51
|
Ainslie GR, Davis M, Ewart L, Lieberman LA, Rowlands DJ, Thorley AJ, Yoder G, Ryan AM. Microphysiological lung models to evaluate the safety of new pharmaceutical modalities: a biopharmaceutical perspective. LAB ON A CHIP 2019; 19:3152-3161. [PMID: 31469131 DOI: 10.1039/c9lc00492k] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The lung is a complex organ; it is both the initial barrier for inhaled agents and the site of metabolism and therapeutic effect for a subset of systemically administered drugs. Comprised of more than 40 cell types that are responsible for various important functions, the lung's complexity contributes to the subsequent challenges in developing complex in vitro co-culture models (also called microphysiological systems (MPS), complex in vitro models or organs-on-a-chip). Although there are multiple considerations and limitations in the development and qualification of such in vitro systems, MPS exhibit great promise in the fields of pharmacology and toxicology. Successful development and implementation of MPS models may enable mechanistic bridging between non-clinical species and humans, and increase clinical relevance of safety endpoints, while decreasing overall animal use. This article summarizes, from a biopharmaceutical industry perspective, essential elements for the development and qualification of lung MPS models. Its purpose is to guide MPS developers and manufacturers to expedite MPS utilization for safety assessment in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Garrett R Ainslie
- Drug Metabolism and Pharmacokinetics, Theravance Biopharma US, Inc, South San Francisco, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Yin LM, Xu YD, Peng LL, Duan TT, Liu JY, Xu Z, Wang WQ, Guan N, Han XJ, Li HY, Pang Y, Wang Y, Chen Z, Zhu W, Deng L, Wu YL, Ge GB, Huang S, Ulloa L, Yang YQ. Transgelin-2 as a therapeutic target for asthmatic pulmonary resistance. Sci Transl Med 2019; 10:10/427/eaam8604. [PMID: 29437149 DOI: 10.1126/scitranslmed.aam8604] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/11/2017] [Accepted: 11/01/2017] [Indexed: 12/24/2022]
Abstract
There is a clinical need for new bronchodilator drugs in asthma, because more than half of asthmatic patients do not receive adequate control with current available treatments. We report that inhibition of metallothionein-2 protein expression in lung tissues causes the increase of pulmonary resistance. Conversely, metallothionein-2 protein is more effective than β2-agonists in reducing pulmonary resistance in rodent asthma models, alleviating tension in tracheal spirals, and relaxing airway smooth muscle cells (ASMCs). Metallothionein-2 relaxes ASMCs via transgelin-2 (TG2) and induces dephosphorylation of myosin phosphatase target subunit 1 (MYPT1). We identify TSG12 as a nontoxic, specific TG2-agonist that relaxes ASMCs and reduces asthmatic pulmonary resistance. In vivo, TSG12 reduces pulmonary resistance in both ovalbumin- and house dust mite-induced asthma in mice. TSG12 induces RhoA phosphorylation, thereby inactivating the RhoA-ROCK-MYPT1-MLC pathway and causing ASMCs relaxation. TSG12 is more effective than β2-agonists in relaxing human ASMCs and pulmonary resistance with potential clinical advantages. These results suggest that TSG12 could be a promising therapeutic approach for treating asthma.
Collapse
Affiliation(s)
- Lei-Miao Yin
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Yu-Dong Xu
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Ling-Ling Peng
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Ting-Ting Duan
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Jia-Yuan Liu
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Zhijian Xu
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen-Qian Wang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Nan Guan
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Xiao-Jie Han
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Hai-Yan Li
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Yu Pang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Yu Wang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Zhaoqiang Chen
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weiliang Zhu
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Jiangsu 213164, China
| | - Ying-Li Wu
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Shuang Huang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Luis Ulloa
- International Laboratory of Neuro-Immunomodulation, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China. .,Center of Immunology and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Yong-Qing Yang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China.
| |
Collapse
|
53
|
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
|
54
|
Modeling Pneumonic Plague in Human Precision-Cut Lung Slices Highlights a Role for the Plasminogen Activator Protease in Facilitating Type 3 Secretion. Infect Immun 2019; 87:IAI.00175-19. [PMID: 31085709 PMCID: PMC6652753 DOI: 10.1128/iai.00175-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/06/2019] [Indexed: 12/30/2022] Open
Abstract
Pneumonic plague is the deadliest form of disease caused by Yersinia pestis Key to the progression of infection is the activity of the plasminogen activator protease Pla. Deletion of Pla results in a decreased Y. pestis bacterial burden in the lung and failure to progress into the lethal proinflammatory phase of disease. While a number of putative functions have been attributed to Pla, its precise role in the pathogenesis of pneumonic plague is yet to be defined. Here, we show that Pla facilitates type 3 secretion into primary alveolar macrophages but not into the commonly used THP-1 cell line. We also establish human precision-cut lung slices as a platform for modeling early host/pathogen interactions during pneumonic plague and solidify the role of Pla in promoting optimal type 3 secretion using primary human tissue with relevant host cell heterogeneity. These results position Pla as a key player in the early host/pathogen interactions that define pneumonic plague and showcase the utility of human precision-cut lung slices as a platform to evaluate pulmonary infection by bacterial pathogens.
Collapse
|
55
|
Liu G, Betts C, Cunoosamy DM, Åberg PM, Hornberg JJ, Sivars KB, Cohen TS. Use of precision cut lung slices as a translational model for the study of lung biology. Respir Res 2019; 20:162. [PMID: 31324219 PMCID: PMC6642541 DOI: 10.1186/s12931-019-1131-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/09/2019] [Indexed: 12/28/2022] Open
Abstract
Animal models remain invaluable for study of respiratory diseases, however, translation of data generated in genetically homogeneous animals housed in a clean and well-controlled environment does not necessarily provide insight to the human disease situation. In vitro human systems such as air liquid interface (ALI) cultures and organ-on-a-chip models have attempted to bridge the divide between animal models and human patients. However, although 3D in nature, these models struggle to recreate the architecture and complex cellularity of the airways and parenchyma, and therefore cannot mimic the complex cell-cell interactions in the lung. To address this issue, lung slices have emerged as a useful ex vivo tool for studying the respiratory responses to inflammatory stimuli, infection, and novel drug compounds. This review covers the practicality of precision cut lung slice (PCLS) generation and benefits of this ex vivo culture system in modeling human lung biology and disease pathogenesis.
Collapse
Affiliation(s)
- Guanghui Liu
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine Betts
- Pathology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Danen M Cunoosamy
- Bioscience, Respiratory Inflammation and Autoimmunity, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Present Address: Sanofi, Cambridge, MA, USA
| | - Per M Åberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jorrit J Hornberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kinga Balogh Sivars
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Taylor S Cohen
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20877, USA.
| |
Collapse
|
56
|
Wu X, van Dijk EM, Bos IST, Kistemaker LEM, Gosens R. Mouse Lung Tissue Slice Culture. Methods Mol Biol 2019; 1940:297-311. [PMID: 30788834 DOI: 10.1007/978-1-4939-9086-3_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Precision-cut lung slices (PCLS) represent an ex vivo model widely used in visualizing interactions between lung structure and function. The major advantage of this technique is that the presence, differentiation state, and localization of the more than 40 cell types that make up the lung are in accordance with the physiological situation found in lung tissue, including the right localization and patterning of extracellular matrix elements. Here we describe the methodology involved in preparing and culturing PCLS followed by detailed practical information about their possible applications.
Collapse
Affiliation(s)
- Xinhui Wu
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eline M van Dijk
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - I Sophie T Bos
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Faculty of Science and Engineering, Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
57
|
Rieg AD, Bünting NA, Cranen C, Suleiman S, Spillner JW, Schnöring H, Schröder T, von Stillfried S, Braunschweig T, Manley PW, Schälte G, Rossaint R, Uhlig S, Martin C. Tyrosine kinase inhibitors relax pulmonary arteries in human and murine precision-cut lung slices. Respir Res 2019; 20:111. [PMID: 31170998 PMCID: PMC6555704 DOI: 10.1186/s12931-019-1074-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/16/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) inhibit the platelet derived growth factor receptor (PDGFR) and gain increasing significance in the therapy of proliferative diseases, e.g. pulmonary arterial hypertension (PAH). Moreover, TKIs relax pulmonary vessels of rats and guinea pigs. So far, it is unknown, whether TKIs exert relaxation in human and murine pulmonary vessels. Thus, we studied the effects of TKIs and the PDGFR-agonist PDGF-BB in precision-cut lung slices (PCLS) from both species. METHODS The vascular effects of imatinib (mice/human) or nilotinib (human) were studied in Endothelin-1 (ET-1) pre-constricted pulmonary arteries (PAs) or veins (PVs) by videomicroscopy. Baseline initial vessel area (IVA) was defined as 100%. With regard to TKI-induced relaxation, K+-channel activation was studied in human PAs (PCLS) and imatinib/nilotinib-related changes of cAMP and cGMP were analysed in human PAs/PVs (ELISA). Finally, the contractile potency of PDGF-BB was explored in PCLS (mice/human). RESULTS Murine PCLS: Imatinib (10 μM) relaxed ET-1-pre-constricted PAs to 167% of IVA. Vice versa, 100 nM PDGF-BB contracted PAs to 60% of IVA and pre-treatment with imatinib or amlodipine prevented PDGF-BB-induced contraction. Murine PVs reacted only slightly to imatinib or PDGF-BB. Human PCLS: 100 μM imatinib or nilotinib relaxed ET-1-pre-constricted PAs to 166% or 145% of IVA, respectively, due to the activation of KATP-, BKCa2+- or Kv-channels. In PVs, imatinib exerted only slight relaxation and nilotinib had no effect. Imatinib and nilotinib increased cAMP in human PAs, but not in PVs. In addition, PDGF-BB contracted human PAs/PVs, which was prevented by imatinib. CONCLUSIONS TKIs relax pre-constricted PAs/PVs from both, mice and humans. In human PAs, the activation of K+-channels and the generation of cAMP are relevant for TKI-induced relaxation. Vice versa, PDGF-BB contracts PAs/PVs (human/mice) due to PDGFR. In murine PAs, PDGF-BB-induced contraction depends on intracellular calcium. So, PDGFR regulates the tone of PAs/PVs. Since TKIs combine relaxant and antiproliferative effects, they may be promising in therapy of PAH.
Collapse
Affiliation(s)
- Annette D Rieg
- Department of Anaesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany.
| | - Nina A Bünting
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Christian Cranen
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Jan W Spillner
- Department of Cardiac and Thoracic Surgery, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Heike Schnöring
- Department of Cardiac and Thoracic Surgery, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Thomas Schröder
- Department of Surgery, Luisenhospital Aachen, Aachen, Germany
| | | | - Till Braunschweig
- Institute of Pathology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | | | - Gereon Schälte
- Department of Anaesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Rolf Rossaint
- Department of Anaesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| |
Collapse
|
58
|
Fernández-Colino A, Iop L, Ventura Ferreira MS, Mela P. Fibrosis in tissue engineering and regenerative medicine: treat or trigger? Adv Drug Deliv Rev 2019; 146:17-36. [PMID: 31295523 DOI: 10.1016/j.addr.2019.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/11/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Fibrosis is a life-threatening pathological condition resulting from a dysfunctional tissue repair process. There is no efficient treatment and organ transplantation is in many cases the only therapeutic option. Here we review tissue engineering and regenerative medicine (TERM) approaches to address fibrosis in the cardiovascular system, the kidney, the lung and the liver. These strategies have great potential to achieve repair or replacement of diseased organs by cell- and material-based therapies. However, paradoxically, they might also trigger fibrosis. Cases of TERM interventions with adverse outcome are also included in this review. Furthermore, we emphasize the fact that, although organ engineering is still in its infancy, the advances in the field are leading to biomedically relevant in vitro models with tremendous potential for disease recapitulation and development of therapies. These human tissue models might have increased predictive power for human drug responses thereby reducing the need for animal testing.
Collapse
|
59
|
Gray ME, Meehan J, Sullivan P, Marland JRK, Greenhalgh SN, Gregson R, Clutton RE, Ward C, Cousens C, Griffiths DJ, Murray A, Argyle D. Ovine Pulmonary Adenocarcinoma: A Unique Model to Improve Lung Cancer Research. Front Oncol 2019; 9:335. [PMID: 31106157 PMCID: PMC6498990 DOI: 10.3389/fonc.2019.00335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer represents a major worldwide health concern; although advances in patient management have improved outcomes for some patients, overall 5-year survival rates are only around 15%. In vitro studies and mouse models are commonly used to study lung cancer and their use has increased the molecular understanding of the disease. Unfortunately, mouse models are poor predictors of clinical outcome and seldom mimic advanced stages of the human disease. Animal models that more accurately reflect human disease are required for progress to be made in improving treatment outcomes and prognosis. Similarities in pulmonary anatomy and physiology potentially make sheep better models for studying human lung function and disease. Ovine pulmonary adenocarcinoma (OPA) is a naturally occurring lung cancer that is caused by the jaagsiekte sheep retrovirus. The disease is endemic in many countries throughout the world and has several features in common with human lung adenocarcinomas, including histological classification and activation of common cellular signaling pathways. Here we discuss the in vivo and in vitro OPA models that are currently available and describe the advantages of using pre-clinical naturally occurring OPA cases as a translational animal model for human lung adenocarcinoma. The challenges and options for obtaining these OPA cases for research purposes, along with their use in developing novel techniques for the evaluation of chemotherapeutic agents or for monitoring the tumor microenvironment in response to treatment, are also discussed.
Collapse
Affiliation(s)
- Mark E. Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
- School of Engineering and Physical Sciences, Institute of Sensors, Signals and Systems, Heriot-Watt University, Edinburgh, United Kingdom
| | - Paul Sullivan
- School of Engineering, Institute for Integrated Micro and Nano Systems, The King's Buildings, Edinburgh, United Kingdom
| | - Jamie R. K. Marland
- School of Engineering, Institute for Integrated Micro and Nano Systems, The King's Buildings, Edinburgh, United Kingdom
| | - Stephen N. Greenhalgh
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rachael Gregson
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard Eddie Clutton
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Carol Ward
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Chris Cousens
- Moredun Research Institute, Pentlands Science Park, Midlothian, United Kingdom
| | - David J. Griffiths
- Moredun Research Institute, Pentlands Science Park, Midlothian, United Kingdom
| | - Alan Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, The King's Buildings, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
60
|
Live imaging of alveologenesis in precision-cut lung slices reveals dynamic epithelial cell behaviour. Nat Commun 2019; 10:1178. [PMID: 30862802 PMCID: PMC6414680 DOI: 10.1038/s41467-019-09067-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/20/2019] [Indexed: 01/14/2023] Open
Abstract
Damage to alveoli, the gas-exchanging region of the lungs, is a component of many chronic and acute lung diseases. In addition, insufficient generation of alveoli results in bronchopulmonary dysplasia, a disease of prematurity. Therefore visualising the process of alveolar development (alveologenesis) is critical for our understanding of lung homeostasis and for the development of treatments to repair and regenerate lung tissue. Here we show live alveologenesis, using long-term, time-lapse imaging of precision-cut lung slices. We reveal that during this process, epithelial cells are highly mobile and we identify specific cell behaviours that contribute to alveologenesis: cell clustering, hollowing and cell extension. Using the cytoskeleton inhibitors blebbistatin and cytochalasin D, we show that cell migration is a key driver of alveologenesis. This study reveals important novel information about lung biology and provides a new system in which to manipulate alveologenesis genetically and pharmacologically. The process of alveologenesis is incompletely understood, partly due to the lack of applicable real-time imaging methods. Here the authors describe the process of alveologenesis and the behaviour of epithelial cells in real-time, using widefield microscopy and image deconvolution in precision-cut lung slices, revealing the dominant role of epithelial cell migration.
Collapse
|
61
|
Yang L, Feuchtinger A, Möller W, Ding Y, Kutschke D, Möller G, Schittny JC, Burgstaller G, Hofmann W, Stoeger T, Walch A, Schmid O. Three-Dimensional Quantitative Co-Mapping of Pulmonary Morphology and Nanoparticle Distribution with Cellular Resolution in Nondissected Murine Lungs. ACS NANO 2019; 13:1029-1041. [PMID: 30566327 DOI: 10.1021/acsnano.8b07524] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Deciphering biodistribution, biokinetics, and biological effects of nanoparticles (NPs) in entire organs with cellular resolution remains largely elusive due to the lack of effective imaging tools. Here, light sheet fluorescence microscopy in combination with optical tissue clearing was validated for concomitant three-dimensional mapping of lung morphology and NP biodistribution with cellular resolution in nondissected ex vivo murine lungs. Tissue autofluorescence allowed for label-free, quantitative morphometry of the entire bronchial tree, acinar structure, and blood vessels. Co-registration of fluorescent NPs with lung morphology revealed significant differences in pulmonary NP distribution depending on the means of application (intratracheal instillation and ventilator-assisted aerosol inhalation under anesthetized conditions). Inhalation exhibited a more homogeneous NP distribution in conducting airways and acini indicated by a central-to-peripheral (C/P) NP deposition ratio of unity (0.98 ± 0.13) as compared to a 2-fold enhanced central deposition (C/P = 1.98 ± 0.37) for instillation. After inhalation most NPs were observed in the proximal part of the acini as predicted by computational fluid dynamics simulations. At cellular resolution patchy NP deposition was visualized in bronchioles and acini, but more pronounced for instillation. Excellent linearity of the fluorescence intensity-dose response curve allowed for accurate NP dosimetry and revealed ca. 5% of the inhaled aerosol was deposited in the lungs. This single-modality imaging technique allows for quantitative co-registration of tissue architecture and NP biodistribution, which could accelerate elucidation of NP biokinetics and bioactivity within intact tissues, facilitating both nanotoxicology studies and the development of nanomedicines.
Collapse
Affiliation(s)
- Lin Yang
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
- Faculty of Medicine , Technical University of Munich , Munich , 80333 , Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology , Helmholtz Zentrum München , Neuherberg , 85764 , Germany
| | - Winfried Möller
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| | - Yaobo Ding
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| | - David Kutschke
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| | - Gabriele Möller
- Department Genome Analysis Center , Institute of Experimental Genetics, Helmholtz Zentrum München , Neuherberg , 85764 , Germany
| | | | - Gerald Burgstaller
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| | - Werner Hofmann
- Department of Chemistry and Physics of Materials , University of Salzburg , Salzburg , A-5020 , Austria
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| | - Alex Walch
- Research Unit Analytical Pathology , Helmholtz Zentrum München , Neuherberg , 85764 , Germany
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC-M) , Member of the German Center for Lung Research (DZL) , Munich , 81377 , Germany
- Institute of Lung Biology and Disease , Helmholtz Zentrum München-German Research Center for Environmental Health , Neuherberg , 85764 , Germany
| |
Collapse
|
62
|
Hung L, Obernolte H, Sewald K, Eiwegger T. Human ex vivo and in vitro disease models to study food allergy. Asia Pac Allergy 2019; 9:e4. [PMID: 30740352 PMCID: PMC6365658 DOI: 10.5415/apallergy.2019.9.e4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Food allergy is a growing global public health concern. As treatment strategies are currently limited to allergen avoidance and emergency interventions, there is an increasing demand for appropriate models of food allergy for the development of new therapeutics. Many models of food allergy rely heavily on the use of animals, and while useful, many are unable to accurately reflect the human system. In order to bridge the gap between in vivo animal models and clinical trials with human patients, human models of food allergy are of great importance. This review will summarize the commonly used human ex vivo and in vitro models of food allergy and highlight their advantages and limitations regarding how accurately they represent the human in vivo system. We will cover biopsy-based systems, precision cut organ slices, and coculture systems as well as organoids and organ-on-a-chip. The availability of appropriate experimental models will allow us to move forward in the field of food allergy research, to search for effective treatment options and to further explore the cause and progression of this disorder.
Collapse
Affiliation(s)
- Lisa Hung
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Helena Obernolte
- Department of Preclinical Pharmacology and In-Vitro Toxicology, Fraunhofer ITEM, Hannover, Germany
| | - Katherina Sewald
- Department of Preclinical Pharmacology and In-Vitro Toxicology, Fraunhofer ITEM, Hannover, Germany
| | - Thomas Eiwegger
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Clinical Immunology and Allergy, Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
63
|
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
|
64
|
Yilmaz Y, Williams G, Walles M, Manevski N, Krähenbühl S, Camenisch G. Comparison of Rat and Human Pulmonary Metabolism Using Precision-cut Lung Slices (PCLS). Drug Metab Lett 2019; 13:53-63. [PMID: 30345935 DOI: 10.2174/1872312812666181022114622] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/28/2018] [Accepted: 10/08/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Although the liver is the primary organ of drug metabolism, the lungs also contain drug-metabolizing enzymes and may, therefore, contribute to the elimination of drugs. In this investigation, the Precision-cut Lung Slice (PCLS) technique was standardized with the aims of characterizing and comparing rat and human pulmonary drug metabolizing activity. METHOD Due to the limited availability of human lung tissue, standardization of the PCLS method was performed with rat lung tissue. Pulmonary enzymatic activity was found to vary significantly with rat age and rat strain. The Dynamic Organ Culture (DOC) system was superior to well-plates for tissue incubations, while oxygen supply appeared to have a limited impact within the 4h incubation period used here. RESULTS The metabolism of a range of phase I and phase II probe substrates was assessed in rat and human lung preparations. Cytochrome P450 (CYP) activity was relatively low in both species, whereas phase II activity appeared to be more significant. CONCLUSION PCLS is a promising tool for the investigation of pulmonary drug metabolism. The data indicates that pulmonary CYP activity is relatively low and that there are significant differences in enzyme activity between rat and human lung.
Collapse
Affiliation(s)
- Yildiz Yilmaz
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Gareth Williams
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Nenad Manevski
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Stephan Krähenbühl
- Clinical Pharmacology and Toxicology, University Hospital, Basel, Switzerland
| | - Gian Camenisch
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
65
|
Chachi L, Alzahrani A, Koziol-White C, Biddle M, Bagadood R, Panettieri RA, Bradding P, Amrani Y. Increased β2-adrenoceptor phosphorylation in airway smooth muscle in severe asthma: possible role of mast cell-derived growth factors. Clin Exp Immunol 2018; 194:253-258. [PMID: 30069878 DOI: 10.1111/cei.13191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The purpose of this study was to investigate whether growth factors produced by activated human lung mast cells (HLMCs) impair β2 -adrenoceptor (β2 -AR) function in human airway smooth muscle (ASM) cells. Protein array analysis confirmed the presence of various growth factors, including transforming growth factor (TGF)-β1, in the supernatants of high-affinity IgE receptor (FcεRI)-activated HLMCs which, when applied to ASM cells, impaired albuterol-induced cyclic adenosine monophosphate (cAMP) production, an effect that was prevented following neutralization of TGF-β1. This blunted β2 -AR response was reproduced by treating ASM cells with TGF-β1 or fibroblast growth factor (FGF)-2, which induced β2 -AR phosphorylation at tyrosine residues Tyr141 and Tyr350 , and significantly reduced the maximal bronchorelaxant responses to isoproterenol in human precision cut lung slices (PCLS). Finally, ASM cells isolated from severe asthmatics displayed constitutive elevated β2 -AR phosphorylation at both Tyr141 and Tyr350 and a reduced relaxant response to albuterol. This study shows for the first time that abnormal β2 -AR phosphorylation/function in ASM cells that is induced rapidly by HLMC-derived growth factors, is present constitutively in cells from severe asthmatics.
Collapse
Affiliation(s)
- L Chachi
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
| | - A Alzahrani
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK.,Faculty of Applied Medical Sciences, Albaha University, Albaha, Kingdom of Saudi Arabia
| | - C Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ, USA
| | - M Biddle
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
| | - R Bagadood
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
| | - R A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers Biomedical and Health Sciences, Rutgers University, New Brunswick, NJ, USA
| | - P Bradding
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Y Amrani
- Department of Infection, Immunity and Inflammation, Clinical Sciences, University of Leicester, Glenfield Hospital, Leicester, UK
| |
Collapse
|
66
|
Rosales Gerpe MC, van Vloten JP, Santry LA, de Jong J, Mould RC, Pelin A, Bell JC, Bridle BW, Wootton SK. Use of Precision-Cut Lung Slices as an Ex Vivo Tool for Evaluating Viruses and Viral Vectors for Gene and Oncolytic Therapy. Mol Ther Methods Clin Dev 2018; 10:245-256. [PMID: 30112421 PMCID: PMC6092314 DOI: 10.1016/j.omtm.2018.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022]
Abstract
Organotypic slice cultures recapitulate many features of an intact organ, including cellular architecture, microenvironment, and polarity, making them an ideal tool for the ex vivo study of viruses and viral vectors. Here, we describe a procedure for generating precision-cut ovine and murine tissue slices from agarose-perfused normal and murine melanoma tumor-bearing lungs. Furthermore, we demonstrate that these precision-cut lung slices can be maintained up to 1 month and can be used for a range of applications, which include characterizing the tissue tropism of viruses that cannot be propagated in cell monolayers, evaluating the transducing properties of gene therapy vectors, and, finally, investigating the tumor specificity of oncolytic viruses. Our results suggest that ex vivo lung slices are an ideal platform for studying the tissue specificity and cancer cell selectivity of gene therapy vectors and oncolytic viruses prior to in vivo studies, providing justification for pre-clinical work.
Collapse
Affiliation(s)
| | - Jacob P. van Vloten
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lisa A. Santry
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jondavid de Jong
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Robert C. Mould
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Adrian Pelin
- Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - John C. Bell
- Ottawa Hospital Research Institute, Centre for Innovative Cancer Research, Ottawa, ON K1H 8L6, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
67
|
Lacroix G, Koch W, Ritter D, Gutleb AC, Larsen ST, Loret T, Zanetti F, Constant S, Chortarea S, Rothen-Rutishauser B, Hiemstra PS, Frejafon E, Hubert P, Gribaldo L, Kearns P, Aublant JM, Diabaté S, Weiss C, de Groot A, Kooter I. Air-Liquid Interface In Vitro Models for Respiratory Toxicology Research: Consensus Workshop and Recommendations. ACTA ACUST UNITED AC 2018; 4:91-106. [PMID: 32953944 PMCID: PMC7500038 DOI: 10.1089/aivt.2017.0034] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In vitro air-liquid interface (ALI) cell culture models can potentially be used to assess inhalation toxicology endpoints and are usually considered, in terms of relevancy, between classic (i.e., submerged) in vitro models and animal-based models. In some situations that need to be clearly defined, ALI methods may represent a complement or an alternative option to in vivo experimentations or classic in vitro methods. However, it is clear that many different approaches exist and that only very limited validation studies have been carried out to date. This means comparison of data from different methods is difficult and available methods are currently not suitable for use in regulatory assessments. This is despite inhalation toxicology being a priority area for many governmental organizations. In this setting, a 1-day workshop on ALI in vitro models for respiratory toxicology research was organized in Paris in March 2016 to assess the situation and to discuss what might be possible in terms of validation studies. The workshop was attended by major parties in Europe and brought together more than 60 representatives from various academic, commercial, and regulatory organizations. Following plenary, oral, and poster presentations, an expert panel was convened to lead a discussion on possible approaches to validation studies for ALI inhalation models. A series of recommendations were made and the outcomes of the workshop are reported.
Collapse
Affiliation(s)
- Ghislaine Lacroix
- Chronic Risks Division, Institut National de l'Environnement Industriel et des RISques, Verneuil-en-Halatte, France
| | - Wolfgang Koch
- In Vitro und Mechanistische Toxikologie, Fraunhofer ITEM, Hannover, Germany
| | - Detlef Ritter
- In Vitro und Mechanistische Toxikologie, Fraunhofer ITEM, Hannover, Germany
| | - Arno C Gutleb
- Environmental Research and Innovation (ERIN) Department, Luxembourg Institute of Science and Technology, Esch-sur-Alzette, Luxembourg
| | - Søren Thor Larsen
- Inhalation Toxicology Group, National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Thomas Loret
- Chronic Risks Division, Institut National de l'Environnement Industriel et des RISques, Verneuil-en-Halatte, France
| | - Filippo Zanetti
- Systems Toxicology Department, Philip Morris International R&D, Neuchâtel, Switzerland
| | | | - Savvina Chortarea
- BioNanomaterials, Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland.,Laboratory for Materials-Biology Interactions, EMPA, Swiss Federal Laboratories for Materials, Science and Technology, St Gallen, Switzerland
| | | | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emeric Frejafon
- Chronic Risks Division, Institut National de l'Environnement Industriel et des RISques, Verneuil-en-Halatte, France
| | - Philippe Hubert
- Chronic Risks Division, Institut National de l'Environnement Industriel et des RISques, Verneuil-en-Halatte, France
| | - Laura Gribaldo
- Directorate F-Health, Consumers and Reference Materials Chemicals Safety and Alternative Methods Unit (F.3), EURL ECVAM, JRC, Ispra, Italy
| | - Peter Kearns
- Environment, Health and Safety Division, OECD, Paris, France
| | - Jean-Marc Aublant
- European Affairs and Standardization, Laboratoire National de Métrologie et d'Essais, Paris, France
| | - Silvia Diabaté
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Carsten Weiss
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Antoinette de Groot
- Toxicological and Environmental Risk Assessment (TERA) Department, Solvay, Brussels, Belgium
| | - Ingeborg Kooter
- Department of Circular Environment and Environment (CEE), TNO, Utrecht, The Netherlands
| |
Collapse
|
68
|
Niemeyer BF, Zhao P, Tuder RM, Benam KH. Advanced Microengineered Lung Models for Translational Drug Discovery. SLAS DISCOVERY 2018; 23:777-789. [PMID: 29447055 DOI: 10.1177/2472555218760217] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung diseases impose a significant socioeconomic burden and are a leading cause of morbidity and mortality worldwide. Moreover, respiratory medicine, unlike several other therapeutic areas, faces a disappointingly low number of new approved therapies. This is partly due to lack of reliable in vitro or in vivo models that can reproduce organ-level complexity and pathophysiological responses of human lung. Here, we examine new opportunities in application of recently emerged organ-on-chip technology to model human lung alveolus and small airway in preclinical drug development and biomarker discovery. We also discuss challenges that need to be addressed in coming years to further enhance the physiological and clinical relevance of these microsystems, enable their increased accessibility, and support their leap into personalized medicine.
Collapse
Affiliation(s)
- Brian F Niemeyer
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Peng Zhao
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Rubin M Tuder
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Kambez H Benam
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,2 Department of Bioengineering, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
69
|
Lam M, Royce SG, Samuel CS, Bourke JE. Serelaxin as a novel therapeutic opposing fibrosis and contraction in lung diseases. Pharmacol Ther 2018; 187:61-70. [PMID: 29447958 DOI: 10.1016/j.pharmthera.2018.02.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common therapies for asthma and other chronic lung diseases are anti-inflammatory agents and bronchodilators. While these drugs oppose disease symptoms, they do not reverse established structural changes in the airways and their therapeutic efficacy is reduced with increasing disease severity. The peptide hormone, relaxin, is a Relaxin Family Peptide Receptor 1 (RXFP1) receptor agonist with unique combined effects in the lung that differentiates it from these existing therapies. Relaxin has previously been reported to have cardioprotective effects in acute heart failure as well anti-fibrotic actions in several organs. This review focuses on recent experimental evidence of the beneficial effects of chronic relaxin treatment in animal models of airways disease demonstrating inhibition of airway hyperresponsiveness and reversal of established fibrosis, consistent with potential therapeutic benefit. Of particular interest, accumulating evidence demonstrates that relaxin can also acutely oppose contraction by reducing the release of mast cell-derived bronchoconstrictors and by directly eliciting bronchodilation. When used in combination, chronic and acute treatment with relaxin has been shown to enhance responsiveness to both glucocorticoids and β2-adrenoceptor agonists respectively. While the mechanisms underlying these beneficial actions remain to be fully elucidated, translation of these promising combined preclinical findings is critical in the development of relaxin as a novel alternative or adjunct therapeutic opposing multiple aspects of airway pathology in lung diseases.
Collapse
Affiliation(s)
- Maggie Lam
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Department of Pharmacology, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Simon G Royce
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Chrishan S Samuel
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Department of Pharmacology, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Jane E Bourke
- Biomedicine Discovery Institute, Monash University, Melbourne, Australia; Department of Pharmacology, School of Biomedical Sciences, Monash University, Melbourne, Australia.
| |
Collapse
|
70
|
Temann A, Golovina T, Neuhaus V, Thompson C, Chichester JA, Braun A, Yusibov V. Evaluation of inflammatory and immune responses in long-term cultured human precision-cut lung slices. Hum Vaccin Immunother 2017; 13:351-358. [PMID: 27929748 DOI: 10.1080/21645515.2017.1264794] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development of systems that are more accurate and time-efficient in predicting safety and efficacy of target products in humans are critically important in reducing the cost and duration of pharmaceutical development. To circumvent some of the limitations imposed by the use of animal models, ex vivo systems, such as precision-cut lung slices (PCLS), have been proposed as an alternative for evaluating safety, immunogenicity and efficacy of vaccines and pharmaceuticals. In this study, we have established a human PCLS system and methodology for PCLS cultivation that can provide long-term viability and functionality in culture. Using these techniques, we found that cultured PCLS remained viable for at least 14 d in culture and maintained normal metabolic activity, tissue homeostasis and structural integrity. To investigate whether cultured PCLS remained functional, lipopolysaccharide (LPS) was used as a target stimulating compound. We observed that after an 18-hour incubation with LPS, cultured PCLS produced a set of pro-inflammatory cytokines, including TNF-α, IL-1β, IL-6 and IL-10 as well as the enzyme COX-2. Furthermore, cultured PCLS were shown to be capable of generating re-call immune responses, characterized by cytokine production, against antigens commonly found in routine vaccinations against influenza virus and tetanus toxoid. Taken together, these results suggest that human PCLS have the potential to be used as an alternative, high-throughput, ex vivo system for evaluating the safety, and potentially immunogenicity, of vaccines and pharmaceuticals.
Collapse
Affiliation(s)
- Angela Temann
- a Fraunhofer USA Center for Molecular Biotechnology , Newark , DE , USA
| | - Tatiana Golovina
- a Fraunhofer USA Center for Molecular Biotechnology , Newark , DE , USA
| | - Vanessa Neuhaus
- b Fraunhofer Institute for Toxicology and Experimental Medicine , Hannover , Germany
| | - Carolann Thompson
- a Fraunhofer USA Center for Molecular Biotechnology , Newark , DE , USA
| | | | - Armin Braun
- b Fraunhofer Institute for Toxicology and Experimental Medicine , Hannover , Germany
| | - Vidadi Yusibov
- a Fraunhofer USA Center for Molecular Biotechnology , Newark , DE , USA
| |
Collapse
|
71
|
Cook DP, Adam RJ, Zarei K, Deonovic B, Stroik MR, Gansemer ND, Meyerholz DK, Au KF, Stoltz DA. CF airway smooth muscle transcriptome reveals a role for PYK2. JCI Insight 2017; 2:95332. [PMID: 28878137 DOI: 10.1172/jci.insight.95332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 12/17/2022] Open
Abstract
Abnormal airway smooth muscle function can contribute to cystic fibrosis (CF) airway disease. We previously found that airway smooth muscle from newborn CF pigs had increased basal tone, an increased bronchodilator response, and abnormal calcium handling. Since CF pigs lack airway infection and inflammation at birth, these findings suggest intrinsic airway smooth muscle dysfunction in CF. In this study, we tested the hypothesis that CFTR loss in airway smooth muscle would produce a distinct set of changes in the airway smooth muscle transcriptome that we could use to develop novel therapeutic targets. Total RNA sequencing of newborn wild-type and CF airway smooth muscle revealed changes in muscle contraction-related genes, ontologies, and pathways. Using connectivity mapping, we identified several small molecules that elicit transcriptional signatures opposite of CF airway smooth muscle, including NVP-TAE684, an inhibitor of proline-rich tyrosine kinase 2 (PYK2). In CF airway smooth muscle tissue, PYK2 phosphorylation was increased and PYK2 inhibition decreased smooth muscle contraction. In vivo NVP-TAE684 treatment of wild-type mice reduced methacholine-induced airway smooth muscle contraction. These findings suggest that studies in the newborn CF pig may provide an important approach to enhance our understanding of airway smooth muscle biology and for discovery of novel airway smooth muscle therapeutics for CF and other diseases of airway hyperreactivity.
Collapse
Affiliation(s)
- Daniel P Cook
- Department of Internal Medicine.,Department of Molecular Physiology and Biophysics, and
| | - Ryan J Adam
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Keyan Zarei
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Benjamin Deonovic
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | | | - David K Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Kin Fai Au
- Department of Internal Medicine.,Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - David A Stoltz
- Department of Internal Medicine.,Department of Molecular Physiology and Biophysics, and.,Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
72
|
Bai Y, Krishnamoorthy N, Patel KR, Rosas I, Sanderson MJ, Ai X. Cryopreserved Human Precision-Cut Lung Slices as a Bioassay for Live Tissue Banking. A Viability Study of Bronchodilation with Bitter-Taste Receptor Agonists. Am J Respir Cell Mol Biol 2017; 54:656-63. [PMID: 26550921 DOI: 10.1165/rcmb.2015-0290ma] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Human precision-cut lung slices (hPCLSs) provide a unique ex vivo model for translational research. However, the limited and unpredictable availability of human lung tissue greatly impedes their use. Here, we demonstrate that cryopreservation of hPCLSs facilitates banking of live human lung tissue for routine use. Our results show that cryopreservation had little effect on overall cell viability and vital functions of immune cells, including phagocytes and T lymphocytes. In addition, airway contraction and relaxation in response to specific agonists and antagonists, respectively, were unchanged after cryopreservation. At the subcellular level, cryopreserved hPCLSs maintained Ca(2+)-dependent regulatory mechanisms for the control of airway smooth muscle cell contractility. To exemplify the use of cryopreserved hPCLSs in smooth muscle research, we provide evidence that bitter-taste receptor (TAS2R) agonists relax airways by blocking Ca(2+) oscillations in airway smooth muscle cells. In conclusion, the banking of cryopreserved hPCLSs provides a robust bioassay for translational research of lung physiology and disease.
Collapse
Affiliation(s)
- Yan Bai
- 1 Pulmonary, Allergy, Sleep and Critical Care Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Nandini Krishnamoorthy
- 2 Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts; and
| | - Kruti R Patel
- 1 Pulmonary, Allergy, Sleep and Critical Care Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Ivan Rosas
- 2 Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts; and
| | - Michael J Sanderson
- 3 Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Xingbin Ai
- 1 Pulmonary, Allergy, Sleep and Critical Care Medicine, Boston University School of Medicine, Boston, Massachusetts.,2 Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts; and
| |
Collapse
|
73
|
Lyons-Cohen MR, Thomas SY, Cook DN, Nakano H. Precision-cut Mouse Lung Slices to Visualize Live Pulmonary Dendritic Cells. J Vis Exp 2017. [PMID: 28448013 DOI: 10.3791/55465] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inhalation of allergens and pathogens elicits multiple changes in a variety of immune cell types in the lung. Flow cytometry is a powerful technique for quantitative analysis of cell surface proteins on immune cells, but it provides no information on the localization and migration patterns of these cells within the lung. Similarly, chemotaxis assays can be performed to study the potential of cells to respond to chemotactic factors in vitro, but these assays do not reproduce the complex environment of the intact lung. In contrast to these aforementioned techniques, the location of individual cell types within the lung can be readily visualized by generating Precision-cut Lung Slices (PCLS), staining them with commercially available, fluorescently tagged antibodies, and visualizing the sections by confocal microscopy. PCLS can be used for both live and fixed lung tissue, and the slices can encompass areas as large as a cross section of an entire lobe. We have used this protocol to successfully visualize the location of a wide variety of cell types in the lung, including distinct types of dendritic cells, macrophages, neutrophils, T cells and B cells, as well as structural cells such as lymphatic, endothelial, and epithelial cells. The ability to visualize cellular interactions, such as those between dendritic cells and T cells, in live, three-dimensional lung tissue, can reveal how cells move within the lung and interact with one another at steady state and during inflammation. Thus, when used in combination with other procedures, such as flow cytometry and quantitative PCR, PCLS can contribute to a comprehensive understanding of cellular events that underlie allergic and inflammatory diseases of the lung.
Collapse
Affiliation(s)
- Miranda R Lyons-Cohen
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH
| | - Seddon Y Thomas
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH
| | - Donald N Cook
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH
| | - Hideki Nakano
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH;
| |
Collapse
|
74
|
Rosner SR, Pascoe CD, Blankman E, Jensen CC, Krishnan R, James AL, Elliot JG, Green FH, Liu JC, Seow CY, Park JA, Beckerle MC, Paré PD, Fredberg JJ, Smith MA. The actin regulator zyxin reinforces airway smooth muscle and accumulates in airways of fatal asthmatics. PLoS One 2017; 12:e0171728. [PMID: 28278518 PMCID: PMC5344679 DOI: 10.1371/journal.pone.0171728] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/24/2017] [Indexed: 01/21/2023] Open
Abstract
Bronchospasm induced in non-asthmatic human subjects can be easily reversed by a deep inspiration (DI) whereas bronchospasm that occurs spontaneously in asthmatic subjects cannot. This physiological effect of a DI has been attributed to the manner in which a DI causes airway smooth muscle (ASM) cells to stretch, but underlying molecular mechanisms-and their failure in asthma-remain obscure. Using cells and tissues from wild type and zyxin-/- mice we report responses to a transient stretch of physiologic magnitude and duration. At the level of the cytoskeleton, zyxin facilitated repair at sites of stress fiber fragmentation. At the level of the isolated ASM cell, zyxin facilitated recovery of contractile force. Finally, at the level of the small airway embedded with a precision cut lung slice, zyxin slowed airway dilation. Thus, at each level zyxin stabilized ASM structure and contractile properties at current muscle length. Furthermore, when we examined tissue samples from humans who died as the result of an asthma attack, we found increased accumulation of zyxin compared with non-asthmatics and asthmatics who died of other causes. Together, these data suggest a biophysical role for zyxin in fatal asthma.
Collapse
Affiliation(s)
- Sonia R. Rosner
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Christopher D. Pascoe
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Elizabeth Blankman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Christopher C. Jensen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, West Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - John G. Elliot
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, West Australia, Australia
| | - Francis H. Green
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeffrey C. Liu
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Chun Y. Seow
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Jin-Ah Park
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Mary C. Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Peter D. Paré
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Jeffrey J. Fredberg
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Mark A. Smith
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
75
|
Maihöfer NA, Suleiman S, Dreymüller D, Manley PW, Rossaint R, Uhlig S, Martin C, Rieg AD. Imatinib relaxes the pulmonary venous bed of guinea pigs. Respir Res 2017; 18:32. [PMID: 28178968 PMCID: PMC5299687 DOI: 10.1186/s12931-017-0514-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/19/2017] [Indexed: 12/15/2022] Open
Abstract
Background Recently, the IMPRES study revealed that systemic imatinib improves exercise capacity in patients with advanced pulmonary arterial hypertension. Imatinib blocks the tyrosine kinase activity of the platelet-derived growth factor (PDGF)-receptor (PDGFR), acts antiproliferative and relaxes pulmonary arteries. However so far, the relaxant effects of imatinib on pulmonary veins (PVs) and on the postcapillary resistance are unknown, although pulmonary hypertension (PH) due to left heart disease (LHD) is most common and primarily affects PVs. Next, it is unknown whether activation of PDGFR alters the pulmonary venous tone. Due to the reported adverse effects of systemic imatinib, we evaluated the effects of nebulized imatinib on the postcapillary resistance. Methods Precision-cut lung slices (PCLS) were prepared from guinea pigs. PVs were pre-constricted with Endothelin-1 (ET-1) and the imatinib-induced relaxation was studied by videomicroscopy; PDGF-BB-related vascular properties were evaluated as well. The effects of perfused/nebulized imatinib on the postcapillary resistance were studied in cavine isolated perfused lungs (IPL). Intracellular cAMP/cGMP was measured by ELISA in PVs. Results In PCLS, imatinib (100 μM) relaxed pre-constricted PVs (126%). In PVs, imatinib increased cAMP, but not cGMP and inhibition of adenyl cyclase or protein kinase A reduced the imatinib-induced relaxation. Further, inhibition of KATP-channels, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$ {\mathrm{BK}}_{\mathrm{Ca}}^{2+} $$\end{document}BKCa2+-channels or Kv-channels diminished the imatinib-induced relaxation, whereas inhibition of NO-signaling was without effect. In the IPL, perfusion or nebulization of imatinib reduced the ET-1-induced increase of the postcapillary resistance. In PCLS, PDGF-BB contracted PVs, which was blocked by imatinib and by the PDGFR-β kinase inhibitor SU6668, whereas inhibition of PDGFR-α (ponatinib) had no significant effect. Conversely, PDGFR-β kinase inhibitors (SU6668/DMPQ) relaxed PVs pre-constricted with ET-1 comparable to imatinib, whereas the PDGFR-α kinase inhibitor ponatinib did not. Conclusions Imatinib-induced relaxation depends on cAMP and on the activation of K+-channels. Perfused or nebulized imatinib significantly reduces the postcapillary resistance in the pre-constricted (ET-1) pulmonary venous bed. Hence, nebulization of imatinib is feasible and might reduce systemic side effects. Conversely, PDGF-BB contracts PVs by activation of PDGFR-β suggesting that imatinib-induced relaxation depends on PDGFR-β-antagonism. Imatinib combines short-term relaxant and long-term antiproliferative effects. Thus, imatinib might be a promising therapy for PH due to LHD.
Collapse
Affiliation(s)
- Nina A Maihöfer
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Daniela Dreymüller
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | | | - Rolf Rossaint
- Department of Anesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany
| | - Annette D Rieg
- Institute of Pharmacology and Toxicology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany. .,Department of Anesthesiology, Medical Faculty Aachen, RWTH-Aachen, Aachen, Germany.
| |
Collapse
|
76
|
Lohsiriwat V, Scholefield JH, Wilson VG, Dashwood MR. Endothelin-1 and its receptors on haemorrhoidal tissue: a potential site for therapeutic intervention. Br J Pharmacol 2017; 174:569-579. [PMID: 28095606 DOI: 10.1111/bph.13719] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/15/2016] [Accepted: 07/10/2016] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Haemorrhoids is a common anorectal condition affecting millions worldwide. We have studied the effect of endothelin-1 (ET-1) and the role of endothelin ETA and ETB receptors in haemorrhoid tissue. EXPERIMENTAL APPROACH Protein expression of ET-1, ETA and ETB receptors were compared between haemorrhoids and normal rectal submucosa using Western blot analysis, with the localization of proteins determined by autoradiography and immunohistochemistry. Effects of ET-1 and sarafotoxin 6a on human colonic and rectal arteries and veins was assessed by wire myography and the involvement of receptor subtypes established by selective antagonists. KEY RESULTS Dense binding of [125 I]-ET-1 to haemorrhoidal sections was reduced by selective receptor antagonists. A higher density of ETB than ETA receptors was found in haemorrhoidal, than in control rectal tissue and confirmed by Western blot analysis. ETA and ETB receptors were localized to smooth muscle of haemorrhoidal arteries and veins, with ETB receptors on the endothelium. Human colonic and rectal arteries and veins were similarly sensitive to ET-1 and affected by the ETA selective antagonist, but sarafotoxin S6a-induced contractions were more pronounced in veins and antagonized by a selective ETB receptor antagonist. CONCLUSIONS AND IMPLICATIONS ETA and ETB receptors are present in human haemorrhoids with ETB receptors predominating. ETA receptors are activated by ET-1 to mediate a contraction in arteries and veins, but the latter are selectively activated by sarafotoxin S6a - a response that involves ETB receptors at low concentrations. Selective ETB agonists may have therapeutic potential to reduce congestion of the haemorrhoidal venous sinusoids.
Collapse
Affiliation(s)
- Varut Lohsiriwat
- Division of Gastrointestinal Surgery, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - John H Scholefield
- Division of Gastrointestinal Surgery, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Vincent G Wilson
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Michael R Dashwood
- Royal Free Hospital Campus, University College Medical School, London, UK
| |
Collapse
|
77
|
Lilburn DML, Tatler AL, Six JS, Lesbats C, Habgood A, Porte J, Hughes-Riley T, Shaw DE, Jenkins G, Meersmann T. Investigating lung responses with functional hyperpolarized xenon-129 MRI in an ex vivo rat model of asthma. Magn Reson Med 2016; 76:1224-35. [PMID: 26507239 PMCID: PMC5026173 DOI: 10.1002/mrm.26003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 08/26/2015] [Accepted: 09/08/2015] [Indexed: 01/08/2023]
Abstract
PURPOSE Asthma is a disease of increasing worldwide importance that calls for new investigative methods. Ex vivo lung tissue is being increasingly used to study functional respiratory parameters independent of confounding systemic considerations but also to reduce animal numbers and associated research costs. In this work, a straightforward laboratory method is advanced to probe dynamic changes in gas inhalation patterns by using an ex vivo small animal ovalbumin (OVA) model of human asthma. METHODS Hyperpolarized (hp) (129) Xe was actively inhaled by the excised lungs exposed to a constant pressure differential that mimicked negative pleural cavity pressure. The method enabled hp (129) Xe MRI of airway responsiveness to intravenous methacholine (MCh) and airway challenge reversal through salbutamol. RESULTS Significant differences were demonstrated between control and OVA challenged animals on global lung hp (129) Xe gas inhalation with P < 0.05 at MCh dosages above 460 μg. Spatial mapping of the regional hp gas distribution revealed an approximately three-fold increase in heterogeneity for the asthma model organs. CONCLUSION The experimental results from this proof of concept work suggest that the ex vivo hp noble gas imaging arrangement and the applied image analysis methodology may be useful as an adjunct to current diagnostic techniques. Magn Reson Med 76:1224-1235, 2016. © 2015 The Authors. Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- David M L Lilburn
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Amanda L Tatler
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Joseph S Six
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Clémentine Lesbats
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Anthony Habgood
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Joanne Porte
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Theodore Hughes-Riley
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Dominick E Shaw
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Gisli Jenkins
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, University of Nottingham, Nottingham, United Kingdom
| | - Thomas Meersmann
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
78
|
Hiorns JE, Bidan CM, Jensen OE, Gosens R, Kistemaker LEM, Fredberg JJ, Butler JP, Krishnan R, Brook BS. Airway and Parenchymal Strains during Bronchoconstriction in the Precision Cut Lung Slice. Front Physiol 2016; 7:309. [PMID: 27559314 PMCID: PMC4989902 DOI: 10.3389/fphys.2016.00309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/07/2016] [Indexed: 01/25/2023] Open
Abstract
The precision-cut lung slice (PCLS) is a powerful tool for studying airway reactivity, but biomechanical measurements to date have largely focused on changes in airway caliber. Here we describe an image processing tool that reveals the associated spatio-temporal changes in airway and parenchymal strains. Displacements of sub-regions within the PCLS are tracked in phase-contrast movies acquired after addition of contractile and relaxing drugs. From displacement maps, strains are determined across the entire PCLS or along user-specified directions. In a representative mouse PCLS challenged with 10(-4)M methacholine, as lumen area decreased, compressive circumferential strains were highest in the 50 μm closest to the airway lumen while expansive radial strains were highest in the region 50-100 μm from the lumen. However, at any given distance from the airway the strain distribution varied substantially in the vicinity of neighboring small airways and blood vessels. Upon challenge with the relaxant agonist chloroquine, although most strains disappeared, residual positive strains remained a long time after addition of chloroquine, predominantly in the radial direction. Taken together, these findings establish strain mapping as a new tool to elucidate local dynamic mechanical events within the constricting airway and its supporting parenchyma.
Collapse
Affiliation(s)
- Jonathan E Hiorns
- School of Mathematical Sciences, University of Nottingham Nottingham, UK
| | - Cécile M Bidan
- Laboratoire Interdisciplinaire de Physique, Centre National de la Recherche Scientifique, Université Grenoble AlpesGrenoble, France; Department of Molecular Pharmacology, University of GroningenGroningen, Netherlands; Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Oliver E Jensen
- School of Mathematics, University of Manchester Manchester, UK
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands
| | - Jeffrey J Fredberg
- Department of Environmental Health, Harvard School of Public Health Boston, MA, USA
| | - Jim P Butler
- Department of Environmental Health, Harvard School of Public Health Boston, MA, USA
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, MA, USA
| | - Bindi S Brook
- School of Mathematical Sciences, University of Nottingham Nottingham, UK
| |
Collapse
|
79
|
Cook DP, Rector MV, Bouzek DC, Michalski AS, Gansemer ND, Reznikov LR, Li X, Stroik MR, Ostedgaard LS, Abou Alaiwa MH, Thompson MA, Prakash YS, Krishnan R, Meyerholz DK, Seow CY, Stoltz DA. Cystic Fibrosis Transmembrane Conductance Regulator in Sarcoplasmic Reticulum of Airway Smooth Muscle. Implications for Airway Contractility. Am J Respir Crit Care Med 2016; 193:417-26. [PMID: 26488271 DOI: 10.1164/rccm.201508-1562oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE An asthma-like airway phenotype has been described in people with cystic fibrosis (CF). Whether these findings are directly caused by loss of CF transmembrane conductance regulator (CFTR) function or secondary to chronic airway infection and/or inflammation has been difficult to determine. OBJECTIVES Airway contractility is primarily determined by airway smooth muscle. We tested the hypothesis that CFTR is expressed in airway smooth muscle and directly affects airway smooth muscle contractility. METHODS Newborn pigs, both wild type and with CF (before the onset of airway infection and inflammation), were used in this study. High-resolution immunofluorescence was used to identify the subcellular localization of CFTR in airway smooth muscle. Airway smooth muscle function was determined with tissue myography, intracellular calcium measurements, and regulatory myosin light chain phosphorylation status. Precision-cut lung slices were used to investigate the therapeutic potential of CFTR modulation on airway reactivity. MEASUREMENTS AND MAIN RESULTS We found that CFTR localizes to the sarcoplasmic reticulum compartment of airway smooth muscle and regulates airway smooth muscle tone. Loss of CFTR function led to delayed calcium reuptake following cholinergic stimulation and increased myosin light chain phosphorylation. CFTR potentiation with ivacaftor decreased airway reactivity in precision-cut lung slices following cholinergic stimulation. CONCLUSIONS Loss of CFTR alters porcine airway smooth muscle function and may contribute to the airflow obstruction phenotype observed in human CF. Airway smooth muscle CFTR may represent a therapeutic target in CF and other diseases of airway narrowing.
Collapse
Affiliation(s)
- Daniel P Cook
- 1 Department of Internal Medicine.,2 Department of Molecular Physiology and Biophysics
| | | | | | | | | | | | | | | | | | | | - Michael A Thompson
- 3 Department of Anesthesiology and.,4 Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- 3 Department of Anesthesiology and.,4 Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Ramaswamy Krishnan
- 5 Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts; and
| | | | - Chun Y Seow
- 7 Department of Pathology and Laboratory Medicine, James Hogg Research Centre/St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - David A Stoltz
- 1 Department of Internal Medicine.,2 Department of Molecular Physiology and Biophysics.,8 Department of Biomedical Engineering, and.,9 Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa
| |
Collapse
|
80
|
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
|
81
|
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
|
82
|
Konar D, Devarasetty M, Yildiz DV, Atala A, Murphy SV. Lung-On-A-Chip Technologies for Disease Modeling and Drug Development. Biomed Eng Comput Biol 2016; 7:17-27. [PMID: 27127414 PMCID: PMC4839966 DOI: 10.4137/becb.s34252] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 01/13/2023] Open
Abstract
Animal and two-dimensional cell culture models have had a profound impact on not only lung research but also medical research at large, despite inherent flaws and differences when compared with in vivo and clinical observations. Three-dimensional (3D) tissue models are a natural progression and extension of existing techniques that seek to plug the gaps and mitigate the drawbacks of two-dimensional and animal technologies. In this review, we describe the transition of historic models to contemporary 3D cell and organoid models, the varieties of current 3D cell and tissue culture modalities, the common methods for imaging these models, and finally, the applications of these models and imaging techniques to lung research.
Collapse
Affiliation(s)
- Dipasri Konar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Didem V Yildiz
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
83
|
Mapping physiological G protein-coupled receptor signaling pathways reveals a role for receptor phosphorylation in airway contraction. Proc Natl Acad Sci U S A 2016; 113:4524-9. [PMID: 27071102 DOI: 10.1073/pnas.1521706113] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are known to initiate a plethora of signaling pathways in vitro. However, it is unclear which of these pathways are engaged to mediate physiological responses. Here, we examine the distinct roles of Gq/11-dependent signaling and receptor phosphorylation-dependent signaling in bronchial airway contraction and lung function regulated through the M3-muscarinic acetylcholine receptor (M3-mAChR). By using a genetically engineered mouse expressing a G protein-biased M3-mAChR mutant, we reveal the first evidence, to our knowledge, of a role for M3-mAChR phosphorylation in bronchial smooth muscle contraction in health and in a disease state with relevance to human asthma. Furthermore, this mouse model can be used to distinguish the physiological responses that are regulated by M3-mAChR phosphorylation (which include control of lung function) from those responses that are downstream of G protein signaling. In this way, we present an approach by which to predict the physiological/therapeutic outcome of M3-mAChR-biased ligands with important implications for drug discovery.
Collapse
|
84
|
Youssef G, Wallace WAH, Dagleish MP, Cousens C, Griffiths DJ. Ovine pulmonary adenocarcinoma: a large animal model for human lung cancer. ILAR J 2016; 56:99-115. [PMID: 25991702 DOI: 10.1093/ilar/ilv014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Recent progress in understanding the molecular pathogenesis of this disease has resulted in novel therapeutic strategies targeting specific groups of patients. Further studies are required to provide additional advances in diagnosis and treatment. Animal models are valuable tools for studying oncogenesis in lung cancer, particularly during the early stages of disease where tissues are rarely available from human cases. Mice have traditionally been used for studying lung cancer in vivo, and a variety of spontaneous and transgenic models are available. However, it is recognized that other species may also be informative for studies of cancer. Ovine pulmonary adenocarcinoma (OPA) is a naturally occurring lung cancer of sheep caused by retrovirus infection and has several features in common with adenocarcinoma of humans, including a similar histological appearance and activation of common cell signaling pathways. Additionally, the size and organization of human lungs are much closer to those of sheep lungs than to those of mice, which facilitates experimental approaches in sheep that are not available in mice. Thus OPA presents opportunities for studying lung tumor development that can complement conventional murine models. Here we describe the potential applications of OPA as a model for human lung adenocarcinoma with an emphasis on the various in vivo and in vitro experimental systems available.
Collapse
Affiliation(s)
- Gehad Youssef
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - William A H Wallace
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - Mark P Dagleish
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - Chris Cousens
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - David J Griffiths
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| |
Collapse
|
85
|
Pulmonary vein sleeve cell excitation-contraction-coupling becomes dysynchronized by spontaneous calcium transients. Biochem Soc Trans 2016; 43:410-6. [PMID: 26009184 DOI: 10.1042/bst20140299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Atrial fibrillation (AF) is the most common form of sustained cardiac arrhythmia. Substantial evidence indicates that cardiomyocytes located in the pulmonary veins [pulmonary vein sleeve cells (PVCs)] cause AF by generating ectopic electrical activity. Electrical ablation, isolating PVCs from their left atrial junctions, is a major treatment for AF. In small rodents, the sleeve of PVCs extends deep inside the lungs and is present in lung slices. Here we present data, using the lung slice preparation, characterizing how spontaneous Ca2+ transients in PVCs affect their capability to respond to electrical pacing. Immediately after a spontaneous Ca2+ transient the cell is in a refractory period and it cannot respond to electrical stimulation. Consequently, we observe that the higher the level of spontaneous activity in an individual PVC, the less likely it is that this PVC responds to electrical field stimulation. The spontaneous activity of neighbouring PVCs can be different from each other. Heterogeneity in the Ca2+ signalling of cells and in their responsiveness to electrical stimuli are known pro-arrhythmic events. The tendency of PVCs to show spontaneous Ca2+ transients and spontaneous action potentials (APs) underlies their potential to cause AF.
Collapse
|
86
|
Bäckström E, Lundqvist A, Boger E, Svanberg P, Ewing P, Hammarlund-Udenaes M, Fridén M. Development of a Novel Lung Slice Methodology for Profiling of Inhaled Compounds. J Pharm Sci 2016; 105:838-845. [DOI: 10.1002/jps.24575] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 01/11/2023]
|
87
|
Watson CY, Damiani F, Ram-Mohan S, Rodrigues S, de Moura Queiroz P, Donaghey TC, Rosenblum Lichtenstein JH, Brain JD, Krishnan R, Molina RM. Screening for Chemical Toxicity Using Cryopreserved Precision Cut Lung Slices. Toxicol Sci 2015; 150:225-33. [PMID: 26719368 DOI: 10.1093/toxsci/kfv320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
To assess chemical toxicity, current high throughput screening (HTS) assays rely primarily on in vitro measurements using cultured cells. Responses frequently differ from in vivo results due to the lack of physical and humoral interactions provided by the extracellular matrix, cell-cell interactions, and other molecular components of the native organ. To more accurately reproduce organ complexity in HTS, we developed an organotypic assay using the cryopreserved precision cut lung slice (PCLS) from rats and mice. Compared to the never-frozen PCLS, their frozen-thawed counterpart slices showed viability or metabolic activity that is decreased to an extent comparable to that observed in other cryopreserved cells and tissues, but shows no differences in further changes in cell viability, mitochondrial integrity, and glutathione activity in response to the model toxin zinc chloride (ZnCl2). Notably, these measurements were successfully miniaturized so as to establish HTS capacity in a 96-well plate format. Finally, PCLS responses correlated with common markers of lung injury measured in lavage fluid from rats intratracheally instilled with ZnCl2. In summary, we establish that the cryopreserved PCLS is a feasible approach for HTS investigations in predictive toxicology.
Collapse
Affiliation(s)
- Christa Y Watson
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Flavia Damiani
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Sumati Ram-Mohan
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
| | - Sylvia Rodrigues
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Priscila de Moura Queiroz
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Thomas C Donaghey
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Jamie H Rosenblum Lichtenstein
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Joseph D Brain
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
| | - Ramon M Molina
- *Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115 and
| |
Collapse
|
88
|
Siemsen DW, Dobrinen E, Han S, Chiocchi K, Meissner N, Swain SD. Vascular Dysfunction in Pneumocystis-Associated Pulmonary Hypertension Is Related to Endothelin Response and Adrenomedullin Concentration. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 186:259-69. [PMID: 26687815 DOI: 10.1016/j.ajpath.2015.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/29/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022]
Abstract
Pulmonary hypertension subsequent to an infectious disease can be due to vascular structural remodeling or to functional alterations within various vascular cell types. In our previous mouse model of Pneumocystis-associated pulmonary hypertension, we found that vascular remodeling was not responsible for observed increases in right ventricular pressures. Here, we report that the vascular dysfunction we observed could be explained by an enhanced response to endothelin-1 (20% greater reduction in lumen diameter, P ≤ 0.05), corresponding to an up-regulation of similar magnitude (P ≤ 0.05) of the endothelin A receptor in the lung tissue. This effect was potentially augmented by a decrease in production of the pulmonary vasodilator adrenomedullin of almost 70% (P ≤ 0.05). These changes did not occur in interferon-γ knockout mice similarly treated, which do not develop pulmonary hypertension under these circumstances. Surprisingly, we did not observe any relevant changes in the vascular endothelial nitric oxide synthase vasodilatory response, which is a common potential site of inflammatory alterations to pulmonary vascular function. Our results indicate the diverse mechanisms by which inflammatory responses to prior infections can cause functionally relevant changes in vascular responses in the lung, promoting the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Dan W Siemsen
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Erin Dobrinen
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Soo Han
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Kari Chiocchi
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Nicole Meissner
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Steve D Swain
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana.
| |
Collapse
|
89
|
Bidan CM, Veldsink AC, Meurs H, Gosens R. Airway and Extracellular Matrix Mechanics in COPD. Front Physiol 2015; 6:346. [PMID: 26696894 PMCID: PMC4667091 DOI: 10.3389/fphys.2015.00346] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common lung diseases worldwide, and is characterized by airflow obstruction that is not fully reversible with treatment. Even though airflow obstruction is caused by airway smooth muscle contraction, the extent of airway narrowing depends on a range of other structural and functional determinants that impact on active and passive tissue mechanics. Cells and extracellular matrix in the airway and parenchymal compartments respond both passively and actively to the mechanical stimulation induced by smooth muscle contraction. In this review, we summarize the factors that regulate airway narrowing and provide insight into the relative contributions of different constituents of the extracellular matrix and their biomechanical impact on airway obstruction. We then review the changes in extracellular matrix composition in the airway and parenchymal compartments at different stages of COPD, and finally discuss how these changes impact airway narrowing and the development of airway hyperresponsiveness. Finally, we position these data in the context of therapeutic research focused on defective tissue repair. As a conclusion, we propose that future works should primarily target mild or early COPD, prior to the widespread structural changes in the alveolar compartment that are more characteristic of severe COPD.
Collapse
Affiliation(s)
- Cécile M Bidan
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands ; Laboratoire Interdisciplinaire de Physique (LIPhy), Université Grenoble Alpes Grenoble, France ; Centre National de la Recherche Scientifique, LIPhy Grenoble, France
| | - Annemiek C Veldsink
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen Groningen, Netherlands ; Groningen Research Institute for Asthma and COPD, University of Groningen Netherlands
| |
Collapse
|
90
|
Abstract
The 2nd Cross Company Respiratory Symposium (CCRS), held in Horsham, U.K. in 2012, brought together representatives from across the pharmaceutical industry with expert academics, in the common interest of improving the design and translational predictiveness of in vivo models of respiratory disease. Organized by the respiratory representatives of the European Federation of Pharmaceutical Industries and Federations (EFPIA) group of companies involved in the EU-funded project (U-BIOPRED), the aim of the symposium was to identify state-of-the-art improvements in the utility and design of models of respiratory disease, with a view to improving their translational potential and reducing wasteful animal usage. The respiratory research and development community is responding to the challenge of improving translation in several ways: greater collaboration and open sharing of data, careful selection of the species, complexity and chronicity of the models, improved practices in preclinical research, continued refinement in models of respiratory diseases and their sub-types, greater understanding of the biology underlying human respiratory diseases and their sub-types, and finally greater use of human (and especially disease-relevant) cells, tissues and explants. The present review highlights these initiatives, combining lessons from the symposium and papers published in Clinical Science arising from the symposium, with critiques of the models currently used in the settings of asthma, idiopathic pulmonary fibrosis and COPD. The ultimate hope is that this will contribute to a more rational, efficient and sustainable development of a range of new treatments for respiratory diseases that continue to cause substantial morbidity and mortality across the world.
Collapse
|
91
|
Romberger DJ, Heires AJ, Nordgren TM, Souder CP, West W, Liu XD, Poole JA, Toews ML, Wyatt TA. Proteases in agricultural dust induce lung inflammation through PAR-1 and PAR-2 activation. Am J Physiol Lung Cell Mol Physiol 2015; 309:L388-99. [PMID: 26092994 DOI: 10.1152/ajplung.00025.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/09/2015] [Indexed: 01/31/2023] Open
Abstract
Workers exposed to aerosolized dust present in concentrated animal feeding operations (CAFOs) are susceptible to inflammatory lung diseases, such as chronic obstructive pulmonary disease. Extracts of dust collected from hog CAFOs [hog dust extract (HDE)] are potent stimulators of lung inflammatory responses in several model systems. The observation that HDE contains active proteases prompted the present study, which evaluated the role of CAFO dust proteases in lung inflammatory processes and tested whether protease-activated receptors (PARs) are involved in the signaling pathway for these events. We hypothesized that the damaging proinflammatory effect of HDE is due, in part, to the proteolytic activation of PARs, and inhibiting the proteases in HDE or disrupting PAR activation would attenuate HDE-mediated inflammatory indexes in bronchial epithelial cells (BECs), in mouse lung slices in vitro, and in a murine in vivo exposure model. Human BECs and mouse lung slice cultures stimulated with 5% HDE released significantly more of each of the cytokines measured (IL-6, IL-8, TNF-α, keratinocyte-derived chemokine/CXC chemokine ligand 1, and macrophage inflammatory protein-2/CXC chemokine ligand 2) than controls, and these effects were markedly diminished by protease inhibition. Inhibition of PARs also blunted the HDE-induced cytokine release from BECs. In addition, protease depletion inhibited HDE-induced BEC intracellular PKCα and PKCε activation. C57BL/6J mice administered 12.5% HDE intranasally, either once or daily for 3 wk, exhibited increased total cellular and neutrophil influx, bronchial alveolar fluid inflammatory cytokines, lung histopathology, and inflammatory scores compared with mice receiving protease-depleted HDE. These data suggest that proteases in dust from CAFOs are important mediators of lung inflammation, and these proteases and their receptors may provide novel targets for therapeutic intervention in CAFO dust-induced airways disease.
Collapse
Affiliation(s)
- Debra J Romberger
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska; Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, Nebraska;
| | - Art J Heires
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Tara M Nordgren
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Chelsea P Souder
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - William West
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Xiang-de Liu
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jill A Poole
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Todd A Wyatt
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska; Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, Nebraska; Department of Environmental, Agricultural, and Occupational Health, University of Nebraska Medical Center, Omaha, Nebraska; and
| |
Collapse
|
92
|
Gilmour MI, Kim YH, Hays MD. Comparative chemistry and toxicity of diesel and biomass combustion emissions. Anal Bioanal Chem 2015; 407:5869-75. [DOI: 10.1007/s00216-015-8797-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 01/12/2023]
|
93
|
Liu R, An L, Liu G, Li X, Tang W, Chen X. Mouse lung slices: An ex vivo model for the evaluation of antiviral and anti-inflammatory agents against influenza viruses. Antiviral Res 2015; 120:101-11. [PMID: 26022197 PMCID: PMC7125926 DOI: 10.1016/j.antiviral.2015.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 11/20/2022]
Abstract
Mouse lung slices stay alive for at least 5 days ex vivo. Influenza viruses can replicate in mouse lung slices and trigger robust cytokine and chemokine responses. A positive correlation in cytokine and chemokine responses between ex vivo and in vivo exists. Neuraminidase and IP-10 can serve as readouts for antiviral and anti-inflammation activities, respectively. This ex vivo model may predict efficacy of drug candidates in antiviral and anti-inflammation activities in vivo.
The influenza A virus is notoriously known for its ability to cause recurrent epidemics and global pandemics. Antiviral therapy is effective when treatment is initiated within 48 h of symptom onset, and delaying treatment beyond this time frame is associated with decreased efficacy. Research on anti-inflammatory therapy to ameliorate influenza-induced inflammation is currently underway and seems important to the impact on the clinical outcome. Both antiviral and anti-inflammatory drugs with novel mechanisms of action are urgently needed. Current methods for evaluating the efficacy of anti-influenza drugs rely mostly on transformed cells and animals. Transformed cell models are distantly related to physiological and pathological conditions. Although animals are the best choices for preclinical drug testing, they are not time- or cost-efficient. In this study, we established an ex vivo model using mouse lung slices to evaluate both antiviral and anti-inflammatory agents against influenza virus infection. Both influenza virus PR8 (H1N1) and A/Human/Hubei/3/2005 (H3N2) can replicate efficiently in mouse lung slices and trigger significant cytokine and chemokine responses. The induction of selected cytokines and chemokines were found to have a positive correlation between ex vivo and in vivo experiments, suggesting that the ex vivo cultured lung slices may closely resemble the lung functionally in an in vivo configuration when challenged by influenza virus. Furthermore, a set of agents with known antiviral and/or anti-inflammatory activities were tested to validate the ex vivo model. Our results suggested that mouse lung slices provide a robust, convenient and cost-efficient model for the assessment of both antiviral and anti-inflammatory agents against influenza virus infection in one assay. This ex vivo model may predict the efficacy of drug candidates’ antiviral and anti-inflammatory activities in vivo.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liwei An
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Anatomy, The University of Hong Kong, Hong Kong, China(1)
| | - Ge Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Tang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China
| | - Xulin Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 43001, Hubei, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
94
|
Yarova PL, Stewart AL, Sathish V, Britt RD, Thompson MA, P Lowe AP, Freeman M, Aravamudan B, Kita H, Brennan SC, Schepelmann M, Davies T, Yung S, Cholisoh Z, Kidd EJ, Ford WR, Broadley KJ, Rietdorf K, Chang W, Bin Khayat ME, Ward DT, Corrigan CJ, T Ward JP, Kemp PJ, Pabelick CM, Prakash YS, Riccardi D. Calcium-sensing receptor antagonists abrogate airway hyperresponsiveness and inflammation in allergic asthma. Sci Transl Med 2015; 7:284ra60. [PMID: 25904744 DOI: 10.1126/scitranslmed.aaa0282] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/24/2015] [Indexed: 12/27/2022]
Abstract
Airway hyperresponsiveness and inflammation are fundamental hallmarks of allergic asthma that are accompanied by increases in certain polycations, such as eosinophil cationic protein. Levels of these cations in body fluids correlate with asthma severity. We show that polycations and elevated extracellular calcium activate the human recombinant and native calcium-sensing receptor (CaSR), leading to intracellular calcium mobilization, cyclic adenosine monophosphate breakdown, and p38 mitogen-activated protein kinase phosphorylation in airway smooth muscle (ASM) cells. These effects can be prevented by CaSR antagonists, termed calcilytics. Moreover, asthmatic patients and allergen-sensitized mice expressed more CaSR in ASMs than did their healthy counterparts. Indeed, polycations induced hyperreactivity in mouse bronchi, and this effect was prevented by calcilytics and absent in mice with CaSR ablation from ASM. Calcilytics also reduced airway hyperresponsiveness and inflammation in allergen-sensitized mice in vivo. These data show that a functional CaSR is up-regulated in asthmatic ASM and targeted by locally produced polycations to induce hyperresponsiveness and inflammation. Thus, calcilytics may represent effective asthma therapeutics.
Collapse
Affiliation(s)
- Polina L Yarova
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Alecia L Stewart
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Rodney D Britt
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Alexander P P Lowe
- Division of Pharmacology, Cardiff University, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK
| | - Michelle Freeman
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Hirohito Kita
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Sarah C Brennan
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | | | - Thomas Davies
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Sun Yung
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Zakky Cholisoh
- Division of Pharmacology, Cardiff University, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK
| | - Emma J Kidd
- Division of Pharmacology, Cardiff University, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK
| | - William R Ford
- Division of Pharmacology, Cardiff University, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK
| | - Kenneth J Broadley
- Division of Pharmacology, Cardiff University, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3XF, UK
| | - Katja Rietdorf
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Wenhan Chang
- Department of Medicine, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Mohd E Bin Khayat
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Donald T Ward
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | | | - Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, King's College London, London SE1 9RT, UK
| | - Paul J Kemp
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | | | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Daniela Riccardi
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.
| |
Collapse
|
95
|
Anaparti V, Ilarraza R, Orihara K, Stelmack GL, Ojo OO, Mahood TH, Unruh H, Halayko AJ, Moqbel R. NMDA receptors mediate contractile responses in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1253-64. [PMID: 25888577 DOI: 10.1152/ajplung.00402.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/15/2015] [Indexed: 01/12/2023] Open
Abstract
Human airway smooth muscle (HASM) exhibits enhanced contractility in asthma. Inflammation is associated with airway hypercontractility, but factors that underpin these features are not fully elucidated. Glutamate toxicity associated with increased plasma glutamate concentrations was observed in airway inflammation, suggesting that multisubunit glutamate receptors, N-methyl-d-aspartate receptors (NMDA-R) contribute to airway hyperreactivity. We tested the hypothesis that HASM expresses NMDA-R subunits that can form functional receptors to mediate contractile responses to specific extracellular ligands. In cultured HASM cells, we measured NMDA-R subunit mRNA and protein abundance by quantitative PCR, immunoblotting, flow cytometry, and epifluorescence immunocytochemistry. We measured mRNA for a number of NMDA-R subunits, including the obligatory NR1 subunit, which we confirmed to be present as a protein. In vitro and ex vivo functional NMDA-R activation in HASM cells was measured using intracellular calcium flux (fura-2 AM), collagen gel contraction assays, and murine thin-cut lung slices (TCLS). NMDA, a pharmacological glutamate analog, induced cytosolic calcium mobilization in cultured HASM cells. We detected three different temporal patterns of calcium response, suggesting the presence of heterogeneous myocyte subpopulations. NMDA-R activation also induced airway contraction in murine TCLS and soft collagen gels seeded with HASM cells. Responses in cells, lung slices, and collagen gels were mediated by NMDA-R, as they could be blocked by (2R)-amino-5-phosphonopentanoate, a specific NMDA-R inhibitor. In summary, we reveal the presence of NMDA-R in HASM that mediate contractile responses via glutamatergic mechanisms. These findings suggest that accumulation of glutamate-like ligands for NMDA-R associated with airway inflammation contributes directly to airway hyperreactivity.
Collapse
Affiliation(s)
- Vidyanand Anaparti
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Ramses Ilarraza
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kanami Orihara
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gerald L Stelmack
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Oluwaseun O Ojo
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Thomas H Mahood
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Helmut Unruh
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Thoracic Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada;
| | - Redwan Moqbel
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
96
|
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
|
97
|
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
|
98
|
Yoo HY, Park SJ, Kim HJ, Kim WK, Kim SJ. Integrative understanding of hypoxic pulmonary vasoconstriction using in vitro models: from ventilated/perfused lung to single arterial myocyte. Integr Med Res 2014; 3:180-188. [PMID: 28664095 PMCID: PMC5481745 DOI: 10.1016/j.imr.2014.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 10/25/2022] Open
Abstract
Contractile response of a pulmonary artery (PA) to hypoxia (hypoxic pulmonary vasoconstriction; HPV) is a unique physiological reaction. HPV is beneficial for the optimal distribution of blood flow to differentially ventilated alveolar regions in the lung, thereby preventing systemic hypoxemia. Numerous in vitro studies have been conducted to elucidate the mechanisms underlying HPV. These studies indicate that PA smooth muscle cells (PASMCs) sense lowers the oxygen partial pressure (PO2) and contract under hypoxia. As for the PO2-sensing molecules, a variety of ion channels in PASMCs had been suggested. Nonetheless, the modulator(s) of the ion channels alone cannot mimic HPV in the experiments using PA segments and/or isolated organs. We compared the hypoxic responses of PASMCs, PAs, lung slices, and total lungs using a variety of methods (e.g., patch-clamp technique, isometric contraction measurement, video analysis of precision-cut lung slices, and PA pressure measurement in ventilated/perfused lungs). In this review, the relevant results are compared to provide a comprehensive understanding of HPV. Integration of the influences from surrounding tissues including blood cells as well as the hypoxic regulation of ion channels in PASMCs are indispensable for insights into HPV and other related clinical conditions.
Collapse
Affiliation(s)
- Hae Young Yoo
- Red Cross College of Nursing, Chung-Ang University, Seoul, Korea
| | - Su Jung Park
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Hae Jin Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Woo Kyung Kim
- Department of Internal Medicine and Channelopathy Research Institute (CRC), College of Medicine, Dongguk University, Goyang, Korea
| | - Sung Joon Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.,Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
99
|
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
|
100
|
Vijayaraj P, Sivaprakasam C, Varthini LV, Sarkar M, Nachiappan V. In vitro exposure of tobacco specific nitrosamines decreases the rat lung phospholipids by enhanced phospholipase A2 activity. Toxicol In Vitro 2014; 28:1097-105. [PMID: 24835565 DOI: 10.1016/j.tiv.2014.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 04/10/2014] [Accepted: 05/05/2014] [Indexed: 01/23/2023]
Abstract
Tobacco-specific nitrosamines (TSNA) have implications in the pathogenesis of various lung diseases and conditions are prevalent even in non-smokers. N-nitrosonornicotine (NNN) and 4-(methyl nitrosamino)-1-(3-pyridyl)-1-butanone (NNK) are potent pulmonary carcinogens present in tobacco product and are mainly responsible for lung cancer. TSNA reacts with pulmonary surfactants, and alters the surfactant phospholipid. The present study was undertaken to investigate the in vitro exposure of rat lung tissue slices to NNK or NNN and to monitor the phospholipid alteration by [(32)P]orthophosphate labeling. Phospholipid content decreased significantly in the presence of either NNK or NNN with concentration and time dependent manner. Phosphatidylcholine (PC) is the main phospholipid of lung and significant reduction was observed in PC ∼61%, followed by phosphatidylglycerol (PG) with 100μM of NNK, whereas NNN treated tissues showed a reduction in phosphatidylserine (PS) ∼60% and PC at 250μM concentration. The phospholipase A2 assays and expression studies reveal that both compounds enhanced phospholipid hydrolysis, thereby reducing the phospholipid content. Collectively, our data demonstrated that both NNK and NNN significantly influenced the surfactant phospholipid level by enhanced phospholipase A2 activity.
Collapse
Affiliation(s)
- Panneerselvam Vijayaraj
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Chinnarasu Sivaprakasam
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | | | - Mary Sarkar
- Biochemistry Department, Indian Institute of Science, Bangalore 560012, India
| | - Vasanthi Nachiappan
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.
| |
Collapse
|