89%), whereas dilator responses to salbutamol were maintained. IL-1Ra treatment prevented hyperoxia-induced alveolar disruption and airway fibrosis but, surprisingly, not the increase in methacholine-induced airway contraction. The current study is the first to demonstrate ex vivo airway hyperreactivity caused by systemic maternal inflammation and postnatal hyperoxia, and it reveals further preclinical mechanistic insights into IL-1Ra as a treatment targeting key pathophysiological features of BPD.
Collapse
Affiliation(s)
- Simon G Royce
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Marcel F Nold
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Christine Bui
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Chantal Donovan
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Maggie Lam
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Emma Lamanna
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Ina Rudloff
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Jane E Bourke
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Claudia A Nold-Petry
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
14
|
Pendergraff HM, Krishnamurthy PM, Debacker AJ, Moazami MP, Sharma VK, Niitsoo L, Yu Y, Tan YN, Haitchi HM, Watts JK. Locked Nucleic Acid Gapmers and Conjugates Potently Silence ADAM33, an Asthma-Associated Metalloprotease with Nuclear-Localized mRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:158-168. [PMID: 28918018 PMCID: PMC5498289 DOI: 10.1016/j.omtn.2017.06.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/15/2017] [Accepted: 06/16/2017] [Indexed: 12/22/2022]
Abstract
Two mechanisms dominate the clinical pipeline for oligonucleotide-based gene silencing, namely, the antisense approach that recruits RNase H to cleave target RNA and the RNAi approach that recruits the RISC complex to cleave target RNA. Multiple chemical designs can be used to elicit each pathway. We compare the silencing of the asthma susceptibility gene ADAM33 in MRC-5 lung fibroblasts using four classes of gene silencing agents, two that use each mechanism: traditional duplex small interfering RNAs (siRNAs), single-stranded small interfering RNAs (ss-siRNAs), locked nucleic acid (LNA) gapmer antisense oligonucleotides (ASOs), and novel hexadecyloxypropyl conjugates of the ASOs. Of these designs, the gapmer ASOs emerged as lead compounds for silencing ADAM33 expression: several gapmer ASOs showed subnanomolar potency when transfected with cationic lipid and low micromolar potency with no toxicity when delivered gymnotically. The preferential susceptibility of ADAM33 mRNA to silencing by RNase H may be related to the high degree of nuclear retention observed for this mRNA. Dynamic light scattering data showed that the hexadecyloxypropyl ASO conjugates self-assemble into clusters. These conjugates showed reduced potency relative to unconjugated ASOs unless the lipophilic tail was conjugated to the ASO using a biocleavable linkage. Finally, based on the lead ASOs from (human) MRC-5 cells, we developed a series of homologous ASOs targeting mouse Adam33 with excellent activity. Our work confirms that ASO-based gene silencing of ADAM33 is a useful tool for asthma research and therapy.
Collapse
Affiliation(s)
- Hannah M Pendergraff
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Pranathi Meda Krishnamurthy
- RNA Therapeutics Institute, UMass Medical School, Worcester, MA 01605, USA; Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605, USA
| | - Alexandre J Debacker
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; RNA Therapeutics Institute, UMass Medical School, Worcester, MA 01605, USA; Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605, USA
| | - Michael P Moazami
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; RNA Therapeutics Institute, UMass Medical School, Worcester, MA 01605, USA; Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605, USA
| | - Vivek K Sharma
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; RNA Therapeutics Institute, UMass Medical School, Worcester, MA 01605, USA; Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605, USA
| | - Liisa Niitsoo
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Yong Yu
- Institute of Materials Research and Engineering, A*STAR, Singapore 138634, Singapore
| | - Yen Nee Tan
- Institute of Materials Research and Engineering, A*STAR, Singapore 138634, Singapore
| | - Hans Michael Haitchi
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; NIHR Southampton Respiratory Biomedical Research Unit at University Hospital Southampton NHS Foundation Trust, Southampton, Southampton SO16 6YD, UK.
| | - Jonathan K Watts
- Department of Chemistry, University of Southampton, Southampton SO17 1BJ, UK; Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; RNA Therapeutics Institute, UMass Medical School, Worcester, MA 01605, USA; Department of Biochemistry and Molecular Pharmacology, UMass Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
15
|
Liu MW, Liu R, Wu HY, Chen M, Dong MN, Huang YQ, Zhang CH, Wang YZ, Xia J, Shi Y, Xie FM, Luo H, Zhao XY, Wei W, Su MX. Atorvastatin has a protective effect in a mouse model of bronchial asthma through regulating tissue transglutaminase and triggering receptor expressed on myeloid cells-1 expression. Exp Ther Med 2017; 14:917-930. [PMID: 28810543 PMCID: PMC5526119 DOI: 10.3892/etm.2017.4576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023] Open
Abstract
Airway remodeling in asthma contributes to airway hyperreactivity, loss of lung function and persistent symptoms. Current therapies do not adequately treat the structural airway changes associated with asthma. Statin drugs have improved respiratory health and their therapeutic potential in asthma has been tested in clinical trials. However, the mechanism of action of statins in this context has remained elusive. The present study hypothesized that atorvastatin treatment of ovalbumin-exposed mice attenuates early features of airway remodeling via a mevalonate-dependent mechanism. BALB/c mice were sensitized with ovalbumin and atorvastatin was delivered via oral gavage prior to each ovalbumin exposure. Reverse transcription-semi-quantitative polymerase chain reaction (RT-semi-qPCR), ELISA and western blot analysis were used to assess the expression of a number of relevant genes, including tissue transglutaminase (tTG), triggering receptor expressed on myeloid cells (TREM)-1, nuclear factor erythroid 2-related factor (Nrf) 2, hypoxia-inducible factor (HIF)-1α, transforming growth factor (TGF)-β1, matrix metalloproteinase (MMP)-9 and tissue inhibitors of metalloproteinases (TIMP)-1 in lung tissue. α-Smooth muscle actin (α-SMA) activity was measured by immunohistochemistry. Airway hyperresponsiveness, lung collagen deposition, airway wall area, airway smooth muscle thickness and lung pathology were also assessed. Atorvastatin treatment led to downregulation of tTG and TREM-1 expression in lung tissue after ovalbumin sensitization, blocked the activity of MMP-9, vascular endothelial growth factor, nuclear factor-κB p65, α-SMA, HIF-α and TGF-β1 and up-regulated Nrf2 expression. Furthermore, the number of lymphocytes and eosinophils in the atorvastatin group was significantly lower than that in the control group. In addition, airway hyperresponsiveness, lung collagen deposition, airway wall area, airway smooth muscle thickness and pathological changes in the lung were significantly decreased in the atorvastatin group, and tumor necrosis factor-α, interleukin (IL)-8, IL-13 and IL-17 in serum were significantly decreased. Histological results demonstrated the attenuating effect of atorvastatin on ovalbumin-induced airway remodeling in asthma. In conclusion, the present study indicated that atorvastatin significantly alleviated ovalbumin-induced airway remodeling in asthma by downregulating tTG and TREM-1 expression. The marked protective effects of atorvastatin suggest its therapeutic potential in ovalbumin-induced airway remodeling in asthma treatment.
Collapse
Affiliation(s)
- Ming-Wei Liu
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Rong Liu
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Hai-Ying Wu
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Mei Chen
- Department of Respiratory Medicine, The Yan'An Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, P.R. China
| | - Min-Na Dong
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yun-Qiao Huang
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Chun-Hai Zhang
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yin-Zhong Wang
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jing Xia
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yang Shi
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Feng-Mei Xie
- Department of Gastroenterology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650106, P.R. China
| | - Hua Luo
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xin-Yuan Zhao
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wei Wei
- Department of Emergency, The First Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Mei-Xian Su
- Department of Emergency, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650106, P.R. China
| |
Collapse
|
16
|
Thymic stromal lymphopoietin and apocynin alter the expression of airway remodeling factors in human rhinovirus-infected cells. Immunobiology 2017; 222:892-899. [PMID: 28545810 DOI: 10.1016/j.imbio.2017.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/21/2017] [Accepted: 05/14/2017] [Indexed: 12/30/2022]
Abstract
Airway remodeling is a characteristic of bronchial asthma. The process involves the expression of many genes, such as transforming growth factor-beta (TGF-β), tissue inhibitors of metalloproteinases (TIMP-1), MMP and arginase. Human rhinovirus (HRV) is known to cause asthma exacerbations, and viral infections might be involved in the development of airway remodeling. Therefore, the aim of this study was to determine the influence of HRV on the genes involved in airway remodeling and to examine the impact of thymic stromal lymphopoietin (TSLP) and contribution of oxidative stress on airway remodeling in the context of HRV infection. Peripheral blood mononuclear cells, isolated from blood collected from 10 healthy volunteers, and human lung fibroblasts were infected with HRV-16. The cells were treated with apocynin or TSLP 48h after infection. The expression of TGF-β1, TIMP-1 and arginase I mRNA and protein were determined by real-time PCR, immunoblotting and ELISA, respectively. Rhinovirus infection significantly increased the expression of TGF-β1 and arginase I, on the mRNA and protein levels. This effect was inhibited by apocynin, though only on the mRNA level. TIMP-1 expression was not influenced by HRV; however, apocynin caused a significant increase of TIMP-1 mRNA expression. TSLP increased the expression of TGF-β1 and arginase I mRNA in fibroblasts, but not in PBMC.
Collapse
|
17
|
Gombedza F, Kondeti V, Al-Azzam N, Koppes S, Duah E, Patil P, Hexter M, Phillips D, Thodeti CK, Paruchuri S. Mechanosensitive transient receptor potential vanilloid 4 regulates Dermatophagoides farinae-induced airway remodeling via 2 distinct pathways modulating matrix synthesis and degradation. FASEB J 2017; 31:1556-1570. [PMID: 28073835 DOI: 10.1096/fj.201601045r] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022]
Abstract
Contributions of mechanical signals to airway remodeling during asthma are poorly understood. Transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel, has been implicated in cardiac and pulmonary fibrosis; however, its role in asthma remains elusive. Employing a Dermatophagoides farinae-induced asthma model, we report here that TRPV4-knockout mice were protected from D. farinae-induced airway remodeling. Furthermore, lung fibroblasts that were isolated from TRPV4-knockout mice showed diminished differentiation potential compared with wild-type mice. Fibroblasts from asthmatic lung exhibited increased TRPV4 activity and enhanced differentiation potential compared with normal human lung fibroblasts. Of interest, TGF-β1 treatment enhanced TRPV4 activation in a PI3K-dependent manner in normal human lung fibroblasts in vitro Mechanistically, TRPV4 modulated matrix remodeling in the lung via 2 distinct but dependent pathways: one enhances matrix deposition by fibrotic gene activation, whereas the other slows down matrix degradation by increased plasminogen activator inhibitor 1. Of importance, both pathways are regulated by Rho/myocardin-related transcription factor-A and contribute to fibroblast differentiation and matrix remodeling in the lung. Thus, our results support a unique role for TRPV4 in D. farinae-induced airway remodeling and warrant further studies in humans for it to be used as a novel therapeutic target in the treatment of asthma.-Gombedza, F., Kondeti, V., Al-Azzam, N., Koppes, S., Duah, E., Patil, P., Hexter, M., Phillips, D., Thodeti, C. K., Paruchuri, S. Mechanosensitive transient receptor potential vanilloid 4 regulates Dermatophagoides farinae-induced airway remodeling via 2 distinct pathways modulating matrix synthesis and degradation.
Collapse
Affiliation(s)
- Farai Gombedza
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Vinay Kondeti
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Nosayba Al-Azzam
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Stephanie Koppes
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Ernest Duah
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Prachi Patil
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Madison Hexter
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Daniel Phillips
- Department of Chemistry, University of Akron, Akron, Ohio, USA; and
| | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | | |
Collapse
|
18
|
Chen J, Deng L, Dreymüller D, Jiang X, Long J, Duan Y, Wang Y, Luo M, Lin F, Mao L, Müller B, Koller G, Bartsch JW. A novel peptide ADAM8 inhibitor attenuates bronchial hyperresponsiveness and Th2 cytokine mediated inflammation of murine asthmatic models. Sci Rep 2016; 6:30451. [PMID: 27458083 PMCID: PMC4960557 DOI: 10.1038/srep30451] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/06/2016] [Indexed: 12/31/2022] Open
Abstract
A disintegrin and metalloproteinase 8 (ADAM8) has been identified as a signature gene associated with moderate and severe asthma. Studies in mice have demonstrated that the severity of asthma can be reduced by either transgenic knock-out or by antibodies blocking ADAM8 function, highlighting ADAM8 as potential drug target for asthma therapy. Here, we examined the therapeutic effect of an ADAM8 inhibitor peptide (BK-1361) that specifically blocks cellular ADAM8 activity in ovalbumin-sensitized and challenged Balb/c mice. We found that BK-1361 (25 μg/g body weight) attenuated airway responsiveness to methacholine stimulation by up to 42%, concomitantly reduced tissue remodeling by 50%, and decreased inflammatory cells (e.g. eosinophils down by 54%)/inflammatory factors (e.g. sCD23 down by 50%)/TH2 cytokines (e.g. IL-5 down by 70%)/ADAM8-positive eosinophils (down by 60%) in the lung. We further verified that BK-1361 specifically targets ADAM8 in vivo as the peptide caused significantly reduced levels of soluble CD23 in wild-type but not in ADAM8-deficient mice. These findings suggest that BK-1361 blocks ADAM8-dependent asthma effects in vivo by inhibiting infiltration of eosinophils and TH2 lymphocytes, thus leading to reduction of TH2-mediated inflammation, tissue remodeling and bronchial hyperresponsiveness. Taken together, pharmacological ADAM8 inhibition appears as promising novel therapeutic strategy for the treatment of asthma.
Collapse
Affiliation(s)
- Jun Chen
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China.,Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China.,Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Daniela Dreymüller
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Xuemei Jiang
- Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Jiaoyue Long
- Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Yiyuan Duan
- Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Yue Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China
| | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, China
| | - Feng Lin
- Key Lab of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China
| | - Lizhen Mao
- Jiangsu Asialand Bio-med Technology Co. Ltd., Changzhou, Jiangsu, China
| | - Bernd Müller
- Laboratory of Respiratory Cell Biology, Division of Pneumology, Philipps-University Marburg, Marburg, Germany
| | - Garrit Koller
- KCLDI Biomaterials, Biomimetics and Biophotonics Group. King's College London, London SE1 9RT, United Kingdom.,Department of Neurosurgery, Philipps-University Marburg, Baldinger Str., 35033 Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps-University Marburg, Baldinger Str., 35033 Marburg, Germany
| |
Collapse
|
19
|
Patel KP, Giraud AS, Samuel CS, Royce SG. Combining an epithelial repair factor and anti-fibrotic with a corticosteroid offers optimal treatment for allergic airways disease. Br J Pharmacol 2016; 173:2016-29. [PMID: 27060978 DOI: 10.1111/bph.13494] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE We evaluated the extent to which individual versus combination treatments that specifically target airway epithelial damage [trefoil factor-2 (TFF2)], airway fibrosis [serelaxin (RLX)] or airway inflammation [dexamethasone (DEX)] reversed the pathogenesis of chronic allergic airways disease (AAD). EXPERIMENTAL APPROACH Following induction of ovalbumin (OVA)-induced chronic AAD in 6–8 week female Balb/c mice, animals were i.p. administered naphthalene (NA) on day 64 to induce epithelial damage, then received daily intranasal administration of RLX (0.8 mg·mL(−1)), TFF2 (0.5 mg·mL(−1)), DEX (0.5 mg·mL(−1)), RLX + TFF2 or RLX + TFF2 + DEX from days 67–74. On day 75, lung function was assessed by invasive plethysmography, before lung tissue was isolated for analyses of various measures. The control group was treated with saline + corn oil (vehicle for NA). KEY RESULTS OVA + NA-injured mice demonstrated significantly increased airway inflammation, airway remodelling (AWR) (epithelial damage/thickness; subepithelial myofibroblast differentiation, extracellular matrix accumulation and fibronectin deposition; total lung collagen concentration), and significantly reduced airway dynamic compliance (cDyn). RLX + TFF2 markedly reversed several measures of OVA + NA-induced AWR and normalized the reduction in cDyn. The combined effects of RLX + TFF2 + DEX significantly reversed peribronchial inflammation score, airway epithelial damage, subepithelial extracellular matrix accumulation/fibronectin deposition and total lung collagen concentration (by 50–90%) and also normalized the reduction of cDyn. CONCLUSIONS AND IMPLICATIONS Combining an epithelial repair factor and anti-fibrotic provides an effective means of treating the AWR and dysfunction associated with AAD/asthma and may act as an effective adjunct therapy to anti-inflammatory corticosteroids
Collapse
Affiliation(s)
- K P Patel
- Fibrosis Laboratory, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Vic., Australia
| | - A S Giraud
- Murdoch Children's Research Institute, University of Melbourne, Vic., Australia.,Department of Paediatrics, University of Melbourne, Vic., Australia
| | - C S Samuel
- Fibrosis Laboratory, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Vic., Australia
| | - S G Royce
- Fibrosis Laboratory, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Vic., Australia.,Respiratory Pharmacology Laboratory, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Vic., Australia
| |
Collapse
|
20
|
Gao T, Ng CL, Li C, Li YY, Duan C, Shen L, Jiao YF, Liu M, Wang D. Smoking is an independent association of squamous metaplasia in Chinese nasal polyps. Int Forum Allergy Rhinol 2015; 6:66-74. [DOI: 10.1002/alr.21631] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 07/11/2015] [Accepted: 07/21/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Tian Gao
- Department of Otolaryngology; Second Affiliated Hospital of Harbin Medical University; Heilongjiang China
| | - Chew Lip Ng
- Department of Otolaryngology; National University of Singapore, National University Health System; Singapore
| | - Chunwei Li
- Department of Otolaryngology; National University of Singapore, National University Health System; Singapore
| | - Ying Ying Li
- Department of Otolaryngology; National University of Singapore, National University Health System; Singapore
| | - Chen Duan
- Department of Otolaryngology; National University of Singapore, National University Health System; Singapore
| | - Liang Shen
- Biostatistics Unit, National University of Singapore; National University Health System; Singapore
| | - Yu Fei Jiao
- Department of Pathology; Second Affiliated Hospital of Harbin Medical University; Heilongjiang China
| | - Ming Liu
- Department of Otolaryngology; Second Affiliated Hospital of Harbin Medical University; Heilongjiang China
| | - De Yun Wang
- Department of Otolaryngology; National University of Singapore, National University Health System; Singapore
| |
Collapse
|
21
|
Impaired Cell Cycle Regulation in a Natural Equine Model of Asthma. PLoS One 2015; 10:e0136103. [PMID: 26292153 PMCID: PMC4546272 DOI: 10.1371/journal.pone.0136103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 07/29/2015] [Indexed: 12/14/2022] Open
Abstract
Recurrent airway obstruction (RAO) is a common and potentially debilitating lower airway disease in horses, which shares many similarities with human asthma. In susceptible horses RAO exacerbation is caused by environmental allergens and irritants present in hay dust. The objective of this study was the identification of genes and pathways involved in the pathology of RAO by global transcriptome analyses in stimulated peripheral blood mononuclear cells (PBMCs). We performed RNA-seq on PBMCs derived from 40 RAO affected and 45 control horses belonging to three cohorts of Warmblood horses: two half-sib families and one group of unrelated horses. PBMCs were stimulated with hay dust extract, lipopolysaccharides, a recombinant parasite antigen, or left unstimulated. The total dataset consisted of 561 individual samples. We detected significant differences in the expression profiles between RAO and control horses. Differential expression (DE) was most marked upon stimulation with hay dust extract. An important novel finding was a strong upregulation of CXCL13 together with many genes involved in cell cycle regulation in stimulated samples from RAO affected horses, in addition to changes in the expression of several HIF-1 transcription factor target genes. The RAO condition alters systemic changes observed as differential expression profiles of PBMCs. Those changes also depended on the cohort and stimulation of the samples and were dominated by genes involved in immune cell trafficking, development, and cell cycle regulation. Our findings indicate an important role of CXCL13, likely macrophage or Th17 derived, and the cell cycle regulator CDC20 in the immune response in RAO.
Collapse
|
22
|
Royce SG, Lim CXF, Patel KP, Wang B, Samuel CS, Tang MLK. Intranasally administered serelaxin abrogates airway remodelling and attenuates airway hyperresponsiveness in allergic airways disease. Clin Exp Allergy 2015; 44:1399-408. [PMID: 25113628 DOI: 10.1111/cea.12391] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 03/03/2014] [Accepted: 05/08/2014] [Indexed: 01/24/2023]
Abstract
BACKGROUND The peptide hormone relaxin plays a key role in the systemic hemodynamic and renovascular adaptive changes that occur during pregnancy, which is linked to its antiremodelling effects. Serelaxin (a recombinant form of human gene-2 relaxin) has been shown to inhibit lung fibrosis in various disease models and reverse airway remodelling and airway hyperresponsiveness (AHR) in allergic airways disease (AAD). OBJECTIVE Although continuous systemic delivery of exogenous serelaxin alleviates allergic fibrosis and AHR, more direct routes for administration into the lung have not been investigated. Thus, intranasal administration of serelaxin was evaluated for its ability to reverse airway remodelling and AHR associated with AAD. METHODS Female Balb/c mice were subjected to a 9-week model of chronic AAD. Subgroups of animals (n = 12/group) were then treated intranasally with serelaxin (0.8 mg/mL) or vehicle once daily for 14 days (from weeks 9-11). Saline-sensitized/challenged mice treated with intranasal saline served as additional controls. Differential bronchoalveolar lavage (BAL) cell counts, ovalbumin (OVA)-specific IgE levels, tissue inflammation, parameters of airway remodelling and AHR were then assessed. RESULTS Chronic AAD was associated with significant increases in differential BAL cell counts, OVA-specific IgE levels, inflammation, epithelial thickening, goblet cell metaplasia, TGF-β1 expression, epithelial Smad2 phosphorylation (pSmad2), subepithelial collagen thickness, total lung collagen concentration and AHR (all P < 0.05 vs. respective measurements from saline-treated mice). Daily intranasal delivery of serelaxin significantly diminished AAD-induced epithelial thickening, epithelial pSmad2, subepithelial and total lung collagen content (fibrosis) and AHR (all P < 0.05 vs. vehicle-treated AAD mice). CONCLUSIONS AND CLINICAL RELEVANCE Intranasal delivery of serelaxin can effectively reduce airway remodelling and AHR, when administered once daily. Respirable preparations of serelaxin may have therapeutic potential for the prevention and/or reversal of established airway remodelling and AHR in asthma.
Collapse
Affiliation(s)
- S G Royce
- Allergy and Immune Disorders, Murdoch Children's Research Institute, Melbourne, Vic., Australia; Department of Pharmacology, Monash University, Melbourne, Vic., Australia
| | | | | | | | | | | |
Collapse
|
23
|
Characterization of a novel model incorporating airway epithelial damage and related fibrosis to the pathogenesis of asthma. J Transl Med 2014; 94:1326-39. [PMID: 25264707 DOI: 10.1038/labinvest.2014.119] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 08/05/2014] [Accepted: 08/29/2014] [Indexed: 12/26/2022] Open
Abstract
Asthma develops from injury to the airways/lungs, stemming from airway inflammation (AI) and airway remodeling (AWR), both contributing to airway hyperresponsiveness (AHR). Airway epithelial damage has been identified as a new etiology of asthma but is not targeted by current treatments. Furthermore, it is poorly studied in currently used animal models of AI and AWR. Therefore, this study aimed to incorporate epithelial damage/repair with the well-established ovalbumin (OVA)-induced model of chronic allergic airway disease (AAD), which presents with AI, AWR, and AHR, mimicking several features of human asthma. A 3-day naphthalene (NA)-induced model of epithelial damage/repair was superimposed onto the 9-week OVA-induced model of chronic AAD, before 6 weeks of OVA nebulization (NA+OVA group), during the second last OVA nebulization period (OVA/NA group) or 1 day after the 6-week OVA nebulization period (OVA+NA group), using 6-8-week-old female Balb/c mice (n=6-12/group). Mice subjected to the 9-week OVA model, 3-day NA model or respective vehicle treatments (saline and corn oil) were used as appropriate controls. OVA alone significantly increased epithelial thickness and apoptosis, goblet cell metaplasia, TGF-β1, subepithelial collagen (assessed by morphometric analyses of various histological stains), total lung collagen (hydroxyproline analysis), and AHR (invasive plethysmography) compared with that in saline-treated mice (all P<0.05 vs saline treatment). NA alone caused a significant increase in epithelial denudation and apoptosis, TGF-β1, subepithelial, and total lung collagen compared with respective measurements from corn oil-treated controls (all P<0.01 vs corn oil treatment). All three combined models underwent varying degrees of epithelial damage and AWR, with the OVA+NA model demonstrating the greatest increase in subepithelial/total lung collagen and AHR (all P<0.05 vs OVA alone or NA alone). These combined models of airway epithelial damage/AAD demonstrated that epithelial damage is a key contributor to AWR, fibrosis and related AHR, and augments the effects of AI on these parameters.
Collapse
|
24
|
Chen J, Jiang X, Duan Y, Long J, Bartsch JW, Deng L. ADAM8 in asthma. Friend or foe to airway inflammation? Am J Respir Cell Mol Biol 2014; 49:875-84. [PMID: 23837412 DOI: 10.1165/rcmb.2013-0168tr] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway inflammation has been suggested as the pathological basis in asthma pathogenesis. Recruitment of leukocytes from the vasculature into airway sites is essential for induction of airway inflammation, a process thought to be mediated by a disintegrin and metalloprotease 8 (ADAM8). However, there is an apparent controversy about whether ADAM8 helps or hampers transmigration of leukocytes through endothelium in airway inflammation of asthma. This review outlines the current contradictory concepts concerning the role of ADAM8 in airway inflammation, particularly focusing on the recruitment of leukocytes during asthma, and attempts to bridge the existing experimental data on the basis of the functional analysis of different domains of ADAM8 and their endogenous processing in vivo. We suggest a possible hypothesis for the specific mechanism by which ADAM8 regulates the transmigration of leukocytes to explain the disparity existing in current studies, and we also raise some questions that require future investigations.
Collapse
Affiliation(s)
- Jun Chen
- 1 Key Lab of Biorheological Science and Technology, Ministry of Education, "National 985 Project" Institute of Biorheology and Gene Regulation, Bioengineering College, Chongqing University, Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
25
|
Young IH, Bye PTP. Gas exchange in disease: asthma, chronic obstructive pulmonary disease, cystic fibrosis, and interstitial lung disease. Compr Physiol 2013; 1:663-97. [PMID: 23737199 DOI: 10.1002/cphy.c090012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ventilation-perfusion (VA/Q) inequality is the underlying abnormality determining hypoxemia and hypercapnia in lung diseases. Hypoxemia in asthma is characterized by the presence of low VA/Q units, which persist despite improvement in airway function after an attack. This hypoxemia is generally attenuated by compensatory redistribution of blood flow mediated by hypoxic vasoconstriction and changes in cardiac output, however, mediator release and bronchodilator therapy may cause deterioration. Patients with chronic obstructive pulmonary disease have more complex patterns of VA/Q inequality, which appear more fixed, and changes in blood flow and ventilation have less benefit in improving gas exchange efficiency. The inability of ventilation to match increasing cardiac output limits exercise capacity as the disease progresses. Deteriorating hypoxemia during exacerbations reflects the falling mixed venous oxygen tension from increased respiratory muscle activity, which is not compensated by any redistribution of VA/Q ratios. Shunt is not a feature of any of these diseases. Patients with cystic fibrosis (CF) have no substantial shunt when managed according to modern treatment regimens. Interstitial lung diseases demonstrate impaired oxygen diffusion across the alveolar-capillary barrier, particularly during exercise, although VA/Q inequality still accounts for most of the gas exchange abnormality. Hypoxemia may limit exercise capacity in these diseases and in CF. Persistent hypercapnic respiratory failure is a feature of advancing chronic obstructive pulmonary disease and CF, closely associated with sleep disordered breathing, which is not a prominent feature of the other diseases. Better understanding of the mechanisms of hypercapnic respiratory failure, and of the detailed mechanisms controlling the distribution of ventilation and blood flow in the lung, are high priorities for future research.
Collapse
Affiliation(s)
- Iven H Young
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, and The University of Sydney, Australia.
| | | |
Collapse
|
26
|
Royce SG, Moodley Y, Samuel CS. Novel therapeutic strategies for lung disorders associated with airway remodelling and fibrosis. Pharmacol Ther 2013; 141:250-60. [PMID: 24513131 DOI: 10.1016/j.pharmthera.2013.10.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/01/2013] [Indexed: 01/11/2023]
Abstract
Inflammatory cell infiltration, cytokine release, epithelial damage, airway/lung remodelling and fibrosis are central features of inflammatory lung disorders, which include asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome and idiopathic pulmonary fibrosis. Although the lung has some ability to repair itself from acute injury, in the presence of ongoing pathological stimuli and/or insults that lead to chronic disease, it no longer retains the capacity to heal, resulting in fibrosis, the final common pathway that causes an irreversible loss of lung function. Despite inflammation, genetic predisposition/factors, epithelial-mesenchymal transition and mechanotransduction being able to independently contribute to airway remodelling and fibrosis, current therapies for inflammatory lung diseases are limited by their ability to only target the inflammatory component of the disease without having any marked effects on remodelling (epithelial damage and fibrosis) that can cause lung dysfunction independently of inflammation. Furthermore, as subsets of patients suffering from these diseases are resistant to currently available therapies (such as corticosteroids), novel therapeutic approaches are required to combat all aspects of disease pathology. This review discusses emerging therapeutic approaches, such as trefoil factors, relaxin, histone deacetylase inhibitors and stem cells, amongst others that have been able to target airway inflammation and airway remodelling while improving related lung dysfunction. A better understanding of the mode of action of these therapies and their possible combined effects may lead to the identification of their clinical potential in the setting of lung disease, either as adjunct or alternative therapies to currently available treatments.
Collapse
Affiliation(s)
- Simon G Royce
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Departments of Pathology and Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Yuben Moodley
- Department of Respiratory and Sleep Medicine, School of Medicine and Pharmacology, Royal Perth Hospital, University of Western Australia, Perth 6000, Western Australia, Australia
| | - Chrishan S Samuel
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3010, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
27
|
Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo. Int J Mol Med 2013; 32:629-36. [PMID: 23807697 DOI: 10.3892/ijmm.2013.1425] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 06/20/2013] [Indexed: 11/05/2022] Open
Abstract
The inhibition of the proliferation of airway smooth muscle cells (ASMCs) is crucial for the prevention and treatment of asthma. Recent studies have revealed some important functions of curcumin; however, its effects on the proliferation of ASMCs in asthma remain unknown. Therefore, in this study, we performed in vitro and in vivo experiments to investigate the effects of curcumin on the proliferation of ASMCs in asthma. The thickness of the airway wall, the airway smooth muscle layer, the number of ASMCs and the expression of extracellular signal-regulated kinase (ERK) were significantly reduced in the curcumin-treated group as compared with the model group. Curcumin inhibited the cell proliferation induced by platelet-derived growth factor (PDGF) and decreased the PDGF-induced phosphorylation of ERK1/2 in the rat ASMCs. Moreover, the disruption of caveolae using methyl-β-cyclodextrin (MβCD) attenuated the anti-proliferative effects of curcumin in the ASMCs, which suggests that caveolin is involved in this process. Curcumin upregulated the mRNA and protein expression of caveolin-1. The data presented in this study demonstrate that the proliferation of ASMCs is inhibited by curcumin in vitro and in vivo; curcumin exerts these effects by upregulating the expression of caveolin-1 and blocking the activation of the ERK pathway.
Collapse
|
28
|
Karimi K, Forsythe P. Natural killer cells in asthma. Front Immunol 2013; 4:159. [PMID: 23801996 PMCID: PMC3689036 DOI: 10.3389/fimmu.2013.00159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 06/09/2013] [Indexed: 12/23/2022] Open
Abstract
The worldwide prevalence, morbidity, and mortality of asthma have dramatically increased over the last few decades and there is a clear need to identify new effective therapeutic and prophylactic strategies. Despite high numbers of NK cells in the lung and their ability to generate a variety of immunomodulatory mediators, the potential of NK cells as therapeutic targets in allergic airway disease has been largely overlooked. The fact that IgE, acting through FcγRIII, can activate NK cells resulting in cytokine/chemokine production implies that NK cells may contribute to IgE-mediated allergic responses. Indeed, current evidence suggests that NK cells can promote allergic airway responses during sensitization and ongoing inflammation. In animal models, increased NK cells are observed in the lung following antigen challenge and depletion of the cells before immunization inhibits allergic airway inflammation. Moreover, in asthmatics, NK cell phenotype is altered and may contribute to the promotion of a pro-inflammatory Th2-type environment. Conversely, driving NK cells toward an IFN-γ-secreting phenotype can reduce features of the allergic airway response in animal models. However, we have limited knowledge of the signals that drive the development of distinct subsets and functional phenotypes of NK cells in the lung and thus the role and therapeutic potential of NK cells in the allergic airway remains unclear. Here we review the potentially diverse role of NK cells in allergic airway disease, identify gaps in current knowledge, and discuss the potential of modulating NK cell function as a treatment strategy in asthma.
Collapse
Affiliation(s)
- Khalil Karimi
- Institut für Experimentelle Immunologie und Hepatologie, Universitätsklinikum Hamburg-Eppendorf , Hamburg , Germany ; Department of Medicine, Brain-Body Institute, McMaster University , Hamilton, ON , Canada
| | | |
Collapse
|
29
|
Sexton P, Black P, Wu L, Sommerville F, Hamed M, Metcalf P, Kolbe J. Fixed airflow obstruction among nonsmokers with asthma:a case-comparison study. J Asthma 2013; 50:606-12. [PMID: 23574362 DOI: 10.3109/02770903.2013.793706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Airflow obstruction in asthma is usually reversible, but fixed obstruction develops in some individuals. Little is known about risk factors for development of fixed airflow obstruction in nonsmokers with asthma. METHODS This case-comparison study recruited nonsmokers aged over 45 years with physician-diagnosed asthma from specialist outpatient clinics and primary care. Two age-matched groups were recruited on the basis of spirometry: anobstructed group (post-bronchodilator FEV(1) ≤ 70% predicted, FEV1/FVC ratio < lower limit of normal) and a control group with normal lung function. Subjects completed a questionnaire and interview, and underwent spirometry, venesection, exhaled nitric oxide (ENO) measurement, allergen skinprick testing, and formal lung function testing. RESULTS Thirty-four obstructed subjects and 40 controls participated in the study. Obstructed subjects exhibited greater evidence of systemic inflammation, abnormal glucose homeostasis, and central obesity than controls. Obstructed subjects reported longer duration of asthma, and childhood respiratory infection was commoner in that group. Metabolic syndrome prevalence was similar between groups, but several features of insulin resistance were associated with reduced FEV(1). Cough and sputum were common among controls. CONCLUSIONS Risk of fixed airflow obstruction may correlate with lifetime asthma duration. Individuals with coexisting asthma and fixed airflow obstruction have heightened systemic inflammation. A variety of chronic respiratory symptoms are common among "healthy" nonsmokers with asthma.
Collapse
Affiliation(s)
- Paul Sexton
- Department of Medicine, University of Auckland, Auckland, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
30
|
Leonardi S, Coco A, Del Giudice MM, Marseglia GL, La Rosa M. The airway epithelium dysfunction in the pathogenesis of asthma: The evidence. Health (London) 2013. [DOI: 10.4236/health.2013.52a044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Tefé-Silva C, Beneli CT, Celes MR, Machado ER, Ueta MT, Sorgi CA, Floriano EM, Faccioli LH, Ramos SG. Dexamethasone reduces bronchial wall remodeling during pulmonary migration of Strongyloides venezuelensis larvae in rats. Parasitol Int 2012; 61:425-30. [PMID: 22808527 DOI: 10.1016/j.parint.2012.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Strongyloidiasis is an intestinal parasitosis with an obligatory pulmonary cycle. A Th2-type immune response is induced and amplifies the cellular response through the secretion of inflammatory mediators. Although this response has been described as being similar to asthma, airway remodeling during pulmonary migration of larvae has not yet been established. The aim of this study was to identify the occurrence of airway remodeling during Strongyloides venezuelensis (S. v.) infection and to determine the ability of dexamethasone treatment to interfere with the mechanisms involved in this process. Rats were inoculated with 9,000 S. v. larvae, treated with dexamethasone (2 mg/kg) and killed at 1, 3, 5, 7, 14 and 21 days. Morphological and morphometric analyzes with routine stains and immunohistochemistry were conducted, and some inflammatory mediators were evaluated using ELISA. Goblet cell hyperplasia and increased bronchiolar thickness, characterized by edema, neovascularization, inflammatory infiltrate, collagen deposition and enlargement of the smooth muscle cell layer were observed. VEGF, IL1-β and IL-4 levels were elevated throughout the course of the infection. The morphological findings and the immunomodulatory response to the infection were drastically reduced in dexamethasone-treated rats. The pulmonary migration of S. venezuelensis larvae produced a transitory, but significant amount of airway remodeling with a slight residual bronchiolar fibrosis. The exact mechanisms involved in this process require further study.
Collapse
Affiliation(s)
- Cristiane Tefé-Silva
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Royce SG, Dang W, Yuan G, Tran J, El-Osta A, Karagiannis TC, Tang MLK. Effects of the histone deacetylase inhibitor, trichostatin A, in a chronic allergic airways disease model in mice. Arch Immunol Ther Exp (Warsz) 2012; 60:295-306. [PMID: 22684086 DOI: 10.1007/s00005-012-0180-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2011] [Accepted: 02/20/2012] [Indexed: 01/26/2023]
Abstract
There is a need for new asthma therapies that can concurrently address airway remodeling, airway hyperresponsiveness and progressive irreversible loss of lung function, in addition to inhibiting inflammation. Histone deacetylase inhibitors (HDACi) alter gene expression by interfering with the removal of acetyl groups from histones. The HDACi trichostatin A (TSA) has pleiotropic effects targeting key pathological processes in asthma including inflammation, proliferation, angiogenesis and fibrosis. The aim was to evaluate the effects of TSA treatment in a mouse model of chronic allergic airways disease (AAD). Wild-type BALB/c mice with AAD were treated intraperitoneally with 5 mg/kg TSA or vehicle control. Airway inflammation was assessed by bronchoalveolar lavage fluid (BALF) cell counts and histological examination of lung tissue sections. Remodeling was assessed by morphometric analysis and airway hyperresponsiveness was assessed by invasive plethysmography. TSA-treated mice had a reduced number of total inflammatory cells and eosinophils within the BALF as compared to vehicle-treated mice (both p < 0.05). Furthermore, airway remodeling changes were significantly reduced with TSA compared to vehicle-treated mice, with fewer goblet cells (p < 0.05), less subepithelial collagen deposition (p < 0.05) and attenuated airway hyperresponsiveness at the highest methacholine dose. These findings demonstrate that treatment with an HDACi can concurrently reduce structural airway remodeling changes and airway hyperresponsiveness, in addition to attenuating airway inflammation in a chronic AAD model. This has important implications for the development of novel treatments for severe asthma.
Collapse
Affiliation(s)
- Simon G Royce
- Allergy and Immune Disorders, Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| | | | | | | | | | | | | |
Collapse
|
33
|
Royce SG, Lim C, Muljadi RC, Samuel CS, Ververis K, Karagiannis TC, Giraud AS, Tang MLK. Trefoil factor-2 reverses airway remodeling changes in allergic airways disease. Am J Respir Cell Mol Biol 2012; 48:135-44. [PMID: 22652198 DOI: 10.1165/rcmb.2011-0320oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Trefoil factor 2 (TFF2) is a small peptide with an important role in mucosal repair. TFF2 is up-regulated in asthma, suggesting a role in asthma pathogenesis. Given its known biological role in promoting epithelial repair, TFF2 might be expected to exert a protective function in limiting the progression of airway remodeling in asthma. The contribution of TFF2 to airway remodeling in asthma was investigated by examining the expression of TFF2 in the airway and lung, and evaluating the effects of recombinant TFF2 treatment on established airway remodeling in a murine model of chronic allergic airways disease (AAD). BALB/c mice were sensitized and challenged with ovalbumin (OVA) or saline for 9 weeks, whereas mice with established OVA-induced AAD were treated with TFF2 or vehicle control (intranasally for 14 d). Effects on airway remodeling, airway inflammation, and airway hyperresponsiveness were then assessed, whereas TFF2 expression was determined by immunohistochemistry. TFF2 expression was significantly increased in the airways of mice with AAD, compared with expression levels in control mice. TFF2 treatment resulted in reduced epithelial thickening, subepithelial collagen deposition, goblet-cell metaplasia, bronchial epithelium apoptosis, and airway hyperresponsiveness (all P < 0.05, versus vehicle control), but TFF2 treatment did not influence airway inflammation. The increased expression of endogenous TFF2 in response to chronic allergic inflammation is insufficient to prevent the progression of airway inflammation and remodeling in a murine model of chronic AAD. However, exogenous TFF2 treatment is effective in reversing aspects of established airway remodeling. TFF2 has potential as a novel treatment for airway remodeling in asthma.
Collapse
Affiliation(s)
- Simon G Royce
- Department of Allergy and Immune Disorders, Murdoch Children's Research Institute Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Mahmoud YI. Grape seed extract attenuates lung parenchyma pathology in ovalbumin-induced mouse asthma model: an ultrastructural study. Micron 2012; 43:1050-9. [PMID: 22609098 DOI: 10.1016/j.micron.2012.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 04/07/2012] [Accepted: 04/24/2012] [Indexed: 01/31/2023]
Abstract
Due to the growing incidence of asthma and because of the non-specificity and side effects of the conventional drugs, the development of novel agents for the treatment of asthma has become considerably important. Natural plant products offer promising alternatives for the development of effective and safe treatments. Grape seed extract (GSE) is one such phytochemical supplement that has been shown to have potent antioxidant and anti-inflammatory effects. Thus, the present study aimed to investigate the effect of GSE to suppress lung parenchyma pathology and inflammation in ovalbumin-induced murine asthma model. Ovalbumin exposure was associated with many pathological and morphometric alterations in the lungs of asthmatic mice. The alterations involved alveolar size reduction, alveolar wall thickening, cellular infiltration and blood capillary congestion, as well as significant increase in the number of type II pneumocytes and lamellar bodies. However, GSE significantly ameliorated of the pathological changes of ovalbumin-induced asthma. The results support the possibility of GSE as an effective, safe anti-inflammatory dietary supplement to attenuate the pathogenicity of asthma. While these preliminary results appear promising, further studies are required to elucidate the precise mechanism of the modulatory effect of GSE on asthma remodeling.
Collapse
Affiliation(s)
- Yomna Ibrahim Mahmoud
- Zoology Department, Faculty of Science, Ain Shams University, P.O. Box 11566, Abbassia, Cairo, Egypt. yomna
| |
Collapse
|
35
|
Zhang WX, Liang YF, Wang XM, Nie Y, Chong L, Lin L, Chen C, Li CC. Urotensin upregulates transforming growth factor-β1 expression of asthma airway through ERK-dependent pathway. Mol Cell Biochem 2012; 364:291-8. [PMID: 22270542 DOI: 10.1007/s11010-012-1229-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 01/04/2012] [Indexed: 12/22/2022]
Abstract
Airway smooth muscle cells (ASMCs) play a key role in the process of asthma airway remodeling. Urotensin II (UII) and transforming growth factor (TGF)-β are potent mitogens for ASMCs proliferation. The study was aimed to determine whether UII-upregulated TGF-β-mediated ASMCs proliferation and extracellular signal-regulated kinase (ERK) was required for such an effect. OVA-sensitized rats were challenged to induce asthma. Lung morphology and airway dynamic parameters were monitored. ASMCs from control and asthma rats were purified for the measurement of UII and TGF-β1 expression. In vitro experiments were conducted to determine the direct effect of UII on TGF-β1 expression by ASMCs. Finally, U0126, an ERK inhibitor was used to examine the role of ERK pathway in UII mediated TGF-β1 upregulation. We found that both UII and TGF-β1 were upregulated in asthma lung tissues. In vitro study on ASMCs further revealed that UII may render its effect on ASMCs cells through the upregulation of TGF-β1. Data also supported the conclusion that ERK pathway was required, but not sufficient in UII-induced TGF-β1 upregulation. The current study provides new evidence that UII is involved in the TGF-β mediated mitogenic effect on ASMCs. UII, at least partially, uses ERK pathway to render such effect.
Collapse
Affiliation(s)
- Wei-Xi Zhang
- Department of Pediatric Pulmonology, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical College, Wenzhou 325027, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Yoon CH, Park HJ, Cho YW, Kim EJ, Lee JD, Kang KR, Han J, Kang D. Cigarette Smoke Extract-induced Reduction in Migration and Contraction in Normal Human Bronchial Smooth Muscle Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:397-403. [PMID: 22359478 PMCID: PMC3282228 DOI: 10.4196/kjpp.2011.15.6.397] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 11/21/2011] [Accepted: 12/01/2011] [Indexed: 11/18/2022]
Abstract
The proliferation, migration, cytokine release, and contraction of airway smooth muscle cells are key events in the airway remodeling process that occur in lung disease such as asthma, chronic obstruction pulmonary disease, and cancer. These events can be modulated by a number of factors, including cigarette smoke extract (CSE). CSE-induced alterations in the viability, migration, and contractile abilities of normal human airway cells remain unclear. This study investigated the effect of CSE on cell viability, migration, tumor necrosis factor (TNF)-α secretion, and contraction in normal human bronchial smooth muscle cells (HBSMCs). Treatment of HBSMCs with 10% CSE induced cell death, and the death was accompanied by the generation of reactive oxygen species (ROS). CSE-induced cell death was reduced by N-acetyl-l-cysteine (NAC), an ROS scavenger. In addition, CSE reduced the migration ability of HBSMCs by 75%. The combination of NAC with CSE blocked the CSE-induced reduction of cell migration. However, CSE had no effect on TNF-α secretion and NF-κB activation. CSE induced an increase in intracellular Ca2+ concentration in 64% of HBSMCs. CSE reduced the contractile ability of HBSMCs, and the ability was enhanced by NAC treatment. These results demonstrate that CSE treatment induces cell death and reduces migration and contraction by increasing ROS generation in normal HBSMCs. These results suggest that CSE may induce airway change through cell death and reduction in migration and contraction of normal HBSMCs.
Collapse
Affiliation(s)
- Chul Ho Yoon
- Department of Rehabilitation Medicine, Gyeongsang National University School of Medicine, Jinju 660-751, Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Lin YT, Hsu JY, Chen CJ, Chu JJ, Fu LS. Caffeic acid phenethyl ester suppresses eotaxin secretion and nuclear p-STAT6 in human lung fibroblast cells. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2011; 44:435-41. [DOI: 10.1016/j.jmii.2011.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 10/28/2010] [Accepted: 11/22/2010] [Indexed: 11/16/2022]
|
38
|
Abstract
Airway remodeling is a central feature of asthma. It is exemplified by thickening of the lamina reticularis and structural changes to the epithelium, submucosa, smooth muscle, and vasculature of the airway wall. Airway remodeling may result from persistent airway inflammation. Immunoglobulin E (IgE) is an important mediator of allergic reactions and has a central role in airway inflammation and asthma-related symptoms. Anti-IgE therapies (such as omalizumab) have the potential to block an early step in the allergic cascade and therefore have the potential to reduce airway remodeling. The reduction in free IgE levels following anti-IgE therapy leads to reductions in high-affinity IgE receptor (FcεRI) expression on mast cells, basophils, and dendritic cells. This combined effect results in attenuation of several markers of inflammation, including peripheral and bronchial tissue eosinophilia and levels of granulocyte macrophage colony-stimulating factor, interleukin (IL)-2, IL-4, IL-5, and IL-13. Considering the previously demonstrated anti-inflammatory effects of anti-IgE therapy, along with results from a small study showing continued benefit after discontinuation of long-term treatment, a larger study to assess its effect on markers of airway remodeling is underway.
Collapse
Affiliation(s)
- K F Rabe
- Department of Medicine, University Kiel, Kiel, Germany and Krankenhaus Grosshansdorf, Center for Pulmonology and Thoracic Surgery, Wöhrendamm 80, Grosshansdorf, Germany.
| | | | | | | |
Collapse
|
39
|
Buckland GL. Harnessing opportunities in non-animal asthma research for a 21st-century science. Drug Discov Today 2011; 16:914-27. [PMID: 21875684 DOI: 10.1016/j.drudis.2011.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 06/29/2011] [Accepted: 08/02/2011] [Indexed: 10/17/2022]
Abstract
The incidence of asthma is on the increase and calls for research are growing, yet asthma is a disease that scientists are still trying to come to grips with. Asthma research has relied heavily on animal use; however, in light of increasingly robust in vitro and computational models and the need to more fully incorporate the 'Three Rs' principles of Replacement, Reduction and Refinement, is it time to reassess the asthma research paradigm? Progress in non-animal research techniques is reaching a level where commitment and integration are necessary. Many scientists believe that progress in this field rests on linking disciplines to make research directly translatable from the bench to the clinic; a '21st-century' scientific approach to address age-old questions.
Collapse
|
40
|
Rowe RG, Keena D, Sabeh F, Willis AL, Weiss SJ. Pulmonary fibroblasts mobilize the membrane-tethered matrix metalloprotease, MT1-MMP, to destructively remodel and invade interstitial type I collagen barriers. Am J Physiol Lung Cell Mol Physiol 2011; 301:L683-92. [PMID: 21840960 DOI: 10.1152/ajplung.00187.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In acute and chronic lung disease, widespread disruption of tissue architecture underlies compromised pulmonary function. Pulmonary fibroblasts have been implicated as critical effectors of tissue-destructive extracellular matrix (ECM) remodeling by mobilizing a spectrum of proteolytic enzymes. Although efforts to date have focused on the catabolism of type I collagen, the predominant component of the lung interstitial matrix, the key collagenolytic enzymes employed by pulmonary fibroblasts remain unidentified. Herein, membrane type-1 matrix metalloprotease (MT1-MMP) is identified as the dominant and direct-acting protease responsible for the type I collagenolytic activity mediated by both mouse and human pulmonary fibroblasts. Furthermore, MT1-MMP is shown to be essential for pulmonary fibroblast migration within three-dimensional (3-D) hydrogels of cross-linked type I collagen that recapitulate ECM barriers encountered in the in vivo environment. Together, these findings demonstrate that MT1-MMP serves as a key effector of type I collagenolytic activity in pulmonary fibroblasts and earmark this pericellular collagenase as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- R Grant Rowe
- Divisions of Molecular Medicine & Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109-2216, USA
| | | | | | | | | |
Collapse
|
41
|
Evidence of a genetic contribution to lung function decline in asthma. J Allergy Clin Immunol 2011; 128:479-84. [PMID: 21752436 DOI: 10.1016/j.jaci.2011.05.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/25/2011] [Accepted: 05/25/2011] [Indexed: 11/21/2022]
Abstract
There has been great progress in identifying new asthma susceptibility genes. In asthmatic subjects there is variable airway remodeling that includes features such as smooth muscle hypertrophy/hyperplasia, basement membrane thickening, and increased extracellular matrix deposition. Does airway remodeling have a genetic contribution in asthma? Data from different murine strains suggest there is a genetic contribution to the development and progression of airway remodeling. In human subjects it is important to consider what surrogate markers of remodeling have been used in genetic studies. Baseline FEV(1) and airway hyperresponsiveness are determined by a complex interplay of factors, including nonremodeling mechanisms; however, we consider a decline in FEV(1) as a robust marker of remodeling. To date, single nucleotide polymorphisms spanning ADAM33, ESR1, PLAUR, and VEGF have been associated with an excess decline in lung function in asthmatic subjects carrying the rare alleles (FEV(1), -13.0 to 55.2 mL/y excess). Interestingly these genes have overlapping functions in proteolytic pathways in the airways. There is accumulating evidence that genetic factors are important in the development of airway remodeling in asthmatic subjects, and further longitudinal studies with additional remodeling phenotypes and genome-wide association studies will identify novel susceptibility genes, leading to new approaches to target remodeling in asthmatic subjects.
Collapse
|
42
|
Royce SG, Dang W, Yuan G, Tran J, El Osta A, Karagiannis TC, Tang MLK. Resveratrol has protective effects against airway remodeling and airway hyperreactivity in a murine model of allergic airways disease. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2011; 1:PBA-1-7134. [PMID: 22953028 PMCID: PMC3417665 DOI: 10.3402/pba.v1i0.7134] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 12/14/2022]
Abstract
Background New therapies for asthma which can address three main interrelated features of the disease, airway inflammation, airway remodeling and airway hyperreactivity, are urgently required. Resveratrol, a well known red wine polyphenol has received much attention due to its potential anti-aging properties. This compound is an agonist of silent information regulator two histone deacetylases and has many effects that are relevant to key aspects of the pathophysiology of asthma including inflammation, cell proliferation and fibrosis. Therefore, resveratrol may offer a novel asthma therapy that simultaneously inhibits airway inflammation, and airway remodeling which are the main contributors to airway hyperreactivity and irreversible lung function loss. Methods We evaluated the effects of systemic resveratrol treatment in a murine model of chronic allergic airways disease which displays most of the clinicopathological features of severe human asthma. Wild-type Balb/c mice with allergic airways disease were treated with 12.5 mg/kg resveratrol or vehicle control. Airway inflammation was assessed by bronchoalveolar lavage fluid cell counts and histological examination of lung tissue sections. Further, remodeling was assessed by morphometric analysis and lung function was assessed by invasive plethysmography measurement of airway resistance and dynamic compliance. Results Mice treated with resveratrol exhibited reduced tissue inflammation as compared to vehicle treated mice (p<0.05). Additionally, resveratrol treatment resulted in reduced subepithelial collagen deposition as compared to vehicle treated mice (p<0.05) and attenuated airway hyperreactivity (p<0.05). Conclusions These novel findings demonstrate that treatment with resveratrol can reduce structural airway remodeling changes and hyperreactivity. This has important implications for the development of new therapeutic approaches to asthma.
Collapse
Affiliation(s)
- Simon G Royce
- Allergy and Immune Disorders, Murdoch Children's Research Institute, Melbourne, Vic., Australia
| | | | | | | | | | | | | |
Collapse
|
43
|
Airway remodeling: a potential therapeutic target in asthma. World J Pediatr 2011; 7:124-8. [PMID: 21574028 DOI: 10.1007/s12519-011-0264-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 10/15/2010] [Indexed: 10/18/2022]
Abstract
BACKGROUND Apart from airway inflammation, airway remodeling is one of the main pathological features of asthma. However, it remains unclear when airway remodeling starts in children and whether it could be a potential therapeutic target in asthma. DATA SOURCES We have reviewed the recent literature regarding structural changes after airway remodeling, the relationship between airway inflammation and airway remodeling, the relationship between childhood asthma and airway remodeling, and the role of long-term medication in asthma treatment for airway remodeling. RESULTS The relationship between airway inflammation and airway remodeling is still controversial. A number of morphological and pathological studies have confirmed that airway remodeling occurs not only in adult asthma, but also in childhood asthma. It develops early in the disease process of asthma. At present, long-term medication in asthma treatment mainly focuses on anti-inflammation. However, there are no therapeutic interventions that revert airway remodeling once it is established. CONCLUSIONS Airway remodeling may provide a possible new therapeutic target in the management of asthma. It is imperative to strengthen the research in developing new medications specifically for asthma airway remodeling. Prevention and treatment of airway remodeling become top priority in future asthma research.
Collapse
|
44
|
Todorova L, Bjermer L, Westergren-Thorsson G, Miller-Larsson A. TGFβ-induced matrix production by bronchial fibroblasts in asthma: budesonide and formoterol effects. Respir Med 2011; 105:1296-307. [PMID: 21514131 DOI: 10.1016/j.rmed.2011.03.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 03/27/2011] [Accepted: 03/29/2011] [Indexed: 01/09/2023]
Abstract
To investigate the mechanisms of enhanced airway deposition of subepithelial collagen in asthma and its sensitivity to drug therapy with combination of an inhaled glucocorticosteroid (GC) and a long-acting β(2)-agonist (LABA), a cell model system involving bronchial fibroblasts derived from biopsies from patients with stable mild-to-moderate asthma has been used. To mimic unstable conditions and severe asthma, fibroblasts were stimulated ex vivo with TGFβ1. Primary fibroblasts established from central bronchial biopsies from 8 asthmatic patients were incubated for 24 h with 0.4% serum or TGFβ1 (10 ng/ml) with/without the GC budesonide (BUD; 10 nM) and/or the LABA formoterol (FORM; 0.1 nM). Procollagen peptide I (PICP), metalloproteinase (MMP)-1 and tissue inhibitor of MMPs (TIMP-1) were determined in culture media using ELISA while the activity of MMP-2, -3, -9 by zymography. Metabolically labeled proteoglycans, biglycan and decorin, associated with collagen fibrillation/deposition, were separated using chromatography and SDS-PAGE. The levels of PICP and biglycan were increased 2-fold by TGFβ1 (p < 0.05). The BUD and FORM combination reduced the PICP increase by 58% (p < 0.01) and the biglycan by 36% (p < 0.05) while each drug alone had no effect. Decorin levels were reduced by TGFβ1 in fibroblasts of most patients; BUD alone and BUD and FORM completely counteracted this decrease. MMPs and TIMP-1 were not affected by TGFβ1 or the drugs. These results suggest that BUD and FORM combination therapy, without affecting metalloproteolytic balance, has a potential to counteract enhanced collagen production by bronchial fibroblasts in asthma and to normalize the production of small proteoglycans which may affect collagen fibrillation and deposition.
Collapse
Affiliation(s)
- Lizbet Todorova
- Department of Experimental Medical Sciences, Division of Lung Biology, Lund University, BMC D12, 221 84 Lund, Sweden
| | | | | | | |
Collapse
|
45
|
AKHABIR LOUBNA, SANDFORD ANDREWJ. Genome-wide association studies for discovery of genes involved in asthma. Respirology 2011; 16:396-406. [DOI: 10.1111/j.1440-1843.2011.01939.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
Effect of short-term oral and inhaled corticosteroids on airway inflammation and responsiveness in a feline acute asthma model. Vet J 2011; 192:41-8. [PMID: 21354836 DOI: 10.1016/j.tvjl.2011.01.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 12/17/2022]
Abstract
The objective of this study was to investigate whether high-dose inhaled fluticasone propionate (FP), alone or in combination with salmeterol (SAL), is as effective as oral prednisolone in reducing airway inflammation and obstruction in cats with experimentally-induced acute asthma. Six cats sensitised to Ascaris suum (AS) were enrolled in a prospective controlled therapeutic trial and underwent four aerosol challenges, at 1-month intervals with AS allergen. The allergen - stimulated animals received four consecutive days treatment with either oral prednisolone at 1mg/kg twice daily, 500 μg of FP inhaled twice daily, or a combination of FP/SAL at 500 μg/50 μg inhaled twice daily, respectively, according to a randomised cross-over design. Treatment-related changes in lung function, airway responsiveness (AR) and bronchoalveolar lavage fluid (BALF) cytology were assessed. Barometric whole-body plethysmography (BWBP) was used for the assessment of respiratory variables and AR. No significant differences in respiratory rate or Penh (an estimate of airflow limitation measured by BWBP) were detected among treatment groups. Allergen-induced airway hyper-responsiveness was significantly inhibited by all three steroid treatments (P<0.05). The mean BALF eosinophil percentage (±SEM) was lower after oral and inhaled corticosteroid treatment and these changes were significant for groups receiving prednisolone and the FP/SAL combination. Findings suggest high-dose FP, particularly in combination with SAL, is effective in ameliorating airway inflammation and hyper-responsiveness in this model of acute feline asthma, and highlight the potential use of these drugs in cats experiencing acute exacerbations of the naturally occurring disease.
Collapse
|
47
|
Girodet PO, Ozier A, Bara I, Tunon de Lara JM, Marthan R, Berger P. Airway remodeling in asthma: new mechanisms and potential for pharmacological intervention. Pharmacol Ther 2011; 130:325-37. [PMID: 21334378 DOI: 10.1016/j.pharmthera.2011.02.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 02/02/2011] [Indexed: 01/10/2023]
Abstract
The chronic inflammatory response within the airways of asthmatics is associated with structural changes termed airway remodeling. This remodeling process is a key feature of severe asthma. The 5-10% of patients with a severe form of the disease account for the higher morbidity and health costs related to asthma. Among the histopathological characteristics of airway remodeling, recent reports indicate that the increased mass of airway smooth muscle (ASM) plays a critical role. ASM cell proliferation in severe asthma implicates a gallopamil-sensitive calcium influx and the activation of calcium-calmodulin kinase IV leading to enhanced mitochondrial biogenesis through the activation of various transcription factors (PGC-1α, NRF-1 and mt-TFA). The altered expression and function of sarco/endoplasmic reticulum Ca(2+) pump could play a role in ASM remodeling in moderate to severe asthma. Additionally, aberrant communication between an injured airway epithelium and ASM could also contribute to disease severity. Airway remodeling is insensitive to corticosteroids and anti-leukotrienes whereas the effect of monoclonal antibodies (the anti-IgE omalizumab, the anti-interleukin-5 mepolizumab or anti-tumor necrosis factor-alpha) remains to be investigated. This review focuses on potential new therapeutic strategies targeting ASM cells, especially Ca(2+) and mitochondria-dependent pathways.
Collapse
|
48
|
McKenzie R, Burton MD, Royce SG, Tang MLK. Age and sex influences on airway hyperresponsiveness. J Asthma 2010; 47:651-4. [PMID: 20632782 DOI: 10.3109/02770901003692801] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The precise relationship between age and gender and their influence on airway reactivity has not been clearly defined. Previous studies of age and gender influences on airway reactivity have been confounded by environmental influences such as cigarette smoking. The objective of this study was to examine the effect of age and gender on airway reactivity in C57BL/6 mice housed under controlled conditions, independent of confounding environmental factors. METHODS Mice were separated into four experimental groups based on age and gender: males at 6 and 12 weeks of age and females at 6 and 12 weeks of age. Airway reactivity to inhaled methacholine was examined in each group. RESULTS Significant differences in methacholine-induced airway reactivity were observed between the sexes at both age groups. At 6 weeks of age, the males demonstrated a significantly increased airway reactivity to methacholine as compared to females (p <.001). Paradoxically, at 12 weeks of age, the males demonstrated a significantly lower response to methacholine as compared to females (p <.001). Hence the relationship between age and airway reactivity is markedly different in males as compared to females. Examination of intragender differences revealed that in female mice, airway reactivity increased significantly with age (p <.001). In contrast, males demonstrated a significant reduction in airway reactivity with age (p <.001). CONCLUSION These findings demonstrate important differences in airway reactivity related to age and gender that are observed independent of any environmental influences. Furthermore, these findings highlight the importance of careful age and sex matching in studies of airway reactivity.
Collapse
Affiliation(s)
- Ross McKenzie
- Department of Allergy and Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
49
|
Rock JR, Randell SH, Hogan BLM. Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis Model Mech 2010; 3:545-56. [PMID: 20699479 DOI: 10.1242/dmm.006031] [Citation(s) in RCA: 542] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The small airways of the human lung undergo pathological changes in pulmonary disorders, such as chronic obstructive pulmonary disease (COPD), asthma, bronchiolitis obliterans and cystic fibrosis. These clinical problems impose huge personal and societal healthcare burdens. The changes, termed 'pathological airway remodeling', affect the epithelium, the underlying mesenchyme and the reciprocal trophic interactions that occur between these tissues. Most of the normal human airway is lined by a pseudostratified epithelium of ciliated cells, secretory cells and 6-30% basal cells, the proportion of which varies along the proximal-distal axis. Epithelial abnormalities range from hypoplasia (failure to differentiate) to basal- and goblet-cell hyperplasia, squamous- and goblet-cell metaplasia, dysplasia and malignant transformation. Mesenchymal alterations include thickening of the basal lamina, smooth muscle hyperplasia, fibrosis and inflammatory cell accumulation. Paradoxically, given the prevalence and importance of airway remodeling in lung disease, its etiology is poorly understood. This is due, in part, to a lack of basic knowledge of the mechanisms that regulate the differentiation, maintenance and repair of the airway epithelium. Specifically, little is known about the proliferation and differentiation of basal cells, a multipotent stem cell population of the pseudostratified airway epithelium. This Perspective summarizes what we know, and what we need to know, about airway basal cells to evaluate their contributions to normal and abnormal airway remodeling. We contend that exploiting well-described model systems using both human airway epithelial cells and the pseudostratified epithelium of the genetically tractable mouse trachea will enable crucial discoveries regarding the pathogenesis of airway disease.
Collapse
Affiliation(s)
- Jason R Rock
- Department of Cell Biology, Nanaline Duke Building, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
50
|
Mehrad B, Keane MP, Gomperts BN, Strieter RM. Circulating progenitor cells in chronic lung disease. Expert Rev Respir Med 2010; 1:157-65. [PMID: 20477275 DOI: 10.1586/17476348.1.1.157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Tissue regeneration and repair are fundamental both to recovery of the lung from injury and to the pathology of many chronic lung diseases. There are two potential sources for the adult progenitor cells that participate in this reparative process: resident lung progenitors and bone marrow-derived circulating cells. Bone marrow-derived cells, in particular, have been shown to give rise to airway and alveolar epithelial cells, as well as lung mesenchymal cells. Emerging data have linked specific chemokine ligand-receptor interactions to the recruitment of these cells to the lung and has implicated these cells in chronic lung disorders such as asthma and interstitial lung diseases. In this review, we summarize the current understanding of the biology of adult circulating progenitors as related to lung disease.
Collapse
Affiliation(s)
- Borna Mehrad
- University of Virginia, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Box 800546, Charlottesville, VA 22908, USA.
| | | | | | | |
Collapse
|