1
|
Teitz-Tennenbaum S, Marinetti KN, Lahiri S, Siddiqui K, Flory C, Tennenbaum K, Hicks HG, Song B, Ganguly A, Osterholzer JJ. Sulfur dioxide exposure of mice induces peribronchiolar fibrosis-A defining feature of deployment-related constrictive bronchiolitis. PLoS One 2025; 20:e0313992. [PMID: 39854594 PMCID: PMC11761160 DOI: 10.1371/journal.pone.0313992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/01/2024] [Indexed: 01/26/2025] Open
Abstract
Deployment-related constrictive bronchiolitis (DRCB) has emerged as a health concern in military personnel returning from Southwest Asia. Exposure to smoke from a fire at the Al-Mishraq sulfur enrichment facility and/or burn pits was reported by a subset of Veterans diagnosed with this disorder. DRCB is characterized by thickening and fibrosis of small airways (SA) in the lung, but whether these are related to toxin inhalation remains uncertain. The aim of this study was to determine whether sulfur dioxide (SO2) exposure can induce histopathological features of DRCB. C57BL/6J mice were exposed to 50 ± 5 ppm SO2 for one hour/day for five consecutive days. Lungs from exposed and unexposed mice were evaluated on day 5, 10, and 20. Lung sections were stained using hematoxylin and eosin, Masson's trichrome, picrosirius red (PSR), and immunofluorescence for club cell secretory protein, acetylated-α-tubulin, and Ki67. Small airway wall thickness was determined by morphometric analysis and collagen content was quantified by measuring PSR fluorescence intensity. CurveAlign and CT-FIRE were used to enumerate collagen fibers and assess fibers' width and length, respectively. Leukocyte subpopulations were quantified by flow cytometry analysis. This protocol of SO2 exposure of mice: 1) Triggered club cell proliferation and differentiation; 2) Increased SA wall thickness by inducing subepithelial collagen deposition; and 3) Increased width, length, and number, but not density, of collagen fibers within the wall of SA. 4) Induced no peribronchiolar inflammation or respiratory bronchiolitis. Collectively, these findings implicate club cell proliferation and differentiation in the profibrotic response to SO2 and identify this SO2 exposure as a potentially effective though imperfect model for studying SA fibrosis in DRCB.
Collapse
Affiliation(s)
- Seagal Teitz-Tennenbaum
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kayla N. Marinetti
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shayanki Lahiri
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Khadijah Siddiqui
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Celia Flory
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karinne Tennenbaum
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Helen G. Hicks
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Brian Song
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
| | - Anutosh Ganguly
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John J. Osterholzer
- Research Service and Pulmonary Section Medical Service, Veterans Affairs Ann Arbor Health System, Ann Arbor, Michigan, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
2
|
Wang J, Peng X, Yuan N, Wang B, Chen S, Wang B, Xie L. Interplay between pulmonary epithelial stem cells and innate immune cells contribute to the repair and regeneration of ALI/ARDS. Transl Res 2024; 272:111-125. [PMID: 38897427 DOI: 10.1016/j.trsl.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
Mammalian lung is the important organ for ventilation and exchange of air and blood. Fresh air and venous blood are constantly delivered through the airway and vascular tree to the alveolus. Based on this, the airways and alveolis are persistently exposed to the external environment and are easily suffered from toxins, irritants and pathogens. For example, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common cause of respiratory failure in critical patients, whose typical pathological characters are diffuse epithelial and endothelial damage resulting in excessive accumulation of inflammatory fluid in the alveolar cavity. The supportive treatment is the main current treatment for ALI/ARDS with the lack of targeted effective treatment strategies. However, ALI/ARDS needs more targeted treatment measures. Therefore, it is extremely urgent to understand the cellular and molecular mechanisms that maintain alveolar epithelial barrier and airway integrity. Previous researches have shown that the lung epithelial cells with tissue stem cell function have the ability to repair and regenerate after injury. Also, it is able to regulate the phenotype and function of innate immune cells involving in regeneration of tissue repair. Meanwhile, we emphasize that interaction between the lung epithelial cells and innate immune cells is more supportive to repair and regenerate in the lung epithelium following acute lung injury. We reviewed the recent advances in injury and repair of lung epithelial stem cells and innate immune cells in ALI/ARDS, concentrating on alveolar type 2 cells and alveolar macrophages and their contribution to post-injury repair behavior of ALI/ARDS through the latest potential molecular communication mechanisms. This will help to develop new research strategies and therapeutic targets for ALI/ARDS.
Collapse
Affiliation(s)
- Jiang Wang
- College of Pulmonary & Critical Care Medicine, the Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Xinyue Peng
- Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Na Yuan
- Department of Pulmonary & Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Bin Wang
- Department of Thoracic Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Siyu Chen
- Department of Thoracic Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Bo Wang
- Department of Thoracic Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, the Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China; Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
3
|
Wong IG, Stark J, Ya V, Moye AL, Vazquez AB, Dang SM, Shehaj A, Rouhani MJ, Bronson R, Janes SM, Rowbotham SP, Paschini M, Franklin RA, Kim CF. Airway injury induces alveolar epithelial and mesenchymal responses mediated by macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587596. [PMID: 38617297 PMCID: PMC11014629 DOI: 10.1101/2024.04.02.587596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Acute injury in the airways or the lung activates local progenitors and stimulates changes in cell-cell interactions to restore homeostasis, but it is not appreciated how more distant niches are impacted. We utilized mouse models of airway-specific epithelial injury to examine secondary tissue-wide alveolar, immune, and mesenchymal responses. Single-cell transcriptomics and in vivo validation revealed transient, tissue-wide proliferation of alveolar type 2 (AT2) progenitor cells after club cell-specific ablation. The AT2 cell proliferative response was reliant on alveolar macrophages (AMs) via upregulation of Spp1 which encodes the secreted factor Osteopontin. A previously uncharacterized mesenchymal population we termed Mesenchymal Airway/Adventitial Niche Cell 2 (MANC2) also exhibited dynamic changes in abundance and a pro-fibrotic transcriptional signature after club cell ablation in an AM-dependent manner. Overall, these results demonstrate that acute airway damage can trigger distal lung responses including altered cell-cell interactions that may contribute to potential vulnerabilities for further dysregulation and disease.
Collapse
|
4
|
Beach TA, Finkelstein JN, Chang PY. Epithelial Responses in Radiation-Induced Lung Injury (RILI) Allow Chronic Inflammation and Fibrogenesis. Radiat Res 2023; 199:439-451. [PMID: 37237442 PMCID: PMC10498477 DOI: 10.1667/rade-22-00103.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 02/20/2023] [Indexed: 05/28/2023]
Abstract
Radiation models, such as whole thorax lung irradiation (WTLI) or partial-body irradiation (PBI) with bone-marrow sparing, have shown that affected lung tissue displays a continual progression of injury, often for months after the initial insult. Undoubtably, a variety of resident and infiltrating cell types either contribute to or fail to resolve this type of progressive injury, which in lung tissue, often develops into lethal and irreversible radiation-induced pulmonary fibrosis (RIPF), indicating a failure of the lung to return to a homeostatic state. Resident pulmonary epithelium, which are present at the time of irradiation and persist long after the initial insult, play a key role in the maintenance of homeostatic conditions in the lung and have often been described as contributing to the progression of radiation-induced lung injury (RILI). In this study, we took an unbiased approach through RNA sequencing to determine the in vivo response of the lung epithelium in the progression of RIPF. In our methodology, we isolated CD326+ epithelium from the lungs of 12.5 Gy WTLI C57BL/6J female mice (aged 8-10 weeks and sacrificed at regular intervals) and compared irradiated and non-irradiated CD326+ cells and whole lung tissue. We subsequently verified our findings by qPCR and immunohistochemistry. Transcripts associated with epithelial regulation of immune responses and fibroblast activation were significantly reduced in irradiated animals at 4 weeks postirradiation. Additionally, alveolar type-2 epithelial cells (AEC2) appeared to be significantly reduced in number at 4 weeks and thereafter based on the diminished expression of pro-surfactant protein C (pro-SPC). This change is associated with a reduction of Cd200 and cyclooxygenase 2 (COX2), which are expressed within the CD326 populations of cells and function to suppress macrophage and fibroblast activation under steady-state conditions, respectively. These data indicate that either preventing epithelial cell loss that occurs after irradiation or replacing important mediators of immune and fibroblast activity produced by the epithelium are potentially important strategies for preventing or treating this unique injury.
Collapse
Affiliation(s)
- Tyler A. Beach
- SRI Biosciences, SRI International, Menlo Park, Calfornia 94025-3493
| | - Jacob N. Finkelstein
- University of Rochester Medical Center, Departments of Pediatrics and Neonatology, and Environmental Medicine, Rochester, New York 14642
| | - Polly Y. Chang
- SRI Biosciences, SRI International, Menlo Park, Calfornia 94025-3493
| |
Collapse
|
5
|
Blackburn JB, Li NF, Bartlett NW, Richmond BW. An update in club cell biology and its potential relevance to chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2023; 324:L652-L665. [PMID: 36942863 PMCID: PMC10110710 DOI: 10.1152/ajplung.00192.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Club cells are found in human small airways where they play an important role in immune defense, xenobiotic metabolism, and repair after injury. Over the past few years, data from single-cell RNA sequencing (scRNA-seq) studies has generated new insights into club cell heterogeneity and function. In this review, we integrate findings from scRNA-seq experiments with earlier in vitro, in vivo, and microscopy studies and highlight the many ways club cells contribute to airway homeostasis. We then discuss evidence for loss of club cells or club cell products in the airways of patients with chronic obstructive pulmonary disease (COPD) and discuss potential mechanisms through which this might occur.
Collapse
Affiliation(s)
- Jessica B Blackburn
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Ngan Fung Li
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease Group, University of Newcastle, Callaghan, New South Wales, Australia
| | - Bradley W Richmond
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
6
|
McGraw MD, Yee M, Kim SY, Dylag AM, Lawrence BP, O'Reilly MA. Diacetyl inhalation impairs airway epithelial repair in mice infected with influenza A virus. Am J Physiol Lung Cell Mol Physiol 2022; 323:L578-L592. [PMID: 36068185 PMCID: PMC9639765 DOI: 10.1152/ajplung.00124.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/12/2022] [Accepted: 08/29/2022] [Indexed: 01/11/2023] Open
Abstract
Bronchiolitis obliterans (BO) is a debilitating disease of the small airways that can develop following exposure to toxic chemicals as well as respiratory tract infections. BO development is strongly associated with diacetyl (DA) inhalation exposures at occupationally relevant concentrations or severe influenza A viral (IAV) infections. However, it remains unclear whether lower dose exposures or more mild IAV infections can result in similar pathology. In the current work, we combined these two common environmental exposures, DA and IAV, to test whether shorter DA exposures followed by sublethal IAV infection would result in similar airways disease. Adult mice exposed to DA vapors 1 h/day for 5 consecutive days followed by infection with the airway-tropic IAV H3N2 (HKx31) resulted in increased mortality, increased bronchoalveolar lavage (BAL) neutrophil percentage, mixed obstruction and restriction by lung function, and subsequent airway remodeling. Exposure to DA or IAV alone failed to result in significant pathology, whereas mice exposed to DA + IAV showed increased α-smooth muscle actin (αSMA) and epithelial cells coexpressing the basal cell marker keratin 5 (KRT5) with the club cell marker SCGB1A1. To test whether DA exposure impairs epithelial repair after IAV infection, mice were infected first with IAV and then exposed to DA during airway epithelial repair. Mice exposed to IAV + DA developed similar airway remodeling with increased subepithelial αSMA and epithelial cells coexpressing KRT5 and SCGB1A1. Our findings reveal an underappreciated concept that common environmental insults while seemingly harmless by themselves can have catastrophic implications on lung function and long-term respiratory health when combined.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Min Yee
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - So-Young Kim
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Andrew M Dylag
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Michael A O'Reilly
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
7
|
Corteselli E, Aboushousha R, Janssen-Heininger Y. S-Glutathionylation-Controlled Apoptosis of Lung Epithelial Cells; Potential Implications for Lung Fibrosis. Antioxidants (Basel) 2022; 11:antiox11091789. [PMID: 36139863 PMCID: PMC9495907 DOI: 10.3390/antiox11091789] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Glutathione (GSH), a major antioxidant in mammalian cells, regulates several vital cellular processes, such as nutrient metabolism, protein synthesis, and immune responses. In addition to its role in antioxidant defense, GSH controls biological processes through its conjugation to reactive protein cysteines in a post-translational modification known as protein S-glutathionylation (PSSG). PSSG has recently been implicated in the pathogenesis of multiple diseases including idiopathic pulmonary fibrosis (IPF). Hallmarks of IPF include repeated injury to the alveolar epithelium with aberrant tissue repair, epithelial cell apoptosis and fibroblast resistance to apoptosis, and the accumulation of extracellular matrix and distortion of normal lung architecture. Several studies have linked oxidative stress and PSSG to the development and progression of IPF. Additionally, it has been suggested that the loss of epithelial cell homeostasis and increased apoptosis, accompanied by the release of various metabolites, creates a vicious cycle that aggravates disease progression. In this short review, we highlight some recent studies that link PSSG to epithelial cell apoptosis and highlight the potential implication of metabolites secreted by apoptotic cells.
Collapse
|
8
|
Kumar A, Elko E, Bruno SR, Mark ZF, Chamberlain N, Mihavics BK, Chandrasekaran R, Walzer J, Ruban M, Gold C, Lam YW, Ghandikota S, Jegga AG, Gomez JL, Janssen-Heininger YM, Anathy V. Inhibition of PDIA3 in club cells attenuates osteopontin production and lung fibrosis. Thorax 2022; 77:669-678. [PMID: 34400514 PMCID: PMC8847543 DOI: 10.1136/thoraxjnl-2021-216882] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND The role of club cells in the pathology of idiopathic pulmonary fibrosis (IPF) is not well understood. Protein disulfide isomerase A3 (PDIA3), an endoplasmic reticulum-based redox chaperone required for the functions of various fibrosis-related proteins; however, the mechanisms of action of PDIA3 in pulmonary fibrosis are not fully elucidated. OBJECTIVES To examine the role of club cells and PDIA3 in the pathology of pulmonary fibrosis and the therapeutic potential of inhibition of PDIA3 in lung fibrosis. METHODS Role of PDIA3 and aberrant club cells in lung fibrosis was studied by analyses of human transcriptome dataset from Lung Genomics Research Consortium, other public resources, the specific deletion or inhibition of PDIA3 in club cells and blocking SPP1 downstream of PDIA3 in mice. RESULTS PDIA3 and club cell secretory protein (SCGB1A1) signatures are upregulated in IPF compared with control patients. PDIA3 or SCGB1A1 increases also correlate with a decrease in lung function in patients with IPF. The bleomycin (BLM) model of lung fibrosis showed increases in PDIA3 in SCGB1A1 cells in the lung parenchyma. Ablation of Pdia3, specifically in SCGB1A1 cells, decreases parenchymal SCGB1A1 cells along with fibrosis in mice. The administration of a PDI inhibitor LOC14 reversed the BLM-induced parenchymal SCGB1A1 cells and fibrosis in mice. Evaluation of PDIA3 partners revealed that SPP1 is a major interactor in fibrosis. Blocking SPP1 attenuated the development of lung fibrosis in mice. CONCLUSIONS Our study reveals a new relationship with distally localised club cells, PDIA3 and SPP1 in lung fibrosis and inhibition of PDIA3 or SPP1 attenuates lung fibrosis.
Collapse
Affiliation(s)
- Amit Kumar
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Evan Elko
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sierra R Bruno
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Zoe F Mark
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Nicolas Chamberlain
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | | | - Ravishankar Chandrasekaran
- Department of Pulmonary, Critical Care Medicine, Larner College of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Joseph Walzer
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Mona Ruban
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Clarissa Gold
- Department of Biology & Vermont Biomedical Research Network Proteomics Facility, University of Vermont, Burlington, Vermont, USA
| | - Ying Wai Lam
- Department of Biology & Vermont Biomedical Research Network Proteomics Facility, University of Vermont, Burlington, Vermont, USA
| | - Sudhir Ghandikota
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Computer Science, University of Cincinnati College of Engineering and Applied Science, Cincinnati, Ohio, USA
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Computer Science, University of Cincinnati College of Engineering and Applied Science, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jose L Gomez
- Internal Medicine-Pulmonary, Critical Care and Sleep Section, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Vikas Anathy
- Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
9
|
Garcia-Arcos I, Park SS, Mai M, Alvarez-Buve R, Chow L, Cai H, Baumlin-Schmid N, Agudelo CW, Martinez J, Kim MD, Dabo AJ, Salathe M, Goldberg IJ, Foronjy RF. LRP1 loss in airway epithelium exacerbates smoke-induced oxidative damage and airway remodeling. J Lipid Res 2022; 63:100185. [PMID: 35202607 PMCID: PMC8953659 DOI: 10.1016/j.jlr.2022.100185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
The LDL receptor-related protein 1 (LRP1) partakes in metabolic and signaling events regulated in a tissue-specific manner. The function of LRP1 in airways has not been studied. We aimed to study the function of LRP1 in smoke-induced disease. We found that bronchial epithelium of patients with chronic obstructive pulmonary disease and airway epithelium of mice exposed to smoke had increased LRP1 expression. We then knocked out LRP1 in human bronchial epithelial cells in vitro and in airway epithelial club cells in mice. In vitro, LRP1 knockdown decreased cell migration and increased transforming growth factor β activation. Tamoxifen-inducible airway-specific LRP1 knockout mice (club Lrp1-/-) induced after complete lung development had increased inflammation in the bronchoalveolar space and lung parenchyma at baseline. After 6 months of smoke exposure, club Lrp1-/- mice showed a combined restrictive and obstructive phenotype, with lower compliance, inspiratory capacity, and forced expiratory volume0.05/forced vital capacity than WT smoke-exposed mice. This was associated with increased values of Ashcroft fibrotic index. Proteomic analysis of room air exposed-club Lrp1-/- mice showed significantly decreased levels of proteins involved in cytoskeleton signaling and xenobiotic detoxification as well as decreased levels of glutathione. The proteome fingerprint created by smoke eclipsed many of the original differences, but club Lrp1-/- mice continued to have decreased lung glutathione levels and increased protein oxidative damage and airway cell proliferation. Therefore, LRP1 deficiency leads to greater lung inflammation and damage and exacerbates smoke-induced lung disease.
Collapse
Affiliation(s)
- Itsaso Garcia-Arcos
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA.
| | - Sangmi S Park
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michelle Mai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Roger Alvarez-Buve
- Respiratory Department, Hospital University Arnau de Vilanova and Santa Maria, IRB Lleida, University of Lleida, Lleida, Catalonia, Spain
| | - Lillian Chow
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Huchong Cai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | | | - Christina W Agudelo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Jennifer Martinez
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michael D Kim
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Abdoulaye J Dabo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Matthias Salathe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ira J Goldberg
- Department of Medicine, NYU Langone School of Medicine, New York, NY, USA
| | - Robert F Foronjy
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| |
Collapse
|
10
|
Teitz-Tennenbaum S, Viglianti SP, Jomma A, Palone Q, Andrews H, Selbmann KN, Lahiri S, Subbotina N, Walker N, Perl AKT, Lama VN, Sisson TH, Osterholzer JJ. Sustained Club Cell Injury in Mice Induces Histopathologic Features of Deployment-Related Constrictive Bronchiolitis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:410-425. [PMID: 34954211 PMCID: PMC8895425 DOI: 10.1016/j.ajpath.2021.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/29/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
Histopathologic evidence of deployment-related constrictive bronchiolitis (DRCB) has been identified in soldiers deployed to Southwest Asia. While inhalational injury to the airway epithelium is suspected, relatively little is known about the pathogenesis underlying this disabling disorder. Club cells are local progenitors critical for repairing the airway epithelium after exposure to various airborne toxins, and a prior study using an inducible transgenic murine model reported that 10 days of sustained targeted club cell injury causes constrictive bronchiolitis. To further understand the mechanisms leading to small airway fibrosis, a murine model was employed to show that sustained club cell injury elicited acute weight loss, caused increased local production of proinflammatory cytokines, and promoted accumulation of numerous myeloid cell subsets in the lung. Transition to a chronic phase was characterized by up-regulated expression of oxidative stress-associated genes, increased activation of transforming growth factor-β, accumulation of alternatively activated macrophages, and enhanced peribronchiolar collagen deposition. Comparative histopathologic analysis demonstrated that sustained club cell injury was sufficient to induce epithelial metaplasia, airway wall thickening, peribronchiolar infiltrates, and clusters of intraluminal airway macrophages that recapitulated key abnormalities observed in DRCB. Depletion of alveolar macrophages in mice decreased activation of transforming growth factor-β and ameliorated constrictive bronchiolitis. Collectively, these findings implicate sustained club cell injury in the development of DRCB and delineate pathways that may yield biomarkers and treatment targets for this disorder.
Collapse
Affiliation(s)
- Seagal Teitz-Tennenbaum
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Steven P Viglianti
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan
| | - Ahmad Jomma
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Quentin Palone
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Halia Andrews
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Kayla N Selbmann
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shayanki Lahiri
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Natalia Subbotina
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Natalie Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Anne-Karina T Perl
- Pulmonary Biology, The Perinatal Institute and Section of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Vibha N Lama
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Thomas H Sisson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - John J Osterholzer
- Research Service and the Pulmonary Section Medical Service, VA Ann Arbor Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
11
|
Peng Y, Wang ZN, Xu AR, Fang ZF, Chen SY, Hou XT, Zhou ZQ, Lin HM, Xie JX, Tang XX, Wang DY, Zhong NS. Mucus Hypersecretion and Ciliary Impairment in Conducting Airway Contribute to Alveolar Mucus Plugging in Idiopathic Pulmonary Fibrosis. Front Cell Dev Biol 2022; 9:810842. [PMID: 35174169 PMCID: PMC8842394 DOI: 10.3389/fcell.2021.810842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease attributed to the complex interplay of genetic and environmental risks. The muco-ciliary clearance (MCC) system plays a critical role in maintaining the conduit for air to and from the alveoli, but it remains poorly understood whether the MCC abnormalities in conducting airway are involved in IPF pathogenesis. In this study, we obtained the surgically resected bronchi and peripheral lung tissues from 31 IPF patients and 39 control subjects, and we sought to explore the morphologic characteristics of MCC in conducting airway by using immunostaining and scanning and transmission electron microscopy. In the submucosal regions of the bronchi, we found that the areas of mucus glands (MUC5B+) were significantly larger in IPF patients as compared with control subjects (p < 0.05). In the surface epithelium of three airway regions (bronchi, proximal bronchioles, and distal bronchioles), increased MUC5B and MUC5AC expression of secretory cells, decreased number of ciliated cells, and increased ciliary length were observed in IPF patients than control subjects (all p < 0.05). In addition, the mRNA expression levels of MUC5B were up-regulated in both the bronchi and peripheral lung of IPF patients than those of control subjects (p < 0.05), accompanied with 93.55% IPF subjects who had obvious MUC5B+ mucus plugs in alveolar regions. No MUC5B rs35705950 single-nucleotide polymorphism allele was detected in both IPF patients and control subjects. Our study shows that mucus hypersecretion and ciliary impairment in conducting airway are major causes of mucus plugs in alveolar regions and may be closely related to the alveolar injuries in IPF patients.
Collapse
Affiliation(s)
- Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhao-Ni Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ai-Ru Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhang-Fu Fang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shi-Ying Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Tao Hou
- Guangzhou KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Zi-Qing Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Hui-Min Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jia-Xing Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Bertho A, Dos Santos M, Braga-Cohen S, Buard V, Paget V, Guipaud O, Tarlet G, Milliat F, François A. Preclinical Model of Stereotactic Ablative Lung Irradiation Using Arc Delivery in the Mouse: Is Fractionation Worthwhile? Front Med (Lausanne) 2022; 8:794324. [PMID: 35004768 PMCID: PMC8739220 DOI: 10.3389/fmed.2021.794324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Lung stereotactic body radiation therapy is characterized by a reduction in target volumes and the use of severely hypofractionated schedules. Preclinical modeling became possible thanks to rodent-dedicated irradiation devices allowing accurate beam collimation and focal lung exposure. Given that a great majority of publications use single dose exposures, the question we asked in this study was as follows: in incremented preclinical models, is it worth using fractionated protocols or should we continue focusing solely on volume limitation? The left lungs of C57BL/6JRj mice were exposed to ionizing radiation using arc therapy and 3 × 3 mm beam collimation. Three-fraction schedules delivered over a period of 1 week were used with 20, 28, 40, and 50 Gy doses per fraction. Lung tissue opacification, global histological damage and the numbers of type II pneumocytes and club cells were assessed 6 months post-exposure, together with the gene expression of several lung cells and inflammation markers. Only the administration of 3 × 40 Gy or 3 × 50 Gy generated focal lung fibrosis after 6 months, with tissue opacification visible by cone beam computed tomography, tissue scarring and consolidation, decreased club cell numbers and a reactive increase in the number of type II pneumocytes. A fractionation schedule using an arc-therapy-delivered three fractions/1 week regimen with 3 × 3 mm beam requires 40 Gy per fraction for lung fibrosis to develop within 6 months, a reasonable time lapse given the mouse lifespan. A comparison with previously published laboratory data suggests that, in this focal lung irradiation configuration, administering a Biological Effective Dose ≥ 1000 Gy should be recommended to obtain lung fibrosis within 6 months. The need for such a high dose per fraction challenges the appropriateness of using preclinical highly focused fractionation schedules in mice.
Collapse
Affiliation(s)
- Annaïg Bertho
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Laboratory of Radiobiology of Accidental Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Sarah Braga-Cohen
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Valérie Buard
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Vincent Paget
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Agnès François
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| |
Collapse
|
13
|
Lin J, Deng H, Zhang Y, Zou L, Fu Z, Dai J. Effect of human umbilical cord-derived mesenchymal stem cells on murine model of bronchiolitis obliterans like injury. Pediatr Pulmonol 2021; 56:129-137. [PMID: 33085211 DOI: 10.1002/ppul.25128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND Bronchiolitis obliterans is a fatal respiratory disease characterized by the obliteration of small airways. Mesenchymal stem cells (MSCs) is a promising candidate for cell-based therapy. OBJECTIVE To evaluate the therapeutic effect of human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) on a murine model of bronchiolitis obliterans like injury (BOLI). METHOD The murine model of BOLI was established by administrating of diacetyl (DA) via intratracheal instillation. Treatment of HUC-MSCs or HUC-MSCs culture medium (HUC-MSCs-CM) was conducted in the BOLI model. RESULTS The pathogenic manifestations, lung function, and the number of neutrophils were similar between the oropharyngeal inhalation DA group (OPI-DA), intratracheal instillation group (ITI-DA); however, less reduction of weight and higher survival rate were observed in ITI-DA groups. Compared with the control groups, the trend of weight loss was significantly reduced (p < .05), and the pulmonary function was significantly improved (p < .05) in HUC-MSCs and HUC-MSCs-CM groups. Masson staining and hematoxylin and eosin staining showed that the deposition of collagen around bronchioles and blood vessels is less and airway epithelial cells and basal cells in lung tissue repaired better in HUC-MSCs and HUC-MSCs-CM groups compared with the control groups. Immunofluorescence shows the expression of E-cadherin and cytokeratin 5 (CK-5) were significantly higher in HUC-MSCs and HUC-MSCs-CM groups compared with control groups, while HUC-MSCs themselves did not express E-cadherin or CK-5. The DiI label showed HUC-MSCs gradually reduced after 2 days in the bronchus and 4 days in bronchiole. CONCLUSION HUC-MSCs could help to repair airway epithelial cells in a murine model of BOLI. It might be related to paracrine factors of HUC-MSCs.
Collapse
Affiliation(s)
- Jilei Lin
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Huarong Deng
- Guangzhou Women and Children's Medical Center, Guangdong, China
| | - Yin Zhang
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lin Zou
- Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhou Fu
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jihong Dai
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
14
|
Tan Q, Ma XY, Liu W, Meridew JA, Jones DL, Haak AJ, Sicard D, Ligresti G, Tschumperlin DJ. Nascent Lung Organoids Reveal Epithelium- and Bone Morphogenetic Protein-mediated Suppression of Fibroblast Activation. Am J Respir Cell Mol Biol 2020; 61:607-619. [PMID: 31050552 DOI: 10.1165/rcmb.2018-0390oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Reciprocal epithelial-mesenchymal interactions are pivotal in lung development, homeostasis, injury, and repair. Organoids have been used to investigate such interactions, but with a major focus on epithelial responses to mesenchyme and less attention to epithelial effects on mesenchyme. In the present study, we used nascent organoids composed of human and mouse lung epithelial and mesenchymal cells to demonstrate that healthy lung epithelium dramatically represses transcriptional, contractile, and matrix synthetic functions of lung fibroblasts. Repression of fibroblast activation requires signaling via the bone morphogenetic protein (BMP) pathway. BMP signaling is diminished after epithelial injury in vitro and in vivo, and exogenous BMP4 restores fibroblast repression in injured organoids. In contrast, inhibition of BMP signaling in healthy organoids is sufficient to derepress fibroblast matrix synthetic function. Our results reveal potent repression of fibroblast activation by healthy lung epithelium and a novel mechanism by which epithelial loss or injury is intrinsically coupled to mesenchymal activation via loss of repressive BMP signaling.
Collapse
Affiliation(s)
- Qi Tan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Xiao Yin Ma
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Wei Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Jeffrey A Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Dakota L Jones
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Giovanni Ligresti
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Bertho A, Dos Santos M, Buard V, Paget V, Guipaud O, Tarlet G, Milliat F, François A. Preclinical Model of Stereotactic Ablative Lung Irradiation Using Arc Delivery in the Mouse: Effect of Beam Size Changes and Dose Effect at Constant Collimation. Int J Radiat Oncol Biol Phys 2020; 107:548-562. [PMID: 32278852 DOI: 10.1016/j.ijrobp.2020.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Stereotactic body radiation therapy is a therapeutic option offered to high surgical risk patients with lung cancer. Focal lung irradiation in mice is a new preclinical model to help understand the development of lung damage in this context. Here we developed a mouse model of lung stereotactic therapy using arc delivery and monitored the development of lung damage while varying the beam size and dose delivered. METHODS AND MATERIALS C57BL/6JRj mice were exposed to 90 Gy focal irradiation on the left lung using 1-mm diameter, 3 × 3 mm2, 7 × 7 mm2, or 10 × 10 mm2 beam collimation for beam size effect and using 3 × 3 mm2 beam collimation delivering 20 to 120 Gy for dose effect. Long-term lung damage was monitored with micro-computed tomography imaging with anatomopathologic and gene expression measurements in the injured patch and the ipsilateral and contralateral lungs. RESULTS Both 1-mm diameter and 3 × 3 mm2 beam collimation allow long-term studies, but only 3-mm beam collimation generates lung fibrosis when delivering 90 Gy. Dose-effect studies with constant 3-mm beam collimation revealed a dose of 60 Gy as the minimum to obtain lung fibrosis 6 months postexposure. Lung fibrosis development was associated with club cell depletion and increased type II pneumocyte numbers. Lung injury developed with ipsilateral and contralateral consequences such as parenchymal thickening and gene expression modifications. CONCLUSIONS Arc therapy allows long-term studies and dose escalation without lethality. In our dose-delivery conditions, dose-effect studies revealed that 3 × 3 mm2 beam collimation to a minimum single dose of 60 Gy enables preclinical models for the assessment of lung injury within a 6-month period. This model of lung tissue fibrosis in a time length compatible with mouse life span may offer good prospects for future mechanistic studies.
Collapse
Affiliation(s)
- Annaïg Bertho
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Accidentelles, Fontenay-aux-Roses, France
| | - Valérie Buard
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Vincent Paget
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France
| | - Agnès François
- Institut de Radioprotection et de Sûreté Nucléaire, Service de Recherche en Radiobiologie et en Médecine régénérative, Laboratoire de Radiobiologie des expositions Médicales, Fontenay-aux-Roses, France.
| |
Collapse
|
16
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
17
|
Beach TA, Groves AM, Williams JP, Finkelstein JN. Modeling radiation-induced lung injury: lessons learned from whole thorax irradiation. Int J Radiat Biol 2020; 96:129-144. [PMID: 30359147 PMCID: PMC6483900 DOI: 10.1080/09553002.2018.1532619] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/25/2022]
Abstract
Models of thoracic irradiation have been developed as clinicians and scientists have attempted to decipher the events that led up to the pulmonary toxicity seen in human subjects following radiation treatment. The most common model is that of whole thorax irradiation (WTI), applied in a single dose. Mice, particularly the C57BL/6J strain, has been frequently used in these investigations, and has greatly informed our current understanding of the initiation and progression of radiation-induced lung injury (RILI). In this review, we highlight the sequential progression and dynamic nature of RILI, focusing primarily on the vast array of information that has been gleaned from the murine model. Ample evidence indicates a wide array of biological responses that can be seen following irradiation, including DNA damage, oxidative stress, cellular senescence and inflammation, all triggered by the initial exposure to ionizing radiation (IR) and heterogeneously maintained throughout the temporal progression of injury, which manifests as acute pneumonitis and later fibrosis. It appears that the early responses of specific cell types may promote further injury, disrupting the microenvironment and preventing a return to homeostasis, although the exact mechanisms driving these responses remains somewhat unclear. Attempts to either prevent or treat RILI in preclinical models have shown some success by targeting these disparate radiobiological processes. As our understanding of the dynamic cellular responses to radiation improves through the use of such models, so does the likelihood of preventing or treating RILI.
Collapse
Affiliation(s)
- Tyler A. Beach
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Angela M. Groves
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
18
|
Sagiv A, Bar-Shai A, Levi N, Hatzav M, Zada L, Ovadya Y, Roitman L, Manella G, Regev O, Majewska J, Vadai E, Eilam R, Feigelson SW, Tsoory M, Tauc M, Alon R, Krizhanovsky V. p53 in Bronchial Club Cells Facilitates Chronic Lung Inflammation by Promoting Senescence. Cell Rep 2019; 22:3468-3479. [PMID: 29590616 DOI: 10.1016/j.celrep.2018.03.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 11/28/2017] [Accepted: 02/28/2018] [Indexed: 12/31/2022] Open
Abstract
The tumor suppressor p53 limits tumorigenesis by inducing apoptosis, cell cycle arrest, and senescence. Although p53 is known to limit inflammation during tumor development, its role in regulating chronic lung inflammation is less well understood. To elucidate the function of airway epithelial p53 in such inflammation, we subjected genetically modified mice, whose bronchial epithelial club cells lack p53, to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to severe chronic bronchitis and airway senescence in wild-type mice. Surprisingly, the club cell p53 knockout mice exhibited reduced airway senescence and bronchitis in response to chronic LPS exposure and were significantly protected from global lung destruction. Furthermore, pharmacological elimination of senescent cells also protected wild-type mice from chronic LPS-induced bronchitis. Our results implicate p53 in induction of club-cell senescence and correlate epithelial cell senescence of chronic airway inflammation and lung destruction.
Collapse
Affiliation(s)
- Adi Sagiv
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Amir Bar-Shai
- Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Naama Levi
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Miki Hatzav
- Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Lior Zada
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yossi Ovadya
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Lior Roitman
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gal Manella
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ofer Regev
- Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Julia Majewska
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ezra Vadai
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Raya Eilam
- Department of Veterinary Resources, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sara W Feigelson
- Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Michel Tauc
- University of Nice Sophia Antipolis, Nice, France
| | - Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Valery Krizhanovsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
19
|
Liu Z, Liao F, Scozzi D, Furuya Y, Pugh KN, Hachem R, Chen DL, Cano M, Green JM, Krupnick AS, Kreisel D, Perl AKT, Huang HJ, Brody SL, Gelman AE. An obligatory role for club cells in preventing obliterative bronchiolitis in lung transplants. JCI Insight 2019; 5:124732. [PMID: 30990794 DOI: 10.1172/jci.insight.124732] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Obliterative bronchiolitis (OB) is a poorly understood airway disease characterized by the generation of fibrotic bronchiolar occlusions. In the lung transplant setting, OB is a pathological manifestation of bronchiolitis obliterans syndrome (BOS), which is a major impediment to long-term recipient survival. Club cells play a key role in bronchiolar epithelial repair, but whether they promote lung transplant tolerance through preventing OB remains unclear. We determined if OB occurs in mouse orthotopic lung transplants following conditional transgene-targeted club cell depletion. In syngeneic lung transplants club cell depletion leads to transient epithelial injury followed by rapid club cell-mediated repair. In contrast, allogeneic lung transplants develop severe OB lesions and poorly regenerate club cells despite immunosuppression treatment. Lung allograft club cell ablation also triggers the recognition of alloantigens, and pulmonary restricted self-antigens reported associated with BOS development. However, CD8+ T cell depletion restores club cell reparative responses and prevents OB. In addition, ex-vivo analysis reveals a specific role for alloantigen-primed effector CD8+ T cells in preventing club cell proliferation and maintenance. Taken together, we demonstrate a vital role for club cells in maintaining lung transplant tolerance and propose a new model to identify the underlying mechanisms of OB.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fuyi Liao
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Davide Scozzi
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Kaitlyn N Pugh
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | - Jonathan M Green
- Department of Medicine.,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexander S Krupnick
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anne Karina T Perl
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Howard J Huang
- Houston Methodist J.C. Walter Jr. Transplant Center, Houston, Texas, USA
| | | | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
20
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
21
|
Post S, Heijink IH, Hesse L, Koo HK, Shaheen F, Fouadi M, Kuchibhotla VNS, Lambrecht BN, Van Oosterhout AJM, Hackett TL, Nawijn MC. Characterization of a lung epithelium specific E-cadherin knock-out model: Implications for obstructive lung pathology. Sci Rep 2018; 8:13275. [PMID: 30185803 PMCID: PMC6125431 DOI: 10.1038/s41598-018-31500-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/13/2018] [Indexed: 12/20/2022] Open
Abstract
The airway epithelium regulates responses to aeroallergens, acting as a physical and immunological barrier. In asthma, epithelial barrier function and the expression of adherens junction protein E-cadherin is compromised, but it is unknown whether this is cause or consequence of the disease. We hypothesized that airway epithelial loss of E-cadherin is a critical step in the development of manifestations of asthma. We generated a transgenic mouse model with conditional loss of E-cadherin in lung epithelial cells at birth and onwards. We observed normal lung development at the time of birth in mice lacking E-cadherin in the lung epithelium. However, E-cadherin deficiency led to progressive epithelial damage in mice growing into adulthood, as evidenced by airway epithelial denudation, decreased zonula occludens (ZO)-1 expression, loss of ciliated cells, and enlarged alveolar spaces. In addition, spontaneous goblet cell metaplasia with mucus production was observed. These epithelial changes were accompanied by elevated levels of the epithelial-derived chemokine CCL17, infiltration of eosinophils and dendritic cells, and mucus production. In conclusion, loss of E-cadherin induces features in the lung reminiscent of those observed in asthma, indicating that the disruption of E-cadherin-mediated cell-cell contacts may play a key role in the development of asthma manifestations.
Collapse
Affiliation(s)
- S Post
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University of British Columbia, Centre for Heart and Lung Innovation, Department of Anesthesiology, Pharmacology and Therapeutics, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - I H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands.
| | - L Hesse
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - H K Koo
- University of British Columbia, Centre for Heart and Lung Innovation, Department of Anesthesiology, Pharmacology and Therapeutics, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - F Shaheen
- University of British Columbia, Centre for Heart and Lung Innovation, Department of Anesthesiology, Pharmacology and Therapeutics, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - M Fouadi
- University of British Columbia, Centre for Heart and Lung Innovation, Department of Anesthesiology, Pharmacology and Therapeutics, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - V N S Kuchibhotla
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - B N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, Department for Molecular Biomedical Research, Inflammation Research Centre (IRC), Ghent, Belgium
- Department of Pulmonary Medicine, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A J M Van Oosterhout
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - T L Hackett
- University of British Columbia, Centre for Heart and Lung Innovation, Department of Anesthesiology, Pharmacology and Therapeutics, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - M C Nawijn
- University of Groningen, University Medical Center Groningen, Department of Pathology & Medical Biology, laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| |
Collapse
|
22
|
van der Velden JL, Wagner DE, Lahue KG, Abdalla ST, Lam YW, Weiss DJ, Janssen-Heininger YMW. TGF-β1-induced deposition of provisional extracellular matrix by tracheal basal cells promotes epithelial-to-mesenchymal transition in a c-Jun NH 2-terminal kinase-1-dependent manner. Am J Physiol Lung Cell Mol Physiol 2018; 314:L984-L997. [PMID: 29469614 PMCID: PMC6032072 DOI: 10.1152/ajplung.00053.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/16/2022] Open
Abstract
Epithelial cells have been suggested as potential drivers of lung fibrosis, although the epithelial-dependent pathways that promote fibrogenesis remain unknown. Extracellular matrix is increasingly recognized as an environment that can drive cellular responses in various pulmonary diseases. In this study, we demonstrate that transforming growth factor-β1 (TGF-β1)-stimulated mouse tracheal basal (MTB) cells produce provisional matrix proteins in vitro, which initiate mesenchymal changes in subsequently freshly plated MTB cells via Rho kinase- and c-Jun NH2-terminal kinase (JNK1)-dependent processes. Repopulation of decellularized lung scaffolds, derived from mice with bleomycin-induced fibrosis or from patients with idiopathic pulmonary fibrosis, with wild-type MTB cells resulted in a loss of epithelial gene expression and augmentation of mesenchymal gene expression compared with cells seeded into decellularized normal lungs. In contrast, Jnk1-/- basal cells seeded into fibrotic lung scaffolds retained a robust epithelial expression profile, failed to induce mesenchymal genes, and differentiated into club cell secretory protein-expressing cells. This new paradigm wherein TGF-β1-induced extracellular matrix derived from MTB cells activates a JNK1-dependent mesenchymal program, which impedes subsequent normal epithelial cell homeostasis, provides a plausible scenario of chronic aberrant epithelial repair, thought to be critical in lung fibrogenesis. This study identifies JNK1 as a possible target for inhibition in settings wherein reepithelialization is desired.
Collapse
Affiliation(s)
- Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Darcy E Wagner
- Department of Medicine, University of Vermont , Burlington, Vermont
- Department of Experimental Medical Sciences, Lung Bioengineering, and Regeneration, Lund University , Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University , Lund, Sweden
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Sarah T Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Ying-Wai Lam
- Department of Biology, University of Vermont , Burlington, Vermont
- Vermont Genetics Networks Proteomics Facility, University of Vermont , Burlington, Vermont
| | - Daniel J Weiss
- Department of Medicine, University of Vermont , Burlington, Vermont
| | | |
Collapse
|
23
|
McGraw MD, Rioux JS, Garlick RB, Rancourt RC, White CW, Veress LA. From the Cover: ImpairedProliferation and Differentiation of the Conducting Airway Epithelium Associated With Bronchiolitis Obliterans After Sulfur Mustard Inhalation Injury in Rats. Toxicol Sci 2018; 157:399-409. [PMID: 28402575 DOI: 10.1093/toxsci/kfx057] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sulfur mustard (SM) is a chemical warfare agent that causes chronic airway remodeling. This study's objective was to assess for changes to the bronchiolar epithelium after SM exposure to explain its contribution to chronic airway remodeling. Materials and methods Adult male rats were exposed to a sublethal dose of SM inhalation (1.0-1.2 mg/kg) for 50 min. Histological sections of the bronchiolar epithelium were analyzed for changes using hematoxylin and eosin, trichrome, and immunofluorescent staining for acetylated tubulin (AT) and club cell secretory protein (CCSP). CCSP in bronchoalveolar lavage fluid was assessed using western blot. A bromodeoxyuridine (BRDU) assay was used to assess for epithelial proliferation, and real-time PCR measured changes in Notch mRNA expression. Results SM caused significant proximal bronchiolar epithelial injury with epithelial denudation, loss of acetylated tubulin and CCSP staining, and reduced bronchoalveolar lavage fluid CCSP levels. bromodeoxyuridine (BRDU) + staining of proximal bronchiolar epithelial cells was not increased, but staining was increased in the distal bronchiolar epithelium. One month after injury, the proximal bronchiolar epithelium was not fully repaired. Significant collagen deposition surrounded proximal bronchioles with luminal obstruction, consistent with bronchiolitis obliterans. These changes corresponded with a downregulation of Notch1, Notch3, and Hes1 mRNA expressions. Conclusions This study demonstrates that SM exposure resulted in severe proximal airway epithelial injury, persistent morphological changes, impaired epithelial proliferation and, ultimately, bronchiolitis obliterans. These changes occurred at the same time that the Notch signaling genes were downregulated. Thus, the lung epithelium and the Notch signaling pathway may be worthy targets for the prevention of chronic airway remodeling after SM inhalation injury.
Collapse
Affiliation(s)
- Matthew D McGraw
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA.,Pediatric Pulmonary Division, The Breathing Institute at Children's Hospital Colorado, Aurora, Colorado, USA
| | - Jaqueline S Rioux
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA
| | - Rhonda B Garlick
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA
| | - Raymond C Rancourt
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA
| | - Carl W White
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA.,Pediatric Pulmonary Division, The Breathing Institute at Children's Hospital Colorado, Aurora, Colorado, USA
| | - Livia A Veress
- Department of Pediatric Pulmonology, University of Colorado Denver, Aurora, Colorado, USA.,Pediatric Pulmonary Division, The Breathing Institute at Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
24
|
Ahmed E, Sansac C, Assou S, Gras D, Petit A, Vachier I, Chanez P, De Vos J, Bourdin A. Lung development, regeneration and plasticity: From disease physiopathology to drug design using induced pluripotent stem cells. Pharmacol Ther 2017; 183:58-77. [PMID: 28987320 DOI: 10.1016/j.pharmthera.2017.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lungs have a complex structure composed of different cell types that form approximately 17 million airway branches of gas-delivering bronchioles connected to 500 million gas-exchanging alveoli. Airways and alveoli are lined by epithelial cells that display a low rate of turnover at steady-state, but can regenerate the epithelium in response to injuries. Here, we review the key points of lung development, homeostasis and epithelial cell plasticity in response to injury and disease, because this knowledge is required to develop new lung disease treatments. Of note, canonical signaling pathways that are essential for proper lung development during embryogenesis are also involved in the pathophysiology of most chronic airway diseases. Moreover, the perfect control of these interconnected pathways is needed for the successful differentiation of induced pluripotent stem cells (iPSC) into lung cells. Indeed, differentiation of iPSC into airway epithelium and alveoli is based on the use of biomimetics of normal embryonic and fetal lung development. In vitro iPSC-based models of lung diseases can help us to better understand the impaired lung repair capacity and to identify new therapeutic targets and new approaches, such as lung cell therapy.
Collapse
Affiliation(s)
- Engi Ahmed
- Department of Respiratory Diseases, Hôpital Arnaud de Villeneuve, Montpellier F34000, France; CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France
| | - Caroline Sansac
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France
| | - Said Assou
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France
| | - Delphine Gras
- Dept of Respiratory Diseases APHM, INSERM CNRS U 1067, UMR7333, Aix-Marseille University, Marseille, France
| | - Aurélie Petit
- INSERM, U1046, PhyMedExp, Montpellier F34000, France
| | | | - Pascal Chanez
- Dept of Respiratory Diseases APHM, INSERM CNRS U 1067, UMR7333, Aix-Marseille University, Marseille, France
| | - John De Vos
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France; CHU Montpellier, Unit for Cellular Therapy, Hospital Saint-Eloi, Montpellier F 34000, France.
| | - Arnaud Bourdin
- Department of Respiratory Diseases, Hôpital Arnaud de Villeneuve, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France; INSERM, U1046, PhyMedExp, Montpellier F34000, France.
| |
Collapse
|
25
|
Sinha M, Lowell CA. Efficiency and Specificity of Gene Deletion in Lung Epithelial Doxycycline-Inducible Cre Mice. Am J Respir Cell Mol Biol 2017; 57:248-257. [PMID: 28287822 DOI: 10.1165/rcmb.2016-0208oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The transgenic mouse strains surfactant protein C-reverse tetracycline transactivator (SP-C-rtTA), club cell secretory protein (CCSP)-rtTA, and tetracycline operator (TetO)-Cre have been invaluable for spatiotemporally regulating gene deletion in the pulmonary epithelium. In this study, we measured the efficiency and specificity of gene deletion that can be achieved in these mice using the Rosa26-eYFP reporter. Triple-transgenic mice (tTg or rtTA/TetO-Cre/Rosa-eYFP) were bred and treated with various doxycycline (dox) regimens to induce gene deletion, which was then quantified in various cell populations by flow cytometry. In these crosses, we found that the TetO-Cre transgene must be transmitted through the female parent to avoid germline gene deletion. With dox exposure during lung development, SP-C-tTg mice deleted in ∼65-75% of alveolar epithelial type II (ATII) cells, but in only ∼45-50% of the integrin β4+ population, which consisted of club cells and distal lung progenitor cells. In contrast, CCSP-tTg mice deleted in ∼50% of ATII cells and ∼80% of integrin β4+ cells. Upon dox treatment of adults, deletion in ATII cells and integrin β4+ cells in SP-C-tTg mice dropped significantly to ∼20% and ∼6%, respectively, whereas CCSP-tTg mice deleted in ∼57% of ATII and ∼40% of integrin β4+ cells. Interestingly, untreated CCSP-tTg mice also deleted in ∼40% of integrin β4+ cells, indicating significant leakiness of CCSP-tTg in β4+ cells. In all mouse groups, minimal deletion occurred in mouse tracheal epithelial cells or in mesenchymal or hematopoietic cells. These data provide the first quantitative, side-by-side comparison of the deletion efficiency for these widely used transgenic mouse strains.
Collapse
Affiliation(s)
- Meenal Sinha
- Department of Laboratory Medicine and Program in Immunology, University of California, San Francisco, California
| | - Clifford A Lowell
- Department of Laboratory Medicine and Program in Immunology, University of California, San Francisco, California
| |
Collapse
|
26
|
Tang X, Sun L, Jin X, Chen Y, Zhu H, Liang Y, Wu Q, Han X, Liang J, Liu X, Liang Z, Wang G, Luo F. Runt-Related Transcription Factor 1 Regulates LPS-Induced Acute Lung Injury via NF-κB Signaling. Am J Respir Cell Mol Biol 2017; 57:174-183. [PMID: 28314106 DOI: 10.1165/rcmb.2016-0319oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Runt-related transcription factor 1 (RUNX1), a transcription factor expressed in multiple organs, plays important roles in embryonic development and hematopoiesis. Although RUNX1 is highly expressed in pulmonary tissues, its roles in lung function and homeostasis are unknown. We sought to assess the role of RUNX1 in lung development and inflammation after LPS challenge. Expression of RUNX1 was assessed in the developing and postnatal lung. RUNX1 was conditionally deleted in pulmonary epithelial cells. Pulmonary maturation was evaluated in the developing and postnatal lung, and lung inflammation was investigated in adult mice after LPS challenge. Interactions between RUNX1 and inflammatory signaling via NF-κB-IkB kinase β were assessed in vitro. RUNX1 was expressed in both mesenchymal and epithelial compartments of the developing and postnatal lung. The RUNX1 gene was efficiently deleted from respiratory epithelial cells producing Runx1∆/∆ mice. Although lung maturation was delayed, Runx1∆/∆ mice survived postnatally and subsequent growth and maturation of the lung proceeded normally. Increased respiratory distress, inflammation, and proinflammatory cytokines were observed in the Runx1-deleted mice after pulmonary LPS exposure. RUNX1 deletion was associated with the activation of NF-κB in respiratory epithelial cells. RUNX1 was required for the suppression of NF-κB signaling pathway via inhibition of IkB kinase β in in vitro studies. RUNX1 plays a critical role in the lung inflammation after LPS-induced injury.
Collapse
Affiliation(s)
- Xiaoju Tang
- 1 Department of Respiratory Medicine.,2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Ling Sun
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | - Xiaodong Jin
- 3 Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hui Zhu
- 1 Department of Respiratory Medicine
| | - Yasha Liang
- 4 First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingbo Wu
- 1 Department of Respiratory Medicine
| | - Xing Han
- 5 Fourth People's Hospital of Sichuan Province, Chengdu, China
| | - Jianing Liang
- 6 Department of Respiratory Medicine, Fourth Military Medical University, Xian, China
| | - Xiaojing Liu
- 2 Laboratory of Cardiovascular Diseases, Research Center of Regeneration Medicine, and
| | | | - Gang Wang
- 1 Department of Respiratory Medicine
| | | |
Collapse
|
27
|
Ilic N, Mihailovic N. Serum Clara cell protein and atopic phenotype in children up to 2 years of age. J Clin Lab Anal 2017; 31. [PMID: 28146340 DOI: 10.1002/jcla.22151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/26/2016] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Low value of serum Clara cell protein (CC16) is associated with bronchial hyperreactivity in children. OBJECTIVE To evaluate the serum CC16 in relation to atopy and previously manifested LRTD. METHODS In the population of 163 healthy 5- to 24-month-old children, atopy was determined by Phadiatop-infant (serum-specific IgE≥0.35 kUA/L), serum CC16 by ELISA, while data on previously manifested low respiratory tract diseases (LRTD) were collected from the Health Care Center database. RESULTS In atopic children, serum CC16 negatively correlated with age (r -.281, P=.041, n=53), while in nonatopic children, this correlation was positive (r .200, P=.036, n=110). Atopic ≥8-month-old children with previously manifested LRTD had lower level of CC16 (3.07 ng/mL) in relation to atopic children without LRTD at the same age (6.51 ng/mL), P=.029 (value of serum CC16≥4.8 ng/mL indicates atopic phenotype without LRTD 75% sensitivity, 87.5% specificity). In 8- to 24-month-old children with previously manifested pneumonia, serum CC16 was lower in atopic (2.9 ng/mL) in relation to nonatopic children (3.7 ng/mL), P=.029 (serum CC16 ≤3.4 ng/mL indicating atopy in the group of children with pneumonia, sensitivity 100%, and specificity 77%). Atopic 8- to 24-month-old children with previously manifested pneumonia had lower CC16 in relation to other atopic children in this age (P=.021) (for cutoff CC16≤3.4 ng/mL sensitivity 100%, specificity 77%), and also often chronic wheezing (atopic with pneumonia 83.3%, n=5/6 vs atopic without pneumonia 21.4%, n=3/14), P=.018. CONCLUSION Low serum CC16 is associated with previously expressed pneumonia and chronic wheezing in atopic children.
Collapse
Affiliation(s)
- Nevenka Ilic
- Department of Allergology and Immunology, Public Health Institute, Kragujevac, Serbia
| | - Natasa Mihailovic
- Department of Biostatistics, Public Health Institute, Kragujevac, Serbia
| |
Collapse
|
28
|
Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol 2016; 138:16-27. [PMID: 27373322 DOI: 10.1016/j.jaci.2016.05.011] [Citation(s) in RCA: 995] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with chronic inflammation affecting predominantly the lung parenchyma and peripheral airways that results in largely irreversible and progressive airflow limitation. This inflammation is characterized by increased numbers of alveolar macrophages, neutrophils, T lymphocytes (predominantly TC1, TH1, and TH17 cells), and innate lymphoid cells recruited from the circulation. These cells and structural cells, including epithelial and endothelial cells and fibroblasts, secrete a variety of proinflammatory mediators, including cytokines, chemokines, growth factors, and lipid mediators. Although most patients with COPD have a predominantly neutrophilic inflammation, some have an increase in eosinophil counts, which might be orchestrated by TH2 cells and type 2 innate lymphoid cells though release of IL-33 from epithelial cells. These patients might be more responsive to corticosteroids and bronchodilators. Oxidative stress plays a key role in driving COPD-related inflammation, even in ex-smokers, and might result in activation of the proinflammatory transcription factor nuclear factor κB (NF-κB), impaired antiprotease defenses, DNA damage, cellular senescence, autoantibody generation, and corticosteroid resistance though inactivation of histone deacetylase 2. Systemic inflammation is also found in patients with COPD and can worsen comorbidities, such as cardiovascular diseases, diabetes, and osteoporosis. Accelerated aging in the lungs of patients with COPD can also generate inflammatory protein release from senescent cells in the lung. In the future, it will be important to recognize phenotypes of patients with optimal responses to more specific therapies, and development of biomarkers that identify the therapeutic phenotypes will be important.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom.
| |
Collapse
|
29
|
Yang C, Jiang J, Yang X, Wang H, Du J. Stem/progenitor cells in endogenous repairing responses: new toolbox for the treatment of acute lung injury. J Transl Med 2016; 14:47. [PMID: 26865361 PMCID: PMC4750219 DOI: 10.1186/s12967-016-0804-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023] Open
Abstract
The repair of organs and tissues has stepped into a prospective era of regenerative medicine. However, basic research and clinical practice in the lung regeneration remains crawling. Owing to the complicated three dimensional structures and above 40 types of pulmonary cells, the regeneration of lung tissues becomes a great challenge. Compelling evidence has showed that distinct populations of intrapulmonary and extrapulmonary stem/progenitor cells can regenerate epithelia as well as endothelia in various parts of the respiratory tract. Recently, the discovery of human lung stem cells and their relevant studies has opened the door of hope again, which might put us on the path to repair our injured body parts, lungs on demand. Herein, we emphasized the role of endogenous and exogenous stem/progenitor cells in lungs as well as artificial tissue repair for the injured lungs, which constitute a marvelous toolbox for the treatment of acute lung injury. Finally, we further discussed the potential problems in the pulmonary remodeling and regeneration.
Collapse
Affiliation(s)
- Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Xuetao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Haiyan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, 400042, Chongqing, China.
| |
Collapse
|
30
|
An official American Thoracic Society workshop report: stem cells and cell therapies in lung biology and diseases. Ann Am Thorac Soc 2016; 12:S79-97. [PMID: 25897748 DOI: 10.1513/annalsats.201502-086st] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The University of Vermont College of Medicine and the Vermont Lung Center, in collaboration with the NHLBI, Alpha-1 Foundation, American Thoracic Society, European Respiratory Society, International Society for Cell Therapy, and the Pulmonary Fibrosis Foundation, convened a workshop, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," held July 29 to August 1, 2013 at the University of Vermont. The conference objectives were to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are all rapidly expanding areas of study that both provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases. This conference was a follow-up to four previous biennial conferences held at the University of Vermont in 2005, 2007, 2009, and 2011. Each of those conferences, also sponsored by the National Institutes of Health, American Thoracic Society, and Respiratory Disease Foundations, has been important in helping guide research and funding priorities. The major conference recommendations are summarized at the end of the report and highlight both the significant progress and major challenges in these rapidly progressing fields.
Collapse
|
31
|
Yang C, Yang X, Du J, Wang H, Li H, Zeng L, Gu W, Jiang J. Retinoic acid promotes the endogenous repair of lung stem/progenitor cells in combined with simvastatin after acute lung injury: a stereological analysis. Respir Res 2015; 16:140. [PMID: 26561298 PMCID: PMC4642746 DOI: 10.1186/s12931-015-0300-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 11/03/2015] [Indexed: 11/10/2022] Open
Abstract
Background The treatment of acute respiratory distress syndrome (ARDS), most commonly seen during the organ dysfunction remains unsatisfied. Presently, the stem/progenitor cell-based endogenous repair has been aroused attention enormously. This report investigated the effects of retinoic acid (RA) plus simvastatin (SS) with respect to dynamics of lung repair cells as well as to elucidate the underlying mechanism. Materials and methods The experimental Sprague–Dawley rats were divided randomly into normal control (control), sham operated (sham), ARDS, ARDS + vehicle and ARDS + RA + SS groups. ARDS was reproduced through hemorrhagic shock/resuscitation (shock) and subsequent intratracheal LPS (4.5 mg/kg, Escherichia coli serotype O55: B5) injection. The rats were treated by intragastric administration of RA (2 mg/kg/day) and SS (2 mg/kg/day) for 5 days in the ARDS + RA + SS group. Seven days after the first RA-SS injection, a right lower lobe of lung was sampled for histological analysis concerning systemic uniform random sampling method. Immunohistochemistry of inflation-fixed lungs for alveolar type 1 (AT1), alveolar type 2 (AT2) and Clara cells was measured by AQP5, Pro-SPC and CCSP staining respectively. The alveolar cell proliferation and apoptosis were analyzed with Ki67 staining and terminal deoxylnucleotidyl transferase mediated-dUTP nick end labeling (TUNEL) method. Meanwhile, the alveolar cell numerical and surface density (alveolar cells, AT1, AT2, Clara, proliferating and apoptotic cells) were evaluated by stereology. Results RA-SS compound exerted anti-inflammatory and pro-repairing effects on respiratory tracts in ARDS induced by hemorrhagic-endotoxin shock. The numerical density and surface density of alveolar cells, AT1 cell fraction, and numerical density of AT2 and Clara cells were significantly increased after treatment with RA-SS compound in ARDS. Concurrently, the Ki67+ alveolar cells were obviously increased while the TUNEL+ alveolar cells were reduced, which was correlated with the attenuation of inflammatory injury and functional repair in injured lung tissues. Conclusions Our data convincingly indicated that the prophylactic and therapeutic treatment of RA plus SS had obvious beneficial effect on the remodeling/regeneration of injured pulmonary tissues, suggesting that the underlying mechanisms are related to the re-balance between regeneration and apoptosis in lung stem/progenitor cells.
Collapse
Affiliation(s)
- Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China. .,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, Chongqing, 400042, China.
| | - Xuetao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Haiyan Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Haisheng Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Wei Gu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China. .,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Changjiang Zhilu, Daping, Chongqing, 400042, China.
| |
Collapse
|
32
|
Ortega-Martínez M, Rodríguez-Flores LE, de-la-Garza-González C, Ancer-Rodríguez J, Jaramillo-Rangel G. Detection of a novel stem cell probably involved in normal turnover of the lung airway epithelium. J Cell Mol Med 2015; 19:2679-81. [PMID: 26257389 PMCID: PMC4627572 DOI: 10.1111/jcmm.12653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/09/2015] [Indexed: 01/22/2023] Open
Abstract
Regeneration of the lung airway epithelium after injury has been extensively studied. In contrast, analysis of its turnover in healthy adulthood has received little attention. In the classical view, this epithelium is maintained in the steady-state by the infrequent proliferation of basal or Clara cells. The intermediate filament protein nestin was initially identified as a marker for neural stem cells, but its expression has also been detected in other stem cells. Lungs from CD1 mice at the age of 2, 6, 12, 18 or 24 months were fixed in neutral-buffered formalin and paraffin-embedded. Nestin expression was examined by an immunohistochemical peroxidase-based method. Nestin-positive cells were detected in perivascular areas and in connective tissue that were in close proximity of the airway epithelium. Also, nestin-positive cells were found among the cells lining the airway epithelium. These findings suggest that nestin-positive stem cells circulate in the bloodstream, transmigrate through blood vessels and localize in the lung airway epithelium to participate in its turnover. We previously reported the existence of similar cells able to differentiate into lung chondrocytes. Thus, the stem cell reported here might be a bone marrow-derived mesenchymal stem cell (BMDMSC) able to generate several types of lung tissues. In conclusion, our findings indicate that there exist a BMDMSC in healthy adulthood that participates in the turnover of the lung airway epithelium. These findings may improve our knowledge about the lung stem cell biology and also provide novel approaches to therapy for devastating pulmonary diseases.
Collapse
Affiliation(s)
- Marta Ortega-Martínez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| | - Laura E Rodríguez-Flores
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| | - Carlos de-la-Garza-González
- Department of Embryology, School of Medicine, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| | - Jesús Ancer-Rodríguez
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo LeonMonterrey, Nuevo Leon, Mexico
| |
Collapse
|
33
|
Affiliation(s)
- Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, England.
| |
Collapse
|
34
|
Epa AP, Thatcher TH, Pollock SJ, Wahl LA, Lyda E, Kottmann RM, Phipps RP, Sime PJ. Normal Human Lung Epithelial Cells Inhibit Transforming Growth Factor-β Induced Myofibroblast Differentiation via Prostaglandin E2. PLoS One 2015; 10:e0135266. [PMID: 26248335 PMCID: PMC4527711 DOI: 10.1371/journal.pone.0135266] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 07/20/2015] [Indexed: 11/18/2022] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a chronic progressive disease with very few effective treatments. The key effector cells in fibrosis are believed to be fibroblasts, which differentiate to a contractile myofibroblast phenotype with enhanced capacity to proliferate and produce extracellular matrix. The role of the lung epithelium in fibrosis is unclear. While there is evidence that the epithelium is disrupted in IPF, it is not known whether this is a cause or a result of the fibroblast pathology. We hypothesized that healthy epithelial cells are required to maintain normal lung homeostasis and can inhibit the activation and differentiation of lung fibroblasts to the myofibroblast phenotype. To investigate this hypothesis, we employed a novel co-culture model with primary human lung epithelial cells and fibroblasts to investigate whether epithelial cells inhibit myofibroblast differentiation. Measurements and Main Results In the presence of transforming growth factor (TGF)-β, fibroblasts co-cultured with epithelial cells expressed significantly less α-smooth muscle actin and collagen and showed marked reduction in cell migration, collagen gel contraction, and cell proliferation compared to fibroblasts grown without epithelial cells. Epithelial cells from non-matching tissue origins were capable of inhibiting TGF-β induced myofibroblast differentiation in lung, keloid and Graves’ orbital fibroblasts. TGF-β promoted production of prostaglandin (PG) E2 in lung epithelial cells, and a PGE2 neutralizing antibody blocked the protective effect of epithelial cell co-culture. Conclusions We provide the first direct experimental evidence that lung epithelial cells inhibit TGF-β induced myofibroblast differentiation and pro-fibrotic phenotypes in fibroblasts. This effect is not restricted by tissue origin, and is mediated, at least in part, by PGE2. Our data support the hypothesis that the epithelium plays a crucial role in maintaining lung homeostasis, and that damaged and/ or dysfunctional epithelium contributes to the development of fibrosis.
Collapse
Affiliation(s)
- Amali P. Epa
- Department of Pathology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Thomas H. Thatcher
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Stephen J. Pollock
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Lindsay A. Wahl
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Elizabeth Lyda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - R. M. Kottmann
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
| | - Richard P. Phipps
- Department of Pathology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester NY, 14642, United States of America
| | - Patricia J. Sime
- Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY 14642, United States of America
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester NY, 14642, United States of America
- * E-mail:
| |
Collapse
|
35
|
Gamez AS, Gras D, Petit A, Knabe L, Molinari N, Vachier I, Chanez P, Bourdin A. Supplementing Defect in Club Cell Secretory Protein Attenuates Airway Inflammation in COPD. Chest 2015; 147:1467-1476. [DOI: 10.1378/chest.14-1174] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
36
|
Emura M, Aufderheide M, Mohr U. Target cell types with stem/progenitor function to isolate for in vitro reconstruction of human bronchiolar epithelia. ACTA ACUST UNITED AC 2015; 67:81-8. [DOI: 10.1016/j.etp.2014.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/19/2022]
|
37
|
Lange AW, Sridharan A, Xu Y, Stripp BR, Perl AK, Whitsett JA. Hippo/Yap signaling controls epithelial progenitor cell proliferation and differentiation in the embryonic and adult lung. J Mol Cell Biol 2014; 7:35-47. [PMID: 25480985 DOI: 10.1093/jmcb/mju046] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Hippo/Yap pathway is a well-conserved signaling cascade that regulates cell proliferation and differentiation to control organ size and stem/progenitor cell behavior. Following airway injury, Yap was dynamically regulated in regenerating airway epithelial cells. To determine the role of Hippo signaling in the lung, the mammalian Hippo kinases, Mst1 and Mst2, were deleted in epithelial cells of the embryonic and mature mouse lung. Mst1/2 deletion in the fetal lung enhanced proliferation and inhibited sacculation and epithelial cell differentiation. The transcriptional inhibition of cell proliferation and activation of differentiation during normal perinatal lung maturation were inversely regulated following embryonic Mst1/2 deletion. Ablation of Mst1/2 from bronchiolar epithelial cells in the adult lung caused airway hyperplasia and altered differentiation. Inhibitory Yap phosphorylation was decreased and Yap nuclear localization and transcriptional targets were increased after Mst1/2 deletion, consistent with canonical Hippo/Yap signaling. YAP potentiated cell proliferation and inhibited differentiation of human bronchial epithelial cells in vitro. Loss of Mst1/2 and expression of YAP regulated transcriptional targets controlling cell proliferation and differentiation, including Ajuba LIM protein. Ajuba was required for the effects of YAP on cell proliferation in vitro. Hippo/Yap signaling regulates Ajuba and controls proliferation and differentiation of lung epithelial progenitor cells.
Collapse
Affiliation(s)
- Alexander W Lange
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Anusha Sridharan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Yan Xu
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | | | - Anne-Karina Perl
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| |
Collapse
|
38
|
Aoshiba K, Tsuji T, Itoh M, Semba S, Yamaguchi K, Nakamura H, Watanabe H. A murine model of airway fibrosis induced by repeated naphthalene exposure. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 2014; 66:169-177. [PMID: 24480153 DOI: 10.1016/j.etp.2014.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/16/2013] [Accepted: 01/03/2014] [Indexed: 02/07/2023]
Abstract
The airway epithelium serves as a biological barrier essential for host defense against inhaled pollutants. While chronic epithelial injury, commonly associated with chronic obstructive pulmonary disease and bronchiolitis obliterans syndrome, often results in airway fibrosis, limited animal models of airway fibrosis have been established. Club cells (Clara cells) in the small airways represent an important population of epithelial progenitor cells and also the principal site of localization of the cytochrome P-450 monooxygenase system, which metabolically activates xenobiotic chemicals such as naphthalene by converting them to toxic epoxide intermediates. We hypothesized that repeated exposure to naphthalene may cause prolonged loss of club cells, triggering aberrant local epithelial repair mechanisms that lead to peribronchial fibrosis. We administered intraperitoneal injections of naphthalene to C57/BL6J mice once a week for 14 consecutive weeks. Repeated club cell injury caused by naphthalene triggered regional hyperproliferation of epithelial progenitor cells, while other regions remained denuded or squamated, resulting in fibroblast proliferation and peribronchial collagen deposition associated with upregulation of the fibrogenic cytokines transforming growth factor-β and connective tissue growth factor. The total collagen content of the lung assessed by measurement of the hydroxyproline content was also increased after repeated exposure to naphthalene. These results lend support to the relevance of repeated injury of airway epithelial cells as a trigger for resting fibroblast proliferation and airway fibrosis. This model of airway fibrosis is simple and easy to reproduce, and may be expected to advance our understanding of the pathogenesis and potential treatment of airway fibrotic disorders.
Collapse
Affiliation(s)
- Kazutetsu Aoshiba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan.
| | - Takao Tsuji
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan
| | - Masayuki Itoh
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan
| | - Seitaro Semba
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan
| | - Kazuhiro Yamaguchi
- Comprehensive and Internal Medicine, Tokyo Women's Medical University Medical Center East, Japan
| | - Hiroyuki Nakamura
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan
| | - Hidehiro Watanabe
- Department of Respiratory Medicine, Tokyo Medical University Ibaraki Medical Center, Japan
| |
Collapse
|
39
|
Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac Soc 2014; 10:S45-97. [PMID: 23869446 DOI: 10.1513/annalsats.201304-090aw] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A conference, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," was held July 25 to 28, 2011 at the University of Vermont to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, to discuss and debate current controversies, and to identify future research directions and opportunities for basic and translational research in cell-based therapies for lung diseases. The goal of this article, which accompanies the formal conference report, is to provide a comprehensive review of the published literature in lung regenerative medicine from the last conference report through December 2012.
Collapse
|
40
|
Hamvas A, Deterding R, Balch WE, Schwartz DA, Albertine KH, Whitsett JA, Cardoso WV, Kotton DN, Kourembanas S, Hagood JS. Diffuse lung disease in children: summary of a scientific conference. Pediatr Pulmonol 2014; 49:400-9. [PMID: 23798474 PMCID: PMC4145861 DOI: 10.1002/ppul.22805] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 02/24/2013] [Indexed: 12/14/2022]
Abstract
A multi-disciplinary scientific conference focused on diffuse and interstitial lung diseases in children was held in La Jolla, CA in June 2012. The conference brought together clinicians (including Pediatric and Adult Pulmonologists, Neonatologists, Pathologists, and Radiologists), clinical researchers, basic scientists, government agency representatives, patient advocates, as well as children affected by diffuse lung disease (DLD) and their families, to review recent scientific developments and emerging concepts in the pathophysiology of childhood DLD. Invited speakers discussed translational approaches, including genetics and proteomics, epigenetics and epigenomics, models of DLD, including animal models and induced pluripotent stem cells, and regenerative medicine approaches. The presentations of the invited speakers are summarized here.
Collapse
Affiliation(s)
- Aaron Hamvas
- Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
O'Koren EG, Hogan BLM, Gunn MD. Loss of basal cells precedes bronchiolitis obliterans-like pathological changes in a murine model of chlorine gas inhalation. Am J Respir Cell Mol Biol 2013; 49:788-97. [PMID: 23742075 DOI: 10.1165/rcmb.2012-0369oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchiolitis obliterans (BO) is a major cause of chronic airway dysfunction after toxic chemical inhalation. The pathophysiology of BO is not well understood, but epithelial cell injury has been closely associated with the development of fibrotic lesions in human studies and in animal models of both toxin-induced and transplant-induced BO. However, whereas almost all cases and models of BO include epithelial injury, not all instances of epithelial injury result in BO, suggesting that epithelial damage per se is not the critical event leading to the development of BO. Here, we describe a model of chlorine-induced BO in which mice develop tracheal and large airway obliterative lesions within 10 days of exposure to high (350 parts per million [ppm]), but not low (200 ppm), concentrations of chlorine gas. Importantly, these lesions arise only under conditions and in areas in which basal cells, the resident progenitor cells for large airway epithelium, are eliminated by chlorine exposure. In areas of basal cell loss, epithelial regeneration does not occur, resulting in persistent regions of epithelial denudation. Obliterative airway lesions arise specifically from regions of epithelial denudation in a process that includes inflammatory cell infiltration by Day 2 after exposure, fibroblast infiltration and collagen deposition by Day 5, and the ingrowth of blood vessels by Day 7, ultimately leading to lethal airway obstruction by Days 9-12. We conclude that the loss of epithelial progenitor cells constitutes a critical factor leading to the development of obliterative airway lesions after chemical inhalation.
Collapse
|
42
|
Development of lysophosphatidic acid pathway modulators as therapies for fibrosis. Future Med Chem 2013; 5:1935-52. [DOI: 10.4155/fmc.13.154] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a class of bioactive phospholipid that displays a wide range of cellular effects via LPA receptors, of which six have been identified (LPAR1–6). In serum and plasma, LPA production occurs mainly by the hydrolysis of lysophosphatidylcholine by the phospholipase D activity of autotaxin (ATX). The involvement of the LPA pathway in driving chronic wound-healing conditions, such as idiopathic pulmonary fibrosis, has suggested targets in this pathway could provide potential therapeutic approaches. Mice with LPAR1 knockout or tissue-specific ATX deletion have demonstrated reduced lung fibrosis following bleomycin challenge. Therefore, strategies aimed at antagonizing LPA receptors or inhibiting ATX have gained considerable attention. This Review will summarize the current status of identifying small-molecule modulators of the LPA pathway. The therapeutic utility of LPA modulators for the treatment of fibrotic diseases will soon be revealed as clinical trials are already in progress in this area.
Collapse
|
43
|
Manning CM, Johnston CJ, Hernady E, Miller JNH, Reed CK, Lawrence BP, Williams JP, Finkelstein JN. Exacerbation of lung radiation injury by viral infection: the role of Clara cells and Clara cell secretory protein. Radiat Res 2013; 179:617-29. [PMID: 23621375 DOI: 10.1667/rr3279.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Viral infections have been associated with exacerbation of disease in human cases of idiopathic pulmonary fibrosis. Since pulmonary fibrosis is a common outcome after irradiation to the lung, we hypothesized that viral infection after radiation exposure would exacerbate radiation-induced lung injury. Epithelial injury, a frequent outcome after infection, has been hypothesized to contribute to the pathogenesis of pulmonary fibrosis and bronchiolar epithelial Clara cells participate in epithelial repair. Therefore, it was further hypothesized that altered responses after irradiation involve the bronchiolar epithelial Clara cells. C57BL/6J or CCSP(-/-) mice were irradiated with 0 (sham), 5, 10 or 15 Gy to the whole thorax. At ten weeks post-irradiation, animals were mock infected or infected with influenza A virus and body weight and survival were monitored. Pulmonary function was assessed by whole-body plethysmography. The Clara cell markers, CCSP and Cyp2f2, were measured in the lung by qRT-PCR, and protein expression was visualized in the lung by immunofluorescence. Following pulmonary function tests, mice were sacrificed and tissues were collected for pathological analysis. In 15 Gy irradiated animals infected with influenza A virus, accelerated respiratory rates, reduced pulmonary function, and exacerbated lung pathology occurred earlier post-irradiation than previously observed after irradiation alone, suggesting infection accelerates the development of radiation injury. After irradiation alone, CCSP and Cyp2f2 mRNA levels were reduced, correlating with reductions in the number of Clara cells lining the airways. When combined with infection, these markers further declined and an apparent delay in recovery of mRNA expression was observed, suggesting that radiation injury leads to a chronic reduction in the number of Clara cells that may potentiate the epithelial injury observed after influenza A virus infection. This novel finding may have considerable therapeutic implications with respect to both thoracic tumor patients and recipients of bone marrow transplants.
Collapse
Affiliation(s)
- Casey M Manning
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Balli D, Ustiyan V, Zhang Y, Wang IC, Masino AJ, Ren X, Whitsett JA, Kalinichenko VV, Kalin TV. Foxm1 transcription factor is required for lung fibrosis and epithelial-to-mesenchymal transition. EMBO J 2013; 32:231-44. [PMID: 23288041 DOI: 10.1038/emboj.2012.336] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 11/27/2012] [Indexed: 12/18/2022] Open
Abstract
Alveolar epithelial cells (AECs) participate in the pathogenesis of pulmonary fibrosis, producing pro-inflammatory mediators and undergoing epithelial-to-mesenchymal transition (EMT). Herein, we demonstrated the critical role of Forkhead Box M1 (Foxm1) transcription factor in radiation-induced pulmonary fibrosis. Foxm1 was induced in AECs following lung irradiation. Transgenic expression of an activated Foxm1 transcript in AECs enhanced radiation-induced pneumonitis and pulmonary fibrosis, and increased the expression of IL-1β, Ccl2, Cxcl5, Snail1, Zeb1, Zeb2 and Foxf1. Conditional deletion of Foxm1 from respiratory epithelial cells decreased radiation-induced pulmonary fibrosis and prevented the increase in EMT-associated gene expression. siRNA-mediated inhibition of Foxm1 prevented TGF-β-induced EMT in vitro. Foxm1 bound to and increased promoter activity of the Snail1 gene, a critical transcriptional regulator of EMT. Expression of Snail1 restored TGF-β-induced loss of E-cadherin in Foxm1-deficient cells in vitro. Lineage-tracing studies demonstrated that Foxm1 increased EMT during radiation-induced pulmonary fibrosis in vivo. Foxm1 is required for radiation-induced pulmonary fibrosis by enhancing the expression of genes critical for lung inflammation and EMT.
Collapse
Affiliation(s)
- David Balli
- Department of Pediatrics, Division of Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mo Y, Chen J, Schlueter CF, Hoyle GW. Differential susceptibility of inbred mouse strains to chlorine-induced airway fibrosis. Am J Physiol Lung Cell Mol Physiol 2012; 304:L92-102. [PMID: 23171502 DOI: 10.1152/ajplung.00272.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Chlorine is a reactive gas that is considered a chemical threat agent. Humans who develop acute lung injury from chlorine inhalation typically recover normal lung function; however, a subset can experience chronic airway disease. To examine pathological changes following chlorine-induced lung injury, mice were exposed to a single high dose of chlorine, and repair of the lung was analyzed at multiple times after exposure. In FVB/NJ mice, chlorine inhalation caused pronounced fibrosis of larger airways that developed by day 7 after exposure and was associated with airway hyperreactivity. In contrast, A/J mice had little or no airway fibrosis and had normal lung function at day 7. Unexposed FVB/NJ mice had less keratin 5 staining (basal cell marker) than A/J mice in large intrapulmonary airways where epithelial repair was poor and fibrosis developed after chlorine exposure. FVB/NJ mice had large areas devoid of epithelium on day 1 after exposure leading to fibroproliferative lesions on days 4 and 7. A/J mice had airways covered by squamous keratin 5-stained cells on day 1 that transitioned to a highly proliferative reparative epithelium by day 4 followed by the reappearance of ciliated and Clara cells by day 7. The data suggest that lack of basal cells in the large intrapulmonary airways and failure to effect epithelial repair at these sites are factors contributing to the development of airway fibrosis in FVB/NJ mice. The observed differences in susceptibility to chlorine-induced airway disease provide a model in which mechanisms and treatment of airway fibrosis can be investigated.
Collapse
Affiliation(s)
- Yiqun Mo
- Dept. of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 701 HSC-A, 319 Abraham Flexner Way, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
46
|
Vadász I, Brochard L. Update in acute lung injury and mechanical ventilation 2011. Am J Respir Crit Care Med 2012; 186:17-23. [PMID: 22753685 DOI: 10.1164/rccm.201203-0582up] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center, Klinikstrasse 33, Giessen, Germany.
| | | |
Collapse
|
47
|
Luckhardt TR, Müller-Quernheim J, Thannickal VJ. Update in diffuse parenchymal lung disease 2011. Am J Respir Crit Care Med 2012; 186:24-9. [PMID: 22753686 DOI: 10.1164/rccm.201203-0509up] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Tracy R Luckhardt
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, 1900 University Boulevard, Birmingham, AL 35294, USA.
| | | | | |
Collapse
|
48
|
Budd DC, Holmes AM. Targeting TGFβ superfamily ligand accessory proteins as novel therapeutics for chronic lung disorders. Pharmacol Ther 2012; 135:279-91. [PMID: 22722064 DOI: 10.1016/j.pharmthera.2012.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Dysregulation of the transforming growth factor β (TGFβ) pathway has been implicated to underlie a number of disease indications including chronic lung disorders such as asthma, chronic obstructive pulmonary disease (COPD), interstitial pneumonias, and pulmonary arterial hypertension (PAH). Consequently, the pharmaceutical industry has devoted significant resources in the pursuit of TGFβ pathway inhibitors that target the cognate type I and II receptors and respective ligands. The progress of these approaches has been painfully slow, due in part to dose-limiting safety issues that result from the antagonism of a pathway that is responsible for regulating many fundamental biological processes including immune surveillance and cardiovascular responses. These disappointments have led many in the field to conclude that modulating the TGFβ pathway for chronic indications with a sufficient safety window using conventional approaches may be extremely difficult to achieve. Here we review the rationale and limitations of the use of TGFβ pathway inhibitors in chronic lung disorders and the possibility of targeting TGFβ superfamily ligand accessory proteins to allow rheostatic regulation of signaling to achieve efficacy while maintaining a sufficient therapeutic index.
Collapse
Affiliation(s)
- David C Budd
- Respiratory Drug Discovery, Inflammation, Hoffmann-La Roche Inc., Nutley, NJ, USA.
| | | |
Collapse
|
49
|
Sato A, Xu Y, Whitsett JA, Ikegami M. CCAAT/enhancer binding protein-α regulates the protease/antiprotease balance required for bronchiolar epithelium regeneration. Am J Respir Cell Mol Biol 2012; 47:454-63. [PMID: 22652201 DOI: 10.1165/rcmb.2011-0239oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many transcription factors that regulate lung morphogenesis during development are reactivated to mediate repairs of the injured adult lung. We hypothesized that CCAAT/enhancer binding protein-α (C/EBPα), a transcription factor critical for perinatal lung maturation, regulates genes required for the normal repair of the bronchiolar epithelium after injury. Transgenic Cebpα(Δ/Δ) mice, in which Cebpa was conditionally deleted from Clara cells and Type II cells after birth, were used in this study. Airway injury was induced in mice by the intraperitoneal administration of naphthalene to ablate bronchiolar epithelial cells. Although the deletion of C/EBPα did not influence lung structure and function under unstressed conditions, C/EBPα was required for the normal repair of terminal bronchiolar epithelium after naphthalene injury. To identify cellular processes that are influenced by C/EBPα during repair, mRNA microarray was performed on terminal bronchiolar epithelial cells isolated by laser-capture microdissection. Normal repair of the terminal bronchiolar epithelium was highly associated with the mRNAs regulating antiprotease activities, and their induction required C/EBPα. The defective deposition of fibronectin in Cebpα(Δ/Δ) mice was associated with increased protease activity and delayed differentiation of FoxJ1-expressing ciliated cells. The fibronectin and ciliated cells were restored by the intratracheal treatment of Cebpα(Δ/Δ) mice with the serine protease inhibitor. In conclusion, C/EBPα regulates the expression of serine protease inhibitors that are required for the normal increase of fibronectin and the restoration of ciliated cells after injury. Treatment with serine protease inhibitor may aid in the recovery of injured bronchiolar epithelial cells, and prevent common chronic lung diseases.
Collapse
Affiliation(s)
- Atsuyasu Sato
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | |
Collapse
|
50
|
Wansleeben C, Barkauskas CE, Rock JR, Hogan BLM. Stem cells of the adult lung: their development and role in homeostasis, regeneration, and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:131-48. [DOI: 10.1002/wdev.58] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|