101
|
Abstract
Asthma is a genetically and phenotypically complex disease that has a major impact on global health. Signs and symptoms of asthma are caused by the obstruction of airflow through the airways. The epithelium that lines the airways plays a major role in maintaining airway patency and in host defense. The epithelium initiates responses to inhaled or aspirated substances, including allergens, viruses, and bacteria, and epithelial-derived cytokines are important in the recruitment and activation of immune cells in the airway. Changes in the structure and function of the airway epithelium are a prominent feature of asthma. Approximately half of individuals with asthma have evidence of active type 2 immune responses in the airway. In these individuals, epithelial cytokines promote type 2 responses, and responses to type 2 cytokines result in increased epithelial mucus production and other effects that cause airway obstruction. Recent work also implicates other epithelial responses, including interleukin-17, interferon and ER stress responses, that may contribute to asthma pathogenesis and provide new targets for therapy.
Collapse
Affiliation(s)
- Luke R Bonser
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States
| | - David J Erle
- Lung Biology Center, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
102
|
A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med 2019; 25:1153-1163. [PMID: 31209336 DOI: 10.1038/s41591-019-0468-5] [Citation(s) in RCA: 584] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 04/25/2019] [Indexed: 11/09/2022]
Abstract
Human lungs enable efficient gas exchange and form an interface with the environment, which depends on mucosal immunity for protection against infectious agents. Tightly controlled interactions between structural and immune cells are required to maintain lung homeostasis. Here, we use single-cell transcriptomics to chart the cellular landscape of upper and lower airways and lung parenchyma in healthy lungs, and lower airways in asthmatic lungs. We report location-dependent airway epithelial cell states and a novel subset of tissue-resident memory T cells. In the lower airways of patients with asthma, mucous cell hyperplasia is shown to stem from a novel mucous ciliated cell state, as well as goblet cell hyperplasia. We report the presence of pathogenic effector type 2 helper T cells (TH2) in asthmatic lungs and find evidence for type 2 cytokines in maintaining the altered epithelial cell states. Unbiased analysis of cell-cell interactions identifies a shift from airway structural cell communication in healthy lungs to a TH2-dominated interactome in asthmatic lungs.
Collapse
|
103
|
Ng-Blichfeldt JP, Gosens R, Dean C, Griffiths M, Hind M. Regenerative pharmacology for COPD: breathing new life into old lungs. Thorax 2019; 74:890-897. [PMID: 30940772 DOI: 10.1136/thoraxjnl-2018-212630] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/09/2019] [Accepted: 02/25/2019] [Indexed: 11/04/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global health concern with few effective treatments. Widespread destruction of alveolar tissue contributes to impaired gas exchange in severe COPD, and recent radiological evidence suggests that destruction of small airways is a major contributor to increased peripheral airway resistance in disease. This important finding might in part explain the failure of conventional anti-inflammatory treatments to restore lung function even in patients with mild disease. There is a clear need for alternative pharmacological strategies for patients with COPD/emphysema. Proposed regenerative strategies such as cell therapy and tissue engineering are hampered by poor availability of exogenous stem cells, discouraging trial results, and risks and cost associated with surgery. An alternative therapeutic approach is augmentation of lung regeneration and/or repair by biologically active factors, which have potential to be employed on a large scale. In favour of this strategy, the healthy adult lung is known to possess a remarkable endogenous regenerative capacity. Numerous preclinical studies have shown induction of regeneration in animal models of COPD/emphysema. Here, we argue that given the widespread and irreversible nature of COPD, serious consideration of regenerative pharmacology is necessary. However, for this approach to be feasible, a better understanding of the cell-specific molecular control of regeneration, the regenerative potential of the human lung and regenerative competencies of patients with COPD are required.
Collapse
Affiliation(s)
- John-Poul Ng-Blichfeldt
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK .,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen, Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen, Netherlands
| | - Charlotte Dean
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mark Griffiths
- National Heart and Lung Institute, Imperial College London, London, UK.,Barts Heart Centre, St Bartholomews Hospital, London, UK
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, UK.,Respiratory Medicine, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
104
|
Abstract
INTRODUCTION Lifelong maintenance of a healthy lung requires resident stem cells to proliferate according to tissue requirements. Once thought to be a quiescent tissue, evolving views of the complex differentiation landscape of lung stem and progenitor cells have broad implications for our understanding of how the lung is maintained, as well as the development of new therapies for promoting endogenous regeneration in lung disease. AREAS COVERED This review collates a large body of research relating to the hierarchical organization of epithelial stem cells in the adult lung and their role in tissue homeostasis and regeneration after injury. To identify relevant studies, PubMed was queried using one or a combination of the terms 'lung', 'airway', 'alveoli', 'stem cells', 'progenitor', 'repair' and 'regeneration'. EXPERT OPINION This review discusses how new technologies and injury models have challenged the demarcations between stem and progenitor cell populations.
Collapse
Affiliation(s)
- Jonathan L McQualter
- a School of Health and Biomedical Sciences , RMIT University , Melbourne , Australia
| |
Collapse
|
105
|
Non-lytic clearance of influenza B virus from infected cells preserves epithelial barrier function. Nat Commun 2019; 10:779. [PMID: 30770807 PMCID: PMC6377627 DOI: 10.1038/s41467-019-08617-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/22/2019] [Indexed: 01/11/2023] Open
Abstract
Influenza B virus (IBV) is an acute, respiratory RNA virus that has been assumed to induce the eventual death of all infected cells. We and others have shown however, that infection with apparently cytopathic viruses does not necessarily lead to cell death; some cells can intrinsically clear the virus and persist in the host long-term. To determine if any cells can survive direct IBV infection, we here generate a recombinant IBV capable of activating a host-cell reporter to permanently label all infected cells. Using this system, we demonstrate that IBV infection leads to the formation of a survivor cell population in the proximal airways that are ciliated-like, but transcriptionally and phenotypically distinct from both actively infected and bystander ciliated cells. We also show that survivor cells are critical to maintain respiratory barrier function. These results highlight a host response pathway that preserves the epithelium to limit the severity of IBV disease. Infection of a cell with influenza B virus (IBV) often results in cell death and the role of surviving cells in pathogenesis is unclear. Here, Dumm et al. generate a recombinant IBV that activates a host-cell reporter to permanently label infected cells, and show that surviving cells are important to preserve epithelial barrier function.
Collapse
|
106
|
Berical A, Lee RE, Randell SH, Hawkins F. Challenges Facing Airway Epithelial Cell-Based Therapy for Cystic Fibrosis. Front Pharmacol 2019; 10:74. [PMID: 30800069 PMCID: PMC6376457 DOI: 10.3389/fphar.2019.00074] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause the life-limiting hereditary disease, cystic fibrosis (CF). Decreased or absent functional CFTR protein in airway epithelial cells leads to abnormally viscous mucus and impaired mucociliary transport, resulting in bacterial infections and inflammation causing progressive lung damage. There are more than 2000 known variants in the CFTR gene. A subset of CF individuals with specific CFTR mutations qualify for pharmacotherapies of variable efficacy. These drugs, termed CFTR modulators, address key defects in protein folding, trafficking, abundance, and function at the apical cell membrane resulting from specific CFTR mutations. However, some CFTR mutations result in little or no CFTR mRNA or protein expression for which a pharmaceutical strategy is more challenging and remote. One approach to rescue CFTR function in the airway epithelium is to replace cells that carry a mutant CFTR sequence with cells that express a normal copy of the gene. Cell-based therapy theoretically has the potential to serve as a one-time cure for CF lung disease regardless of the causative CFTR mutation. In this review, we explore major challenges and recent progress toward this ambitious goal. The ideal therapeutic cell would: (1) be autologous to avoid the complications of rejection and immune-suppression; (2) be safely modified to express functional CFTR; (3) be expandable ex vivo to generate sufficient cell quantities to restore CFTR function; and (4) have the capacity to engraft, proliferate and persist long-term in recipient airways without complications. Herein, we explore human bronchial epithelial cells (HBECs) and induced pluripotent stem cells (iPSCs) as candidate cell therapies for CF and explore the challenges facing their delivery to the human airway.
Collapse
Affiliation(s)
- Andrew Berical
- Center for Regenerative Medicine, Boston Medical Center and Boston University, Boston, MA, United States.,The Pulmonary Center, Boston University School of Medicine, Boston, MA, United States
| | - Rhianna E Lee
- Cystic Fibrosis Research Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Scott H Randell
- Cystic Fibrosis Research Center, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Finn Hawkins
- Center for Regenerative Medicine, Boston Medical Center and Boston University, Boston, MA, United States.,The Pulmonary Center, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
107
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
108
|
Duval C, Watanabe M, Donati G. Buried myoepithelial stem cells as a reservoir for repairing the exposed airway epithelium. Stem Cell Investig 2019; 5:45. [PMID: 30701180 DOI: 10.21037/sci.2018.11.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/09/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Carlotta Duval
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy.,Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Mika Watanabe
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy.,Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Giacomo Donati
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy.,Molecular Biotechnology Center, University of Turin, Torino, Italy
| |
Collapse
|
109
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
110
|
Burgess JK, Heijink IH. Chronic Lung Pathologies That Require Repair and Regeneration. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7122311 DOI: 10.1007/978-3-030-29403-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, are a major cause of mortality worldwide. With the increasing incidence with ageing, the full impact of these diseases is yet to be realised. For most chronic lung diseases there are limited treatments options, with the existing approaches mainly addressing symptom relief. Little progress has been made, in recent years, in the development of new therapeutic strategies for managing these burdensome pathologies. There is an urgent need to increase our understanding of the mechanisms underlying these diseases. Endogenous progenitor cells (stem cells) have been recognised in many organs, including the lungs where they are suggested to maintain a population of cells that are able to facilitate the endogenous repair processes. Emerging knowledge of how these repair processes are disrupted in chronic lung diseases and the potential to capitalise upon the regenerative capacity of stem cell populations raise the hopes of the field worldwide for innovative treatment approaches for these devastating diseases in the future. This chapter outlines the series of diseases that may benefit from these emerging new therapeutic outlooks.
Collapse
Affiliation(s)
- Janette K. Burgess
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Irene H. Heijink
- The University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| |
Collapse
|
111
|
Guenthart BA, Chen YW, Bacchetta M. New insights and therapeutic targets: Lung injury and disease. J Thorac Cardiovasc Surg 2018; 157:416-420. [PMID: 30557958 DOI: 10.1016/j.jtcvs.2018.09.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Brandon A Guenthart
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Weill Cornell Medical Center, New York, NY
| | - Ya-Wen Chen
- Columbia Center for Human Development, Columbia University Medical Center, Columbia University, New York, NY; Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY; Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, Calif; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Matthew Bacchetta
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, Tenn.
| |
Collapse
|
112
|
Su TT. Cellular plasticity, caspases and autophagy; that which does not kill us, well, makes us different. Open Biol 2018; 8:rsob.180157. [PMID: 30487302 PMCID: PMC6282069 DOI: 10.1098/rsob.180157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023] Open
Abstract
The ability to regenerate is a fundamental requirement for tissue homeostasis. Regeneration draws on three sources of cells. First and best-studied are dedicated stem/progenitor cells. Second, existing cells may proliferate to compensate for the lost cells of the same type. Third, a different cell type may change fate to compensate for the lost cells. This review focuses on regeneration of the third type and will discuss the contributions by post-transcriptional mechanisms including the emerging evidence for cell-autonomous and non-lethal roles of cell death pathways.
Collapse
Affiliation(s)
- Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309-0347, USA .,University of Colorado Comprehensive Cancer Center, Anschutz Medical Campus, 13001 E. 17th Pl., Aurora, CO 80045, USA
| |
Collapse
|
113
|
Abdullah C, Duronio RJ. The many fates of tissue regeneration. PLoS Genet 2018; 14:e1007728. [PMID: 30462634 PMCID: PMC6248895 DOI: 10.1371/journal.pgen.1007728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Christopher Abdullah
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- SPIRE Postdoctoral Fellowship Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Robert J. Duronio
- Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
114
|
Verghese S, Su TT. Ionizing radiation induces stem cell-like properties in a caspase-dependent manner in Drosophila. PLoS Genet 2018; 14:e1007659. [PMID: 30462636 PMCID: PMC6248896 DOI: 10.1371/journal.pgen.1007659] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/27/2018] [Indexed: 11/18/2022] Open
Abstract
Cancer treatments including ionizing radiation (IR) can induce cancer stem cell-like properties in non-stem cancer cells, an outcome that can interfere with therapeutic success. Yet, we understand little about what consequences of IR induces stem cell like properties and why some cancer cells show this response but not others. In previous studies, we identified a pool of epithelial cells in Drosophila larval wing discs that display IR-induced stem cell-like properties. These cells are resistant to killing by IR and, after radiation damage, change fate and translocate to regenerate parts of the disc that suffered more cell death. Here, we report the identification of two new pools of cells with IR-induced regenerative capability. We addressed how IR exposure results in the induction of stem cell-like behavior, and found a requirement for IR-induced caspase activity and for Zfh2, a transcription factor and an effector in the JAK/STAT pathway. Unexpectedly, the requirement for caspase activity was cell-autonomous within cell populations that display regenerative behavior. We propose a model in which the requirement for caspase activity and Zfh2 can be explained by apoptotic and non-apoptotic functions of caspases in the induction of stem cell-like behavior. Ionizing Radiation (IR), alone or in combination with other therapies, is used to treat an estimated half of all cancer patients. Yet, we understand little about why some tumors cells respond to treatment while others grow back (regenerate). We identified specific pools of cells within a Drosophila organ that are capable of regeneration after damage by IR. We also identified what it is about IR damage that allows these cells to regenerate. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States of America
| | - Tin Tin Su
- University of Colorado Comprehensive Cancer Center, Anschutz Medical Campus, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
115
|
Hewlett JC, Kropski JA, Blackwell TS. Idiopathic pulmonary fibrosis: Epithelial-mesenchymal interactions and emerging therapeutic targets. Matrix Biol 2018; 71-72:112-127. [PMID: 29625182 PMCID: PMC6146058 DOI: 10.1016/j.matbio.2018.03.021] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic disease of the lung that is marked by progressive decline in pulmonary function and ultimately respiratory failure. Genetic and environmental risk factors have been identified that indicate injury to, and dysfunction of the lung epithelium is central to initiating the pathogenic process. Following injury to the lung epithelium, growth factors, matrikines and extracellular matrix driven signaling together activate a variety of repair pathways that lead to inflammatory cell recruitment, fibroblast proliferation and expansion of the extracellular matrix, culminating in tissue fibrosis. This tissue fibrosis then leads to changes in the biochemical and biomechanical properties of the extracellular matrix, which potentiate profibrotic mechanisms through a "feed-forward cycle." This review provides an overview of the interactions of the pathogenic mechanisms of IPF with a focus on epithelial-mesenchymal crosstalk and the extracellular matrix as a therapeutic target for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin C Hewlett
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States.
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Veterans Affairs Medical Center, Nashville, TN, United States; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States.
| |
Collapse
|
116
|
van Haasteren J, Hyde SC, Gill DR. Lessons learned from lung and liver in-vivo gene therapy: implications for the future. Expert Opin Biol Ther 2018; 18:959-972. [PMID: 30067117 PMCID: PMC6134476 DOI: 10.1080/14712598.2018.1506761] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Ex-vivo gene therapy has had significant clinical impact over the last couple of years and in-vivo gene therapy products are being approved for clinical use. Gene therapy and gene editing approaches have huge potential to treat genetic disease and chronic illness. AREAS COVERED This article provides a review of in-vivo approaches for gene therapy in the lung and liver, exploiting non-viral and viral vectors with varying serotypes and pseudotypes to target-specific cells. Antibody responses inhibiting viral vectors continue to constrain effective repeat administration. Lessons learned from ex-vivo gene therapy and genome editing are also discussed. EXPERT OPINION The fields of lung and liver in-vivo gene therapy are thriving and a comparison highlights obstacles and opportunities for both. Overcoming immunological issues associated with repeated administration of viral vectors remains a key challenge. The addition of targeted small molecules in combination with viral vectors may offer one solution. A substantial bottleneck to the widespread adoption of in-vivo gene therapy is how to ensure sufficient capacity for clinical-grade vector production. In the future, the exploitation of gene editing approaches for in-vivo disease treatment may facilitate the resurgence of non-viral gene transfer approaches, which tend to be eclipsed by more efficient viral vectors.
Collapse
Affiliation(s)
- Joost van Haasteren
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen C. Hyde
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Deborah R. Gill
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Science, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
117
|
Chacón-Martínez CA, Koester J, Wickström SA. Signaling in the stem cell niche: regulating cell fate, function and plasticity. Development 2018; 145:145/15/dev165399. [PMID: 30068689 DOI: 10.1242/dev.165399] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cells have the ability to self-renew and differentiate along multiple lineages, driving tissue homeostasis and regeneration. Paradigms of unidirectional, hierarchical differentiation trajectories observed in embryonic and hematopoietic stem cells have traditionally been applied to tissue-resident stem cells. However, accumulating evidence implicates stemness as a bidirectional, dynamic state that is largely governed by the niche, which facilitates plasticity and adaptability to changing conditions. In this Review, we discuss mechanisms of cell fate regulation through niche-derived cues, with a particular focus on epithelial stem cells of the mammalian skin, intestine and lung. We discuss a spectrum of niche-derived biochemical, mechanical and architectural inputs that define stem cell states during morphogenesis, homeostasis and regeneration, and highlight how these diverse inputs influence stem cell plasticity.
Collapse
Affiliation(s)
- Carlos Andrés Chacón-Martínez
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany
| | - Janis Koester
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany
| | - Sara A Wickström
- Paul Gerson Unna Group 'Skin Homeostasis and Ageing', Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany .,Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland.,Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland
| |
Collapse
|
118
|
Plasschaert LW, Žilionis R, Choo-Wing R, Savova V, Knehr J, Roma G, Klein AM, Jaffe AB. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 2018; 560:377-381. [PMID: 30069046 PMCID: PMC6108322 DOI: 10.1038/s41586-018-0394-6] [Citation(s) in RCA: 722] [Impact Index Per Article: 103.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Lindsey W Plasschaert
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA.,Respiratory Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Rapolas Žilionis
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.,Institute of Biotechnology, Vilnius University, Vilnius, Lithuania
| | - Rayman Choo-Wing
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA.,Respiratory Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Virginia Savova
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.,Precision Immunology, Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Judith Knehr
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Guglielmo Roma
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Aron B Jaffe
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA, USA. .,Respiratory Diseases, Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| |
Collapse
|
119
|
Lynch TJ, Ahlers BA, Parekh KR. Lung transplantation: chronic rejection and stem cell depletion. J Thorac Dis 2018; 10:E666-E668. [PMID: 30233912 PMCID: PMC6129905 DOI: 10.21037/jtd.2018.07.70] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 07/12/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Thomas J Lynch
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Bethany A Ahlers
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kalpaj R Parekh
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
120
|
Diverse mechanisms for endogenous regeneration and repair in mammalian organs. Nature 2018; 557:322-328. [PMID: 29769669 DOI: 10.1038/s41586-018-0073-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
Abstract
Mammalian organs comprise an extraordinary diversity of cell and tissue types. Regenerative organs, such as the skin and gastrointestinal tract, use resident stem cells to maintain tissue function. Organs with a lower cellular turnover, such as the liver and lungs, mostly rely on proliferation of committed progenitor cells. In many organs, injury reveals the plasticity of both resident stem cells and differentiated cells. The ability of resident cells to maintain and repair organs diminishes with age, whereas, paradoxically, the risk of cancer increases. New therapeutic approaches aim to harness cell plasticity for tissue repair and regeneration while avoiding the risk of malignant transformation of cells.
Collapse
|
121
|
Lynch TJ, Anderson PJ, Rotti PG, Tyler SR, Crooke AK, Choi SH, Montoro DT, Silverman CL, Shahin W, Zhao R, Jensen-Cody CW, Adamcakova-Dodd A, Evans TIA, Xie W, Zhang Y, Mou H, Herring BP, Thorne PS, Rajagopal J, Yeaman C, Parekh KR, Engelhardt JF. Submucosal Gland Myoepithelial Cells Are Reserve Stem Cells That Can Regenerate Mouse Tracheal Epithelium. Cell Stem Cell 2018; 22:653-667.e5. [PMID: 29656941 PMCID: PMC5935589 DOI: 10.1016/j.stem.2018.03.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 01/25/2018] [Accepted: 03/21/2018] [Indexed: 12/18/2022]
Abstract
The mouse trachea is thought to contain two distinct stem cell compartments that contribute to airway repair-basal cells in the surface airway epithelium (SAE) and an unknown submucosal gland (SMG) cell type. Whether a lineage relationship exists between these two stem cell compartments remains unclear. Using lineage tracing of glandular myoepithelial cells (MECs), we demonstrate that MECs can give rise to seven cell types of the SAE and SMGs following severe airway injury. MECs progressively adopted a basal cell phenotype on the SAE and established lasting progenitors capable of further regeneration following reinjury. MECs activate Wnt-regulated transcription factors (Lef-1/TCF7) following injury and Lef-1 induction in cultured MECs promoted transition to a basal cell phenotype. Surprisingly, dose-dependent MEC conditional activation of Lef-1 in vivo promoted self-limited airway regeneration in the absence of injury. Thus, modulating the Lef-1 transcriptional program in MEC-derived progenitors may have regenerative medicine applications for lung diseases.
Collapse
Affiliation(s)
- Thomas J Lynch
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Preston J Anderson
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Pavana G Rotti
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Scott R Tyler
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Adrianne K Crooke
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Soon H Choi
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Daniel T Montoro
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carolyn L Silverman
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Weam Shahin
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Rui Zhao
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52242, USA
| | - T Idil Apak Evans
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Weiliang Xie
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Yulong Zhang
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - B Paul Herring
- Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, IN 46202, USA
| | - Peter S Thorne
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA 52242, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Charles Yeaman
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Kalpaj R Parekh
- Department of Cardiothoracic Surgery, University of Iowa, Iowa City, IA 52242, USA
| | - John F Engelhardt
- Department of Anatomy & Cell Biology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
122
|
Zhu Y, Chen X, Yang X, El-Hashash A. Stem cells in lung repair and regeneration: Current applications and future promise. J Cell Physiol 2018; 233:6414-6424. [PMID: 29271480 DOI: 10.1002/jcp.26414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022]
Abstract
Lung diseases are major cause of morbidity and mortality worldwide. The progress in regenerative medicine and stem cell research in the lung are currently a fast-growing research topic that can provide solutions to these major health problems. Under normal conditions, the rate of cellular proliferation is relatively low in the lung in vivo, compared to other major organ systems. Lung injury leads to the activation of stem/progenitor cell populations that re-enter the cell cycle. Yet, little is known about stem cells in the lung, despite common thoughts that these cells could play a critical role in the repair of lung injuries. Nor do we fully understand the cellular and architectural complexity of the respiratory tract, and the diverse stem/progenitor cells that are involved in the lung repair and regeneration. In this review, we discuss the conceptual framework of lung stem/progenitor cell biology, and describe lung diseases, in which stem cell manipulations may be physiologically significant. In addition, we highlight the challenges of lung stem cell-based therapy.
Collapse
Affiliation(s)
- Yuqing Zhu
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiao Chen
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xu Yang
- Section of Environmental Biomedicine, School of Life Science, Central China Normal University, Wuhan, Hubei, China
| | - Ahmed El-Hashash
- Centre of Stem cell and Regenerative Medicine, Schools of Medicine and Basic Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,University of Edinburgh-Zhejiang University Institute (UoE-ZJU Institute), Haining, Zhejiang, China.,Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
123
|
Tata A, Kobayashi Y, Chow RD, Tran J, Desai A, Massri AJ, McCord TJ, Gunn MD, Tata PR. Myoepithelial Cells of Submucosal Glands Can Function as Reserve Stem Cells to Regenerate Airways after Injury. Cell Stem Cell 2018; 22:668-683.e6. [PMID: 29656943 DOI: 10.1016/j.stem.2018.03.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/01/2018] [Accepted: 03/21/2018] [Indexed: 01/12/2023]
Abstract
Cells demonstrate plasticity following injury, but the extent of this phenomenon and the cellular mechanisms involved remain underexplored. Using single-cell RNA sequencing (scRNA-seq) and lineage tracing, we uncover that myoepithelial cells (MECs) of the submucosal glands (SMGs) proliferate and migrate to repopulate the airway surface epithelium (SE) in multiple injury models. Specifically, SMG-derived cells display multipotency and contribute to basal and luminal cell types of the SMGs and SE. Ex vivo expanded MECs have the potential to repopulate and differentiate into SE cells when grafted onto denuded airway scaffolds. Significantly, we find that SMG-like cells appear on the SE of both extra- and intra-lobular airways of large animal lungs following severe injury. We find that the transcription factor SOX9 is necessary for MEC plasticity in airway regeneration. Because SMGs are abundant and present deep within airways, they may serve as a reserve cell source for enhancing human airway regeneration.
Collapse
Affiliation(s)
- Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ryan D Chow
- Department of Genetics, Systems Biology Institute, Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jasmine Tran
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Avani Desai
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Abdull J Massri
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy J McCord
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael Dee Gunn
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
124
|
Yao E, Lin C, Wu Q, Zhang K, Song H, Chuang PT. Notch Signaling Controls Transdifferentiation of Pulmonary Neuroendocrine Cells in Response to Lung Injury. Stem Cells 2018; 36:377-391. [PMID: 29148109 DOI: 10.1002/stem.2744] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/21/2017] [Accepted: 11/04/2017] [Indexed: 12/23/2022]
Abstract
Production of an appropriate number of distinct cell types in precise locations during embryonic development is critical for proper tissue function. Homeostatic renewal or repair of damaged tissues in adults also requires cell expansion and transdifferentiation to replenish lost cells. However, the responses of diverse cell types to tissue injury are not fully elucidated. Moreover, the molecular mechanisms underlying transdifferentiation remain poorly understood. This knowledge is essential for harnessing the regenerative potential of individual cell types. This study investigated the fate of pulmonary neuroendocrine cells (PNECs) following lung damage to understand their plasticity and potential. PNECs are proposed to carry out diverse physiological functions in the lung and can also be the cells of origin of human small cell lung cancer. We found that Notch signaling is activated in proliferating PNECs in response to epithelial injury. Forced induction of high levels of Notch signaling in PNECs in conjunction with lung injury results in extensive proliferation and transdifferentiation of PNECs toward the fate of club cells, ciliated cells and goblet cells. Conversely, inactivating Notch signaling in PNECs abolishes their ability to switch cell fate following lung insult. We also established a connection between PNEC transdifferentiation and epigenetic modification mediated by the polycomb repressive complex 2 and inflammatory responses that involve the IL6-STAT3 pathway. These studies not only reveal a major pathway that controls PNEC fate change following lung injury but also provide tools to uncover the molecular basis of cell proliferation and fate determination in response to lung injury. Stem Cells 2018;36:377-391.
Collapse
Affiliation(s)
- Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Chuwen Lin
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Qingzhe Wu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, People's Republic of China
| | - Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Hai Song
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, People's Republic of China
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| |
Collapse
|
125
|
Abstract
Central to the classical hematopoietic stem cell (HSC) paradigm is the concept that the maintenance of blood cell numbers is exclusively executed by a discrete physical entity: the transplantable HSC. The HSC paradigm has served as a stereotypic template in stem cell biology, yet the search for rare, hardwired professional stem cells has remained futile in most other tissues. In a more open approach, the focus on the search for stem cells as a physical entity may need to be replaced by the search for stem cell function, operationally defined as the ability of an organ to replace lost cells. The nature of such a cell may be different under steady state conditions and during tissue repair. We discuss emerging examples including the renewal strategies of the skin, gut epithelium, liver, lung, and mammary gland in comparison with those of the hematopoietic system. While certain key housekeeping and developmental signaling pathways are shared between different stem cell systems, there may be no general, deeper principles underlying the renewal mechanisms of the various individual tissues.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, 3584CT Utrecht, The Netherlands;
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, United Kingdom;
| |
Collapse
|
126
|
Taylor MS, Chivukula RR, Myers LC, Jeck WR, Waghray A, Tata PR, Selig MK, O'Donnell WJ, Farver CF, Thompson BT, Rajagopal J, Kradin RL. A Conserved Distal Lung Regenerative Pathway in Acute Lung Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1149-1160. [PMID: 29476724 DOI: 10.1016/j.ajpath.2018.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 12/05/2017] [Accepted: 01/23/2018] [Indexed: 10/18/2022]
Abstract
Improved tools have led to a burgeoning understanding of lung regeneration in mice, but it is not yet known how these insights may be relevant to acute lung injury in humans. We report in detail two cases of fulminant idiopathic acute lung injury requiring extracorporeal membrane oxygenation in previously healthy young adults with acute respiratory distress syndrome, one of whom required lung transplantation. Biopsy specimens showed diffuse alveolar injury with a striking paucity of alveolar epithelial regeneration, rare hyaline membranes, and diffuse contiguous airspace lining by macrophages. This novel constellation was termed diffuse alveolar injury with delayed epithelization. In addition, mirroring data from murine models of lung injury/regeneration, peribronchiolar basaloid pods (previously described as squamous metaplasia) and ciliated bronchiolarization were identified in these patients and in 39% of 57 historical cases with diffuse alveolar damage. These findings demonstrate a common and clinically relevant human disease correlate for murine models of severe acute lung injury. Evidence suggests that peribronchiolar basaloid pods and bronchiolarization are related spatially and temporally and likely represent overlapping sequential stages of the response to severe distal airway injury.
Collapse
Affiliation(s)
- Martin S Taylor
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Raghu R Chivukula
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Laura C Myers
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - William R Jeck
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Avinash Waghray
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Purushothama R Tata
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Martin K Selig
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Walter J O'Donnell
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Carol F Farver
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jayaraj Rajagopal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard L Kradin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
127
|
Taylor MS, Chivukula RR, Myers LC, Jeck WR, Tata PR, O'Donnell WJ, Farver CF, Thompson BT, Rajagopal J, Kradin RL. Delayed Alveolar Epithelialization: A Distinct Pathology in Diffuse Acute Lung Injury. Am J Respir Crit Care Med 2018; 197:522-524. [PMID: 28696778 PMCID: PMC5821904 DOI: 10.1164/rccm.201706-1094le] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | - Laura C Myers
- 1 Massachusetts General Hospital Boston, Massachusetts and
| | - William R Jeck
- 1 Massachusetts General Hospital Boston, Massachusetts and
| | | | | | | | | | | | | |
Collapse
|
128
|
Developmental mechanisms and adult stem cells for therapeutic lung regeneration. Dev Biol 2018; 433:166-176. [DOI: 10.1016/j.ydbio.2017.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022]
|
129
|
Abstract
The meeting covered a plethora of rapidly evolving approaches and areas, such as organoid cultures modeling tissues and organs; stem cell-specific metabolites revealing new signaling pathways; single-cell technologies discovering new cell types and exploring stem cell niche interactions; novel methods studying stem cells in aging and cancer; lineage-tracing experiments exploring cell plasticity of tissues before and after injury; epigenetic studies illuminating cell reprogramming; new protocols improving cells for regenerative purposes; and several other timely and exciting topics.
Collapse
Affiliation(s)
- Thomas Graf
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
130
|
Stem and Progenitor Cells in Human Cardiopulmonary Development and Regeneration. Stem Cells Int 2017; 2017:2653142. [PMID: 29075297 PMCID: PMC5623785 DOI: 10.1155/2017/2653142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/15/2017] [Indexed: 12/26/2022] Open
Abstract
Already during embryonic development, the heart and the lung are thoroughly connected organs. Their interdependence allows our survival in the terrestrial environment by coupling cardiac output and gas exchange. The knowledge on developmental processes involving stem and progenitor cells is crucial to understand the onset of human cardiopulmonary diseases. The precise identification of various adult endogenous progenitors is still incomplete. Thus, caution should be exercised on newly available stem cell-based treatments until specific mechanisms of action are disclosed. The objective is to provide in the nearest future feasible and safer cell therapeutics for the complex pathological condition of human cardiopulmonary diseases. In this paper, we highlight the significant knowledge advancement concerning stem and progenitor cells in the cardiopulmonary field: from embryonic development to adult progenitors until early preclinical models for cardiopulmonary regeneration.
Collapse
|
131
|
Barkauskas CE, Chung MI, Fioret B, Gao X, Katsura H, Hogan BLM. Lung organoids: current uses and future promise. Development 2017; 144:986-997. [PMID: 28292845 DOI: 10.1242/dev.140103] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lungs are composed of a system of highly branched tubes that bring air into the alveoli, where gas exchange takes place. The proximal and distal regions of the lung contain epithelial cells specialized for different functions: basal, secretory and ciliated cells in the conducting airways and type II and type I cells lining the alveoli. Basal, secretory and type II cells can be grown in three-dimensional culture, with or without supporting stromal cells, and under these conditions they give rise to self-organizing structures known as organoids. This Review summarizes the different methods for generating organoids from cells isolated from human and mouse lungs, and compares their final structure and cellular composition with that of the airways or alveoli of the adult lung. We also discuss the potential and limitations of organoids for addressing outstanding questions in lung biology and for developing new drugs for disorders such as cystic fibrosis and asthma.
Collapse
Affiliation(s)
- Christina E Barkauskas
- Department of Medicine, Division of Pulmonary and Critical Medicine, Duke Medicine, Durham, NC 27710, USA
| | - Mei-I Chung
- Department of Cell Biology, Duke Medicine, Durham, NC 27710, USA
| | - Bryan Fioret
- Department of Cell Biology, Duke Medicine, Durham, NC 27710, USA
| | - Xia Gao
- Department of Cell Biology, Duke Medicine, Durham, NC 27710, USA
| | - Hiroaki Katsura
- Department of Cell Biology, Duke Medicine, Durham, NC 27710, USA
| | - Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, NC 27710, USA
| |
Collapse
|
132
|
Abstract
A new source of progenitor cells can now be used to study hidden aspects of human lung development and pediatric lung disease.
Collapse
Affiliation(s)
- Avinash Waghray
- Massachusetts General HospitalHarvard UniversityBostonUnited States
| | | |
Collapse
|
133
|
Geiger S, Hirsch D, Hermann FG. Cell therapy for lung disease. Eur Respir Rev 2017; 26:26/144/170044. [PMID: 28659506 DOI: 10.1183/16000617.0044-2017] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/08/2017] [Indexed: 12/11/2022] Open
Abstract
Besides cancer and cardiovascular diseases, lung disorders are a leading cause of morbidity and death worldwide. For many disease conditions no effective and curative treatment options are available. Cell therapies offer a novel therapeutic approach due to their inherent anti-inflammatory and anti-fibrotic properties. Mesenchymal stem/stromal cells (MSC) are the most studied cell product. Numerous preclinical studies demonstrate an improvement of disease-associated parameters after MSC administration in several lung disorders, including chronic obstructive pulmonary disease, acute respiratory distress syndrome and idiopathic pulmonary fibrosis. Furthermore, results from clinical studies using MSCs for the treatment of various lung diseases indicate that MSC treatment in these patients is safe. In this review we summarise the results of preclinical and clinical studies that indicate that MSCs are a promising therapeutic approach for the treatment of lung diseases. Nevertheless, further investigations are required.
Collapse
Affiliation(s)
- Sabine Geiger
- Preclinical Development, Apceth Biopharma GmbH, Munich, Germany
| | - Daniela Hirsch
- Preclinical Development, Apceth Biopharma GmbH, Munich, Germany
| | - Felix G Hermann
- Preclinical Development, Apceth Biopharma GmbH, Munich, Germany
| |
Collapse
|