1
|
Martín-Vicente P, López-Martínez C, López-Alonso I, Exojo-Ramírez SM, Duarte-Herrera ID, Amado-Rodríguez L, Ordoñez I, Cuesta-Llavona E, Gómez J, Campo N, O'Kane CM, McAuley DF, Huidobro C, Albaiceta GM. Mechanical Stretch Induces Senescence of Lung Epithelial Cells and Drives Fibroblast Activation by Paracrine Mechanisms. Am J Respir Cell Mol Biol 2025; 72:195-205. [PMID: 39133930 DOI: 10.1165/rcmb.2023-0449oc] [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: 12/19/2023] [Accepted: 08/12/2024] [Indexed: 02/01/2025] Open
Abstract
Severe lung injury requiring mechanical ventilation may lead to secondary fibrosis. Senescence, a cell response characterized by cell cycle arrest and a shift toward a proinflammatory/profibrotic phenotype, is one of the involved mechanisms. In this study, we explore the contribution of mechanical stretch as a trigger of senescence of the respiratory epithelium and its link with fibrosis. Human lung epithelial cells and fibroblasts were exposed in vitro to mechanical stretch, and senescence was assessed. In addition, fibroblasts were exposed to culture media preconditioned by senescent epithelial cells, and their activation was studied. Transcriptomic profiles from stretched, senescent epithelial cells and activated fibroblasts were combined to identify potential activated pathways. Finally, the senolytic effects of digoxin were tested in these models. Mechanical stretch induced senescence in lung epithelial cells, but not in fibroblasts. This stretch-induced senescence has specific features compared with senescence induced by doxorubicin. Fibroblasts were activated after exposure to supernatants conditioned by epithelial senescent cells. Transcriptomic analyses revealed Notch signaling as potentially responsible for the epithelial-mesenchymal cross-talk, because blockade of this pathway inhibits fibroblast activation. Treatment with digoxin reduced the percentage of senescent cells after stretch and ameliorated the fibroblast response to preconditioned media. These results suggest that lung fibrosis in response to mechanical stretch may be caused by the paracrine effects of senescent cells. This pathogenetic mechanism can be pharmacologically manipulated to improve lung repair.
Collapse
Affiliation(s)
- Paula Martín-Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
| | - Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
| | - Inés López-Alonso
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
| | - Sara M Exojo-Ramírez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
| | - Israel David Duarte-Herrera
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
- Departamento de Medicina
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
- Unidad de Cuidados Intensivos Cardiológicos and
| | - Irene Ordoñez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
| | - Elias Cuesta-Llavona
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Genética, Hospital Universitario Central de Asturias, Oviedo, Spain; and
| | - Juan Gómez
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Servicio de Genética, Hospital Universitario Central de Asturias, Oviedo, Spain; and
| | - Natalia Campo
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | | | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Respiratorias, Madrid, Spain
- Unidad de Cuidados Intensivos Cardiológicos and
| |
Collapse
|
2
|
Gautam RK, Laltanpuia, Singh N, Kushwaha S. A particle of concern: explored and proposed underlying mechanisms of microplastic-induced lung damage and pulmonary fibrosis. Inhal Toxicol 2025; 37:1-17. [PMID: 39932476 DOI: 10.1080/08958378.2025.2461048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE In the past decade, microplastics (MPs) have drawn significant attention as widespread environmental contaminants, with research increasingly highlighting their harmful effects on respiratory health in aquatic and terrestrial organisms. Findings revealed microplastics in human lung tissues, raising concerns about their potential role in damaging lung tissue integrity and contributing to pulmonary fibrosis-a chronic inflammatory condition characterized by scarring of lung epithelial tissues due to accumulated extracellular matrix, triggered by factors such as alcohol, pathogens, genetic mutations, and environmental pollutants. OBJECTIVE In this review, we explore both well-studied and lesser-studied mechanisms and signaling pathways, aiming to shed light on how microplastics might act as mediators that activate distinct, often overlooked signaling cascades. MATERIALS AND METHODS This review searched PubMed and Google Scholar using keywords like "plastic," "microplastic," "lung fibrosis," "pulmonary system," "exposure route," and "signaling pathways," combined with "OR" and "AND" in singular and plural forms. RESULTS These pathways could not only induce lung damage but also play a significant role in the development of pulmonary fibrosis. DISCUSSION AND CONCLUSIONS These signaling pathways could also be targeted to reduce microplastic-induced pulmonary fibrosis, opening new avenues for future treatments.
Collapse
Affiliation(s)
- Rohit Kumar Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| | - Laltanpuia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| | - Nishant Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow, India
| |
Collapse
|
3
|
Jones DL, Morley MP, Li X, Ying Y, Zhao G, Schaefer SE, Rodriguez LR, Cardenas-Diaz FL, Li S, Zhou S, Chembazhi UV, Kim M, Shen C, Nottingham A, Lin SM, Cantu E, Diamond JM, Basil MC, Vaughan AE, Morrisey EE. An injury-induced mesenchymal-epithelial cell niche coordinates regenerative responses in the lung. Science 2024; 386:eado5561. [PMID: 39666855 DOI: 10.1126/science.ado5561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/07/2024] [Accepted: 10/14/2024] [Indexed: 12/14/2024]
Abstract
Severe lung injury causes airway basal stem cells to migrate and outcompete alveolar stem cells, resulting in dysplastic repair. We found that this "stem cell collision" generates an injury-induced tissue niche containing keratin 5+ epithelial cells and plastic Pdgfra+ mesenchymal cells. Single-cell analysis revealed that the injury-induced niche is governed by mesenchymal proliferation and Notch signaling, which suppressed Wnt/Fgf signaling in the injured niche. Conversely, loss of Notch signaling rewired alveolar signaling patterns to promote functional regeneration and gas exchange. Signaling patterns in injury-induced niches can differentiate fibrotic from degenerative human lung diseases through altering the direction of Wnt/Fgf signaling. Thus, we have identified an injury-induced niche in the lung with the ability to discriminate human lung disease phenotypes.
Collapse
Affiliation(s)
- Dakota L Jones
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinyuan Li
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yun Ying
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gan Zhao
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E Schaefer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Luis R Rodriguez
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fabian L Cardenas-Diaz
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ullas V Chembazhi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mijeong Kim
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chen Shen
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana Nottingham
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Lin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
He Y, Yang F, Yang L, Yuan H, You Y, Chen Y, Wu X, Min H, Chen J, Li C. Mechanics-activated fibroblasts promote pulmonary group 2 innate lymphoid cell plasticity propelling silicosis progression. Nat Commun 2024; 15:9770. [PMID: 39532893 PMCID: PMC11557922 DOI: 10.1038/s41467-024-54174-5] [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: 10/17/2023] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Crystalline silica (CS) particle exposure leads to silicosis which is characterized as progressive fibrosis. Fibroblasts are vital effector cells in fibrogenesis. Emerging studies have identified immune sentinel roles for fibroblasts in chronic disease, while their immune-modulatory roles in silicosis remain unclear. Herein, we show that group 2 innate lymphoid cell (ILC2) conversion to ILC1s is closely involved in silicosis progression, which is mediated by activated fibroblasts via interleukin (IL)-18. Mechanistically, Notch3 signaling in mechanics-activated fibroblasts modulates IL-18 production via caspase 1 activity. The mouse-specific Notch3 knockout in fibroblasts retards pulmonary fibrosis progression that is linked to attenuated ILC conversion. Our results indicate that activated fibroblasts in silicotic lungs are regulators of ILC2-ILC1 conversion, associated with silicosis progression via the Notch3-IL-18 signaling axis. This finding broadens our understanding of immune-modulatory mechanisms in silicosis, and indicates potential therapeutic targets for lung fibrotic diseases.
Collapse
Affiliation(s)
- Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Fan Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Lin Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Yinghui Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Xiulin Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
5
|
Zhang X, Xu Z, Chen Q, Zhou Z. Notch signaling regulates pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1450038. [PMID: 39450276 PMCID: PMC11499121 DOI: 10.3389/fcell.2024.1450038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Pulmonary fibrosis is a progressive interstitial lung disease associated with aging. The pathogenesis of pulmonary fibrosis remains unclear, however, alveolar epithelial cell injury, myofibroblast activation, and extracellular matrix (ECM) accumulation are recognized as key contributors. Moreover, recent studies have implicated cellular senescence, endothelial-mesenchymal transition (EndMT), and epigenetic modifications in the pathogenesis of fibrotic diseases. Various signaling pathways regulate pulmonary fibrosis, including the TGF-β, Notch, Wnt, Hedgehog, and mTOR pathways. Among these, the TGF-β pathway is extensively studied, while the Notch pathway has emerged as a recent research focus. The Notch pathway influences the fibrotic process by modulating immune cell differentiation (e.g., macrophages, lymphocytes), inhibiting autophagy, and promoting interstitial transformation. Consequently, inhibiting Notch signaling represents a promising approach to mitigating pulmonary fibrosis. In this review, we discuss the role of Notch signaling pathway in pulmonary fibrosis, aiming to offer insights for future therapeutic investigations.
Collapse
Affiliation(s)
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | | | | |
Collapse
|
6
|
Bakalenko N, Kuznetsova E, Malashicheva A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int J Mol Sci 2024; 25:10803. [PMID: 39409132 PMCID: PMC11477142 DOI: 10.3390/ijms251910803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Fibrosis is a major medical challenge, as it leads to irreversible tissue remodeling and organ dysfunction. Its progression contributes significantly to morbidity and mortality worldwide, with limited therapeutic options available. Extensive research on the molecular mechanisms of fibrosis has revealed numerous factors and signaling pathways involved. However, the interactions between these pathways remain unclear. A comprehensive understanding of the entire signaling network that drives fibrosis is still missing. The TGF-β and Notch signaling pathways play a key role in fibrogenesis, and this review focuses on their functional interplay and molecular mechanisms. Studies have shown synergy between TGF-β and Notch cascades in fibrosis, but antagonistic interactions can also occur, especially in cardiac fibrosis. The molecular mechanisms of these interactions vary depending on the cell context. Understanding these complex and context-dependent interactions is crucial for developing effective strategies for treating fibrosis.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia; (N.B.); (E.K.)
| |
Collapse
|
7
|
Wang J, Li K, Hao D, Li X, Zhu Y, Yu H, Chen H. Pulmonary fibrosis: pathogenesis and therapeutic strategies. MedComm (Beijing) 2024; 5:e744. [PMID: 39314887 PMCID: PMC11417429 DOI: 10.1002/mco2.744] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive lung disease characterized by extensive alterations of cellular fate and function and excessive accumulation of extracellular matrix, leading to lung tissue scarring and impaired respiratory function. Although our understanding of its pathogenesis has increased, effective treatments remain scarce, and fibrotic progression is a major cause of mortality. Recent research has identified various etiological factors, including genetic predispositions, environmental exposures, and lifestyle factors, which contribute to the onset and progression of PF. Nonetheless, the precise mechanisms by which these factors interact to drive fibrosis are not yet fully elucidated. This review thoroughly examines the diverse etiological factors, cellular and molecular mechanisms, and key signaling pathways involved in PF, such as TGF-β, WNT/β-catenin, and PI3K/Akt/mTOR. It also discusses current therapeutic strategies, including antifibrotic agents like pirfenidone and nintedanib, and explores emerging treatments targeting fibrosis and cellular senescence. Emphasizing the need for omni-target approaches to overcome the limitations of current therapies, this review integrates recent findings to enhance our understanding of PF and contribute to the development of more effective prevention and management strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Jianhai Wang
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Kuan Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - De Hao
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
| | - Xue Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Yu Zhu
- Department of Clinical LaboratoryNankai University Affiliated Third Central HospitalTianjinChina
- Department of Clinical LaboratoryThe Third Central Hospital of TianjinTianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesArtificial Cell Engineering Technology Research Center of TianjinTianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Hongzhi Yu
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Huaiyong Chen
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| |
Collapse
|
8
|
Kooistra T, Saez B, Roche M, Egea-Zorrilla A, Li D, Anketell D, Nguyen N, Villoria J, Gillis J, Petri E, Vera L, Blasco-Iturri Z, Smith NP, Alladina J, Zhang Y, Vinarsky V, Shivaraju M, Sheng SL, Gonzalez-Celeiro M, Mou H, Waghray A, Lin B, Paksa A, Yanger K, Tata PR, Zhao R, Causton B, Zulueta JJ, Prosper F, Cho JL, Villani AC, Haber A, Rajagopal J, Medoff BD, Pardo-Saganta A. Airway basal stem cells are necessary for the maintenance of functional intraepithelial airway macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600501. [PMID: 38979172 PMCID: PMC11230263 DOI: 10.1101/2024.06.25.600501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Adult stem cells play a crucial role in tissue homeostasis and repair through multiple mechanisms. In addition to being able to replace aged or damaged cells, stem cells provide signals that contribute to the maintenance and function of neighboring cells. In the lung, airway basal stem cells also produce cytokines and chemokines in response to inhaled irritants, allergens, and pathogens, which affect specific immune cell populations and shape the nature of the immune response. However, direct cell-to-cell signaling through contact between airway basal stem cells and immune cells has not been demonstrated. Recently, a unique population of intraepithelial airway macrophages (IAMs) has been identified in the murine trachea. Here, we demonstrate that IAMs require Notch signaling from airway basal stem cells for maintenance of their differentiated state and function. Furthermore, we demonstrate that Notch signaling between airway basal stem cells and IAMs is required for antigen-induced allergic inflammation only in the trachea where the basal stem cells are located whereas allergic responses in distal lung tissues are preserved consistent with a local circuit linking stem cells to proximate immune cells. Finally, we demonstrate that IAM-like cells are present in human conducting airways and that these cells display Notch activation, mirroring their murine counterparts. Since diverse lung stem cells have recently been identified and localized to specific anatomic niches along the proximodistal axis of the respiratory tree, we hypothesize that the direct functional coupling of local stem cell-mediated regeneration and immune responses permits a compartmentalized inflammatory response.
Collapse
|
9
|
Wang X, Zhou J, Li X, Liu C, Liu L, Cui H. The Role of Macrophages in Lung Fibrosis and the Signaling Pathway. Cell Biochem Biophys 2024; 82:479-488. [PMID: 38536578 DOI: 10.1007/s12013-024-01253-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/13/2024] [Indexed: 08/25/2024]
Abstract
Lung fibrosis is a dysregulated repair process caused by excessive deposition of extracellular matrix that can severely affect respiratory function. Macrophages are a group of immune cells that have multiple functions and can perform a variety of roles. Lung fibrosis develops with the involvement of pro-inflammatory and pro-fibrotic factors secreted by macrophages. The balance between M1 and M2 macrophages has been proposed to play a role in determining the trend and severity of lung fibrosis. New avenues and concepts for preventing and treating lung fibrosis have emerged in recent years through research on mitochondria, Gab proteins, and exosomes. The main topic of this essay is the impact that mitochondria, Gab proteins, and exosomes have on macrophage polarization. In addition, the potential of these factors as targets to enhance lung fibrosis is also explored. We have also collated the functions and mechanisms of signaling pathways associated with the regulation of macrophage polarization such as Notch, TGF-β/Smad, JAK-STAT and cGAS-STING. The goal of this article is to explain the potential benefits of focusing on macrophage polarization as a way to relieve lung fibrosis. We aspire to provide valuable insights that could lead to enhancements in the treatment of this condition.
Collapse
Affiliation(s)
- Xingmei Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, China
- Center of Medical Functional Experiment, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Jiaxu Zhou
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, China
- Center of Medical Functional Experiment, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Xinrui Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, China
- Center of Medical Functional Experiment, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Chang Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, China
- Center of Medical Functional Experiment, Yanbian University Medical College, Yanji, 133002, Jilin, China
| | - Lan Liu
- Department of Pathology, Affiliated Hospital of Yanbian University, Yanji, 133002, Jilin, China.
| | - Hong Cui
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, 133002, Jilin, China.
- Center of Medical Functional Experiment, Yanbian University Medical College, Yanji, 133002, Jilin, China.
| |
Collapse
|
10
|
Henneke I, Pilz C, Wilhelm J, Alexopoulos I, Ezaddoustdar A, Mukhametshina R, Weissmann N, Ghofrani HA, Grimminger F, Seeger W, Schermuly RT, Wygrecka M, Kojonazarov B. Microscopic computed tomography with AI-CNN-powered image analysis: the path to phenotype bleomycin-induced lung injury. Am J Physiol Cell Physiol 2024; 326:C1637-C1647. [PMID: 38646782 DOI: 10.1152/ajpcell.00708.2023] [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: 12/20/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Bleomycin (BLM)-induced lung injury in mice is a valuable model for investigating the molecular mechanisms that drive inflammation and fibrosis and for evaluating potential therapeutic approaches to treat the disease. Given high variability in the BLM model, it is critical to accurately phenotype the animals in the course of an experiment. In the present study, we aimed to demonstrate the utility of microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation for rapid phenotyping of BLM mice. µCT was performed in freely breathing C57BL/6J mice under isoflurane anesthesia on days 7 and 21 after BLM administration. Terminal invasive lung function measurement and histological assessment of the left lung collagen content were conducted as well. µCT image analysis demonstrated gradual and time-dependent development of lung injury as evident by alterations in the lung density, air-to-tissue volume ratio, and lung aeration in mice treated with BLM. The right and left lung were unequally affected. µCT-derived parameters such as lung density, air-to-tissue volume ratio, and nonaerated lung volume correlated well with the invasive lung function measurement and left lung collagen content. Our study demonstrates the utility of AI-CNN-powered µCT image analysis for rapid and accurate phenotyping of BLM mice in the course of disease development and progression.NEW & NOTEWORTHY Microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation is a rapid and powerful tool for noninvasive phenotyping of bleomycin mice over the course of the disease. This, in turn, allows earlier and more reliable identification of therapeutic effects of new drug candidates, ultimately leading to the reduction of unnecessary procedures in animals in pharmacological research.
Collapse
Affiliation(s)
- Ingrid Henneke
- Experimental Lung Disease Models Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Christina Pilz
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Jochen Wilhelm
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Genomics and Bioinformatics Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Multyscale Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Aysan Ezaddoustdar
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Center for Infection and Genomics of the Lung (CIGL), Faculty of Medicine, Justus Liebig University (JLU), Giessen, Germany
| | - Regina Mukhametshina
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Small Animal Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Norbert Weissmann
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph T Schermuly
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
| | - Malgorzata Wygrecka
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Center for Infection and Genomics of the Lung (CIGL), Faculty of Medicine, Justus Liebig University (JLU), Giessen, Germany
- CSL Behring Innovation GmbH, Marburg, Germany
| | - Baktybek Kojonazarov
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Murburg Lung Center (UGMLC), Justus Liebig University (JLU), Giessen, Germany
- Small Animal Imaging Platform, Institute for Lung Health (ILH), Justus Liebig University (JLU), Giessen, Germany
| |
Collapse
|
11
|
Xu S, Jiemy WF, Brouwer E, Burgess JK, Heeringa P, van der Geest KSM, Alba-Rovira R, Corbera-Bellalta M, Boots AH, Cid MC, Sandovici M. Current evidence on the role of fibroblasts in large-vessel vasculitides: From pathogenesis to therapeutics. Autoimmun Rev 2024; 23:103574. [PMID: 38782083 DOI: 10.1016/j.autrev.2024.103574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Large-vessel vasculitides (LVV) comprise a group of chronic inflammatory diseases of the aorta and its major branches. The most common forms of LVV are giant cell arteritis (GCA) and Takayasu arteritis (TAK). Both GCA and TAK are characterized by granulomatous inflammation of the vessel wall accompanied by a maladaptive immune and vascular response that promotes vascular damage and remodeling. The inflammatory process in LVV starts in the adventitia where fibroblasts constitute the dominant cell population. Fibroblasts are traditionally recognized for synthesizing and renewing the extracellular matrix thereby being major players in maintenance of normal tissue architecture and in tissue repair. More recently, fibroblasts have emerged as a highly plastic cell population exerting various functions, including the regulation of local immune processes and organization of immune cells at the site of inflammation through production of cytokines, chemokines and growth factors as well as cell-cell interaction. In this review, we summarize and discuss the current knowledge on fibroblasts in LVV. Furthermore, we identify key questions that need to be addressed to fully understand the role of fibroblasts in the pathogenesis of LVV.
Collapse
Affiliation(s)
- Shuang Xu
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - William F Jiemy
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Elisabeth Brouwer
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Peter Heeringa
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Kornelis S M van der Geest
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Roser Alba-Rovira
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Corbera-Bellalta
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Annemieke H Boots
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Maria C Cid
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Sandovici
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands.
| |
Collapse
|
12
|
Bernard M, Menet R, Lecordier S, ElAli A. Endothelial PDGF-D contributes to neurovascular protection after ischemic stroke by rescuing pericyte functions. Cell Mol Life Sci 2024; 81:225. [PMID: 38769116 PMCID: PMC11106055 DOI: 10.1007/s00018-024-05244-w] [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: 11/28/2023] [Revised: 03/29/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024]
Abstract
Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)β controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRβ is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.
Collapse
Affiliation(s)
- Maxime Bernard
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Romain Menet
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Sarah Lecordier
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Ayman ElAli
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
13
|
Xiang H, Pan Y, Sze MA, Wlodarska M, Li L, van de Mark KA, Qamar H, Moure CJ, Linn DE, Hai J, Huo Y, Clarke J, Tan TG, Ho S, Teng KW, Ramli MN, Nebozhyn M, Zhang C, Barlow J, Gustafson CE, Gornisiewicz S, Albertson TP, Korle SL, Bueno R, Moy LY, Vollmann EH, Chiang DY, Brandish PE, Loboda A. Single-Cell Analysis Identifies NOTCH3-Mediated Interactions between Stromal Cells That Promote Microenvironment Remodeling and Invasion in Lung Adenocarcinoma. Cancer Res 2024; 84:1410-1425. [PMID: 38335304 PMCID: PMC11063690 DOI: 10.1158/0008-5472.can-23-1183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/15/2023] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
Cancer immunotherapy has revolutionized the treatment of lung adenocarcinoma (LUAD); however, a significant proportion of patients do not respond. Recent transcriptomic studies to understand determinants of immunotherapy response have pinpointed stromal-mediated resistance mechanisms. To gain a better understanding of stromal biology at the cellular and molecular level in LUAD, we performed single-cell RNA sequencing of 256,379 cells, including 13,857 mesenchymal cells, from 9 treatment-naïve patients. Among the mesenchymal cell subsets, FAP+PDPN+ cancer-associated fibroblasts (CAF) and ACTA2+MCAM+ pericytes were enriched in tumors and differentiated from lung-resident fibroblasts. Imaging mass cytometry revealed that both subsets were topographically adjacent to the perivascular niche and had close spatial interactions with endothelial cells (EC). Modeling of ligand and receptor interactomes between mesenchymal and ECs identified that NOTCH signaling drives these cell-to-cell interactions in tumors, with pericytes and CAFs as the signal receivers and arterial and PLVAPhigh immature neovascular ECs as the signal senders. Either pharmacologically blocking NOTCH signaling or genetically depleting NOTCH3 levels in mesenchymal cells significantly reduced collagen production and suppressed cell invasion. Bulk RNA sequencing data demonstrated that NOTCH3 expression correlated with poor survival in stroma-rich patients and that a T cell-inflamed gene signature only predicted survival in patients with low NOTCH3. Collectively, this study provides valuable insights into the role of NOTCH3 in regulating tumor stroma biology, warranting further studies to elucidate the clinical implications of targeting NOTCH3 signaling. SIGNIFICANCE NOTCH3 signaling activates tumor-associated mesenchymal cells, increases collagen production, and augments cell invasion in lung adenocarcinoma, suggesting its critical role in remodeling tumor stroma.
Collapse
Affiliation(s)
- Handan Xiang
- Discovery Immunology, Merck & Co., Inc., Cambridge, Massachusetts
| | - Yidan Pan
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | - Marc A. Sze
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | - Marta Wlodarska
- Discovery Oncology, Merck & Co., Inc., Boston, Massachusetts
| | - Ling Li
- Quantitative Bioscience, MSD, Singapore
| | | | - Haleema Qamar
- Discovery Immunology, Merck & Co., Inc., Cambridge, Massachusetts
| | - Casey J. Moure
- Discovery Oncology, Merck & Co., Inc., Boston, Massachusetts
| | - Douglas E. Linn
- Quantitative Bioscience, Merck & Co., Inc., Boston, Massachusetts
| | - Josephine Hai
- Quantitative Bioscience, Merck & Co., Inc., Boston, Massachusetts
| | - Ying Huo
- Quantitative Bioscience, Merck & Co., Inc., Boston, Massachusetts
| | - James Clarke
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | - Tze Guan Tan
- Discovery Cardiometabolic Diseases, MSD, Singapore
| | - Samantha Ho
- Discovery Cardiometabolic Diseases, MSD, Singapore
| | | | | | - Michael Nebozhyn
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | - Chunsheng Zhang
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | - Julianne Barlow
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Corinne E. Gustafson
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Savanna Gornisiewicz
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas P. Albertson
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stephanie L. Korle
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raphael Bueno
- The Division of Thoracic Surgery, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lily Y. Moy
- Quantitative Bioscience, Merck & Co., Inc., Boston, Massachusetts
| | | | - Derek Y. Chiang
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| | | | - Andrey Loboda
- Data and Genome Sciences, Merck & Co., Inc., Boston, Massachusetts
| |
Collapse
|
14
|
Zhao W, Li Y, Cheng H, Wang M, Zhang Z, Cai M, Zhao C, Xi X, Zhao X, Zhao W, Yang Y, Shao R. Myofibrillogenesis Regulator-1 Regulates the Ubiquitin Lysosomal Pathway of Notch3 Intracellular Domain Through E3 Ubiquitin-Protein Ligase Itchy Homolog in the Metastasis of Non-Small Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306472. [PMID: 38342606 PMCID: PMC11022719 DOI: 10.1002/advs.202306472] [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: 09/07/2023] [Revised: 12/28/2023] [Indexed: 02/13/2024]
Abstract
Myofibrillogenesis regulator-1 (MR-1) is a multifunctional protein involved in the development of various human tumors. The study is the first to report the promoting effect of MR-1 on the development and metastasis of non-small cell lung cancer (NSCLC). MR-1 is upregulated in NSCLC and positively associated with poor prognosis. The overexpression of MR-1 promotes the metastasis of NSCLC cells by stabilizing the expression of Notch3-ICD (NICD3) in the cytoplasm through enrichment analysis, in vitro and in vivo experimental researches. And Notch3 signaling can upregulate many genes related to metastasis. The stabilizing effect of MR-1 on NICD3 is achieved through the mono-ubiquitin lysosomal pathway and the specific E3 ubiquitin ligase is Itchy homolog (ITCH). There is a certain interaction between MR-1 and NICD3. Elevated MR-1 can affect the level of ITCH phosphorylation, reduce its E3 enzyme activity, and thus lead to reduce the ubiquitination and degradation of NICD3. Interference with the interaction between MR-1 and NICD3 can increase the degradation of NICD3 and impair the metastatic ability of NSCLC cells, which is a previously overlooked treatment option in NSCLC. In summary, interference with the interaction between MR-1 and NICD3 in the progression of lung cancer may be a promising therapeutic target.
Collapse
Affiliation(s)
- Wenxia Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Yang Li
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Hanzeng Cheng
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia MedicaPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100050P. R. China
| | - Mengyan Wang
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
- Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510280P. R. China
| | - Zhishuo Zhang
- Department of EmergencyXinhua HospitalShanghai Jiaotong University School of MedicineShanghai200092P. R. China
- Department of Organ Transplantation and Hepatobiliary SurgeryThe First Hospital of China Medical UniversityShenyangLiaoning110001P. R. China
| | - Meilian Cai
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Cong Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Xiaoming Xi
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Xiaojun Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Wuli Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| | - Yajun Yang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia MedicaPeking Union Medical College and Chinese Academy of Medical SciencesBeijing100050P. R. China
| | - Rongguang Shao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of OncologyInstitute of Medicinal Biotechnology Chinese Academy of Medical Sciences & Peking Union Medical College Beijing100050BeijingP. R. China
| |
Collapse
|
15
|
Zhou BW, Liu HM, Xu F, Jia XH. The role of macrophage polarization and cellular crosstalk in the pulmonary fibrotic microenvironment: a review. Cell Commun Signal 2024; 22:172. [PMID: 38461312 PMCID: PMC10924385 DOI: 10.1186/s12964-024-01557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/03/2024] [Indexed: 03/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive interstitial inflammatory disease with a high mortality rate. Patients with PF commonly experience a chronic dry cough and progressive dyspnoea for years without effective mitigation. The pathogenesis of PF is believed to be associated with dysfunctional macrophage polarization, fibroblast proliferation, and the loss of epithelial cells. Thus, it is of great importance and necessity to explore the interactions among macrophages, fibroblasts, and alveolar epithelial cells in lung fibrosis, as well as in the pro-fibrotic microenvironment. In this review, we discuss the latest studies that have investigated macrophage polarization and activation of non-immune cells in the context of PF pathogenesis and progression. Next, we discuss how profibrotic cellular crosstalk is promoted in the PF microenvironment by multiple cytokines, chemokines, and signalling pathways. And finally, we discuss the potential mechanisms of fibrogenesis development and efficient therapeutic strategies for the disease. Herein, we provide a comprehensive summary of the vital role of macrophage polarization in PF and its profibrotic crosstalk with fibroblasts and alveolar epithelial cells and suggest potential treatment strategies to target their cellular communication in the microenvironment.
Collapse
Affiliation(s)
- Bo-Wen Zhou
- The First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Hua-Man Liu
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Fei Xu
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Xin-Hua Jia
- Department of Pneumology and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
16
|
Lang NJ, Gote-Schniering J, Porras-Gonzalez D, Yang L, De Sadeleer LJ, Jentzsch RC, Shitov VA, Zhou S, Ansari M, Agami A, Mayr CH, Hooshiar Kashani B, Chen Y, Heumos L, Pestoni JC, Molnar ES, Geeraerts E, Anquetil V, Saniere L, Wögrath M, Gerckens M, Lehmann M, Yildirim AÖ, Hatz R, Kneidinger N, Behr J, Wuyts WA, Stoleriu MG, Luecken MD, Theis FJ, Burgstaller G, Schiller HB. Ex vivo tissue perturbations coupled to single-cell RNA-seq reveal multilineage cell circuit dynamics in human lung fibrogenesis. Sci Transl Med 2023; 15:eadh0908. [PMID: 38055803 DOI: 10.1126/scitranslmed.adh0908] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
Pulmonary fibrosis develops as a consequence of failed regeneration after injury. Analyzing mechanisms of regeneration and fibrogenesis directly in human tissue has been hampered by the lack of organotypic models and analytical techniques. In this work, we coupled ex vivo cytokine and drug perturbations of human precision-cut lung slices (hPCLS) with single-cell RNA sequencing and induced a multilineage circuit of fibrogenic cell states in hPCLS. We showed that these cell states were highly similar to the in vivo cell circuit in a multicohort lung cell atlas from patients with pulmonary fibrosis. Using micro-CT-staged patient tissues, we characterized the appearance and interaction of myofibroblasts, an ectopic endothelial cell state, and basaloid epithelial cells in the thickened alveolar septum of early-stage lung fibrosis. Induction of these states in the hPCLS model provided evidence that the basaloid cell state was derived from alveolar type 2 cells, whereas the ectopic endothelial cell state emerged from capillary cell plasticity. Cell-cell communication routes in patients were largely conserved in hPCLS, and antifibrotic drug treatments showed highly cell type-specific effects. Our work provides an experimental framework for perturbational single-cell genomics directly in human lung tissue that enables analysis of tissue homeostasis, regeneration, and pathology. We further demonstrate that hPCLS offer an avenue for scalable, high-resolution drug testing to accelerate antifibrotic drug development and translation.
Collapse
Affiliation(s)
- Niklas J Lang
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Janine Gote-Schniering
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Department of Rheumatology and Immunology, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Lung Precision Medicine Program, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Diana Porras-Gonzalez
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Lin Yang
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Laurens J De Sadeleer
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium
| | - R Christoph Jentzsch
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Vladimir A Shitov
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany
| | - Shuhong Zhou
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Meshal Ansari
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany
| | - Ahmed Agami
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Christoph H Mayr
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Baharak Hooshiar Kashani
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Yuexin Chen
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Lukas Heumos
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany
| | - Jeanine C Pestoni
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Eszter Sarolta Molnar
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | | | | | | | - Melanie Wögrath
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Michael Gerckens
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Mareike Lehmann
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute for Lung Research, Philipps-University Marburg, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), 35043 Marburg, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Rudolf Hatz
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Medical Center, Munich-Gauting, 82131 Gauting, Germany
| | - Nikolaus Kneidinger
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Wim A Wuyts
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, 3000 Leuven, Belgium
| | - Mircea-Gabriel Stoleriu
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Medical Center, Munich-Gauting, 82131 Gauting, Germany
| | - Malte D Luecken
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764 Munich, Germany
- Department of Mathematics, Technische Universität München, 85748 Garching bei München, Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive/Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 81377 Munich, Germany
| |
Collapse
|
17
|
Yuan H, You Y, He Y, Wei Y, Zhang Y, Min H, Li C, Chen J. Crystalline Silica-Induced Proinflammatory Interstitial Macrophage Recruitment through Notch3 Signaling Promotes the Pathogenesis of Silicosis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:14502-14514. [PMID: 37721423 DOI: 10.1021/acs.est.3c03980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
Crystalline silica (CS) particles are ubiquitous in the environment, especially in occupational conditions, and exposure to respirable CS causes silicosis. The initial response to CS is mediated by innate immunity, where pulmonary macrophages act as central orchestrators. However, the repercussions of CS on functionally distinct macrophage subsets remain to be inconclusive. Herein, to study the effects of inhaled CS, we divided macrophages into three subsets: circulating monocytes, interstitial macrophages (IMs), and alveolar macrophages (AMs). CS-induced massive IMs increase in the lung, the phenotype and function of which differed from those of tissue-resident AMs and circulating monocytes. The augmented IMs were driven by recruitment of circulating macrophages rather than cell proliferation in situ. Moreover, the IMs predominantly exerted a classic activated (M1) phenotype and expressed proinflammatory cytokines, contributing to CS-induced lung injury. Notably, we demonstrated that IMs augmented Notch3 expression. Mechanistically, using myeloid-specific Notch3-knockout mice, we demonstrated that Notch3 signaling not only promoted IMs recruitment by regulating CCR2 expression but also manipulated the proinflammatory phenotype. Mice with conditional Notch3-knockout exhibited alleviation of CS-induced inflammation and fibrosis in lung. Overall, our study identifies IMs as critical mediators in response to CS and highlights the role of Notch3 in IMs recruitment and activation, providing new insights into CS toxicological effects in the lung.
Collapse
Affiliation(s)
- Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yungeng Wei
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yuting Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, (China Medical University), Ministry of Education, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| |
Collapse
|
18
|
Liu D, Zhang C, Zhang J, Xu GT, Zhang J. Molecular pathogenesis of subretinal fibrosis in neovascular AMD focusing on epithelial-mesenchymal transformation of retinal pigment epithelium. Neurobiol Dis 2023; 185:106250. [PMID: 37536385 DOI: 10.1016/j.nbd.2023.106250] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss among elderly people in developed countries. Neovascular AMD (nAMD) accounts for more than 90% of AMD-related vision loss. At present, intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) is widely used as the first-line therapy to decrease the choroidal and retinal neovascularizations, and thus to improve or maintain the visual acuity of the patients with nAMD. However, about 1/3 patients still progress to irreversible visual impairment due to subretinal fibrosis even with adequate anti-VEGF treatment. Extensive literatures support the critical role of epithelial-mesenchymal transformation (EMT) of retinal pigment epithelium (RPE) in the pathogenesis of subretinal fibrosis in nAMD, but the underlying mechanisms still remain largely unknown. This review summarized the molecular pathogenesis of subretinal fibrosis in nAMD, especially focusing on the transforming growth factor-β (TGF-β)-induced EMT pathways. It was also discussed how these pathways crosstalk and respond to signals from the microenvironment to mediate EMT and contribute to the progression of nAMD-related subretinal fibrosis. Targeting EMT signaling pathways might provide a promising and effective therapeutic strategy to treat subretinal fibrosis secondary to nAMD.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| |
Collapse
|
19
|
Pi P, Zeng Z, Zeng L, Han B, Bai X, Xu S. Molecular mechanisms of COVID-19-induced pulmonary fibrosis and epithelial-mesenchymal transition. Front Pharmacol 2023; 14:1218059. [PMID: 37601070 PMCID: PMC10436482 DOI: 10.3389/fphar.2023.1218059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
As the outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first broke out in Hubei Province, China, at the end of 2019. It has brought great challenges and harms to global public health. SARS-CoV-2 mainly affects the lungs and is mainly manifested as pulmonary disease. However, one of the biggest crises arises from the emergence of COVID-19-induced fibrosis. At present, there are still many questions about how COVID-19 induced pulmonary fibrosis (PF) occurs and how to treat and regulate its long-term effects. In addition, as an important process of fibrosis, the effect of COVID-19 on epithelial-mesenchymal transition (EMT) may be an important factor driving PF. This review summarizes the main pathogenesis and treatment mechanisms of COVID-19 related to PF. Starting with the basic mechanisms of PF, such as EMT, transforming growth factor-β (TGF-β), fibroblasts and myofibroblasts, inflammation, macrophages, innate lymphoid cells, matrix metalloproteinases and tissue inhibitors of metalloproteinases, hedgehog pathway as well as Notch signaling. Further, we highlight the importance of COVID-19-induced EMT in the process of PF and provide an overview of the related molecular mechanisms, which will facilitate future research to propose new clinical therapeutic solutions for the treatment of COVID-19-induced PF.
Collapse
Affiliation(s)
- Peng Pi
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Zhipeng Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Liqing Zeng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Bing Han
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Xizhe Bai
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shousheng Xu
- School of Sports Engineering, Beijing Sport University, Beijing, China
| |
Collapse
|
20
|
Ng B, Xie C, Su L, Kuthubudeen FF, Kwek XY, Yeong D, Pua CJ, Cook SA, Lim WW. IL11 (Interleukin-11) Causes Emphysematous Lung Disease in a Mouse Model of Marfan Syndrome. Arterioscler Thromb Vasc Biol 2023; 43:739-754. [PMID: 36924234 PMCID: PMC10125130 DOI: 10.1161/atvbaha.122.318802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Marfan Syndrome (MFS) is an inherited connective tissue disorder caused by mutations in the FBN1 (fibrillin-1) gene. Lung abnormalities are common in MFS, but their pathogenesis is poorly understood. IL11 (interleukin-11) causes aortic disease in a mouse model of MFS and was studied here in the lung. METHODS We examined histological and molecular phenotypes in the lungs of Fbn1C1041G/+ mice (mouse model of Marfan Syndrome [mMFS]), an established mouse model of MFS. To identify IL11-expressing cells, we used immunohistochemistry on lungs of 4- and 16-week-old Fbn1C1041G/+:Il11EGFP/+ reporter mice. We studied the effects of IL11 inhibition by RT-qPCR, immunoblots and histopathology in lungs from genetic or pharmacologic models: (1) 16-week-old IL11 receptor (IL11RA) knockout mMFS mice (Fbn1C1041G/+:Il11ra1-/- mice) and (2) in mMFS mice administered IgG control or interleukin-11 receptor antibodies twice weekly from 4 to 24 weeks of age. RESULTS mMFS lungs showed progressive loss and enlargement of distal airspaces associated with increased proinflammatory and profibrotic gene expression as well as matrix metalloproteinases 2, 9, and 12. IL11 was increased in mMFS lungs and localized to smooth muscle and endothelial cells in young mMFS mice in the Fbn1C1041G/+:Il11EGFP/+ reporter strain and in fibroblasts, in older mice. In mMFS mice, genetic (Fbn1C1041G/+:Il11ra1-/-) or pharmacologic (anti-interleukin-11 receptor) inhibition of IL11 signaling reduced lung emphysema, fibrosis, and inflammation. This protective effect was associated with reduced pathogenic ERK1/2 signaling and lower metalloproteinase 2, 9, and 12 expression. CONCLUSIONS IL11 causes lung disease in mMFS. This reveals a shared IL11-driven disease mechanism in lung and aorta in MFS and suggests inhibition of IL11 signaling as a holistic approach for treating multiorgan morbidity in MFS.
Collapse
Affiliation(s)
- Benjamin Ng
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| | - Chen Xie
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Liping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Fathima F. Kuthubudeen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| | - Xiu-Yi Kwek
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Daryl Yeong
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Chee Jian Pua
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
| | - Stuart A. Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
- MRC-London Institute of Medical Sciences, United Kingdom (S.A.C.)
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore (B.N., C.X., L.S., X.-Y.K., D.Y., C.J.P., S.A.C., W.-W.L.)
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School (B.N., F.F.K., S.A.C., W.-W.L.)
| |
Collapse
|
21
|
Abstract
The mammalian respiratory system or lung is a tree-like branching structure, and the main site of gas exchange with the external environment. Structurally, the lung is broadly classified into the proximal (or conducting) airways and the distal alveolar region, where the gas exchange occurs. In parallel with the respiratory tree, the pulmonary vasculature starts with large pulmonary arteries that subdivide rapidly ending in capillaries adjacent to alveolar structures to enable gas exchange. The NOTCH signalling pathway plays an important role in lung development, differentiation and regeneration post-injury. Signalling via the NOTCH pathway is mediated through activation of four NOTCH receptors (NOTCH1-4), with each receptor capable of regulating unique biological processes. Dysregulation of the NOTCH pathway has been associated with development and pathophysiology of multiple adult acute and chronic lung diseases. This includes accumulating evidence that alteration of NOTCH3 signalling plays an important role in the development and pathogenesis of chronic obstructive pulmonary disease, lung cancer, asthma, idiopathic pulmonary fibrosis and pulmonary arterial hypertension. Herein, we provide a comprehensive summary of the role of NOTCH3 signalling in regulating repair/regeneration of the adult lung, its association with development of lung disease and potential therapeutic strategies to target its signalling activity.
Collapse
|
22
|
Li R, Jia Y, Kong X, Nie Y, Deng Y, Liu Y. Novel drug delivery systems and disease models for pulmonary fibrosis. J Control Release 2022; 348:95-114. [PMID: 35636615 DOI: 10.1016/j.jconrel.2022.05.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/17/2022] [Accepted: 05/21/2022] [Indexed: 12/19/2022]
Abstract
Pulmonary fibrosis (PF) is a serious and progressive lung disease which is possibly life-threatening. It causes lung scarring and affects lung functions including epithelial cell injury, massive recruitment of immune cells and abnormal accumulation of extracellular matrix (ECM). There is currently no cure for PF. Treatment for PF is aimed at slowing the course of the disease and relieving symptoms. Pirfenidone (PFD) and nintedanib (NDNB) are currently the only two FDA-approved oral medicines to slow down the progress of idiopathic pulmonary fibrosis, a specific type of PF. Novel drug delivery systems and therapies have been developed to improve the prognosis of the disease, as well as reduce or minimize the toxicities during drug treatment. The drug delivery routes for these therapies are various including oral, intravenous, nasal, inhalant, intratracheal and transdermal; although this is dependent on specific treatment mechanisms. In addition, researchers have also expanded current animal models that could not fully restore the clinicopathology, and developed a series of in vitro models such as organoids to study the pathogenesis and treatment of PF. This review describes recent advances on pathogenesis exploration, classifies and specifies the progress of drug delivery systems by their delivery routes, as well as an overview on the in vitro and in vivo models for PF research.
Collapse
Affiliation(s)
- Rui Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yizhen Jia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaohan Kong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yichu Nie
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan 528000, China
| | - Yang Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Yang Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
23
|
McCarthy C, Keane MP. Contemporary Concise Review 2021: Interstitial lung disease. Respirology 2022; 27:539-548. [PMID: 35513341 PMCID: PMC9320947 DOI: 10.1111/resp.14278] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/28/2022] [Indexed: 11/30/2022]
Abstract
The last 2 years have presented previously unforeseen challenges in pulmonary medicine. Despite the significant impact of the SARS‐CoV‐2 pandemic on patients, clinicians and communities, advances in the care and understanding of interstitial lung disease (ILD) continued unabated. Recent studies have led to improved guidelines, better understanding of the role for antifibrotics in fibrosing ILDs, prognostic indicators and novel biomarkers. In this concise contemporary review, we summarize many of the important studies published in 2021, highlighting their relevance and impact to the management and knowledge of ILD.
Collapse
Affiliation(s)
- Cormac McCarthy
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Michael P Keane
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
24
|
Dhamodaran K, Baidouri H, Nartey A, Staverosky J, Keller K, Acott T, Vranka J, Raghunathan V. Endogenous expression of Notch pathway molecules in human trabecular meshwork cells. Exp Eye Res 2022; 216:108935. [PMID: 35033558 PMCID: PMC8885976 DOI: 10.1016/j.exer.2022.108935] [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: 11/11/2021] [Revised: 12/23/2021] [Accepted: 01/05/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE Cells in the trabecular meshwork sense and respond to a myriad of physical forces through a process known as mechanotransduction. Whilst the effect of substratum stiffness or stretch on TM cells have been investigated in the context of transforming growth factor (TGF-β), Wnt and YAP/TAZ pathways, the role of Notch signaling, an evolutionarily conserved pathway, recently implicated in mechanotransduction, has not been investigated in trabecular meshwork (TM) cells. Here, we compare the endogenous expression of Notch pathway molecules in TM cells from glaucomatous and non-glaucomatous donors, segmental flow regions, and when subjected to cyclical strain, or grown on hydrogels of varying rigidity. METHODS Primary TM from glaucomatous (GTM), non-glaucomatous (NTM) donors, and from segmental flow regions [high flow (HF), low flow (LF)], were utilized between passages 2-6. Cells were (i) plated on tissue culture plastic, (ii) subjected to cyclical strain (6 h and 24 h), or (iii) cultured on 3 kPa and 80 kPa hydrogels. mRNA levels of Notch receptors/ligands/effectors in the TM cells was determined by qRT-PCR. Phagocytosis was determined as a function of substratum stiffness in NTM-HF/LF cells in the presence or absence of 100 nM Dexamethasone treatment. RESULTS Innate expression of Notch pathway genes were significantly overexpressed in GTM cells with no discernible differences observed between HF/LF cells in either NTM or GTM cells cultured on plastic substrates. With 6 h of cyclical strain, a subset of Notch pathway genes presented with altered expression. Expression of Notch receptors/ligands/receptors/inhibitors progressively declined with increasing stiffness and this correlated with phagocytic ability of NTM cells. Dexamethasone treatment decreased phagocytosis regardless of stiffness or cells isolated from segmental outflow regions. CONCLUSIONS We demonstrate here that the Notch expression in cultured TM cells differ intrinsically between GTM vs NTM, and by substratum cues (cyclical strain and stiffness). Of import, the most apparent differences in gene expression were observed as a function of substratum stiffness which closely followed phagocytic ability of cells. Interestingly, on soft substrates (mimicking normal TM stiffness) Notch expression and phagocytosis was highest, while both expression and phagocytosis was significantly lower on stiffer substrates (mimicking glaucomatous stiffness) regardless of DEX treatment. Such context dependent changes suggest Notch pathway may play differing roles in disease vs homeostasis. Studies focused on understanding the mechanistic role of Notch (if any) in outflow homeostasis are thus warranted.
Collapse
Affiliation(s)
- Kamesh Dhamodaran
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, USA
| | - Hasna Baidouri
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, USA
| | - Andrews Nartey
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, USA
| | - Julia Staverosky
- Casey Eye Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Kate Keller
- Casey Eye Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Ted Acott
- Casey Eye Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Janice Vranka
- Casey Eye Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - VijayKrishna Raghunathan
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, TX, USA,Department of Biomedical Engineering, University of Houston, Houston, TX, USA,Correspondence should be sent to: VijayKrishna Raghunathan, Ph.D., University of Houston, College of Optometry, 4901 Calhoun Rd, Houston, TX, 77204, Phone: (713)-743-8331,
| |
Collapse
|
25
|
The NOTCH3 Downstream Target HEYL Is Required for Efficient Human Airway Basal Cell Differentiation. Cells 2021; 10:cells10113215. [PMID: 34831437 PMCID: PMC8620267 DOI: 10.3390/cells10113215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Basal cells (BCs) are stem/progenitor cells of the mucociliary airway epithelium, and their differentiation is orchestrated by the NOTCH signaling pathway. NOTCH3 receptor signaling regulates BC to club cell differentiation; however, the downstream responses that regulate this process are unknown. Overexpression of the active NOTCH3 intracellular domain (NICD3) in primary human bronchial epithelial cells (HBECs) on in vitro air–liquid interface culture promoted club cell differentiation. Bulk RNA-seq analysis identified 692 NICD3-responsive genes, including the classical NOTCH target HEYL, which increased in response to NICD3 and positively correlated with SCGB1A1 (club cell marker) expression. siRNA knockdown of HEYL decreased tight junction formation and cell proliferation. Further, HEYL knockdown reduced club, goblet and ciliated cell differentiation. In addition, we observed decreased expression of HEYL in HBECs from donors with chronic obstructive pulmonary disease (COPD) vs. normal donors which correlates with the impaired differentiation capacity of COPD cells. Finally, overexpression of HEYL in COPD HBECs promoted differentiation into club, goblet and ciliated cells, suggesting the impaired capacity of COPD cells to generate a normal airway epithelium is a reversible phenotype that can be regulated by HEYL. Overall, our data identify the NOTCH3 downstream target HEYL as a key regulator of airway epithelial differentiation.
Collapse
|
26
|
Abstract
Fibroblasts are important cells for the support of homeostatic tissue function. In inflammatory diseases such as rheumatoid arthritis and inflammatory bowel disease, fibroblasts take on different roles (a) as inflammatory cells themselves and (b) in recruiting leukocytes, driving angiogenesis, and enabling chronic inflammation in tissues. Recent advances in single-cell profiling techniques have transformed the ability to examine fibroblast states and populations in inflamed tissues, providing evidence of previously underappreciated heterogeneity and disease-associated fibroblast populations. These studies challenge the preconceived notion that fibroblasts are homogeneous and provide new insights into the role of fibroblasts in inflammatory pathology. In addition, new molecular insights into the mechanisms of fibroblast activation reveal powerful cell-intrinsic amplification loops that synergize with primary fibroblast stimuli to result in striking responses. In this Review, we focus on recent developments in our understanding of fibroblast heterogeneity and fibroblast pathology across tissues and diseases in rheumatoid arthritis and inflammatory bowel diseases. We highlight new approaches to, and applications of, single-cell profiling techniques and what they teach us about fibroblast biology. Finally, we address how these insights could lead to the development of novel therapeutic approaches to targeting fibroblasts in disease.
Collapse
|
27
|
Gajjala PR, Madala SK. Notch3: A New Culprit in Fibrotic Lung Disease. Am J Respir Cell Mol Biol 2021; 64:403-404. [PMID: 33591242 PMCID: PMC8008798 DOI: 10.1165/rcmb.2021-0024ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Prathibha R Gajjala
- Department of Pediatrics University of Cincinnati Cincinnati, Ohio and.,Division of Pulmonary Medicine Cincinnati Children's Hospital Medical Center Cincinnati, Ohio
| | - Satish K Madala
- Department of Pediatrics University of Cincinnati Cincinnati, Ohio and.,Division of Pulmonary Medicine Cincinnati Children's Hospital Medical Center Cincinnati, Ohio
| |
Collapse
|