1
|
Vierhout M, Ayoub A, Ali P, Kumaran V, Naiel S, Isshiki T, Koenig JFE, Kolb MRJ, Ask K. A novel ex vivo approach for investigating profibrotic macrophage polarization using murine precision-cut lung slices. Biochem Biophys Res Commun 2024; 741:151038. [PMID: 39603027 DOI: 10.1016/j.bbrc.2024.151038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is fatal interstitial lung disease characterized by excessive scarring of the lung tissue and declining respiratory function. Given its short prognosis and limited treatment options, novel strategies to investigate emerging experimental treatments are urgently needed. Macrophages, as the most abundant immune cell in the lung, have key implications in wound healing and lung fibrosis. However, they are highly plastic and adaptive to their surrounding microenvironment, and thus to maximize translation of research to lung disease, there is a need to study macrophages in multifaceted, complex systems that are representative of the lung. Precision-cut lung slices (PCLS) are living tissue preparations derived from the lung that are cultured ex vivo, which bypass the need for artificial recapitulation of the lung milieu and architecture. Macrophage programming studies are traditionally conducted using isolated cells in vitro, thus our objective was to establish and validate a moderate-throughput, biologically-translational, viable model to study profibrotic polarization of pulmonary-resident macrophages using murine PCLS. To achieve this, we used a polarization cocktail (PC), consisting of IL-4, IL-13, and IL-6, over a 72-h time course. We first demonstrated no adverse effects of the PC on PCLS viability and architecture. Next, we showed that multiple markers of macrophage profibrotic polarization, including Arginase-1, CD206, YM1, and CCL17 were induced in PCLS following PC treatment. Through tissue microarray-based histological assessments, we directly visualized and quantified Arginase-1 and CD206 staining in PCLS in a moderate-throughput manner. We further delineated phenotype of polarized macrophages, and using high-plex immunolabelling with the Iterative Bleaching Extends Multiplexity (IBEX) method, showed that the PC effects both interstitial and alveolar macrophages. Substantiating the profibrotic properties of the system, we also showed expression of extracellular matrix components and fibrotic markers in stimulated PCLS. Finally, we demonstrated that clodronate treatment diminishes the PC effects on profibrotic macrophage readouts. Overall, our findings support a suitable complex model for studying ex vivo profibrotic macrophage programming in the lung, with future capacity for investigating experimental therapeutic candidates and disease mechanisms in pulmonary fibrosis.
Collapse
Affiliation(s)
- Megan Vierhout
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Anmar Ayoub
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Pareesa Ali
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Vaishnavi Kumaran
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Safaa Naiel
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Takuma Isshiki
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada; Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omorinishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Joshua F E Koenig
- Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omorinishi, Ota-ku, Tokyo, 143-8540, Japan; Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Martin R J Kolb
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada.
| |
Collapse
|
2
|
Budeus B, Kroepel C, Stasch LM, Klein D. Matrix-free human lung organoids derived from induced pluripotent stem cells to model lung injury. Stem Cell Res Ther 2024; 15:468. [PMID: 39696649 DOI: 10.1186/s13287-024-04106-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Organoids, as near-physiological 3D culture systems, offer new opportunities to study the pathogenesis of various organs in mimicking the cellular complexity and functionality of human organs. METHOD Here we used a quite simple and very practicable method to successfully generate induced pluripotent stem cell (iPSC)-derived human lung organoids (LuOrg) in a matrix-free manner as an alternative to the widely used preclinical mouse models in order to investigate normal lung damage in detail and as close as possible to the patient. We performed detailed morphological and molecular analyses, including bulk and single cell RNA sequencing, of generated lung organoids and evaluated the quality and robustness of our model as a potential in vitro platform for lung diseases, namely radiation-induced lung injury. RESULTS A matrix-free method for differentiation of iPSCs can be used to obtain lung organoids that morphologically reflect the target tissue of the human lung very well, especially with regard to the cellular composition. The different cellular fates were investigated following the genotoxic stress induced by radiation and revealed further insights in the radiation-sensitivity of the different lung cells. Finally, we provide cellular gene sets found to be induced in the different lung organoid cellular subsets after irradiation, which could be used as additional RT response and particularly senescence gene sets in future studies. CONCLUSION By establishing these free-floating LuOrgs for the investigation of cancer therapeutic approaches as a new and patient-oriented in vitro platform particularly in experimental radiooncology, not only a reduction in the number of experimental animals, but also an adequately and meaningfully replacement of corresponding animal experiments can be achieved.
Collapse
Affiliation(s)
- Bettina Budeus
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chiara Kroepel
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lisa Marie Stasch
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
3
|
Bammert MT, Ansari M, Haag L, Ahmad Z, Schröder V, Birch J, Santacruz D, Rust W, Viollet C, Strobel B, Dick A, Gantner F, Schlüter H, Ramirez F, Lizé M, Thomas MJ, Le HQ. JUNB O-GlcNAcylation-Mediated Promoter Accessibility of Metabolic Genes Modulates Distinct Epithelial Lineage in Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406751. [PMID: 39676507 DOI: 10.1002/advs.202406751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/15/2024] [Indexed: 12/17/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal disease with substantial unmet medical needs. While aberrant epithelial remodeling is a key factor in IPF progression, the molecular mechanisms behind this process remain elusive. Harnessing a 3D patient-derived organoid model and multi-omics approach, the first inventory of the connection between metabolic alteration, chromatin accessibility, and transcriptional regulation in IPF aberrant epithelial remodeling is provided. This remodeling is characterized by an increase in chromatin accessibility, particularly at JUNB motif-enriched promoter regions proximal to transcription start sites of metabolic and pro-fibrotic genes. Mechanistically, JUNB undergoes O-linked β-N-acetylglucosamine modification (O-GlcNAcylation), a critical step in modulating pro-fibrotic responses to chronic injury. This modification is pivotal in fostering the emergence of aberrant epithelial basal cells in the alveolar niche, a proposed driver of IPF pathology. Specific deletion of O-GlcNAcylation sites on JUNB attenuates the metaplastic differentiation of basal cells, thereby aiding in the restoration of the alveolar lineage. Together, the findings reveal a novel link between metabolic dysregulation and cell fate regulation at the chromatin level in fibrosis, mediated by the O-GlcNAc-JUNB axis, suggesting avenues for the development of new therapeutic strategies in IPF.
Collapse
Affiliation(s)
- Marie-Therese Bammert
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
- Faculty of Biology, University of Konstanz, 78457, Konstanz, Germany
| | - Meshal Ansari
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Leoni Haag
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Zuhdi Ahmad
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Victoria Schröder
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Joseph Birch
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Diana Santacruz
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Werner Rust
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Coralie Viollet
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Benjamin Strobel
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Alec Dick
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Florian Gantner
- Faculty of Biology, University of Konstanz, 78457, Konstanz, Germany
- C.H. Boehringer Sohn AG and Co. KG, 55218, Ingelheim, Germany
| | - Holger Schlüter
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Fidel Ramirez
- Global Computational Biology and Digital Science, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Muriel Lizé
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Matthew J Thomas
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
- University of Bath, Bath, BA27JX, UK
| | - Huy Q Le
- Lung Repair & Regeneration Department, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| |
Collapse
|
4
|
Kollareth DJM, Sharma AK. Precision cut lung slices: an innovative tool for lung transplant research. Front Immunol 2024; 15:1504421. [PMID: 39669559 PMCID: PMC11634892 DOI: 10.3389/fimmu.2024.1504421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
Lung ischemia-reperfusion injury (IRI), a common complication after lung transplantation (LTx), plays a crucial role in both primary graft dysfunction (PGD) and chronic lung allograft dysfunction (CLAD) thereby adversely impacting the clinical outcomes in these patient cohorts. Lung IRI is characterized by several molecular events including immune cell infiltration, reactive oxygen species (ROS) generation, calcium overload, inflammation and various forms of cell death pathways. Currently, no therapeutic agents are available to clinically prevent lung IRI. While animal and cell culture models are highly valuable in understanding the pathophysiology of lung IRI, they may not completely recapitulate the complexity of human lung tissue pathology. This limitation necessitates the requirement for developing innovative preclinical human research tools that can supplement available scientific modalities. Emerging evidence suggests that precision-cut lung slices (PCLS) have become an indispensable tool in scientific research to study lung biology in an ex vivo tissue system. Recent studies using PCLS have investigated lung diseases including asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. Although PCLS can be successfully employed to determine the deleterious events in the pathogenesis of lung IRI, including cell-cell interactions as well as hallmarks of inflammation and oxidative stress-dependent pathways, detailed studies employing PCLS to decipher these molecular events in post-LTx injury are currently limited. This review focuses on the applicability and unexplored potential of PCLS as a powerful tool in lung IRI research for understanding the pathophysiology and consequent development of new therapeutic modalities.
Collapse
Affiliation(s)
| | - Ashish K. Sharma
- Department of Surgery, University of Florida, Gainesville, FL, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Gonzales-Huerta LE, Williams TJ, Aljohani R, Robertson BD, Evans CA, Armstrong-James D. Precision-cut lung slices in air-liquid interface (PCLS-ALI): A novel ex-vivo model for the study of Pulmonary Aspergillosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.615211. [PMID: 39605594 PMCID: PMC11601337 DOI: 10.1101/2024.11.15.615211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pulmonary Aspergillosis is a respiratory infection with a high mortality rate, which affects patients with immunosuppression or structural lung defects. Antifungal treatment options are few and many have narrow therapeutic margins and potentially serious side effects. In recent years, there are growing numbers of reports of antifungal resistance. Thus, there is an urgent need for effective models to study fungal pathogenesis and test antifungal therapies in the respiratory system. Here, we present a novel ex-vivo model using precision-cut lung slices in an air-liquid interface platform to evaluate lung tissue responses to fungal infection and antifungal treatment. Readouts assessed were lactate dehydrogenase for tissue damage, release of inflammatory cytokines (TNF-α, IL-1β, CXCL1), and histology for confirmation of hyphal invasion. Overall, the PCLS-ALI model is a promising approach for understanding lung tissue responses to fungal infections, which fulfils the reduction and refinement components of the 3Rs guiding principles for ethical use of experimental animals.
Collapse
Affiliation(s)
- L E Gonzales-Huerta
- Department of Infectious Disease, Imperial College London, SW7 2AZ, UK
- Carrera de Medicina Humana, Facultad de Ciencias de la Salud, Universidad San Ignacio de Loyola, Lima 15024 Peru
| | - T J Williams
- Department of Infectious Disease, Imperial College London, SW7 2AZ, UK
| | - R Aljohani
- Department of Infectious Disease, Imperial College London, SW7 2AZ, UK
| | - B D Robertson
- Department of Infectious Disease, Imperial College London, SW7 2AZ, UK
| | - C A Evans
- Department of Infectious Disease, Imperial College London, SW7 2AZ, UK
- Innovación Por la Salud Y Desarrollo (IPSYD), Asociación Benéfica PRISMA, Lima, 15073, Peru4 IFHAD: Innovation for Health and Development, Laboratorio de Investigación y Desarrollo, Universidad Peruana Cayetano Heredia, Lima 150135 Peru
- IFHAD: Innovation For Health And Development, Laboratorio de Investigación y Desarrollo, Universidad Peruana Cayetano Heredia, Lima 150135 Peru
- IFHAD: Innovation For Health And Development, Department of infectious disease, Imperial College London, London, UK
| | | |
Collapse
|
6
|
De Luca S, Gunatilaka A, Coward-Smith M, Gomez HM, Kim RY, Stenekes A, Chan SMH, Wang W, Tan D, Vlahos R, Stewart AG, Donovan C. Understanding Comorbidities of Respiratory Models as Novel Platforms for Drug Discovery. ACS Pharmacol Transl Sci 2024; 7:3385-3393. [PMID: 39539266 PMCID: PMC11555503 DOI: 10.1021/acsptsci.4c00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Chronic respiratory diseases affect over 450 million people worldwide and result in 4 million deaths per year. The majority of lung diseases are treated with drugs delivered directly to the lungs. However, there is bidirectional crosstalk between the lung and other organs/tissues in health and disease. This crosstalk supports targeting of extrapulmonary sites in addition to the lung to improve the comorbidities associated with lung disease. However, new preclinical in vivo and in vitro assays that model the human pathophysiology are required. In this review, we showcase the latest knowledge of the bidirectional relationship between the respiratory system and organs affected by comorbidities such as obesity and atherosclerosis. We also discuss the impact of new cell culture systems, including complex 3D culture models that may be used as platforms to generate disease insights and for drug discovery. This review highlights work presented by Respiratory and Inflammation Special Interest Group researchers as part of the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists (ASCEPT) annual scientific meeting in 2023.
Collapse
Affiliation(s)
- Simone
N. De Luca
- Respiratory
Research Group, Centre for Respiratory Science and Health, School
of Health and Biomedical Sciences, RMIT
University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Avanka Gunatilaka
- Department
of Biochemistry and Pharmacology, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Madison Coward-Smith
- Respiratory
Research Group, Centre for Respiratory Science and Health, School
of Health and Biomedical Sciences, RMIT
University, Bundoora, Melbourne, Victoria 3083, Australia
- School
of Life Sciences, University of Technology
Sydney, Sydney, New South Wales 2007, Australia
| | - Henry M. Gomez
- School
of Biomedical Sciences and Pharmacy, University of Newcastle and Immune
Health Program, Hunter Medical Research
Institute, Newcastle, New South Wales 2308, Australia
| | - Richard Y. Kim
- School
of Life Sciences, University of Technology
Sydney, Sydney, New South Wales 2007, Australia
- School
of Biomedical Sciences and Pharmacy, University of Newcastle and Immune
Health Program, Hunter Medical Research
Institute, Newcastle, New South Wales 2308, Australia
- Woolcock
Institute of Medical Research, Macquarie Park, New South Wales 2113, Australia
| | - Aimee Stenekes
- School
of Life Sciences, University of Technology
Sydney, Sydney, New South Wales 2007, Australia
| | - Stanley M. H. Chan
- Respiratory
Research Group, Centre for Respiratory Science and Health, School
of Health and Biomedical Sciences, RMIT
University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Wei Wang
- Respiratory
Research Group, Centre for Respiratory Science and Health, School
of Health and Biomedical Sciences, RMIT
University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Daniel Tan
- Department
of Biochemistry and Pharmacology, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ross Vlahos
- Respiratory
Research Group, Centre for Respiratory Science and Health, School
of Health and Biomedical Sciences, RMIT
University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Alastair G. Stewart
- Department
of Biochemistry and Pharmacology, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Chantal Donovan
- School
of Life Sciences, University of Technology
Sydney, Sydney, New South Wales 2007, Australia
- School
of Biomedical Sciences and Pharmacy, University of Newcastle and Immune
Health Program, Hunter Medical Research
Institute, Newcastle, New South Wales 2308, Australia
- Woolcock
Institute of Medical Research, Macquarie Park, New South Wales 2113, Australia
| |
Collapse
|
7
|
Nowakowska J, Gvazava N, Langwiński W, Ziarniak K, da Silva IAN, Stegmayr J, Wagner DE, Szczepankiewicz A. Optimizing miRNA transfection for screening in precision cut lung slices. Am J Physiol Lung Cell Mol Physiol 2024; 327:L712-L723. [PMID: 39254091 PMCID: PMC11563635 DOI: 10.1152/ajplung.00138.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/31/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Precision cut lung slices (PCLS) are complex three-dimensional (3-D) lung tissue models, which preserve the native microenvironment, including cell diversity and cell-matrix interactions. They are an innovative ex vivo platform that allows studying disease as well as the effects of therapeutic agents or regulatory molecules [e.g., microRNA (miRNA)]. The aim of our study was to develop a protocol to transfect PCLS with miRNA using lipid nanoparticles (LNPs) to enable higher throughput screening of miRNA, obviating the need for custom stabilization and internalization approaches. PCLS of 4 mm diameter were generated using agarose-filled rodent lungs and a vibratome. TYE665-labeled scrambled miRNA was used to evaluate transfection efficacy of six different commercially available LNPs. Transfection efficacy was visualized using live high-content fluorescence microscopy, followed by higher-resolution confocal fluorescence microscopy in fixed PCLS. Metabolic activity and cellular damage were assessed using water-soluble tetrazolium salt (WST-1) and lactate dehydrogenase (LDH) release. Using a live staining kit containing a cell membrane impermeant nuclear dye, RedDot2, we established that cellular membranes in PCLS are permeable in the initial 24 h of slicing but diminished thereafter. Therefore, all transfection experiments occurred at least 24 h after slicing. All six commercially available LNPs enabled transfection without inducing significant cytotoxicity or impaired metabolic function. However, RNAiMAX and INTERFERin led to increases in transfection efficacy as compared with other LNPs, with detection possible as low as 25 nM. Therefore, LNP-based transfection of miRNA is possible and can be visualized in live or fixed PCLS, enabling future higher throughput studies using diverse miRNAs.NEW & NOTEWORTHY RNA-based therapeutics hold significant promise for disease treatment; however, limited research exists on miRNA transfection specifically within PCLS. miRNA transfection has thus far required custom functionalization for stabilization and internalization. We aimed to optimize a transfection protocol for rapid screening approaches of miRNA sequences. We show that transfecting miRNA in PCLS is possible using lipid nanoparticles. In addition, we show that 25 nM of TYE665-miRNA is sufficient for detection in a high-content imaging system.
Collapse
Affiliation(s)
- Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | - Nika Gvazava
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kamil Ziarniak
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Iran Augusto N da Silva
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - John Stegmayr
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - Darcy E Wagner
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
- Meakins-Christie Laboratories, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
8
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Nelson TM, Mariano CA, Ramirez GO, Badrou A, Quiros KAM, Shankel M, Eskandari M. Lung Mechanics: Material Characterization of Pulmonary Constituents for an Experimentally Informed Computational Pipeline. Curr Protoc 2024; 4:e70001. [PMID: 39240156 DOI: 10.1002/cpz1.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
The lung comprises multiple components including the parenchyma, airways, and visceral pleura, where each constituent displays specific material properties that together govern the whole organ's properties. The structural and mechanical complexity of the lung has historically undermined its comprehensive characterization, especially compared to other biological organs, such as the heart or bones. This knowledge void is particularly remarkable when considering that pulmonary disease is one of the leading causes of morbidity and mortality across the globe. Establishing the mechanical properties of the lung is central to formulating a baseline understanding of its operation, which can facilitate investigations of diseased states and how the lung will potentially respond to clinical interventions. Here, we present established and widely accepted experimental protocols for pulmonary material quantification, specifying how to extract, prepare, and test each type of lung constituent under planar biaxial tensile loading to investigate the mechanical properties, such as physiological stress-strain profiles, anisotropy, and viscoelasticity. These methods are presented across an array of commonly studied species (murine, rat, and porcine). Additionally, we highlight how such material properties may inform the construction of an inverse finite element model, which is central to implementing predictive computational tools for accurate disease diagnostics and optimized medical treatments. These presented methodologies are aimed at supporting research advancements in the field of pulmonary biomechanics and to help inaugurate future novel studies. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: General procedures in lung biaxial testing Alternate Protocol 1: Parenchymal-specific preparation and loading procedures Alternate Protocol 2: Airway-specific preparation and loading procedures Alternate Protocol 3: Visceral pleura-specific preparation and loading procedures Basic Protocol 2: Computational analysis.
Collapse
Affiliation(s)
- Talyah M Nelson
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Crystal A Mariano
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Gustavo O Ramirez
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Arif Badrou
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Kathrine A M Quiros
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Matthew Shankel
- Department of Mechanical Engineering, University of California, Riverside, California
| | - Mona Eskandari
- Department of Mechanical Engineering, University of California, Riverside, California
- BREATHE Center, School of Medicine University of California, Riverside, California
- Department of Bioengineering, University of California, Riverside, California
| |
Collapse
|
10
|
Marimoutou M, Patel V, Kim JH, Schaible N, Alvarez J, Hughes J, Obermok M, Rodríguez CI, Kallarakal T, Suki B, Amin K, Krishnan R, Behrsing HP. The Fibrotic Phenotype of Human Precision-Cut Lung Slices Is Maintained after Cryopreservation. TOXICS 2024; 12:637. [PMID: 39330565 PMCID: PMC11436228 DOI: 10.3390/toxics12090637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Human precision-cut lung slices (hPCLS) prepared from fibrotic lungs recapitulate the pathophysiological hallmarks of fibrosis. These hallmark features can also be induced by treating non-fibrotic hPCLS with a fibrotic cocktail (FC). As a result, the fibrotic and fibrosis-induced hPCLS are rapidly emerging as preferred models for disease modeling and drug discovery. However, current hPCLS models are limited by tissue viability in culture, as they are usually only viable for one week after harvesting. Here, we demonstrate that the fibrotic hPCLS can be cryopreserved, stored for months, and then thawed on demand without loss of hPCLS viability or protein content for 14 days post-thawing. Cryopreservation also preserves the pro-fibrotic potential of non-fibrotic hPCLS. Specifically, when we treated the thawed non-fibrotic hPCLS with an FC, we observed significant pro-fibrotic cytokine secretion and elevated tissue stiffness. These pro-fibrotic changes were inhibited by the small-molecule tyrosine kinase inhibitor, Nintedanib. Taken together, our work indicates that a feasible solution to prolong the pre-clinical utility of fibrotic and fibrosis-induced hPCLS is cryopreservation. We anticipate that cryopreserved hPCLS will serve as an advantageous predictive model for the evaluation of pro-fibrotic pathways during acute and chronic toxicity testing.
Collapse
Affiliation(s)
- Méry Marimoutou
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Vivek Patel
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Jae Hun Kim
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Niccole Schaible
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jose Alvarez
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Joseph Hughes
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - McKenzie Obermok
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | | | | | - Béla Suki
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Khalid Amin
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ramaswamy Krishnan
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
11
|
Farrell LA, O’Rourke MB, Padula MP, Souza-Fonseca-Guimaraes F, Caramori G, Wark PAB, Dharmage SC, Hansbro PM. The Current Molecular and Cellular Landscape of Chronic Obstructive Pulmonary Disease (COPD): A Review of Therapies and Efforts towards Personalized Treatment. Proteomes 2024; 12:23. [PMID: 39189263 PMCID: PMC11348234 DOI: 10.3390/proteomes12030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks as the third leading cause of global illness and mortality. It is commonly triggered by exposure to respiratory irritants like cigarette smoke or biofuel pollutants. This multifaceted condition manifests through an array of symptoms and lung irregularities, characterized by chronic inflammation and reduced lung function. Present therapies primarily rely on maintenance medications to alleviate symptoms, but fall short in impeding disease advancement. COPD's diverse nature, influenced by various phenotypes, complicates diagnosis, necessitating precise molecular characterization. Omics-driven methodologies, including biomarker identification and therapeutic target exploration, offer a promising avenue for addressing COPD's complexity. This analysis underscores the critical necessity of improving molecular profiling to deepen our comprehension of COPD and identify potential therapeutic targets. Moreover, it advocates for tailoring treatment strategies to individual phenotypes. Through comprehensive exploration-based molecular characterization and the adoption of personalized methodologies, innovative treatments may emerge that are capable of altering the trajectory of COPD, instilling optimism for efficacious disease-modifying interventions.
Collapse
Affiliation(s)
- Luke A. Farrell
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew B. O’Rourke
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | | | - Gaetano Caramori
- Pulmonology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Peter A. B. Wark
- School of Translational Medicine, Monash University, Melbourne, VIC 3000, Australia;
| | - Shymali C. Dharmage
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Phillip M. Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| |
Collapse
|
12
|
Bellomo A, Herbert J, Kudlak MJ, Laskin JD, Gow AJ, Laskin DL. Identification of early events in nitrogen mustard pulmonary toxicity that are independent of infiltrating inflammatory cells using precision cut lung slices. Toxicol Appl Pharmacol 2024; 486:116941. [PMID: 38677601 DOI: 10.1016/j.taap.2024.116941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Nitrogen mustard (NM; mechlorethamine) is a cytotoxic vesicant known to cause acute lung injury which can progress to chronic disease. Due to the complex nature of NM injury, it has been difficult to analyze early responses of resident lung cells that initiate inflammation and disease progression. To investigate this, we developed a model of acute NM toxicity using murine precision cut lung slices (PCLS), which contain all resident lung cell populations. PCLS were exposed to NM (1-100 μM) for 0.5-3 h and analyzed 1 and 3 d later. NM caused a dose-dependent increase in cytotoxicity and a reduction in metabolic activity, as measured by LDH release and WST-1 activity, respectively. Optimal responses were observed with 50 μM NM after 1 h incubation and these conditions were used in further experiments. Analysis of PCLS bioenergetics using an Agilent Seahorse showed that NM impaired both glycolytic activity and mitochondrial respiration. This was associated with injury to the bronchial epithelium and a reduction in methacholine-induced airway contraction. NM was also found to cause DNA damage in bronchial epithelial cells in PCLS, as measured by expression of γ-H2AX, and to induce oxidative stress, which was evident by a reduction in glutathione levels and upregulation of the antioxidant enzyme catalase. Cleaved caspase-3 was also upregulated in airway smooth muscle cells indicating apoptotic cell death. Characterizing early events in NM toxicity is key in identifying therapeutic targets for the development of efficacious countermeasures.
Collapse
Affiliation(s)
- Alyssa Bellomo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Julia Herbert
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Melissa J Kudlak
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
13
|
Saglam-Metiner P, Yildiz-Ozturk E, Tetik-Vardarli A, Cicek C, Goksel O, Goksel T, Tezcanli B, Yesil-Celiktas O. Organotypic lung tissue culture as a preclinical model to study host- influenza A viral infection: A case for repurposing of nafamostat mesylate. Tissue Cell 2024; 87:102319. [PMID: 38359705 DOI: 10.1016/j.tice.2024.102319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/11/2024] [Accepted: 01/29/2024] [Indexed: 02/17/2024]
Abstract
Reliable and effective models for recapitulation of host-pathogen interactions are imperative for the discovery of potential therapeutics. Ex vivo models can fulfill these requirements as the multicellular native environment in the tissue is preserved and be utilized for toxicology, vaccine, infection and drug efficacy studies due to the presence of immune cells. Drug repurposing involves the identification of new applications for already approved drugs that are not related to the prime medical indication and emerged as a strategy to cope with slow pace of drug discovery due to high costs and necessary phases to reach the patients. Within the scope of the study, broad-spectrum serine protease inhibitor nafamostat mesylate was repurposed to inhibit influenza A infection and evaluated by a translational ex vivo organotypic model, in which human organ-level responses can be achieved in preclinical safety studies of potential antiviral agents, along with in in vitro lung airway culture. The safe doses were determined as 10 µM for in vitro, whereas 22 µM for ex vivo to be applied for evaluation of host-pathogen interactions, which reduced virus infectivity, increased cell/tissue viability, and protected total protein content by reducing cell death with the inflammatory response. When the gene expression levels of specific pro-inflammatory, anti-inflammatory and cell surface markers involved in antiviral responses were examined, the significant inflammatory response represented by highly elevated mRNA gene expression levels of cytokines and chemokines combined with CDH5 downregulated by 5.1-fold supported the antiviral efficacy of NM and usability of ex vivo model as a preclinical infection model.
Collapse
Affiliation(s)
- Pelin Saglam-Metiner
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey; Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey
| | - Ece Yildiz-Ozturk
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey; Department of Food Processing, Food Technology Programme, Yasar University, 35100 Izmir, Turkey
| | - Aslı Tetik-Vardarli
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey; Department of Medical Biology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Candan Cicek
- Department of Medical Microbiology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Ozlem Goksel
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey; Department of Pulmonary Medicine, Division of Allergy and Immunology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Tuncay Goksel
- Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey; Department of Pulmonary Medicine, Division of Allergy and Immunology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | | | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey; Translational Pulmonary Research Center (EgeSAM), Ege University, Izmir 35100, Turkey.
| |
Collapse
|
14
|
López-Posadas R, Bagley DC, Pardo-Pastor C, Ortiz-Zapater E. The epithelium takes the stage in asthma and inflammatory bowel diseases. Front Cell Dev Biol 2024; 12:1258859. [PMID: 38529406 PMCID: PMC10961468 DOI: 10.3389/fcell.2024.1258859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
The epithelium is a dynamic barrier and the damage to this epithelial layer governs a variety of complex mechanisms involving not only epithelial cells but all resident tissue constituents, including immune and stroma cells. Traditionally, diseases characterized by a damaged epithelium have been considered "immunological diseases," and research efforts aimed at preventing and treating these diseases have primarily focused on immuno-centric therapeutic strategies, that often fail to halt or reverse the natural progression of the disease. In this review, we intend to focus on specific mechanisms driven by the epithelium that ensure barrier function. We will bring asthma and Inflammatory Bowel Diseases into the spotlight, as we believe that these two diseases serve as pertinent examples of epithelium derived pathologies. Finally, we will argue how targeting the epithelium is emerging as a novel therapeutic strategy that holds promise for addressing these chronic diseases.
Collapse
Affiliation(s)
- Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universtiy Eralngen-Nürnberg, Erlangen, Germany
| | - Dustin C. Bagley
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Carlos Pardo-Pastor
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, School of Basic and Medical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Universitat de Valencia, Valencia, Spain
- Instituto Investigación Hospital Clínico-INCLIVA, Valencia, Spain
| |
Collapse
|
15
|
Calzetta L, Page C, Matera MG, Cazzola M, Rogliani P. Use of human airway smooth muscle in vitro and ex vivo to investigate drugs for the treatment of chronic obstructive respiratory disorders. Br J Pharmacol 2024; 181:610-639. [PMID: 37859567 DOI: 10.1111/bph.16272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023] Open
Abstract
Isolated airway smooth muscle has been extensively investigated since 1840 to understand the pharmacology of airway diseases. There has often been poor predictability from murine experiments to drugs evaluated in patients with asthma or chronic obstructive pulmonary disease (COPD). However, the use of isolated human airways represents a sensible strategy to optimise the development of innovative molecules for the treatment of respiratory diseases. This review aims to provide updated evidence on the current uses of isolated human airways in validated in vitro methods to investigate drugs in development for the treatment of chronic obstructive respiratory disorders. This review also provides historical notes on the pioneering pharmacological research on isolated human airway tissues, the key differences between human and animal airways, as well as the pivotal differences between human medium bronchi and small airways. Experiments carried out with isolated human bronchial tissues in vitro and ex vivo replicate many of the main anatomical, pathophysiological, mechanical and immunological characteristics of patients with asthma or COPD. In vitro models of asthma and COPD using isolated human airways can provide information that is directly translatable into humans with obstructive lung diseases. Regardless of the technique used to investigate drugs for the treatment of chronic obstructive respiratory disorders (i.e., isolated organ bath systems, videomicroscopy and wire myography), the most limiting factors to produce high-quality and repeatable data remain closely tied to the manual skills of the researcher conducting experiments and the availability of suitable tissue.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Clive Page
- Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
16
|
Reddy KD, Bizymi N, Schweikert A, Ananth S, Lim CX, Lodge KM, Joannes A, Ubags N, van der Does AM, Cloonan SM, Mailleux A, Mansouri N, Reynaert NL, Heijink IH, Cuevas-Ocaña S. ERS International Congress 2023: highlights from the Basic and Translational Sciences Assembly. ERJ Open Res 2024; 10:00875-2023. [PMID: 38686182 PMCID: PMC11057505 DOI: 10.1183/23120541.00875-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 05/02/2024] Open
Abstract
Early career members of Assembly 3 (Basic and Translational Sciences) of the European Respiratory Society (ERS) summarise the key messages discussed during six selected sessions that took place at the ERS International Congress 2023 in Milan, Italy. Aligned with the theme of the congress, the first session covered is "Micro- and macro-environments and respiratory health", which is followed by a summary of the "Scientific year in review" session. Next, recent advances in experimental methodologies and new technologies are discussed from the "Tissue modelling and remodelling" session and a summary provided of the translational science session, "What did you always want to know about omics analyses for clinical practice?", which was organised as part of the ERS Translational Science initiative's aims. The "Lost in translation: new insights into cell-to-cell crosstalk in lung disease" session highlighted how next-generation sequencing can be integrated with laboratory methods, and a final summary of studies is presented from the "From the transcriptome landscape to innovative preclinical models in lung diseases" session, which links the transcriptome landscape with innovative preclinical models. The wide range of topics covered in the selected sessions and the high quality of the research discussed demonstrate the strength of the basic and translational science being presented at the international respiratory conference organised by the ERS.
Collapse
Affiliation(s)
- Karosham Diren Reddy
- Epigenetics of Chronic Lung Disease Group, Forschungszentrum Borstel Leibniz Lungenzentrum, Borstel, Germany
- Division of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- These authors contributed equally
| | - Nikoleta Bizymi
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, Heraklion, Greece
- These authors contributed equally
| | - Anja Schweikert
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- These authors contributed equally
| | - Sachin Ananth
- London North West University Healthcare NHS Trust, London, UK
- These authors contributed equally
| | - Clarice X. Lim
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Lung Health, Clinic Penzing, Vienna, Austria
- These authors contributed equally
| | - Katharine M. Lodge
- National Heart and Lung Institute, Imperial College London, London, UK
- These authors contributed equally
| | - Audrey Joannes
- Université de Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) – UMR_S 1085, Rennes, France
| | - Niki Ubags
- Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne M. Cloonan
- School of Medicine, Trinity Biosciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Arnaud Mailleux
- Université Paris Cité, Inserm, Physiopathologie et épidémiologie des maladies respiratoires, Paris, France
| | - Nahal Mansouri
- Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Niki L. Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonary Diseases, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| | - Sara Cuevas-Ocaña
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
17
|
Kageyama T, Ito T, Tanaka S, Nakajima H. Physiological and immunological barriers in the lung. Semin Immunopathol 2024; 45:533-547. [PMID: 38451292 PMCID: PMC11136722 DOI: 10.1007/s00281-024-01003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
The lungs serve as the primary organ for respiration, facilitating the vital exchange of gases with the bloodstream. Given their perpetual exposure to external particulates and pathogens, they possess intricate protective barriers. Cellular adhesion in the lungs is robustly maintained through tight junctions, adherens junctions, and desmosomes. Furthermore, the pulmonary system features a mucociliary clearance mechanism that synthesizes mucus and transports it to the outside. This mucus is enriched with chemical barriers like antimicrobial proteins and immunoglobulin A (IgA). Additionally, a complex immunological network comprising epithelial cells, neural cells, and immune cells plays a pivotal role in pulmonary defense. A comprehensive understanding of these protective systems offers valuable insights into potential pathologies and their therapeutic interventions.
Collapse
Affiliation(s)
- Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan.
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan.
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba, 260-8670, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| |
Collapse
|