1
|
Boucher M, Henry C, Gélinas L, Packwood R, Rojas-Ruiz A, Fereydoonzad L, Graham P, Bossé Y. High throughput screening of airway constriction in mouse lung slices. Sci Rep 2024; 14:20133. [PMID: 39210022 PMCID: PMC11362152 DOI: 10.1038/s41598-024-71170-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The level of airway constriction in thin slices of lung tissue is highly variable. Owing to the labor-intensive nature of these experiments, determining the number of airways to be analyzed in order to allocate a reliable value of constriction in one mouse is challenging. Herein, a new automated device for physiology and image analysis was used to facilitate high throughput screening of airway constriction in lung slices. Airway constriction was first quantified in slices of lungs from male BALB/c mice with and without experimental asthma that were inflated with agarose through the trachea or trans-parenchymal injections. Random sampling simulations were then conducted to determine the number of airways required per mouse to quantify maximal constriction. The constriction of 45 ± 12 airways per mouse in 32 mice were analyzed. Mean maximal constriction was 37.4 ± 32.0%. The agarose inflating technique did not affect the methacholine response. However, the methacholine constriction was affected by experimental asthma (p = 0.003), shifting the methacholine concentration-response curve to the right, indicating a decreased sensitivity. Simulations then predicted that approximately 35, 16 and 29 airways per mouse are needed to quantify the maximal constriction mean, standard deviation and coefficient of variation, respectively; these numbers varying between mice and with experimental asthma.
Collapse
Affiliation(s)
- Magali Boucher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Cyndi Henry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Louis Gélinas
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Rosalie Packwood
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Andrés Rojas-Ruiz
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | | | | | - Ynuk Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada.
| |
Collapse
|
2
|
Gomez HM, Haw TJ, Ilic D, Robinson P, Donovan C, Croft AJ, Vanka KS, Small E, Carroll OR, Kim RY, Mayall JR, Beyene T, Palanisami T, Ngo DTM, Zosky GR, Holliday EG, Jensen ME, McDonald VM, Murphy VE, Gibson PG, Horvat JC. Landscape fire smoke airway exposure impairs respiratory and cardiac function and worsens experimental asthma. J Allergy Clin Immunol 2024; 154:209-221.e6. [PMID: 38513838 DOI: 10.1016/j.jaci.2024.02.022] [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: 05/09/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Millions of people are exposed to landscape fire smoke (LFS) globally, and inhalation of LFS particulate matter (PM) is associated with poor respiratory and cardiovascular outcomes. However, how LFS affects respiratory and cardiovascular function is less well understood. OBJECTIVE We aimed to characterize the pathophysiologic effects of representative LFS airway exposure on respiratory and cardiac function and on asthma outcomes. METHODS LFS was generated using a customized combustion chamber. In 8-week-old female BALB/c mice, low (25 μg/m3, 24-hour equivalent) or moderate (100 μg/m3, 24-hour equivalent) concentrations of LFS PM (10 μm and below [PM10]) were administered daily for 3 (short-term) and 14 (long-term) days in the presence and absence of experimental asthma. Lung inflammation, gene expression, structural changes, and lung function were assessed. In 8-week-old male C57BL/6 mice, low concentrations of LFS PM10 were administered for 3 days. Cardiac function and gene expression were assessed. RESULTS Short- and long-term LFS PM10 airway exposure increased airway hyperresponsiveness and induced steroid insensitivity in experimental asthma, independent of significant changes in airway inflammation. Long-term LFS PM10 airway exposure also decreased gas diffusion. Short-term LFS PM10 airway exposure decreased cardiac function and expression of gene changes relating to oxidative stress and cardiovascular pathologies. CONCLUSIONS We characterized significant detrimental effects of physiologically relevant concentrations and durations of LFS PM10 airway exposure on lung and heart function. Our study provides a platform for assessment of mechanisms that underpin LFS PM10 airway exposure on respiratory and cardiovascular disease outcomes.
Collapse
Affiliation(s)
- Henry M Gomez
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tatt J Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Dusan Ilic
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Peter Robinson
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Chantal Donovan
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Kanth S Vanka
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Ellen Small
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tesfalidet Beyene
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Thava Palanisami
- Global Innovative Centre for Advanced Nanomaterials, University of Newcastle, Callaghan, Australia
| | - Doan T M Ngo
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Australia; College of Health and Medicine, Tasmanian School of Medicine, University of Tasmania, Hobart, Australia
| | - Elizabeth G Holliday
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Megan E Jensen
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa M McDonald
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa E Murphy
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Peter G Gibson
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia.
| |
Collapse
|
3
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [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: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
4
|
Thorpe AE, Donovan C, Kim RY, Vindin HJ, Zakarya R, Miyai H, Chan YL, van Reyk D, Chen H, Oliver BG. Third-Hand Exposure to E-Cigarette Vapour Induces Pulmonary Effects in Mice. TOXICS 2023; 11:749. [PMID: 37755759 PMCID: PMC10536515 DOI: 10.3390/toxics11090749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/18/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023]
Abstract
In the last decade, e-cigarette usage has increased, with an estimated 82 million e-cigarette users globally. This is, in part, due to the common opinion that they are "healthier" than tobacco cigarettes or simply "water vapour". Third-hand e-vapour exposure is the chemical residue left behind from e-cigarette aerosols, which is of concern due to its invisible nature, especially among young children. However, there is limited information surrounding third-hand e-vapour exposure. This study aimed to investigate the pulmonary effects of sub-chronic third-hand e-vapour exposure in a murine model. BALB/c mice (4 weeks of age) were exposed to a towel containing nicotine free (0 mg) e-vapour, nicotine (18 mg) e-vapour, or no e-vapour (sham) and replaced daily for 4 weeks. At the endpoint, lung function was assessed, and bronchoalveolar lavage fluid and lungs were collected to measure inflammation and fibrosis. Mice exposed to third-hand e-vapour without nicotine had alveolar enlargement compared to sham exposed controls. Mice exposed to third-hand e-vapour with nicotine had reduced bronchial responsiveness to provocation, increased epithelial thickening in large airways, increased epithelial layers in small airways, alveolar enlargement, and increased small airway collagen deposition, compared to sham exposed controls. In conclusion, our study shows that third-hand e-vapour exposure, particularly in the presence of nicotine, negatively affects the lung health of mice and highlights the need for greater public awareness surrounding the dangers of third-hand exposure to e-cigarette vapour.
Collapse
Affiliation(s)
- Andrew E. Thorpe
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
| | - Chantal Donovan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW 2000, Australia
| | - Richard Y. Kim
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
- Immune Health Program, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW 2000, Australia
| | - Howard J. Vindin
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
- School of Life and Environmental Sciences, Faculty of Science, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Razia Zakarya
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
- Epigenetics of Chronic Disease, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
| | - Hanna Miyai
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
| | - Yik L. Chan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
| | - David van Reyk
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
| | - Brian G. Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia (Y.L.C.); (H.C.)
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie University, Glebe, NSW 2037, Australia
| |
Collapse
|
5
|
Lam M, Lamanna E, Organ L, Donovan C, Bourke JE. Perspectives on precision cut lung slices-powerful tools for investigation of mechanisms and therapeutic targets in lung diseases. Front Pharmacol 2023; 14:1162889. [PMID: 37261291 PMCID: PMC10228656 DOI: 10.3389/fphar.2023.1162889] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/19/2023] [Indexed: 06/02/2023] Open
Abstract
Precision cut lung slices (PCLS) have emerged as powerful experimental tools for respiratory research. Pioneering studies using mouse PCLS to visualize intrapulmonary airway contractility have been extended to pulmonary arteries and for assessment of novel bronchodilators and vasodilators as therapeutics. Additional disease-relevant outcomes, including inflammatory, fibrotic, and regenerative responses, are now routinely measured in PCLS from multiple species, including humans. This review provides an overview of established and innovative uses of PCLS as an intermediary between cellular and organ-based studies and focuses on opportunities to increase their application to investigate mechanisms and therapeutic targets to oppose excessive airway contraction and fibrosis in lung diseases.
Collapse
Affiliation(s)
- Maggie Lam
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Emma Lamanna
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Institut Pasteur, Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Paris, France
| | - Louise Organ
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chantal Donovan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
6
|
Carroll OR, Pillar AL, Brown AC, Feng M, Chen H, Donovan C. Advances in respiratory physiology in mouse models of experimental asthma. Front Physiol 2023; 14:1099719. [PMID: 37008013 PMCID: PMC10060990 DOI: 10.3389/fphys.2023.1099719] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023] Open
Abstract
Recent advances in mouse models of experimental asthma coupled with vast improvements in systems that assess respiratory physiology have considerably increased the accuracy and human relevance of the outputs from these studies. In fact, these models have become important pre-clinical testing platforms with proven value and their capacity to be rapidly adapted to interrogate emerging clinical concepts, including the recent discovery of different asthma phenotypes and endotypes, has accelerated the discovery of disease-causing mechanisms and increased our understanding of asthma pathogenesis and the associated effects on lung physiology. In this review, we discuss key distinctions in respiratory physiology between asthma and severe asthma, including the magnitude of airway hyperresponsiveness and recently discovered disease drivers that underpin this phenomenon such as structural changes, airway remodeling, airway smooth muscle hypertrophy, altered airway smooth muscle calcium signaling, and inflammation. We also explore state-of-the-art mouse lung function measurement techniques that accurately recapitulate the human scenario as well as recent advances in precision cut lung slices and cell culture systems. Furthermore, we consider how these techniques have been applied to recently developed mouse models of asthma, severe asthma, and asthma-chronic obstructive pulmonary disease overlap, to examine the effects of clinically relevant exposures (including ovalbumin, house dust mite antigen in the absence or presence of cigarette smoke, cockroach allergen, pollen, and respiratory microbes) and to increase our understanding of lung physiology in these diseases and identify new therapeutic targets. Lastly, we focus on recent studies that examine the effects of diet on asthma outcomes, including high fat diet and asthma, low iron diet during pregnancy and predisposition to asthma development in offspring, and environmental exposures on asthma outcomes. We conclude our review with a discussion of new clinical concepts in asthma and severe asthma that warrant investigation and how we could utilize mouse models and advanced lung physiology measurement systems to identify factors and mechanisms with potential for therapeutic targeting.
Collapse
Affiliation(s)
- Olivia R. Carroll
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Amber L. Pillar
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Alexandra C. Brown
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Min Feng
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Chantal Donovan
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Chantal Donovan,
| |
Collapse
|
7
|
Tatler AL, Philp CJ, Hill MR, Cox S, Bullock AM, Habgood A, John A, Middlewick R, Stephenson KE, Goodwin AT, Billington CK, O'Dea RD, Johnson SR, Brook BS. Differential remodeling in small and large murine airways revealed by novel whole lung airway analysis. Am J Physiol Lung Cell Mol Physiol 2023; 324:L271-L284. [PMID: 36594851 PMCID: PMC9970660 DOI: 10.1152/ajplung.00034.2022] [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: 01/28/2022] [Revised: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Airway remodeling occurs in chronic asthma leading to increased airway smooth muscle (ASM) mass and extracellular matrix (ECM) deposition. Although extensively studied in murine airways, studies report only selected larger airways at one time-point meaning the spatial distribution and resolution of remodeling are poorly understood. Here we use a new method allowing comprehensive assessment of the spatial and temporal changes in ASM, ECM, and epithelium in large numbers of murine airways after allergen challenge. Using image processing to analyze 20-50 airways per mouse from a whole lung section revealed increases in ASM and ECM after allergen challenge were greater in small and large rather than intermediate airways. ASM predominantly accumulated adjacent to the basement membrane, whereas ECM was distributed across the airway wall. Epithelial hyperplasia was most marked in small and intermediate airways. After challenge, ASM changes resolved over 7 days, whereas ECM and epithelial changes persisted. The new method suggests large and small airways remodel differently, and the long-term consequences of airway inflammation may depend more on ECM and epithelial changes than ASM. The improved quantity and quality of unbiased data provided by the method reveals important spatial differences in remodeling and could set new analysis standards for murine asthma models.
Collapse
Affiliation(s)
- Amanda L Tatler
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Christopher J Philp
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Michael R Hill
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Sam Cox
- Digital Research Service, University of Nottingham, Nottingham, United Kingdom
| | - Andrew M Bullock
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Anthony Habgood
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Alison John
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Robert Middlewick
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Katherine E Stephenson
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Amanda T Goodwin
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte K Billington
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Reuben D O'Dea
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Simon R Johnson
- Centre for Respiratory Research, NIHR Biomedical Research Centre and Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
8
|
Tu X, Gomez HM, Kim RY, Brown AC, de Jong E, Galvao I, Faiz A, Bosco A, Horvat JC, Hansbro P, Donovan C. Airway and parenchyma transcriptomics in a house dust mite model of experimental asthma. Respir Res 2023; 24:32. [PMID: 36698141 PMCID: PMC9878882 DOI: 10.1186/s12931-022-02298-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/15/2022] [Indexed: 01/26/2023] Open
Abstract
Lung transcriptomics studies in asthma have provided valuable information in the whole lung context, however, deciphering the individual contributions of the airway and parenchyma in disease pathogenesis may expedite the development of novel targeted treatment strategies. In this study, we performed transcriptomics on the airway and parenchyma using a house dust mite (HDM)-induced model of experimental asthma that replicates key features of the human disease. HDM exposure increased the expression of 3,255 genes, of which 212 were uniquely increased in the airways, 856 uniquely increased in the parenchyma, and 2187 commonly increased in both compartments. Further interrogation of these genes using a combination of network and transcription factor enrichment analyses identified several transcription factors that regulate airway and/or parenchymal gene expression, including transcription factor EC (TFEC), transcription factor PU.1 (SPI1), H2.0-like homeobox (HLX), metal response element binding transcription factor-1 (MTF1) and E74-like factor 4 (ets domain transcription factor, ELF4) involved in controlling innate immune responses. We next assessed the effects of inhibiting lung SPI1 responses using commercially available DB1976 and DB2313 on key disease outcomes. We found that both compounds had no protective effects on airway inflammation, however DB2313 (8 mg/kg) decreased mucus secreting cell number, and both DB2313 (1 mg/kg) and DB1976 (2.5 mg/kg and 1 mg/kg) reduced small airway collagen deposition. Significantly, both compounds decreased airway hyperresponsiveness. This study demonstrates that SPI1 is important in HDM-induced experimental asthma and that its pharmacological inhibition reduces HDM-induced airway collagen deposition and hyperresponsiveness.
Collapse
Affiliation(s)
- Xiaofan Tu
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Henry M. Gomez
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Richard Y. Kim
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| | - Alexandra C. Brown
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Emma de Jong
- Centre for Health Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA Australia
| | - Izabela Galvao
- grid.117476.20000 0004 1936 7611Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Alen Faiz
- grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| | - Anthony Bosco
- grid.134563.60000 0001 2168 186XAsthma and Airway Disease Research Center, University of Arizona, Arizona, USA
| | - Jay C. Horvat
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Philip Hansbro
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Chantal Donovan
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| |
Collapse
|
9
|
Royce SG, Licciardi PV, Beh RC, Bourke JE, Donovan C, Hung A, Khurana I, Liang JJ, Maxwell S, Mazarakis N, Pitsillou E, Siow YY, Snibson KJ, Tobin MJ, Ververis K, Vongsvivut J, Ziemann M, Samuel CS, Tang MLK, El-Osta A, Karagiannis TC. Sulforaphane prevents and reverses allergic airways disease in mice via anti-inflammatory, antioxidant, and epigenetic mechanisms. Cell Mol Life Sci 2022; 79:579. [PMID: 36319916 PMCID: PMC11803010 DOI: 10.1007/s00018-022-04609-3] [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: 07/01/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022]
Abstract
Sulforaphane has been investigated in human pathologies and preclinical models of airway diseases. To provide further mechanistic insights, we explored L-sulforaphane (LSF) in the ovalbumin (OVA)-induced chronic allergic airways murine model, with key hallmarks of asthma. Histological analysis indicated that LSF prevented or reversed OVA-induced epithelial thickening, collagen deposition, goblet cell metaplasia, and inflammation. Well-known antioxidant and anti-inflammatory mechanisms contribute to the beneficial effects of LSF. Fourier transform infrared microspectroscopy revealed altered composition of macromolecules, following OVA sensitization, which were restored by LSF. RNA sequencing in human peripheral blood mononuclear cells highlighted the anti-inflammatory signature of LSF. Findings indicated that LSF may alter gene expression via an epigenetic mechanism which involves regulation of protein acetylation status. LSF resulted in histone and α-tubulin hyperacetylation in vivo, and cellular and enzymatic assays indicated decreased expression and modest histone deacetylase (HDAC) inhibition activity, in comparison with the well-known pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). Molecular modeling confirmed interaction of LSF and LSF metabolites with the catalytic domain of metal-dependent HDAC enzymes. More generally, this study confirmed known mechanisms and identified potential epigenetic pathways accounting for the protective effects and provide support for the potential clinical utility of LSF in allergic airways disease.
Collapse
Affiliation(s)
- Simon G Royce
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Raymond C Beh
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, 2305, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, 2050, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Ishant Khurana
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Julia J Liang
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Scott Maxwell
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Nadia Mazarakis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Eleni Pitsillou
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Ya Yun Siow
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Kenneth J Snibson
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Mark J Tobin
- ANSTO-Australian Synchrotron, Clayton, VIC, 3168, Australia
| | - Katherine Ververis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia
| | | | - Mark Ziemann
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Warrnambool, VIC, 3216, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Mimi L K Tang
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Population Allergy Group, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Allergy and Immunology, Royal Children's Hospital, Parkville, VIC, 3052, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine Laboratory, Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
10
|
Gomez HM, Pillar AL, Brown AC, Kim RY, Ali MK, Essilfie AT, Vanders RL, Frazer DM, Anderson GJ, Hansbro PM, Collison AM, Jensen ME, Murphy VE, Johnstone DM, Reid D, Milward EA, Donovan C, Horvat JC. Investigating the Links between Lower Iron Status in Pregnancy and Respiratory Disease in Offspring Using Murine Models. Nutrients 2021; 13:nu13124461. [PMID: 34960012 PMCID: PMC8708709 DOI: 10.3390/nu13124461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022] Open
Abstract
Maternal iron deficiency occurs in 40-50% of all pregnancies and is associated with an increased risk of respiratory disease and asthma in children. We used murine models to examine the effects of lower iron status during pregnancy on lung function, inflammation and structure, as well as its contribution to increased severity of asthma in the offspring. A low iron diet during pregnancy impairs lung function, increases airway inflammation, and alters lung structure in the absence and presence of experimental asthma. A low iron diet during pregnancy further increases these major disease features in offspring with experimental asthma. Importantly, a low iron diet increases neutrophilic inflammation, which is indicative of more severe disease, in asthma. Together, our data demonstrate that lower dietary iron and systemic deficiency during pregnancy can lead to physiological, immunological and anatomical changes in the lungs and airways of offspring that predispose to greater susceptibility to respiratory disease. These findings suggest that correcting iron deficiency in pregnancy using iron supplements may play an important role in preventing or reducing the severity of respiratory disease in offspring. They also highlight the utility of experimental models for understanding how iron status in pregnancy affects disease outcomes in offspring and provide a means for testing the efficacy of different iron supplements for preventing disease.
Collapse
Affiliation(s)
- Henry M. Gomez
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Amber L. Pillar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Alexandra C. Brown
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Richard Y. Kim
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Md Khadem Ali
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Ama-Tawiah Essilfie
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
| | - Rebecca L. Vanders
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - David M. Frazer
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Gregory J. Anderson
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Philip M. Hansbro
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Adam M. Collison
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Megan E. Jensen
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Vanessa E. Murphy
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, and Priority Research Centre for GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (A.M.C.); (M.E.J.); (V.E.M.)
| | - Daniel M. Johnstone
- School of Medical Sciences, University of Sydney, Camperdown, NSW 2050, Australia;
| | - David Reid
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (A.-T.E.); (D.M.F.); (G.J.A.); (D.R.)
| | - Elizabeth A. Milward
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
| | - Chantal Donovan
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jay C. Horvat
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW 2308, Australia; (H.M.G.); (A.L.P.); (A.C.B.); (R.Y.K.); (M.K.A.); (R.L.V.); (P.M.H.); (E.A.M.); (C.D.)
- Correspondence: ; Tel.: +612-4042-0220
| |
Collapse
|
11
|
Xue Y, Bao W, Zhou Y, Fu Q, Hao H, Han L, Yin D, Zhang Y, Zhang X, Zhang M. Small-Airway Dysfunction is Involved in the Pathogenesis of Asthma: Evidence from Two Mouse Models. J Asthma Allergy 2021; 14:883-896. [PMID: 34285515 PMCID: PMC8286250 DOI: 10.2147/jaa.s312361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
Background There has been growing evidence of small-airway dysfunction in patients with asthma. Few studies have evaluated the mechanism of small-airway dysfunction in mouse models of asthma. Purpose We explored the correlation between small-airway spirometric variables and large-airway function or inflammation in different endotypes of asthma. Methods Ovalbumin (OVA) sensitization/challenge was used to produce a type 2 (T2)-high asthma model, and OVA combined with ozone exposure (OVA + ozone) was used for the T2-low asthma model with increased neutrophils. Spirometry, airway responsiveness, cytokine levels in bronchoalveolar lavage fluid (BALF), and pathological analyses of lung slices stained with hematoxylin-eosin, periodic acid–Schiff, and Masson’s trichrome stain were all determined. Muc5ac expression in lung tissue was evaluated by the reverse transcription-polymerase chain reaction (RT-PCR), and alpha-smooth muscle actin was measured by immunohistochemistry. Results Inflammatory cells infiltrated the lung tissue and inflammatory cytokines were increased in the BALF of both the OVA and OVA + ozone groups, compared with the control group. Peribronchial hypersecretion and collagen deposition were evident in the models. The OVA + ozone group showed greater neutrophilic infiltration and peribronchial smooth muscle proliferation than the OVA group. Large-airway obstruction, small-airway dysfunction, and airway hyperresponsiveness were confirmed in both models. Small-airway functional variables, such as MMEF (mean midexpiratory flow, average flow from 25 to 75% forced vital capacity [FVC]) and FEF50 (forced expiratory flow at 50% of FVC), were positively correlated with large-airway function and had a stronger negative correlation with airway inflammation, mucus secretion, and responsiveness than large-airway function. Conclusion Small-airway dysfunction was evident in the two endotypes of asthma and was correlated with severe airway inflammation, mucus hypersecretion, and airway hyperresponsiveness. The small airways may be an important target in asthma treatment, and further research in the role of small-airway variables in the pathogenesis of asthma is warranted.
Collapse
Affiliation(s)
- Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Qiang Fu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Huijuan Hao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Dongning Yin
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yingying Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Xue Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| |
Collapse
|
12
|
Boucher M, Henry C, Dufour-Mailhot A, Khadangi F, Bossé Y. Smooth Muscle Hypocontractility and Airway Normoresponsiveness in a Mouse Model of Pulmonary Allergic Inflammation. Front Physiol 2021; 12:698019. [PMID: 34267677 PMCID: PMC8277197 DOI: 10.3389/fphys.2021.698019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/04/2021] [Indexed: 01/25/2023] Open
Abstract
The contractility of airway smooth muscle (ASM) is labile. Although this feature can greatly modulate the degree of airway responsiveness in vivo, the extent by which ASM's contractility is affected by pulmonary allergic inflammation has never been compared between strains of mice exhibiting a different susceptibility to develop airway hyperresponsiveness (AHR). Herein, female C57BL/6 and BALB/c mice were treated intranasally with either saline or house dust mite (HDM) once daily for 10 consecutive days to induce pulmonary allergic inflammation. The doses of HDM were twice greater in the less susceptible C57BL/6 strain. All outcomes, including ASM contractility, were measured 24 h after the last HDM exposure. As expected, while BALB/c mice exposed to HDM became hyperresponsive to a nebulized challenge with methacholine in vivo, C57BL/6 mice remained normoresponsive. The lack of AHR in C57BL/6 mice occurred despite exhibiting more than twice as much inflammation than BALB/c mice in bronchoalveolar lavages, as well as similar degrees of inflammatory cell infiltrates within the lung tissue, goblet cell hyperplasia and thickening of the epithelium. There was no enlargement of ASM caused by HDM exposure in either strain. Unexpectedly, however, excised tracheas derived from C57BL/6 mice exposed to HDM demonstrated a decreased contractility in response to both methacholine and potassium chloride, while tracheas from BALB/c mice remained normocontractile following HDM exposure. These results suggest that the lack of AHR in C57BL/6 mice, at least in an acute model of HDM-induced pulmonary allergic inflammation, is due to an acquired ASM hypocontractility.
Collapse
Affiliation(s)
- Magali Boucher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Cyndi Henry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Alexis Dufour-Mailhot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Fatemeh Khadangi
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Ynuk Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| |
Collapse
|
13
|
Effects of (a Combination of) the Beta 2-Adrenoceptor Agonist Indacaterol and the Muscarinic Receptor Antagonist Glycopyrrolate on Intrapulmonary Airway Constriction. Cells 2021; 10:cells10051237. [PMID: 34069899 PMCID: PMC8157597 DOI: 10.3390/cells10051237] [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: 04/20/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/01/2022] Open
Abstract
Expression of bronchodilatory β2-adrenoceptors and bronchoconstrictive muscarinic M3-receptors alter with airway size. In COPD, (a combination of) β2-agonists and muscarinic M3-antagonists (anticholinergics) are used as bronchodilators. We studied whether differential receptor expression in large and small airways affects the response to β2-agonists and anticholinergics in COPD. Bronchoprotection by indacaterol (β2-agonist) and glycopyrrolate (anticholinergic) against methacholine- and EFS-induced constrictions of large and small airways was measured in guinea pig and human lung slices using video-assisted microscopy. In guinea pig lung slices, glycopyrrolate (1, 3 and 10 nM) concentration-dependently protected against methacholine- and EFS-induced constrictions, with no differences between large and small intrapulmonary airways. Indacaterol (0.01, 0.1, 1 and 10 μM) also provided concentration-dependent protection, which was greater in large airways against methacholine and in small airways against EFS. Indacaterol (10 μM) and glycopyrrolate (10 nM) normalized small airway hyperresponsiveness in COPD lung slices. Synergy of low indacaterol (10 nM) and glycopyrrolate (1 nM) concentrations was greater in LPS-challenged guinea pigs (COPD model) compared to saline-challenged controls. In conclusion, glycopyrrolate similarly protects large and small airways, whereas the protective effect of indacaterol in the small, but not the large, airways depends on the contractile stimulus used. Moreover, findings in a guinea pig model indicate that the synergistic bronchoprotective effect of indacaterol and glycopyrrolate is enhanced in COPD.
Collapse
|
14
|
Yarova PL, Huang P, Schepelmann MW, Bruce R, Ecker R, Nica R, Telezhkin V, Traini D, Gomes Dos Reis L, Kidd EJ, Ford WR, Broadley KJ, Kariuki BM, Corrigan CJ, Ward JPT, Kemp PJ, Riccardi D. Characterization of Negative Allosteric Modulators of the Calcium-Sensing Receptor for Repurposing as a Treatment of Asthma. J Pharmacol Exp Ther 2021; 376:51-63. [PMID: 33115824 DOI: 10.1124/jpet.120.000281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is still an incurable disease, and there is a recognized need for novel small-molecule therapies for people with asthma, especially those poorly controlled by current treatments. We previously demonstrated that calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs), calcilytics, uniquely suppress both airway hyperresponsiveness (AHR) and inflammation in human cells and murine asthma surrogates. Here we assess the feasibility of repurposing four CaSR NAMs, which were originally developed for oral therapy for osteoporosis and previously tested in the clinic as a novel, single, and comprehensive topical antiasthma therapy. We address the hypotheses, using murine asthma surrogates, that topically delivered CaSR NAMs 1) abolish AHR; 2) are unlikely to cause unwanted systemic effects; 3) are suitable for topical application; and 4) inhibit airway inflammation to the same degree as the current standard of care, inhaled corticosteroids, and, furthermore, inhibit airway remodeling. All four CaSR NAMs inhibited poly-L-arginine-induced AHR in naïve mice and suppressed both AHR and airway inflammation in a murine surrogate of acute asthma, confirming class specificity. Repeated exposure to inhaled CaSR NAMs did not alter blood pressure, heart rate, or serum calcium concentrations. Optimal candidates for repurposing were identified based on anti-AHR/inflammatory activities, pharmacokinetics/pharmacodynamics, formulation, and micronization studies. Whereas both inhaled CaSR NAMs and inhaled corticosteroids reduced airways inflammation, only the former prevented goblet cell hyperplasia in a chronic asthma model. We conclude that inhaled CaSR NAMs are likely a single, safe, and effective topical therapy for human asthma, abolishing AHR, suppressing airways inflammation, and abrogating some features of airway remodeling. SIGNIFICANCE STATEMENT: Calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs) reduce airway smooth muscle hyperresponsiveness, reverse airway inflammation as efficiently as topical corticosteroids, and suppress airway remodeling in asthma surrogates. CaSR NAMs, which were initially developed for oral therapy of osteoporosis proved inefficacious for this indication despite being safe and well tolerated. Here we show that structurally unrelated CaSR NAMs are suitable for inhaled delivery and represent a one-stop, steroid-free approach to asthma control and prophylaxis.
Collapse
Affiliation(s)
- Polina L Yarova
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Ping Huang
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Martin W Schepelmann
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Richard Bruce
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Rupert Ecker
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Robert Nica
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Vsevolod Telezhkin
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Daniela Traini
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Larissa Gomes Dos Reis
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Emma J Kidd
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - William R Ford
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Kenneth J Broadley
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Benson M Kariuki
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Christopher J Corrigan
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Jeremy P T Ward
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Paul J Kemp
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Daniela Riccardi
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| |
Collapse
|
15
|
Wang S, Xie Y, Huo YW, Li Y, Abel PW, Jiang H, Zou X, Jiao HZ, Kuang X, Wolff DW, Huang YG, Casale TB, Panettieri RA, Wei T, Cao Z, Tu Y. Airway relaxation mechanisms and structural basis of osthole for improving lung function in asthma. Sci Signal 2020; 13:eaax0273. [PMID: 33234690 PMCID: PMC8720283 DOI: 10.1126/scisignal.aax0273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Overuse of β2-adrenoceptor agonist bronchodilators evokes receptor desensitization, decreased efficacy, and an increased risk of death in asthma patients. Bronchodilators that do not target β2-adrenoceptors represent a critical unmet need for asthma management. Here, we characterize the utility of osthole, a coumarin derived from a traditional Chinese medicine, in preclinical models of asthma. In mouse precision-cut lung slices, osthole relaxed preconstricted airways, irrespective of β2-adrenoceptor desensitization. Osthole administered in murine asthma models attenuated airway hyperresponsiveness, a hallmark of asthma. Osthole inhibited phosphodiesterase 4D (PDE4D) activity to amplify autocrine prostaglandin E2 signaling in airway smooth muscle cells that eventually triggered cAMP/PKA-dependent relaxation of airways. The crystal structure of the PDE4D complexed with osthole revealed that osthole bound to the catalytic site to prevent cAMP binding and hydrolysis. Together, our studies elucidate a specific molecular target and mechanism by which osthole induces airway relaxation. Identification of osthole binding sites on PDE4D will guide further development of bronchodilators that are not subject to tachyphylaxis and would thus avoid β2-adrenoceptor agonist resistance.
Collapse
Affiliation(s)
- Sheng Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Xie
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yan-Wu Huo
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Li
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Peter W Abel
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Haihong Jiang
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Xiaohan Zou
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China
| | - Hai-Zhan Jiao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaolin Kuang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dennis W Wolff
- Kansas City University of Medicine and Biosciences-Joplin, Joplin, MO 64804, USA
| | - You-Guo Huang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Thomas B Casale
- Department of Internal Medicine, University of South Florida School of Medicine, Tampa, FL 33612, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Taotao Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Yaping Tu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
16
|
Dragunas G, Woest ME, Nijboer S, Bos ST, van Asselt J, de Groot AP, Vohlídalová E, Vermeulen CJ, Ditz B, Vonk JM, Koppelman GH, van den Berge M, Ten Hacken NHT, Timens W, Munhoz CD, Prakash YS, Gosens R, Kistemaker LEM. Cholinergic neuroplasticity in asthma driven by TrkB signaling. FASEB J 2020; 34:7703-7717. [PMID: 32277855 PMCID: PMC7302963 DOI: 10.1096/fj.202000170r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Parasympathetic neurons in the airways control bronchomotor tone. Increased activity of cholinergic neurons are mediators of airway hyperresponsiveness (AHR) in asthma, however, mechanisms are not elucidated. We describe remodeling of the cholinergic neuronal network in asthmatic airways driven by brain‐derived neurotrophic factor (BDNF) and Tropomyosin receptor kinase B (TrkB). Human bronchial biopsies were stained for cholinergic marker vesicular acetylcholine transporter (VAChT). Human lung gene expression and single nucleotide polymorphisms (SNP) in neuroplasticity‐related genes were compared between asthma and healthy patients. Wild‐type (WT) and mutated TrkB knock‐in mice (Ntrk2tm1Ddg/J) with impaired BDNF signaling were chronically exposed to ovalbumin (OVA). Neuronal VAChT staining and airway narrowing in response to electrical field stimulation in precision cut lung slices (PCLS) were assessed. Increased cholinergic fibers in asthmatic airway biopsies was found, paralleled by increased TrkB gene expression in human lung tissue, and SNPs in the NTRK2 [TrkB] and BDNF genes linked to asthma. Chronic allergen exposure in mice resulted in increased density of cholinergic nerves, which was prevented by inhibiting TrkB. Increased nerve density resulted in AHR in vivo and in increased nerve‐dependent airway reactivity in lung slices mediated via TrkB. These findings show cholinergic neuroplasticity in asthma driven by TrkB signaling and suggest that the BDNF‐TrkB pathway may be a potential target.
Collapse
Affiliation(s)
- Guilherme Dragunas
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Manon E Woest
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Susan Nijboer
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janet van Asselt
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne P de Groot
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eva Vohlídalová
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Corneel J Vermeulen
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Benedikt Ditz
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Judith M Vonk
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Epidemiology, UMCG, Groningen, the Netherlands
| | - Gerard H Koppelman
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, University of Groningen, Beatrix Children's Hospital, Groningen, the Netherlands
| | - Maarten van den Berge
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Nick H T Ten Hacken
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, UMCG, Groningen, the Netherlands
| | - Wim Timens
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pathology, UMCG, Groningen, the Netherlands
| | - Carolina D Munhoz
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands.,GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
17
|
Ali MK, Kim RY, Brown AC, Donovan C, Vanka KS, Mayall JR, Liu G, Pillar AL, Jones-Freeman B, Xenaki D, Borghuis T, Karim R, Pinkerton JW, Aryal R, Heidari M, Martin KL, Burgess JK, Oliver BG, Trinder D, Johnstone DM, Milward EA, Hansbro PM, Horvat JC. Critical role for iron accumulation in the pathogenesis of fibrotic lung disease. J Pathol 2020; 251:49-62. [PMID: 32083318 DOI: 10.1002/path.5401] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/19/2019] [Accepted: 02/13/2020] [Indexed: 12/18/2022]
Abstract
Increased iron levels and dysregulated iron homeostasis, or both, occur in several lung diseases. Here, the effects of iron accumulation on the pathogenesis of pulmonary fibrosis and associated lung function decline was investigated using a combination of murine models of iron overload and bleomycin-induced pulmonary fibrosis, primary human lung fibroblasts treated with iron, and histological samples from patients with or without idiopathic pulmonary fibrosis (IPF). Iron levels are significantly increased in iron overloaded transferrin receptor 2 (Tfr2) mutant mice and homeostatic iron regulator (Hfe) gene-deficient mice and this is associated with increases in airway fibrosis and reduced lung function. Furthermore, fibrosis and lung function decline are associated with pulmonary iron accumulation in bleomycin-induced pulmonary fibrosis. In addition, we show that iron accumulation is increased in lung sections from patients with IPF and that human lung fibroblasts show greater proliferation and cytokine and extracellular matrix responses when exposed to increased iron levels. Significantly, we show that intranasal treatment with the iron chelator, deferoxamine (DFO), from the time when pulmonary iron levels accumulate, prevents airway fibrosis and decline in lung function in experimental pulmonary fibrosis. Pulmonary fibrosis is associated with an increase in Tfr1+ macrophages that display altered phenotype in disease, and DFO treatment modified the abundance of these cells. These experimental and clinical data demonstrate that increased accumulation of pulmonary iron plays a key role in the pathogenesis of pulmonary fibrosis and lung function decline. Furthermore, these data highlight the potential for the therapeutic targeting of increased pulmonary iron in the treatment of fibrotic lung diseases such as IPF. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University, Stanford, CA, USA.,Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Alexandra C Brown
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Kanth S Vanka
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Amber L Pillar
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Bernadette Jones-Freeman
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Dikaia Xenaki
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Theo Borghuis
- Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Rafia Karim
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - James W Pinkerton
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Respiratory Pharmacology & Toxicology Group, National Heart & Lung Institute, Imperial College London, London, UK
| | - Ritambhara Aryal
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Moones Heidari
- Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Kristy L Martin
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Janette K Burgess
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia.,Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney and School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Debbie Trinder
- Medical School and, Harry Perkins Institute of Medical Research, University of Western Australia, Perth, Australia
| | - Daniel M Johnstone
- Discipline of Physiology and Bosch Institute, University of Sydney, Sydney, Australia
| | - Elizabeth A Milward
- Priority Research Centre for Brain and Mental Health and School of Biomedical Sciences, University of Newcastle, Newcastle, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs and School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| |
Collapse
|
18
|
Regulation of Airway Smooth Muscle Contraction in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:381-422. [PMID: 31183836 DOI: 10.1007/978-981-13-5895-1_16] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Airway smooth muscle (ASM) extends from the trachea throughout the bronchial tree to the terminal bronchioles. In utero, spontaneous phasic contraction of fetal ASM is critical for normal lung development by regulating intraluminal fluid movement, ASM differentiation, and release of key growth factors. In contrast, phasic contraction appears to be absent in the adult lung, and regulation of tonic contraction and airflow is under neuronal and humoral control. Accumulating evidence suggests that changes in ASM responsiveness contribute to the pathophysiology of lung diseases with lifelong health impacts.Functional assessments of fetal and adult ASM and airways have defined pharmacological responses and signaling pathways that drive airway contraction and relaxation. Studies using precision-cut lung slices, in which contraction of intrapulmonary airways and ASM calcium signaling can be assessed simultaneously in situ, have been particularly informative. These combined approaches have defined the relative importance of calcium entry into ASM and calcium release from intracellular stores as drivers of spontaneous phasic contraction in utero and excitation-contraction coupling.Increased contractility of ASM in asthma contributes to airway hyperresponsiveness. Studies using animal models and human ASM and airways have characterized inflammatory and other mechanisms underlying increased reactivity to contractile agonists and reduced bronchodilator efficacy of β2-adrenoceptor agonists in severe diseases. Novel bronchodilators and the application of bronchial thermoplasty to ablate increased ASM within asthmatic airways have the potential to overcome limitations of current therapies. These approaches may directly limit excessive airway contraction to improve outcomes for difficult-to-control asthma and other chronic lung diseases.
Collapse
|
19
|
Maarsingh H, Bidan CM, Brook BS, Zuidhof AB, Elzinga CRS, Smit M, Oldenburger A, Gosens R, Timens W, Meurs H. Small airway hyperresponsiveness in COPD: relationship between structure and function in lung slices. Am J Physiol Lung Cell Mol Physiol 2019; 316:L537-L546. [PMID: 30628486 PMCID: PMC6459292 DOI: 10.1152/ajplung.00325.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The direct relationship between pulmonary structural changes and airway hyperresponsiveness (AHR) in chronic obstructive pulmonary disease (COPD) is unclear. We investigated AHR in relation to airway and parenchymal structural changes in a guinea pig model of COPD and in COPD patients. Precision-cut lung slices (PCLS) were prepared from guinea pigs challenged with lipopolysaccharide or saline two times weekly for 12 wk. Peripheral PCLS were obtained from patients with mild to moderate COPD and non-COPD controls. AHR to methacholine was measured in large and small airways using video-assisted microscopy. Airway smooth muscle mass and alveolar airspace size were determined in the same slices. A mathematical model was used to identify potential changes in biomechanical properties underlying AHR. In guinea pigs, lipopolysaccharide increased the sensitivity of large (>150 μm) airways toward methacholine by 4.4-fold and the maximal constriction of small airways (<150 μm) by 1.5-fold. Similarly increased small airway responsiveness was found in COPD patients. In both lipopolysaccharide-challenged guinea pigs and patients, airway smooth muscle mass was unaltered, whereas increased alveolar airspace correlated with small airway hyperresponsiveness in guinea pigs. Fitting the parameters of the model indicated that COPD weakens matrix mechanical properties and enhances stiffness differences between the airway and the parenchyma, in both species. In conclusion, this study demonstrates small airway hyperresponsiveness in PCLS from COPD patients. These changes may be related to reduced parenchymal retraction forces and biomechanical changes in the airway wall. PCLS from lipopolysaccharide-exposed guinea pigs may be useful to study mechanisms of small airway hyperresponsiveness in COPD.
Collapse
Affiliation(s)
- Harm Maarsingh
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University , West Palm Beach, Florida.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Cécile M Bidan
- Laboratoire Interdisciplinaire de Physique, Centre for Scientific Research, Université Grenoble Alpes , Grenoble , France.,Department of Biomaterials, Max Planck Institute of Colloids and Interfaces , Potsdam , Germany
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham , Nottingham , United Kingdom
| | - Annet B Zuidhof
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Carolina R S Elzinga
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Marieke Smit
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Department of Pathology and Medical Biology, University Medical Center Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Anouk Oldenburger
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Asthma and Chronic Obstructive Pulmonary Disease, University Medical Center Groningen, University of Groningen , Groningen , The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
20
|
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
|
21
|
Dubsky S, Zosky GR, Perks K, Samarage CR, Henon Y, Hooper SB, Fouras A. Assessment of airway response distribution and paradoxical airway dilation in mice during methacholine challenge. J Appl Physiol (1985) 2017; 122:503-510. [DOI: 10.1152/japplphysiol.00476.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 11/22/2022] Open
Abstract
Detailed information on the distribution of airway diameters during bronchoconstriction in situ is required to understand the regional response of the lungs. Imaging studies using computed tomography (CT) have previously measured airway diameters and changes in response to bronchoconstricting agents, but the manual measurements used have severely limited the number of airways measured per subject. Hence, the detailed distribution and heterogeneity of airway responses are unknown. We have developed and applied dynamic imaging and advanced image-processing methods to quantify and compare hundreds of airways in vivo. The method, based on CT, was applied to house dust-mite-sensitized and control mice during intravenous methacholine (MCh) infusion. Airway diameters were measured pre- and post-MCh challenge, and the results compared demonstrate the distribution of airway response throughout the lungs during mechanical ventilation. Forced oscillation testing was used to measure the global response in lung mechanics. We found marked heterogeneity in the response, with paradoxical dilation of airways present at all airway sizes. The probability of paradoxical dilation decreased with decreasing baseline airway diameter and was not affected by pre-existing inflammation. The results confirm the importance of considering the lung as an entire interconnected system rather than a collection of independent units. It is hoped that the response distribution measurements can help to elucidate the mechanisms that lead to heterogeneous airway response in vivo. NEW & NOTEWORTHY Information on the distribution of airway diameters during bronchoconstriction in situ is critical for understanding the regional response of the lungs. We have developed an imaging method to quantify and compare the size of hundreds of airways in vivo during bronchoconstriction in mice. The results demonstrate large heterogeneity with both constriction and paradoxical dilation of airways, confirming the importance of considering the lung as an interconnected system rather than a collection of independent units.
Collapse
Affiliation(s)
- S. Dubsky
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria, Australia
| | - G. R. Zosky
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - K. Perks
- Harry Perkins Institute for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - C. R. Samarage
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria, Australia
- 4Dx Limited, Melbourne, Victoria, Australia
| | - Y. Henon
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria, Australia
- 4Dx Limited, Melbourne, Victoria, Australia
| | - S. B. Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia; and
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - A. Fouras
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria, Australia
- 4Dx Limited, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Lam M, Royce SG, Donovan C, Jelinic M, Parry LJ, Samuel CS, Bourke JE. Serelaxin Elicits Bronchodilation and Enhances β-Adrenoceptor-Mediated Airway Relaxation. Front Pharmacol 2016; 7:406. [PMID: 27833558 PMCID: PMC5081476 DOI: 10.3389/fphar.2016.00406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/13/2016] [Indexed: 12/28/2022] Open
Abstract
Treatment with β-adrenoceptor agonists does not fully overcome the symptoms associated with severe asthma. Serelaxin elicits potent uterine and vascular relaxation via its cognate receptor, RXFP1, and nitric oxide (NO) signaling, and is being clinically evaluated for the treatment of acute heart failure. However, its direct bronchodilator efficacy has yet to be explored. Tracheal rings were prepared from male Sprague-Dawley rats (250–350 g) and tricolor guinea pigs, and precision cut lung slices (PCLSs) containing intrapulmonary airways were prepared from rats only. Recombinant human serelaxin (rhRLX) alone and in combination with rosiglitazone (PPARγ agonist; recently described as a novel dilator) or β-adrenoceptor agonists (isoprenaline, salbutamol) were added either to pre-contracted airways, or before contraction with methacholine or endothelin-1. Regulation of rhRLX responses by epithelial removal, indomethacin (cyclooxygenase inhibitor), L-NAME (nitric oxide synthase inhibitor), SQ22536 (adenylate cyclase inhibitor) and ODQ (guanylate cyclase inhibitor) were also evaluated. Immunohistochemistry was used to localize RXFP1 to airway epithelium and smooth muscle. rhRLX elicited relaxation in rat trachea and PCLS, more slowly than rosiglitazone or isoprenaline, but potentiated relaxation to both these dilators. It markedly increased β-adrenoceptor agonist potency in guinea pig trachea. rhRLX, rosiglitazone, and isoprenaline pretreatment also inhibited the development of rat tracheal contraction. Bronchoprotection by rhRLX increased with longer pre-incubation time, and was partially reduced by epithelial removal, indomethacin and/or L-NAME. SQ22536 and ODQ also partially inhibited rhRLX-mediated relaxation in both intact and epithelial-denuded trachea. RXFP1 expression in the airways was at higher levels in epithelium than smooth muscle. In summary, rhRLX elicits large and small airway relaxation via epithelial-dependent and -independent mechanisms, likely via RXFP1 activation and generation of NO, prostaglandins and cAMP/cGMP. rhRLX also enhanced responsiveness to other dilators, suggesting its potential as an alternative or add-on therapy for severe asthma.
Collapse
Affiliation(s)
- Maggie Lam
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton VIC, Australia
| | - Simon G Royce
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton VIC, Australia
| | - Chantal Donovan
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, ClaytonVIC, Australia; Department of Pharmacology, Lung Health Research Centre, The University of Melbourne, ParkvilleVIC, Australia
| | - Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | - Laura J Parry
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | - Chrishan S Samuel
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton VIC, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, ClaytonVIC, Australia; Department of Pharmacology, Lung Health Research Centre, The University of Melbourne, ParkvilleVIC, Australia
| |
Collapse
|
23
|
Khan MA. Dynamics of airway response in lung microsections: a tool for studying airway-extra cellular matrix interactions. J Biomed Sci 2016; 23:43. [PMID: 27176036 PMCID: PMC4865010 DOI: 10.1186/s12929-016-0263-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/06/2016] [Indexed: 01/27/2023] Open
Abstract
The biological configuration of extracellular matrix (ECM) plays a key role in how mechanical interactions of the airway with its parenchymal attachments affect the dynamics of airway responses in different pulmonary disorders including asthma, emphysema and chronic bronchitis. It is now recognized that mechanical interactions between airway tissue and ECM play a key regulatory role on airway physiology and kinetics that can lead to the reorganization and remodeling of airway connective tissue. A connective tissue is composed of airway smooth muscle cells (ASM) and the ECM, which includes variety of glycoproteins and therefore the extent of interactions between ECM and ASM affects airway dynamics during exacerbations of major pulmonary disorders. Measurement of the velocity and magnitude of airway closure or opening provide important insights into the functions of the airway contractile apparatus and the interactions with its surrounding connective tissues. This review highlights suitability of lung microsection technique in studying measurements of airway dynamics (narrowing/opening) and associated structural distortions in airway compartments.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh, 11211, Kingdom of Saudi Arabiana.
| |
Collapse
|
24
|
Donovan C, Seow HJ, Bourke JE, Vlahos R. Influenza A virus infection and cigarette smoke impair bronchodilator responsiveness to β-adrenoceptor agonists in mouse lung. Clin Sci (Lond) 2016; 130:829-37. [PMID: 27128803 PMCID: PMC5233570 DOI: 10.1042/cs20160093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/14/2016] [Accepted: 02/23/2016] [Indexed: 11/27/2022]
Abstract
β2-adrenoceptor agonists are the mainstay therapy for patients with asthma but their effectiveness in cigarette smoke (CS)-induced lung disease such as chronic obstructive pulmonary disease (COPD) is limited. In addition, bronchodilator efficacy of β2-adrenoceptor agonists is decreased during acute exacerbations of COPD (AECOPD), caused by respiratory viruses including influenza A. Therefore, the aim of the present study was to assess the effects of the β2-adrenoceptor agonist salbutamol (SALB) on small airway reactivity using mouse precision cut lung slices (PCLS) prepared from CS-exposed mice and from CS-exposed mice treated with influenza A virus (Mem71, H3N1). CS exposure alone reduced SALB potency and efficacy associated with decreased β2-adrenoceptor mRNA expression, and increased tumour necrosis factor α (TNFα) and interleukin-1β (IL-1β) expression. This impaired relaxation was restored by day 12 in the absence of further CS exposure. In PCLS prepared after Mem71 infection alone, responses to SALB were transient and were not well maintained. CS exposure prior to Mem71 infection almost completely abolished relaxation, although β2-adrenoceptor and TNFα and IL-1β expression were unaltered. The present study has shown decreased sensitivity to SALB after CS or a combination of CS and Mem71 occurs by different mechanisms. In addition, the PCLS technique and our models of CS and influenza infection provide a novel setting for assessment of alternative bronchodilators.
Collapse
Affiliation(s)
- Chantal Donovan
- Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Huei Jiunn Seow
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| |
Collapse
|
25
|
Methacholine-Induced Variations in Airway Volume and the Slope of the Alveolar Capnogram Are Distinctly Associated with Airflow Limitation and Airway Closure. PLoS One 2015; 10:e0143550. [PMID: 26599006 PMCID: PMC4658077 DOI: 10.1371/journal.pone.0143550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022] Open
Abstract
Mechanisms driving alteration of lung function in response to inhalation of a methacholine aerosol are incompletely understood. To explore to what extent large and small airways contribute to airflow limitation and airway closure in this context, volumetric capnography was performed before (n = 93) and after (n = 78) methacholine provocation in subjects with an intermediate clinical probability of asthma. Anatomical dead space (VDaw), reflecting large airway volume, and the slope of the alveolar capnogram (slope3), an index of ventilation heterogeneity linked to small airway dysfunction, were determined. At baseline, VDaw was positively correlated with lung volumes, FEV1 and peak expiratory flow, while slope3 was not correlated with any lung function index. Variations in VDaw and slope3 following methacholine stimulation were correlated to a small degree (R2 = -0.20). Multivariate regression analysis identified independent associations between variation in FEV1 and variations in both VDaw (Standardized Coefficient-SC = 0.66) and Slope3 (SC = 0.35). By contrast, variation in FVC was strongly associated with variations in VDaw (SC = 0.8) but not Slope3. Thus, alterations in the geometry and/or function of large and small airways were weakly correlated and contributed distinctly to airflow limitation. While both large and small airways contributed to airflow limitation as assessed by FEV1, airway closure as assessed by FVC reduction mostly involved the large airways.
Collapse
|
26
|
Donovan C, Seow HJ, Royce SG, Bourke JE, Vlahos R. Alteration of Airway Reactivity and Reduction of Ryanodine Receptor Expression by Cigarette Smoke in Mice. Am J Respir Cell Mol Biol 2015; 53:471-8. [DOI: 10.1165/rcmb.2014-0400oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
27
|
Donovan C, Bailey SR, Tran J, Haitsma G, Ibrahim ZA, Foster SR, Tang MLK, Royce SG, Bourke JE. Rosiglitazone elicits in vitro relaxation in airways and precision cut lung slices from a mouse model of chronic allergic airways disease. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1219-28. [PMID: 26386117 DOI: 10.1152/ajplung.00156.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/04/2015] [Indexed: 12/14/2022] Open
Abstract
Rosiglitazone (RGZ), a peroxisome proliferator-activated receptor-γ (PPARγ) ligand, is a novel dilator of small airways in mouse precision cut lung slices (PCLS). In this study, relaxation to RGZ and β-adrenoceptor agonists were compared in trachea from naïve mice and guinea pigs and trachea and PCLS from a mouse model of chronic allergic airways disease (AAD). Airways were precontracted with methacholine before addition of PPARγ ligands [RGZ, ciglitazone (CGZ), or 15-deoxy-(Δ12,14)-prostaglandin J2 (15-deoxy-PGJ2)] or β-adrenoceptor agonists (isoprenaline and salbutamol). The effects of T0070907 and GW9662 (PPARγ antagonists) or epithelial removal on relaxation were assessed. Changes in force of trachea and lumen area in PCLS were measured using preparations from saline-challenged mice and mice sensitized (days 0 and 14) and challenged with ovalbumin (3 times/wk, 6 wk). RGZ and CGZ elicited complete relaxation with greater efficacy than β-adrenoceptor agonists in mouse airways but not guinea pig trachea, while 15-deoxy-PGJ2 did not mediate bronchodilation. Relaxation to RGZ was not prevented by T0070907 or GW9662 or by epithelial removal. RGZ-induced relaxation was preserved in the trachea and increased in PCLS after ovalbumin-challenge. Although RGZ was less potent than β-adrenoceptor agonists, its effects were additive with salbutamol and isoprenaline and only RGZ maintained potency and full efficacy in maximally contracted airways or after allergen challenge. Acute PPARγ-independent, epithelial-independent airway relaxation to RGZ is resistant to functional antagonism and maintained in both trachea and PCLS from a model of chronic AAD. These novel efficacious actions of RGZ support its therapeutic potential in asthma when responsiveness to β-adrenoceptor agonists is limited.
Collapse
Affiliation(s)
- Chantal Donovan
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Australia; Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Simon R Bailey
- Faculty of Veterinary Science, University of Melbourne, Parkville, Australia; and
| | - Jenny Tran
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Gertruud Haitsma
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Zaridatul A Ibrahim
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Simon R Foster
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Mimi L K Tang
- Department of Allergy and Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Simon G Royce
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Australia; Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia; Department of Allergy and Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Australia; Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia;
| |
Collapse
|
28
|
Donovan C, Royce SG, Vlahos R, Bourke JE. Lipopolysaccharide does not alter small airway reactivity in mouse lung slices. PLoS One 2015; 10:e0122069. [PMID: 25822969 PMCID: PMC4379153 DOI: 10.1371/journal.pone.0122069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/13/2015] [Indexed: 11/19/2022] Open
Abstract
The bacterial endotoxin, lipopolysaccharide (LPS) has been associated with occupational airway diseases with asthma-like symptoms and in acute exacerbations of COPD. The direct and indirect effects of LPS on small airway reactivity have not been fully elucidated. We tested the hypothesis that both in vitro and in vivo LPS treatment would increase contraction and impair relaxation of mouse small airways. Lung slices were prepared from naïve Balb/C mice and cultured in the absence or presence of LPS (10 μg/ml) for up to 48 h for measurement of TNFα levels in conditioned media. Alternatively, mice were challenged with PBS or LPS in vivo once a day for 4 days for preparation of lung slices or for harvest of lungs for Q-PCR analysis of gene expression of pro-inflammatory cytokines and receptors involved in airway contraction. Reactivity of small airways to contractile agonists, methacholine and serotonin, and bronchodilator agents, salbutamol, isoprenaline and rosiglitazone, were assessed using phase-contrast microscopy. In vitro LPS treatment of slices increased TNFα release 6-fold but did not alter contraction or relaxation to any agonists tested. In vivo LPS treatment increased lung gene expression of TNFα, IL-1β and ryanodine receptor isoform 2 more than 5-fold. However there were no changes in reactivity in lung slices from these mice, even when also incubated with LPS ex vivo. Despite evidence of LPS-induced inflammation, neither airway hyperresponsiveness or impaired dilator reactivity were evident. The increase in ryanodine receptor isoform 2, known to regulate calcium signaling in vascular smooth muscle, warrants investigation. Since LPS failed to elicit changes in small airway reactivity in mouse lung slices following in vitro or in vivo treatment, alternative approaches are required to define the potential contribution of this endotoxin to altered small airway reactivity in human lung diseases.
Collapse
Affiliation(s)
- Chantal Donovan
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Simon G. Royce
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
- School of Health Sciences, Health Innovations Research Institute, RMIT University Bundoora, Victoria, Australia
| | - Jane E. Bourke
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
29
|
Onugha H, MacFarlane PM, Mayer CA, Abrah A, Jafri A, Martin RJ. Airway Hyperreactivity Is Delayed after Mild Neonatal Hyperoxic Exposure. Neonatology 2015; 108:65-72. [PMID: 26021677 PMCID: PMC4475446 DOI: 10.1159/000380758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/06/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Wheezing disorders are prominent in former preterm infants beyond the neonatal period. OBJECTIVES We used a neonatal mouse model to investigate the time course of airway hyperreactivity in response to mild (40% oxygen) or severe (70% oxygen) neonatal hyperoxia. METHODS After hyperoxic exposure during the first week of postnatal life, we measured changes in airway reactivity using the in vitro living lung slice preparation at the end of exposure [postnatal day 8 (P8)] and 2 weeks later (P21). This was accompanied by measures of smooth muscle actin, myosin light chain (MLC) and alveolar morphology. RESULTS Neither mild nor severe hyperoxia exposure affected airway reactivity to methacholine at P8 compared to normoxic controls. In contrast, airway reactivity was enhanced at P21 in mice exposed to mild (but not severe) hyperoxia, 2 weeks after exposure ended. This was associated with increased airway α-smooth muscle actin expression at P21 after 40% oxygen exposure without a significant increase in MLC. Alveolar morphology via radial alveolar counts was comparably diminished by both 40 and 70% oxygen at both P8 and P21. CONCLUSIONS These data demonstrate that early mild hyperoxia exposure causes a delayed augmentation of airway reactivity, suggesting a long-term alteration in the trajectory of airway smooth muscle development and consistent with resultant symptomatology.
Collapse
Affiliation(s)
- Harris Onugha
- Division of Neonatology, Rainbow Babies and Children's Hospital, and Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | |
Collapse
|
30
|
Uhlík J, Šimůnková P, Žaloudíková M, Partlová S, Jarkovský J, Vajner L. Airway wall remodeling in young and adult rats with experimentally provoked bronchial asthma. Int Arch Allergy Immunol 2014; 164:289-300. [PMID: 25228052 DOI: 10.1159/000366278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/01/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Airway wall remodeling is a typical finding in patients suffering from bronchial asthma. While morphological changes have been thoroughly described in adults, less is known about such changes in children because of the limited accessibility of relevant material. To overcome this constraint, animal asthma models may be used instead of human specimens. This study examined rats with artificially stimulated chronic asthma-like symptoms. METHODS Brown Norway rats of two age categories (young and adult) were sensitized by ovalbumin (OA), and their intrapulmonary airways (IA) were studied using morphometric and histochemical methods. RESULTS OA administration induced a significant increase in lung resistance in young animals but not in adults. The total IA wall area was significantly increased in both young and adult OA rats. In young animals, thickening of the adventitia played a more crucial role in this increase than it did in adults, in which the mucosa and the submucosa participated to a higher degree. The IA walls of young OA rats had significantly higher levels of infiltrating eosinophils than those of adult OA animals. The multiplication of goblet cells was more pronounced in adult rats, which was associated with a tendency to produce a higher proportion of acidic glycoconjugates. CONCLUSIONS OA stimulation affected the IA of young rats differently than those of adult animals. Changes in the outer IA layer of young rats can be triggered by activated eosinophils; however, stimulated airway epithelium can be a source of factors that influence the inner IA layers in adult rats.
Collapse
Affiliation(s)
- Jiří Uhlík
- Department of Histology and Embryology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | | | | | | | | |
Collapse
|
31
|
Alkhouri H, Rumzhum NN, Rahman MM, FitzPatrick M, de Pedro M, Oliver BG, Bourke JE, Ammit AJ. TLR2 activation causes tachyphylaxis to β2 -agonists in vitro and ex vivo: modelling bacterial exacerbation. Allergy 2014; 69:1215-22. [PMID: 24862376 DOI: 10.1111/all.12449] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Asthma is a widespread chronic health problem exacerbated by common viral and bacterial infections. Further research is required to understand how infection worsens asthma control in order to advance therapeutic options in the future. Recent research has revealed that β2 -adrenergic receptor (β2 -AR) agonists lose bronchodilatory efficacy because the receptor-mediated molecular pathways responsible for their beneficial actions are desensitized by infection. To date, most studies have focussed on viral infection, leaving the impact of bacterial infection on β2 -AR desensitization relatively under-investigated. We address this in this study. METHODS AND RESULTS Utilizing an in vitro model of bacterial exacerbation in airway smooth muscle (ASM) cells, we show that activation of toll-like receptor 2 (TLR2; mimicking bacterial infection) in the presence of an inflammatory stimulus leads to β2 -AR desensitization. This occurs via TLR2-dependent upregulation of cyclooxygenase 2 (COX-2) mRNA expression and increased secretion of PGE2 . Importantly, PGE2 causes heterologous β2 -AR desensitization and reduces cAMP production in response to short-acting (salbutamol) and long-acting (formoterol) β2 -agonists. Thus, bacterial infectious stimuli act in a PGE2 -dependent manner to severely curtail the beneficial actions of β2 -agonists. The impact of β2 -AR desensitization is demonstrated by reduced gene expression of the critical anti-inflammatory molecule MKP-1 in response to β2 -agonists, as well as impaired bronchodilation in a mouse lung slices. CONCLUSIONS Taken together, our results show that, like viruses, bacteria induce prostanoid-dependent β2 -AR desensitization on ASM cells. Notably, COX-2 inhibition with the specific inhibitor celecoxib represses PGE2 secretion, presenting a feasible pharmacological option for treatment of infectious exacerbation in asthma in the future.
Collapse
Affiliation(s)
- H. Alkhouri
- Faculty of Pharmacy; University of Sydney; Sydney NSW Australia
| | - N. N. Rumzhum
- Faculty of Pharmacy; University of Sydney; Sydney NSW Australia
| | - M. M. Rahman
- Faculty of Pharmacy; University of Sydney; Sydney NSW Australia
| | - M. FitzPatrick
- Respiratory Pharmacology Laboratory; Department of Pharmacology and Therapeutics; Lung Health Research Centre; University of Melbourne; Melbourne Vic. Australia
| | - M. de Pedro
- Woolcock Institute of Medical Research; University of Sydney; Sydney NSW Australia
| | - B. G. Oliver
- Woolcock Institute of Medical Research; University of Sydney; Sydney NSW Australia
| | - J. E. Bourke
- Respiratory Pharmacology Laboratory; Department of Pharmacology and Therapeutics; Lung Health Research Centre; University of Melbourne; Melbourne Vic. Australia
- Respiratory Pharmacology Laboratory; Department of Pharmacology; Monash University; Melbourne Vic. Australia
| | - A. J. Ammit
- Faculty of Pharmacy; University of Sydney; Sydney NSW Australia
| |
Collapse
|
32
|
Bourke JE, Bai Y, Donovan C, Esposito JG, Tan X, Sanderson MJ. Novel small airway bronchodilator responses to rosiglitazone in mouse lung slices. Am J Respir Cell Mol Biol 2014; 50:748-56. [PMID: 24188042 DOI: 10.1165/rcmb.2013-0247oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There is a need to identify novel agents that elicit small airway relaxation when β2-adrenoceptor agonists become ineffective in difficult-to-treat asthma. Because chronic treatment with the synthetic peroxisome proliferator activated receptor (PPAR)γ agonist rosiglitazone (RGZ) inhibits airway hyperresponsiveness in mouse models of allergic airways disease, we tested the hypothesis that RGZ causes acute airway relaxation by measuring changes in small airway size in mouse lung slices. Whereas the β-adrenoceptor agonists albuterol (ALB) and isoproterenol induced partial airway relaxation, RGZ reversed submaximal and maximal contraction to methacholine (MCh) and was similarly effective after precontraction with serotonin or endothelin-1. Concentration-dependent relaxation to RGZ was not altered by the β-adrenoceptor antagonist propranolol and was enhanced by ALB. RGZ-induced relaxation was mimicked by other synthetic PPARγ agonists but not by the putative endogenous agonist 15-deoxy-PGJ2 and was not prevented by the PPARγ antagonist GW9662. To induce airway relaxation, RGZ inhibited the amplitude and frequency of MCh-induced Ca(2+) oscillations of airway smooth muscle cells (ASMCs). In addition, RGZ reduced MCh-induced Ca(2+) sensitivity of the ASMCs. Collectively, these findings demonstrate that acute bronchodilator responses induced by RGZ are PPARγ independent, additive with ALB, and occur by the inhibition of ASMC Ca(2+) signaling and Ca(2+) sensitivity. Because RGZ continues to elicit relaxation when β-adrenoceptor agonists have a limited effect, RGZ or related compounds may have potential as bronchodilators for the treatment of difficult asthma.
Collapse
Affiliation(s)
- Jane E Bourke
- 1 Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia; and
| | | | | | | | | | | |
Collapse
|
33
|
Royce SG, Le Saux CJ. Role of caveolin-1 in asthma and chronic inflammatory respiratory diseases. Expert Rev Respir Med 2014; 8:339-47. [PMID: 24742020 DOI: 10.1586/17476348.2014.905915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caveolin-1 (Cav-1) is the major protein present in invaginations of the plasma membrane of cells known as caveolae. Cav-1 is expressed in numerous resident and inflammatory cells implicated in the pathogenesis of asthma and chronic inflammatory respiratory diseases including chronic obstructive pulmonary disease. A remarkable repertoire of functions has been identified for Cav-1 and these extend to, and have relevance to, asthma and chronic inflammatory respiratory diseases. Important processes influenced by Cav-1 include inflammation, fibrosis, smooth muscle contractility, regulation of apoptosis and cell senescence as well as epithelial barrier function and homeostasis. A better understanding of Cav-1 may be useful in developing new therapies for chronic inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Simon G Royce
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
34
|
Donovan C, Simoons M, Esposito J, Ni Cheong J, Fitzpatrick M, Bourke JE. Rosiglitazone is a superior bronchodilator compared to chloroquine and β-adrenoceptor agonists in mouse lung slices. Respir Res 2014; 15:29. [PMID: 24621080 PMCID: PMC3995634 DOI: 10.1186/1465-9921-15-29] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/25/2014] [Indexed: 11/20/2022] Open
Abstract
Background Current therapy for relieving bronchoconstriction may be ineffective in severe asthma, particularly in the small airways. The aim of this study was to further characterise responses to the recently identified novel bronchodilators rosiglitazone (RGZ) and chloroquine (CQ) under conditions where β-adrenoceptor agonist efficacy was limited or impaired in mouse small airways within lung slices. Methods Relaxation to RGZ and CQ was assessed following submaximal methacholine (MCh) pre-contraction, in slices treated overnight with either RGZ, CQ or albuterol (ALB) (to induce β-adrenoceptor desensitization), and in slices treated with caffeine/ryanodine in which contraction is associated with increases in Ca2+ sensitivity in the absence of contractile agonist-induced Ca2+ oscillations. Furthermore, the effects of RGZ, CQ, ALB and isoproterenol (ISO) on the initiation and development of methacholine-induced contraction were also compared. Results RGZ and CQ, but not ALB or ISO, elicited complete relaxation with increasing MCh pre-contraction and maintained their potency and efficacy following β-adrenoceptor desensitization. RGZ, CQ and ALB maintained efficacy following overnight incubation with RGZ or CQ. Relaxation responses to all dilators were generally maintained but delayed after caffeine/ryanodine. Pre-treatment with RGZ, but not CQ, ALB or ISO, reduced MCh potency. Conclusions This study demonstrates the superior effectiveness of RGZ in comparison to CQ and β-adrenoceptor agonists as a dilator of mouse small airways. Further investigation of the mechanisms underlying the relatively greater efficacy of RGZ under these conditions are warranted and should be extended to include studies in human asthmatic airways.
Collapse
Affiliation(s)
| | | | | | | | | | - Jane Elizabeth Bourke
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
35
|
FitzPatrick M, Donovan C, Bourke JE. Prostaglandin E2 elicits greater bronchodilation than salbutamol in mouse intrapulmonary airways in lung slices. Pulm Pharmacol Ther 2013; 28:68-76. [PMID: 24291048 DOI: 10.1016/j.pupt.2013.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/23/2013] [Accepted: 11/18/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Current asthma therapy may not adequately target contraction of smaller intrapulmonary airways, which are a major site of airway obstruction and inflammation. The aim of this study was to characterise responses of mouse intrapulmonary airways to prostaglandin E(2) (PGE(2)) and compare its dilator efficacy with the β(2)-adrenoceptor agonist salbutamol in situ, using lung slices. METHODS Lung slices (150 μm) were prepared from male Balb/C mice. Changes in intrapulmonary airway lumen area were recorded and analysed by phase-contrast microscopy. Relaxation to PGE(2) and salbutamol were assessed following various levels of pre-contraction with methacholine, serotonin or endothelin-1, as well as following overnight incubation with PGE(2) or salbutamol. The mechanism of PGE(2)-mediated relaxation was explored using selective EP antagonists (EP(1/2) AH6809; EP(4) L-161982) and Ca(2+)-permeabilized slices, where airway responses are due to regulation of Ca(2+)-sensitivity alone. RESULTS PGE2 elicited EP(1/2)-mediated relaxation of intrapulmonary airways. PGE(2) was more potent than salbutamol in opposing submaximal pre-contraction to all constrictors tested, and only PGE(2) opposed maximal pre-contraction with endothelin-1. Relaxation to PGE(2) was maintained when contraction to methacholine was mediated via increased Ca(2+)-sensitivity alone. PGE(2) was less sensitive to homologous or heterologous desensitization of its receptors than salbutamol. CONCLUSION The greater efficacy and potency of PGE(2) compared to salbutamol in mouse intrapulmonary airways supports further investigation of the mechanisms underlying this improved dilator responsiveness for the treatment of severe asthma.
Collapse
Affiliation(s)
- M FitzPatrick
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - C Donovan
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.
| | - J E Bourke
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|