1
|
Garcia da Silva AC, Carvalho Filho SDM, Furtado de Mendonça IC, Valadares MC. Identification of toxicity-induced biomarkers in human non-immune airway cells exposed to respiratory sensitizers: A mechanistic approach. Toxicology 2024; 503:153750. [PMID: 38360295 DOI: 10.1016/j.tox.2024.153750] [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: 12/21/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
Occupational asthma covers a group of work-related diseases whose clinical manifestations include airway hyperresponsiveness and airflow limitation. Although the chemical respiratory allergy (CRA) induced by Low Molecular Weight (LMW) sensitizers is a major concern, especially in terms of the regulatory framework, to date there are no methods available for preclinically addressing this toxicological outcome, as its mechanistic background is not fully understood at molecular or cellular levels. This paper proposes a mechanistic study applying New Approach Methodologies (NAM) of the pro-inflammatory and functional effects triggered by LMW respiratory allergens in different respiratory tract cell lines, including bronchial epithelial (BEAS-2B), lung fibroblast (MRC-5), and endothelial cells (EA.hy926), and an analysis of the capacity of such chemicals to interact with the mucin protein, to address certain toxicodynamic aspects of such compounds. The results showed that some of the sensitizers evaluated interact with mucin, the main protein mucus component, but the toxicant-mucin complex formation does not seem to be a common feature of different chemical classes of allergens. At a cellular level, sensitizers promoted an increase in IL-8, IL-6, and IL-1β production in the evaluated cell types. It also impaired the MUC1 expression by bronchial cells and activated endothelial cells, thereby increasing the ICAM-I surface expression. Taken together, our results showed that these aforementioned cell types participate in the CRA Adverse Outcome Pathway and must be considered when developing preclinical testing strategies, particularly investigating danger signal production after exposure to LMW sensitizers in different tissue compartments.
Collapse
Affiliation(s)
- Artur Christian Garcia da Silva
- Laboratory of Education and Research in In vitro Toxicology, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Sérgio de Morais Carvalho Filho
- Laboratory of Education and Research in In vitro Toxicology, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Marize Campos Valadares
- Laboratory of Education and Research in In vitro Toxicology, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
2
|
Zhao S, Luo J, Hu J, Wang H, Zhao N, Cao M, Zhang C, Hu R, Liu L. Role of Ezrin in Asthma-Related Airway Inflammation and Remodeling. Mediators Inflamm 2022; 2022:6255012. [PMID: 36530558 PMCID: PMC9750775 DOI: 10.1155/2022/6255012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 08/13/2023] Open
Abstract
Ezrin is an actin binding protein connecting the cell membrane and the cytoskeleton, which is crucial to maintaining cell morphology, intercellular adhesion, and cytoskeleton remodeling. Asthma involves dysfunction of inflammatory cells, cytokines, and airway structural cells. Recent studies have shown that ezrin, whose function is affected by extensive phosphorylation and protein interactions, is closely associated with asthma, may be a therapeutic target for asthma treatment. In this review, we summarize studies on ezrin and discuss its role in asthma-related airway inflammation and remodeling.
Collapse
Affiliation(s)
- Shumei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jiaqi Luo
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jun Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hesheng Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Shimadzu Biomedical Research Laboratory, Shanghai 200233, China
| | - Meng Cao
- Nanjing University of Chinese Medicine, Nanjing 210029, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Cong Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Rongkui Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Lanying Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
3
|
Sex Steroids Effects on Asthma: A Network Perspective of Immune and Airway Cells. Cells 2022; 11:cells11142238. [PMID: 35883681 PMCID: PMC9318292 DOI: 10.3390/cells11142238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022] Open
Abstract
A multitude of evidence has suggested the differential incidence, prevalence and severity of asthma between males and females. A compilation of recent literature recognized sex differences as a significant non-modifiable risk factor in asthma pathogenesis. Understanding the cellular and mechanistic basis of sex differences remains complex and the pivotal point of this ever elusive quest, which remains to be clarified in the current scenario. Sex steroids are an integral part of human development and evolution while also playing a critical role in the conditioning of the immune system and thereby influencing the function of peripheral organs. Classical perspectives suggest a pre-defined effect of sex steroids, generalizing estrogens popularly under the “estrogen paradox” due to conflicting reports associating estrogen with a pro- and anti-inflammatory role. On the other hand, androgens are classified as “anti-inflammatory,” serving a protective role in mitigating inflammation. Although considered mainstream and simplistic, this observation remains valid for numerous reasons, as elaborated in the current review. Women appear immune-favored with stronger and more responsive immune elements than men. However, the remarkable female predominance of diverse autoimmune and allergic diseases contradicts this observation suggesting that hormonal differences between the sexes might modulate the normal and dysfunctional regulation of the immune system. This review illustrates the potential relationship between key elements of the immune cell system and their interplay with sex steroids, relevant to structural cells in the pathophysiology of asthma and many other lung diseases. Here, we discuss established and emerging paradigms in the clarification of observed sex differences in asthma in the context of the immune system, which will deepen our understanding of asthma etiopathology.
Collapse
|
4
|
Asthmatic Eosinophils Promote Contractility and Migration of Airway Smooth Muscle Cells and Pulmonary Fibroblasts In Vitro. Cells 2021; 10:cells10061389. [PMID: 34199925 PMCID: PMC8229663 DOI: 10.3390/cells10061389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022] Open
Abstract
Enhanced contractility and migration of airway smooth muscle cells (ASMC) and pulmonary fibroblasts (PF) are part of airway remodeling in asthma. Eosinophils are the central inflammatory cells that participate in airway inflammation. However, the role of asthmatic eosinophils in ASMC and PF contractility, migration, and differentiation to contractile phenotype has not yet been precisely described. A total of 38 individuals were included in this study: 13 steroid-free non-severe allergic asthma (AA) patients, 11 severe non-allergic eosinophilic asthma (SNEA) patients, and 14 healthy subjects (HS). For AA patients and HS groups, a bronchial allergen challenge with D. pteronyssinus was performed. Individual combined cell cultures were prepared from isolated peripheral blood eosinophils and immortalized ASMC or commercial PF cell lines separately. The migration of ASMC and PF was evaluated using wound healing assay and contractility using collagen gel assay. Gene expression of contractile apparatus proteins, COL1A1, COL5A1, and FN, in ASMC and PF was evaluated using qRT-PCR. We found that contractility and migration of ASMC and PF significantly increased after incubation with asthmatic eosinophils compared to HS eosinophils, p < 0.05, and SNEA eosinophils demonstrated the highest effect on contractility of ASMC and migration of both cell lines, p < 0.05. AA and SNEA eosinophils significantly increased gene expression of contractile apparatus proteins, COL1A1 and FN, in both cell lines, p < 0.05. Furthermore, the allergen-activated AA eosinophils significantly increased the contractility of ASMC, and migration and gene expression in ASMC and PF, p < 0.05. Thus, asthmatic eosinophils change ASMC and PF behavior by increasing their contractility and migration, contributing to airway remodeling.
Collapse
|
5
|
Leukotriene D 4 role in allergic asthma pathogenesis from cellular and therapeutic perspectives. Life Sci 2020; 260:118452. [PMID: 32956660 DOI: 10.1016/j.lfs.2020.118452] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 01/05/2023]
Abstract
Asthma is a chronic inflammatory and allergic disease that is mainly characterized by reversible airway obstruction and bronchial hyperresponsiveness. The incidence of asthma is increasing with more than 350 million people worldwide are affected. Up to now, there is no therapeutic option for asthma and most of the prescribed drugs aim to ameliorate the symptoms of the disease especially during the acute exacerbations after trigger exposure. Asthma is a heterogonous disease that involves interactions between inflammatory mediators and cellular components within the disease microenvironment including inflammatory and structural cells. Cysteinyl leukotrienes (cys-LTs) are inflammatory lipid mediators that have potent roles in asthma pathogenesis. CysLTs consisting of LTC4, LTD4, and LTE4 are mainly secreted by leukocytes and act through three main G-protein coupled receptors (CysLT1R, CysLT2R, and CysLT3R). LTD4 is the most potent bronchoconstrictor which gives it the priority to be discussed in detail in this review. LTD4 binds with high affinity to CysLT1R and many studies showed that using CysLT1R antagonists such as montelukast has a beneficial effect for asthmatics especially in corticosteroid refractory cases. Since asthma is a heterogeneous inflammatory disease of many cell types involved in the disease pathogenies and LTD4 has a special role in inflammation and bronchoconstriction, this review highlights the role of LTD4 on each cellular component in asthma and the benefits of using CysLT1R antagonists in ameliorating LTD4-induced effects.
Collapse
|
6
|
Liu Y, Li X, He C, Chen R, Wei L, Meng L, Zhang C. Emodin ameliorates ovalbumin-induced airway remodeling in mice by suppressing airway smooth muscle cells proliferation. Int Immunopharmacol 2020; 88:106855. [PMID: 32777676 DOI: 10.1016/j.intimp.2020.106855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 01/15/2023]
Abstract
Increased number of airway smooth muscle cells (ASMCs) is a characteristic of airway remodeling in asthma. In this study we investigated whether emodin alleviated airway remodeling in a murine asthma model and reduced the proliferation of ASMCs in vitro. We provided in vivo evidence suggesting that intraperitoneal injection of emodin (20 mg/kg) 1 h prior to OVA challenge apparently alleviated the thickness of airway smooth muscle, the mass of alpha-smooth muscle actin (α-SMA), collagen deposition, epithelial damage, goblet cell hyperplasia, airway inflammation and airway hyperresponsiveness (AHR) in lung tissue. Meanwhile, we found that emodin suppressed the activation of the Akt pathway in lungtissue of allergic mouse models. Additionally, we found that emodin inhibited cellular proliferation and Akt activation in a dose-dependent manner in vitro. Furthermore, LY294002, an inhibitor for PI3K, abrogated serum-induced phosphorylation of Akt, and decreased the proliferation of ASMCs. These findings indicated that emodin alleviated ASMCs proliferation by inhibiting PI3K/Akt pathway in vivo and in vitro, which may provide a potential therapeutic option for airway smooth muscle remodeling in asthma.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Xin Li
- Department of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Chao He
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Ran Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Li Wei
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Ling Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China.
| | - Caiqing Zhang
- Department of Respiratory Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
7
|
What Have In Vitro Co-Culture Models Taught Us about the Contribution of Epithelial-Mesenchymal Interactions to Airway Inflammation and Remodeling in Asthma? Cells 2020; 9:cells9071694. [PMID: 32679790 PMCID: PMC7408556 DOI: 10.3390/cells9071694] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
As the lung develops, epithelial-mesenchymal crosstalk is essential for the developmental processes that drive cell proliferation, differentiation, and extracellular matrix (ECM) production within the lung epithelial-mesenchymal trophic unit (EMTU). In asthma, a number of the lung EMTU developmental signals have been associated with airway inflammation and remodeling, which has led to the hypothesis that aberrant activation of the asthmatic EMTU may lead to disease pathogenesis. Monoculture studies have aided in the understanding of the altered phenotype of airway epithelial and mesenchymal cells and their contribution to the pathogenesis of asthma. However, 3-dimensional (3D) co-culture models are needed to enable the study of epithelial-mesenchymal crosstalk in the setting of the in vivo environment. In this review, we summarize studies using 3D co-culture models to assess how defective epithelial-mesenchymal communication contributes to chronic airway inflammation and remodeling within the asthmatic EMTU.
Collapse
|
8
|
Yap JMG, Ueda T, Takeda N, Fukumitsu K, Fukuda S, Uemura T, Tajiri T, Ohkubo H, Maeno K, Ito Y, Kanemitsu Y, Niimi A. An inflammatory stimulus sensitizes TRPA1 channel to increase cytokine release in human lung fibroblasts. Cytokine 2020; 129:155027. [PMID: 32050145 DOI: 10.1016/j.cyto.2020.155027] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 01/08/2023]
Abstract
External stimuli such as cigarette smoke and house dust mite are often involved in the development and exacerbation of asthma. These risk factors could activate or sensitize transient receptor potential channel ankyrin 1 (TRPA1), which are primarily expressed in neuronal structures but also in non-neuronal cells such as fibroblasts. However, the role of non-neuronal TRPA1 in the pathophysiology of airway diseases including asthma remains unclear. We investigated TRPA1 expression on human fibroblast cells and whether inflammatory mediators could modulate its function. This study utilized human lung fibroblast cell lines, Medical Research Council cell strain 5 (MRC-5) and HF19 cells frequently used on experimental studies regarding allergic and respiratory disorders. The human lung fibroblasts were stimulated with house dust mite (Der p1) or tumor necrosis factor alpha (TNF-α) for 24 h, and we quantified TRPA1 mRNA and protein by qRT-PCR and western blot analysis, respectively. TRPA1 mRNA expressions were upregulated after TNF-α treatment. Calcium imaging analysis revealed that TNF-α treatment apparently sensitized TRPA1-mediated calcium influx by TRPA1 agonist allyl isothiocyanate (AITC) and the selective TRPA1 channel blocker HC-030031 effectively reduced the calcium response. Lastly, TRPA1 activation was not only involved in increased IL-8 cytokine release, but also in upregulating gene expression of matrix metalloprotease 9 (MMP9) in the human lung fibroblasts treated with TNF-α Together, these results indicate that presence of inflammatory mediators such as TNF-α could upregulate the non-neuronal expression of TRPA1 on fibroblasts which may aggravate further the release of inflammatory cytokines observed in human airway diseases.
Collapse
Affiliation(s)
- Jennifer Maries Go Yap
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Takashi Ueda
- Department of Anatomy and Neuroscience, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Norihisa Takeda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Kensuke Fukumitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Satoshi Fukuda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Tomoko Tajiri
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Hirotsugu Ohkubo
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Ken Maeno
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Yutaka Ito
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Yoshihiro Kanemitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan.
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| |
Collapse
|
9
|
Mostaço-Guidolin LB, Osei ET, Ullah J, Hajimohammadi S, Fouadi M, Li X, Li V, Shaheen F, Yang CX, Chu F, Cole DJ, Brandsma CA, Heijink IH, Maksym GN, Walker D, Hackett TL. Defective Fibrillar Collagen Organization by Fibroblasts Contributes to Airway Remodeling in Asthma. Am J Respir Crit Care Med 2019; 200:431-443. [PMID: 30950644 DOI: 10.1164/rccm.201810-1855oc] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Histologic stains have been used as the gold standard to visualize extracellular matrix (ECM) changes associated with airway remodeling in asthma, yet they provide no information on the biochemical and structural characteristics of the ECM, which are vital to understanding alterations in tissue function.Objectives: To demonstrate the use of nonlinear optical microscopy (NLOM) and texture analysis algorithms to image fibrillar collagen (second harmonic generation) and elastin (two-photon excited autofluorescence), to obtain biochemical and structural information on the remodeled ECM environment in asthma.Methods: Nontransplantable donor lungs from donors with asthma (n = 13) and control (n = 12) donors were used for the assessment of airway collagen and elastin fibers by NLOM, and extraction of lung fibroblasts for in vitro experiments.Measurements and Main Results: Fibrillar collagen is not only increased but also highly disorganized and fragmented within large and small asthmatic airways compared with control subjects, using NLOM imaging. Furthermore, such structural alterations are present in pediatric and adult donors with asthma, irrespective of fatal disease. In vitro studies demonstrated that asthmatic airway fibroblasts are deficient in their packaging of fibrillar collagen-I and express less decorin, important for collagen fibril packaging. Packaging of collagen fibrils was found to be more disorganized in asthmatic airways compared with control subjects, using transmission electron microscopy.Conclusions: NLOM imaging enabled the structural assessment of the ECM, and the data suggest that airway remodeling in asthma involves the progressive accumulation of disorganized fibrillar collagen by airway fibroblasts. This study highlights the future potential clinical application of NLOM to assess airway remodeling in vivo.
Collapse
Affiliation(s)
- Leila B Mostaço-Guidolin
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emmanuel T Osei
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jari Ullah
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Soheil Hajimohammadi
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - May Fouadi
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xian Li
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vicky Li
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Furquan Shaheen
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Chen Xi Yang
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Fanny Chu
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Darren J Cole
- 3School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - Corry-Anke Brandsma
- 4Department of Pathology and Medical Biology
- 5Groningen Research Institute of Asthma and COPD, and
| | - Irene H Heijink
- 4Department of Pathology and Medical Biology
- 5Groningen Research Institute of Asthma and COPD, and
- 6Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Geoffrey N Maksym
- 3School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; and
| | - David Walker
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- 1Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
- 2Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Rutting S, Xenaki D, Malouf M, Horvat JC, Wood LG, Hansbro PM, Oliver BG. Short-chain fatty acids increase TNFα-induced inflammation in primary human lung mesenchymal cells through the activation of p38 MAPK. Am J Physiol Lung Cell Mol Physiol 2018; 316:L157-L174. [PMID: 30407866 DOI: 10.1152/ajplung.00306.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs), produced as by-products of dietary fiber metabolism by gut bacteria, have anti-inflammatory properties and could potentially be used for the treatment of inflammatory diseases, including asthma. The direct effects of SCFAs on inflammatory responses in primary human lung mesenchymal cells have not been assessed. We investigated whether SCFAs can protect against tumor necrosis factor (TNF)α-induced inflammation in primary human lung fibroblasts (HLFs) and airway smooth muscle (ASM) cells in vitro. HLFs and ASM cells were exposed to SCFAs, acetate (C2:0), propionate (C3:0), and butyrate (C4:0) (0.01-25 mM) with or without TNFα, and the release of proinflammatory cytokines, IL-6, and CXCL8 was measured using ELISA. We found that none of the SCFAs suppressed TNFα-induced cytokine release. On the contrary, challenge with supraphysiological concentrations (10-25 mM), as might be used therapeutically, of propionate or butyrate in combination with TNFα resulted in substantially greater IL-6 and CXCL8 release from HLFs and ASM cells than challenge with TNFα alone, demonstrating synergistic effects. In ASM cells, challenge with acetate also enhanced TNFα-induced IL-6, but not CXCL8 release. Synergistic upregulation of IL-6 and CXCL8 was mediated through the activation of free fatty acid receptor (FFAR)3, but not FFAR2. The signaling pathways involved were further examined using specific inhibitors and immunoblotting, and responses were found to be mediated through p38 MAPK signaling. This study demonstrates that proinflammatory, rather than anti-inflammatory effects of SCFAs are evident in lung mesenchymal cells.
Collapse
Affiliation(s)
- Sandra Rutting
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Dia Xenaki
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia
| | - Monique Malouf
- Thoracic Medicine and Lung Transplantation, Saint Vincent's Hospital , Sydney, New South Wales , Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle, New South Wales , Australia.,Graduate School of Health, Discipline of Pharmacy, University of Technology Sydney , Sydney, New South Wales , Australia
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney , Sydney, New South Wales , Australia.,School of Life Sciences, University of Technology Sydney , Sydney, New South Wales , Australia
| |
Collapse
|
11
|
Majors AK, Chakravarti R, Ruple LM, Leahy R, Stuehr DJ, Lauer M, Erzurum SC, Janocha A, Aronica MA. Nitric oxide alters hyaluronan deposition by airway smooth muscle cells. PLoS One 2018; 13:e0200074. [PMID: 29966020 PMCID: PMC6028120 DOI: 10.1371/journal.pone.0200074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 06/19/2018] [Indexed: 12/04/2022] Open
Abstract
Asthma is a chronic inflammatory disease that is known to cause changes in the extracellular matrix, including changes in hyaluronan (HA) deposition. However, little is known about the factors that modulate its deposition or the potential consequences. Asthmatics with high levels of exhaled nitric oxide (NO) are characterized by greater airway reactivity and greater evidence of airway inflammation. Based on these data and our previous work we hypothesized that excessive NO promotes the pathologic production of HA by airway smooth muscle cells (SMCs). Exposure of cultured SMCs to various NO donors results in the accumulation of HA in the form of unique, cable-like structures. HA accumulates rapidly after exposure to NO and can be seen as early as one hour after NO treatment. The cable-like HA in NO-treated SMC cultures supports the binding of leukocytes. In addition, NO produced by murine macrophages (RAW cells) and airway epithelial cells also induces SMCs to produce HA cables when grown in co-culture. The modulation of HA by NO appears to be independent of soluble guanylate cyclase. Taken together, NO-induced production of leukocyte-binding HA by SMCs provides a new potential mechanism for the non-resolving airway inflammation in asthma and suggests a key role of non-immune cells in driving the chronic inflammation of the submucosa. Modulation of NO, HA and the consequent immune cell interactions may serve as potential therapeutic targets in asthma.
Collapse
Affiliation(s)
- Alana K. Majors
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ritu Chakravarti
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lisa M. Ruple
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Rachel Leahy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark Lauer
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C. Erzurum
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison Janocha
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark A. Aronica
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
12
|
Nayak AP, Deshpande DA, Penn RB. New targets for resolution of airway remodeling in obstructive lung diseases. F1000Res 2018; 7. [PMID: 29904584 PMCID: PMC5981194 DOI: 10.12688/f1000research.14581.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 12/17/2022] Open
Abstract
Airway remodeling (AR) is a progressive pathological feature of the obstructive lung diseases, including asthma and chronic obstructive pulmonary disease (COPD). The pathology manifests itself in the form of significant, progressive, and (to date) seemingly irreversible changes to distinct respiratory structural compartments. Consequently, AR correlates with disease severity and the gradual decline in pulmonary function associated with asthma and COPD. Although current asthma/COPD drugs manage airway contraction and inflammation, none of these effectively prevent or reverse features of AR. In this review, we provide a brief overview of the features and putative mechanisms affecting AR. We further discuss recently proposed strategies with promise for deterring or treating AR.
Collapse
Affiliation(s)
- Ajay P Nayak
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Deepak A Deshpande
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Raymond B Penn
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
13
|
Rutting S, Xenaki D, Lau E, Horvat J, Wood LG, Hansbro PM, Oliver BG. Dietary omega-6, but not omega-3, polyunsaturated or saturated fatty acids increase inflammation in primary lung mesenchymal cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L922-L935. [PMID: 29368548 DOI: 10.1152/ajplung.00438.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Obesity is an important risk factor for developing severe asthma. Dietary fatty acids, which are increased in sera of obese individuals and after high-fat meals, activate the innate immune system and induce inflammation. This study investigated whether dietary fatty acids directly cause inflammation and/or synergize with obesity-induced cytokines in primary human pulmonary fibroblasts in vitro. Fibroblasts were challenged with BSA-conjugated fatty acids [ω-6 polyunsaturated fatty acids (PUFAs) and ω-3 PUFAs or saturated fatty acids (SFAs)], with or without TNF-α, and release of the proinflammatory cytokines, IL-6 and CXCL8, was measured. We found that the ω-6 PUFA arachidonic acid (AA), but not ω-3 PUFAs or SFAs, upregulates IL-6 and CXCL8 release. Combined AA and TNF-α challenge resulted in substantially greater cytokine release than either alone, demonstrating synergy. Synergistic upregulation of IL-6, but not CXCL8, was mainly mediated via cyclooxygenase (COX). Inhibition of p38 MAPK reduced CXCL8 release, induced by AA and TNF-α alone, but not in combination. Synergistic CXCL8 release, following AA and TNF-α challenge, was not medicated via a single signaling pathway (MEK1, JNK, phosphoinositide 3-kinase, and NF-κB) nor by hyperactivation of NF-κB or p38. To investigate if these findings occur in other airway cells, effects of AA in primary human airway smooth muscle (ASM) cells and human bronchial epithelial cells were also investigated. We found proinflammatory effects in ASM cells but not epithelial cells. This study suggests that diets rich in ω-6 PUFAs might promote airway inflammation via multiple pathways, including COX-dependent and -independent pathways, and in an obese person, may lead to more severe airway inflammation.
Collapse
Affiliation(s)
- Sandra Rutting
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia.,Department of Respiratory Medicine, Royal Prince Alfred Hospital, University of Sydney, Sydney, Australia
| | - Dia Xenaki
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia
| | - Edmund Lau
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, University of Sydney, Sydney, Australia
| | - Jay Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and University of Newcastle , Newcastle , Australia
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney , Sydney , Australia.,School of Life Sciences, University of Technology Sydney , Sydney , Australia
| |
Collapse
|
14
|
King GG, James A, Harkness L, Wark PAB. Pathophysiology of severe asthma: We've only just started. Respirology 2018; 23:262-271. [PMID: 29316003 DOI: 10.1111/resp.13251] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 12/01/2022]
Abstract
Severe asthma is defined by the high treatment requirements to partly or fully control the clinical manifestations of disease. It remains a problem worldwide with a large burden for individuals and health services. The key to improving targeted treatments, reducing disease burden and improving patient outcomes is a better understanding of the pathophysiology and mechanisms of severe disease. The heterogeneity, complexity and difficulties in undertaking clinical studies in severe asthma remain challenges to achieving better understanding and better outcomes. In this review, we focus on the structural, mechanical and inflammatory abnormalities that are relevant in severe asthma.
Collapse
Affiliation(s)
- Gregory G King
- NHMRC Centre for Excellence in Severe Asthma, Newcastle, NSW, Australia.,Department of Respiratory Medicine, Royal North Shore Hospital, Sydney, NSW, Australia.,The Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Alan James
- NHMRC Centre for Excellence in Severe Asthma, Newcastle, NSW, Australia.,Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia
| | - Louise Harkness
- NHMRC Centre for Excellence in Severe Asthma, Newcastle, NSW, Australia.,The Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Peter A B Wark
- NHMRC Centre for Excellence in Severe Asthma, Newcastle, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Respiratory Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
15
|
Al-Alwan LA, Chang Y, Rousseau S, Martin JG, Eidelman DH, Hamid Q. CXCL1 inhibits airway smooth muscle cell migration through the decoy receptor Duffy antigen receptor for chemokines. THE JOURNAL OF IMMUNOLOGY 2014; 193:1416-26. [PMID: 24981451 DOI: 10.4049/jimmunol.1302860] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway smooth muscle cell (ASMC) migration is an important mechanism postulated to play a role in airway remodeling in asthma. CXCL1 chemokine has been linked to tissue growth and metastasis. In this study, we present a detailed examination of the inhibitory effect of CXCL1 on human primary ASMC migration and the role of the decoy receptor, Duffy AgR for chemokines (DARC), in this inhibition. Western blots and pathway inhibitors showed that this phenomenon was mediated by activation of the ERK-1/2 MAPK pathway, but not p38 MAPK or PI3K, suggesting a biased selection in the signaling mechanism. Despite being known as a nonsignaling receptor, small interference RNA knockdown of DARC showed that ERK-1/2 MAPK activation was significantly dependent on DARC functionality, which, in turn, was dependent on the presence of heat shock protein 90 subunit α. Interestingly, DARC- or heat shock protein 90 subunit α-deficient ASMCs responded to CXCL1 stimulation by enhancing p38 MAPK activation and ASMC migration through the CXCR2 receptor. In conclusion, we demonstrated DARC's ability to facilitate CXCL1 inhibition of ASMC migration through modulation of the ERK-1/2 MAPK-signaling pathway.
Collapse
Affiliation(s)
- Laila A Al-Alwan
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Ying Chang
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Simon Rousseau
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - James G Martin
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Qutayba Hamid
- Meakins-Christie Laboratories, Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| |
Collapse
|
16
|
Ayars AG, Altman LC, Potter-Perigo S, Radford K, Wight TN, Nair P. Sputum hyaluronan and versican in severe eosinophilic asthma. Int Arch Allergy Immunol 2012; 161:65-73. [PMID: 23257685 DOI: 10.1159/000343031] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 08/27/2012] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND We examined levels of hyaluronan, a matrix glycosaminoglycan and versican, a matrix proteoglycan, in the sputum of asthmatics treated with mepolizumab (anti-IL-5 monoclonal antibody) versus placebo to evaluate the utility of these measurements as possible biomarkers of asthma control and airway remodeling. METHODS Patients with severe, prednisone-dependent asthma received either mepolizumab or placebo as described in a previously published randomized, double-blind, placebo-controlled study. We measured hyaluronan and versican levels by enzyme-linked immunosorbent assay in sputum collected before and after the 16-week treatment phase. Patients underwent a predefined prednisone tapering schedule if they remained exacerbation free, and sputum eosinophil percentage, asthma control questionnaire (ACQ) and spirometry were monitored. RESULTS After 6 months of mepolizumab therapy and prednisone tapering, there was a significant increase in sputum hyaluronan in the placebo group compared with baseline (p = 0.003). In contrast, there was a significant decrease in sputum hyaluronan in the active treatment group compared with placebo (p = 0.007), which correlated with improvements in percent forced expiratory volume in 1 s (FEV1%) (p = 0.001) and ACQ scores (p = 0.009) as well as a decrease in sputum eosinophils (p = 0.02). There was a nonsignificant increase in sputum versican in the placebo group (p = 0.16), a decrease in the mepolizumab group (p = 0.13) and a significant inverse correlation between versican reduction and FEV1% improvement (p = 0.03). CONCLUSIONS Sputum hyaluronan values are reduced with mepolizumab therapy and correlate with improved clinical and spirometry values, suggesting this measurement may serve as a noninvasive biomarker of asthma control.
Collapse
Affiliation(s)
- Andrew G Ayars
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Wash., USA.
| | | | | | | | | | | |
Collapse
|
17
|
Halwani R, Vazquez-Tello A, Sumi Y, Pureza MA, Bahammam A, Al-Jahdali H, Soussi-Gounni A, Mahboub B, Al-Muhsen S, Hamid Q. Eosinophils induce airway smooth muscle cell proliferation. J Clin Immunol 2012. [PMID: 23180361 DOI: 10.1007/s10875-012-9836-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Asthma is characterized by eosinophilic airway inflammation and remodeling of the airway wall. Features of airway remodeling include increased airway smooth muscle (ASM) mass. However, little is known about the interaction between inflammatory eosinophils and ASM cells. In this study, we investigated the effect of eosinophils on ASM cell proliferation. Eosinophils were isolated from peripheral blood of mild asthmatics and non-asthmatic subjects and co-cultured with human primary ASM cells. ASM proliferation was estimated using Ki-67 expression assay. The expression of extracellular matrix (ECM) mRNA in ASM cells was measured using quantitative real-time PCR. The role of eosinophil derived Cysteinyl Leukotrienes (CysLTs) in enhancing ASM proliferation was estimated by measuring the release of leukotrienes from eosinophils upon their direct contact with ASM cells using ELISA. This role was confirmed either by blocking eosinophil-ASM contact or co-culturing them in the presence of leukotrienes antagonist. ASM cells co-cultured with eosinophils, isolated from asthmatics, but not non-asthmatics, had a significantly higher rate of proliferation compared to controls. This increase in ASM proliferation was independent of their release of ECM proteins but dependent upon eosinophils release of CysLTs. Eosinophil-ASM cell to cell contact was required for CysLTs release. Preventing eosinophil contact with ASM cells using anti-adhesion molecules antibodies, or blocking the activity of eosinophil derived CysLTs using montelukast inhibited ASM proliferation. Our results indicated that eosinophils contribute to airway remodeling during asthma by enhancing ASM cell proliferation and hence increasing ASM mass. Direct contact of eosinophils with ASM cells triggers their release of CysLTs which enhance ASM proliferation. Eosinophils, and their binding to ASM cells, constitute a potential therapeutic target to interfere with the series of biological events leading to airway remodeling and Asthma.
Collapse
Affiliation(s)
- Rabih Halwani
- Asthma Research Chair and Prince Naif Center for Immunology Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Fourtounis J, Wang IM, Mathieu MC, Claveau D, Loo T, Jackson AL, Peters MA, Therien AG, Boie Y, Crackower MA. Gene expression profiling following NRF2 and KEAP1 siRNA knockdown in human lung fibroblasts identifies CCL11/Eotaxin-1 as a novel NRF2 regulated gene. Respir Res 2012; 13:92. [PMID: 23061798 PMCID: PMC3546844 DOI: 10.1186/1465-9921-13-92] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/10/2012] [Indexed: 01/21/2023] Open
Abstract
Background Oxidative Stress contributes to the pathogenesis of many diseases. The NRF2/KEAP1 axis is a key transcriptional regulator of the anti-oxidant response in cells. Nrf2 knockout mice have implicated this pathway in regulating inflammatory airway diseases such as asthma and COPD. To better understand the role the NRF2 pathway has on respiratory disease we have taken a novel approach to define NRF2 dependent gene expression in a relevant lung system. Methods Normal human lung fibroblasts were transfected with siRNA specific for NRF2 or KEAP1. Gene expression changes were measured at 30 and 48 hours using a custom Affymetrix Gene array. Changes in Eotaxin-1 gene expression and protein secretion were further measured under various inflammatory conditions with siRNAs and pharmacological tools. Results An anti-correlated gene set (inversely regulated by NRF2 and KEAP1 RNAi) that reflects specific NRF2 regulated genes was identified. Gene annotations show that NRF2-mediated oxidative stress response is the most significantly regulated pathway, followed by heme metabolism, metabolism of xenobiotics by Cytochrome P450 and O-glycan biosynthesis. Unexpectedly the key eosinophil chemokine Eotaxin-1/CCL11 was found to be up-regulated when NRF2 was inhibited and down-regulated when KEAP1 was inhibited. This transcriptional regulation leads to modulation of Eotaxin-1 secretion from human lung fibroblasts under basal and inflammatory conditions, and is specific to Eotaxin-1 as NRF2 or KEAP1 knockdown had no effect on the secretion of a set of other chemokines and cytokines. Furthermore, the known NRF2 small molecule activators CDDO and Sulphoraphane can also dose dependently inhibit Eotaxin-1 release from human lung fibroblasts. Conclusions These data uncover a previously unknown role for NRF2 in regulating Eotaxin-1 expression and further the mechanistic understanding of this pathway in modulating inflammatory lung disease.
Collapse
Affiliation(s)
- Jimmy Fourtounis
- Department of Respiratory and Immunology, Merck Research Laboratories, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Budd DC, Holmes AM. Targeting TGFβ superfamily ligand accessory proteins as novel therapeutics for chronic lung disorders. Pharmacol Ther 2012; 135:279-91. [PMID: 22722064 DOI: 10.1016/j.pharmthera.2012.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Dysregulation of the transforming growth factor β (TGFβ) pathway has been implicated to underlie a number of disease indications including chronic lung disorders such as asthma, chronic obstructive pulmonary disease (COPD), interstitial pneumonias, and pulmonary arterial hypertension (PAH). Consequently, the pharmaceutical industry has devoted significant resources in the pursuit of TGFβ pathway inhibitors that target the cognate type I and II receptors and respective ligands. The progress of these approaches has been painfully slow, due in part to dose-limiting safety issues that result from the antagonism of a pathway that is responsible for regulating many fundamental biological processes including immune surveillance and cardiovascular responses. These disappointments have led many in the field to conclude that modulating the TGFβ pathway for chronic indications with a sufficient safety window using conventional approaches may be extremely difficult to achieve. Here we review the rationale and limitations of the use of TGFβ pathway inhibitors in chronic lung disorders and the possibility of targeting TGFβ superfamily ligand accessory proteins to allow rheostatic regulation of signaling to achieve efficacy while maintaining a sufficient therapeutic index.
Collapse
Affiliation(s)
- David C Budd
- Respiratory Drug Discovery, Inflammation, Hoffmann-La Roche Inc., Nutley, NJ, USA.
| | | |
Collapse
|
20
|
Bergeron C, Tulic MK, Hamid Q. Airway remodelling in asthma: from benchside to clinical practice. Can Respir J 2010; 17:e85-93. [PMID: 20808979 PMCID: PMC2933777 DOI: 10.1155/2010/318029] [Citation(s) in RCA: 213] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Airway remodelling refers to the structural changes that occur in both large and small airways relevant to miscellaneous diseases including asthma. In asthma, airway structural changes include subepithelial fibrosis, increased smooth muscle mass, gland enlargement, neovascularization and epithelial alterations. Although controversial, airway remodelling is commonly attributed to an underlying chronic inflammatory process. These remodelling changes contribute to thickening of airway walls and, consequently, lead to airway narrowing, bronchial hyper-responsiveness, airway edema and mucous hypersecretion. Airway remodelling is associated with poor clinical outcomes among asthmatic patients. Early diagnosis and prevention of airway remodelling has the potential to decrease disease severity, improve control and prevent disease expression. The relationship between structural changes and clinical and functional abnormalities clearly deserves further investigation. The present review briefly describes the characteristic features of airway remodelling observed in asthma, its clinical consequences and relevance for physicians, and its modulation by therapeutic approaches used in the treatment of asthmatic patients.
Collapse
Affiliation(s)
- Céline Bergeron
- Hotel-Dieu Hospital, Centre Hospitalier de l’Université de Montréal, University of Montreal, Montreal, Quebec
| | - Meri K Tulic
- Division of Cell Biology, Telethon Institute for Child Health Research, Centre for Child Health Research, Perth, Australia
| | - Qutayba Hamid
- Meakins-Christie Laboratories, McGilll University, Montreal, Quebec
| |
Collapse
|
21
|
Abstract
Proteomic approaches have already been successfully implemented in areas such as cancer research. Surprisingly, only a few proteomics analyses have been published reporting on the protein profiles associated with asthma. Although proteomics has its limitations and experimental challenges, it can successfully contribute to the understanding of a complex disease such as asthma. We have reviewed the current literature that has reported the use of proteomic techniques to identify proteins that may contribute to altered lung function in asthma. Only a few of these studies have used proteomic techniques on human tissues associated with asthma, while most research has been performed with animal models of asthma. Proteomic applications have been used as a complimentary technique to verify the suspected candidate proteins involved in asthma. In addition, novel proteins have been identified as potential therapeutic targets. Future collaboration between the different scientific disciplines using proteomic studies of animal models of asthma and confirmation of these findings in human tissues will significantly contribute to the understanding of the etiology of asthma and lead to the development of new therapeutic strategies for this highly prevalent disease.
Collapse
Affiliation(s)
- Annette Osei-Kumah
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, University of Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
22
|
Schauer IG, Ressler SJ, Tuxhorn JA, Dang TD, Rowley DR. Elevated epithelial expression of interleukin-8 correlates with myofibroblast reactive stroma in benign prostatic hyperplasia. Urology 2008; 72:205-13. [PMID: 18314176 PMCID: PMC2528841 DOI: 10.1016/j.urology.2007.11.083] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 10/30/2007] [Accepted: 11/12/2007] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Numerous inflammatory diseases display elevated interleukin (IL)-8, and most are associated with a reactive stroma. IL-8 expression is also elevated in benign prostatic hyperplasia (BPH), yet little is known about reactive stroma in BPH. Whether a reactive stroma response exists in BPH, whether this correlates with elevated IL-8, and whether IL-8 can induce a reactive stroma phenotype have not been determined. This study was designed to specifically address these issues. METHODS Normal prostate transition zone tissue and BPH specimens, as identified by the Baylor College of Medicine pathology department, were examined by quantitative immunohistochemistry to correlate IL-8, smooth muscle alpha-actin, vimentin, calponin, and tenascin-C. In addition, human prostate stromal cell cultures were used to evaluate the effect of IL-8 on the expression of reactive stroma biomarkers. RESULTS BPH nodules exhibited elevated epithelial IL-8 immunoreactivity, and this correlated with elevated smooth muscle alpha-actin, reduced calponin, and altered deposition of tenascin-C, relative to the normal prostate transition zone tissue (P <0.05). Multiple vimentin-positive prostate stromal fibroblast cultures were induced by IL-8 to also co-express smooth muscle alpha-actin and tenascin-C, typical of a reactive stroma myofibroblast phenotype. CONCLUSIONS These data show that BPH reactive stroma is fundamentally different from normal prostate fibromuscular stroma and is typified by the emergence of a reactive stroma myofibroblast phenotype. This reactive stroma pattern correlated spatially with IL-8 elevation in adjacent epithelium. Additionally, IL-8 induced expression of myofibroblast markers in human prostate fibroblasts in vitro. These results suggest that IL-8 acts as a regulator of BPH reactive stroma and is therefore a potential therapeutic target.
Collapse
Affiliation(s)
- Isaiah G. Schauer
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Steven J. Ressler
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | | | - Truong D. Dang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - David R. Rowley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| |
Collapse
|
23
|
Stolz D, Anderson SD, Gysin C, Miedinger D, Surber C, Tamm M, Leuppi JD. Airway reactivity to inhaled mannitol in cigarette smokers: a longitudinal study. Respir Med 2007; 101:1470-6. [PMID: 17324566 DOI: 10.1016/j.rmed.2007.01.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2006] [Accepted: 01/11/2007] [Indexed: 10/23/2022]
Abstract
Smoking induces airway hyperresponsiveness (AHR). Bronchial provocation with mannitol is used to identify AHR in subjects with asthma. This study aimed to determine the prevalence of airway hyperresponsiveness to mannitol in asymptomatic smokers compared to non-smokers and to assess if airway responsiveness to mannitol changes after smoking cessation. Airway responsiveness to inhaled mannitol was measured in smokers (n=42), and non-smokers (n=45). In smokers, the mannitol test was repeated 3 months after smoking cessation. Demographics including age, lung function and atopy status were similar for smokers and non-smokers (p=ns). Compared with non-smokers (2.2%), AHR to mannitol expressed by 15%> or = fall in FEV(1) was significantly more common in smokers (26.2%) (p=0.001). The provoking dose to induce a 15%> or = fall in FEV(1) (PD(15)), a measure of sensitivity, was median [IQR] 291 mg [207-377] in the 11 positive smokers. The response-dose ratio (RDR) (% fall in FEV(1)/cumulative dose), a measure of reactivity, was significantly higher in smokers (0.013 [0.006-0.029]) compared with non-smokers (0.004 [0.002-0.007]), (p<0.0001). After successful smoking cessation, the RDR decreased in most cases (p=0.01) and only one patient still recorded a 15% fall in FEV(1). None of the patients with a negative mannitol test turned positive, irrespective of the outcome of smoking cessation. AHR to mannitol is quite common in smokers compared to non-smokers and decreases significantly after smoking cessation. Thus, the mannitol test may be sensitive to non-asthmatic inflammation of the airways.
Collapse
Affiliation(s)
- Daiana Stolz
- Pulmonary Medicine and Respiratory Cell Research and Hospital Pharmacy, University Hospital Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
24
|
Bossé Y, Maghni K, Hudson TJ. 1alpha,25-dihydroxy-vitamin D3 stimulation of bronchial smooth muscle cells induces autocrine, contractility, and remodeling processes. Physiol Genomics 2007; 29:161-8. [PMID: 17213369 DOI: 10.1152/physiolgenomics.00134.2006] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genetic variants in the vitamin D receptor (VDR) gene were recently associated with asthma. The biological mechanisms explaining this association are unknown but are likely to involve many cell types given the pleiotropic effect of its ligand, 1alpha,25-dihydroxy-vitamin D3 [1alpha,25(OH)2D3]. Considering the prominent role of bronchial smooth muscle cells (BSMCs) in the pathogenesis of asthma, experiments were conducted to explore the gene regulatory effects of 1alpha,25(OH)2D3 in these cells. Using RT-PCR and Western blot, we showed that VDR is present both at the mRNA transcript and protein levels in human BSMCs. The functionality of the receptor was then demonstrated by showing a >200-fold change in the expression of the 24-hydroxylase (CYP24A1) gene following 1alpha,25(OH)2D3 stimulation. Microarray experiments were then performed to identify differentially regulated genes and pathways in BMSCs treated or not with 1alpha,25(OH)2D3. A total of 729 probe sets on the U133 plus 2.0 Affymetrix GeneChip showed fold-change differences above the 1.5 threshold using the Robust Multichip Average intensities. This corresponds to 231 unique genes that were upregulated and 215 unique genes that were down-regulated following 1alpha,25(OH)2D3 stimulation. A high similarity between microarray and real-time PCR results was observed for 13 random genes, with a concordance correlation coefficient of 0.91. Real-time PCR was also performed to confirm the regulation of asthma candidate genes. To identify the biological relevance of this regulation, biological pathways analyses were performed. The most significant network of upregulated genes included genes involved in morphogenesis, cell growth, and survival as well as genes encoding structural proteins, which are potentially involved in airway remodeling.
Collapse
Affiliation(s)
- Yohan Bossé
- McGill University and Génome Québec Innovation Centre, Montréal, Québec, Canada.
| | | | | |
Collapse
|
25
|
Choe MM, Sporn PHS, Swartz MA. Extracellular matrix remodeling by dynamic strain in a three-dimensional tissue-engineered human airway wall model. Am J Respir Cell Mol Biol 2006; 35:306-13. [PMID: 16601241 PMCID: PMC2643283 DOI: 10.1165/rcmb.2005-0443oc] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Airway wall remodeling is a hallmark of asthma, characterized by subepithelial thickening and extracellular matrix (ECM) remodeling. Mechanical stress due to hyperresponsive smooth muscle cells may contribute to this remodeling, but its relevance in a three-dimensional environment (where the ECM plays an important role in modulating stresses felt by cells) is unclear. To characterize the effects of dynamic compression in ECM remodeling in a physiologically relevant three-dimensional environment, a tissue-engineered human airway wall model with differentiated bronchial epithelial cells atop a collagen gel containing lung fibroblasts was used. Lateral compressive strain of 10 or 30% at 1 or 60 cycles per hour was applied using a novel straining device. ECM remodeling was assessed by immunohistochemistry and zymography. Dynamic strain, particularly at the lower magnitude, induced airway wall remodeling, as indicated by increased deposition of types III and IV collagen and increased secretion of matrix metalloproteinase-2 and -9. These changes paralleled increased myofibroblast differentiation and were fibroblast-dependent. Furthermore, the spatial pattern of type III collagen deposition correlated with that of myofibroblasts; both were concentrated near the epithelium and decreased diffusely away from the surface, indicating some epithelial control of the remodeling response. Thus, in a physiologically relevant three-dimensional model of the bronchial wall, dynamic compressive strain induced tissue remodeling that mimics many features of remodeling seen in asthma, in the absence of inflammation and dependent on epithelial-fibroblast signaling.
Collapse
Affiliation(s)
- Melanie M Choe
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, USA
| | | | | |
Collapse
|
26
|
Marlow SL, Blennerhassett MG. Deficient innervation characterizes intestinal strictures in a rat model of colitis. Exp Mol Pathol 2005; 80:54-66. [PMID: 15990093 DOI: 10.1016/j.yexmp.2005.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Accepted: 04/14/2005] [Indexed: 12/16/2022]
Abstract
Intestinal strictures are a common complication of Crohn's disease leading to serious consequences. With unknown etiology and cellular composition, strictures can be neither prevented nor reversed by current therapeutic strategies, and research has been limited by the lack of a well-developed animal model. We observed the sporadic occurrence of intestinal strictures at Day 35 in the TNBS rat model of colitis, which persisted beyond Day 90. Strictured tissue showed fusion, thickening, and disorganization of the smooth muscle layers. Immunocytochemistry revealed that all strictures were characterized by deficient innervation with a complete loss of intrinsic neurons, and a 92% loss of total axons per area. The number of alpha-smooth muscle actin-positive smooth muscle cells (SMC) increased in strictures, but immunolabeling showed phenotypic modulation of these cells, with the SMC phenotype (desmin-positive, vimentin-negative) entirely replaced by a myofibroblast phenotype (desmin-negative, vimentin-positive). Although cellular structure still predominated in the strictured regions, histochemistry showed increased extracellular matrix collagen, from 6 +/- 0.9% to 22 +/- 4% of total area. With previous evidence for neural loss in colitis, and in vitro studies showing neural regulation of smooth muscle cell (SMC) growth, we conclude that the regional loss of innervation may initiate tissue re-modeling that is characteristic of stricture formation.
Collapse
MESH Headings
- Animals
- Axons/metabolism
- Axons/pathology
- Biomarkers/metabolism
- Colitis/complications
- Colitis/pathology
- Collagen/metabolism
- Constriction, Pathologic/metabolism
- Constriction, Pathologic/pathology
- Constriction, Pathologic/physiopathology
- Desmin/metabolism
- Disease Models, Animal
- Enteric Nervous System/pathology
- Intestinal Obstruction/etiology
- Intestinal Obstruction/metabolism
- Intestinal Obstruction/pathology
- Male
- Muscle, Smooth/innervation
- Muscle, Smooth/pathology
- Myoblasts, Smooth Muscle/metabolism
- Myoblasts, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neurons/metabolism
- Neurons/pathology
- Rats
- Rats, Sprague-Dawley
- Vimentin/metabolism
Collapse
Affiliation(s)
- Stacey L Marlow
- Gastrointestinal Diseases Research Unit, Queen's University, Hotel Dieu Hospital, 166 Brock Street, Kingston, Ontario, Canada K7L 5G2
| | | |
Collapse
|
27
|
Anderson SD, Kippelen P. Exercise-induced bronchoconstriction: pathogenesis. Curr Allergy Asthma Rep 2005; 5:116-22. [PMID: 15683611 DOI: 10.1007/s11882-005-0084-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
There is still active debate on the acute mechanism of exercise-induced bronchoconstriction (EIB). Although it is unlikely that vasoconstriction and hyperemia of the bronchial vasculature are essential events for EIB, it is likely that this vasculature enhances the airway response to dehydration and contributes to the pathogenesis of EIB, particularly in elite athletes. Accumulating evidence suggests that airway smooth muscle (ASM) becomes more sensitive as a result of repeated exposure to bulk plasma in response to airway injury from dehydration. Recent evidence also demonstrates sufficient concentrations of mediators that could affect ASM. Paradoxically, mediator release from mast cells may be enhanced and their contractile effects greater when beta(2)-receptor agonists are taken daily. The effect of drugs that have the potential to reduce microvascular leak and reduce or inhibit release or action of these mediators needs to be investigated in elite athletes.
Collapse
Affiliation(s)
- Sandra D Anderson
- Department of Respiratory Medicine, 11 West, Royal Prince Alfred Hospital, Missenden Road, Camperdown NSW 2050, Australia.
| | | |
Collapse
|
28
|
Abstract
Asthma is now the commonest chronic disease in industrialised countries and its prevalence is rising throughout the world. However, despite the availability of very effective treatments many patients with asthma have poorly controlled symptoms and frequent exacerbations, some of which require hospital admission. Important factors contributing to poor asthma control are failure to recognise asthma as a diagnosis, non-implementation of asthma management guidelines and patient non-compliance with therapy. The guidelines themselves have many inherent limitations. Chief among these is that the evidence upon which they are based may be misleading. The evidence used to support recommendations in guidelines is derived from double-blind, placebo-controlled studies in very carefully selected patients who are usually compliant with therapy and instructed in the use of inhalers. In the 'real world' things are very different and patients frequently fail to understand how to use their therapy and are often non-compliant. Reasons for non-compliance are complex and include complicated drug and dosing regimens and inability to use inhaler devices correctly; particularly pressurised metered dose inhalers (pMDIs) due to difficulty co-ordinating device activation and inhalation. Dry powder inhalers (DPIs) preclude the need to co-ordinate inspiration with activation, are easy and convenient to use and are environmentally friendly. There is no doubt that the type of inhaler is just as important as the class of drug in the long-term management of asthma. Improvements in inhaled drug delivery will continue to be important in improving asthma management.
Collapse
Affiliation(s)
- Peter J Barnes
- Department of Thoracic Medicine, National Heart & Lung Institute, Imperial College School of Medicine, Dovehouse Street, London SW3 6LY, UK.
| |
Collapse
|