1
|
Yang X, Zhou N, Cao J. Role of Small Airway Epithelial-Mesenchymal Transition and CXCL13 in Pulmonary Lymphoid Follicle Formation in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:2559-2569. [PMID: 39629182 PMCID: PMC11613702 DOI: 10.2147/copd.s487539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder characterized by inflammation and airway remodeling. Lymphoid follicles play a crucial role in acquired immunity and the development of COPD. However, the precise mechanisms of lymphoid follicle formation in COPD and the effects of cigarette smoke (CS) exposure on this process remain unclear. Epithelial-mesenchymal transition (EMT) is implicated in the progression of COPD and may serves as a source of stromal cells that produce chemokines crucial for lymphoid follicle formation. This study aims to clarify the contributions and mechanisms of EMT in lymphoid follicle genesis in COPD, focusing specifically on the role of CXCL13. Methods Lung tissue samples were obtained from patients with COPD, smokers, and non-smokers. Immunohistochemistry was performed to assess the lymphoid follicles, EMT-related markers, and CXCL13 expression. In vitro experiments were conducted using CS extract (CSE)-stimulated immortalized human bronchial epithelial cells (iHBECs) to induce EMT. The expression of EMT-related markers and CXCL13 in CSE-stimulated iHBECs was analyzed using Western blotting, real-time PCR, and immunofluorescence staining. The effect of an EMT inhibitor on CXCL13 expression was also examined. Results Patients with COPD and lymphoid follicles exhibited significantly lower forced expiratory volume in 1 s (% predicted) values than those without lymphoid follicles. Enhanced EMT changes were observed in patients with COPD and lymphoid follicles. Increased EMT-related markers and CXCL13 expression were observed in CSE-stimulated iHBECs, and CXCL13 expression gradually increased over time. Inhibiting EMT downregulated CXCL13 expression in iHBECs. Conclusion Lymphoid follicles are associated with enhanced EMT in COPD. EMT may act as a key driver of the adaptive immune response in COPD by promoting a microenvironment conducive to lymphoid follicles formation through the production of CXCL13. This study provides valuable insights into the mechanisms underlying lymphoid follicle formation in COPD and identifies potential therapeutic targets.
Collapse
Affiliation(s)
- Xia Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Ning Zhou
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Jie Cao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
2
|
Duong A, Wong A, Ramendra R, Sebben D, Moshkelgosha S, MacParland S, Liu M, Juvet S, Martinu T. A Rapid Human Lung Tissue Dissociation Protocol Maximizing Cell Yield and Minimizing Cellular Stress. Am J Respir Cell Mol Biol 2024; 71:509-518. [PMID: 38959415 DOI: 10.1165/rcmb.2023-0343ma] [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: 09/21/2023] [Accepted: 07/03/2024] [Indexed: 07/05/2024] Open
Abstract
The human lung is a complex organ that comprises diverse populations of epithelial, mesenchymal, vascular, and immune cells, which gains even greater complexity during disease states. To effectively study the lung at a single-cell level, a dissociation protocol that achieves the highest yield of viable cells of interest with minimal dissociation-associated protein or transcription changes is key. Here, we detail a rapid collagenase-based dissociation protocol (Col-Short) that provides a high-yield single-cell suspension that is suitable for a variety of downstream applications. Diseased human lung explants were obtained and dissociated through the Col-Short protocol and compared with four other dissociation protocols. Resulting single-cell suspensions were then assessed with flow cytometry, differential staining, and quantitative real-time PCR to identify major hematopoietic and nonhematopoietic cell populations, as well as their activation states. We observed that the Col-Short protocol provides the greatest number of cells per gram of lung tissue, with no reduction in viability when compared with previously described dissociation protocols. Col-Short had no observable surface protein marker cleavage as well as lower expression of protein activation markers and stress-related transcripts compared with four other protocols. The Col-Short dissociation protocol can be used as a rapid strategy to generate single cells for respiratory cell biology research.
Collapse
Affiliation(s)
- Allen Duong
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
- Institute of Medical Science
| | - Aaron Wong
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
- Institute of Medical Science
| | - Rayoun Ramendra
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
| | - David Sebben
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
| | - Sajad Moshkelgosha
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
| | - Sonya MacParland
- Toronto General Hospital Research Institute, and
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; and
- Institute of Medical Science
- Department of Laboratory Medicine and Pathobiology, and
| | - Mingyao Liu
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; and
- Institute of Medical Science
| | - Stephen Juvet
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; and
- Institute of Medical Science
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Toronto Lung Transplant Program
- Toronto General Hospital Research Institute, and
- Ajmera Transplant Centre, University Health Network, Toronto, Ontario, Canada; and
- Institute of Medical Science
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Takahashi M, Isagawa T, Sato T, Takeda N, Kawakami K. Lineage tracing using Wnt2b-2A-CreERT2 knock-in mice reveals the contributions of Wnt2b-expressing cells to novel subpopulations of mesothelial/epicardial cell lineages during mouse development. Genes Cells 2024; 29:854-875. [PMID: 39109760 DOI: 10.1111/gtc.13147] [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: 06/08/2023] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 10/04/2024]
Abstract
Mesothelial and epicardial cells give rise to various types of mesenchymal cells via epithelial (mesothelial)-to-mesenchymal transition during development. However, the genes controlling the differentiation and diversification of mesothelial/epicardial cells remain unclear. Here, we examined Wnt2b expression in the embryonic mesothelium and epicardium and performed lineage tracing of Wnt2b-expressing cells by using novel Wnt2b-2A-CreERT2 knock-in and LacZ-reporter mice. Wnt2b was expressed in mesothelial cells covering visceral organs, but the expression was restricted in their subpopulations. Wnt2b-expressing cells labeled at embryonic day (E) 10.5 were distributed to the mesothelium and mesenchyme in the lungs, abdominal wall, stomach, and spleen in Wnt2b2A-CreERT2/+;R26RLacZ/+ mice at E13.0. Wnt2b was initially expressed in the proepicardial organ (PEO) at E9.5 and then in the epicardium after E10.0. Wnt2b-expressing PEO cells labeled at E9.5 differentiated into a small fraction of cardiac fibroblasts and preferentially localized at the left side of the postnatal heart. LacZ+ epicardium-derived cells labeled at E10.5 differentiated into a small fraction of fibroblasts and smooth muscle cells in the postnatal heart. Taken together, our results reveal novel subpopulations of PEO and mesothelial/epicardial cells that are distinguishable by Wnt2b expression and elucidate the unique contribution of Wnt2b-expressing PEO and epicardial cells to the postnatal heart.
Collapse
Affiliation(s)
- Masanori Takahashi
- Department of Anatomy, Division of Bioimaging and Neuro-cell Science, Jichi Medical University, Shimotsuke, Japan
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Takayuki Isagawa
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Data Science Center, Jichi Medical University, Shimotsuke, Japan
| | - Tatsuyuki Sato
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | | |
Collapse
|
4
|
Andreozzi P, Gussoni G, Sesti G, Montano N, Pietrangelo A. Impact of electronic cigarettes (e-cigs) and heat-not-burn/heated tobacco products (HnB/HTP) on asthma and chronic obstructive pulmonary disease: a viewpoint of the Italian Society of Internal Medicine. Intern Emerg Med 2024; 19:1829-1837. [PMID: 38806787 PMCID: PMC11467123 DOI: 10.1007/s11739-024-03648-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
The association of cigarette smoking with several severe and very severe diseases (oncological, cardiovascular, respiratory) which have dramatic epidemiological, medical, and financial impact, is a well-known public threat. Asthma and chronic obstructive pulmonary disease (COPD) are highly prevalent diseases in Italy, posing significant public health challenges. Tobacco smoking, a primary risk factor for COPD and a common asthma trigger, remains a critical preventable public health issue. While universally acknowledged that quitting smoking drastically reduces the risk of smoking-related health issues, a significant portion of smokers and patients find quitting challenging or undesirable, hence a need for new ways to deal with it. A worth considering alternative might be the switch to electronic cigarettes (e-cig), and heat-not-burn/heated tobacco products (HnB/HTP). Emerging evidence suggests potential benefits in asthma and COPD management when transitioning from traditional smoking to e-cigs or HnB devices. However, the effectiveness of these products in facilitating smoking cessation is still debated, alongside concerns about their role in promoting smoking initiation among non-smokers. Internists are among the physicians who most frequently assist patients with smoking-related diseases, and in this perspective they cannot avoid paying attention to the progressive diffusion of smoking products alternative to the traditional cigarette, and to the controversies with respect to their use. In this context, the Italian Society of Internal Medicine, also recognizing a growing need for clarity for healthcare providers, has undertaken a comprehensive analysis of existing literature to offer an informed perspective on the health impact of e-cigs and HnB/HTP on asthma and COPD.
Collapse
Affiliation(s)
- Paola Andreozzi
- Predictive Medicine Unit, Department of Internal Medicine, Endocrine-Metabolic Sciences and Infectious Diseases, Azienda Ospedaliero Universitaria Policlinico Umberto I, Rome, Italy
| | | | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, 00189, Rome, Italy
| | - Nicola Montano
- Department of Clinical Community Sciences, University of Milan, 20122, Milan, Italy
| | - Antonello Pietrangelo
- Internal Medicine Unit, Department of Medical and Surgical Sciences, University Hospital of Modena, Modena, Italy.
| |
Collapse
|
5
|
Le NT, Dunleavy MW, Kumar RD, Zhou W, Bhatia SS, El-Hashash AH. Cellular therapies for idiopathic pulmonary fibrosis: current progress and future prospects. AMERICAN JOURNAL OF STEM CELLS 2024; 13:191-211. [PMID: 39308764 PMCID: PMC11411253 DOI: 10.62347/daks5508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial, fibrotic lung disease characterized by progressive damage. Lung tissues with IPF are replaced by fibrotic tissues with increased collagen deposition, modified extracellular matrix, all which overall damages the alveoli. These changes eventually impede the gas exchange function of the alveoli, and eventually leads to fatal respiratory failure of the lung. Investigations have been conducted to further understand IPF's pathogenesis, and significant progress in understanding its development has been made. Additionally, two therapeutic treatments, Nintedanib and Pirfenidone, have been approved and are currently used in medical applications. Moreover, cell-based treatments have recently come to the forefront of developing disease therapeutics and are the focus of many current studies. Furthermore, a sizable body of research encompassing basic, pre-clinical, and even clinical trials have all been amassed in recent years and hold a great potential for more widespread applications in patient care. Herein, this article reviews the progress in understanding the pathogenesis and pathophysiology of IPF. Additionally, different cell types used in IPF therapy were reviewed, including alveolar epithelial cells (AECs), circulating endothelial progenitors (EPCs), mixed lung epithelial cells, different types of stem cells, and endogenous lung tissue-specific stem cells. Finally, we discussed the contemporary trials that employ or explore cell-based therapy for IPF.
Collapse
Affiliation(s)
- Nicholas T Le
- Biology Department, Texas A&M University College Station, TX, USA
| | | | - Rebecca D Kumar
- Biology Department, Texas A&M University College Station, TX, USA
| | - William Zhou
- The University of Texas at Austin Austin, TX, USA
| | | | | |
Collapse
|
6
|
Attia SH, Saadawy SF, El-Mahroky SM, Nageeb MM. Alleviation of pulmonary fibrosis by the dual PPAR agonist saroglitazar and breast milk mesenchymal stem cells via modulating TGFß/SMAD pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5953-5974. [PMID: 38376539 PMCID: PMC11329427 DOI: 10.1007/s00210-024-03004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/09/2024] [Indexed: 02/21/2024]
Abstract
Pulmonary fibrosis (PF) is a complex disorder with high morbidity and mortality. Limited efficacies of the available drugs drive researchers to seek for new therapies. Saroglitazar (Saro), a full (PPAR α/γ) agonist, is devoid of known PPAR-mediated adverse effects. Breast milk mesenchymal stem cells (BrMSCs) are contemplated to be the ideal cell type harboring differentiation/anti-inflammatory/immunosuppressive properties. Accordingly, our aims were to investigate the potential roles of Saro and/or BrMSCs in PF and to spot their underlying protective mechanisms. In this study, PF was induced by bleomycin (BLM) via intratracheal instillation. Treatment started 14 days later. Animals were treated with oral saroglitazar (3 mg/kg daily) or intraperitoneal single BrMSCs injection (0.5 ml phosphate buffer saline (PBS) containing 2 × 107 cells) or their combination with same previous doses. At the work end, 24 h following the 6 weeks of treatment period, the levels of oxidative (MDA, SOD), inflammatory (IL-1ß, IL-10), and profibrotic markers (TGF-ß, αSMA) were assessed. The autophagy-related genes (LC3, Beclin) and the expression of PPAR-α/γ and SMAD-3/7 were evaluated. Furthermore, immunohistochemical and histological work were evaluated. Our study revealed marked lung injury influenced by BLM with severe oxidative/inflammatory/fibrotic damage, autophagy inhibition, and deteriorated lung histology. Saro and BrMSCs repaired the lung structure worsened by BLM. Treatments greatly declined the oxidative/inflammatory markers. The pro-fibrotic TGF-ß, αSMA, and SMAD-3 were decreased. Contrarily, autophagy markers were increased. SMAD-7 and PPAR α/γ were activated denoting their pivotal antifibrotic roles. Co-administration of Saro and BrMSCs revealed the top results. Our findings support the study hypothesis that Saro and BrMSCs can be proposed as potential treatments for IPF.
Collapse
Affiliation(s)
- Seba Hassan Attia
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Sara F Saadawy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samaa M El-Mahroky
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mahitab M Nageeb
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
7
|
Joo H, Min S, Cho SW. Advanced lung organoids for respiratory system and pulmonary disease modeling. J Tissue Eng 2024; 15:20417314241232502. [PMID: 38406820 PMCID: PMC10894554 DOI: 10.1177/20417314241232502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Amidst the recent coronavirus disease 2019 (COVID-19) pandemic, respiratory system research has made remarkable progress, particularly focusing on infectious diseases. Lung organoid, a miniaturized structure recapitulating lung tissue, has gained global attention because of its advantages over other conventional models such as two-dimensional (2D) cell models and animal models. Nevertheless, lung organoids still face limitations concerning heterogeneity, complexity, and maturity compared to the native lung tissue. To address these limitations, researchers have employed co-culture methods with various cell types including endothelial cells, mesenchymal cells, and immune cells, and incorporated bioengineering platforms such as air-liquid interfaces, microfluidic chips, and functional hydrogels. These advancements have facilitated applications of lung organoids to studies of pulmonary diseases, providing insights into disease mechanisms and potential treatments. This review introduces recent progress in the production methods of lung organoids, strategies for improving maturity, functionality, and complexity of organoids, and their application in disease modeling, including respiratory infection and pulmonary fibrosis.
Collapse
Affiliation(s)
- Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| |
Collapse
|
8
|
Zhang F, Gao H, Jiang X, Yang F, Zhang J, Song S, Shen J. Biomedical Application of Decellularized Scaffolds. ACS APPLIED BIO MATERIALS 2023; 6:5145-5168. [PMID: 38032114 DOI: 10.1021/acsabm.3c00778] [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] [Indexed: 12/01/2023]
Abstract
Tissue loss and end-stage organ failure are serious health problems across the world. Natural and synthetic polymer scaffold material based artificial organs play an important role in the field of tissue engineering and organ regeneration, but they are not from the body and may cause side effects such as rejection. In recent years, the biomimetic decellularized scaffold based materials have drawn great attention in the tissue engineering field for their good biocompatibility, easy modification, and excellent organism adaptability. Therefore, in this review, we comprehensively summarize the application of decellularized scaffolds in tissue engineering and biomedicine in recent years. The preparation methods, modification strategies, construction of artificial tissues, and application in biomedical applications are discussed. We hope that this review will provide a useful reference for research on decellularized scaffolds and promote their application tissue engineering.
Collapse
Affiliation(s)
- Fang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Huimin Gao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xuefeng Jiang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fang Yang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jun Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Saijie Song
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
- Jiangsu Engineering Research Center of Interfacial Chemistry, Nanjing University, Nanjing 210023, China
| |
Collapse
|
9
|
Mason EC, Menon S, Schneider BR, Gaskill CF, Dawson MM, Moore CM, Armstrong LC, Cho O, Richmond BW, Kropski JA, West JD, Geraghty P, Gomperts BN, Ess KC, Gally F, Majka SM. Activation of mTOR signaling in adult lung microvascular progenitor cells accelerates lung aging. J Clin Invest 2023; 133:e171430. [PMID: 37874650 PMCID: PMC10721153 DOI: 10.1172/jci171430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023] Open
Abstract
Reactivation and dysregulation of the mTOR signaling pathway are a hallmark of aging and chronic lung disease; however, the impact on microvascular progenitor cells (MVPCs), capillary angiostasis, and tissue homeostasis is unknown. While the existence of an adult lung vascular progenitor has long been hypothesized, these studies show that Abcg2 enriches for a population of angiogenic tissue-resident MVPCs present in both adult mouse and human lungs using functional, lineage, and transcriptomic analyses. These studies link human and mouse MVPC-specific mTORC1 activation to decreased stemness, angiogenic potential, and disruption of p53 and Wnt pathways, with consequent loss of alveolar-capillary structure and function. Following mTOR activation, these MVPCs adapt a unique transcriptome signature and emerge as a venous subpopulation in the angiodiverse microvascular endothelial subclusters. Thus, our findings support a significant role for mTOR in the maintenance of MVPC function and microvascular niche homeostasis as well as a cell-based mechanism driving loss of tissue structure underlying lung aging and the development of emphysema.
Collapse
Affiliation(s)
- Emma C. Mason
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Swapna Menon
- Pulmonary Vascular Research Institute Kochi and AnalyzeDat Consulting Services, Kerala, India
| | - Benjamin R. Schneider
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Christa F. Gaskill
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maggie M. Dawson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Camille M. Moore
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Craig Armstrong
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Okyong Cho
- Genomics and Microarray Core, University of Colorado Cancer Center, Anschutz Medical Center, Aurora, Colorado, USA
| | - Bradley W. Richmond
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Jonathan A. Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - James D. West
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center and Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Brigitte N. Gomperts
- Translational Research, UCLA Broad Stem Cell Research Center; Pediatrics Division of Pulmonary Medicine, University of California, Los Angeles, California, USA
| | - Kevin C. Ess
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Fabienne Gally
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan M. Majka
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
10
|
Tung VSK, Mathews F, Boruk M, Suppa G, Foronjy R, Pato MT, Pato CN, Knowles JA, Evgrafov OV. Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology. Int J Mol Sci 2023; 24:15339. [PMID: 37895019 PMCID: PMC10607243 DOI: 10.3390/ijms242015339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The study of neurodevelopmental molecular mechanisms in schizophrenia requires the development of adequate biological models such as patient-derived cells and their derivatives. We previously utilized cell lines with neural progenitor properties (CNON) derived from the superior or middle turbinates of patients with schizophrenia and control groups to study schizophrenia-specific gene expression. In this study, we analyzed single-cell RNA seq data from two CNON cell lines (one derived from an individual with schizophrenia (SCZ) and the other from a control group) and two biopsy samples from the middle turbinate (MT) (also from an individual with SCZ and a control). We compared our data with previously published data regarding the olfactory neuroepithelium and demonstrated that CNON originated from a single cell type present both in middle turbinate and the olfactory neuroepithelium and expressed in multiple markers of mesenchymal cells. To define the relatedness of CNON to the developing human brain, we also compared CNON datasets with scRNA-seq data derived from an embryonic brain and found that the expression profile of the CNON closely matched the expression profile one of the cell types in the embryonic brain. Finally, we evaluated the differences between SCZ and control samples to assess the utility and potential benefits of using CNON single-cell RNA seq to study the etiology of schizophrenia.
Collapse
Affiliation(s)
- Victoria Sook Keng Tung
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Fasil Mathews
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Marina Boruk
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Gabrielle Suppa
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Robert Foronjy
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Michele T. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - Carlos N. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - James A. Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| | - Oleg V. Evgrafov
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| |
Collapse
|
11
|
Phogat S, Thiam F, Al Yazeedi S, Abokor FA, Osei ET. 3D in vitro hydrogel models to study the human lung extracellular matrix and fibroblast function. Respir Res 2023; 24:242. [PMID: 37798767 PMCID: PMC10552248 DOI: 10.1186/s12931-023-02548-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
The pulmonary extracellular matrix (ECM) is a macromolecular structure that provides mechanical support, stability and elastic recoil for different pulmonary cells including the lung fibroblasts. The ECM plays an important role in lung development, remodeling, repair, and the maintenance of tissue homeostasis. Biomechanical and biochemical signals produced by the ECM regulate the phenotype and function of various cells including fibroblasts in the lungs. Fibroblasts are important lung structural cells responsible for the production and repair of different ECM proteins (e.g., collagen and fibronectin). During lung injury and in chronic lung diseases such as asthma, idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD), an abnormal feedback between fibroblasts and the altered ECM disrupts tissue homeostasis and leads to a vicious cycle of fibrotic changes resulting in tissue remodeling. In line with this, using 3D hydrogel culture models with embedded lung fibroblasts have enabled the assessment of the various mechanisms involved in driving defective (fibrotic) fibroblast function in the lung's 3D ECM environment. In this review, we provide a summary of various studies that used these 3D hydrogel models to assess the regulation of the ECM on lung fibroblast phenotype and function in altered lung ECM homeostasis in health and in chronic respiratory disease.
Collapse
Affiliation(s)
- Sakshi Phogat
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Fama Thiam
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Safiya Al Yazeedi
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Filsan Ahmed Abokor
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada
| | - Emmanuel Twumasi Osei
- Department of Biology, Okanagan Campus, University of British Columbia, 3187 University Way, ASC366, Kelowna, BC, V1V1V7, Canada.
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
12
|
Belgacemi R, Ribeiro Baptista B, Justeau G, Toigo M, Frauenpreis A, Yilmaz R, Der Vartanian A, Cazaunau M, Pangui E, Bergé A, Gratien A, Macias Rodriguez JC, Bellusci S, Derumeaux G, Boczkowski J, Al Alam D, Coll P, Lanone S, Boyer L. Complex urban atmosphere alters alveolar stem cells niche properties and drives lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2023; 325:L447-L459. [PMID: 37529852 PMCID: PMC10639009 DOI: 10.1152/ajplung.00061.2023] [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: 02/21/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
There is growing evidence suggesting that urban pollution has adverse effects on lung health. However, how urban pollution affects alveolar mesenchymal and epithelial stem cell niches remains unknown. This study aimed to determine how complex representative urban atmospheres alter alveolar stem cell niche properties. Mice were placed in an innovative chamber realistically simulating the atmosphere of a megalopolis, or "clean air," for 7 days. Lungs were collected, and fibroblasts and epithelial cells (EpCAM+) were isolated. Proliferative capacities of fibroblasts were tested by population doubling levels (PDL), and microarray analyses were performed. Fibroblasts and EpCAM+ cells from exposed, nonexposed, or naive mice were cocultured in organoid assays to assess the stem cell properties. Collagen deposition (Sirius red), lipofibroblasts (ADRP, COL1A1), myofibroblasts (αSMA), alveolar type 2 cells (AT2, SFTPC+), and alveolar differentiation intermediate cell [ADI, keratin-8-positive (KRT8+)/claudin-4-positive (CLDN4+)] markers were quantified in the lungs. Fibroblasts obtained from mice exposed to urban atmosphere had lower PDL and survival and produced fewer and smaller organoids. Microarray analysis showed a decrease of adipogenesis and an increase of genes associated with fibrosis, suggesting a lipofibroblast to myofibroblast transition. Collagen deposition and myofibroblast number increased in the lungs of urban atmosphere-exposed mice. AT2 number was reduced and associated with an increase in ADI cells KRT8+/CLDN4+. Furthermore, EpCAM+ cells from exposed mice also produced fewer and smaller organoids. In conclusion, urban atmosphere alters alveolar mesenchymal stem cell niche properties by inducing a lipofibroblast to myofibroblast shift. It also results in alveolar epithelial dysfunction and a fibrotic-like phenotype.NEW & NOTEWORTHY Urban pollution is known to have major adverse effects on lung health. To assess the effect of pollution on alveolar regeneration, we exposed adult mice to a simulated high-pollution urban atmosphere, using an innovative CESAM simulation chamber (Multiphase Atmospheric Experimental Simulation Chamber, https://cesam.cnrs.fr/). We demonstrated that urban atmosphere alters alveolar mesenchymal stem cell niche properties by inducing a lipofibroblast to myofibroblast shift and induces alveolar epithelial dysfunction.
Collapse
Affiliation(s)
- Randa Belgacemi
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, California, United States
| | | | - Grégoire Justeau
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
| | - Marylène Toigo
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
| | - Andrew Frauenpreis
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Rojda Yilmaz
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
| | | | - Mathieu Cazaunau
- Université Paris Est Créteil and Université Paris Cité, CNRS, LISA, Créteil, France
| | - Edouard Pangui
- Université Paris Est Créteil and Université Paris Cité, CNRS, LISA, Créteil, France
| | - Antonin Bergé
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, Paris, France
| | - Aline Gratien
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, Paris, France
| | | | - Saverio Bellusci
- German Center for Lung Research (DZL), Department of Pulmonary and Critical Care Medicine and Infectious Diseases, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University Giessen, Giessen, Germany
| | - Geneviève Derumeaux
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
- Service de Physiologie-Explorations Fonctionnelles, Hôpital Henri Mondor, Créteil, France
| | - Jorge Boczkowski
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, California, United States
| | - Patrice Coll
- Université Paris Cité and Université Paris Est Créteil, CNRS, LISA, Paris, France
| | - Sophie Lanone
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
| | - Laurent Boyer
- Université Paris Est Créteil, INSERM, IMRB, FHU Senec, Créteil, France
- Service de Physiologie-Explorations Fonctionnelles, Hôpital Henri Mondor, Créteil, France
| |
Collapse
|
13
|
Belgacemi R, Cherry C, El Alam I, Frauenpreis A, Glass I, Bellusci S, Danopoulos S, Al Alam D. Preferential FGF18/FGFR activity in pseudoglandular versus canalicular stage human lung fibroblasts. Front Cell Dev Biol 2023; 11:1220002. [PMID: 37701781 PMCID: PMC10493313 DOI: 10.3389/fcell.2023.1220002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/17/2023] [Indexed: 09/14/2023] Open
Abstract
Fibroblast growth factor (FGF) signaling is necessary for proper lung branching morphogenesis, alveolarization, and vascular development. Dysregulation of FGF activity has been implicated in various lung diseases. Recently, we showed that FGF18 promotes human lung branching morphogenesis by regulating mesenchymal progenitor cells. However, the underlying mechanisms remain unclear. Thus, we aimed to determine the role of FGF18 and its receptors (FGFR) in regulating mesenchymal cell proliferation, migration, and differentiation from pseudoglandular to canalicular stage. We performed siRNA assays to identify the specific FGFR(s) associated with FGF18-induced biological processes. We found that FGF18 increased proliferation and migration in human fetal lung fibroblasts (HFLF) from both stages. FGFR2/FGFR4 played a significant role in pseudoglandular stage. HFLF proliferation, while FGFR3/FGFR4 were involved in canalicular stage. FGF18 enhanced HFLF migration through FGFR2 and FGFR4 in pseudoglandular and canalicular stage, respectively. Finally, we provide evidence that FGF18 treatment leads to reduced expression of myofibroblast markers (ACTA2 and COL1A1) and increased expression of lipofibroblast markers (ADRP and PPARγ) in both stages HFLF. However, the specific FGF18/FGFR complex involved in this process varies depending on the stage. Our findings suggest that in context of human lung development, FGF18 tends to associate with distinct FGFRs to initiate specific biological processes on mesenchymal cells.
Collapse
Affiliation(s)
- Randa Belgacemi
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Caroline Cherry
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Imad El Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Andrew Frauenpreis
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Ian Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Saverio Bellusci
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UG-MLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
14
|
Roman J. Fibroblasts-Warriors at the Intersection of Wound Healing and Disrepair. Biomolecules 2023; 13:945. [PMID: 37371525 DOI: 10.3390/biom13060945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/07/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023] Open
Abstract
Wound healing is triggered by inflammation elicited after tissue injury. Mesenchymal cells, specifically fibroblasts, accumulate in the injured tissues, where they engage in tissue repair through the expression and assembly of extracellular matrices that provide a scaffold for cell adhesion, the re-epithelialization of tissues, the production of soluble bioactive mediators that promote cellular recruitment and differentiation, and the regulation of immune responses. If appropriately deployed, these processes promote adaptive repair, resulting in the preservation of the tissue structure and function. Conversely, the dysregulation of these processes leads to maladaptive repair or disrepair, which causes tissue destruction and a loss of organ function. Thus, fibroblasts not only serve as structural cells that maintain tissue integrity, but are key effector cells in the process of wound healing. The review will discuss the general concepts about the origins and heterogeneity of this cell population and highlight the specific fibroblast functions disrupted in human disease. Finally, the review will explore the role of fibroblasts in tissue disrepair, with special attention to the lung, the role of aging, and how alterations in the fibroblast phenotype underpin disorders characterized by pulmonary fibrosis.
Collapse
Affiliation(s)
- Jesse Roman
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care and The Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
15
|
Cao S, Feng H, Yi H, Pan M, Lin L, Zhang YS, Feng Z, Liang W, Cai B, Li Q, Xiong Z, Shen Q, Ke M, Zhao X, Chen H, He Q, Min M, Cai Q, Liu H, Wang J, Pei D, Chen J, Ma Y. Single-cell RNA sequencing reveals the developmental program underlying proximal-distal patterning of the human lung at the embryonic stage. Cell Res 2023:10.1038/s41422-023-00802-6. [PMID: 37085732 PMCID: PMC10119843 DOI: 10.1038/s41422-023-00802-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/21/2023] [Indexed: 04/23/2023] Open
Abstract
The lung is the primary respiratory organ in human, in which the proximal airway and the distal alveoli are responsible for air conduction and gas exchange, respectively. However, the regulation of proximal-distal patterning at the embryonic stage of human lung development is largely unknown. Here we investigated the early lung development of human embryos at weeks 4-8 post fertilization (Carnegie stages 12-21) using single-cell RNA sequencing, and obtained a transcriptomic atlas of 169,686 cells. We observed discernible gene expression patterns of proximal and distal epithelia at week 4, upon the initiation of lung organogenesis. Moreover, we identified novel transcriptional regulators of the patterning of proximal (e.g., THRB and EGR3) and distal (e.g., ETV1 and SOX6) epithelia. Further dissection revealed various stromal cell populations, including an early-embryonic BDNF+ population, providing a proximal-distal patterning niche with spatial specificity. In addition, we elucidated the cell fate bifurcation and maturation of airway and vascular smooth muscle progenitor cells at the early stage of lung development. Together, our study expands the scope of human lung developmental biology at early embryonic stages. The discovery of intrinsic transcriptional regulators and novel niche providers deepens the understanding of epithelial proximal-distal patterning in human lung development, opening up new avenues for regenerative medicine.
Collapse
Affiliation(s)
- Shangtao Cao
- Guangzhou Laboratory, Guangzhou, Guangdong, China.
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Huijian Feng
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Hongyan Yi
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Mengjie Pan
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Lihui Lin
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Yao Santo Zhang
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Ziyu Feng
- Guangzhou Laboratory, Guangzhou, Guangdong, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weifang Liang
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Baomei Cai
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Key Laboratory of the Ministry of Education for Reproductive Health Diseases Research and Translation, Hainan Medical University, Haikou, Hainan, China
| | - Zhi Xiong
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Qingmei Shen
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Minjing Ke
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xing Zhao
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Key Laboratory of the Ministry of Education for Reproductive Health Diseases Research and Translation, Hainan Medical University, Haikou, Hainan, China
| | - Huilin Chen
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qina He
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Key Laboratory of the Ministry of Education for Reproductive Health Diseases Research and Translation, Hainan Medical University, Haikou, Hainan, China
| | - Mingwei Min
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Quanyou Cai
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - He Liu
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jie Wang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China.
| | - Jiekai Chen
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China.
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China.
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Hainan Provincial Clinical Research Center for Thalassemia, Reproductive Medical Center, International Technology Cooperation Base "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
- Key Laboratory of the Ministry of Education for Reproductive Health Diseases Research and Translation, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
16
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
17
|
Tung VSK, Mathews F, Boruk M, Suppa G, Foronjy R, Pato M, Pato C, Knowles JA, Evgrafov OV. Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534295. [PMID: 37034711 PMCID: PMC10081251 DOI: 10.1101/2023.03.28.534295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Study of the neurodevelopmental molecular mechanisms of schizophrenia requires the development of adequate biological models such as patient-derived cells and their derivatives. We previously used cell lines with neural progenitor properties (CNON) derived from superior or middle turbinates of patients with schizophrenia and control groups to study gene expression specific to schizophrenia. In this study, we compared single cell-RNA seq data from two CNON cell lines, one derived from an individual with schizophrenia (SCZ) and the other from a control group, with two biopsy samples from the middle turbinate (MT), also from an individual with SCZ and a control. In addition, we compared our data with previously published data from olfactory neuroepithelium (1). Our data demonstrated that CNON originated from a single cell type which is present both in middle turbinate and olfactory neuroepithelium. CNON express multiple markers of mesenchymal cells. In order to define relatedness of CNON to the developing human brain, we also compared CNON datasets with scRNA-seq data of embryonic brain (2) and found that the expression profile of CNON very closely matched one of the cell types in the embryonic brain. Finally, we evaluated differences between SCZ and control samples to assess usability and potential benefits of using single cell RNA-seq of CNON to study etiology of schizophrenia.
Collapse
Affiliation(s)
| | - Fasil Mathews
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Marina Boruk
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Gabrielle Suppa
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Robert Foronjy
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| | | | - Carlos Pato
- Department of Psychiatry, Rutgers University
| | - James A. Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Oleg V. Evgrafov
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| |
Collapse
|
18
|
Fuentes-Mateos R, Santos E, Fernández-Medarde A. Optimized Protocol for Isolation and Culture of Murine Neonatal Primary Lung Fibroblasts. Methods Protoc 2023; 6:14. [PMID: 36827501 PMCID: PMC9966303 DOI: 10.3390/mps6010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
During all the stages of lung development, the lung mesoderm (or mesenchyme) is closely related to the endoderm, and their cross-regulation promotes, controls, and drives all lung developmental processes. Generation of 3D organoids in vitro, RNA assays, and mitochondrial respiration studies are used to analyze lung development and regeneration to better understand the interactions between epithelium and mesenchyme, as well as for the study of redox alterations and the metabolic status of the cells. Moreover, to avoid using immortalized cell lines in these in vitro approaches, standardized murine neonatal primary lung fibroblast isolation techniques are essential. Here, we present an optimized method to isolate, culture, and freeze primary lung fibroblasts from neonatal lungs. Our current method includes step-by-step instructions accompanied by graphical explanations and critical steps.
Collapse
Affiliation(s)
| | - Eugenio Santos
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca/CSIC and CIBERONC, 37007 Salamanca, Spain
| | - Alberto Fernández-Medarde
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca/CSIC and CIBERONC, 37007 Salamanca, Spain
| |
Collapse
|
19
|
Tanneberger AE, Weiss DJ, Magin CM. An Introduction to Engineering and Modeling the Lung. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:1-13. [PMID: 37195523 DOI: 10.1007/978-3-031-26625-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Over the last decade, the field of lung biology has evolved considerably due to many advancements, including the advent of single-cell RNA (scRNA) sequencing, induced pluripotent stem cell (iPSC) reprogramming, and 3D cell and tissue culture. Despite rigorous research and tireless efforts, chronic pulmonary diseases remain the third leading cause of death globally, with transplantation being the only option for treating end-stage disease. This chapter will introduce the broader impacts of understanding lung biology in health and disease, provide an overview of lung physiology and pathophysiology, and summarize the key takeaways from each chapter describing engineering translational models of lung homeostasis and disease. This book is divided into broad topic areas containing chapters covering basic biology, engineering approaches, and clinical perspectives related to (1) the developing lung, (2) the large airways, (3) the mesenchyme and parenchyma, (4) the pulmonary vasculature, and (5) the interface between lungs and medical devices. Each section highlights the underlying premise that engineering strategies, when applied in collaboration with cell biologists and pulmonary physicians, will address critical challenges in pulmonary health care.
Collapse
Affiliation(s)
- Alicia E Tanneberger
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, USA
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO, USA.
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Lahmar Z, Ahmed E, Fort A, Vachier I, Bourdin A, Bergougnoux A. Hedgehog pathway and its inhibitors in chronic obstructive pulmonary disease (COPD). Pharmacol Ther 2022; 240:108295. [PMID: 36191777 DOI: 10.1016/j.pharmthera.2022.108295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
COPD affects millions of people and is now ranked as the third leading cause of death worldwide. This largely untreatable chronic airway disease results in irreversible destruction of lung architecture. The small lung hypothesis is now supported by epidemiological, physiological and clinical studies. Accordingly, the early and severe COPD phenotype carries the most dreadful prognosis and finds its roots during lung growth. Pathophysiological mechanisms remain poorly understood and implicate individual susceptibility (genetics), a large part of environmental factors (viral infections, tobacco consumption, air pollution) and the combined effects of those triggers on gene expression. Genetic susceptibility is most likely involved as the disease is severe and starts early in life. The latter observation led to the identification of Mendelian inheritance via disease-causing variants of SERPINA1 - known as the basis for alpha-1 anti-trypsin deficiency, and TERT. In the last two decades multiple genome wide association studies (GWAS) identified many single nucleotide polymorphisms (SNPs) associated with COPD. High significance SNPs are located in 4q31 near HHIP which encodes an evolutionarily highly conserved physiological inhibitor of the Hedgehog signaling pathway (HH). HHIP is critical to several in utero developmental lung processes. It is also implicated in homeostasis, injury response, epithelial-mesenchymal transition and tumor resistance to apoptosis. A few studies have reported decreased HHIP RNA and protein levels in human adult COPD lungs. HHIP+/- murine models led to emphysema. HH pathway inhibitors, such as vismodegib and sonidegib, are already validated in oncology, whereas other drugs have evidenced in vitro effects. Targeting the Hedgehog pathway could lead to a new therapeutic avenue in COPD. In this review, we focused on the early and severe COPD phenotype and the small lung hypothesis by exploring genetic susceptibility traits that are potentially treatable, thus summarizing promising therapeutics for the future.
Collapse
Affiliation(s)
- Z Lahmar
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France
| | - E Ahmed
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Fort
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - I Vachier
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bourdin
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bergougnoux
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France; Laboratoire de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
21
|
Wang Y, Dong X, Pan C, Zhu C, Qi H, Wang Y, Wei H, Xie Q, Wu L, Shen H, Li S, Xie Y. Single-cell transcriptomic characterization reveals the landscape of airway remodeling and inflammation in a cynomolgus monkey model of asthma. Front Immunol 2022; 13:1040442. [PMID: 36439114 PMCID: PMC9685410 DOI: 10.3389/fimmu.2022.1040442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/20/2022] [Indexed: 06/22/2024] Open
Abstract
Monkey disease models, which are comparable to humans in terms of genetic, anatomical, and physiological characteristics, are important for understanding disease mechanisms and evaluating the efficiency of biological treatments. Here, we established an A.suum-induced model of asthma in cynomolgus monkeys to profile airway inflammation and remodeling in the lungs by single-cell RNA sequencing (scRNA-seq). The asthma model results in airway hyperresponsiveness and remodeling, demonstrated by pulmonary function test and histological characterization. scRNA-seq reveals that the model elevates the numbers of stromal, epithelial and mesenchymal cells (MCs). Particularly, the model increases the numbers of endothelial cells (ECs), fibroblasts (Fibs) and smooth muscle cells (SMCs) in the lungs, with upregulated gene expression associated with cell functions enriched in cell migration and angiogenesis in ECs and Fibs, and VEGF-driven cell proliferation, apoptotic process and complement activation in SMCs. Interestingly, we discover a novel Fib subtype that mediates type I inflammation in the asthmatic lungs. Moreover, MCs in the asthmatic lungs are found to regulate airway remodeling and immunological responses, with elevated gene expression enriched in cell migration, proliferation, angiogenesis and innate immunological responses. Not only the numbers of epithelial cells in the asthmatic lungs change at the time of lung tissue collection, but also their gene expressions are significantly altered, with an enrichment in the biological processes of IL-17 signaling pathway and apoptosis in the majority of subtypes of epithelial cells. Moreover, the ubiquitin process and DNA repair are more prevalent in ciliated epithelial cells. Last, cell-to-cell interaction analysis reveals a complex network among stromal cells, MCs and macrophages that contribute to the development of asthma and airway remodeling. Our findings provide a critical resource for understanding the principle underlying airway remodeling and inflammation in a monkey model of asthma, as well as valuable hints for the future treatment of asthma, especially the airway remodeling-characterized refractory asthma.
Collapse
Affiliation(s)
- Yingshuo Wang
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyan Dong
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Caizhe Pan
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Cihang Zhu
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hantao Qi
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Wang
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wei
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiangmin Xie
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Respiratory Drugs Research, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Wu
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijuan Shen
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuxian Li
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicheng Xie
- Department of Pulmonology, The Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Su X, Wu W, Zhu Z, Lin X, Zeng Y. The effects of epithelial-mesenchymal transitions in COPD induced by cigarette smoke: an update. Respir Res 2022; 23:225. [PMID: 36045410 PMCID: PMC9429334 DOI: 10.1186/s12931-022-02153-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Cigarette smoke is a complex aerosol containing a large number of compounds with a variety of toxicity and carcinogenicity. Long-term exposure to cigarette smoke significantly increases the risk of a variety of diseases, including chronic obstructive pulmonary disease (COPD) and lung cancer. Epithelial–mesenchymal transition (EMT) is a unique biological process, that refers to epithelial cells losing their polarity and transforming into mobile mesenchymal cells, playing a crucial role in organ development, fibrosis, and cancer progression. Numerous recent studies have shown that EMT is an important pathophysiological process involved in airway fibrosis, airway remodeling, and malignant transformation of COPD. In this review, we summarized the effects of cigarette smoke on the development and progression of COPD and focus on the specific changes and underlying mechanisms of EMT in COPD induced by cigarette smoke. We spotlighted the signaling pathways involved in EMT induced by cigarette smoke and summarize the current research and treatment approaches for EMT in COPD, aiming to provide ideas for potential new treatment and research directions.
Collapse
Affiliation(s)
- Xiaoshan Su
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Weijing Wu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Zhixing Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Xiaoping Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Respirology Medicine Centre of Fujian Province, Quanzhou, China.
| |
Collapse
|
23
|
Guo P, Li R, Piao TH, Wang CL, Wu XL, Cai HY. Pathological Mechanism and Targeted Drugs of COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:1565-1575. [PMID: 35855746 PMCID: PMC9288175 DOI: 10.2147/copd.s366126] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/04/2022] [Indexed: 01/17/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) includes chronic bronchitis, emphysema, and small airway obstruction. Incompletely reversible airflow limitation, inflammation, excessive mucus secretion and bronchial mucosal epithelial lesions are the main pathological basis of the disease. The prevalence of COPD is increasingly worldwide, which has caused the burden on individuals and society. This paper summarizes the pathogenesis of COPD and clarifies the effect and mechanism of the latest targeted drugs for COPD. Besides, we focus on NOD-like receptor thermal protein domain associated protein 3 inflammasome (NLRP3 inflammasome). NLRP3 can promote production of interleukin-1β (IL-1β) and interleukin-18 (IL-18). NLRP3 is an important factor in the migratory aggregation of macrophages and neutrophils and the generation of oxidative stress. Inhibition of NLRP3 inflammasome indirectly blocks the inflammatory effects of IL-1β and IL-18, which may be regarded as an ideal target for COPD treatment.
Collapse
Affiliation(s)
- Peng Guo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Changchun, 130000, People's Republic of China
| | - Rui Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Tie Hua Piao
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Chun Lan Wang
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Xiao Lu Wu
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| | - Hong Yan Cai
- Pulmonology Department, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, People's Republic of China
| |
Collapse
|
24
|
Lin CR, Bahmed K, Kosmider B. Impaired Alveolar Re-Epithelialization in Pulmonary Emphysema. Cells 2022; 11:2055. [PMID: 35805139 PMCID: PMC9265977 DOI: 10.3390/cells11132055] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/24/2023] Open
Abstract
Alveolar type II (ATII) cells are progenitors in alveoli and can repair the alveolar epithelium after injury. They are intertwined with the microenvironment for alveolar epithelial cell homeostasis and re-epithelialization. A variety of ATII cell niches, transcription factors, mediators, and signaling pathways constitute a specific environment to regulate ATII cell function. Particularly, WNT/β-catenin, YAP/TAZ, NOTCH, TGF-β, and P53 signaling pathways are dynamically involved in ATII cell proliferation and differentiation, although there are still plenty of unknowns regarding the mechanism. However, an imbalance of alveolar cell death and proliferation was observed in patients with pulmonary emphysema, contributing to alveolar wall destruction and impaired gas exchange. Cigarette smoking causes oxidative stress and is the primary cause of this disease development. Aberrant inflammatory and oxidative stress responses result in loss of cell homeostasis and ATII cell dysfunction in emphysema. Here, we discuss the current understanding of alveolar re-epithelialization and altered reparative responses in the pathophysiology of this disease. Current therapeutics and emerging treatments, including cell therapies in clinical trials, are addressed as well.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
25
|
Merimi M, Rahmani S, Afailal Tribak A, Bouhtit F, Fahmi H, Najar M. Fundamental and Applied Advances in Stem Cell Therapeutic Research. Cells 2022; 11:cells11121976. [PMID: 35741105 PMCID: PMC9221776 DOI: 10.3390/cells11121976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
We are pleased to present this Special Issue of Cells, entitled 'Feature Papers in Stem Cells' [...].
Collapse
Affiliation(s)
- Makram Merimi
- Experimental Hematology, Jules Bordet Institute, Unive and nd nd ité Libre de Bruxelles, 1070 Bruxelles, Belgium; (M.M.); (S.R.); (A.A.T.); (F.B.)
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Saida Rahmani
- Experimental Hematology, Jules Bordet Institute, Unive and nd nd ité Libre de Bruxelles, 1070 Bruxelles, Belgium; (M.M.); (S.R.); (A.A.T.); (F.B.)
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Ahmed Afailal Tribak
- Experimental Hematology, Jules Bordet Institute, Unive and nd nd ité Libre de Bruxelles, 1070 Bruxelles, Belgium; (M.M.); (S.R.); (A.A.T.); (F.B.)
| | - Fatima Bouhtit
- Experimental Hematology, Jules Bordet Institute, Unive and nd nd ité Libre de Bruxelles, 1070 Bruxelles, Belgium; (M.M.); (S.R.); (A.A.T.); (F.B.)
- Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada;
| | - Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Department of Medicine, University of Montreal, Montreal, QC H2X 0A9, Canada;
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium
- Correspondence:
| |
Collapse
|