1
|
Polkoff KM, Lampe R, Gupta NK, Murphy Y, Chung J, Carter A, Simon JM, Gleason K, Moatti A, Murthy PK, Edwards L, Greenbaum A, Tata A, Tata PR, Piedrahita JA. Novel Porcine Model Reveals Two Distinct LGR5 Cell Types during Lung Development and Homeostasis. Am J Respir Cell Mol Biol 2025; 72:496-509. [PMID: 39499850 PMCID: PMC12051919 DOI: 10.1165/rcmb.2024-0040oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 11/05/2024] [Indexed: 11/07/2024] Open
Abstract
Cells expressing leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) play a pivotal role in homeostasis, repair, and regeneration in multiple organs, including skin and gastrointestinal tract, but little is known about their role in the lung. Findings from mice, a widely used animal model, suggest that lung LGR5 expression differs from that of humans. In this work, using a new transgenic pig model, we identify two main populations of LGR5+ cells in the lung that are conserved in human but not mouse lungs. Using RNA sequencing, three-dimensional imaging, and organoid models, we determine that in the fetal lung, epithelial LGR5 expression is transient in a subpopulation of SOX9+/ETV5+/SFTPC+ progenitor lung tip cells. In contrast, epithelial LGR5 expression is absent from postnatal lung but is reactivated in bronchioalveolar organoids derived from basal airway cells. We also describe a separate population of mesenchymal LGR5+ cells that surrounds developing and mature airways, lies adjacent to airway basal cells, and is closely associated with nerve fibers. Transcriptionally, mesenchymal LGR5+ cells include a subset of peribronchial fibroblasts that express unique patterns of SHH, FGF, WNT, and TGF-β signaling pathway genes. These results support distinct roles for LGR5+ cells in the lung and describe a physiologically relevant animal model for further studies on the function of these cells in repair and regeneration.
Collapse
Affiliation(s)
- Kathryn M. Polkoff
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Ross Lampe
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Nithin K. Gupta
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- School of Osteopathic Medicine, Campbell University, Lillington, North Carolina
| | - Yanet Murphy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Jaewook Chung
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Amber Carter
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Jeremy M. Simon
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Katherine Gleason
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh and Chapel Hill, North Carolina; and
| | - Preetish K. Murthy
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Laura Edwards
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh and Chapel Hill, North Carolina; and
| | - Aleksandra Tata
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Purushothama Rao Tata
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Jorge A. Piedrahita
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
2
|
Wen H, Chandrasekaran P, Jin A, Pankin J, Lu M, Liberti DC, Zepp JA, Jain R, Morrisey EE, Michki SN, Frank DB. A spatiotemporal cell atlas of cardiopulmonary progenitor cell allocation during development. Cell Rep 2025; 44:115513. [PMID: 40178979 DOI: 10.1016/j.celrep.2025.115513] [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: 02/06/2024] [Revised: 01/10/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
The heart and lung co-orchestrate their development during organogenesis. The mesoderm surrounding both the developing heart and anterior foregut endoderm provides instructive cues guiding cardiopulmonary development. Additionally, it serves as a source of cardiopulmonary progenitor cells (CPPs) expressing Wnt2 that give rise to both cardiac and lung mesodermal cell lineages. Despite the mesoderm's critical importance to both heart and lung development, mechanisms guiding CPP specification are unclear. To address this, we lineage traced Wnt2+ CPPs at E8.5 and performed single-cell RNA sequencing on collected progeny across the developmental lifespan. Using computational analyses, we created a CPP-derived cell atlas that revealed a previously underappreciated spectrum of CPP-derived cell lineages, including all lung mesodermal lineages, ventricular cardiomyocytes, and epicardial and pericardial cells. By integrating spatial mapping with computational cell trajectory analysis and transcriptional profiling, we have provided a potential molecular and cellular roadmap for cardiopulmonary development.
Collapse
Affiliation(s)
- Hongbo Wen
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Prashant Chandrasekaran
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Annabelle Jin
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Josh Pankin
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - MinQi Lu
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Derek C Liberti
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Jarod A Zepp
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, CHOP, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Department of Cell and Developmental Biology, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sylvia N Michki
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, CHOP, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA.
| | - David B Frank
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia (CHOP), Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, CHOP Cardiovascular Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Chen YT, Lohia GK, Chen S, Liu Z, Wong Fok Lung T, Wang C, Riquelme SA. A host-pathogen metabolic synchrony that facilitates disease tolerance. Nat Commun 2025; 16:3729. [PMID: 40253414 PMCID: PMC12009439 DOI: 10.1038/s41467-025-59134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Disease tolerance mitigates organ damage from non-resolving inflammation during persistent infections, yet its underlying mechanisms remain unclear. Here we show, in a Pseudomonas aeruginosa pneumonia mouse model, that disease tolerance depends on the mitochondrial metabolite itaconate, which mediates cooperative host-pathogen interactions. In P. aeruginosa, itaconate modifies key cysteine residues in TCA cycle enzymes critical for succinate metabolism, inducing bioenergetic stress and promoting the formation biofilms that are less immunostimulatory and allow the bacteria to integrate into the local microbiome. Itaconate incorporates into the central metabolism of the biofilm, driving exopolysaccharide production-particularly alginate-which amplifies airway itaconate signaling. This itaconate-alginate interplay limits host immunopathology by enabling pulmonary glutamine assimilation, activating glutaminolysis, and thereby restrain detrimental inflammation caused by the inflammasome. Clinical sample analysis reveals that P. aeruginosa adapts to this metabolic environment through compensatory mutations in the anti-sigma-factor mucA, which restore the succinate-driven bioenergetics and disrupt the metabolic synchrony essential for sustaining disease tolerance.
Collapse
Affiliation(s)
- Ying-Tsun Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Samantha Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Zihua Liu
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | | - Chu Wang
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | |
Collapse
|
4
|
Ke X, van Soldt B, Vlahos L, Zhou Y, Qian J, George J, Capdevila C, Glass I, Yan K, Califano A, Cardoso WV. Morphogenesis and regeneration share a conserved core transition cell state program that controls lung epithelial cell fate. Dev Cell 2025; 60:819-836.e7. [PMID: 39667932 PMCID: PMC11945641 DOI: 10.1016/j.devcel.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/07/2024] [Accepted: 11/17/2024] [Indexed: 12/14/2024]
Abstract
Transitional cell states are at the crossroads of crucial developmental and regenerative events, yet little is known about how these states emerge and influence outcomes. The alveolar and airway epithelia arise from distal lung multipotent progenitors, which undergo cell fate transitions to form these distinct compartments. The identification and impact of cell states in the developing lung are poorly understood. Here, we identified a population of Icam1/Nkx2-1 epithelial progenitors harboring a transitional state program remarkably conserved in humans and mice during lung morphogenesis and regeneration. Lineage-tracing and functional analyses reveal their role as progenitors to both airways and alveolar cells and the requirement of this transitional program to make distal lung progenitors competent to undergo airway cell fate specification. The identification of a common progenitor cell state in vastly distinct processes suggests a unified program reiteratively regulating outcomes in development and regeneration.
Collapse
Affiliation(s)
- Xiangyi Ke
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pharmacology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benjamin van Soldt
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lukas Vlahos
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yizhuo Zhou
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Pulmonary & Allergy Critical Care, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jun Qian
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joel George
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Claudia Capdevila
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ian Glass
- Birth Defects Research Laboratory (BDRL), University of Washington, Seattle, WA 98105, USA
| | - Kelley Yan
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Disease, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrea Califano
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Pulmonary & Allergy Critical Care, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
5
|
McSwiggin H, Wang R, Magalhães RDM, Zhu F, Doherty TA, Yan W, Jendzjowsky N. Comprehensive sequencing of the lung neuroimmune landscape in response to asthmatic induction. Front Immunol 2025; 16:1518771. [PMID: 40181989 PMCID: PMC11965707 DOI: 10.3389/fimmu.2025.1518771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/13/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Evidence demonstrates that sensory neurons respond to pathogenic/allergic infiltration and mediate immune responses, forming an integral part of host defense that becomes hypersensitized during allergy. Our objective was to investigate how asthmatic induction alters the pulmonary neuroimmune transcriptome. We hypothesized that asthmatic induction would upregulate genes in the vagal ganglia (nodose/jugular ganglia), which would be associated with asthmatic immunity, and that these would be clustered, primarily in nodose neurons. Furthermore, lungs would increase transcripts associated with nerve activation, and these would be centered in neural and neuroendocrine-like cells. Methods Standard RNA sequencing, single nucleus-RNA sequencing, and spatial RNA sequencing of vagal ganglia. Standard RNA-sequencing and spatial RNA-sequencing of lungs in naïve and mice that have undergone asthmatic induction with Alternaria alternata. Results Bulk RNA-seq revealed that genes related to allergen sensing were increased in asthmatic ganglia nodose/jugular ganglia compared to control ganglia. These genes were associated with nodose clusters as shown by single-nucleus RNA sequencing, and a distinct caudal-to-rostral spatial arrangement was presented as delineated by spatial transcriptomics. The distinct clusters closely match previous identification of nodose neuron clusters. Correspondingly, the lung transcriptome was altered with asthmatic induction such that transcripts associated with neural excitation were upregulated. The spatial distribution of these transcripts was revealed by spatial transcriptomics to illustrate that these were expressed in neuroendocrine-like cells/club cells, and neurons. Conclusions These results show that the neuroimmune transcriptome is altered in response to asthmatic induction in a cell cluster and spatially distinct manner.
Collapse
Affiliation(s)
- Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Rui Wang
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Rubens Daniel Miserani Magalhães
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Fengli Zhu
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Taylor A. Doherty
- Division of Allergy and Immunology, Department of Medicine, University of California, San Diego, Veterans Affairs San Diego Healthcare System, La Jolla, CA, United States
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
- Division of Endocrinology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Nicholas Jendzjowsky
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
- Division of Respiratory and Critical Care Medicine and Physiology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
6
|
Meng X, Li W, Xu J, Yao Y, Gong A, Yang Y, Qu F, Guo C, Zheng H, Cui G, Suo S, Peng G. Spatiotemporal transcriptome atlas of developing mouse lung. Sci Bull (Beijing) 2025:S2095-9273(25)00240-3. [PMID: 40118721 DOI: 10.1016/j.scib.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025]
Abstract
The functional development of the mammalian lung is a complex process that relies on the spatial and temporal organization of multiple cell types and their states. However, a comprehensive spatiotemporal transcriptome atlas of the developing lung has not yet been reported. Here we apply high-throughput spatial transcriptomics to allow for a comprehensive assessment of mouse lung development comprised of two critical developmental events: branching morphogenesis and alveologenesis. We firstly generate a spatial molecular atlas of mouse lung development spanning from E12.5 to P0 based on the integration of published single cell RNA-sequencing data and identify 10 spatial domains critical for functional lung organization. Furthermore, we create a lineage trajectory connecting spatial clusters from adjacent time points in E12.5-P0 lungs and explore TF (transcription factor) regulatory networks for each lineage specification. We observe the establishment of pulmonary airways within the developing lung, accompanied by the proximal-distal patterning with distinct characteristics of gene expression, signaling landscape and transcription factors enrichment. We characterize the alveolar niche heterogeneity with maturation state differences during the later developmental stage around birth and demonstrate differentially expressed genes, such as Angpt2 and Epha3, which may perform a critical role during alveologenesis. In addition, multiple signaling pathways, including ANGPT, VEGF and EPHA, exhibit increased levels in more maturing alveolar niche. Collectively, by integrating the spatial transcriptome with corresponding single-cell transcriptome data, we provide a comprehensive molecular atlas of mouse lung development with detailed molecular domain annotation and communication, which would pave the way for understanding human lung development and respiratory regeneration medicine.
Collapse
Affiliation(s)
- Xiaogao Meng
- Life Science and Medicine, University of Science and Technology of China, Hefei 230026, China; Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenjia Li
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; Guangzhou National Laboratory, Guangzhou 510005, China
| | - Jian Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yao Yao
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - An Gong
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yumeng Yang
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Fangfang Qu
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Chenkai Guo
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Hui Zheng
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China
| | - Guizhong Cui
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Guangzhou National Laboratory, Guangzhou 510005, China.
| | - Shengbao Suo
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; Guangzhou National Laboratory, Guangzhou 510005, China.
| | - Guangdun Peng
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
7
|
Khadim A, Kiliaris G, Vazquez-Armendariz AI, Procida-Kowalski T, Glaser D, Bartkuhn M, Malik T, Chu X, Moiseenko A, Kuznetsova I, Ahmadvand N, Lingampally A, Hadzic S, Alexopoulos I, Chen Y, Günther A, Behr J, Neumann J, Schiller HB, Li X, Weissmann N, Braun T, Seeger W, Wygrecka M, Morty RE, Herold S, El Agha E. Myofibroblasts emerge during alveolar regeneration following influenza-virus-induced lung injury. Cell Rep 2025; 44:115248. [PMID: 39903667 DOI: 10.1016/j.celrep.2025.115248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/24/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025] Open
Abstract
Alveolar regeneration requires the coordinated engagement of epithelial stem cells and mesenchymal niche cells to restore the intricate alveolar architecture of the lung. The current paradigm is that certain aspects of lung organogenesis are mimicked during injury repair in the adult stage. Here, we employ a longitudinal single-cell transcriptomic survey to fate map lung mesenchymal cells throughout development and adulthood. We show that myofibroblasts that are reminiscent of developmental alveolar myofibroblasts (AMFs), termed AMF-like cells, are activated during alveolar regeneration following influenza-virus-induced lung injury. Although AMF-like cells share a similar transcriptomic signature with myofibroblasts that are associated with aberrant repair and fibrosis, these cells do not derive from fibroblast growth factor 10-positive alveolar fibroblasts, and their dysregulation is associated with failed alveolar regeneration in humans. Our data emphasize the role played by developmental mechanisms in alveolar regeneration and highlight the context-dependent nature of myofibroblast biology and function during injury repair.
Collapse
Affiliation(s)
- Ali Khadim
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Georgios Kiliaris
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ana Ivonne Vazquez-Armendariz
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; University of Bonn, Transdisciplinary Research Area Life and Health, Organoid Biology, Life & Medical Sciences Institute, 53115 Bonn, Germany
| | - Tara Procida-Kowalski
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - David Glaser
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Tanya Malik
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Xuran Chu
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Alena Moiseenko
- Immunology and Respiratory Department, Boehringer Ingelheim Pharma GmbH, Biberach 88400, Germany
| | - Irina Kuznetsova
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Negah Ahmadvand
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Arun Lingampally
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Stefan Hadzic
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Yuexin Chen
- Research Unit Precision Regenerative Medicine (PRM), Comprehensive Pneumology Center (CPC), German Center for Lung Research (DZL), 81377 Munich, Germany; Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Andreas Günther
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Jürgen Behr
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Jens Neumann
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center, German Center for Lung Research (DZL), 80336 Munich, Germany
| | - Herbert B Schiller
- Research Unit Precision Regenerative Medicine (PRM), Comprehensive Pneumology Center (CPC), German Center for Lung Research (DZL), 81377 Munich, Germany; Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, 61231 Bad Nauheim, Germany
| | - Werner Seeger
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, 61231 Bad Nauheim, Germany
| | - Malgorzata Wygrecka
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, Heidelberg University Hospital, 69117 Heidelberg, Germany
| | - Susanne Herold
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Elie El Agha
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
8
|
He H, Ma C, Wei W, Wang H, Lai Y, Liu M, Sun S, Ma Q, Lai J, Liu H, Liu H, Sun F, Lin X. Heparan sulfate regulates myofibroblast heterogeneity and function to mediate niche homeostasis during alveolar morphogenesis. Nat Commun 2025; 16:1834. [PMID: 39979343 PMCID: PMC11842828 DOI: 10.1038/s41467-025-57163-4] [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: 05/06/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
Postnatal respiration requires bulk formation of alveoli that produces extensive surface area for gas diffusion from epithelium to the circulatory system. Alveolar morphogenesis initiates at late gestation or postnatal stage during mammalian development and is mediated by coordination among multiple cell types. Here we show that fibroblast-derived Heparan Sulfate Glycosaminoglycan (HS-GAG) is essential for maintaining a niche that supports alveolar formation by modulating both biophysical and biochemical cues. Gli1-CreER mediated deletion of HS synthase gene Ext1 in lung fibroblasts results in enlarged and simplified alveolar structures. Ablation of HS results in loss of a subset of PDGFRαhi αSMA+ alveolar myofibroblasts residing in the distal alveolar region, which exhibit contractile properties and maintain WNT signaling activity to support normal proliferation and differentiation of alveolar epithelial cells. HS is essential for proliferation while preventing precocious apoptosis of alveolar myofibroblasts. We show that these processes are dependent upon FGF/MAPK signaling and forced activation of MAPK/ERK signaling partially corrected alveolar simplification and restored alveolar myofibroblast number and AT2 cell proliferation in HS deficient mice. These data reveal HS-dependent myofibroblast heterogeneity and function as an essential orchestrator for developing alveolar niche critical for the generation of gas exchange units.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Chong Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Wei Wei
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haonan Wang
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yutian Lai
- Department of Lung Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ming Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Shenfei Sun
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jiashuang Lai
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanxiang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Fei Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Xinhua Lin
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Niethamer TK, Planer JD, Morley MP, Babu A, Zhao G, Basil MC, Cantu E, Frank DB, Diamond JM, Nottingham AN, Li S, Sharma A, Hallquist H, Levin LI, Zhou S, Vaughan AE, Morrisey EE. Longitudinal single-cell profiles of lung regeneration after viral infection reveal persistent injury-associated cell states. Cell Stem Cell 2025; 32:302-321.e6. [PMID: 39818203 PMCID: PMC11805657 DOI: 10.1016/j.stem.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
Functional regeneration of the lung's gas exchange surface following injury requires the coordination of a complex series of cell behaviors within the alveolar niche. Using single-cell transcriptomics combined with lineage tracing of proliferating progenitors, we examined mouse lung regeneration after influenza injury, demonstrating an asynchronously phased response across different cellular compartments. This longitudinal atlas of injury responses has produced a catalog of transient and persistent transcriptional alterations in cells as they transit across axes of differentiation. These cell states include an injury-induced capillary endothelial cell (iCAP) that arises after injury, persists indefinitely, and shares hallmarks with developing lung endothelium and endothelial aberrations found in degenerative human lung diseases. This dataset provides a foundational resource to understand the complexity of cellular and molecular responses to injury and correlations to responses found in human development and disease.
Collapse
Affiliation(s)
- Terren K Niethamer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| | - Joseph D Planer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Apoorva Babu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gan Zhao
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Cantu
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Frank
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ana N Nottingham
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shanru Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arnav Sharma
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Hannah Hallquist
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lillian I Levin
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-Children's Hospital of Philadelphia Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Negretti NM, Son Y, Crooke P, Plosa EJ, Benjamin JT, Jetter CS, Bunn C, Mignemi N, Marini J, Hackett AN, Ransom M, Garg S, Nichols D, Guttentag SH, Pua HH, Blackwell TS, Zacharias W, Frank DB, Kozub JA, Mahadevan-Jansen A, Krystofiak E, Kropski JA, Wright CV, Millis B, Sucre JM. Epithelial outgrowth through mesenchymal rings drives lung alveologenesis. JCI Insight 2025; 10:e187876. [PMID: 39773701 PMCID: PMC11949025 DOI: 10.1172/jci.insight.187876] [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: 10/09/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Determining how alveoli are formed and maintained is critical to understanding lung organogenesis and regeneration after injury. To study the cellular dynamics of this critical stage of lung development, we have used scanned oblique-plane illumination microscopy of living lung slices to observe alveologenesis in real time at high resolution over several days. Contrary to the prevailing notion that alveologenesis occurs by airspace subdivision via ingrowing septa, we found that alveoli form by ballooning epithelial outgrowth supported by contracting mesenchymal ring structures. Systematic analysis has produced a computational model of finely timed cellular structural changes that drive normal alveologenesis. With this model, we can now quantify how perturbing known regulatory intercellular signaling pathways and cell migration processes affects alveologenesis. In the future, this paradigm and platform can be leveraged for mechanistic studies and screening for therapies to promote lung regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nicholas Mignemi
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - John Marini
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | | | | - Heather H. Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Medicine, and
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - William Zacharias
- Department of Pediatrics, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - David B. Frank
- Department of Cardiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - John A. Kozub
- Department of Bioengineering
- Vanderbilt Biophotonics Center, and
- Department of Physics, Vanderbilt University, Nashville, Tennessee, USA
| | - Anita Mahadevan-Jansen
- Department of Bioengineering
- Vanderbilt Biophotonics Center, and
- Department of Surgery, Neurological Surgery and Otolaryngology, and
| | - Evan Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jonathan A. Kropski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Medicine, and
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher V.E. Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Bryan Millis
- Vanderbilt Biophotonics Center, and
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jennifer M.S. Sucre
- Department of Pediatrics
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
11
|
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
|
12
|
Wang C, Liu S, Li C, Wang Z, Ming R, Huang L. Monitoring the Cascade of Monocyte-Derived Macrophages to Influenza Virus Infection in Human Alveolus Chips. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60045-60055. [PMID: 39450775 DOI: 10.1021/acsami.4c15125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Respiratory viruses ravage the world and seriously threaten people's health. Despite intense research efforts, the immune mechanism underlying respiratory virus-induced acute lung injury (ALI) and pulmonary fibrosis (PF) has not been fully elucidated. Here, the cascade of monocyte-derived macrophages to influenza A virus infection is monitored on an optimized human alveolus chip to reveal the role of macrophages in the development of ALI and PF. We find that viral infection causes damage to the alveolar air-liquid barrier and the release of inflammatory cytokines, which induce the M0 macrophages to gather and polarize to the M1 phenotype at the damaged site through recruitment, adhesion, migration, and activation, leading to ALI. Afterward, M1 macrophages polarize into the M2 phenotype, and then transform into myofibroblasts, followed by enhanced secretion of various anti-inflammatory cytokines and profibrotic cytokines, to promote PF. Our study provides an insight into the pathogenesis of virus-induced ALI and PF, which will assist in the development of therapeutic strategies and drugs for treating influenza and other respiratory virus infections.
Collapse
Affiliation(s)
- Chenguang Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Shujun Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chuyu Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhongjie Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Ruiqi Ming
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lili Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
- Tangshan Research Institute, Beijing Institute of Technology, Tangshan 063000, P. R. China
| |
Collapse
|
13
|
Spurgin S, Nguimtsop AM, Chaudhry FN, Michki SN, Salvador J, Iruela-Arispe ML, Zepp JA, Mukhopadhyay S, Cleaver O. Spatiotemporal dynamics of primary and motile cilia throughout lung development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620342. [PMID: 39484464 PMCID: PMC11527191 DOI: 10.1101/2024.10.25.620342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Cilia are specialized structures found on a variety of mammalian cells, with variable roles in the transduction of mechanical and biological signals (by primary cilia, PC), as well as the generation of fluid flow (by motile cilia). Their critical role in the establishment of a left-right axis in early development is well described, as is the innate immune function of multiciliated upper airway epithelium. By contrast, the dynamics of ciliary status during organogenesis and postnatal development is largely unknown. In this study, we define the progression of ciliary status within the endothelium, epithelium, and mesenchyme of the lung. Remarkably, we find that endothelial cells (ECs) lack PC at all stages of development, except in low numbers in the most proximal portions of the pulmonary arteries. In the lung epithelium, a proximodistal ciliary gradient is established over time, as the uniformly mono-ciliated epithelium transitions into proximal, multiciliated cells, and the distal alveolar epithelium loses its cilia. Mesenchymal cells, interestingly, are uniformly ciliated in early development, but with restriction to PDGFRα+ fibroblasts in the adult alveoli. This dynamic process in multiple cellular populations both challenges prior assertions that PC are found on all cells, and highlights a need to understand their spatiotemporal functions.
Collapse
Affiliation(s)
- Stephen Spurgin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas, USA 75390
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas, USA 75390
| | - Ange Michelle Nguimtsop
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas, USA 75390
| | - Fatima N. Chaudhry
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania USA 19104
| | - Sylvia N. Michki
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania USA 19104
| | - Jocelynda Salvador
- Department of Cell and Developmental Biology, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA 60611
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA 60611
| | - Jarod A. Zepp
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania USA 19104
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, Texas, USA 75390
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas, USA 75390
| |
Collapse
|
14
|
Jung H, Kim DH, Díaz RE, White JM, Rucknagel S, Mosby L, Wang Y, Reddy S, Winkler ES, Hassan AO, Ying B, Diamond MS, Locksley RM, Fraser JS, Van Dyken SJ. An ILC2-chitinase circuit restores lung homeostasis after epithelial injury. Sci Immunol 2024; 9:eadl2986. [PMID: 39423283 PMCID: PMC11854321 DOI: 10.1126/sciimmunol.adl2986] [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: 10/10/2023] [Revised: 05/15/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Environmental exposures increase the risk for severe lung disease, but specific drivers of persistent epithelial injury and immune dysfunction remain unclear. Here, we identify a feedback circuit triggered by chitin, a common component of airborne particles, that affects lung health after epithelial injury. In mice, epithelial damage disrupts lung chitinase activity, leading to environmental chitin accumulation, impaired epithelial renewal, and group 2 innate lymphoid cell (ILC2) activation. ILC2s, in turn, restore homeostasis by inducing acidic mammalian chitinase (AMCase) in regenerating epithelial cells and promoting chitin degradation, epithelial differentiation, and inflammatory resolution. Mice lacking AMCase or ILC2s fail to clear chitin and exhibit increased mortality and impaired epithelial regeneration after injury. These effects are ameliorated by chitinase replacement therapy, demonstrating that chitin degradation is crucial for recovery after various forms of lung perturbation. Thus, the ILC2-chitinase response circuit may serve as a target for alleviating persistent postinjury lung epithelial and immune dysfunction.
Collapse
Affiliation(s)
- Haerin Jung
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Do-Hyun Kim
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Roberto Efraín Díaz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco; San Francisco, CA, USA
| | - J. Michael White
- Department of Pathology & Immunology, Washington University Gnotobiotic Core Facility, Washington University School of Medicine; St. Louis, MO, USA
| | - Summer Rucknagel
- Department of Pathology & Immunology, Washington University Gnotobiotic Core Facility, Washington University School of Medicine; St. Louis, MO, USA
| | - Lauryn Mosby
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Yilin Wang
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Sanjana Reddy
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Emma S. Winkler
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Ahmed O. Hassan
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Baoling Ying
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Michael S. Diamond
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine; St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine; St. Louis, MO, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine; St. Louis, MO, USA
| | - Richard M. Locksley
- Department of Microbiology & Immunology, University of California, San Francisco; San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco; San Francisco, CA, USA
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco; San Francisco, CA, USA
| | - Steven J. Van Dyken
- Department of Pathology & Immunology, Washington University School of Medicine; St. Louis, MO, USA
| |
Collapse
|
15
|
Xu L, Tan C, Barr J, Talaba N, Verheyden J, Chin JS, Gaboyan S, Kasaraneni N, Elgamal RM, Gaulton KJ, Lin G, Afshar K, Golts E, Meier A, Crotty Alexander LE, Borok Z, Shen Y, Chung WK, McCulley DJ, Sun X. Context-dependent roles of mitochondrial LONP1 in orchestrating the balance between airway progenitor versus progeny cells. Cell Stem Cell 2024; 31:1465-1483.e6. [PMID: 39181129 DOI: 10.1016/j.stem.2024.08.001] [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: 09/16/2023] [Revised: 06/12/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
While all eukaryotic cells are dependent on mitochondria for function, in a complex tissue, which cell type and which cell behavior are more sensitive to mitochondrial deficiency remain unpredictable. Here, we show that in the mouse airway, compromising mitochondrial function by inactivating mitochondrial protease gene Lonp1 led to reduced progenitor proliferation and differentiation during development, apoptosis of terminally differentiated ciliated cells and their replacement by basal progenitors and goblet cells during homeostasis, and failed airway progenitor migration into damaged alveoli following influenza infection. ATF4 and the integrated stress response (ISR) pathway are elevated and responsible for the airway phenotypes. Such context-dependent sensitivities are predicted by the selective expression of Bok, which is required for ISR activation. Reduced LONP1 expression is found in chronic obstructive pulmonary disease (COPD) airways with squamous metaplasia. These findings illustrate a cellular energy landscape whereby compromised mitochondrial function could favor the emergence of pathological cell types.
Collapse
Affiliation(s)
- Le Xu
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chunting Tan
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Justinn Barr
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole Talaba
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jamie Verheyden
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ji Sun Chin
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Samvel Gaboyan
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nikita Kasaraneni
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ruth M Elgamal
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kyle J Gaulton
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Grace Lin
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Kamyar Afshar
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Eugene Golts
- Department of Surgery, Division of Cardiovascular and Thoracic Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Angela Meier
- Department of Anesthesiology, Division of Critical Care, University of California, San Diego, La Jolla, CA, USA
| | - Laura E Crotty Alexander
- Pulmonary and Critical Care Section, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032, USA; JP Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - David J McCulley
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xin Sun
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
16
|
Wang J, Peng X, Yuan N, Wang B, Chen S, Wang B, Xie L. Interplay between pulmonary epithelial stem cells and innate immune cells contribute to the repair and regeneration of ALI/ARDS. Transl Res 2024; 272:111-125. [PMID: 38897427 DOI: 10.1016/j.trsl.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
Mammalian lung is the important organ for ventilation and exchange of air and blood. Fresh air and venous blood are constantly delivered through the airway and vascular tree to the alveolus. Based on this, the airways and alveolis are persistently exposed to the external environment and are easily suffered from toxins, irritants and pathogens. For example, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common cause of respiratory failure in critical patients, whose typical pathological characters are diffuse epithelial and endothelial damage resulting in excessive accumulation of inflammatory fluid in the alveolar cavity. The supportive treatment is the main current treatment for ALI/ARDS with the lack of targeted effective treatment strategies. However, ALI/ARDS needs more targeted treatment measures. Therefore, it is extremely urgent to understand the cellular and molecular mechanisms that maintain alveolar epithelial barrier and airway integrity. Previous researches have shown that the lung epithelial cells with tissue stem cell function have the ability to repair and regenerate after injury. Also, it is able to regulate the phenotype and function of innate immune cells involving in regeneration of tissue repair. Meanwhile, we emphasize that interaction between the lung epithelial cells and innate immune cells is more supportive to repair and regenerate in the lung epithelium following acute lung injury. We reviewed the recent advances in injury and repair of lung epithelial stem cells and innate immune cells in ALI/ARDS, concentrating on alveolar type 2 cells and alveolar macrophages and their contribution to post-injury repair behavior of ALI/ARDS through the latest potential molecular communication mechanisms. This will help to develop new research strategies and therapeutic targets for ALI/ARDS.
Collapse
Affiliation(s)
- Jiang Wang
- College of Pulmonary & Critical Care Medicine, the Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Xinyue Peng
- Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Na Yuan
- Department of Pulmonary & Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Bin Wang
- Department of Thoracic Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Siyu Chen
- Department of Thoracic Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Bo Wang
- Department of Thoracic Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, the Eighth Medical Center of Chinese PLA General Hospital, Beijing 100091, China; Medical School of Chinese PLA, Beijing 100853, China.
| |
Collapse
|
17
|
Pelizzo G, Calcaterra V, Baldassarre P, Marinaro M, Taranto S, Ceresola M, Capelo G, Gazzola C, Zuccotti G. The impact of hormones on lung development and function: an overlooked aspect to consider from early childhood. Front Endocrinol (Lausanne) 2024; 15:1425149. [PMID: 39371928 PMCID: PMC11449876 DOI: 10.3389/fendo.2024.1425149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/29/2024] [Indexed: 10/08/2024] Open
Abstract
The impact of hormones on the respiratory system constitutes a multifaceted and intricate facet of human biology. We propose a comprehensive review of recent advancements in understanding the interactions between hormones and pulmonary development and function, focusing on pediatric populations. We explore how hormones can influence ventilation, perfusion, and pulmonary function, from regulating airway muscle tone to modulating the inflammatory response. Hormones play an important role in the growth and development of lung tissues, influencing them from early stages through infancy, childhood, adolescence, and into adulthood. Glucocorticoids, thyroid hormones, insulin, ghrelin, leptin, glucagon-like peptide 1 (GLP-1), retinoids, cholecalciferol sex steroids, hormones derived from adipose tissue, factors like insulin, granulocyte-macrophage colony-stimulating factor (GM-CSF) and glucagon are key players in modulating respiratory mechanics and inflammation. While ample evidence underscores the impact of hormones on lung development and function, along with sex-related differences in the prevalence of respiratory disorders, further research is needed to clarify their specific roles in these conditions. Further research into the mechanisms underlying hormonal effects is essential for the development of customizing therapeutic approaches for respiratory diseases. Understanding the impact of hormones on lung function could be valuable for developing personalized monitoring approaches in both medical and surgical pediatric settings, in order to improve outcomes and the quality of care for pediatric patients.
Collapse
Affiliation(s)
- Gloria Pelizzo
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| | | | - Michela Marinaro
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | - Silvia Taranto
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| | - Michele Ceresola
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | - Gerson Capelo
- Pediatric Surgery Department, Buzzi Children’s Hospital, Milan, Italy
| | | | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
- Pediatric Department, Buzzi Children’s Hospital, Milan, Italy
| |
Collapse
|
18
|
He H, Bell SM, Davis AK, Zhao S, Sridharan A, Na CL, Guo M, Xu Y, Snowball J, Swarr DT, Zacharias WJ, Whitsett JA. PRDM3/16 regulate chromatin accessibility required for NKX2-1 mediated alveolar epithelial differentiation and function. Nat Commun 2024; 15:8112. [PMID: 39284798 PMCID: PMC11405758 DOI: 10.1038/s41467-024-52154-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024] Open
Abstract
While the critical role of NKX2-1 and its transcriptional targets in lung morphogenesis and pulmonary epithelial cell differentiation is increasingly known, mechanisms by which chromatin accessibility alters the epigenetic landscape and how NKX2-1 interacts with other co-activators required for alveolar epithelial cell differentiation and function are not well understood. Combined deletion of the histone methyl transferases Prdm3 and Prdm16 in early lung endoderm causes perinatal lethality due to respiratory failure from loss of AT2 cells and the accumulation of partially differentiated AT1 cells. Combination of single-cell RNA-seq, bulk ATAC-seq, and CUT&RUN data demonstrate that PRDM3 and PRDM16 regulate chromatin accessibility at NKX2-1 transcriptional targets critical for perinatal AT2 cell differentiation and surfactant homeostasis. Lineage specific deletion of PRDM3/16 in AT2 cells leads to lineage infidelity, with PRDM3/16 null cells acquiring partial AT1 fate. Together, these data demonstrate that NKX2-1-dependent regulation of alveolar epithelial cell differentiation is mediated by epigenomic modulation via PRDM3/16.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
| | - Sheila M Bell
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Kuenzi Davis
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shuyang Zhao
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anusha Sridharan
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cheng-Lun Na
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Minzhe Guo
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yan Xu
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John Snowball
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Daniel T Swarr
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William J Zacharias
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey A Whitsett
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Koc-Gunel S, Liu EC, Gautam LK, Calvert BA, Murthy S, Harriott NC, Nawroth JC, Zhou B, Krymskaya VP, Ryan AL. Targeting Fibroblast-Endothelial Interactions in LAM Pathogenesis: 3D Spheroid and Spatial Transcriptomic Insights for Therapeutic Innovation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.12.544372. [PMID: 37398026 PMCID: PMC10312665 DOI: 10.1101/2023.06.12.544372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a progressive lung disease with limited treatments, largely due to an incomplete understanding of its pathogenesis. Lymphatic endothelial cells (LECs) invade LAM cell clusters, which include HMB-45-positive epithelioid cells and smooth muscle α-actin-expressing LAM-associated fibroblasts (LAMFs). Recent evidence shows that LAMFs resemble cancer-associated fibroblasts, with LAMF-LEC interactions contributing to disease progression. To explore these mechanisms, we used spatial transcriptomics on LAM lung tissues and identified a gene cluster enriched in kinase signaling pathways linked to myofibroblasts and co-expressed with LEC markers. Kinase arrays revealed elevated PDGFR and FGFR in LAMFs. Using a 3D co-culture spheroid model of primary LAMFs and LECs, we observed increased invasion in LAMF-LEC spheroids compared to non-LAM fibroblasts. Treatment with sorafenib, a multikinase inhibitor, significantly reduced invasion, outperforming Rapamycin. We also confirmed TSC2-null AML cells as key VEGF-A secretors, which was suppressed by sorafenib in both AML cells and LAMFs. These findings highlight VEGF-A and bFGF as potential therapeutic targets and suggest multikinase inhibition as a promising strategy for LAM.
Collapse
Affiliation(s)
- Sinem Koc-Gunel
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Internal Medicine II, Infectious Diseases, University Hospital Frankfurt; Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Emily C. Liu
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| | - Lalit K. Gautam
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| | - Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| | - Shubha Murthy
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| | - Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| | - Janna C. Nawroth
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California; Los Angeles, CA, USA
- Helmholtz Pioneer Campus and Institute of Biological and Medical Imaging; Helmholtz Zentrum München, Neuherberg, Germany
| | - Beiyun Zhou
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
| | - Vera P. Krymskaya
- Division of Pulmonary and Critical Care Medicine, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California; Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa; Iowa City, IA, USA
| |
Collapse
|
20
|
Rafikov R, de Jesus Perez V, Dekan A, Kudryashova TV, Rafikova O. Deciphering the Complexities of Pulmonary Hypertension: The Emergent Role of Single-Cell Omics. Am J Respir Cell Mol Biol 2024; 72:32-40. [PMID: 39141563 PMCID: PMC11707669 DOI: 10.1165/rcmb.2024-0145ps] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024] Open
Abstract
Expanding upon the critical advancements brought forth by single-cell omics in pulmonary hypertension (PH) research, this review delves deep into how these technologies have been piloted in a new era of understanding this complex disease. By leveraging the power of single cell transcriptomics (scRNA-seq), researchers can now dissect the complicated cellular ecosystem of the lungs, examining the key players such as endothelial cells, smooth muscle cells, pericytes, and immune cells, and their unique roles in the pathogenesis of PH. This more granular view is beyond the limitations of traditional bulk analysis, allowing for the identification of novel therapeutic targets previously obscured in the aggregated data. Connectome analysis based on single-cell omics of the cells involved in pathological changes can reveal a clearer picture of the cellular interactions and transitions in the cellular subtypes. Furthermore, the review acknowledges the challenges that lie ahead, including the need for enhancing the resolution of scRNA-seq to capture even finer details of cellular changes, overcoming logistical barriers in processing human tissue samples, and the necessity of integrating diverse omics approaches to fully comprehend the molecular underpinnings of PH. The promise of these single-cell technologies is immense, offering the potential for targeted drug development and the discovery of biomarkers for early diagnosis and disease monitoring. Through these advancements, the field moves closer to realizing the goal of precision medicine for patients with PH.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Indiana University School of Medicine, Indianapolis, Indiana, United States;
| | | | - Aleksandr Dekan
- Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Tatiana V Kudryashova
- University of Pittsburgh Department of Medicine, Pittsburgh, Pennsylvania, United States
| | - Olga Rafikova
- Indiana University Purdue University at Indianapolis, Indianapolis, Indiana, United States
| |
Collapse
|
21
|
Wu ML, Wheeler K, Silasi R, Lupu F, Griffin CT. Endothelial Chromatin-Remodeling Enzymes Regulate the Production of Critical ECM Components During Murine Lung Development. Arterioscler Thromb Vasc Biol 2024; 44:1784-1798. [PMID: 38868942 PMCID: PMC11624602 DOI: 10.1161/atvbaha.124.320881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND The chromatin-remodeling enzymes BRG1 (brahma-related gene 1) and CHD4 (chromodomain helicase DNA-binding protein 4) independently regulate the transcription of genes critical for vascular development, but their coordinated impact on vessels in late-stage embryos has not been explored. METHODS In this study, we genetically deleted endothelial Brg1 and Chd4 in mixed background mice (Brg1fl/fl;Chd4fl/fl;VE-Cadherin-Cre), and littermates that were negative for Cre recombinase were used as controls. Tissues were analyzed by immunostaining, immunoblot, and flow cytometry. Quantitative reverse transcription polymerase chain reaction was used to determine gene expression, and chromatin immunoprecipitation revealed gene targets of BRG1 and CHD4 in cultured endothelial cells. RESULTS We found Brg1/Chd4 double mutants grew normally but died soon after birth with small and compact lungs. Despite having normal cellular composition, distal air sacs of the mutant lungs displayed diminished ECM (extracellular matrix) components and TGFβ (transforming growth factor-β) signaling, which typically promotes ECM synthesis. Transcripts for collagen- and elastin-related genes and the TGFβ ligand Tgfb1 were decreased in mutant lung endothelial cells, but genetic deletion of endothelial Tgfb1 failed to recapitulate the small lungs and ECM defects seen in Brg1/Chd4 mutants. We instead found several ECM genes to be direct targets of BRG1 and CHD4 in cultured endothelial cells. CONCLUSIONS Collectively, our data highlight essential roles for endothelial chromatin-remodeling enzymes in promoting ECM deposition in the distal lung tissue during the saccular stage of embryonic lung development.
Collapse
Affiliation(s)
- Meng-Ling Wu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Kate Wheeler
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
22
|
Luo S, Rollins S, Schmitz-Abe K, Tam A, Li Q, Shi J, Lin J, Wang R, Agrawal PB. The solute carrier family 26 member 9 modifies rapidly progressing cystic fibrosis associated with homozygous F508del CFTR mutation. Clin Chim Acta 2024; 561:119765. [PMID: 38852790 DOI: 10.1016/j.cca.2024.119765] [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: 03/26/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND AIMS Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations to the CF transmembrane conductance regulator (CFTR). Symptoms and severity of the disease can be quite variable suggesting modifier genes play an important role. MATERIALS AND METHODS Exome sequencing was performed on six individuals carrying homozygous deltaF508 for CFTR genotype but present with rapidly progressing CF (RPCF). Data was analyzed using an unbiased genome-wide genetic burden test against 3076 controls. Single cell RNA sequencing data from LungMAP was utilized to evaluate unique and co-expression of candidate genes, and structural modeling to evaluate the deleterious effects of identified candidate variants. RESULTS We have identified solute carrier family 26 member 9 (SLC26A9) as a modifier gene to be associated with RPCF. Two rare missense SLC26A9 variants were discovered in three of six individuals deemed to have RPCF: c.229G > A; p.G77S (present in two patients), and c.1885C > T; p.P629S. Co-expression of SLC26A9 and CFTR mRNA is limited across different lung cell types, with the highest level of co-expression seen in human (6.3 %) and mouse (9.0 %) alveolar type 2 (AT2) cells. Structural modeling suggests deleterious effects of these mutations as they are in critical protein domains which might affect the anion transport capability of SLC26A9. CONCLUSION The enrichment of rare and potentially deleterious SLC26A9 mutations in patients with RPCF suggests SLC26A9 may act as an alternative anion transporter in CF and is a modifier gene associated with this lung phenotype.
Collapse
Affiliation(s)
- Shiyu Luo
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stuart Rollins
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA
| | - Klaus Schmitz-Abe
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amy Tam
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Qifei Li
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jiahai Shi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasmine Lin
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ruobing Wang
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA; Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02115, USA.
| | - Pankaj B Agrawal
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Song L, Li K, Chen H, Xie L. Cell Cross-Talk in Alveolar Microenvironment: From Lung Injury to Fibrosis. Am J Respir Cell Mol Biol 2024; 71:30-42. [PMID: 38579159 PMCID: PMC11225874 DOI: 10.1165/rcmb.2023-0426tr] [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: 12/05/2023] [Accepted: 04/05/2024] [Indexed: 04/07/2024] Open
Abstract
Alveoli are complex microenvironments composed of various cell types, including epithelial, fibroblast, endothelial, and immune cells, which work together to maintain a delicate balance in the lung environment, ensuring proper growth, development, and an effective response to lung injuries. However, prolonged inflammation or aging can disrupt normal interactions among these cells, leading to impaired repair processes and a substantial decline in lung function. Therefore, it is essential to understand the key mechanisms underlying the interactions among the major cell types within the alveolar microenvironment. We explored the key mechanisms underlying the interactions among the major cell types within the alveolar microenvironment. These interactions occur through the secretion of signaling factors and play crucial roles in the response to injury, repair mechanisms, and the development of fibrosis in the lungs. Specifically, we focused on the regulation of alveolar type 2 cells by fibroblasts, endothelial cells, and macrophages. In addition, we explored the diverse phenotypes of fibroblasts at different stages of life and in response to lung injury, highlighting their impact on matrix production and immune functions. Furthermore, we summarize the various phenotypes of macrophages in lung injury and fibrosis as well as their intricate interplay with other cell types. This interplay can either contribute to the restoration of immune homeostasis in the alveoli or impede the repair process. Through a comprehensive exploration of these cell interactions, we aim to reveal new insights into the molecular mechanisms that drive lung injury toward fibrosis and identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Licheng Song
- College of Pulmonary and Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China; and
| | - Kuan Li
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe Hospital, Tianjin University, Tianjin, China
| | - Huaiyong Chen
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe Hospital, Tianjin University, Tianjin, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China; and
| |
Collapse
|
24
|
Hill ABT, Murphy YM, Polkoff KM, Edwards L, Walker DM, Moatti A, Greenbaum A, Piedrahita JA. A gene edited pig model for studying LGR5 + stem cells: implications for future applications in tissue regeneration and biomedical research. Front Genome Ed 2024; 6:1401163. [PMID: 38903529 PMCID: PMC11187295 DOI: 10.3389/fgeed.2024.1401163] [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: 03/14/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Recent advancements in genome editing techniques, notably CRISPR-Cas9 and TALENs, have marked a transformative era in biomedical research, significantly enhancing our understanding of disease mechanisms and helping develop novel therapies. These technologies have been instrumental in creating precise animal models for use in stem cell research and regenerative medicine. For instance, we have developed a transgenic pig model to enable the investigation of LGR5-expressing cells. The model was designed to induce the expression of H2B-GFP under the regulatory control of the LGR5 promoter via CRISPR/Cas9-mediated gene knock-in. Notably, advancements in stem cell research have identified distinct subpopulations of LGR5-expressing cells within adult human, mouse, and pig tissues. LGR5, a leucine-rich repeat-containing G protein-coupled receptor, enhances WNT signaling and these LGR5+ subpopulations demonstrate varied roles and anatomical distributions, underscoring the necessity for suitable translational models. This transgenic pig model facilitates the tracking of LGR5-expressing cells and has provided valuable insights into the roles of these cells across different tissues and species. For instance, in pulmonary tissue, Lgr5+ cells in mice are predominantly located in alveolar compartments, driving alveolar differentiation of epithelial progenitors via Wnt pathway activation. In contrast, in pigs and humans, these cells are situated in a unique sub-basal position adjacent to the airway epithelium. In fetal stages a pattern of LGR5 expression during lung bud tip formation is evident in humans and pigs but is lacking in mice. Species differences with respect to LGR5 expression have also been observed in the skin, intestines, and cochlea further reinforcing the need for careful selection of appropriate translational animal models. This paper discusses the potential utility of the LGR5+ pig model in exploring the role of LGR5+ cells in tissue development and regeneration with the goal of translating these findings into human and animal clinical applications.
Collapse
Affiliation(s)
- Amanda B. T. Hill
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Yanet M. Murphy
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Kathryn M. Polkoff
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Laura Edwards
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Derek M. Walker
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| | - Jorge A. Piedrahita
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
25
|
Schmitt P, Duval A, Camus M, Lefrançais E, Roga S, Dedieu C, Ortega N, Bellard E, Mirey E, Mouton-Barbosa E, Burlet-Schiltz O, Gonzalez-de-Peredo A, Cayrol C, Girard JP. TL1A is an epithelial alarmin that cooperates with IL-33 for initiation of allergic airway inflammation. J Exp Med 2024; 221:e20231236. [PMID: 38597952 PMCID: PMC11010340 DOI: 10.1084/jem.20231236] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Epithelium-derived cytokines or alarmins, such as interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP), are major players in type 2 immunity and asthma. Here, we demonstrate that TNF-like ligand 1A (TL1A) is an epithelial alarmin, constitutively expressed in alveolar epithelium at steady state in both mice and humans, which cooperates with IL-33 for early induction of IL-9high ILC2s during the initiation of allergic airway inflammation. Upon synergistic activation by IL-33 and TL1A, lung ILC2s acquire a transient IL-9highGATA3low "ILC9" phenotype and produce prodigious amounts of IL-9. A combination of large-scale proteomic analyses, lung intravital microscopy, and adoptive transfer of ILC9 cells revealed that high IL-9 expression distinguishes a multicytokine-producing state-of-activated ILC2s with an increased capacity to initiate IL-5-dependent allergic airway inflammation. Similar to IL-33 and TSLP, TL1A is expressed in airway basal cells in healthy and asthmatic human lungs. Together, these results indicate that TL1A is an epithelium-derived cytokine and an important cofactor of IL-33 in the airways.
Collapse
Affiliation(s)
- Pauline Schmitt
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anais Duval
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Mylène Camus
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emma Lefrançais
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Stéphane Roga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Cécile Dedieu
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Nathalie Ortega
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Elisabeth Bellard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emilie Mirey
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anne Gonzalez-de-Peredo
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Corinne Cayrol
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
26
|
Yang X, Chen Y, Yang Y, Li S, Mi P, Jing N. The molecular and cellular choreography of early mammalian lung development. MEDICAL REVIEW (2021) 2024; 4:192-206. [PMID: 38919401 PMCID: PMC11195428 DOI: 10.1515/mr-2023-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/08/2024] [Indexed: 06/27/2024]
Abstract
Mammalian lung development starts from a specific cluster of endodermal cells situated within the ventral foregut region. With the orchestrating of delicate choreography of transcription factors, signaling pathways, and cell-cell communications, the endodermal diverticulum extends into the surrounding mesenchyme, and builds the cellular and structural basis of the complex respiratory system. This review provides a comprehensive overview of the current molecular insights of mammalian lung development, with a particular focus on the early stage of lung cell fate differentiation and spatial patterning. Furthermore, we explore the implications of several congenital respiratory diseases and the relevance to early organogenesis. Finally, we summarize the unprecedented knowledge concerning lung cell compositions, regulatory networks as well as the promising prospect for gaining an unbiased understanding of lung development and lung malformations through state-of-the-art single-cell omics.
Collapse
Affiliation(s)
- Xianfa Yang
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Yingying Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| | - Yun Yang
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Shiting Li
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- Institute of Biomedical Research, Yunnan University, Kunming, Yunnan Province, China
| | - Panpan Mi
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Naihe Jing
- Guangzhou National Laboratory, Guangzhou, Guangdong Province, China
| |
Collapse
|
27
|
Li X, Mara AB, Musial SC, Kolling FW, Gibbings SL, Gerebtsov N, Jakubzick CV. Coordinated chemokine expression defines macrophage subsets across tissues. Nat Immunol 2024; 25:1110-1122. [PMID: 38698086 PMCID: PMC11565582 DOI: 10.1038/s41590-024-01826-9] [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: 05/18/2023] [Accepted: 03/20/2024] [Indexed: 05/05/2024]
Abstract
Lung-resident macrophages, which include alveolar macrophages and interstitial macrophages (IMs), exhibit a high degree of diversity, generally attributed to different activation states, and often complicated by the influx of monocytes into the pool of tissue-resident macrophages. To gain a deeper insight into the functional diversity of IMs, here we perform comprehensive transcriptional profiling of resident IMs and reveal ten distinct chemokine-expressing IM subsets at steady state and during inflammation. Similar IM subsets that exhibited coordinated chemokine signatures and differentially expressed genes were observed across various tissues and species, indicating conserved specialized functional roles. Other macrophage types shared specific IM chemokine profiles, while also presenting their own unique chemokine signatures. Depletion of CD206hi IMs in Pf4creR26EYFP+DTR and Pf4creR26EYFPCx3cr1DTR mice led to diminished inflammatory cell recruitment, reduced tertiary lymphoid structure formation and fewer germinal center B cells in models of allergen- and infection-driven inflammation. These observations highlight the specialized roles of IMs, defined by their coordinated chemokine production, in regulating immune cell influx and organizing tertiary lymphoid tissue architecture.
Collapse
Affiliation(s)
- Xin Li
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Arlind B Mara
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Shawn C Musial
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | - Fred W Kolling
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine, Hanover, NH, USA
| | | | - Nikita Gerebtsov
- Lab for Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
28
|
Paramore SV, Trenado-Yuste C, Sharan R, Nelson CM, Devenport D. Vangl-dependent mesenchymal thinning shapes the distal lung during murine sacculation. Dev Cell 2024; 59:1302-1316.e5. [PMID: 38569553 PMCID: PMC11111357 DOI: 10.1016/j.devcel.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 10/18/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The planar cell polarity (PCP) complex is speculated to function in murine lung development, where branching morphogenesis generates an epithelial tree whose distal tips expand dramatically during sacculation. Here, we show that PCP is dispensable in the airway epithelium for sacculation. Rather, we find a Celsr1-independent role for the PCP component Vangl in the pulmonary mesenchyme: loss of Vangl1/2 inhibits mesenchymal thinning and expansion of the saccular epithelium. Further, loss of mesenchymal Wnt5a mimics sacculation defects observed in Vangl2-mutant lungs, implicating mesenchymal Wnt5a/Vangl signaling as a key regulator of late lung morphogenesis. A computational model predicts that sacculation requires a fluid mesenchymal compartment. Lineage-tracing and cell-shape analyses are consistent with the mesenchyme acting as a fluid tissue, suggesting that loss of Vangl1/2 impacts the ability of mesenchymal cells to exchange neighbors. Our data thus identify an explicit function for Vangl and the pulmonary mesenchyme in actively shaping the saccular epithelium.
Collapse
Affiliation(s)
- Sarah V Paramore
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Carolina Trenado-Yuste
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Rishabh Sharan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA.
| | - Danelle Devenport
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
29
|
Hassan D, Chen J. CEBPA restricts alveolar type 2 cell plasticity during development and injury-repair. Nat Commun 2024; 15:4148. [PMID: 38755149 PMCID: PMC11099190 DOI: 10.1038/s41467-024-48632-3] [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: 10/31/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024] Open
Abstract
Cell plasticity theoretically extends to all possible cell types, but naturally decreases as cells differentiate, whereas injury-repair re-engages the developmental plasticity. Here we show that the lung alveolar type 2 (AT2)-specific transcription factor (TF), CEBPA, restricts AT2 cell plasticity in the mouse lung. AT2 cells undergo transcriptional and epigenetic maturation postnatally. Without CEBPA, both neonatal and mature AT2 cells reduce the AT2 program, but only the former reactivate the SOX9 progenitor program. Sendai virus infection bestows mature AT2 cells with neonatal plasticity where Cebpa mutant, but not wild type, AT2 cells express SOX9, as well as more readily proliferate and form KRT8/CLDN4+ transitional cells. CEBPA promotes the AT2 program by recruiting the lung lineage TF NKX2-1. The temporal change in CEBPA-dependent plasticity reflects AT2 cell developmental history. The ontogeny of AT2 cell plasticity and its transcriptional and epigenetic mechanisms have implications in lung regeneration and cancer.
Collapse
Affiliation(s)
- Dalia Hassan
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Pediatrics, Perinatal Institute Division of Pulmonary Biology, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
30
|
Burgess CL, Huang J, Bawa PS, Alysandratos KD, Minakin K, Ayers LJ, Morley MP, Babu A, Villacorta-Martin C, Yampolskaya M, Hinds A, Thapa BR, Wang F, Matschulat A, Mehta P, Morrisey EE, Varelas X, Kotton DN. Generation of human alveolar epithelial type I cells from pluripotent stem cells. Cell Stem Cell 2024; 31:657-675.e8. [PMID: 38642558 PMCID: PMC11147407 DOI: 10.1016/j.stem.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/31/2024] [Accepted: 03/27/2024] [Indexed: 04/22/2024]
Abstract
Alveolar epithelial type I cells (AT1s) line the gas exchange barrier of the distal lung and have been historically challenging to isolate or maintain in cell culture. Here, we engineer a human in vitro AT1 model system via directed differentiation of induced pluripotent stem cells (iPSCs). We use primary adult AT1 global transcriptomes to suggest benchmarks and pathways, such as Hippo-LATS-YAP/TAZ signaling, enriched in these cells. Next, we generate iPSC-derived alveolar epithelial type II cells (AT2s) and find that nuclear YAP signaling is sufficient to promote a broad transcriptomic shift from AT2 to AT1 gene programs. The resulting cells express a molecular, morphologic, and functional phenotype reminiscent of human AT1 cells, including the capacity to form a flat epithelial barrier producing characteristic extracellular matrix molecules and secreted ligands. Our results provide an in vitro model of human alveolar epithelial differentiation and a potential source of human AT1s.
Collapse
Affiliation(s)
- Claire L Burgess
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jessie Huang
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Pushpinder S Bawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Kasey Minakin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Lauren J Ayers
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Michael P Morley
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Apoorva Babu
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | | | - Anne Hinds
- The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Bibek R Thapa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Feiya Wang
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Adeline Matschulat
- The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Pankaj Mehta
- Department of Physics, Boston University, Boston, MA 02215, USA
| | - Edward E Morrisey
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xaralabos Varelas
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA; Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
31
|
Bandyopadhyay G, Jehrio MG, Baker C, Bhattacharya S, Misra RS, Huyck HL, Chu C, Myers JR, Ashton J, Polter S, Cochran M, Bushnell T, Dutra J, Katzman PJ, Deutsch GH, Mariani TJ, Pryhuber GS. Bulk RNA sequencing of human pediatric lung cell populations reveals unique transcriptomic signature associated with postnatal pulmonary development. Am J Physiol Lung Cell Mol Physiol 2024; 326:L604-L617. [PMID: 38442187 PMCID: PMC11381037 DOI: 10.1152/ajplung.00385.2023] [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: 12/07/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
Postnatal lung development results in an increasingly functional organ prepared for gas exchange and pathogenic challenges. It is achieved through cellular differentiation and migration. Changes in the tissue architecture during this development process are well-documented and increasing cellular diversity associated with it are reported in recent years. Despite recent progress, transcriptomic and molecular pathways associated with human postnatal lung development are yet to be fully understood. In this study, we investigated gene expression patterns associated with healthy pediatric lung development in four major enriched cell populations (epithelial, endothelial, and nonendothelial mesenchymal cells, along with lung leukocytes) from 1-day-old to 8-yr-old organ donors with no known lung disease. For analysis, we considered the donors in four age groups [less than 30 days old neonates, 30 days to < 1 yr old infants, toddlers (1 to < 2 yr), and children 2 yr and older] and assessed differentially expressed genes (DEG). We found increasing age-associated transcriptional changes in all four major cell types in pediatric lung. Transition from neonate to infant stage showed highest number of DEG compared with the number of DEG found during infant to toddler- or toddler to older children-transitions. Profiles of differential gene expression and further pathway enrichment analyses indicate functional epithelial cell maturation and increased capability of antigen presentation and chemokine-mediated communication. Our study provides a comprehensive reference of gene expression patterns during healthy pediatric lung development that will be useful in identifying and understanding aberrant gene expression patterns associated with early life respiratory diseases.NEW & NOTEWORTHY This study presents postnatal transcriptomic changes in major cell populations in human lung, namely endothelial, epithelial, mesenchymal cells, and leukocytes. Although human postnatal lung development continues through early adulthood, our results demonstrate that greatest transcriptional changes occur in first few months of life during neonate to infant transition. These early transcriptional changes in lung parenchyma are particularly notable for functional maturation and activation of alveolar type II cell genes.
Collapse
Affiliation(s)
- Gautam Bandyopadhyay
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew G Jehrio
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Cameron Baker
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Ravi S Misra
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Heidie L Huyck
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - ChinYi Chu
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Jason R Myers
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - John Ashton
- UR Genomics Research Center, University of Rochester Medical Center, Rochester, New York, United States
| | - Steven Polter
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew Cochran
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Timothy Bushnell
- UR Flow Cytometry Core Facility, University of Rochester Medical Center, Rochester, New York, United States
| | - Jennifer Dutra
- UR Clinical & Translational Science Institute Informatics, University of Rochester Medical Center, Rochester, New York, United States
| | - Philip J Katzman
- Department of Pathology, University of Rochester Medical Center, Rochester, New York, United States
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital, Seattle, Washington, United States
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Program in Pediatric Molecular and Personalized Medicine, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
32
|
Yin Y, Koenitzer JR, Patra D, Dietmann S, Bayguinov P, Hagan AS, Ornitz DM. Identification of a myofibroblast differentiation program during neonatal lung development. Development 2024; 151:dev202659. [PMID: 38602479 PMCID: PMC11165721 DOI: 10.1242/dev.202659] [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: 12/26/2023] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFBs) and a stable but poorly described population of lipid-rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFBs). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single-cell RNA sequencing and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a MyoFB differentiation program that is distinct from other mesenchymal cell types and increases the known repertoire of mesenchymal cell types in the neonatal lung.
Collapse
Affiliation(s)
- Yongjun Yin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey R. Koenitzer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sabine Dietmann
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Peter Bayguinov
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S. Hagan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
33
|
Chaudhry FN, Michki NS, Shirmer DL, McGrath-Morrow S, Young LR, Frank DB, Zepp JA. Dynamic Hippo pathway activity underlies mesenchymal differentiation during lung alveolar morphogenesis. Development 2024; 151:dev202430. [PMID: 38602485 PMCID: PMC11112347 DOI: 10.1242/dev.202430] [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: 03/26/2024] [Indexed: 04/12/2024]
Abstract
Alveologenesis, the final stage in lung development, substantially remodels the distal lung, expanding the alveolar surface area for efficient gas exchange. Secondary crest myofibroblasts (SCMF) exist transiently in the neonatal distal lung and are crucial for alveologenesis. However, the pathways that regulate SCMF function, proliferation and temporal identity remain poorly understood. To address this, we purified SCMFs from reporter mice, performed bulk RNA-seq and found dynamic changes in Hippo-signaling components during alveologenesis. We deleted the Hippo effectors Yap/Taz from Acta2-expressing cells at the onset of alveologenesis, causing a significant arrest in alveolar development. Using single cell RNA-seq, we identified a distinct cluster of cells in mutant lungs with altered expression of marker genes associated with proximal mesenchymal cell types, airway smooth muscle and alveolar duct myofibroblasts. In vitro studies confirmed that Yap/Taz regulates myofibroblast-associated gene signature and contractility. Together, our findings show that Yap/Taz is essential for maintaining functional myofibroblast identity during postnatal alveologenesis.
Collapse
Affiliation(s)
- Fatima N. Chaudhry
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nigel S. Michki
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Dain L. Shirmer
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sharon McGrath-Morrow
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lisa R. Young
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David B. Frank
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jarod A. Zepp
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
34
|
Ohnishi Y, Masui A, Suezawa T, Mikawa R, Hirai T, Hagiwara M, Gotoh S. Screening of factors inducing alveolar type 1 epithelial cells using human pluripotent stem cells. Stem Cell Reports 2024; 19:529-544. [PMID: 38552636 PMCID: PMC11096435 DOI: 10.1016/j.stemcr.2024.02.009] [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: 09/03/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/12/2024] Open
Abstract
Alveolar type 2 (AT2) epithelial cells are tissue stem cells capable of differentiating into alveolar type 1 (AT1) cells for injury repair and maintenance of lung homeostasis. However, the factors involved in human AT2-to-AT1 cell differentiation are not fully understood. Here, we established SFTPCGFP and AGERmCherry-HiBiT dual-reporter induced pluripotent stem cells (iPSCs), which detected AT2-to-AT1 cell differentiation with high sensitivity and identified factors inducing AT1 cell differentiation from AT2 and their progenitor cells. We also established an "on-gel" alveolar epithelial spheroid culture suitable for medium-throughput screening. Among the 274 chemical compounds, several single compounds, including LATS-IN-1, converted AT1 cells from AT2 and their progenitor cells. Moreover, YAP/TAZ signaling activation and AKT signaling suppression synergistically recapitulated the induction of transcriptomic, morphological, and functionally mature AT1 cells. Our findings provide novel insights into human lung development and lung regenerative medicine.
Collapse
Affiliation(s)
- Yuko Ohnishi
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Atsushi Masui
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Takahiro Suezawa
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ryuta Mikawa
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Shimpei Gotoh
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
35
|
Basil MC, Alysandratos KD, Kotton DN, Morrisey EE. Lung repair and regeneration: Advanced models and insights into human disease. Cell Stem Cell 2024; 31:439-454. [PMID: 38492572 PMCID: PMC11070171 DOI: 10.1016/j.stem.2024.02.009] [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: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
36
|
Zheng S, Ye L. Hemodynamic Melody of Postnatal Cardiac and Pulmonary Development in Children with Congenital Heart Diseases. BIOLOGY 2024; 13:234. [PMID: 38666846 PMCID: PMC11048247 DOI: 10.3390/biology13040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Hemodynamics is the eternal theme of the circulatory system. Abnormal hemodynamics and cardiac and pulmonary development intertwine to form the most important features of children with congenital heart diseases (CHDs), thus determining these children's long-term quality of life. Here, we review the varieties of hemodynamic abnormalities that exist in children with CHDs, the recently developed neonatal rodent models of CHDs, and the inspirations these models have brought us in the areas of cardiomyocyte proliferation and maturation, as well as in alveolar development. Furthermore, current limitations, future directions, and clinical decision making based on these inspirations are highlighted. Understanding how CHD-associated hemodynamic scenarios shape postnatal heart and lung development may provide a novel path to improving the long-term quality of life of children with CHDs, transplantation of stem cell-derived cardiomyocytes, and cardiac regeneration.
Collapse
Affiliation(s)
- Sixie Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| |
Collapse
|
37
|
Chandran RR, Adams TS, Kabir I, Gallardo-Vara E, Kaminski N, Gomperts BN, Greif DM. Dedifferentiated early postnatal lung myofibroblasts redifferentiate in adult disease. Front Cell Dev Biol 2024; 12:1335061. [PMID: 38572485 PMCID: PMC10987733 DOI: 10.3389/fcell.2024.1335061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Alveolarization ensures sufficient lung surface area for gas exchange, and during bulk alveolarization in mice (postnatal day [P] 4.5-14.5), alpha-smooth muscle actin (SMA)+ myofibroblasts accumulate, secrete elastin, and lay down alveolar septum. Herein, we delineate the dynamics of the lineage of early postnatal SMA+ myofibroblasts during and after bulk alveolarization and in response to lung injury. SMA+ lung myofibroblasts first appear at ∼ P2.5 and proliferate robustly. Lineage tracing shows that, at P14.5 and over the next few days, the vast majority of SMA+ myofibroblasts downregulate smooth muscle cell markers and undergo apoptosis. Of note, ∼8% of these dedifferentiated cells and another ∼1% of SMA+ myofibroblasts persist to adulthood. Single cell RNA sequencing analysis of the persistent SMA- cells and SMA+ myofibroblasts in the adult lung reveals distinct gene expression profiles. For instance, dedifferentiated SMA- cells exhibit higher levels of tissue remodeling genes. Most interestingly, these dedifferentiated early postnatal myofibroblasts re-express SMA upon exposure of the adult lung to hypoxia or the pro-fibrotic drug bleomycin. However, unlike during alveolarization, these cells that re-express SMA do not proliferate with hypoxia. In sum, dedifferentiated early postnatal myofibroblasts are a previously undescribed cell type in the adult lung and redifferentiate in response to injury.
Collapse
Affiliation(s)
- Rachana R. Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Inamul Kabir
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| | - Eunate Gallardo-Vara
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Brigitte N. Gomperts
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Genetics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
38
|
Li R. Disrupted TGF-β signaling: a link between bronchopulmonary dysplasia and alveolar type 1 cells. J Clin Invest 2024; 134:e178562. [PMID: 38488005 PMCID: PMC10940082 DOI: 10.1172/jci178562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease common in extreme preterm infants and is characterized by alveolar simplification. Current BPD research mainly focuses on alveolar type 2 (AT2) cells, myofibroblasts, and the endothelium. However, a notable gap exists in the involvement of AT1 cells, which constitute a majority of the alveolar surface area. In this issue of the JCI, Callaway and colleagues explored the role of TGF-β signaling in AT1 cells for managing the AT1-to-AT2 transition and its involvement in the integration of mechanical forces with the pulmonary matrisome during development. The findings implicate AT1 cells in the pathogenesis of BPD.
Collapse
|
39
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
40
|
Li D, Hong H, Li M, Xu X, Wang S, Xiao Y, Zheng S, Wang Z, Yan Y, Chen H, Zhou C, Zhang H, Sun Q, Ye L. A surgical mouse model of neonatal right ventricular outflow tract obstruction by pulmonary artery banding. J Heart Lung Transplant 2024; 43:496-507. [PMID: 37839791 DOI: 10.1016/j.healun.2023.10.009] [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: 06/08/2023] [Revised: 09/29/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUD Diseased animal models play an extremely important role in preclinical research. Lacking the corresponding animal models, many basic research studies cannot be carried out, and the conclusions obtained are incomplete or even incorrect. Right ventricular (RV) outflow tract (RVOT) obstruction leads to RV pressure overload (PO) and reduced pulmonary blood flow (RPF), which are 2 of the most important pathophysiological characteristics in pediatric cardiovascular diseases and seriously affect the survival rate and long-term quality of life of many children. Due to the lack of a neonatal mouse model for RVOT obstruction, it is largely unknown how RV PO and RPF regulate postnatal RV and pulmonary development. The aim of this study was to construct a neonatal RVOT obstruction mouse model. METHODS AND RESULTS Here, we first introduced a neonatal mouse model of RVOT obstruction by pulmonary artery banding (PAB) on postnatal day 1. PAB induced neonatal RVOT obstruction, RV PO, and RPF. Neonatal RV PO induced cardiomyocyte proliferation, and neonatal RPF induced pulmonary dysplasia, the 2 features that are not observed in adult RVOT obstruction. As a result, PAB neonates exhibited overall developmental dysplasia, a sign similar to that of children with RVOT obstruction. CONCLUSIONS Because many pediatric cardiovascular diseases are associated with RV PO and RPF, the introduction of a neonatal mouse model of RVOT obstruction may greatly enhance our understanding of these diseases and eventually improve or save the lives of many children.
Collapse
Affiliation(s)
- Debao Li
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Haifa Hong
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Minghui Li
- Department of Cardiovascular Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuxia Xu
- Department of Radiology, Huangpu Branch, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shoubao Wang
- Department of Plastic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Xiao
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sixie Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Yan
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Chen
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxia Zhou
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Zhang
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qi Sun
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
41
|
Chen W, Zhu Y, Liu R, Kong B, Xia N, Zhao Y, Sun L. Screening Therapeutic Effects of MSC-EVs to Acute Lung Injury Model on A Chip. Adv Healthc Mater 2024; 13:e2303123. [PMID: 38084928 DOI: 10.1002/adhm.202303123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/29/2023] [Indexed: 12/19/2023]
Abstract
Acute lung injury (ALI) is a lethal disease with high mortality rate, and its physiologically relevant models that could mimic human disease processes are urgently needed to study pathophysiology and predict drug efficacy. Here, this work presents a novel lipopolysaccharide (LPS) based ALI model on a microfluidic chip that reconstitutes an air-liquid interface lined by human alveolar epithelium and microvascular endothelium for screening the therapeutic effects of mesenchymal stem cells (MSC) derived extracellular vesicles (MSC-EVs) to the biomimetic ALI. The air-liquid interface is established by coculture of alveolar epithelium and microvascular endothelium on the opposite sides of the porous membrane. The functionalized architecture is characterized by integrate cell layers and suitable permeability. Using this biomimetic microsystem, LPS based ALI model is established, which exhibits the disrupted alveolar-capillary barrier, reduced transepithelial/transendothelial electrical resistance (TEER), and impaired expression of junction proteins. As a reliable disease model, this work examines the effects of MSC-EVs, and the data indicate the therapeutic potential of EVs for severe ALI. MSC-EVs can alleviate barrier disruption by restoring both the epithelial and endothelial barrier integrity. They hope this study can become a unique approach to study human pathophysiology of ALI and advance drug development.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Nan Xia
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, China
| |
Collapse
|
42
|
Jones DL, Morley MP, Li X, Ying Y, Cardenas-Diaz FL, Li S, Zhou S, Schaefer SE, Chembazhi UV, Nottingham A, Lin S, Cantu E, Diamond JM, Basil MC, Vaughan AE, Morrisey EE. An injury-induced tissue niche shaped by mesenchymal plasticity coordinates the regenerative and disease response in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582147. [PMID: 38529490 PMCID: PMC10962740 DOI: 10.1101/2024.02.26.582147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Severe lung injury causes basal stem cells to migrate and outcompete alveolar stem cells resulting in dysplastic repair and a loss of gas exchange function. This "stem cell collision" is part of a multistep process that is now revealed to generate an injury-induced tissue niche (iTCH) containing Keratin 5+ epithelial cells and plastic Pdgfra+ mesenchymal cells. Temporal and spatial single cell analysis reveals that iTCHs are governed by mesenchymal proliferation and Notch signaling, which suppresses Wnt and Fgf signaling in iTCHs. Conversely, loss of Notch in iTCHs rewires alveolar signaling patterns to promote euplastic regeneration and gas exchange. The signaling patterns of iTCHs can differentially phenotype fibrotic from degenerative human lung diseases, through apposing flows of FGF and WNT signaling. These data reveal the emergence of an injury and disease associated iTCH in the lung and the ability of using iTCH specific signaling patterns 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 Biomedical Sciences, School of Veterinary 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
| | - 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
| | - 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
| | - 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
| | - 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 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 Medicine, 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
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
43
|
Gaddis N, Fortriede J, Guo M, Bardes EE, Kouril M, Tabar S, Burns K, Ardini-Poleske ME, Loos S, Schnell D, Jin K, Iyer B, Du Y, Huo BX, Bhattacharjee A, Korte J, Munshi R, Smith V, Herbst A, Kitzmiller JA, Clair GC, Carson JP, Adkins J, Morrisey EE, Pryhuber GS, Misra R, Whitsett JA, Sun X, Heathorn T, Paten B, Prasath VBS, Xu Y, Tickle T, Aronow BJ, Salomonis N. LungMAP Portal Ecosystem: Systems-level Exploration of the Lung. Am J Respir Cell Mol Biol 2024; 70:129-139. [PMID: 36413377 PMCID: PMC10848697 DOI: 10.1165/rcmb.2022-0165oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022] Open
Abstract
An improved understanding of the human lung necessitates advanced systems models informed by an ever-increasing repertoire of molecular omics, cellular imaging, and pathological datasets. To centralize and standardize information across broad lung research efforts, we expanded the LungMAP.net website into a new gateway portal. This portal connects a broad spectrum of research networks, bulk and single-cell multiomics data, and a diverse collection of image data that span mammalian lung development and disease. The data are standardized across species and technologies using harmonized data and metadata models that leverage recent advances, including those from the Human Cell Atlas, diverse ontologies, and the LungMAP CellCards initiative. To cultivate future discoveries, we have aggregated a diverse collection of single-cell atlases for multiple species (human, rhesus, and mouse) to enable consistent queries across technologies, cohorts, age, disease, and drug treatment. These atlases are provided as independent and integrated queryable datasets, with an emphasis on dynamic visualization, figure generation, reanalysis, cell-type curation, and automated reference-based classification of user-provided single-cell genomics datasets (Azimuth). As this resource grows, we intend to increase the breadth of available interactive interfaces, supported data types, data portals and datasets from LungMAP, and external research efforts.
Collapse
Affiliation(s)
- Nathan Gaddis
- RTI International, Research Triangle Park, North Carolina
| | - Joshua Fortriede
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Minzhe Guo
- Division of Pulmonary Biology, The Perinatal Institute, and
| | - Eric E. Bardes
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Michal Kouril
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Scott Tabar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Kevin Burns
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | | | - Stephanie Loos
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Daniel Schnell
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Kang Jin
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Balaji Iyer
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, Ohio
| | - Yina Du
- Division of Pulmonary Biology, The Perinatal Institute, and
| | - Bing-Xing Huo
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Anukana Bhattacharjee
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Jeff Korte
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Ruchi Munshi
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Victoria Smith
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Andrew Herbst
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | - Geremy C. Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington
| | - James P. Carson
- Texas Advanced Computing Center, University of Texas at Austin, Austin, Texas
| | - Joshua Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Edward E. Morrisey
- Department of Medicine and
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gloria S. Pryhuber
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Ravi Misra
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Jeffrey A. Whitsett
- Division of Pulmonary Biology, The Perinatal Institute, and
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Xin Sun
- Department of Pediatrics and
- Department of Biological Sciences, University of California, San Diego, San Diego, California; and
| | - Trevor Heathorn
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, California
| | - Benedict Paten
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, California
| | - V. B. Surya Prasath
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Yan Xu
- Division of Pulmonary Biology, The Perinatal Institute, and
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Tim Tickle
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Bruce J. Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
| |
Collapse
|
44
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
45
|
Callaway DA, Penkala IJ, Zhou S, Knowlton JJ, Cardenas-Diaz F, Babu A, Morley MP, Lopes M, Garcia BA, Morrisey EE. TGF-β controls alveolar type 1 epithelial cell plasticity and alveolar matrisome gene transcription in mice. J Clin Invest 2024; 134:e172095. [PMID: 38488000 PMCID: PMC10947970 DOI: 10.1172/jci172095] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/05/2024] [Indexed: 03/19/2024] Open
Abstract
Premature birth disrupts normal lung development and places infants at risk for bronchopulmonary dysplasia (BPD), a disease disrupting lung health throughout the life of an individual and that is increasing in incidence. The TGF-β superfamily has been implicated in BPD pathogenesis, however, what cell lineage it impacts remains unclear. We show that TGFbr2 is critical for alveolar epithelial (AT1) cell fate maintenance and function. Loss of TGFbr2 in AT1 cells during late lung development leads to AT1-AT2 cell reprogramming and altered pulmonary architecture, which persists into adulthood. Restriction of fetal lung stretch and associated AT1 cell spreading through a model of oligohydramnios enhances AT1-AT2 reprogramming. Transcriptomic and proteomic analyses reveal the necessity of TGFbr2 expression in AT1 cells for extracellular matrix production. Moreover, TGF-β signaling regulates integrin transcription to alter AT1 cell morphology, which further impacts ECM expression through changes in mechanotransduction. These data reveal the cell intrinsic necessity of TGF-β signaling in maintaining AT1 cell fate and reveal this cell lineage as a major orchestrator of the alveolar matrisome.
Collapse
Affiliation(s)
- Danielle A. Callaway
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute
| | - Ian J. Penkala
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
| | - Su Zhou
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan J. Knowlton
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute
| | - Fabian Cardenas-Diaz
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Apoorva Babu
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael P. Morley
- Penn-CHOP Lung Biology Institute
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mariana Lopes
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin A. Garcia
- Epigenetics Institute, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward E. Morrisey
- Penn-CHOP Lung Biology Institute
- Department of Cell and Developmental Biology, and
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
46
|
Yin Y, Koenitzer JR, Patra D, Dietmann S, Bayguinov P, Hagan AS, Ornitz DM. Identification of a myofibroblast differentiation program during neonatal lung development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.28.573370. [PMID: 38234814 PMCID: PMC10793446 DOI: 10.1101/2023.12.28.573370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFB) and a stable but poorly described population of lipid rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFB). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single cell RNA sequencing, and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a myofibroblast differentiation program that is distinct form other mesenchymal cells types and increases the known repertoire of mesenchymal cell types in the neonatal lung.
Collapse
Affiliation(s)
- Yongjun Yin
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - Debabrata Patra
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Sabine Dietmann
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine, St. Louis, MO 63110
| | - Peter Bayguinov
- Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew S. Hagan
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - David M. Ornitz
- Departments of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
47
|
He H, Bell SM, Davis AK, Zhao S, Sridharan A, Na CL, Guo M, Xu Y, Snowball J, Swarr DT, Zacharias WJ, Whitsett JA. PRDM3/16 Regulate Chromatin Accessibility Required for NKX2-1 Mediated Alveolar Epithelial Differentiation and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.570481. [PMID: 38187557 PMCID: PMC10769259 DOI: 10.1101/2023.12.20.570481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Differential chromatin accessibility accompanies and mediates transcriptional control of diverse cell fates and their differentiation during embryogenesis. While the critical role of NKX2-1 and its transcriptional targets in lung morphogenesis and pulmonary epithelial cell differentiation is increasingly known, mechanisms by which chromatin accessibility alters the epigenetic landscape and how NKX2-1 interacts with other co-activators required for alveolar epithelial cell differentiation and function are not well understood. Here, we demonstrate that the paired domain zinc finger transcriptional regulators PRDM3 and PRDM16 regulate chromatin accessibility to mediate cell differentiation decisions during lung morphogenesis. Combined deletion of Prdm3 and Prdm16 in early lung endoderm caused perinatal lethality due to respiratory failure from loss of AT2 cell function. Prdm3/16 deletion led to the accumulation of partially differentiated AT1 cells and loss of AT2 cells. Combination of single cell RNA-seq, bulk ATAC-seq, and CUT&RUN demonstrated that PRDM3 and PRDM16 enhanced chromatin accessibility at NKX2-1 transcriptional targets in peripheral epithelial cells, all three factors binding together at a multitude of cell-type specific cis-active DNA elements. Network analysis demonstrated that PRDM3/16 regulated genes critical for perinatal AT2 cell differentiation, surfactant homeostasis, and innate host defense. Lineage specific deletion of PRDM3/16 in AT2 cells led to lineage infidelity, with PRDM3/16 null cells acquiring partial AT1 fate. Together, these data demonstrate that NKX2-1-dependent regulation of alveolar epithelial cell differentiation is mediated by epigenomic modulation via PRDM3/16.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, West China Second University Hospital Sichuan University, Chengdu, Sichuan, 610041, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Sichuan 610041, China
| | - Sheila M. Bell
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Ashley Kuenzi Davis
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Shuyang Zhao
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Anusha Sridharan
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Cheng-Lun Na
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Minzhe Guo
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Yan Xu
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - John Snowball
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
| | - Daniel T. Swarr
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - William J. Zacharias
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| | - Jeffrey A. Whitsett
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center
- Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
48
|
Toth A, Kannan P, Snowball J, Kofron M, Wayman JA, Bridges JP, Miraldi ER, Swarr D, Zacharias WJ. Alveolar epithelial progenitor cells require Nkx2-1 to maintain progenitor-specific epigenomic state during lung homeostasis and regeneration. Nat Commun 2023; 14:8452. [PMID: 38114516 PMCID: PMC10775890 DOI: 10.1038/s41467-023-44184-0] [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: 09/22/2022] [Accepted: 12/04/2023] [Indexed: 12/21/2023] Open
Abstract
Lung epithelial regeneration after acute injury requires coordination cellular coordination to pattern the morphologically complex alveolar gas exchange surface. During adult lung regeneration, Wnt-responsive alveolar epithelial progenitor (AEP) cells, a subset of alveolar type 2 (AT2) cells, proliferate and transition to alveolar type 1 (AT1) cells. Here, we report a refined primary murine alveolar organoid, which recapitulates critical aspects of in vivo regeneration. Paired scRNAseq and scATACseq followed by transcriptional regulatory network (TRN) analysis identified two AT1 transition states driven by distinct regulatory networks controlled in part by differential activity of Nkx2-1. Genetic ablation of Nkx2-1 in AEP-derived organoids was sufficient to cause transition to a proliferative stressed Krt8+ state, and AEP-specific deletion of Nkx2-1 in adult mice led to rapid loss of progenitor state and uncontrolled growth of Krt8+ cells. Together, these data implicate dynamic epigenetic maintenance via Nkx2-1 as central to the control of facultative progenitor activity in AEPs.
Collapse
Affiliation(s)
- Andrea Toth
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paranthaman Kannan
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - John Snowball
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Bio-Imaging and Analysis Facility, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joseph A Wayman
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James P Bridges
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - Emily R Miraldi
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Daniel Swarr
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William J Zacharias
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
49
|
Ahmed DW, Eiken MK, DePalma SJ, Helms AS, Zemans RL, Spence JR, Baker BM, Loebel C. Integrating mechanical cues with engineered platforms to explore cardiopulmonary development and disease. iScience 2023; 26:108472. [PMID: 38077130 PMCID: PMC10698280 DOI: 10.1016/j.isci.2023.108472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024] Open
Abstract
Mechanical forces provide critical biological signals to cells during healthy and aberrant organ development as well as during disease processes in adults. Within the cardiopulmonary system, mechanical forces, such as shear, compressive, and tensile forces, act across various length scales, and dysregulated forces are often a leading cause of disease initiation and progression such as in bronchopulmonary dysplasia and cardiomyopathies. Engineered in vitro models have supported studies of mechanical forces in a number of tissue and disease-specific contexts, thus enabling new mechanistic insights into cardiopulmonary development and disease. This review first provides fundamental examples where mechanical forces operate at multiple length scales to ensure precise lung and heart function. Next, we survey recent engineering platforms and tools that have provided new means to probe and modulate mechanical forces across in vitro and in vivo settings. Finally, the potential for interdisciplinary collaborations to inform novel therapeutic approaches for a number of cardiopulmonary diseases are discussed.
Collapse
Affiliation(s)
- Donia W. Ahmed
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Madeline K. Eiken
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, Division of Pulmonary Sciences and Critical Care Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| |
Collapse
|
50
|
Sucre JM, McCall AS, Kropski JA. "Stuck in the Middle with You": intermediate cell states are not always in transition. J Clin Invest 2023; 133:e174633. [PMID: 37966115 PMCID: PMC10645374 DOI: 10.1172/jci174633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
The era of single-cell multiomics has led to the identification of lung epithelial cells with features of both alveolar type 1 (AT1) and alveolar type 2 (AT2) pneumocytes, leading many to infer that these cells are a distinct cell type in the process of transitioning between AT2 and AT1 cells. In this issue of the JCI, Wang and colleagues demonstrated that many so-called "transitional cells" do not actually contribute to functional repair. The findings warrant a reimagining of these cells as existing in a nondirectional, intermediate cell state, rather than moving through a transitory process from one cell type to another. We look forward to further exploration of diverse cell state expression profiles and a more refined examination of hallmark gene function beyond population labeling.
Collapse
Affiliation(s)
- Jennifer M.S. Sucre
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - A. Scott McCall
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jonathan A. Kropski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|