1
|
Bondeelle L, Clément S, Bergeron A, Tapparel C. Lung stem cells and respiratory epithelial chimerism in transplantation. Eur Respir Rev 2025; 34:240146. [PMID: 39971397 PMCID: PMC11836672 DOI: 10.1183/16000617.0146-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Stem cells are capable of self-renewal and differentiation into specialised types. They range from totipotent cells to multipotent or somatic stem cells and ultimately to unipotent cells. Some adult multipotent stem cells can have the potential to regenerate and colonise diverse tissues. The respiratory airways and lung mucosa, exposed to ambient air, perform vital roles for all human tissues and organs. They serve as barriers against airborne threats and are essential for tissue oxygenation. Despite low steady-state turnover, lungs are vulnerable to injuries and diseases from environmental exposure. Lung stem cells are crucial due to their regenerative potential and ability to replace damaged cells. Lung repair with extrapulmonary stem cells can occur, leading to the coexistence of respiratory cells with different genetic origins, a phenomenon known as airway epithelial chimerism. The impact of such chimerism in lung repair and disease is actively studied. This review explores different stem cell types, focusing on pulmonary stem cells. It discusses airway epithelium models derived from stem cells for studying lung diseases and examines lung chimerism, particularly in lung transplantation and haematopoietic stem cell transplantation, highlighting its significance in understanding tissue repair and chimerism-mediated repair processes in lung pathology.
Collapse
Affiliation(s)
- Louise Bondeelle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, Geneva, Switzerland
- Co-last author
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Co-last author
| |
Collapse
|
2
|
Serna Villa V, Ren X. Lung Progenitor and Stem Cell Transplantation as a Potential Regenerative Therapy for Lung Diseases. Transplantation 2024; 108:e282-e291. [PMID: 38416452 DOI: 10.1097/tp.0000000000004959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Chronic lung diseases are debilitating illnesses ranking among the top causes of death globally. Currently, clinically available therapeutic options capable of curing chronic lung diseases are limited to lung transplantation, which is hindered by donor organ shortage. This highlights the urgent need for alternative strategies to repair damaged lung tissues. Stem cell transplantation has emerged as a promising avenue for regenerative treatment of the lung, which involves delivery of healthy lung epithelial progenitor cells that subsequently engraft in the injured tissue and further differentiate to reconstitute the functional respiratory epithelium. These transplanted progenitor cells possess the remarkable ability to self-renew, thereby offering the potential for sustained long-term treatment effects. Notably, the transplantation of basal cells, the airway stem cells, holds the promise for rehabilitating airway injuries resulting from environmental factors or genetic conditions such as cystic fibrosis. Similarly, for diseases affecting the alveoli, alveolar type II cells have garnered interest as a viable alveolar stem cell source for restoring the lung parenchyma from genetic or environmentally induced dysfunctions. Expanding upon these advancements, the use of induced pluripotent stem cells to derive lung progenitor cells for transplantation offers advantages such as scalability and patient specificity. In this review, we comprehensively explore the progress made in lung stem cell transplantation, providing insights into the current state of the field and its future prospects.
Collapse
Affiliation(s)
- Vanessa Serna Villa
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | | |
Collapse
|
3
|
Milesi J, Gras D, Chanez P, Coiffard B. Airway epithelium in lung transplantation: a potential actor for post-transplant complications? Eur Respir Rev 2024; 33:240093. [PMID: 39603662 PMCID: PMC11600126 DOI: 10.1183/16000617.0093-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/20/2024] [Indexed: 11/29/2024] Open
Abstract
Lung transplantation, a critical intervention for end-stage lung diseases, is frequently challenged by post-transplant complications. Indeed, primary graft dysfunction, anastomotic complications, infections and acute and chronic rejections pose significant hurdles in lung transplantation. While evidence regarding the role of airway epithelium after lung transplantation is still emerging, its importance is becoming increasingly recognised. This review looks at the complex involvement of airway epithelium in various post-transplant complications, while emphasising the utility of airway epithelial culture as a research model. In summary, by elucidating the involvement of airway epithelium in each post-transplant complication and explaining these intricate processes, the review aims to guide specific future research efforts and therapeutic strategies aimed at improving lung transplant outcomes and enhancing patient care.
Collapse
Affiliation(s)
- Jules Milesi
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Delphine Gras
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pascal Chanez
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Benjamin Coiffard
- Aix-Marseille University, APHM, Department of Respiratory Medicine and Lung Transplantation, Marseille, France
- Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
4
|
Ruysseveldt E, Steelant B, Wils T, Cremer J, Bullens DMA, Hellings PW, Martens K. The nasal basal cell population shifts toward a diseased phenotype with impaired barrier formation capacity in allergic rhinitis. J Allergy Clin Immunol 2024; 154:631-643. [PMID: 38705259 DOI: 10.1016/j.jaci.2024.04.021] [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: 12/21/2023] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND The integrity of the airway epithelium is guarded by the airway basal cells that serve as progenitor cells and restore wounds in case of injury. Basal cells are a heterogenous population, and specific changes in their behavior are associated with chronic barrier disruption-mechanisms that have not been studied in detail in allergic rhinitis (AR). OBJECTIVE We aimed to study basal cell subtypes in AR and healthy controls. METHODS Single-cell RNA sequencing (scRNA-Seq) of the nasal epithelium was performed on nonallergic and house dust mite-allergic AR patients to reveal basal cell diversity and to identify allergy-related alterations. Flow cytometry, immunofluorescence staining, and in vitro experiments using primary basal cells were performed to confirm phenotypic findings at the protein level and functionally. RESULTS The scRNA-Seq, flow cytometry, and immunofluorescence staining revealed that basal cells are abundantly and heterogeneously present in the nasal epithelium, suggesting specialized subtypes. The total basal cell fraction within the epithelium in AR is increased compared to controls. scRNA-Seq demonstrated that potentially beneficial basal cells are missing in AR epithelium, while an activated population of allergy-associated basal cells is more dominantly present. Furthermore, our in vitro proliferation, wound healing assay and air-liquid interface cultures show that AR-associated basal cells have altered progenitor capacity compared to nonallergic basal cells. CONCLUSIONS The nasal basal cell population is abundant and diverse, and it shifts toward a diseased state in AR. The absence of potentially protective subtypes and the rise of a proinflammatory population suggest that basal cells are important players in maintaining epithelial barrier defects in AR.
Collapse
Affiliation(s)
- Emma Ruysseveldt
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium.
| | - Brecht Steelant
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Tine Wils
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Jonathan Cremer
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Dominique M A Bullens
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium; Clinical Department of Paediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Peter W Hellings
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium; Clinical Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium; Upper Airways Research Laboratory, University of Ghent, Ghent, Belgium
| | - Katleen Martens
- Department of Microbiology, Immunology, and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium; Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
5
|
Thakur A, Mei S, Zhang N, Zhang K, Taslakjian B, Lian J, Wu S, Chen B, Solway J, Chen HJ. Pulmonary neuroendocrine cells: crucial players in respiratory function and airway-nerve communication. Front Neurosci 2024; 18:1438188. [PMID: 39176384 PMCID: PMC11340541 DOI: 10.3389/fnins.2024.1438188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/04/2024] [Indexed: 08/24/2024] Open
Abstract
Pulmonary neuroendocrine cells (PNECs) are unique airway epithelial cells that blend neuronal and endocrine functions, acting as key sensors in the lung. They respond to environmental stimuli like allergens by releasing neuropeptides and neurotransmitters. PNECs stand out as the only lung epithelial cells innervated by neurons, suggesting a significant role in airway-nerve communication via direct neural pathways and hormone release. Pathological conditions such as asthma are linked to increased PNECs counts and elevated calcitonin gene-related peptide (CGRP) production, which may affect neuroprotection and brain function. CGRP is also associated with neurodegenerative diseases, including Parkinson's and Alzheimer's, potentially due to its influence on inflammation and cholinergic activity. Despite their low numbers, PNECs are crucial for a wide range of functions, highlighting the importance of further research. Advances in technology for producing and culturing human PNECs enable the exploration of new mechanisms and cell-specific responses to targeted therapies for PNEC-focused treatments.
Collapse
Affiliation(s)
- Abhimanyu Thakur
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| | - Shuya Mei
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| | - Noel Zhang
- Canyon Crest Academy, San Diego, CA, United States
| | - Kui Zhang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| | - Boghos Taslakjian
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Jiacee Lian
- School of Health Sciences, Ngee Ann Polytechnic, Singapore, Singapore
| | - Shuang Wu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, United States
| | - Julian Solway
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, United States
| | - Huanhuan Joyce Chen
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
6
|
Zhu H, Leng J, Ju R, Qu S, Tian J, Leng H, Tao S, Liu C, Wu Z, Ren F, Lyu Y, Zhang N. Advantages of pulsed electric field ablation for COPD: Excellent killing effect on goblet cells. Bioelectrochemistry 2024; 158:108726. [PMID: 38733722 DOI: 10.1016/j.bioelechem.2024.108726] [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: 11/18/2023] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Mucus hypersecretion resulting from excessive proliferation and metaplasia of goblet cells in the airways is the pathological foundation for Chronic obstructive pulmonary disease (COPD). Clinical trials have confirmed the clinical efficacy of pulsed electric field ablation (PFA) for COPD, but its underlying mechanisms is poorly understood. Cellular and animal models of COPD (rich in goblet cells) were established in this study to detect goblet cells' sensitivity to PFA. Schwan's equation was adopted to calculate the cells' transmembrane potential and the electroporation areas in the cell membrane. We found that goblet cells are more sensitive to low-intensity PFA (250 V/cm-500 V/cm) than BEAS-2B cells. It is attributed to the larger size of goblet cells, which allows a stronger transmembrane potential formation under the same electric field strength. Additionally, the transmembrane potential of larger-sized cells can reach the cell membrane electroporation threshold in more areas. Trypan blue staining confirmed that the cells underwent IRE rate was higher in goblet cells than in BEAS-2B cells. Animal experiments also confirmed that the airway epithelium of COPD is more sensitive to PFA. We conclude that lower-intensity PFA can selectively kill goblet cells in the COPD airway epithelium, ultimately achieving the therapeutic effect of treating COPD.
Collapse
Affiliation(s)
- Haoyang Zhu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jing Leng
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Ran Ju
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shenao Qu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiawei Tian
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haoze Leng
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shiran Tao
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Chang Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fenggang Ren
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Lyu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Nana Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710049, China; Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
7
|
Kayalar Ö, Rajabi H, Konyalilar N, Mortazavi D, Aksoy GT, Wang J, Bayram H. Impact of particulate air pollution on airway injury and epithelial plasticity; underlying mechanisms. Front Immunol 2024; 15:1324552. [PMID: 38524119 PMCID: PMC10957538 DOI: 10.3389/fimmu.2024.1324552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
Air pollution plays an important role in the mortality and morbidity of chronic airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Particulate matter (PM) is a significant fraction of air pollutants, and studies have demonstrated that it can cause airway inflammation and injury. The airway epithelium forms the first barrier of defense against inhaled toxicants, such as PM. Airway epithelial cells clear airways from inhaled irritants and orchestrate the inflammatory response of airways to these irritants by secreting various lipid mediators, growth factors, chemokines, and cytokines. Studies suggest that PM plays an important role in the pathogenesis of chronic airway diseases by impairing mucociliary function, deteriorating epithelial barrier integrity, and inducing the production of inflammatory mediators while modulating the proliferation and death of airway epithelial cells. Furthermore, PM can modulate epithelial plasticity and airway remodeling, which play central roles in asthma and COPD. This review focuses on the effects of PM on airway injury and epithelial plasticity, and the underlying mechanisms involving mucociliary activity, epithelial barrier function, airway inflammation, epithelial-mesenchymal transition, mesenchymal-epithelial transition, and airway remodeling.
Collapse
Affiliation(s)
- Özgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Hadi Rajabi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Deniz Mortazavi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Gizem Tuşe Aksoy
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
| | - Jun Wang
- Department of Biomedicine and Biopharmacology, School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Türkiye
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Türkiye
| |
Collapse
|
8
|
Raach B, Bundgaard N, Haase MJ, Starruß J, Sotillo R, Stanifer ML, Graw F. Influence of cell type specific infectivity and tissue composition on SARS-CoV-2 infection dynamics within human airway epithelium. PLoS Comput Biol 2023; 19:e1011356. [PMID: 37566610 PMCID: PMC10446191 DOI: 10.1371/journal.pcbi.1011356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/23/2023] [Accepted: 07/13/2023] [Indexed: 08/13/2023] Open
Abstract
Human airway epithelium (HAE) represents the primary site of viral infection for SARS-CoV-2. Comprising different cell populations, a lot of research has been aimed at deciphering the major cell types and infection dynamics that determine disease progression and severity. However, the cell type-specific replication kinetics, as well as the contribution of cellular composition of the respiratory epithelium to infection and pathology are still not fully understood. Although experimental advances, including Air-liquid interface (ALI) cultures of reconstituted pseudostratified HAE, as well as lung organoid systems, allow the observation of infection dynamics under physiological conditions in unprecedented level of detail, disentangling and quantifying the contribution of individual processes and cells to these dynamics remains challenging. Here, we present how a combination of experimental data and mathematical modelling can be used to infer and address the influence of cell type specific infectivity and tissue composition on SARS-CoV-2 infection dynamics. Using a stepwise approach that integrates various experimental data on HAE culture systems with regard to tissue differentiation and infection dynamics, we develop an individual cell-based model that enables investigation of infection and regeneration dynamics within pseudostratified HAE. In addition, we present a novel method to quantify tissue integrity based on image data related to the standard measures of transepithelial electrical resistance measurements. Our analysis provides a first aim of quantitatively assessing cell type specific infection kinetics and shows how tissue composition and changes in regeneration capacity, as e.g. in smokers, can influence disease progression and pathology. Furthermore, we identified key measurements that still need to be assessed in order to improve inference of cell type specific infection kinetics and disease progression. Our approach provides a method that, in combination with additional experimental data, can be used to disentangle the complex dynamics of viral infection and immunity within human airway epithelial culture systems.
Collapse
Affiliation(s)
- Benjamin Raach
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Nils Bundgaard
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Marika J. Haase
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Jörn Starruß
- Center for Information Services and High Performance Computing, TU Dresden, Dresden, Germany
| | - Rocio Sotillo
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Megan L. Stanifer
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
- University of Florida, College of Medicine, Dept. of Molecular Genetics and Microbiology, Gainesville, Florida, United States of America
| | - Frederik Graw
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Medicine 5, Erlangen, Germany
| |
Collapse
|
9
|
Bollenbecker S, Heitman K, Czaya B, Easter M, Hirsch MJ, Vang S, Harris E, Helton ES, Barnes JW, Faul C, Krick S. Phosphate induces inflammation and exacerbates injury from cigarette smoke in the bronchial epithelium. Sci Rep 2023; 13:4898. [PMID: 36966182 PMCID: PMC10039898 DOI: 10.1038/s41598-023-32053-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023] Open
Abstract
An elevation in serum phosphate-also called hyperphosphatemia-is associated with reduced kidney function in chronic kidney disease (CKD). Reports show CKD patients are more likely to develop lung disease and have poorer kidney function that positively correlates with pulmonary obstruction. However, the underlying mechanisms are not well understood. Here, we report that two murine models of CKD, which both exhibit increased serum levels of phosphate and fibroblast growth factor (FGF) 23, a regulator of phosphate homeostasis, develop concomitant airway inflammation. Our in vitro studies point towards a similar increase of phosphate-induced inflammatory markers in human bronchial epithelial cells. FGF23 stimulation alone does not induce a proinflammatory response in the non-COPD bronchial epithelium and phosphate does not cause endogenous FGF23 release. Upregulation of the phosphate-induced proinflammatory cytokines is accompanied by activation of the extracellular-signal regulated kinase (ERK) pathway. Moreover, the addition of cigarette smoke extract (CSE) during phosphate treatments exacerbates inflammation as well as ERK activation, whereas co-treatment with FGF23 attenuates both the phosphate as well as the combined phosphate- and CS-induced inflammatory response, independent of ERK activation. Together, these data demonstrate a novel pathway that potentially explains pathological kidney-lung crosstalk with phosphate as a key mediator.
Collapse
Affiliation(s)
- Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Kylie Heitman
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Brian Czaya
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Shia Vang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Elex Harris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - E Scott Helton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Jarrod W Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA
| | - Christian Faul
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, 1918 University Blvd, MCLM 718, Birmingham, AL, 35294, USA.
| |
Collapse
|
10
|
Byrd AL, Qu X, Lukyanchuk A, Liu J, Chen F, Naughton KJ, DuCote TJ, Song X, Bowman HC, Zhao Y, Edgin AR, Wang C, Liu J, Brainson CF. Dysregulated Polycomb Repressive Complex 2 contributes to chronic obstructive pulmonary disease by rewiring stem cell fate. Stem Cell Reports 2022; 18:289-304. [PMID: 36525966 PMCID: PMC9860081 DOI: 10.1016/j.stemcr.2022.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022] Open
Abstract
Aberrant lung cell differentiation is a hallmark of many lung diseases including chronic obstructive pulmonary disease (COPD). The EZH2-containing Polycomb Repressive Complex 2 (PRC2) regulates embryonic lung stem cell fate, but its role in adult lung is obscure. Histological analysis of patient tissues revealed that loss of PRC2 activity was correlated with aberrant bronchiolar cell differentiation in COPD lung. Histological and single-cell RNA-sequencing analyses showed that loss of EZH2 in mouse lung organoids led to lowered self-renewal capability, increased squamous morphological development, and marked shifts in progenitor cell populations. Evaluation of in vivo models revealed that heterozygosity of Ezh2 in mice with ovalbumin-induced lung inflammation led to epithelial cell differentiation patterns similar to those in COPD lung. We also identified cystathionine-β-synthase as a possible upstream factor for PRC2 destabilization. Our findings suggest that PRC2 is integral to facilitating proper lung stem cell differentiation in humans and mice.
Collapse
Affiliation(s)
- Aria L. Byrd
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Xufeng Qu
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Alexsandr Lukyanchuk
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Jinpeng Liu
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Fan Chen
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Kassandra J. Naughton
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Tanner J. DuCote
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Xiulong Song
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Hannah C. Bowman
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Abigail R. Edgin
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Chi Wang
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA,Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Jinze Liu
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Christine Fillmore Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA; Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
11
|
Osan J, Talukdar SN, Feldmann F, DeMontigny BA, Jerome K, Bailey KL, Feldmann H, Mehedi M. Goblet Cell Hyperplasia Increases SARS-CoV-2 Infection in Chronic Obstructive Pulmonary Disease. Microbiol Spectr 2022; 10:e0045922. [PMID: 35862971 PMCID: PMC9430117 DOI: 10.1128/spectrum.00459-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/29/2022] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the underlying conditions in adults of any age that place them at risk for developing severe illnesses associated with COVID-19. To determine whether SARS-CoV-2's cellular tropism plays a critical role in severe pathophysiology in the lung, we investigated its host cell entry receptor distribution in the bronchial airway epithelium of healthy adults and high-risk adults (those with COPD). We found that SARS-CoV-2 preferentially infects goblet cells in the bronchial airway epithelium, as mostly goblet cells harbor the entry receptor angiotensin-converting enzyme 2 (ACE2) and its cofactor transmembrane serine protease 2 (TMPRSS2). We also found that SARS-CoV-2 replication was substantially increased in the COPD bronchial airway epithelium, likely due to COPD-associated goblet cell hyperplasia. Likewise, SARS-CoV and Middle East respiratory syndrome (MERS-CoV) infection increased disease pathophysiology (e.g., syncytium formation) in the COPD bronchial airway epithelium. Our results reveal that goblet cells play a critical role in SARS-CoV-2-induced pathophysiology in the lung. IMPORTANCE SARS-CoV-2 or COVID-19's first case was discovered in December 2019 in Wuhan, China, and by March 2020 it was declared a pandemic by the WHO. It has been shown that various underlying conditions can increase the chance of having severe COVID-19. COPD, which is the third leading cause of death worldwide, is one of the conditions listed by the CDC which can increase the chance of severe COVID-19. The present study uses a healthy and COPD-derived bronchial airway epithelial model to study the COVID-19 and host factors which could explain the reason for COPD patients developing severe infection due to COVID-19.
Collapse
Affiliation(s)
- Jaspreet Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Sattya N. Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Friederike Feldmann
- Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Beth Ann DeMontigny
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Kailey Jerome
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| | - Kristina L. Bailey
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep and Allergy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Heinz Feldmann
- Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, USA
| |
Collapse
|
12
|
Ancel J, Guecamburu M, Marques Da Silva V, Schilfarth P, Boyer L, Pilette C, Martin C, Devillier P, Berger P, Zysman M, Le Rouzic O, Gonzalez-Bermejo J, Degano B, Burgel PR, Ahmed E, Roche N, Deslee G. [Take-home messages from the COPD 2021 biennial of the French Society of Respiratory Diseases. Understanding to so as to better innovate]. Rev Mal Respir 2022; 39:427-441. [PMID: 35568574 DOI: 10.1016/j.rmr.2022.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The first COPD biennial organized by the French Society of Respiratory Diseases (SPLF) took place on 17 December 2021. STATE OF THE ART The objective of the biennial was to discuss current knowledge regarding COPD pathophysiology, current treatments, research development, and future therapeutic approaches. PERSPECTIVES The different lecturers laid emphasis on the complexity of pathophysiologic mechanisms including bronchial, bronchiolar and parenchymal alterations, and also dwelt on the role of microbiota composition in COPD pathenogenesis. They pointed out that addition to inhaled treatments, ventilatory support and endoscopic approaches have been increasingly optimized. The development of new therapeutic pathways such as biotherapy and cell therapy (stem cells…) call for further exploration. CONCLUSIONS The dynamism of COPD research was repeatedly underlined, and needs to be further reinforced, the objective being to "understand so as to better innovate" so as to develop effective new strategies for treatment and management of COPD.
Collapse
Affiliation(s)
- J Ancel
- Inserm UMRS-1250, service de pneumologie, université Reims Champagne Ardenne, hôpital Maison Blanche, Reims, France
| | - M Guecamburu
- Service des maladies respiratoires, hôpital du Haut-Lévêque, CHU de Bordeaux, Bordeaux, France
| | - V Marques Da Silva
- Inserm U955, FHU SENEC, université Paris-Est Créteil, institut Mondor de recherche biomédicale, équipe GEIC2O, Créteil, France
| | - P Schilfarth
- Service des maladies respiratoires, hôpital du Haut-Lévêque, CHU de Bordeaux, Bordeaux, France; Inserm U1045, centre de recherche cardio-thoracique de Bordeaux, Pessac, France
| | - L Boyer
- Département de physiologie-explorations fonctionnelles, université Paris-Est, hôpital Henri-Mondor, AP-HP, UMR S955, FHU SENEC, UPEC, Créteil, France
| | - C Pilette
- Département de pneumologie, université catholique de Louvain, cliniques universitaires Saint-Luc et institut de recherche expérimentale et clinique, Bruxelles, Belgique
| | - C Martin
- Inserm U1016, service de pneumologie, AP-HP Paris, hôpital Cochin et institut Cochin, université de Paris, Paris, France
| | - P Devillier
- Département des maladies respiratoires, unité de recherche en pharmacologie respiratoire, VIM Suresnes (UMR 0892, université Paris-Saclay), hôpital Foch, Suresnes, France
| | - P Berger
- Service d'exploration fonctionnelle respiratoire, département de pharmacologie, centre de recherche cardiothoracique, U1045, CIC 1401, Pessac, France
| | - M Zysman
- Service des maladies respiratoires, hôpital du Haut-Lévêque, CHU de Bordeaux, Bordeaux, France; Inserm U1045, centre de recherche cardio-thoracique de Bordeaux, Pessac, France
| | - O Le Rouzic
- Inserm, CIIL Center for infection and immunity of Lille, université de Lille, CHU de Lille, pneumologie et immuno-allergologie, Institut Pasteur de Lille, U1019 - UMR9017, Lille, France
| | - J Gonzalez-Bermejo
- Inserm, UMRS115 neurophysiologie respiratoire expérimentale et clinique, service de pneumologie, médecine intensive et réanimation (département R3S), Sorbonne université, groupe hospitalier universitaire AP-HP, Sorbonne Université, site Pitié-Salpêtrière, Paris, France
| | - B Degano
- Inserm 1042, service de pneumologie physiologie, CHU de Grenoble, Grenoble, France
| | - P-R Burgel
- Inserm U1016, service de pneumologie, AP-HP Paris, hôpital Cochin et institut Cochin, université de Paris, Paris, France
| | - E Ahmed
- Département des maladies respiratoires, IRMB, université de Montpellier, CHU de Montpellier, Montpellier, France
| | - N Roche
- Inserm U1016, service de pneumologie, AP-HP Paris, hôpital Cochin et institut Cochin, université de Paris, Paris, France
| | - G Deslee
- Inserm UMRS-1250, service de pneumologie, université Reims Champagne Ardenne, hôpital Maison Blanche, Reims, France.
| |
Collapse
|
13
|
Kanti MM, Striessnig-Bina I, Wieser BI, Schauer S, Leitinger G, Eichmann TO, Schweiger M, Winkler M, Winter E, Lana A, Kufferath I, Marsh LM, Kwapiszewska G, Zechner R, Hoefler G, Vesely PW. Adipose triglyceride lipase-mediated lipid catabolism is essential for bronchiolar regeneration. JCI Insight 2022; 7:e149438. [PMID: 35349484 PMCID: PMC9090255 DOI: 10.1172/jci.insight.149438] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 03/23/2022] [Indexed: 01/18/2023] Open
Abstract
The lung airways are constantly exposed to inhaled toxic substances, resulting in cellular damage that is repaired by local expansion of resident bronchiolar epithelial club cells. Disturbed bronchiolar epithelial damage repair lies at the core of many prevalent lung diseases, including chronic obstructive pulmonary disease, asthma, pulmonary fibrosis, and lung cancer. However, it is still not known how bronchiolar club cell energy metabolism contributes to this process. Here, we show that adipose triglyceride lipase (ATGL), the rate-limiting enzyme for intracellular lipolysis, is critical for normal club cell function in mice. Deletion of the gene encoding ATGL, Pnpla2 (also known as Atgl), induced substantial triglyceride accumulation, decreased mitochondrial numbers, and decreased mitochondrial respiration in club cells. This defect manifested as bronchiolar epithelial thickening and increased airway resistance under baseline conditions. After naphthalene‑induced epithelial denudation, a regenerative defect was apparent. Mechanistically, dysfunctional PPARα lipid-signaling underlies this phenotype because (a) ATGL was needed for PPARα lipid-signaling in regenerating bronchioles and (b) administration of the specific PPARα agonist WY14643 restored normal bronchiolar club cell ultrastructure and regenerative potential. Our data emphasize the importance of the cellular energy metabolism for lung epithelial regeneration and highlight the significance of ATGL-mediated lipid catabolism for lung health.
Collapse
Affiliation(s)
- Manu Manjunath Kanti
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Isabelle Striessnig-Bina
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Beatrix Irene Wieser
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Silvia Schauer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- BioTechMed-Graz, Graz, Austria
- Division of Cell Biology, Histology, and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Thomas O. Eichmann
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Core Facility Mass Spectrometry, Medical University of Graz, Graz, Austria
| | - Martina Schweiger
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Margit Winkler
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biotechnology, NAWI Graz, Graz University of Technology, Graz, Austria
| | - Elke Winter
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Andrea Lana
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Iris Kufferath
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Leigh Matthew Marsh
- BioTechMed-Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- BioTechMed-Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Giessen, Germany
| | - Rudolf Zechner
- BioTechMed-Graz, Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Paul Willibald Vesely
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
14
|
Ruysseveldt E, Martens K, Steelant B. Airway Basal Cells, Protectors of Epithelial Walls in Health and Respiratory Diseases. FRONTIERS IN ALLERGY 2021; 2:787128. [PMID: 35387001 PMCID: PMC8974818 DOI: 10.3389/falgy.2021.787128] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
The airway epithelium provides a critical barrier to the outside environment. When its integrity is impaired, epithelial cells and residing immune cells collaborate to exclude pathogens and to heal tissue damage. Healing is achieved through tissue-specific stem cells: the airway basal cells. Positioned near the basal membrane, airway basal cells sense and respond to changes in tissue health by initiating a pro-inflammatory response and tissue repair via complex crosstalks with nearby fibroblasts and specialized immune cells. In addition, basal cells have the capacity to learn from previous encounters with the environment. Inflammation can indeed imprint a certain memory on basal cells by epigenetic changes so that sensitized tissues may respond differently to future assaults and the epithelium becomes better equipped to respond faster and more robustly to barrier defects. This memory can, however, be lost in diseased states. In this review, we discuss airway basal cells in respiratory diseases, the communication network between airway basal cells and tissue-resident and/or recruited immune cells, and how basal cell adaptation to environmental triggers occurs.
Collapse
Affiliation(s)
- Emma Ruysseveldt
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Katleen Martens
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Brecht Steelant
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Head and Neck Surgery, Department of Otorhinolaryngology, University of Crete School of Medicine, Heraklion, Greece
| |
Collapse
|
15
|
Parsons RF, Baquerizo A, Kirchner VA, Malek S, Desai CS, Schenk A, Finger EB, Brennan TV, Parekh KR, MacConmara M, Brayman K, Fair J, Wertheim JA. Challenges, highlights, and opportunities in cellular transplantation: A white paper of the current landscape. Am J Transplant 2021; 21:3225-3238. [PMID: 34212485 DOI: 10.1111/ajt.16740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023]
Abstract
Although cellular transplantation remains a relatively small field compared to solid organ transplantation, the prospects for advancement in basic science and clinical care remain bountiful. In this review, notable historical events and the current landscape of the field of cellular transplantation are reviewed with an emphasis on islets (allo- and xeno-), hepatocytes (including bioartificial liver), adoptive regulatory immunotherapy, and stem cells (SCs, specifically endogenous organ-specific and mesenchymal). Also, the nascent but rapidly evolving field of three-dimensional bioprinting is highlighted, including its major processing steps and latest achievements. To reach its full potential where cellular transplants are a more viable alternative than solid organ transplants, fundamental change in how the field is regulated and advanced is needed. Greater public and private investment in the development of cellular transplantation is required. Furthermore, consistent with the call of multiple national transplant societies for allo-islet transplants, the oversight of cellular transplants should mirror that of solid organ transplants and not be classified under the unsustainable, outdated model that requires licensing as a drug with the Food and Drug Administration. Cellular transplantation has the potential to bring profound benefit through progress in bioengineering and regenerative medicine, limiting immunosuppression-related toxicity, and providing markedly reduced surgical morbidity.
Collapse
Affiliation(s)
- Ronald F Parsons
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia
| | - Angeles Baquerizo
- Scripps Center for Cell and Organ Transplantation, La Jolla, California
| | - Varvara A Kirchner
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Sayeed Malek
- Division of Transplant Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chirag S Desai
- Division of Transplantation, Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Austin Schenk
- Division of Transplantation, Department of Surgery, Ohio State University, Columbus, Ohio
| | - Erik B Finger
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Todd V Brennan
- Department of Surgery, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kalpaj R Parekh
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Malcolm MacConmara
- Division of Surgical Transplantation, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kenneth Brayman
- Division of Transplantation, Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Jeffrey Fair
- Division of Transplant Surgery, Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Jason A Wertheim
- Departments of Surgery and Biomedical Engineering, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
16
|
Role of Extracellular Vesicles in Compromising Cellular Resilience to Environmental Stressors. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9912281. [PMID: 34337063 PMCID: PMC8321721 DOI: 10.1155/2021/9912281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs), like exosomes, are nanosized membrane-enveloped vesicles containing different bioactive cargo, such as proteins, lipids, mRNA, miRNA, and other small regulatory RNAs. Cell-derived EVs, including EVs originating from stem cells, may capture components from damaged cells or cells impacted by therapeutic treatments. Interestingly, EVs derived from stem cells can be preconditioned to produce and secrete EVs with different therapeutic properties, particularly with respect to heat-shock proteins and other molecular cargo contents. This behavior is consistent with stem cells that also respond differently to various microenvironments. Heat-shock proteins play roles in cellular protection and mediate cellular resistance to radiotherapy, chemotherapy, and heat shock. This review highlights the possible roles EVs play in mediating cellular plasticity and survival when exposed to different physical and chemical stressors, with a special focus on the respiratory distress due to the air pollution.
Collapse
|
17
|
Xiong R, Wu Y, Wu Q, Muskhelishvili L, Davis K, Tripathi P, Chen Y, Chen T, Bryant M, Rosenfeldt H, Healy SM, Cao X. Integration of transcriptome analysis with pathophysiological endpoints to evaluate cigarette smoke toxicity in an in vitro human airway tissue model. Arch Toxicol 2021; 95:1739-1761. [PMID: 33660061 PMCID: PMC8113308 DOI: 10.1007/s00204-021-03008-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/16/2021] [Indexed: 01/04/2023]
Abstract
Exposure to cigarette smoke (CS) is a known risk factor in the pathogenesis of smoking-caused diseases, such as chronic obstructive pulmonary diseases (COPD) and lung cancer. To assess the effects of CS on the function and phenotype of airway epithelial cells, we developed a novel repeated treatment protocol and comprehensively evaluated the progression of key molecular, functional, and structural abnormalities induced by CS in a human in vitro air-liquid-interface (ALI) airway tissue model. Cultures were exposed to CS (diluted with 0.5 L/min, 1.0 L/min, and 4.0 L/min clean air) generated from smoking five 3R4F University of Kentucky reference cigarettes under the International Organization for Standardization (ISO) machine smoking regimen, every other day for 4 weeks (3 days per week, 40 min/day). By integrating the transcriptomics-based approach with the in vitro pathophysiological measurements, we demonstrated CS-mediated effects on oxidative stress, pro-inflammatory cytokines and matrix metalloproteinases (MMPs), ciliary function, expression and secretion of mucins, and squamous cell differentiation that are highly consistent with abnormalities observed in airways of smokers. Enrichment analysis on the transcriptomic profiles of the ALI cultures revealed key molecular pathways, such as xenobiotic metabolism, oxidative stress, and inflammatory responses that were perturbed in response to CS exposure. These responses, in turn, may trigger aberrant tissue remodeling, eventually leading to the onset of respiratory diseases. Furthermore, changes of a panel of genes known to be disturbed in smokers with COPD were successfully reproduced in the ALI cultures exposed to CS. In summary, findings from this study suggest that such an integrative approach may be a useful tool for identifying genes and adverse cellular events caused by inhaled toxicants, like CS.
Collapse
Affiliation(s)
- Rui Xiong
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yue Wu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR, 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hans Rosenfeldt
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Sheila M Healy
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
18
|
Wang Z, Liang W, Ma C, Wang J, Gao X, Wei L. Macrophages Inhibit Ciliary Protein Levels by Secreting BMP-2 Leading to Airway Epithelial Remodeling Under Cigarette Smoke Exposure. Front Mol Biosci 2021; 8:663987. [PMID: 33981724 PMCID: PMC8107431 DOI: 10.3389/fmolb.2021.663987] [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: 02/04/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory disease with high morbidity and mortality worldwide. So far, smoking is still its leading cause. The characteristics of COPD are emphysema and airway remodeling, as well as chronic inflammation, which were predominated by macrophages. Some studies have reported that macrophages were involved in emphysema and chronic inflammation, but whether there is a link between airway remodeling and macrophages remains unclear. In this study, we found that both acute and chronic cigarette smoke exposure led to an increase of macrophages in the lung and a decrease of ciliated cells in the airway epithelium of a mouse model. The results of in vitro experiments showed that the ciliary protein (β-tubulin-IV) levels of BEAS-2B cells could be inhibited when co-cultured with human macrophage line THP-1, and the inhibitory effect was augmented with the stimulation of cigarette smoke extract (CSE). Based on the results of transcriptome sequencing, we focused on the protein, bone morphogenetic protein-2 (BMP-2), secreted by the macrophage, which might mediate this inhibitory effect. Further studies confirmed that BMP-2 protein inhibited β-tubulin-IV protein levels of BEAS-2B cells under the stimulation of CSE. Coincidentally, this inhibitory effect could be nearly blocked by the BMP receptor inhibitor, LDN, or could be interfered with BMP-2 siRNA. This study suggests that activation and infiltration of macrophages in the lung induced by smoke exposure lead to a high expression of BMP-2, which in turn inhibits the ciliary protein levels of the bronchial epithelial cells, contributing to the remodeling of airway epithelium, and aggravates the development of COPD.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China.,Department of Intensive Care Unit, Hebei General Hospital, Shijiazhuang, China
| | - Wenzhang Liang
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Cuiqing Ma
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Jiachao Wang
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Xue Gao
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| | - Lin Wei
- Department of Immunology, Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
19
|
Ben Brahim C, Courageux C, Jolly A, Ouine B, Cartier A, de la Grange P, de Koning L, Leroy P. Proliferation Genes Repressed by TGF-β Are Downstream of Slug/Snail2 in Normal Bronchial Epithelial Progenitors and Are Deregulated in COPD. Stem Cell Rev Rep 2021; 17:703-718. [PMID: 33495975 DOI: 10.1007/s12015-021-10123-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/16/2022]
Abstract
Slug/Snail2 belongs to the Epithelial-Mesenchymal Transition (EMT)-inducing transcription factors involved in development and diseases. Slug is expressed in adult stem/progenitor cells of several epithelia, making it unique among these transcription factors. To investigate Slug role in human bronchial epithelium progenitors, we studied primary bronchial basal/progenitor cells in an air-liquid interface culture system that allows regenerating a bronchial epithelium. To identify Slug downstream genes we knocked down Slug in basal/progenitor cells from normal subjects and subjects with COPD, a respiratory disease presenting anomalies in the bronchial epithelium and high levels of TGF-β in the lungs. We show that normal and COPD bronchial basal/progenitors, even when treated with TGF-β, express both epithelial and mesenchymal markers, and that the epithelial marker E-cadherin is not a target of Slug and, moreover, positively correlates with Slug. We reveal that Slug downstream genes responding to both differentiation and TGF-β are different in normal and COPD progenitors, with in particular a set of proliferation-related genes that are among the genes repressed downstream of Slug in normal but not COPD. In COPD progenitors at the onset of differentiation in presence of TGF-β,we show that there is positive correlations between the effect of differentiation and TGF-β on proliferation-related genes and on Slug protein, and that their expression levels are higher than in normal cells. As well, the expression of Smad3 and β-Catenin, two molecules from TGF-βsignaling pathways, are higher in COPD progenitors, and our results indicate that proliferation-related genes and Slug protein are increased by different TGF-β-induced mechanisms.
Collapse
Affiliation(s)
- Chamseddine Ben Brahim
- INSERM UMR1152, Physiopathology and Epidemiology of Respiratory Diseases, Paris, France
- Faculty of Medicine, Paris Diderot University, Bichat Campus, Paris, France
| | - Charlotte Courageux
- INSERM UMR1152, Physiopathology and Epidemiology of Respiratory Diseases, Paris, France
- Faculty of Medicine, Paris Diderot University, Bichat Campus, Paris, France
| | | | - Bérengère Ouine
- Institut Curie, Department of Translational Research, RPPA platform, PSL Research University, Paris, France
| | - Aurélie Cartier
- Institut Curie, Department of Translational Research, RPPA platform, PSL Research University, Paris, France
| | | | - Leanne de Koning
- Institut Curie, Department of Translational Research, RPPA platform, PSL Research University, Paris, France
| | - Pascale Leroy
- INSERM UMR1152, Physiopathology and Epidemiology of Respiratory Diseases, Paris, France.
- Faculty of Medicine, Paris Diderot University, Bichat Campus, Paris, France.
| |
Collapse
|
20
|
Pineau F, Shumyatsky G, Owuor N, Nalamala N, Kotnala S, Bolla S, Marchetti N, Kelsen S, Criner GJ, Sajjan US. Microarray analysis identifies defects in regenerative and immune response pathways in COPD airway basal cells. ERJ Open Res 2020; 6:00656-2020. [PMID: 33313308 PMCID: PMC7720690 DOI: 10.1183/23120541.00656-2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 01/07/2023] Open
Abstract
Background Airway basal cells are specialised stem cells and regenerate airway epithelium. Airway basal cells isolated from patients with COPD regenerate airway epithelium with an abnormal phenotype. We performed gene expression analysis to gain insights into the defective regenerative programme in COPD basal cells. Methods We conducted microarray analysis and compared COPD versus normal basal cells to identify differentially regulated genes (DEGs) and the enriched biological pathways. We determined the correlation of DEGs with cell polarisation and markers of ciliated and goblet cells. HOXB2 was knocked down in 16HBE14o− cells and monitored for polarisation of cells. HOXB2 expression in the lung sections was determined by immunofluorescence. Results Comparison of normal and COPD basal cell transcriptomic profiles highlighted downregulation of genes associated with tissue development, epithelial cell differentiation and antimicrobial humoral response. Expression of one of the tissue development genes, HOXB2 showed strong correlation with transepithelial resistance and this gene was downregulated in COPD basal cells. Knockdown of HOXB2, abrogated polarisation of epithelial cells in normal cells. Finally, HOXB2 expression was substantially reduced in the bronchial epithelium of COPD patients. Conclusions Defect in gene signatures involved in tissue development and epithelial differentiation were implicated in COPD basal cells. One of the tissue developmental genes, HOXB2, is substantially reduced in bronchial epithelium of COPD patients. Since HOXB2 contributes to airway epithelial cell polarisation, we speculate that reduced expression of HOXB2 in COPD may contribute to abnormal airway epithelial regeneration in COPD. COPD airway basal cells show downregulation of gene sets that are involved in intercellular junctions, epithelial differentiation and immune responses, highlighting the possible mechanisms of defective airway epithelial repair in COPDhttps://bit.ly/3kneloj
Collapse
Affiliation(s)
- Fanny Pineau
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | | | - Nicole Owuor
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Nisha Nalamala
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Sudhir Kotnala
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Sudhir Bolla
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Nathaniel Marchetti
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Steven Kelsen
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Gerard J Criner
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Uma S Sajjan
- Dept of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA.,Dept of Physiology, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| |
Collapse
|
21
|
Rayner RE, Makena P, Prasad GL, Cormet-Boyaka E. Cigarette smoke preparations, not electronic nicotine delivery system preparations, induce features of lung disease in a 3D lung repeat-dose model. Am J Physiol Lung Cell Mol Physiol 2020; 320:L276-L287. [PMID: 33207918 DOI: 10.1152/ajplung.00452.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cigarette smoking is a risk factor for several lung diseases, including chronic obstructive pulmonary disease, cardiovascular disease, and lung cancer. The potential health effects of chronic use of electronic nicotine delivery systems (ENDS) is unclear. This study utilized fully differentiated primary normal human bronchial epithelial (NHBE) cultures in a repeat-dose exposure to evaluate and compare the effect of combustible cigarette and ENDS preparations. We show that 1-h daily exposure of NHBE cultures over a 10-day period to combustible cigarette whole smoke-conditioned media (WS-CM) increased expression of oxidative stress markers, cell proliferation, airway remodeling, and cellular transformation markers and decreased mucociliary function including ion channel function and airway surface liquid. Conversely, aerosol conditioned media (ACM) from ENDS with similar nicotine concentration (equivalent-nicotine units) as WS-CM and nicotine alone had no effect on those parameters. In conclusion, primary NHBE cultures in a repeat-dose exposure system represent a good model to assess the features of lung disease. This study also reveals that cigarette and ENDS preparations differentially elicit several key endpoints, some of which are potential biomarkers for lung cancer or chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Rachael E Rayner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | | | - G L Prasad
- RAI Services Company, Winston-Salem, North Carolina
| | | |
Collapse
|
22
|
Osan JK, Talukdar SN, Feldmann F, DeMontigny BA, Jerome K, Bailey KL, Feldmann H, Mehedi M. Goblet Cell Hyperplasia Increases SARS-CoV-2 Infection in COPD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.11.11.379099. [PMID: 33200131 PMCID: PMC7668735 DOI: 10.1101/2020.11.11.379099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 has become a major problem across the globe, with approximately 50 million cases and more than 1 million deaths and currently no approved treatment or vaccine. Chronic obstructive pulmonary disease (COPD) is one of the underlying conditions in adults of any age that place them at risk for developing severe illness associated with COVID-19. We established an airway epithelium model to study SARS-CoV-2 infection in healthy and COPD lung cells. We found that both the entry receptor ACE2 and the co-factor transmembrane protease TMPRSS2 are expressed at higher levels on nonciliated goblet cell, a novel target for SARS-CoV-2 infection. We observed that SARS-CoV-2 infected goblet cells and induced syncytium formation and cell sloughing. We also found that SARS-CoV-2 replication was increased in the COPD airway epithelium likely due to COPD associated goblet cell hyperplasia. Our results reveal goblet cells play a critical role in SARS-CoV-2 infection in the lung.
Collapse
Affiliation(s)
- Jaspreet K. Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA
- Contributed equally to this study
| | - Sattya N. Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA
- Contributed equally to this study
| | - Friederike Feldmann
- Divison of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Beth Ann DeMontigny
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA
| | - Kailey Jerome
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA
| | - Kristina L. Bailey
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Heinz Feldmann
- Divison of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58202, USA
- Lead contact
| |
Collapse
|
23
|
Chen M, Shen W, Rowan NR, Kulaga H, Hillel A, Ramanathan M, Lane AP. Elevated ACE-2 expression in the olfactory neuroepithelium: implications for anosmia and upper respiratory SARS-CoV-2 entry and replication. Eur Respir J 2020; 56:13993003.01948-2020. [PMID: 32817004 PMCID: PMC7439429 DOI: 10.1183/13993003.01948-2020] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023]
Abstract
The ongoing outbreak of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a major threat to global health [1]. The mechanism of cellular entry by SARS-CoV-2 is through binding to angiotensin-converting enzyme 2 (ACE-2) [2, 3], a metalloproteinase ectoenzyme that primarily functions in the regulation of angiotensin II, but also has non-catalytic roles such as intestinal neutral amino acid transport. The level of ACE-2 protein and its subcellular localisation in the respiratory tract may be a key determinant of susceptibility to infection, symptoms and outcomes in COVID-19. In humans, ACE-2 protein is broadly expressed in the lung, kidney and small intestine [4]. Pathological analysis of COVID-19 post mortem samples shows substantial damage in the lung [5], suggesting that the airway is the principal entry and target of SARS-CoV-2. However, analysis of multiple single cell RNA-seq datasets reveal overall low ACE-2 RNA transcription in nasal airway epithelium, with further reduced expression in lower airway club cells and rare expression in alveolar epithelial cells [6]. This pattern of ACE-2 expression provides evidence that the upper, rather than the lower, airway is the initial site of SARS-CoV-2 infection. ACE2 protein is expressed at high levels in the human olfactory epithelium relative to upper airway epithelial cells. This may explain COVID-19-associated olfactory dysfunction, and suggests a SARS-CoV-2 reservoir site and potential intranasal therapy.https://bit.ly/3hxT0qm
Collapse
Affiliation(s)
- Mengfei Chen
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,M. Chen and W. Shen contributed equally to this article.,M. Chen and A.P. Lane contributed equally to this article as lead authors and supervised the work
| | - Wenjuan Shen
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,M. Chen and W. Shen contributed equally to this article
| | - Nicholas R Rowan
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heather Kulaga
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Hillel
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Murugappan Ramanathan
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew P Lane
- Dept of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA .,M. Chen and A.P. Lane contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
24
|
Brommel CM, Cooney AL, Sinn PL. Adeno-Associated Virus-Based Gene Therapy for Lifelong Correction of Genetic Disease. Hum Gene Ther 2020; 31:985-995. [PMID: 32718227 PMCID: PMC7495917 DOI: 10.1089/hum.2020.138] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
The list of successful gene therapy trials using adeno-associated virus (AAV)-based vectors continues to grow and includes a wide range of monogenic diseases. Replication incompetent AAV genomes typically remain episomal and expression dilutes as cells divide and die. Consequently, long-term transgene expression from AAV is best suited for quiescent cell types, such as retinal cells, myocytes, or neurons. For genetic diseases that involve cells with steady turnover, AAV-conferred correction may require routine readministration, where every dose carries the risk of developing an adaptive immune response that renders treatment ineffective. Here, we discuss innovative approaches to permanently modify the host genome using AAV-based platforms, thus potentially requiring only a single dose. Such approaches include using AAV delivery of DNA transposons, homologous recombination templates into safe harbors, and nucleases for targeting integration. In tissues with continual cell turnover, genetic modification of progenitor cell populations will help ensure persistent therapeutic outcomes. Combining the safety profile of AAV-based gene therapy vectors with the ability to integrate a therapeutic transgene creates novel solutions to the challenge of lifelong curative treatments for human genetic diseases.
Collapse
Affiliation(s)
| | - Ashley L. Cooney
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Patrick L. Sinn
- Program in Molecular Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
25
|
Kikuchi T, Shimizu T. Thickness-wise growth technique for human articular chondrocytes to fabricate three-dimensional cartilage grafts. Regen Ther 2020; 14:119-127. [PMID: 32055650 PMCID: PMC7005340 DOI: 10.1016/j.reth.2019.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/02/2019] [Accepted: 12/03/2019] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Cutting the cost of manufacturing is important for extending the use of tissue-engineered therapeutic products. The present study aimed to develop a simple method for fabrication of cartilaginous tissues for regenerative therapy, utilizing the phenomenon where human articular chondrocytes grow thickness-wise and spontaneously form three-dimensionally thick tissues. METHODS Normal human articular chondrocytes (NHACs) were cultured with varying concentrations of transforming growth factor beta 1 (TGF-β1) and/or fibroblast growth factor-2 (FGF-2) to optimize the culture condition for thickness-wise growth of chondrocytes. Next, the tissues grown in the optimal condition were subjected to re-differentiation culture in attached and detached states to assess differentiation capacity by evaluating secreted factors, histological analysis, and a gene expression assay. RESULTS NHACs grew thickness-wise efficiently in the presence of 1 ng/mL TGF-β1 and 10 ng/mL FGF-2. After two weeks of culture, NHACs grew with 11-fold higher thickness and 16-fold higher cell number compared to cells which were neither treated with TGF-β1 nor with FGF-2. These thickness-wise-grown chondrocytes could be re-differentiated by a differentiation medium according to the increase in melanoma inhibitory activity (MIA) and positive safranin-O staining. Interestingly, the cartilaginous gene expression was considerably different between the attached and detached conditions even in the same culture medium, indicating the necessity of detachment and shrinkage to achieve further differentiation. CONCLUSIONS Spontaneous thickness-wise growth might provide a simple tissue-engineering method for manufacturing cartilaginous 3D tissues.
Collapse
Affiliation(s)
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
26
|
Chen M, Shen W, Rowan NR, Kulaga H, Hillel A, Ramanathan M, Lane AP. Elevated ACE2 expression in the olfactory neuroepithelium: implications for anosmia and upper respiratory SARS-CoV-2 entry and replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32511390 PMCID: PMC7263519 DOI: 10.1101/2020.05.08.084996] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The site of SARS-CoV-2 entry and replication critically impacts strategies for COVID-19 diagnosis, transmission mitigation, and treatment. We determined the cellular location of the SARS-CoV-2 target receptor protein, ACE2, in the human upper airway, finding striking enrichment (200–700 folds) in the olfactory neuroepithelium relative to nasal respiratory or tracheal epithelial cells. This cellular tropism of SARS-CoV-2 may underlie its high transmissibility and association with olfactory dysfunction, while suggesting a viral reservoir potentially amenable to intranasal therapy.
Collapse
Affiliation(s)
- Mengfei Chen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Wenjuan Shen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nicholas R Rowan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Heather Kulaga
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alexander Hillel
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Murugappan Ramanathan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew P Lane
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
27
|
Gouveia L, Kraut S, Hadzic S, Vazquéz-Liébanas E, Kojonazarov B, Wu CY, Veith C, He L, Mermelekas G, Schermuly RT, Weissmann N, Betsholtz C, Andrae J. Lung developmental arrest caused by PDGF-A deletion: consequences for the adult mouse lung. Am J Physiol Lung Cell Mol Physiol 2020; 318:L831-L843. [PMID: 32186397 DOI: 10.1152/ajplung.00295.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PDGF-A is a key contributor to lung development in mice. Its expression is needed for secondary septation of the alveoli and deletion of the gene leads to abnormally enlarged alveolar air spaces in mice. In humans, the same phenotype is the hallmark of bronchopulmonary dysplasia (BPD), a disease that affects premature babies and may have long lasting consequences in adulthood. So far, the knowledge regarding adult effects of developmental arrest in the lung is limited. This is attributable to few follow-up studies of BPD survivors and lack of good experimental models that could help predict the outcomes of this early age disease for the adult individual. In this study, we used the constitutive lung-specific Pdgfa deletion mouse model to analyze the consequences of developmental lung defects in adult mice. We assessed lung morphology, physiology, cellular content, ECM composition and proteomics data in mature mice, that perinatally exhibited lungs with a BPD-like morphology. Histological and physiological analyses both revealed that enlarged alveolar air spaces remained until adulthood, resulting in higher lung compliance and higher respiratory volume in knockout mice. Still, no or only small differences were seen in cellular, ECM and protein content when comparing knockout and control mice. Taken together, our results indicate that Pdgfa deletion-induced lung developmental arrest has consequences for the adult lung at the morphological and functional level. In addition, these mice can reach adulthood with a BPD-like phenotype, which makes them a robust model to further investigate the pathophysiological progression of the disease and test putative regenerative therapies.
Collapse
Affiliation(s)
- Leonor Gouveia
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Simone Kraut
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Elisa Vazquéz-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Baktybek Kojonazarov
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Cheng-Yu Wu
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christine Veith
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Georgios Mermelekas
- Cancer Proteomics Mass Spectrometry, Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ralph Theo Schermuly
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Norbert Weissmann
- Justus-Liebig University of Giessen (JLUG), Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.,Integrated Cardio Metabolic Centre, Karolinska Institutet, Huddinge, Sweden
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
[Modelling the bronchial epithelium in chronic obstructive pulmonary disease using human induced pluripotential stem cells]. Rev Mal Respir 2020; 37:197-200. [PMID: 32146059 DOI: 10.1016/j.rmr.2020.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 01/12/2020] [Indexed: 11/24/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease leading to irreversible destruction of the terminal bronchioles. Although the precise patho-physiological mechanisms remain to be elucidated, the bronchial epithelium seems to play a pivotal role in the disease. Recent studies have highlighted a great heterogeneity among COPD patients, with various disease courses including, in about half the cases, an origin in childhood. Modelling of COPD is a major goal but currently available models are imperfect. Our work aims to create a new in vitro cellular model to study the pathology of the disease. The differentiation of human induced pluripotential stem cells (hiPSCs) in bronchial epithelium is a step towards a better understanding of the developmental origin and the identification of new therapeutic targets.
Collapse
|
29
|
Barbry P, Cavard A, Chanson M, Jaffe AB, Plasschaert LW. Regeneration of airway epithelial cells to study rare cell states in cystic fibrosis. J Cyst Fibros 2020; 19 Suppl 1:S42-S46. [DOI: 10.1016/j.jcf.2019.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 01/09/2023]
|
30
|
Basnet S, Bochkov YA, Brockman-Schneider RA, Kuipers I, Aesif SW, Jackson DJ, Lemanske RF, Ober C, Palmenberg AC, Gern JE. CDHR3 Asthma-Risk Genotype Affects Susceptibility of Airway Epithelium to Rhinovirus C Infections. Am J Respir Cell Mol Biol 2019; 61:450-458. [PMID: 30916989 PMCID: PMC6775945 DOI: 10.1165/rcmb.2018-0220oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
CDHR3 (cadherin-related family member 3) is a transmembrane protein that is highly expressed in airway epithelia and the only known receptor for rhinovirus C (RV-C). A CDHR3 SNP (rs6967330) with G to A base change has been linked to severe exacerbations of asthma and increased susceptibility to RV-C infections in young children. The goals of this study were to determine the subcellular localization of CDHR3 and to test the hypothesis that CDHR3 asthma-risk genotype affects epithelial cell function and susceptibility to RV-C infections of the airway epithelia. We used immunofluorescence imaging, Western blot analysis, and transmission electron microscopy to show CDHR3 subcellular localization in apical cells, including expression in the cilia of airway epithelia. Polymorphisms in CDHR3 rs6967330 locus (G→A) that were previously associated with childhood asthma were related to differences in CDHR3 expression and epithelial cell function. The rs6967330 A allele was associated with higher overall protein expression and RV-C binding and replication compared with the rs6967330 G allele. Furthermore, the rs6967330 A allele was associated with earlier ciliogenesis and higher FOXJ1 expression. Finally, CDHR3 genotype had no significant effects on membrane integrity or ciliary beat function. These findings provide information on the subcellular localization and possible functions of CDHR3 in the airways and link CDHR3 asthma-risk genotype to increased RV-C binding and replication.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Carol Ober
- Department of Human Genetics, University of Chicago, Chicago, Illinois
| | - Ann C. Palmenberg
- Institute of Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin; and
| | | |
Collapse
|
31
|
Widdicombe JH. Early studies on the surface epithelium of mammalian airways. Am J Physiol Lung Cell Mol Physiol 2019; 317:L486-L495. [PMID: 31313615 DOI: 10.1152/ajplung.00240.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
This article traces the beginnings of the various areas of physiological research on airway epithelium. First mentioned in 1600, it was not until 1834 that it was found to be ciliated. Goblet and basal cells were described in 1852, to be followed by ~10 other epithelial cell types (the most recent in 2018). It also contains nerve endings and resident leukocytes. Mucociliary clearance was documented in 1835, but the first studies on the ciliary beat cycle did not appear until 1890, and a definitive description was not published until 1981. It was established in 1932 that goblet cells in the cat trachea were unresponsive to cholinergic agents; but only since 1980 or so has any significant progress been made on what does cause them to degranulate. Active transfer of salts across epithelia creates local osmotic gradients that drive transepithelial water flows. Vectorial salt transport was first described for airway epithelium in 1968, and the associated volume flows were measured in 1981. Evidence that airway epithelium releases signaling molecules first appeared in 1981. Since then, scores of molecules have been identified. The pace of research in most areas increased dramatically after the development of confluent, polarized cultures of airway epithelium in the early 1980s.
Collapse
Affiliation(s)
- Jonathan H Widdicombe
- Department of Physiology and Membrane Biology, University of California, Davis, California
| |
Collapse
|
32
|
Li N, Cooney AL, Zhang W, Ehrhardt A, Sinn PL. Enhanced Tropism of Species B1 Adenoviral-Based Vectors for Primary Human Airway Epithelial Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:228-236. [PMID: 31417941 PMCID: PMC6690641 DOI: 10.1016/j.omtm.2019.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/01/2019] [Indexed: 12/04/2022]
Abstract
Adenoviruses are efficient vehicles for transducing airway epithelial cells. Human adenoviruses (Ads) are classified into seven species termed A–G. Most species use the coxsackie-adenovirus receptor (CAR) as a primary cellular receptor. Ad group B is notable because it is further divided into groups B1 and B2 and its members use CD46 or desmoglein 2 (DSG2) as cellular receptors. To date, human Ad types 2 and 5 have been the predominant choices for preclinical and clinical trials using Ad-based viral vectors in the airways. In this study, we screened 14 Ad types representing species C, B1, B2, D, and E. Using well-differentiated primary cultures of human airway epithelial cells (HAEs), we examined transduction efficiency. Based on GFP or nanoluciferase expression, multiple Ad types transduced HAEs as well as or better than Ad5. Ad3, Ad21, and Ad14 belong to species B and had notable transduction properties. We further examined the transduction properties of conditionally reprogrammed airway basal cells and primary basal cells from human lung donors. Again, the transduction efficiency of species B members outperformed the other types. These data suggest that adenoviral vectors based on species B transduce fully differentiated epithelial cells and progenitor cells in the human airways better than Ad5.
Collapse
Affiliation(s)
- Ni Li
- Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Ashley L Cooney
- Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| | - Wenli Zhang
- Institute of Virology and Microbiology, Department of Human Medicine, Faculty of Health, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany
| | - Anja Ehrhardt
- Institute of Virology and Microbiology, Department of Human Medicine, Faculty of Health, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58453 Witten, Germany
| | - Patrick L Sinn
- Department of Pediatrics, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
33
|
Zhang N, Wang H, Xie Q, Cao H, Wu F, Di Wu DB, Wan Y. Identification of potential diagnostic and therapeutic target genes for lung squamous cell carcinoma. Oncol Lett 2019; 18:169-180. [PMID: 31289486 PMCID: PMC6539486 DOI: 10.3892/ol.2019.10300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
The purpose of this study was to identify potential molecular markers of lung squamous cell carcinoma (LUSC). Three datasets containing LUSC mRNA sequencing data were downloaded from the Gene Expression Omnibus, The Cancer Genome Atlas and the Gene Expression Profiling Interactive Analysis databases. These datasets were used to identify significantly differentially expressed genes (DEGs) in LUSC. A protein-protein interaction network of the DEGs was constructed followed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes and overall survival analyses of the DEGs. A total of 37 DEGs between LUSC and normal tissues were identified, including 26 downregulated genes and 11 upregulated genes. Biological Process enrichment analysis revealed that the DEGs were mainly enriched in ‘cell adhesion’, ‘cell-matrix adhesion’, ‘anatomical structure morphogenesis’, ‘ECM-receptor interaction’ and ‘focal adhesion’. Overall survival analysis demonstrated that transcription factor 21, α-2-macroglobulin, acyl-CoA synthetase long chain family member 5, integrin subunit β8, meiotic nuclear divisions 1 and secretoglobin family 1A member 1 were significantly associated with the occurrence and development of lung cancer, and these genes were selected as hub genes. The results obtained in the present study may aid the elucidation of the molecular mechanisms involved in the development of LUSC and may provide potential targets for LUSC treatment.
Collapse
Affiliation(s)
- Nana Zhang
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hong Wang
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Qiqi Xie
- Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hua Cao
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Fanqi Wu
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Dan Bei Di Wu
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yixin Wan
- Department of Respiration, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
34
|
Phenotypic Analysis of BrdU Label-Retaining Cells during the Maturation of Conducting Airway Epithelium in a Porcine Lung. Stem Cells Int 2019; 2019:7043890. [PMID: 30936924 PMCID: PMC6415319 DOI: 10.1155/2019/7043890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/10/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Stem/progenitor cells have recently been demonstrated to play key roles in the maturation, injury repair, and regeneration of distinct organs or tissues. Porcine has spurred an increased interest in biomedical research models and xenotransplantation, owing to most of its organs share similarities in physiology, cellular composition and size to humans. Therefore, characterization of stem/progenitor cells in porcine organs or tissues may provide a novel avenue to better understand the biology and function of stem cells in humans. In the present study, potential stem/progenitor cells in conducting airway epithelium of a porcine lung were characterized by morphometric analysis of bromodeoxyuridine (BrdU) label-retaining cells (LRCs) during the maturation of the lung. The results showed a pseudostratified mucociliary epithelium comprised of basal, ciliated, goblet, and columnar cells in the conducting airway of a porcine lung. In addition, the majority of primary epithelial cells able to proliferate in vitro expressed keratin 5, a subpopulation of these keratin 5-positive cells, also expressed CD117 (c-Kit) or CD49f (integrin alpha 6, ITGA6), implying that they might be potential epithelial stem/progenitor cells in conducting airway of a porcine lung. Lineage tracing analysis with a BrdU-labeled neonatal piglet showed that the proportion of BrdU-labeled cells in conducting airways decreased over the 90-day period of lung maturation. The BrdU-labeled epithelial cells also expressed keratin 14, mucin 5AC, or prosurfactant protein C (ProSP-C); among them, the keratin 14-positive cells were the most frequent BrdU-labeled epithelial cell type as determined by immunohistochemical and immunofluorescence staining. This study may provide valuable information on the biology and function of epithelial stem/progenitor cells in conducting airway of pigs and humans.
Collapse
|
35
|
Xiong R, Wu Q, Trbojevich R, Muskhelishvili L, Davis K, Bryant M, Richter P, Cao X. Disease-related responses induced by cadmium in an in vitro human airway tissue model. Toxicol Lett 2019; 303:16-27. [DOI: 10.1016/j.toxlet.2018.12.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/18/2018] [Accepted: 12/16/2018] [Indexed: 01/02/2023]
|
36
|
Type VI collagen promotes lung epithelial cell spreading and wound-closure. PLoS One 2018; 13:e0209095. [PMID: 30550606 PMCID: PMC6294368 DOI: 10.1371/journal.pone.0209095] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/29/2018] [Indexed: 11/25/2022] Open
Abstract
Basement membrane (BM) is an essential part of the extracellular matrix (ECM) that plays a crucial role in mechanical support and signaling to epithelial cells during lung development, homeostasis and repair. Abnormal composition and remodeling of the lung ECM have been associated with developmental abnormalities observed in multiple pediatric and adult respiratory diseases. Collagen VI (COL6) is a well-studied muscle BM component, but its role in the lung and its effect on pulmonary epithelium is largely undetermined. We report the presence of COLVI immediately subjacent to human airway and alveolar epithelium in the pediatric lung, in a location where it is likely to interact with epithelial cells. In vitro, both primary human lung epithelial cells and human lung epithelial cell lines displayed an increased rate of “wound healing” in response to a scratch injury when plated on COL6 as compared to other matrices. For the 16HBE cell line, wounds remained >5-fold larger for cells on COL1 (p<0.001) and >6-fold larger on matrigel (p<0.001), a prototypical basement membrane, when compared to COL6 (>96% closure at 10 hr). The effect of COL6 upon lung epithelial cell phenotype was associated with an increase in cell spreading. Three hours after initial plating, 16HBE cells showed >7-fold less spreading on matrigel (p<0.01), and >4-fold less spreading on COL1 (p<0.01) when compared to COL6. Importantly, the addition of COL6 to other matrices also enhanced cell spreading. Similar responses were observed for primary cells. Inhibitor studies indicated both integrin β1 activity and activation of multiple signaling pathways was required for enhanced spreading on all matrices, with the PI3K/AKT pathway (PI3K, CDC42, RAC1) showing both significant and specific effects for spreading on COL6. Genetic gain-of-function experiments demonstrated enhanced PI3K/AKT pathway activity was sufficient to confer equivalent cell spreading on other matrices as compared to COL6. We conclude that COL6 has significant and specific effects upon human lung epithelial cell-autonomous functions.
Collapse
|
37
|
Hodges CA, Conlon RA. Delivering on the promise of gene editing for cystic fibrosis. Genes Dis 2018; 6:97-108. [PMID: 31193992 PMCID: PMC6545485 DOI: 10.1016/j.gendis.2018.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
In this review, we describe a path for translation of gene editing into therapy for cystic fibrosis (CF). Cystic fibrosis results from mutations in the CFTR gene, with one allele predominant in patient populations. This simple, genetic etiology makes gene editing appealing for treatment of this disease. There already have been success in applying this approach to cystic fibrosis in cell and animal models, although these advances have been modest in comparison to advances for other disease. Less than six years after its first demonstration in animals, CRISPR/Cas gene editing is in early clinical trials for several disorders. Most clinical trials, thus far, attempt to edit genes in cells of the blood lineages. The advantage of the blood is that the stem cells are known, can be isolated, edited, selected, expanded, and returned to the body. The likely next trials will be in the liver, which is accessible to many delivery methods. For cystic fibrosis, the biggest hurdle is to deliver editors to other, less accessible organs. We outline a path by which delivery can be improved. The translation of new therapies doesn't occur in isolation, and the development of gene editors is occurring as advances in gene therapy and small molecule therapeutics are being made. The advances made in gene therapy may help develop delivery vehicles for gene editing, although major improvements are needed. Conversely, the approval of effective small molecule therapies for many patients with cystic fibrosis will raise the bar for translation of gene editing.
Collapse
Affiliation(s)
- Craig A Hodges
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Ronald A Conlon
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
38
|
Guo L, Karoubi G, Duchesneau P, Aoki FG, Shutova MV, Rogers I, Nagy A, Waddell TK. Interrupted reprogramming of alveolar type II cells induces progenitor-like cells that ameliorate pulmonary fibrosis. NPJ Regen Med 2018; 3:14. [PMID: 30210809 PMCID: PMC6123410 DOI: 10.1038/s41536-018-0052-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 02/04/2023] Open
Abstract
We describe here an interrupted reprogramming strategy to generate “induced progenitor-like (iPL) cells” from alveolar epithelial type II (AEC-II) cells. A carefully defined period of transient expression of reprogramming factors (Oct4, Sox2, Klf4, and c-Myc (OSKM)) is able to rescue the limited in vitro clonogenic capacity of AEC-II cells, potentially by activation of a bipotential progenitor-like state. Importantly, our results demonstrate that interrupted reprogramming results in controlled expansion of cell numbers yet preservation of the differentiation pathway to the alveolar epithelial lineage. When transplanted to the injured lungs, AEC-II-iPL cells are retained in the lung and ameliorate bleomycin-induced pulmonary fibrosis. Interrupted reprogramming can be used as an alternative approach to produce highly specified functional therapeutic cell populations and may lead to significant advances in regenerative medicine. A modified reprogramming strategy helps expand populations of surfactant-producing lung cells in a dish without altering their cellular function. A team led by Thomas Waddell and Andras Nagy from the University of Toronto, Canada isolated alveolar type II cells from the lungs of mice. They transiently induced expression of four reprogramming factors in these cells for a defined period of time. Before this “interrupted” reprogramming, the lung cells had limited ability to continue replicating themselves. Afterwards, the cells could expand their numbers dramatically without entering a pluripotent state. Rather, the cells maintained their original function while also expressing genes associated with lung precursor cells, which could explain their proliferative ability. The cells, when transplanted into the injured lungs, helped ameliorate pulmonary fibrosis in a mouse model, suggesting that a similar cell-based therapy may be useful in people.
Collapse
Affiliation(s)
- Li Guo
- 1Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON Canada
| | - Golnaz Karoubi
- 1Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON Canada
| | - Pascal Duchesneau
- 1Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON Canada
| | - Fabio Gava Aoki
- 1Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON Canada
| | - Maria V Shutova
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON Canada
| | - Ian Rogers
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON Canada.,3Department of Physiology, University of Toronto, Toronto, ON Canada.,4Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON Canada
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON Canada.,4Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON Canada.,5Institute of Medical Science, University of Toronto, Toronto, ON Canada.,6Monash University, Melbourne, VIC Australia
| | - Thomas K Waddell
- 1Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON Canada.,5Institute of Medical Science, University of Toronto, Toronto, ON Canada
| |
Collapse
|
39
|
Curcumin Attenuates Airway Inflammation and Airway Remolding by Inhibiting NF-κB Signaling and COX-2 in Cigarette Smoke-Induced COPD Mice. Inflammation 2018; 41:1804-1814. [DOI: 10.1007/s10753-018-0823-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
40
|
Swatek AM, Lynch TJ, Crooke AK, Anderson PJ, Tyler SR, Brooks L, Ivanovic M, Klesney-Tait JA, Eberlein M, Pena T, Meyerholz DK, Engelhardt JF, Parekh KR. Depletion of Airway Submucosal Glands and TP63 +KRT5 + Basal Cells in Obliterative Bronchiolitis. Am J Respir Crit Care Med 2018; 197:1045-1057. [PMID: 29236513 PMCID: PMC5909161 DOI: 10.1164/rccm.201707-1368oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/12/2017] [Indexed: 12/24/2022] Open
Abstract
RATIONALE Obliterative bronchiolitis (OB) is a major cause of mortality after lung transplantation. Depletion of airway stem cells (SCs) may lead to fibrosis in OB. OBJECTIVES Two major SC compartments in airways are submucosal glands (SMGs) and surface airway p63 (also known as TP63 [tumor protein 63])-positive/K5 (also known as KRT5 [keratin 5])-positive basal cells (BCs). We hypothesized that depletion of these SC compartments occurs in OB. METHODS Ferret orthotopic left lung transplants were used as an experimental model of OB, and findings were corroborated in human lung allografts. Morphometric analysis was performed in ferret and human lungs to evaluate the abundance of SMGs and changes in the expression of phenotypic BC markers in control, lymphocytic bronchiolitis, and OB airways. The abundance and proliferative capacity of proximal and distal airway SCs was assessed using a clonogenic colony-forming efficiency assay. MEASUREMENTS AND MAIN RESULTS Ferret allografts revealed significant loss of SMGs with development of OB. A progressive decline in p63+/K5+ and increase in K5+/K14+ and K14+ BC phenotypes correlated with the severity of allograft rejection in large and small ferret airways. The abundance and proliferative capacity of basal SCs in large allograft airways declined with severity of OB, and there was complete ablation of basal SCs in distal OB airways. Human allografts mirrored phenotypic BC changes observed in the ferret model. CONCLUSIONS SMGs and basal SC compartments are depleted in large and/or small airways of lung allografts, and basal SC proliferative capacity declines with progression of disease and phenotypic changes. Global airway SC depletion may be a mechanism for pulmonary allograft failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Julia A. Klesney-Tait
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Michael Eberlein
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Tahuanty Pena
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | | | | |
Collapse
|
41
|
Ghosh M, Miller YE, Nakachi I, Kwon JB, Barón AE, Brantley AE, Merrick DT, Franklin WA, Keith RL, Vandivier RW. Exhaustion of Airway Basal Progenitor Cells in Early and Established Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2018; 197:885-896. [PMID: 29211494 PMCID: PMC6020409 DOI: 10.1164/rccm.201704-0667oc] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 12/01/2017] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Up to 40% of smokers develop chronic obstructive pulmonary disease (COPD) over a period that spans decades. Despite the importance of COPD, much remains to be learned about susceptibility and pathogenesis, especially during early, prediagnostic stages of disease. Airway basal progenitor cells are crucial for lung health and resilience because of their ability to repair injured airways. In COPD, the normal airway epithelium is replaced with increased basal and secretory (mucous) cells and decreased ciliated cells, suggesting that progenitors are impaired. OBJECTIVES To examine airway basal progenitor cells and lung function in smokers with and without COPD. METHODS Bronchial biopsies taken from smokers at risk for COPD and lung cancer were used to acquire airway basal progenitor cells. They were evaluated for count, self-renewal, and multipotentiality (ability to differentiate to basal, mucous, and ciliated cells), and progenitor count was examined for its relationship with lung function. MEASUREMENTS AND MAIN RESULTS Basal progenitor count, self-renewal, and multipotentiality were all reduced in COPD versus non-COPD. COPD progenitors produced an epithelium with increased basal and mucous cells and decreased ciliated cells, replicating the COPD phenotype. Progenitor depletion correlated with lung function and identified a subset of subjects without COPD with lung function that was midway between non-COPD with high progenitor counts and those with COPD. CONCLUSIONS Basal progenitor dysfunction relates to the histologic and physiologic manifestations of COPD and identifies a subset that may represent an early, prediagnostic stage of COPD, indicating that progenitor exhaustion is involved in COPD pathogenesis.
Collapse
Affiliation(s)
- Moumita Ghosh
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - York E. Miller
- COPD Program, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
- Veterans Affairs Eastern Colorado Healthcare System, Denver, Colorado
| | - Ichiro Nakachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan; and
| | - Jennifer B. Kwon
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Anna E. Barón
- Department of Biostatistics and Bioinformatics, University of Colorado School of Public Health, Aurora, Colorado
| | - Alexandra E. Brantley
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Daniel T. Merrick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wilbur A. Franklin
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Robert L. Keith
- COPD Program, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
- Veterans Affairs Eastern Colorado Healthcare System, Denver, Colorado
| | - R. William Vandivier
- COPD Program, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
42
|
Kim MS, Kim SH, Jeon D, Kim HY, Lee K. Changes in expression of cytokines in polyhexamethylene guanidine-induced lung fibrosis in mice: Comparison of bleomycin-induced lung fibrosis. Toxicology 2018; 393:185-192. [DOI: 10.1016/j.tox.2017.11.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 01/07/2023]
|
43
|
Klimek L, Koennecke M, Mullol J, Hellings PW, Wang D, Fokkens W, Gevaert P, Wollenberg B. A possible role of stem cells in nasal polyposis. Allergy 2017; 72:1868-1873. [PMID: 28599061 DOI: 10.1111/all.13221] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2017] [Indexed: 12/21/2022]
Abstract
Since its discovery, the understanding of stem/progenitor cells raised dramatically in the last decade. Their regenerative potential is important to develop new therapeutic applications, but the identification advanced much faster than our understanding of stem/progenitor cells. In nasal polyposis, little is known about stem cells/progenitor cells and their ability. However, the further characterization of stem cells/progenitor cells may provide new treatment options for combating nasal polyposis. This review highlights the knowledge of the current literature about stem cells/progenitor cells in nasal polyposis and how this may be exploited in the development of novel treatment strategies.
Collapse
Affiliation(s)
- L. Klimek
- Center for Rhinology and Allergology; Wiesbaden Germany
| | - M. Koennecke
- Department of Otorhinolaryngology; University Hospital Schleswig-Holstein; Lübeck Germany
| | - J. Mullol
- Rhinology Unit and Smell Clinic; Department of Otorhinolaryngology; Hospital Clinic; IDIBAPS; Universitat de Barcelona, CIBERES; Barcelona Catalonia Spain
| | - P. W. Hellings
- Department of Otorhinolaryngology; University Hospitals Leuven; Leuven Belgium
- Department of Otorhinolaryngology; Academic Medical Centre; Amsterdam The Netherlands
| | - D.Y. Wang
- Department of Otolaryngology; National University of Singapore; Singapore
| | - W. Fokkens
- Department of Otorhinolaryngology; Academic Medical Centre; Amsterdam The Netherlands
| | - P. Gevaert
- Department of Otorhinolaryngology; Ghent University; Ghent Belgium
| | - B. Wollenberg
- Department of Otorhinolaryngology; University Hospital Schleswig-Holstein; Lübeck Germany
| |
Collapse
|
44
|
Generation of Induced Progenitor-like Cells from Mature Epithelial Cells Using Interrupted Reprogramming. Stem Cell Reports 2017; 9:1780-1795. [PMID: 29198829 PMCID: PMC5785620 DOI: 10.1016/j.stemcr.2017.10.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/17/2023] Open
Abstract
A suitable source of progenitor cells is required to attenuate disease or affect cure. We present an "interrupted reprogramming" strategy to generate "induced progenitor-like (iPL) cells" using carefully timed expression of induced pluripotent stem cell reprogramming factors (Oct4, Sox2, Klf4, and c-Myc; OSKM) from non-proliferative Club cells. Interrupted reprogramming allowed controlled expansion yet preservation of lineage commitment. Under clonogenic conditions, iPL cells expanded and functioned as a bronchiolar progenitor-like population to generate mature Club cells, mucin-producing goblet cells, and cystic fibrosis transmembrane conductance regulator (CFTR)-expressing ciliated epithelium. In vivo, iPL cells can repopulate CFTR-deficient epithelium. This interrupted reprogramming process could be metronomically applied to achieve controlled progenitor-like proliferation. By carefully controlling the duration of expression of OSKM, iPL cells do not become pluripotent, and they maintain their memory of origin and retain their ability to efficiently return to their original phenotype. A generic technique to produce highly specified populations may have significant implications for regenerative medicine.
Collapse
|
45
|
Schmid A, Sailland J, Novak L, Baumlin N, Fregien N, Salathe M. Modulation of Wnt signaling is essential for the differentiation of ciliated epithelial cells in human airways. FEBS Lett 2017; 591:3493-3506. [PMID: 28921507 PMCID: PMC5683904 DOI: 10.1002/1873-3468.12851] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 12/20/2022]
Abstract
Wnt signaling is essential for the differentiation of airway epithelial cells during development. Here, we examined the role of Wnt signaling during redifferentiation of ciliated airway epithelial cells in vitro at the air liquid interface as a model of airway epithelial repair. Phases of proliferation and differentiation were defined. Markers of squamous metaplasia and epithelial ciliation were followed while enhancing β‐catenin signaling by blocking glycogen synthase kinase 3β with SB216763 and shRNA as well as inhibiting canonical WNT signaling with apical application of Dickkopf 1 (Dkk1). Our findings indicate that enhanced β‐catenin signaling decreases the number of ciliated cells and causes squamous changes in the epithelium, whereas treatment with DDk1 leads to an increased number of ciliated cells.
Collapse
Affiliation(s)
- Andreas Schmid
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami School of Medicine, FL, USA
| | - Juliette Sailland
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami School of Medicine, FL, USA
| | - Lisa Novak
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami School of Medicine, FL, USA
| | - Nathalie Baumlin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami School of Medicine, FL, USA
| | - Nevis Fregien
- Department of Cell Biology, University of Miami School of Medicine, FL, USA
| | - Matthias Salathe
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami School of Medicine, FL, USA
| |
Collapse
|
46
|
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
|
47
|
Martins TL, Campos KKD, Araújo NPDS, Machado DF, Bezerra FS. Extrapulmonary effects of temporal exposure to cigarette smoke. Toxicol Ind Health 2017; 33:717-725. [PMID: 28854870 DOI: 10.1177/0748233717715187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
This study aimed to evaluate the extrapulmonary effects of exposure to cigarette smoke (CS) through the analysis of blood components and histopathological examinations of the trachea and diaphragm muscle (DM) in C57BL/6 mice. Thirty-six animals were exposed to six cigarettes per day for 5 days. The mice were divided into a control group (CG) and groups exposed to CS for 1 (CS1D), 2 (CS2D), 3 (CS3D), 4 (CS4D), and 5 (CS5D) days. The trachea, DM, and blood were collected for morphometric and biochemical analyses. In comparison with the CG, CS4D and CS5D mice showed an increased influx of inflammatory cells into the DM and trachea. Increased glycogen deposits in the tracheal tissue of CS3D mice were observed, compared with that in CG, CS1D, and CS2D mice. In the blood serum, the number of inflammatory cells and the concentration of cholesterol increased in CS1D mice, compared with the CG. Alanine aminotransferase (ALT) levels were elevated in CS5D mice, compared with those in CS3D and CS4D mice. Aspartate aminotransferase (AST) levels were elevated in CS3D and CS5D mice, compared with those in the CG. Urea levels were significantly increased in CS5D mice, compared with CS1D mice. Our results showed extrapulmonary effects of short-term exposure to CS in adult mice.
Collapse
Affiliation(s)
- Thais Lourenço Martins
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Keila Karine Duarte Campos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Natália Pereira da Silva Araújo
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Dafne Fernandes Machado
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
48
|
Koennecke M, Böscke R, Pfannerstill AC, Reers S, Elsner M, Fell B, Richter A, Bruchhage KL, Schumann S, Pries R, Klimek L, Wollenberg B. Neuronal Differentiation Capability of Nasal Polyps of Chronic Rhinosinusitis. Arch Immunol Ther Exp (Warsz) 2017; 65:431-443. [PMID: 28280847 DOI: 10.1007/s00005-017-0456-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
Chronic rhinosinusitis with nasal polyps is considered a subgroup of chronic rhinosinusitis and a significant health problem, but the pathogenesis remains unclear to date. Therefore, we investigated the stemness to determine the role of stem cells in nasal polyps, with additional analysis of the neuronal differentiation potential of nasal polyp cells. We determined gene and protein expression profiles of stem cells in nasal polyp tissues, using whole genome microarray, quantitative real-time PCR (qPCR), immunohistochemistry, and flow cytometry. To evaluate the neuronal differentiation potential of nasal polyp cells, we used an efficient xenogeneic co-culture model with unsliced adult rat brain biopsies, followed by qPCR, immunohistochemistry, and growth factor antibody arrays. During gene expression analysis and immunohistochemistry, we were able to detect different stem cell markers, like Oct-4, Sox2, Klf4, c-Myc, ABCG2, Nanog, CD133, and Nestin, which confirmed the existence of stem cell like cells within nasal polyps. In addition, co-culture experiments give evidence for a guided differentiation into the neuronal lineage by overexpression of Nestin, Neurofilament, and GM-CSF. Our study demonstrated the expression of stem cell-related markers in nasal polyps. Furthermore, we characterized, for the first time, the stemness and neuronal differentiation potential of nasal polyp cells. These results gave new insights into the pathogenesis of nasal polyps and its therapeutic effectiveness could represent a promising strategy in the future.
Collapse
Affiliation(s)
- Michael Koennecke
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
| | - Robert Böscke
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Ann-Christin Pfannerstill
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Stefan Reers
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Martina Elsner
- Fraunhofer Research Institution for Marine Biotechnology, EMB, Lübeck, Germany
| | - Benjamin Fell
- Fraunhofer Research Institution for Marine Biotechnology, EMB, Lübeck, Germany
| | - Anja Richter
- Fraunhofer Research Institution for Marine Biotechnology, EMB, Lübeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Sandra Schumann
- Fraunhofer Research Institution for Marine Biotechnology, EMB, Lübeck, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology, University Medical Center Schleswig-Holstein, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| |
Collapse
|
49
|
Gerovac BJ, Fregien NL. IL-13 Inhibits Multicilin Expression and Ciliogenesis via Janus Kinase/Signal Transducer and Activator of Transcription Independently of Notch Cleavage. Am J Respir Cell Mol Biol 2016; 54:554-61. [PMID: 26414872 DOI: 10.1165/rcmb.2015-0227oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Loss of ciliated cells and increases in goblet cells are seen in respiratory diseases such as asthma. These changes result in part from reduced differentiation of basal progenitor cells to ciliated cells during injury and repair. The T helper 2 cytokine, IL-13, has been shown to inhibit ciliated cell differentiation, but the mechanism is not clearly understood. We recently showed that Notch signaling inhibits ciliated cell differentiation in submerged culture by repressing multicilin and forkhead box J1 (FOXJ1) expression, genes required for ciliogenesis. Using a novel method to study ciliated cell differentiation, we investigated the relationship between IL-13 and Notch signaling pathways. We found that IL-13 inhibits ciliated cell differentiation by repressing multicilin and FOXJ1 expression but does so independent of Notch signaling. In addition, we show that pharmacological inhibition of Janus kinase/signal transducer and activator of transcription, but not mitogen activated protein kinase kinase, signaling rescues multicilin and FOXJ1 expression and ciliated cell differentiation in the presence of IL-13. These findings indicate that regulation of multicilin expression by two distinct signaling pathways affects ciliated cell differentiation. In addition, the requirement for Janus kinase activation in IL-13-induced inhibition of ciliogenesis provides a potential therapeutic target for the treatment of respiratory disease.
Collapse
Affiliation(s)
- Benjamin J Gerovac
- Department of Cell Biology, University of Miami School of Medicine, Miami, Florida
| | - Nevis L Fregien
- Department of Cell Biology, University of Miami School of Medicine, Miami, Florida
| |
Collapse
|
50
|
Pan JH, Adair-Kirk TL, Patel AC, Huang T, Yozamp NS, Xu J, Reddy EP, Byers DE, Pierce RA, Holtzman MJ, Brody SL. Myb permits multilineage airway epithelial cell differentiation. Stem Cells 2015; 32:3245-56. [PMID: 25103188 DOI: 10.1002/stem.1814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
The epithelium of the pulmonary airway is specially differentiated to provide defense against environmental insults, but also subject to dysregulated differentiation that results in lung disease. The current paradigm for airway epithelial differentiation is a one-step program whereby a p63(+) basal epithelial progenitor cell generates a ciliated or secretory cell lineage, but the cue for this transition and whether there are intermediate steps are poorly defined. Here, we identify transcription factor Myb as a key regulator that permits early multilineage differentiation of airway epithelial cells. Myb(+) cells were identified as p63(-) and therefore distinct from basal progenitor cells, but were still negative for markers of differentiation. Myb RNAi treatment of primary-culture airway epithelial cells and Myb gene deletion in mice resulted in a p63(-) population with failed maturation of Foxj1(+) ciliated cells as well as Scbg1a1(+) and Muc5ac(+) secretory cells. Consistent with these findings, analysis of whole genome expression of Myb-deficient cells identified Myb-dependent programs for ciliated and secretory cell differentiation. Myb(+) cells were rare in human airways but were increased in regions of ciliated cells and mucous cell hyperplasia in samples from subjects with chronic obstructive pulmonary disease. Together, the results show that a p63(-) Myb(+) population of airway epithelial cells represents a distinct intermediate stage of differentiation that is required under normal conditions and may be heightened in airway disease.
Collapse
Affiliation(s)
- Jie-Hong Pan
- Department of Medicine, Washington University, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|