1
|
Sahnoon L, Bajbouj K, Mahboub B, Hamoudi R, Hamid Q. Targeting IL-13 and IL-4 in Asthma: Therapeutic Implications on Airway Remodeling in Severe Asthma. Clin Rev Allergy Immunol 2025; 68:44. [PMID: 40257546 PMCID: PMC12011922 DOI: 10.1007/s12016-025-09045-2] [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] [Accepted: 03/19/2025] [Indexed: 04/22/2025]
Abstract
Asthma is a chronic respiratory disorder affecting individuals across all age groups. It is characterized by airway inflammation and remodeling and leads to progressive airflow restriction. While corticosteroids remain a mainstay therapy, their efficacy is limited in severe asthma due to genetic and epigenetic alterations, as well as elevated pro-inflammatory cytokines interleukin-4 (IL-4), interleukin-13 (IL-13), and interleukin-5 (IL-5), which drive structural airway changes including subepithelial fibrosis, smooth muscle hypertrophy, and goblet cell hyperplasia. This underscores the critical need for biologically targeted therapies. This review systematically examines the roles of IL-4 and IL-13, key drivers of type-2 inflammation, in airway remodeling and their potential as therapeutic targets. IL-4 orchestrates eosinophil recruitment, immunoglobulin class switching, and Th2 differentiation, whereas IL-13 directly modulates structural cells, including fibroblasts and epithelial cells, to promote mucus hypersecretion and extracellular matrix (ECM) deposition. Despite shared signaling pathways, IL-13 emerges as the dominant cytokine in remodeling processes including mucus hypersecretion, fibrosis and smooth muscle hypertrophy. While IL-4 primarily amplifies inflammatory cascades by driving IgE switching, promoting Th2 cell polarization that sustain cytokine release, and inducing chemokines to recruit eosinophils. In steroid-resistant severe asthma, biologics targeting IL-4/IL-13 show promise in reducing exacerbations and eosinophilic inflammation. However, their capacity to reverse established remodeling remains inconsistent, as clinical trials prioritize inflammatory biomarkers over long-term structural outcomes. This synthesis highlights critical gaps in understanding the durability of IL-4/IL-13 inhibition on airway structure and advocates for therapies combining biologics with remodeling-specific strategies. Through the integration of mechanistic insights and clinical evidence, this review emphasizes the need for long-term studies utilizing advanced imaging, histopathological techniques, and patient-reported outcomes to evaluate how IL-4/IL-13-targeted therapies alter airway remodeling and symptom burden, thereby informing more effective treatment approaches for severe, steroid-resistant asthma.
Collapse
Affiliation(s)
- Lina Sahnoon
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Bassam Mahboub
- Rashid Hospital, Dubai Health, 4545, Dubai, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Division of Surgery and Interventional Science, University College London, London, UK.
- Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah, United Arab Emirates.
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
2
|
Kumar S, Corkran M, Cheema Y, Scull MA, Duncan GA. AAV-mediated MUC5AC siRNA delivery to prevent mucociliary dysfunction in asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642720. [PMID: 40161599 PMCID: PMC11952410 DOI: 10.1101/2025.03.12.642720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The main structural components of mucus produced in the lung are mucin 5B (MUC5B) and mucin 5AC (MUC5AC) where a relatively higher expression of MUC5B is typical in health. In the lungs of individuals with asthma, there is a shift from MUC5B to MUC5AC as the predominantly secreted mucin which has been shown to impair mucociliary clearance (MCC) and increase mucus plug formation in the airways. Given its role in asthmatic lung disease, MUC5AC represents a potential therapeutic target where a gene delivery approach could be leveraged to modulate its expression. For these purposes, we explored adeno-associated virus serotype 6 (AAV6), as a lung-tropic viral gene vector to target airway epithelial cells and reduce MUC5AC expression via siRNA delivery. We confirmed that AAV6 was able to transduce epithelial cells in the airways of healthy mice with high transgene expression in mucus-secreting goblet cells. Using multiple particle tracking analysis, we observed that AAV6 was capable of penetrating both normal and MUC5AC-enriched mucus barriers. Successful transduction with AAV6 was also achieved in IL-13 stimulated human airway epithelial (HAE) cells differentiated at air-liquid interface (ALI). AAV6 expressing MUC5AC-targeting siRNA was evaluated as a prophylactic treatment in HAE cell cultures before IL-13 challenge. IL-13 stimulated HAE cultures treated with AAV6-MUC5AC siRNA had significantly reduced MUC5AC mRNA and protein expression compared to untreated controls. Mucociliary transport in IL-13 stimulated HAE cultures was also maintained and comparable to healthy controls following AAV6-MUC5AC siRNA treatment. Together, these findings support that AAV6 may be used as an inhaled gene therapy to suppress MUC5AC overexpression and restore normal airway clearance function in asthma.
Collapse
Affiliation(s)
- Sahana Kumar
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
| | - Maria Corkran
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
| | - Yahya Cheema
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Margaret A Scull
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Gregg A Duncan
- Department of Cell Biology & Molecular Genetics, Maryland Pathogen Research Institute (MPRI) University of Maryland, College Park, MD 20742
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| |
Collapse
|
3
|
Zhu X, Cheng F, Duan H, Fu S, Zhao C. Novel insights into the study of goblet cell hypersecretion in allergic rhinitis. Front Immunol 2025; 16:1525928. [PMID: 39958344 PMCID: PMC11825788 DOI: 10.3389/fimmu.2025.1525928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
Goblet cell hypersecretion is a hallmark of airway inflammation and is driven by complex neuroimmune regulation involving submucosal glands and goblet cells. Although studies have focused on mast cell degranulation as a critical driver of nasal secretion, the role of goblet cells in this process is relatively under-researched. In allergic airway inflammation, goblet cells exhibit metaplasia and hypersecretion. However, allergen exposure does not directly trigger goblet cell degranulation, raising questions regarding the underlying mechanisms of these reactions. The activation of enteric neurons promotes goblet cell degranulation by stimulating the calcitonin gene-related peptide (CGRP)-receptor active modification protein-1 (RAMP1) axis. Meanwhile, airway goblet cells express various neuropeptide receptors, and their activation by neuropeptides such as substance P and CGRP induces mucus secretion, exacerbating allergic rhinitis-associated hypersecretion. Thus, although previously less recognised, the neuron-goblet cell signalling axis plays a critical role in allergic rhinitis mucus secretion. This review highlights current research on the neuroimmune mechanisms underlying goblet cell metaplasia and degranulation, focusing on allergic rhinitis, so as to guide clinical treatment strategies.
Collapse
Affiliation(s)
- Xiaojia Zhu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Fengli Cheng
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Hongying Duan
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Sirui Fu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Changqing Zhao
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Poniedziałek B, Rzymski P, Zarębska-Michaluk D, Flisiak R. Viral respiratory infections and air pollution: A review focused on research in Poland. CHEMOSPHERE 2024; 359:142256. [PMID: 38723686 DOI: 10.1016/j.chemosphere.2024.142256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
The COVID-19 pandemic has reinforced an interest in the relationship between air pollution and respiratory viral infections, indicating that their burden can be increased under poor air quality. This paper reviews the pathways through which air pollutants can enhance susceptibility to such infections and aggravate their clinical course and outcome. It also summarizes the research exploring the links between various viral infections and exposure to solid and gaseous pollution in Poland, a region characterized by poor air quality, especially during a heating season. The majority of studies focused on concentrations of particulate matter (PM; 86.7%); the other pollutants, i.e., BaP, benzene, CO, NOx, O3, and SO2, were studied less often and sometimes only in the context of a particular infection type. Most research concerned COVID-19, showing that elevated levels of PM and NO2 correlated with higher morbidity and mortality, while increased PM2.5 and benzo[a]pyrene levels were related to worse clinical course and outcome in hospitalized, regardless of age and dominant SARS-CoV-2 variant. PM10 and PM2.5 levels were also associated with the incidence of influenza-like illness and, along with NO2 concentrations, with a higher rate of children's hospitalizations due to lower respiratory tract RSV infections. Higher levels of air pollutants also increased hospitalization due to bronchitis (PM, NOx, and O3) and emergency department admission due to viral croup (PM10, PM2.5, NOx, CO, and benzene). Although the conducted studies imply only correlations and have other limitations, as discussed in the present paper, it appears that improving air quality through reducing combustion processes in energy production in Poland should be perceived as a part of multilayered protection measures against respiratory viral infections, decreasing the healthcare costs of COVID-19, lower tract RSV infections, influenza, and other respiratory viral diseases prevalent between autumn and early spring, in addition to other health and climate benefits.
Collapse
Affiliation(s)
- Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | | | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
5
|
Shen H, Wei H, Jiang J, Yao H, Jia Y, Shen J, Li Y, Xie Q, Chen X, Xie Y, Dai H. Effects of 101BHG-D01, a novel M receptor antagonism, on allergic rhinitis in animal models and its mechanism. Eur J Pharmacol 2023; 955:175902. [PMID: 37422119 DOI: 10.1016/j.ejphar.2023.175902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Allergic rhinitis (AR) is a nasal mucosal disease with sneezing and nasal itching as the main symptoms. Although AR treatment continues to improve, there remains a lack of effective drugs. There are still controversies regarding whether anticholinergic drugs can effectively and safely relieve the symptoms of AR and reduce inflammation in the nasal mucosa. Here, we synthesized 101BHG-D01, which is a novel anticholinergic drug that mainly targets the M3 receptor and may reduce the adverse effects of other anticholinergic drugs on the heart. We evaluated the effects of 101BHG-D01 on AR and investigated the potential molecular mechanism of anticholinergic therapy for AR. We found that 101BHG-D01 effectively alleviated AR symptoms, reduced the infiltration of inflammatory cells and attenuated the expression of inflammatory factors (IL-4, IL-5, IL-13, etc.) in various AR animal models. In addition, 101BHG-D01 reduced the activation of mast cells and the release of histamine from rat peritoneal mesothelial cells (RPMCs) challenged by IgE. Moreover, 101BHG-D01 reduced the expression of MUC5AC in IL-13-challenged rat nasal epithelial cells (RNECs) and human nasal epithelial cells (HNEpCs). Furthermore, IL-13 stimulation significantly increased JAK1 and STAT6 phosphorylation, which was suppressed by 101BHG-D01. We demonstrated that 101BHG-D01 reduced mucus secretion and inflammatory cell infiltration in the nasal mucosa, which may occur through a reduction in activation of the JAK1-STAT6 signaling pathway, indicating that 101BHG-D01 is a potent and safe anticholinergic therapy for AR.
Collapse
Affiliation(s)
- Huijuan Shen
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wei
- Department of Pulmonology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, China; Department of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Junxia Jiang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongyi Yao
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongliang Jia
- Key Laboratory of Respiratory Drugs Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Key Laboratory of Respiratory Drugs Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanyou Li
- Beijing Showby Pharmaceutical Co., LTD, Beijing, China
| | - Qiangmin Xie
- Department of Pulmonology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, China; Key Laboratory of Respiratory Drugs Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoping Chen
- Beijing Showby Pharmaceutical Co., LTD, Beijing, China.
| | - Yicheng Xie
- Department of Pulmonology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, China.
| | - Haibin Dai
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
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
|
7
|
Boateng E, Kovacevic D, Oldenburg V, Rådinger M, Krauss-Etschmann S. Role of airway epithelial cell miRNAs in asthma. FRONTIERS IN ALLERGY 2022; 3:962693. [PMID: 36203653 PMCID: PMC9530201 DOI: 10.3389/falgy.2022.962693] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/01/2022] [Indexed: 12/07/2022] Open
Abstract
The airway epithelial cells and overlying layer of mucus are the first point of contact for particles entering the lung. The severity of environmental contributions to pulmonary disease initiation, progression, and exacerbation is largely determined by engagement with the airway epithelium. Despite the cellular cross-talk and cargo exchange in the microenvironment, epithelial cells produce miRNAs associated with the regulation of airway features in asthma. In line with this, there is evidence indicating miRNA alterations related to their multifunctional regulation of asthma features in the conducting airways. In this review, we discuss the cellular components and functions of the airway epithelium in asthma, miRNAs derived from epithelial cells in disease pathogenesis, and the cellular exchange of miRNA-bearing cargo in the airways.
Collapse
Affiliation(s)
- Eistine Boateng
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Correspondence: Eistine Boateng
| | - Draginja Kovacevic
- DZL Laboratory for Experimental Microbiome Research, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Vladimira Oldenburg
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Madeleine Rådinger
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Disease, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- DZL Laboratory for Experimental Microbiome Research, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute for Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
8
|
Ehrhardt B, El-Merhie N, Kovacevic D, Schramm J, Bossen J, Roeder T, Krauss-Etschmann S. Airway remodeling: The Drosophila model permits a purely epithelial perspective. FRONTIERS IN ALLERGY 2022; 3:876673. [PMID: 36187164 PMCID: PMC9520053 DOI: 10.3389/falgy.2022.876673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Airway remodeling is an umbrella term for structural changes in the conducting airways that occur in chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). The pathobiology of remodeling involves multiple mesenchymal and lymphoid cell types and finally leads to a variety of hardly reversible changes such as hyperplasia of goblet cells, thickening of the reticular basement membrane, deposition of collagen, peribronchial fibrosis, angiogenesis and hyperplasia of bronchial smooth muscle cells. In order to develop solutions for prevention or innovative therapies, these complex processes must be understood in detail which requires their deconstruction into individual building blocks. In the present manuscript we therefore focus on the role of the airway epithelium and introduce Drosophila melanogaster as a model. The simple architecture of the flies’ airways as well as the lack of adaptive immunity allows to focus exclusively on the importance of the epithelium for the remodeling processes. We will review and discuss genetic and environmentally induced changes in epithelial structures and molecular responses and propose an integrated framework of research for the future.
Collapse
Affiliation(s)
- Birte Ehrhardt
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Natalia El-Merhie
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Draginja Kovacevic
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Juliana Schramm
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Judith Bossen
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Correspondence: Susanne Krauss-Etschmann
| |
Collapse
|
9
|
Calzetta L, Ritondo BL, Zappa MC, Manzetti GM, Perduno A, Shute J, Rogliani P. The impact of long-acting muscarinic antagonists on mucus hypersecretion and cough in chronic obstructive pulmonary disease: a systematic review. Eur Respir Rev 2022; 31:31/164/210196. [PMID: 35508331 DOI: 10.1183/16000617.0196-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/20/2022] [Indexed: 01/18/2023] Open
Abstract
Patients suffering from chronic obstructive pulmonary disease (COPD) clinically manifest airway mucus hypersecretion as sputum expectoration and cough. Evidence accumulated in the past decade has shown that the cholinergic system not only regulates airway smooth muscle contraction but also the activity of inflammatory and airway epithelial cells, including goblet cells, and submucosal gland activity. Long-acting muscarinic antagonists (LAMAs) with the most favourable M3/M2 muscarinic acetylcholine (ACh) receptors residency properties are not only excellent bronchodilators but potentially also mucus-modifying agents, able to positively impact on mucus hypersecretion and cough. The aim of this systematic review was to investigate the impact of LAMAs on mucus hypersecretion and cough in COPD patients. The evidence confirmed that LAMAs, mainly tiotropium and aclidinium, improved sputum production and cough in moderate to severe COPD. Thus, LAMAs not only antagonise the ACh-induced bronchoconstriction of the airways but also appear to limit the production of mucus secreted in response to ACh by airway goblet cells and/or submucosal glands. Further clinical studies are necessary to evaluate the impact of LAMAs exclusively on sputum symptoms and cough as primary end-points and to investigate whether LAMAs have a modulatory action on the rheological properties of mucus.
Collapse
Affiliation(s)
- Luigino Calzetta
- Dept of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Beatrice Ludovica Ritondo
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Gian Marco Manzetti
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Perduno
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Janis Shute
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Paola Rogliani
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
10
|
Lai Y, Fois G, Flores JR, Tuvim MJ, Zhou Q, Yang K, Leitz J, Peters J, Zhang Y, Pfuetzner RA, Esquivies L, Jones P, Frick M, Dickey BF, Brunger AT. Inhibition of calcium-triggered secretion by hydrocarbon-stapled peptides. Nature 2022; 603:949-956. [PMID: 35322233 PMCID: PMC8967716 DOI: 10.1038/s41586-022-04543-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 02/11/2022] [Indexed: 02/06/2023]
Abstract
Membrane fusion triggered by Ca2+ is orchestrated by a conserved set of proteins to mediate synaptic neurotransmitter release, mucin secretion and other regulated exocytic processes1-4. For neurotransmitter release, the Ca2+ sensitivity is introduced by interactions between the Ca2+ sensor synaptotagmin and the SNARE complex5, and sequence conservation and functional studies suggest that this mechanism is also conserved for mucin secretion6. Disruption of Ca2+-triggered membrane fusion by a pharmacological agent would have therapeutic value for mucus hypersecretion as it is the major cause of airway obstruction in the pathophysiology of respiratory viral infection, asthma, chronic obstructive pulmonary disease and cystic fibrosis7-11. Here we designed a hydrocarbon-stapled peptide that specifically disrupts Ca2+-triggered membrane fusion by interfering with the so-called primary interface between the neuronal SNARE complex and the Ca2+-binding C2B domain of synaptotagmin-1. In reconstituted systems with these neuronal synaptic proteins or with their airway homologues syntaxin-3, SNAP-23, VAMP8, synaptotagmin-2, along with Munc13-2 and Munc18-2, the stapled peptide strongly suppressed Ca2+-triggered fusion at physiological Ca2+ concentrations. Conjugation of cell-penetrating peptides to the stapled peptide resulted in efficient delivery into cultured human airway epithelial cells and mouse airway epithelium, where it markedly and specifically reduced stimulated mucin secretion in both systems, and substantially attenuated mucus occlusion of mouse airways. Taken together, peptides that disrupt Ca2+-triggered membrane fusion may enable the therapeutic modulation of mucin secretory pathways.
Collapse
Affiliation(s)
- Ying Lai
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Jose R Flores
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Tuvim
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Department of Cell & Developmental Biology, Vanderbilt Brain Institute, Center for Structural Biology, Vanderbilt University, TN, USA
| | - Kailu Yang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - John Peters
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Yunxiang Zhang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Department of Chemistry, Fudan University, Shanghai, China
| | - Richard A Pfuetzner
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Luis Esquivies
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Philip Jones
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany.
| | - Burton F Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
11
|
Kadur Lakshminarasimha Murthy P, Xi R, Arguijo D, Everitt JI, Kocak DD, Kobayashi Y, Bozec A, Vicent S, Ding S, Crawford GE, Hsu D, Tata PR, Reddy T, Shen X. Epigenetic basis of oncogenic-Kras-mediated epithelial-cellular proliferation and plasticity. Dev Cell 2022; 57:310-328.e9. [PMID: 35134344 PMCID: PMC8938988 DOI: 10.1016/j.devcel.2022.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 09/15/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
Oncogenic Kras induces a hyper-proliferative state that permits cells to progress to neoplasms in diverse epithelial tissues. Depending on the cell of origin, this also involves lineage transformation. Although a multitude of downstream factors have been implicated in these processes, the precise chronology of molecular events controlling them remains elusive. Using mouse models, primary human tissues, and cell lines, we show that, in Kras-mutant alveolar type II cells (AEC2), FOSL1-based AP-1 factor guides the mSWI/SNF complex to increase chromatin accessibility at genomic loci controlling the expression of genes necessary for neoplastic transformation. We identified two orthogonal processes in Kras-mutant distal airway club cells. The first promoted their transdifferentiation into an AEC2-like state through NKX2.1, and the second controlled oncogenic transformation through the AP-1 complex. Our results suggest that neoplasms retain an epigenetic memory of their cell of origin through cell-type-specific transcription factors. Our analysis showed that a cross-tissue-conserved AP-1-dependent chromatin remodeling program regulates carcinogenesis.
Collapse
Affiliation(s)
- Preetish Kadur Lakshminarasimha Murthy
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA. :
| | - Rui Xi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Diana Arguijo
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dewran D Kocak
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Aline Bozec
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Silvestre Vicent
- University of Navarra, Center for Applied Medical Research, Program in Solid Tumors, Pamplona, Spain; University of Navarra, Department of Pathology, Anatomy and Physiology, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Shengli Ding
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Gregory E Crawford
- Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC 27710, USA
| | - David Hsu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Timothy Reddy
- Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Genomics and Computational Biology, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA.
| |
Collapse
|
12
|
Walentek P. Signaling Control of Mucociliary Epithelia: Stem Cells, Cell Fates, and the Plasticity of Cell Identity in Development and Disease. Cells Tissues Organs 2022; 211:736-753. [PMID: 33902038 PMCID: PMC8546001 DOI: 10.1159/000514579] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/19/2021] [Indexed: 01/25/2023] Open
Abstract
Mucociliary epithelia are composed of multiciliated, secretory, and stem cells and line various organs in vertebrates such as the respiratory tract. By means of mucociliary clearance, those epithelia provide a first line of defense against inhaled particles and pathogens. Mucociliary clearance relies on the correct composition of cell types, that is, the proper balance of ciliated and secretory cells. A failure to generate and to maintain correct cell type composition and function results in impaired clearance and high risk to infections, such as in congenital diseases (e.g., ciliopathies) as well as in acquired diseases, including asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). While it remains incompletely resolved how precisely cell types are specified and maintained in development and disease, many studies have revealed important mechanisms regarding the signaling control in mucociliary cell types in various species. Those studies not only provided insights into the signaling contribution to organ development and regeneration but also highlighted the remarkable plasticity of cell identity encountered in mucociliary maintenance, including frequent trans-differentiation events during homeostasis and specifically in disease. This review will summarize major findings and provide perspectives regarding the future of mucociliary research and the treatment of chronic airway diseases associated with tissue remodeling.
Collapse
Affiliation(s)
- Peter Walentek
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
13
|
Johansson K, Woodruff PG, Ansel KM. Regulation of airway immunity by epithelial miRNAs. Immunol Rev 2021; 304:141-153. [PMID: 34549450 PMCID: PMC9135676 DOI: 10.1111/imr.13028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
The airway epithelium is essential to protect the host from inhaled pathogens and particles. It maintains immune homeostasis and mediates tissue repair after injury. Inflammatory diseases of the airways are associated with failure of epithelial functions, including loss of barrier integrity that results in increased tissue permeability and immune activation; excessive mucus secretion and impaired mucociliary clearance that leads to airflow obstruction and microbial overgrowth; and dysregulation of cellular signals that promotes inflammation and alters tissue structure and airway reactivity. MicroRNAs play crucial roles in mounting appropriate cellular responses to environmental stimuli and preventing disease, using a common machinery and mechanism to regulate gene expression in epithelial cells, immune cells of hematopoietic origin, and other cellular components of the airways. Respiratory diseases are accompanied by dramatic changes in epithelial miRNA expression that drive persistent immune dysregulation. In this review, we discuss responses of the epithelium that promote airway immunopathology, with a focus on miRNAs that contribute to the breakdown of essential epithelial functions. We emphasize the emerging role of miRNAs in regulation of epithelial responses in respiratory health and their value as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Prescott G. Woodruff
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
14
|
Hu Y, Cheng X, Qiu Z, Chen X. Identification of Mucus-Associated Molecular Subtypes of Chronic Obstructive Pulmonary Disease: A Latent Profile Analysis Based on MUC5B-Associated Genes. Med Sci Monit 2021; 27:e931222. [PMID: 34389698 PMCID: PMC8372096 DOI: 10.12659/msm.931222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a disease with high heterogeneity, which is a major challenge in clinical individualized treatment. A mucus phenotype is one of the main characteristics of COPD. MATERIAL AND METHODS Gene expression profiles of lung tissue samples were from the Lung Genomics Research Consortium. MUC5B-associated gene signatures were obtained based on a nonlinear feature screening algorithm. These signatures were used to fit a latent profile analysis (LPA) model to identify COPD molecular subtypes and build a subtype classifier to verify the subtypes. Then, we explored the characteristics of cilium assembly and beating signatures, transcriptome features, immune infiltration among the 3 subtypes by xCell, single-sample gene set enrichment analysis, network perturbation amplitude, and weighted gene co-expression network analysis algorithms. An external dataset was used to verify the above COPD subtypes. RESULTS Three subtypes associated with mucus were identified by LPA and verified in an external dataset. Subtype 1 displayed higher T helper type 1 (Th1) and basophil infiltration, higher Th17/regulatory T cells (Tregs) ratio, a higher level of cilium assembly and beating, and lower mast cell and Treg infiltration. The subtypes 2 and 3 demonstrated higher macrophage M2 infiltration in lung tissue, while subtype 3 had higher neutrophil and eosinophil infiltration than subtype 2. CONCLUSIONS Overall, this work identified 3 mucus-associated molecular subtypes related to MUC5B expression, which deepens the understanding of airway mucus secretion in COPD and potentially provides valuable information for precision therapy.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Xiaomeng Cheng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Zhanjun Qiu
- Department of Pulmonary Disease, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Xianhai Chen
- Department of Pulmonary Disease, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|
15
|
Kuchibhotla VNS, Starkey MR, Reid AT, Heijink IH, Nawijn MC, Hansbro PM, Knight DA. Inhibition of β-Catenin/CREB Binding Protein Signaling Attenuates House Dust Mite-Induced Goblet Cell Metaplasia in Mice. Front Physiol 2021; 12:690531. [PMID: 34385933 PMCID: PMC8353457 DOI: 10.3389/fphys.2021.690531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
Abstract
Excessive mucus production is a major feature of allergic asthma. Disruption of epithelial junctions by allergens such as house dust mite (HDM) results in the activation of β-catenin signaling, which has been reported to stimulate goblet cell differentiation. β-catenin interacts with various co-activators including CREB binding protein (CBP) and p300, thereby regulating the expression of genes involved in cell proliferation and differentiation, respectively. We specifically investigated the role of the β-catenin/CBP signaling pathway in goblet cell metaplasia in a HDM-induced allergic airway disease model in mice using ICG-001, a small molecule inhibitor that blocks the binding of CBP to β-catenin. Female 6- 8-week-old BALB/c mice were sensitized to HDM/saline on days 0, 1, and 2, followed by intranasal challenge with HDM/saline with or without subcutaneous ICG-001/vehicle treatment from days 14 to 17, and samples harvested 24 h after the last challenge/treatment. Differential inflammatory cells in bronchoalveolar lavage (BAL) fluid were enumerated. Alcian blue (AB)/Periodic acid–Schiff (PAS) staining was used to identify goblet cells/mucus production, and airway hyperresponsiveness (AHR) was assessed using invasive plethysmography. Exposure to HDM induced airway inflammation, goblet cell metaplasia and increased AHR, with increased airway resistance in response to the non-specific spasmogen methacholine. Inhibition of the β-catenin/CBP pathway using treatment with ICG-001 significantly attenuated the HDM-induced goblet cell metaplasia and infiltration of macrophages, but had no effect on eosinophils, neutrophils, lymphocytes or AHR. Increased β-catenin/CBP signaling may promote HDM-induced goblet cell metaplasia in mice.
Collapse
Affiliation(s)
- Virinchi N S Kuchibhotla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Malcolm R Starkey
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre GrowUpWell and Hunter Medical Research Institute, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Andrew T Reid
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Irene H Heijink
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands.,Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martijn C Nawijn
- GRIAC Research Institute, University Medical Center Groningen, Groningen, Netherlands.,Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research, University of Groningen, Groningen, Netherlands
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Providence Health Care Research Institute, Vancouver, BC, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Bonser LR, Koh KD, Johansson K, Choksi SP, Cheng D, Liu L, Sun DI, Zlock LT, Eckalbar WL, Finkbeiner WE, Erle DJ. Flow-Cytometric Analysis and Purification of Airway Epithelial-Cell Subsets. Am J Respir Cell Mol Biol 2021; 64:308-317. [PMID: 33196316 PMCID: PMC7909335 DOI: 10.1165/rcmb.2020-0149ma] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
The human airway epithelium is essential in homeostasis, and epithelial dysfunction contributes to chronic airway disease. Development of flow-cytometric methods to characterize subsets of airway epithelial cells will enable further dissection of airway epithelial biology. Leveraging single-cell RNA-sequencing data in combination with known cell type-specific markers, we developed panels of antibodies to characterize and isolate the major airway epithelial subsets (basal, ciliated, and secretory cells) from human bronchial epithelial-cell cultures. We also identified molecularly distinct subpopulations of secretory cells and demonstrated cell subset-specific expression of low-abundance transcripts and microRNAs that are challenging to analyze with current single-cell RNA-sequencing methods. These new tools will be valuable for analyzing and separating airway epithelial subsets and interrogating airway epithelial biology.
Collapse
Affiliation(s)
| | - Kyung Duk Koh
- Lung Biology Center
- Cardiovascular Research Institute
| | - Kristina Johansson
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy
- Sandler Asthma Basic Research Center
| | | | - Dan Cheng
- Lung Biology Center
- Cardiovascular Research Institute
- Department of Respiratory and Critical Care Medicine, Renmin Hospital, Wuhan University, Wuhan, China
| | - Leqian Liu
- Department of Bioengineering and Therapeutic Sciences
| | | | | | - Walter L. Eckalbar
- Lung Biology Center
- University of California, San Francisco CoLabs, University of California San Francisco, San Francisco, California; and
| | | | - David J. Erle
- Lung Biology Center
- Cardiovascular Research Institute
- University of California, San Francisco CoLabs, University of California San Francisco, San Francisco, California; and
| |
Collapse
|
17
|
Czekala L, Wieczorek R, Simms L, Yu F, Budde J, Trelles Sticken E, Rudd K, Verron T, Brinster O, Stevenson M, Walele T. Multi-endpoint analysis of human 3D airway epithelium following repeated exposure to whole electronic vapor product aerosol or cigarette smoke. Curr Res Toxicol 2021; 2:99-115. [PMID: 34345855 PMCID: PMC8320624 DOI: 10.1016/j.crtox.2021.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/15/2022] Open
Abstract
Smoking is a cause of serious diseases in smokers including chronic respiratory diseases. This study aimed to evaluate the tobacco harm reduction (THR) potential of an electronic vapor product (EVP, myblu™) compared to a Kentucky Reference Cigarette (3R4F), and assessed endpoints related to chronic respiratory diseases. Endpoints included: cytotoxicity, barrier integrity (TEER), cilia function, immunohistochemistry, and pro-inflammatory markers. In order to more closely represent the user exposure scenario, we have employed the in vitro 3D organotypic model of human airway epithelium (MucilAir™, Epithelix) for respiratory assessment. The model was repeatedly exposed to either whole aerosol of the EVP, or whole 3R4F smoke, at the air liquid interface (ALI), for 4 weeks to either 30, 60 or 90 puffs on 3-exposure-per-week basis. 3R4F smoke generation used the ISO 20778:2018 regime and EVP aerosol used the ISO 20768:2018 vaping regime. Exposure to undiluted whole EVP aerosol did not trigger any significant changes in the level of pro-inflammatory mediators, cilia beating function, barrier integrity and cytotoxicity when compared with air controls. In contrast, exposure to diluted (1:17) whole cigarette smoke caused significant changes to all the endpoints mentioned above. To our knowledge, this is the first study evaluating the effects of repeated whole cigarette smoke and whole EVP aerosol exposure to a 3D lung model at the ALI. Our results add to the growing body of scientific literature supporting the THR potential of EVPs relative to combustible cigarettes and the applicability of the 3D lung models in human-relevant product risk assessments.
Collapse
Key Words
- 2D, Two Dimensional
- 3D, Three Dimensional
- 3R4F, Scientific Reference Tobacco Cigarette (University of Kentucky)
- ALI, Air-Liquid Interface
- ANOVA, Analysis of Variance
- AOP, Adverse Outcome Pathway
- CAA, Cilia Active Area
- CBF, Cilia Beat Frequency
- COPD, Chronic Obstructive Pulmonary Disease
- CYP450, Cytochrome P450
- Cigarette
- Cilia
- DPBS, Dulbecco's phosphate-buffered saline containing Ca2+ and Mg2+
- EGFR, Epidermal Growth Factor Receptor
- EVP, Electronic Vapor Product
- Electronic vapor product
- FOX-J1, Forkhead Box J1 protein
- H&E, Hematoxylin and Eosin
- IIVS, Institute for In Vitro Sciences
- IL-13, Interleukin 13
- IL-1β, Interleukin 1 Beta
- IL-6, Interleukin-6
- IL-8, Interleukin-8
- ISO, International Organization for Standardization
- Immunohistochemistry
- KERs, Key Event Relationships
- KEs, Key Events
- LDH, Lactate Dehydrogenase
- MIE, Molecular Initiating Event
- MMP-1, Matrix Metalloproteinase-1
- MMP-3, Matrix Metalloproteinase-3
- MMP-9, Matrix Metalloproteinase-9
- MUC5AC, Mucin 5AC Protein
- MWP, Multi-Well Plate
- NKT, Natural Killer T Cells
- Organotypic tissue model
- PBS, Phosphate Buffered Saline
- PMN, polymorphonuclear
- Pro-inflammatory markers
- SAEIVS, Smoke Aerosol Exposure In Vitro System
- TEER, Transepithelial Electrical Resistance
- THR, Tobacco Harm Reduction
- TNF-α, Tumor Necrosis Factor Alpha
- TPM, Total Particulate Matter
Collapse
Affiliation(s)
- Lukasz Czekala
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Roman Wieczorek
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Liam Simms
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Fan Yu
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Jessica Budde
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Edgar Trelles Sticken
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Kathryn Rudd
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Thomas Verron
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Oleg Brinster
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Matthew Stevenson
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| | - Tanvir Walele
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL, United Kingdom
| |
Collapse
|
18
|
Walentek P. Xenopus epidermal and endodermal epithelia as models for mucociliary epithelial evolution, disease, and metaplasia. Genesis 2021; 59:e23406. [PMID: 33400364 DOI: 10.1002/dvg.23406] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/08/2022]
Abstract
The Xenopus embryonic epidermis is a powerful model to study mucociliary biology, development, and disease. Particularly, the Xenopus system is being used to elucidate signaling pathways, transcription factor functions, and morphogenetic mechanisms regulating cell fate specification, differentiation and cell function. Thereby, Xenopus research has provided significant insights into potential underlying molecular mechanisms for ciliopathies and chronic airway diseases. Recent studies have also established the embryonic epidermis as a model for mucociliary epithelial remodeling, multiciliated cell trans-differentiation, cilia loss, and mucus secretion. Additionally, the tadpole foregut epithelium is lined by a mucociliary epithelium, which shows remarkable features resembling mammalian airway epithelia, including its endodermal origin and a variable cell type composition along the proximal-distal axis. This review aims to summarize the advantages of the Xenopus epidermis for mucociliary epithelial biology and disease modeling. Furthermore, the potential of the foregut epithelium as novel mucociliary model system is being highlighted. Additional perspectives are presented on how to expand the range of diseases that can be modeled in the frog system, including proton pump inhibitor-associated pneumonia as well as metaplasia in epithelial cells of the airway and the gastroesophageal region.
Collapse
Affiliation(s)
- Peter Walentek
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
19
|
Do AN, Chun Y, Grishina G, Grishin A, Rogers AJ, Raby BA, Weiss ST, Vicencio A, Schadt EE, Bunyavanich S. Network study of nasal transcriptome profiles reveals master regulator genes of asthma. J Allergy Clin Immunol 2020; 147:879-893. [PMID: 32828590 DOI: 10.1016/j.jaci.2020.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nasal transcriptomics can provide an accessible window into asthma pathobiology. OBJECTIVE Our goal was to move beyond gene signatures of asthma to identify master regulator genes that causally regulate genes associated with asthma phenotypes. METHODS We recruited 156 children with severe persistent asthma and controls for nasal transcriptome profiling and applied network-based and probabilistic causal methods to identify severe asthma genes and their master regulators. We then took the same approach in an independent cohort of 190 adults with mild/moderate asthma and controls to identify mild/moderate asthma genes and their master regulators. Comparative analysis of the master regulator genes followed by validation testing in independent children with severe asthma (n = 21) and mild/moderate asthma (n = 154) was then performed. RESULTS Nasal gene signatures for severe persistent asthma and for mild/moderate persistent asthma were identified; both were found to be enriched in coexpression network modules for ciliary function and inflammatory response. By applying probabilistic causal methods to these gene signatures and validation testing in independent cohorts, we identified (1) a master regulator gene common to asthma across severity and ages (FOXJ1); (2) master regulator genes of severe persistent asthma in children (LRRC23, TMEM231, CAPS, PTPRC, and FYB); and (3) master regulator genes of mild/moderate persistent asthma in children and adults (C1orf38 and FMNL1). The identified master regulators were statistically inferred to causally regulate the expression of downstream genes that modulate ciliary function and inflammatory response to influence asthma. CONCLUSION The identified master regulator genes of asthma provide a novel path forward to further uncovering asthma mechanisms and therapy.
Collapse
Affiliation(s)
- Anh N Do
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yoojin Chun
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Galina Grishina
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander Grishin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Angela J Rogers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Benjamin A Raby
- Division of Pulmonary Medicine, Children's Hospital Boston, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass
| | - Alfin Vicencio
- Division of Pulmonary Medicine, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric E Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
20
|
Heijink IH, Kuchibhotla VNS, Roffel MP, Maes T, Knight DA, Sayers I, Nawijn MC. Epithelial cell dysfunction, a major driver of asthma development. Allergy 2020; 75:1902-1917. [PMID: 32460363 PMCID: PMC7496351 DOI: 10.1111/all.14421] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022]
Abstract
Airway epithelial barrier dysfunction is frequently observed in asthma and may have important implications. The physical barrier function of the airway epithelium is tightly interwoven with its immunomodulatory actions, while abnormal epithelial repair responses may contribute to remodelling of the airway wall. We propose that abnormalities in the airway epithelial barrier play a crucial role in the sensitization to allergens and pathogenesis of asthma. Many of the identified susceptibility genes for asthma are expressed in the airway epithelium, supporting the notion that events at the airway epithelial surface are critical for the development of the disease. However, the exact mechanisms by which the expression of epithelial susceptibility genes translates into a functionally altered response to environmental risk factors of asthma are still unknown. Interactions between genetic factors and epigenetic regulatory mechanisms may be crucial for asthma susceptibility. Understanding these mechanisms may lead to identification of novel targets for asthma intervention by targeting the airway epithelium. Moreover, exciting new insights have come from recent studies using single‐cell RNA sequencing (scRNA‐Seq) to study the airway epithelium in asthma. This review focuses on the role of airway epithelial barrier function in the susceptibility to develop asthma and novel insights in the modulation of epithelial cell dysfunction in asthma.
Collapse
Affiliation(s)
- Irene H. Heijink
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Pulmonology University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Virinchi N. S. Kuchibhotla
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan NSW Australia
| | - Mirjam P. Roffel
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent University Ghent Belgium
| | - Tania Maes
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent University Ghent Belgium
| | - Darryl A. Knight
- School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan NSW Australia
- UBC Providence Health Care Research Institute Vancouver BC Canada
- Department of Anesthesiology, Pharmacology and Therapeutics University of British Columbia Vancouver BC Canada
| | - Ian Sayers
- Division of Respiratory Medicine National Institute for Health Research Nottingham Biomedical Research Centre University of Nottingham Biodiscovery Institute University of Nottingham Nottingham UK
| | - Martijn C. Nawijn
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| |
Collapse
|
21
|
Jackson ND, Everman JL, Chioccioli M, Feriani L, Goldfarbmuren KC, Sajuthi SP, Rios CL, Powell R, Armstrong M, Gomez J, Michel C, Eng C, Oh SS, Rodriguez-Santana J, Cicuta P, Reisdorph N, Burchard EG, Seibold MA. Single-Cell and Population Transcriptomics Reveal Pan-epithelial Remodeling in Type 2-High Asthma. Cell Rep 2020; 32:107872. [PMID: 32640237 PMCID: PMC8046336 DOI: 10.1016/j.celrep.2020.107872] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
The type 2 cytokine-high asthma endotype (T2H) is characterized by IL-13-driven mucus obstruction of the airways. To further investigate this incompletely understood pathobiology, we characterize IL-13 effects on human airway epithelial cell cultures using single-cell RNA sequencing, finding that IL-13 generates a distinctive transcriptional state for each cell type. Specifically, we discover a mucus secretory program induced by IL-13 in all cell types which converts both mucus and defense secretory cells into a metaplastic state with emergent mucin production and secretion, while leading to ER stress and cell death in ciliated cells. The IL-13-remodeled epithelium secretes a pathologic, mucin-imbalanced, and innate immunity-depleted proteome that arrests mucociliary motion. Signatures of IL-13-induced cellular remodeling are mirrored by transcriptional signatures characteristic of the nasal airway epithelium within T2H versus T2-low asthmatic children. Our results reveal the epithelium-wide scope of T2H asthma and present candidate therapeutic targets for restoring normal epithelial function.
Collapse
Affiliation(s)
- Nathan D Jackson
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA
| | - Jamie L Everman
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA
| | | | - Luigi Feriani
- Department of Physics, University of Cambridge, Cambridge, CB2 3AX, UK
| | | | - Satria P Sajuthi
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA
| | - Cydney L Rios
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA
| | - Roger Powell
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Michael Armstrong
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Joe Gomez
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Cole Michel
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Celeste Eng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sam S Oh
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Pietro Cicuta
- Department of Physics, University of Cambridge, Cambridge, CB2 3AX, UK
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado-AMC, Aurora, CO 80045, USA
| | - Esteban G Burchard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Max A Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA; Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-AMC, Aurora, CO 80045, USA.
| |
Collapse
|
22
|
Wang Y, Ninaber DK, van Schadewijk A, Hiemstra PS. Tiotropium and Fluticasone Inhibit Rhinovirus-Induced Mucin Production via Multiple Mechanisms in Differentiated Airway Epithelial Cells. Front Cell Infect Microbiol 2020; 10:278. [PMID: 32637364 PMCID: PMC7318795 DOI: 10.3389/fcimb.2020.00278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/12/2020] [Indexed: 01/16/2023] Open
Abstract
Human rhinoviruses (HRVs) are associated with acute exacerbations in patients with chronic obstructive pulmonary disease (COPD) and asthma, which are accompanied by mucus hypersecretion. Whereas, various studies have shown that HRVs increase epithelial mucin production and thus may directly contribute to mucus hypersecretion. The effects of drugs used in the treatment of COPD and asthma on HRV-induced mucin production in epithelial cell cultures have not been studied. In the present study, we assessed effects of HRVs on mucin production and secretion in well-differentiated primary human bronchial epithelial cells (PBEC) and studied the effect of the inhaled corticosteroid fluticasone propionate and the long-acting muscarinic antagonist tiotropium bromide on this process. Differentiated PBEC that were cultured at the air-liquid interface (ALI-PBEC) were infected with HRV-A16 and HRV-1B. Quantitative PCR, immunofluorescence staining, ELISA, periodic acid-Schiff (PAS) staining and immunostaining assays were used to assess the effects of HRV infection. Here we demonstrate that both HRV-A16 and HRV-1B increased mucin (MUC5AC and MUC5B) gene expression and protein release. When exploring this in more detail in HRV-A16-infected epithelial cells, mucin expression was found to be accompanied by increases in expression of SAM-pointed domain-containing Ets-like factor (SPDEF) and SPDEF-regulated genes known to be involved in the regulation of mucin production. We also found that pre-treatment with the purinergic P2R antagonist suramin inhibits HRV-enhanced MUC5AC expression and protein release, implicating involvement of purinergic signaling by extracellular ATP. We furthermore found that both fluticasone and tiotropium decreased HRV-induced mucin production without affecting viral replication, and obtained evidence to suggest that the inhibitory effect of fluticasone involved modulation of SPDEF-regulated genes and extracellular ATP release. These data show that both tiotropium and fluticasone inhibit HRV-induced epithelial mucin production independent of viral clearance, and thus provide insight into the mechanisms underlying beneficial effects of tiotropium and fluticasone in the treatment of COPD, asthma and accompanying exacerbations in these patients. Furthermore, our findings provide additional insight into the mechanisms by which HRV increases epithelial mucin production.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Dennis K Ninaber
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
23
|
He M, Wu B, Ye W, Le DD, Sinclair AW, Padovano V, Chen Y, Li KX, Sit R, Tan M, Caplan MJ, Neff N, Jan YN, Darmanis S, Jan LY. Chloride channels regulate differentiation and barrier functions of the mammalian airway. eLife 2020; 9:e53085. [PMID: 32286221 PMCID: PMC7182432 DOI: 10.7554/elife.53085] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/13/2020] [Indexed: 12/16/2022] Open
Abstract
The conducting airway forms a protective mucosal barrier and is the primary target of airway disorders. The molecular events required for the formation and function of the airway mucosal barrier, as well as the mechanisms by which barrier dysfunction leads to early onset airway diseases, remain unclear. In this study, we systematically characterized the developmental landscape of the mouse airway using single-cell RNA sequencing and identified remarkably conserved cellular programs operating during human fetal development. We demonstrated that in mouse, genetic inactivation of chloride channel Ano1/Tmem16a compromises airway barrier function, results in early signs of inflammation, and alters the airway cellular landscape by depleting epithelial progenitors. Mouse Ano1-/-mutants exhibited mucus obstruction and abnormal mucociliary clearance that resemble the airway defects associated with cystic fibrosis. The data reveal critical and non-redundant roles for Ano1 in organogenesis, and show that chloride channels are essential for mammalian airway formation and function.
Collapse
Affiliation(s)
- Mu He
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Bing Wu
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Wenlei Ye
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Daniel D Le
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Adriane W Sinclair
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
- Division of Pediatric Urology, University of California, San Francisco, Benioff Children's HospitalSan FranciscoUnited States
| | - Valeria Padovano
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HeavenUnited States
| | - Yuzhang Chen
- Department of Anesthesia and Perioperative Care, University of California, San FranciscoSan FranciscoUnited States
| | - Ke-Xin Li
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
| | - Rene Sit
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Michelle Tan
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of MedicineNew HeavenUnited States
| | - Norma Neff
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Yuh Nung Jan
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
| | | | - Lily Yeh Jan
- Department of Physiology, University of California, San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical Institute, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
24
|
Cao Y, Chen M, Dong D, Xie S, Liu M. Environmental pollutants damage airway epithelial cell cilia: Implications for the prevention of obstructive lung diseases. Thorac Cancer 2020; 11:505-510. [PMID: 31975505 PMCID: PMC7049516 DOI: 10.1111/1759-7714.13323] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 01/11/2023] Open
Abstract
Mucociliary epithelium lining the upper and lower respiratory tract constitutes the first line of defense of the airway and lungs against inhaled pollutants and pathogens. The concerted beating of multiciliated cells drives mucociliary clearance. Abnormalities in both the structure and function of airway cilia have been implicated in obstructive lung diseases. Emerging evidence reveals a close correlation between lung diseases and environmental stimuli such as sulfur dioxide and tobacco particles. However, the underlying mechanism remains to be described. In this review, we emphasize the importance of airway cilia in mucociliary clearance and discuss how environmental pollutants affect the structure and function of airway cilia, thus shedding light on the function of airway cilia in preventing obstructive lung diseases and revealing the negative effects of environmental pollutants on human health.
Collapse
Affiliation(s)
- Yu Cao
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Miao Chen
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Dan Dong
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
25
|
Potaczek DP, Miethe S, Schindler V, Alhamdan F, Garn H. Role of airway epithelial cells in the development of different asthma phenotypes. Cell Signal 2020; 69:109523. [PMID: 31904412 DOI: 10.1016/j.cellsig.2019.109523] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 01/06/2023]
Abstract
The term (bronchial) asthma describes a disorder syndrome that comprises several disease phenotypes, all characterized by chronic inflammation in the bronchial epithelium, with a variety of subsequent functional consequences. Thus, the epithelium in the conducting airways is the main localization of the complex pathological changes in the disease. In this regard, bronchial epithelial cells are not passively affected by inflammatory mechanisms induced by immunological processes but rather actively involved in all steps of disease development from initiation and perpetuation to chronification. In recent years it turned out that bronchial epithelial cells show a high level of structural and functional diversity and plasticity with epigenetic mechanisms playing a crucial role in the regulation of these processes. Thus, it is quite reasonable that differential functional activities of the bronchial epithelium are involved in the development of different asthma phenotypes and/or stages of disease. The current knowledge on this topic will be discussed in this review article.
Collapse
Affiliation(s)
- Daniel P Potaczek
- Institute of Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics, Philipps University of Marburg - Medical Faculty, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Marburg, Germany; John Paul II Hospital, Krakow, Poland
| | - Sarah Miethe
- Institute of Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics, Philipps University of Marburg - Medical Faculty, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Marburg, Germany
| | - Viktoria Schindler
- Institute of Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics, Philipps University of Marburg - Medical Faculty, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Marburg, Germany
| | - Fahd Alhamdan
- Institute of Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics, Philipps University of Marburg - Medical Faculty, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics, Philipps University of Marburg - Medical Faculty, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Marburg, Germany.
| |
Collapse
|
26
|
Interleukin-13 Stimulation Reveals the Cellular and Functional Plasticity of the Airway Epithelium. Ann Am Thorac Soc 2019; 15:S98-S102. [PMID: 29676620 DOI: 10.1513/annalsats.201711-868mg] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
About 50% of patients with asthma exhibit chronic airway inflammation driven by the type 2 cytokines interleukin (IL)-4, IL-5, and IL-13. These patients with type 2-high asthma experience more allergic symptoms, greater airway hyperresponsiveness, and more severe mucus obstruction than patients with type 2-low asthma. Mouse models of asthma have shown that much of the airway dysfunction in these models can be generated by IL-13 stimulation of the airway epithelium alone. Both in vivo mouse model studies and in vitro studies of human mucociliary airway epithelial cultures have shown that IL-13 induces cellular remodeling of the airway epithelium through proliferation-independent transdifferentiation processes. In both humans and mice, IL-13 stimulation of the airway epithelium results in generation of hypersecretory mucin 5AC (MUC5AC)-expressing mucus cells. Whereas club cells have been shown to be the source of these mucin 5AC-positive mucus cells in mice, the origin of these mucus cells in humans is unclear. In humans, chronic IL-13 stimulation appears to result in loss of ciliated cells. Moreover, IL-13 stimulation can block ciliated cell differentiation from human basal airway epithelial cells. Coincident with IL-13 cellular remodeling are reported decreases in mucociliary transport and ciliary beat frequency. These IL-13-mediated changes in mucociliary function are accompanied by disorganization of cilia, a decrease in the ratio of mucin 5B (MUC5B) to mucin 5AC, and mucus gel tethering to the epithelial surface by mucin 5AC. These airway epithelial responses to IL-13 are mediated by multiple transcription factors, including signal transducer and activator of transcription-6 (STAT6), SAM pointed domain-containing Ets transcription factor (SPDEF), Forkhead box A2 (FOXA2), and Forkhead box J1 (FOXJ1). In addition, analysis of RNA-sequencing data derived from airway epithelial cells shows how IL-13 stimulation promotes broad changes in gene expression across the transcriptome. These results reveal the plastic nature of airway epithelial cells that enables the epithelium to undergo remodeling and functional shifts in response to IL-13 stimulation. With use of new technology, future studies should lead to greater understanding of how IL-13 and other stimuli of disease bring about airway epithelial remodeling, which may aid in the development of therapies that ameliorate airway dysfunction in asthma and other diseases.
Collapse
|
27
|
Ruiz García S, Deprez M, Lebrigand K, Cavard A, Paquet A, Arguel MJ, Magnone V, Truchi M, Caballero I, Leroy S, Marquette CH, Marcet B, Barbry P, Zaragosi LE. Novel dynamics of human mucociliary differentiation revealed by single-cell RNA sequencing of nasal epithelial cultures. Development 2019; 146:dev.177428. [PMID: 31558434 PMCID: PMC6826037 DOI: 10.1242/dev.177428] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022]
Abstract
The upper airway epithelium, which is mainly composed of multiciliated, goblet, club and basal cells, ensures proper mucociliary function and can regenerate in response to assaults. In chronic airway diseases, defective repair leads to tissue remodeling. Delineating key drivers of differentiation dynamics can help understand how normal or pathological regeneration occurs. Using single-cell transcriptomics and lineage inference, we have unraveled trajectories from basal to luminal cells, providing novel markers for specific populations. We report that: (1) a precursor subgroup of multiciliated cells, which we have entitled deuterosomal cells, is defined by specific markers, such as DEUP1, FOXN4, YPEL1, HES6 and CDC20B; (2) goblet cells can be precursors of multiciliated cells, thus explaining the presence of hybrid cells that co-express markers of goblet and multiciliated cells; and (3) a repertoire of molecules involved in the regeneration process, such as keratins or components of the Notch, Wnt or BMP/TGFβ pathways, can be identified. Confirmation of our results on fresh human and pig airway samples, and on mouse tracheal cells, extend and confirm our conclusions regarding the molecular and cellular choreography at work during mucociliary epithelial differentiation.
Collapse
Affiliation(s)
| | - Marie Deprez
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Kevin Lebrigand
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Amélie Cavard
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Agnès Paquet
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | | | - Virginie Magnone
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Marin Truchi
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | | | - Sylvie Leroy
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France.,Université Côte d'Azur, CHU de Nice, Pulmonology Department, Nice 06000, France
| | | | - Brice Marcet
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, IPMC, Sophia-Antipolis 06560, France
| | | |
Collapse
|
28
|
Ladjemi MZ, Gras D, Dupasquier S, Detry B, Lecocq M, Garulli C, Fregimilicka C, Bouzin C, Gohy S, Chanez P, Pilette C. Bronchial Epithelial IgA Secretion Is Impaired in Asthma. Role of IL-4/IL-13. Am J Respir Crit Care Med 2019; 197:1396-1409. [PMID: 29652177 DOI: 10.1164/rccm.201703-0561oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
RATIONALE Asthma is associated with increased lung IgE production, but whether the secretory IgA system is affected in this disease remains unknown. OBJECTIVES We explored mucosal IgA transport in human asthma and its potential regulation by T-helper cell type 2 inflammation. METHODS Bronchial biopsies from asthma and control subjects were assayed for bronchial epithelial polymeric immunoglobulin receptor (pIgR) expression and correlated to T-helper cell type 2 biomarkers. Bronchial epithelium reconstituted in vitro from these subjects, on culture in air-liquid interface, was assayed for pIgR expression and regulation by IL-4/IL-13. MEASUREMENTS AND MAIN RESULTS Downregulation of pIgR protein was observed in the bronchial epithelium from patients with asthma (P = 0.0002 vs. control subjects). This epithelial defect was not observed ex vivo in the cultured epithelium from patients with asthma. Exogenous IL-13 and IL-4 could inhibit pIgR expression and IgA transcytosis. Mechanistic experiments showed that autocrine transforming growth factor-β mediates the IL-4/IL-13 effect on the pIgR, with a partial contribution of upregulated transforming growth factor-α/epidermal growth factor receptor. CONCLUSIONS This study shows impaired bronchial epithelial pIgR expression in asthma, presumably affecting secretory IgA-mediated frontline defense as a result of type 2 immune activation of the transforming growth factor pathway.
Collapse
Affiliation(s)
- Maha Zohra Ladjemi
- 1 Pôle de Pneumologie, ORL, et Dermatologie and.,2 Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium
| | - Delphine Gras
- 3 INSERM U 1067, CNRS UMR 7333, Université Aix-Marseille, Marseille, France
| | | | - Bruno Detry
- 1 Pôle de Pneumologie, ORL, et Dermatologie and.,2 Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium
| | - Marylène Lecocq
- 1 Pôle de Pneumologie, ORL, et Dermatologie and.,4 Service de Pneumologie, Cliniques universitaires Saint-Luc, Brussels, Belgium; and
| | - Céline Garulli
- 3 INSERM U 1067, CNRS UMR 7333, Université Aix-Marseille, Marseille, France
| | - Chantal Fregimilicka
- 5 Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Caroline Bouzin
- 5 Imaging Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Sophie Gohy
- 1 Pôle de Pneumologie, ORL, et Dermatologie and.,4 Service de Pneumologie, Cliniques universitaires Saint-Luc, Brussels, Belgium; and
| | - Pascal Chanez
- 3 INSERM U 1067, CNRS UMR 7333, Université Aix-Marseille, Marseille, France.,6 Clinique des bronches, de l'allergie et du sommeil, Hôpital Nord, Assistance Publique Hôpitaux de Marseille (APHM), Marseille, France
| | - Charles Pilette
- 1 Pôle de Pneumologie, ORL, et Dermatologie and.,2 Institute for Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium.,4 Service de Pneumologie, Cliniques universitaires Saint-Luc, Brussels, Belgium; and
| |
Collapse
|
29
|
Interleukin-Mediated Pendrin Transcriptional Regulation in Airway and Esophageal Epithelia. Int J Mol Sci 2019; 20:ijms20030731. [PMID: 30744098 PMCID: PMC6386862 DOI: 10.3390/ijms20030731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Pendrin (SLC26A4), a Cl−/anion exchanger, is expressed at high levels in kidney, thyroid, and inner ear epithelia, where it has an essential role in bicarbonate secretion/chloride reabsorption, iodide accumulation, and endolymph ion balance, respectively. Pendrin is expressed at lower levels in other tissues, such as airways and esophageal epithelia, where it is transcriptionally regulated by the inflammatory cytokines interleukin (IL)-4 and IL-13 through a signal transducer and activator of transcription 6 (STAT6)-mediated pathway. In the airway epithelium, increased pendrin expression during inflammatory diseases leads to imbalances in airway surface liquid thickness and mucin release, while, in the esophageal epithelium, dysregulated pendrin expression is supposed to impact the intracellular pH regulation system. In this review, we discuss some of the recent findings on interleukin-mediated transcriptional regulation of pendrin and how this dysregulation impacts airway and esophagus epithelial homeostasis during inflammatory diseases.
Collapse
|
30
|
Pezzulo AA, Tudas RA, Stewart CG, Buonfiglio LGV, Lindsay BD, Taft PJ, Gansemer ND, Zabner J. HSP90 inhibitor geldanamycin reverts IL-13- and IL-17-induced airway goblet cell metaplasia. J Clin Invest 2019; 129:744-758. [PMID: 30640172 PMCID: PMC6355221 DOI: 10.1172/jci123524] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/20/2018] [Indexed: 12/29/2022] Open
Abstract
Goblet cell metaplasia, a disabling hallmark of chronic lung disease, lacks curative treatments at present. To identify novel therapeutic targets for goblet cell metaplasia, we studied the transcriptional response profile of IL-13-exposed primary human airway epithelia in vitro and asthmatic airway epithelia in vivo. A perturbation-response profile connectivity approach identified geldanamycin, an inhibitor of heat shock protein 90 (HSP90) as a candidate therapeutic target. Our experiments confirmed that geldanamycin and other HSP90 inhibitors prevented IL-13-induced goblet cell metaplasia in vitro and in vivo. Geldanamycin also reverted established goblet cell metaplasia. Geldanamycin did not induce goblet cell death, nor did it solely block mucin synthesis or IL-13 receptor-proximal signaling. Geldanamycin affected the transcriptome of airway cells when exposed to IL-13, but not when exposed to vehicle. We hypothesized that the mechanism of action probably involves TGF-β, ERBB, or EHF, which would predict that geldanamycin would also revert IL-17-induced goblet cell metaplasia, a prediction confirmed by our experiments. Our findings suggest that persistent airway goblet cell metaplasia requires HSP90 activity and that HSP90 inhibitors will revert goblet cell metaplasia, despite active upstream inflammatory signaling. Moreover, HSP90 inhibitors may be a therapeutic option for airway diseases with goblet cell metaplasia of unknown mechanism.
Collapse
Affiliation(s)
- Alejandro A. Pezzulo
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Rosarie A. Tudas
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Carley G. Stewart
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | | | - Brian D. Lindsay
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
| | - Peter J. Taft
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Nicholas D. Gansemer
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Joseph Zabner
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, and
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
31
|
Reznikov LR, Liao YSJ, Gu T, Davis KM, Kuan SP, Atanasova KR, Dadural JS, Collins EN, Guevara MV, Vogt K. Sex-specific airway hyperreactivity and sex-specific transcriptome remodeling in neonatal piglets challenged with intra-airway acid. Am J Physiol Lung Cell Mol Physiol 2018; 316:L131-L143. [PMID: 30407862 DOI: 10.1152/ajplung.00417.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Acute airway acidification is a potent stimulus of sensory nerves and occurs commonly with gastroesophageal reflux disease, cystic fibrosis, and asthma. In infants and adults, airway acidification can acutely precipitate asthma-like symptoms, and treatment-resistant asthma can be associated with gastroesophageal reflux disease. Airway protective behaviors, such as mucus secretion and airway smooth muscle contraction, are often exaggerated in asthma. These behaviors are manifested through activation of neural circuits. In some populations, the neural response to acid might be particularly important. For example, the immune response in infants is relatively immature compared with adults. Infants also have a high frequency of gastroesophageal reflux. Thus, in the current study, we compared the transcriptomes of an airway-nervous system circuit (e.g., tracheal epithelia, nodose ganglia, and brain stem) in neonatal piglets challenged with intra-airway acid. We hypothesized that the identification of parallel changes in the transcriptomes of two neutrally connected tissues might reveal the circuit response, and, hence, molecules important for the manifestation of asthma-like features. Intra-airway acid induced airway hyperreactivity and airway obstruction in male piglets. In contrast, female piglets displayed airway obstruction without airway hyperreactivity. Pairwise comparisons revealed parallel changes in genes directly implicated in airway hyperreactivity ( scn10a) in male acid-challenged piglets, whereas acid-challenged females exhibited parallel changes in genes associated with mild asthma ( stat 1 and isg15). These findings reveal sex-specific responses to acute airway acidification and highlight distinct molecules within a neural circuit that might be critical for the manifestation of asthma-like symptoms in pediatric populations.
Collapse
Affiliation(s)
- Leah R Reznikov
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Yan Shin J Liao
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Tongjun Gu
- Bioinformatics, Interdisciplinary Center for Biotechnology Research, University of Florida , Gainesville, Florida
| | - Katelyn M Davis
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Shin Ping Kuan
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Kalina R Atanasova
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Joshua S Dadural
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Emily N Collins
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Maria V Guevara
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| | - Kevin Vogt
- Department of Physiological Sciences, University of Florida , Gainesville, Florida
| |
Collapse
|
32
|
Winkelmann VE, Thompson KE, Neuland K, Jaramillo AM, Fois G, Schmidt H, Wittekindt OH, Han W, Tuvim MJ, Dickey BF, Dietl P, Frick M. Inflammation-induced upregulation of P2X 4 expression augments mucin secretion in airway epithelia. Am J Physiol Lung Cell Mol Physiol 2018; 316:L58-L70. [PMID: 30358443 DOI: 10.1152/ajplung.00157.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mucus clearance provides an essential innate defense mechanism to keep the airways and lungs free of particles and pathogens. Baseline and stimulated mucin secretion from secretory airway epithelial cells need to be tightly regulated to prevent mucus hypersecretion and mucus plugging of the airways. It is well established that extracellular ATP is a potent stimulus for regulated mucus secretion. Previous studies revealed that ATP acts via metabotropic P2Y2 purinoreceptors on goblet cells. Extracellular ATP, however, is also a potent agonist for ionotropic P2X purinoreceptors. Expression of several P2X isoforms has been reported in airways, but cell type-specific expression and the function thereof remained elusive. With this study, we now provide evidence that P2X4 is the predominant P2X isoform expressed in secretory airway epithelial cells. After IL-13 treatment of either human primary tracheal epithelial cells or mice, P2X4 expression is upregulated in vitro and in vivo under conditions of chronic inflammation, mucous metaplasia, and hyperplasia. Upregulation of P2X4 is strongest in MUC5AC-positive goblet cells. Moreover, activation of P2X4 by extracellular ATP augments intracellular Ca2+ signals and mucin secretion, whereas Ca2+ signals and mucin secretion are dampened by inhibition of P2X4 receptors. These data provide new insights into the purinergic regulation of mucin secretion and add to the emerging picture that P2X receptors modulate exocytosis of large secretory organelles and secretion of macromolecular vesicle cargo.
Collapse
Affiliation(s)
| | - Kristin E Thompson
- Centre de Recherche Saint-Antoine, INSERM, Université Pierre et Marie Curie-Université Paris 06, Sorbonne Universités, Paris , France
| | - Kathrin Neuland
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Ana M Jaramillo
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Giorgio Fois
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Hanna Schmidt
- Institute of General Physiology, Ulm University , Ulm , Germany
| | | | - Wei Han
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Michael J Tuvim
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Burton F Dickey
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Paul Dietl
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University , Ulm , Germany
| |
Collapse
|
33
|
Reznikov LR, Meyerholz DK, Kuan SP, Guevara MV, Atanasova KR, Abou Alaiwa MH. Solitary Cholinergic Stimulation Induces Airway Hyperreactivity and Transcription of Distinct Pro-inflammatory Pathways. Lung 2018; 196:219-229. [PMID: 29380034 DOI: 10.1007/s00408-018-0091-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/19/2018] [Indexed: 01/16/2023]
Abstract
Airway hyperreactivity is a hallmark feature of asthma and can be precipitated by airway insults, such as ozone exposure or viral infection. A proposed mechanism linking airway insults to airway hyperreactivity is augmented cholinergic transmission. In the current study, we tested the hypothesis that acute potentiation of cholinergic transmission is sufficient to induce airway hyperreactivity. We atomized the cholinergic agonist bethanechol to neonatal piglets and forty-eight hours later measured airway resistance. Bethanechol-treated piglets displayed increased airway resistance in response to intravenous methacholine compared to saline-treated controls. In the absence of an airway insult, we expected to find no evidence of airway inflammation; however, transcripts for several asthma-associated cytokines, including IL17A, IL1A, and IL8, were elevated in the tracheas of bethanechol-treated piglets. In the lungs, prior bethanechol treatment increased transcripts for IFNγ and its downstream target CXCL10. These findings suggest that augmented cholinergic transmission is sufficient to induce airway hyperreactivity, and raise the possibility that cholinergic-mediated regulation of pro-inflammatory pathways might contribute.
Collapse
Affiliation(s)
- Leah R Reznikov
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA.
| | - David K Meyerholz
- Department of Pathology, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Shin-Ping Kuan
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA
| | - Maria V Guevara
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA
| | - Kalina R Atanasova
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA
| | - Mahmoud H Abou Alaiwa
- Internal Medicine, Roy J and Lucille A Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
34
|
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
|
35
|
Spella M, Lilis I, Stathopoulos GT. Shared epithelial pathways to lung repair and disease. Eur Respir Rev 2017; 26:26/144/170048. [PMID: 28659498 DOI: 10.1183/16000617.0048-2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/19/2017] [Indexed: 01/04/2023] Open
Abstract
Chronic lung diseases present tremendous health burdens and share a common pathobiology of dysfunctional epithelial repair. Lung adenocarcinoma, the leading cancer killer worldwide, is caused mainly by chemical carcinogens of tobacco smoke that induce mutations in pulmonary epithelial cells leading to uncontrolled epithelial proliferation. Lung epithelial cells that possess the capacity for self-renewal and regeneration of other lung cell types are believed to underlie the pathobiology of chronic obstructive, fibrotic and neoplastic lung disorders. However, the understanding of lung epithelial progenitor cell hierarchy and turnover is incomplete and a comprehensive model of the cellular and transcriptional events that underlie lung regeneration and carcinogenesis is missing. The mapping of these processes is extremely important, since their modulation would potentially allow effective cure and/or prevention of chronic lung diseases. In this review we describe current knowledge on cellular and molecular pathways at play during lung repair and carcinogenesis and summarise the critical lung cell populations with regenerative and cancerous potential.
Collapse
Affiliation(s)
- Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Ioannis Lilis
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Rio, Greece .,Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University and Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
36
|
Haswell LE, Baxter A, Banerjee A, Verrastro I, Mushonganono J, Adamson J, Thorne D, Gaça M, Minet E. Reduced biological effect of e-cigarette aerosol compared to cigarette smoke evaluated in vitro using normalized nicotine dose and RNA-seq-based toxicogenomics. Sci Rep 2017; 7:888. [PMID: 28420881 PMCID: PMC5429854 DOI: 10.1038/s41598-017-00852-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/23/2017] [Indexed: 01/10/2023] Open
Abstract
Electronic cigarettes (e-cigarettes) use has increased globally and could potentially offer a lower risk alternative to cigarette smoking. Here, we assessed the transcriptional response of a primary 3D airway model acutely exposed to e-cigarette aerosol and cigarette (3R4F) smoke. Aerosols were generated with standard intense smoking regimens with careful consideration for dose by normalizing the exposures to nicotine. Two e-cigarette aerosol dilutions were tested for equivalent and higher nicotine delivery compared to 3R4F. RNA was extracted at 24 hrs and 48 hrs post exposure for RNA-seq. 873 and 205 RNAs were differentially expressed for 3R4F smoke at 24 hrs and 48 hrs using a pFDR < 0.01 and a [fold change] > 2 threshold. 113 RNAs were differentially expressed at the highest dose of e-cigarette aerosol using a looser threshold of pFDR < 0.05, 3 RNAs exceeded a fold change of 2. Geneset enrichment analysis revealed a clear response from lung cancer, inflammation, and fibrosis associated genes after 3R4F smoke exposure. Metabolic/biosynthetic processes, extracellular membrane, apoptosis, and hypoxia were identified for e-cigarette exposures, albeit with a lower confidence score. Based on equivalent or higher nicotine delivery, an acute exposure to e-cigarette aerosol had a reduced impact on gene expression compared to 3R4F smoke exposure in vitro.
Collapse
Affiliation(s)
- Linsey E Haswell
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Andrew Baxter
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Anisha Banerjee
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Ivan Verrastro
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Jessica Mushonganono
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Jason Adamson
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - David Thorne
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Marianna Gaça
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK
| | - Emmanuel Minet
- British American Tobacco R&D Centre, Regents Park Road, Southampton, SO15 8TL, UK.
| |
Collapse
|
37
|
Luettich K, Talikka M, Lowe FJ, Haswell LE, Park J, Gaca MD, Hoeng J. The Adverse Outcome Pathway for Oxidative Stress-Mediated EGFR Activation Leading to Decreased Lung Function. ACTA ACUST UNITED AC 2017. [DOI: 10.1089/aivt.2016.0032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Karsta Luettich
- Philip Morris International R&D, Philip Morris Products S.A. (Part of Philip Morris International Group of Companies), Neuchâtel, Switzerland
| | - Marja Talikka
- Philip Morris International R&D, Philip Morris Products S.A. (Part of Philip Morris International Group of Companies), Neuchâtel, Switzerland
| | - Frazer J. Lowe
- British American Tobacco (Investments) Ltd., Southampton, United Kingdom
| | - Linsey E. Haswell
- British American Tobacco (Investments) Ltd., Southampton, United Kingdom
| | | | - Marianna D. Gaca
- British American Tobacco (Investments) Ltd., Southampton, United Kingdom
| | - Julia Hoeng
- Philip Morris International R&D, Philip Morris Products S.A. (Part of Philip Morris International Group of Companies), Neuchâtel, Switzerland
| |
Collapse
|
38
|
Gorrieri G, Scudieri P, Caci E, Schiavon M, Tomati V, Sirci F, Napolitano F, Carrella D, Gianotti A, Musante I, Favia M, Casavola V, Guerra L, Rea F, Ravazzolo R, Di Bernardo D, Galietta LJV. Goblet Cell Hyperplasia Requires High Bicarbonate Transport To Support Mucin Release. Sci Rep 2016; 6:36016. [PMID: 27786259 PMCID: PMC5081536 DOI: 10.1038/srep36016] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/05/2016] [Indexed: 12/15/2022] Open
Abstract
Goblet cell hyperplasia, a feature of asthma and other respiratory diseases, is driven by the Th-2 cytokines IL-4 and IL-13. In human bronchial epithelial cells, we find that IL-4 induces the expression of many genes coding for ion channels and transporters, including TMEM16A, SLC26A4, SLC12A2, and ATP12A. At the functional level, we find that IL-4 enhances calcium- and cAMP-activated chloride/bicarbonate secretion, resulting in high bicarbonate concentration and alkaline pH in the fluid covering the apical surface of epithelia. Importantly, mucin release, elicited by purinergic stimulation, requires the presence of bicarbonate in the basolateral solution and is defective in cells derived from cystic fibrosis patients. In conclusion, our results suggest that Th-2 cytokines induce a profound change in expression and function in multiple ion channels and transporters that results in enhanced bicarbonate transport ability. This change is required as an important mechanism to favor release and clearance of mucus.
Collapse
Affiliation(s)
- Giulia Gorrieri
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Paolo Scudieri
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Emanuela Caci
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Marco Schiavon
- Department of Thoracic Surgery, University of Padova, Italy
| | - Valeria Tomati
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Francesco Sirci
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | - Diego Carrella
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Ambra Gianotti
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Ilaria Musante
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Italy
| | - Valeria Casavola
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Italy
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Italy
| | - Federico Rea
- Department of Thoracic Surgery, University of Padova, Italy
| | - Roberto Ravazzolo
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy.,DINOGMI, University of Genova, Italy
| | | | | |
Collapse
|
39
|
Differential Expression Patterns of EGF, EGFR, and ERBB4 in Nasal Polyp Epithelium. PLoS One 2016; 11:e0156949. [PMID: 27285994 PMCID: PMC4902223 DOI: 10.1371/journal.pone.0156949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/23/2016] [Indexed: 11/24/2022] Open
Abstract
Epidermal growth factor receptors play an important role in airway epithelial cell growth and differentiation. The current study investigates the expression profiles of EGF, EGFR and ERBB4 in patients with nasal polyps (NP), and their response to glucocorticosteroid (GC) treatment. Fifty patients with NP (40 without GC treatment and 10 with oral GC) and 20 control subjects with septal deviation were recruited into the study. Protein levels of EGF, EGFR, and ERBB4 were evaluated by immune-staining. In healthy nasal epithelium, EGF and EGFR localized within p63+ basal cells, while ERBB4 localized within ciliated cells. GC-naïve NP epithelium showed weak expression of EGF in 90% of samples versus 5% of controls. EGFR was significantly increased in the epithelium with basal cell hyperplasia from GC-naïve NPs (78%, 31/40) compared to controls (23%, 4/17). EGFR was also found in some degranulating goblet cells. ERBB4 expression was significantly higher in hyperplastic epithelium from GC-naïve NPs (65%, 26/40) than in controls (6%, 1/17). GC treatment restored the EGF expression and normalized the EGFR and ERBB4 expression in NPs. Differential expression patterns of EGF, EGFR, and ERBB4 are essential in epithelial restitution and remodeling in nasal epithelium.
Collapse
|
40
|
Schilders KAA, Eenjes E, van Riet S, Poot AA, Stamatialis D, Truckenmüller R, Hiemstra PS, Rottier RJ. Regeneration of the lung: Lung stem cells and the development of lung mimicking devices. Respir Res 2016; 17:44. [PMID: 27107715 PMCID: PMC4842297 DOI: 10.1186/s12931-016-0358-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/25/2016] [Indexed: 01/07/2023] Open
Abstract
Inspired by the increasing burden of lung associated diseases in society and an growing demand to accommodate patients, great efforts by the scientific community produce an increasing stream of data that are focused on delineating the basic principles of lung development and growth, as well as understanding the biomechanical properties to build artificial lung devices. In addition, the continuing efforts to better define the disease origin, progression and pathology by basic scientists and clinicians contributes to insights in the basic principles of lung biology. However, the use of different model systems, experimental approaches and readout systems may generate somewhat conflicting or contradictory results. In an effort to summarize the latest developments in the lung epithelial stem cell biology, we provide an overview of the current status of the field. We first describe the different stem cells, or progenitor cells, residing in the homeostatic lung. Next, we focus on the plasticity of the different cell types upon several injury-induced activation or repair models, and highlight the regenerative capacity of lung cells. Lastly, we summarize the generation of lung mimics, such as air-liquid interface cultures, organoids and lung on a chip, that are required to test emerging hypotheses. Moreover, the increasing collaboration between distinct specializations will contribute to the eventual development of an artificial lung device capable of assisting reduced lung function and capacity in human patients.
Collapse
Affiliation(s)
- Kim A A Schilders
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Evelien Eenjes
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - André A Poot
- Department of Biomaterials Science and Technology, University of Twente, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, P.O Box 217, 7500 AE, Enschede, The Netherlands
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, University of Twente, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, P.O Box 217, 7500 AE, Enschede, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Regeneration, Maastricht University, Faculty of Health, Medicine and Life Sciences, MERLN Institute for Technology-Inspired Regenerative Medicine, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Emura M, Aufderheide M. Challenge for 3D culture technology: Application in carcinogenesis studies with human airway epithelial cells. ACTA ACUST UNITED AC 2016; 68:255-61. [PMID: 26951634 DOI: 10.1016/j.etp.2016.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 11/18/2022]
Abstract
Lung cancer is still one of the major intractable diseases and we urgently need more efficient preventive and curative measures. Recent molecular studies have provided strong evidence that allows us to believe that classically well-known early airway lesions such as hyperplasia, metaplasia, dysplasia and carcinoma in situ are really precancerous lesions progressing toward cancer but not necessarily transient and reversible alteration. This suggests that adequate early control of the precancerous lesions may lead to improved prevention of lung cancer. This knowledge is encouraging in view of the imminent necessity for additional experimental systems to investigate the causal mechanisms of cancers directly in human cells and tissues. There are many questions with regard to various precancerous lesions of the airways. For example, should cells, before reaching a stage of invasive carcinoma, undergo all precancerous stages such as hyperplasia or metaplasia and dysplasia, or is there any shortcut to bypass one or more of the precancerous stages? For the study of such questions, the emerging 3-dimensional (3D) cell culture technology appears to provide an effective and valuable tool. Though a great challenge, it is expected that this in vitro technology will be rapidly and reliably improved to enable the cultures to be maintained in an in vivo-mimicking state of differentiation for much longer than a period of at best a few months, as is currently the case. With the help of a "causes recombination-Lox" (Cre-lox) technology, it has been possible to trace cells giving rise to specific lung tumor types. In this short review we have attempted to assess the future role of 3D technology in the study of lung carcinogenesis.
Collapse
Affiliation(s)
- M Emura
- Cultex(®) Laboratories GmbH, Feodor-Lynen-Str. 21, 30625 Hannover, Germany.
| | - M Aufderheide
- Cultex(®) Laboratories GmbH, Feodor-Lynen-Str. 21, 30625 Hannover, Germany.
| |
Collapse
|
42
|
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
|
43
|
Upregulation of TMEM16A Protein in Bronchial Epithelial Cells by Bacterial Pyocyanin. PLoS One 2015; 10:e0131775. [PMID: 26121472 PMCID: PMC4486680 DOI: 10.1371/journal.pone.0131775] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/06/2015] [Indexed: 01/22/2023] Open
Abstract
Induction of mucus hypersecretion in the airway epithelium by Th2 cytokines is associated with the expression of TMEM16A, a Ca2+-activated Cl- channel. We asked whether exposure of airway epithelial cells to bacterial components, a condition that mimics the highly infected environment occurring in cystic fibrosis (CF), also results in a similar response. In cultured human bronchial epithelial cells, treatment with pyocyanin or with a P. aeruginosa culture supernatant caused a significant increase in TMEM16A function. The Ca2+-dependent Cl- secretion, triggered by stimulation with UTP, was particularly enhanced by pyocyanin in cells from CF patients. Increased expression of TMEM16A protein and of MUC5AC mucin by bacterial components was demonstrated by immunofluorescence in CF and non-CF cells. We also investigated TMEM16A expression in human bronchi by immunocytochemistry. We found increased TMEM16A staining in the airways of CF patients. The strongest signal was observed in CF submucosal glands. Our results suggest that TMEM16A expression/function is upregulated in CF lung disease, possibly as a response towards the presence of bacteria in the airways.
Collapse
|
44
|
Jenny RA, Hirst C, Lim SM, Goulburn AL, Micallef SJ, Labonne T, Kicic A, Ling KM, Stick SM, Ng ES, Trounson A, Giudice A, Elefanty AG, Stanley EG. Productive Infection of Human Embryonic Stem Cell-Derived NKX2.1+ Respiratory Progenitors with Human Rhinovirus. Stem Cells Transl Med 2015; 4:603-14. [PMID: 25873746 DOI: 10.5966/sctm.2014-0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/09/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Airway epithelial cells generated from pluripotent stem cells (PSCs) represent a resource for research into a variety of human respiratory conditions, including those resulting from infection with common human pathogens. Using an NKX2.1-GFP reporter human embryonic stem cell line, we developed a serum-free protocol for the generation of NKX2.1(+) endoderm that, when transplanted into immunodeficient mice, matured into respiratory cell types identified by expression of CC10, MUC5AC, and surfactant proteins. Gene profiling experiments indicated that day 10 NKX2.1(+) endoderm expressed markers indicative of early foregut but lacked genes associated with later stages of respiratory epithelial cell differentiation. Nevertheless, NKX2.1(+) endoderm supported the infection and replication of the common respiratory pathogen human rhinovirus HRV1b. Moreover, NKX2.1(+) endoderm upregulated expression of IL-6, IL-8, and IL-1B in response to infection, a characteristic of human airway epithelial cells. Our experiments provide proof of principle for the use of PSC-derived respiratory epithelial cells in the study of cell-virus interactions. SIGNIFICANCE This report provides proof-of-principle experiments demonstrating, for the first time, that human respiratory progenitor cells derived from stem cells in the laboratory can be productively infected with human rhinovirus, the predominant cause of the common cold.
Collapse
Affiliation(s)
- Robert A Jenny
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Claire Hirst
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Sue Mei Lim
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Adam L Goulburn
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Suzanne J Micallef
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Tanya Labonne
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Anthony Kicic
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Kak-Ming Ling
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Stephen M Stick
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Elizabeth S Ng
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Alan Trounson
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Antonietta Giudice
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Andrew G Elefanty
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| | - Edouard G Stanley
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia; Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Telethon Kids Institute, Centre for Health Research, School of Paediatrics and Child Health, Centre for Health Research, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia; Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia; Richie Centre, Monash Prince Henry's Medical Research Institute, Clayton, Victoria, Australia
| |
Collapse
|
45
|
Cigarette smoke alters primary human bronchial epithelial cell differentiation at the air-liquid interface. Sci Rep 2015; 5:8163. [PMID: 25641363 PMCID: PMC4313097 DOI: 10.1038/srep08163] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022] Open
Abstract
The differentiated human airway epithelium consists of different cell types forming a polarized and pseudostratified epithelium. This is dramatically altered in chronic obstructive pulmonary disease (COPD), characterized by basal and goblet cell hyperplasia, and squamous cell metaplasia. The effect of cigarette smoke on human bronchial epithelial cell (HBEC) differentiation remains to be elucidated. We analysed whether cigarette smoke extract (CSE) affected primary (p)HBEC differentiation and function. pHBEC were differentiated at the air-liquid interface (ALI) and differentiation was quantified after 7, 14, 21, or 28 days by assessing acetylated tubulin, CC10, or MUC5AC for ciliated, Clara, or goblet cells, respectively. Exposure of differentiating pHBEC to CSE impaired epithelial barrier formation, as assessed by resistance measurements (TEER). Importantly, CSE exposure significantly reduced the number of ciliated cells, while it increased the number of Clara and goblet cells. CSE-dependent cell number changes were reflected by a reduction of acetylated tubulin levels, an increased expression of the basal cell marker KRT14, and increased secretion of CC10, but not by changes in transcript levels of CC10, MUC5AC, or FOXJ1. Our data demonstrate that cigarette smoke specifically alters the cellular composition of the airway epithelium by affecting basal cell differentiation in a post-transcriptional manner.
Collapse
|
46
|
Brinkworth JF, Barreiro LB. The contribution of natural selection to present-day susceptibility to chronic inflammatory and autoimmune disease. Curr Opin Immunol 2014; 31:66-78. [PMID: 25458997 DOI: 10.1016/j.coi.2014.09.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Chronic inflammatory and autoimmune diseases have been the focus of many genome-wide association studies (GWAS) because they represent a significant cause of illness and morbidity, and many are heritable. Almost a decade of GWAS studies suggests that the pathological inflammation associated with these diseases is controlled by a limited number of networked immune system genes. Chronic inflammatory and autoimmune diseases are enigmatic from an evolutionary perspective because they exert a negative affect on reproductive fitness. The persistence of these conditions may be partially explained by the important roles the implicated immune genes play in pathogen defense and other functions thought to be under strong natural selection in humans. The evolutionary reasons for chronic inflammatory and autoimmune disease persistence and uneven distribution across populations are the focus of this review.
Collapse
Affiliation(s)
- Jessica F Brinkworth
- Sainte-Justine Hospital Research Centre, Montréal, Quebec H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Luis B Barreiro
- Sainte-Justine Hospital Research Centre, Montréal, Quebec H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada.
| |
Collapse
|
47
|
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
|
48
|
Paz AC, Soleas J, Poon JC, Trieu D, Waddell TK, McGuigan AP. Challenges and Opportunities for Tissue-Engineering Polarized Epithelium. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:56-72. [DOI: 10.1089/ten.teb.2013.0144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ana C. Paz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - John Soleas
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - James C.H. Poon
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Dennis Trieu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Boucherat O, Boczkowski J, Jeannotte L, Delacourt C. Cellular and molecular mechanisms of goblet cell metaplasia in the respiratory airways. Exp Lung Res 2013; 39:207-16. [DOI: 10.3109/01902148.2013.791733] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
50
|
Pardo-Saganta A, Law BM, Gonzalez-Celeiro M, Vinarsky V, Rajagopal J. Ciliated cells of pseudostratified airway epithelium do not become mucous cells after ovalbumin challenge. Am J Respir Cell Mol Biol 2012; 48:364-73. [PMID: 23239495 DOI: 10.1165/rcmb.2012-0146oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucous cell metaplasia is a hallmark of airway diseases, such as asthma and chronic obstructive pulmonary disease. The majority of human airway epithelium is pseudostratified, but the cell of origin of mucous cells has not been definitively established in this type of airway epithelium. There is evidence that ciliated, club cell (Clara), and basal cells can all give rise to mucus-producing cells in different contexts. Because pseudostratified airway epithelium contains distinct progenitor cells from simple columnar airway epithelium, the lineage relationships of progenitor cells to mucous cells may be different in these two epithelial types. We therefore performed lineage tracing of the ciliated cells of the murine basal cell-containing airway epithelium in conjunction with the ovalbumin (OVA)-induced murine model of allergic lung disease. We genetically labeled ciliated cells with enhanced Yellow Fluorescent Protein (eYFP) before the allergen challenge, and followed the fate of these cells to determine whether they gave rise to newly formed mucous cells. Although ciliated cells increased in number after the OVA challenge, the newly formed mucous cells were not labeled with the eYFP lineage tag. Even small numbers of labeled mucous cells could not be detected, implying that ciliated cells make virtually no contribution to the new goblet cell pool. This demonstrates that, after OVA challenge, new mucous cells do not originate from ciliated cells in a pseudostratified basal cell-containing airway epithelium.
Collapse
Affiliation(s)
- Ana Pardo-Saganta
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|