1
|
Ye D, Feng S, Yang X, Su Y, Zhang J, Feng H, Zhou M, Zhou B, Duan L, Peng T, Wang C. Hedgehog-interacting protein orchestrates alveologenesis and protects against bronchopulmonary dysplasia and emphysema. SCIENCE ADVANCES 2025; 11:eadu2958. [PMID: 40333979 PMCID: PMC12057671 DOI: 10.1126/sciadv.adu2958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025]
Abstract
Most of the lung's gas-exchange surface forms during alveologenesis and its disruption causes bronchopulmonary dysplasia (BPD) in infants, characterized by alveolar simplification and myofibroblast accumulation. BPD also increases the risk of adult emphysema, marked by alveolar loss. Despite this connection, mechanisms linking these conditions and effective treatments are still lacking. We identify hedgehog-interacting protein (HHIP), associated with both BPD and emphysema, as a critical regulator of alveologenesis. During this process, Hhip-expressing cells expanded, accompanied by hedgehog (Hh) signaling inhibition and myofibroblast transition. Stromal-specific Hhip deletion led to hyperactivation of Hh-IGF1 signaling axis, causing persistent SMA+ myofibroblasts and epithelial stem/progenitor cell senescence. Hyperactivation of this pathway was also observed in human BPD and hyperoxia-induced BPD models. Early Hhip deficiency resulted in adult emphysema with myofibroblast accumulation. We developed a therapeutic Fc-fused HHIP protein that mitigated BPD in neonatal mice and prevented adult emphysema. These findings establish HHIP as a critical regulator of alveologenesis and a therapeutic target for BPD and emphysema.
Collapse
Affiliation(s)
- Datian Ye
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Shiyun Feng
- Perfect Life Science Research Institute, Perfect (GuangDong) Co. Ltd., Zhongshan 528402, China
| | - Xinguo Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanjing Su
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Jing Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haixin Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Minqi Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bin Zhou
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lihui Duan
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tien Peng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chaoqun Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
2
|
Liu X, Lam SM, Zheng Y, Mo L, Li M, Sun T, Long X, Peng S, Zhang X, Mei M, Shui G, Bao S. Palmitoyl-carnitine Regulates Lung Development by Promoting Pulmonary Mesenchyme Proliferation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0620. [PMID: 40104443 PMCID: PMC11914330 DOI: 10.34133/research.0620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/20/2025] [Accepted: 01/29/2025] [Indexed: 03/20/2025]
Abstract
Disruption of acylcarnitine homeostasis results in life-threatening outcomes in humans. Carnitine-acylcarnitine translocase deficiency (CACTD) is a scarce autosomal recessive genetic disease and may result in patients' death due to heart arrest or respiratory insufficiency. However, the reasons and mechanism of CACTD inducing respiratory insufficiency have never been elucidated. Herein, we employed lipidomic techniques to create comprehensive lipidomic maps of entire lungs throughout both prenatal and postnatal developmental stages in mice. We found that the acylcarnitines manifested notable variations and coordinated the expression levels of carnitine-acylcarnitine translocase (Cact) across these lung developmental stages. Cact-null mice were all dead with a symptom of respiratory distress and exhibited failed lung development. Loss of Cact resulted in an accumulation of palmitoyl-carnitine (C16-acylcarnitine) in the lungs and promoted the proliferation of mesenchymal progenitor cells. Mesenchymal cells with elevated C16-acylcarnitine levels displayed minimal changes in energy metabolism but, upon investigation, revealed an interaction with sterile alpha motif domain and histidine-aspartate domain-containing protein 1 (Samhd1), leading to decreased protein abundance and enhanced cell proliferation. Thus, our findings present a mechanism addressing respiratory distress in CACTD, offering a valuable reference point for both the elucidation of pathogenesis and the exploration of treatment strategies for neonatal respiratory distress.
Collapse
Affiliation(s)
- Xing Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Respiratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Zheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lesong Mo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Muhan Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianyi Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohui Long
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shulin Peng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinwei Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mei Mei
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Shilai Bao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Rizzoli E, Fievez L, Fastrès A, Roels E, Marichal T, Clercx C. A single-cell RNA sequencing atlas of the healthy canine lung: a foundation for comparative studies. Front Immunol 2025; 16:1501603. [PMID: 40114924 PMCID: PMC11922831 DOI: 10.3389/fimmu.2025.1501603] [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: 09/25/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Single cell RNA sequencing (scRNA-seq) can be used to resolve the cellular and molecular heterogeneity within a tissue by identifying cell populations with an unprecedented granularity along with their transcriptional signatures. Yet, the single cell gene expression profiles of cell populations in the healthy canine lung tissue remain unexplored and such analysis could reveal novel cell populations or markers lacking in dogs and facilitate comparisons with lung diseases. Using fresh healthy lung biopsies from four dogs, we conducted droplet-based scRNA-seq on 26,278 cells. We characterized 46 transcriptionally distinct cell subpopulations across all lung tissue compartments including 23 immune, 13 mesenchymal, five epithelial and five endothelial cell subpopulations. Of note, we captured rare cells such as unconventional T cells or Schwann cells. Differential gene expression profiles identified specific markers across all cell subpopulations. Fibroblasts clusters exhibited a marked transcriptional heterogeneity, some of which might exert immune regulatory functions. Finally, the integration of canine lung cells with an annotated human lung atlas highlighted many similarities in gene expression profiles between species. This study thus provides an extensive molecular cell atlas of the healthy canine lung, expanding our knowledge of lung cell diversity in dogs, and providing the molecular foundation for investigating lung cell identities and functions in canine lung diseases. Besides, the occurrence of spontaneous lung diseases in pet dogs, with phenotypes closely resembling those in humans, may provide a relevant model for advancing research into human lung diseases.
Collapse
Affiliation(s)
- Elodie Rizzoli
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Laurence Fievez
- Department of Functional Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, University of Liège, Liège, Belgium
| | - Aline Fastrès
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Elodie Roels
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Thomas Marichal
- Department of Functional Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Cécile Clercx
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
4
|
Liu B, Sajiki Y, Littlefield N, Hu Y, Stuart WD, Sridharan A, Cui X, Siefert ME, Araki K, Ziady AG, Shi D, Whitsett JA, Maeda Y. PBAE-PEG-based lipid nanoparticles for lung cell-specific gene delivery. Mol Ther 2025; 33:1154-1165. [PMID: 39865654 PMCID: PMC11897763 DOI: 10.1016/j.ymthe.2025.01.037] [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: 07/24/2024] [Revised: 11/13/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
Exemplified by successful use in COVID-19 vaccination, delivery of modified mRNA encapsulated in lipid nanoparticles (LNPs) provides a framework for treating various genetic and acquired disorders. However, LNPs that can deliver mRNA into specific lung cell types have not yet been established. Here, we sought to determine whether poly(β-amino ester)s (PBAE) or PEGylated PBAE (PBAE-PEG) in combination with 4A3-SC8/DOPE/cholesterol/DOTAP LNPs could deliver mRNA into different types of lung cells in vivo. PBAE-PEG/LNP was similar to Lipofectamine MessengerMAX followed by PBAE/LNP for mRNA transfection efficiency in HEK293T cells in vitro. PBAE-PEG/LNP administered by intravenous (IV) injection achieved 73% mRNA transfection efficiency into lung endothelial cells, while PBAE-PEG/LNP administered by intratracheal (IT) injection achieved 55% efficiency in lung alveolar type II (ATII) epithelial cells in mice in vivo. PBAE/LNP administered by IT injection were superior for specific delivery into lung airway club epithelial cells compared to PBAE-PEG/LNP. Lipofectamine MessengerMAX was inactive in vivo. 5-Methoxyuridine-modified mRNA was more efficient than unmodified mRNA in vivo but not in vitro. Our findings indicate that PBAE-PEG/LNP and PBAE/LNP can transfect multiple lung cell types in vivo, which can be applied in gene therapy targeting genetic lung diseases.
Collapse
Affiliation(s)
- Bingxin Liu
- Perinatal Institute, Division of Neonatology, Perinatal, and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, University of Cincinnati College of Medicine, Cincinnati, OH 45221, USA
| | - Yamato Sajiki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nicole Littlefield
- School of Polymer Science and Polymer Engineering, University of Akron, Akron, OH 44325, USA
| | - Yongan Hu
- School of Polymer Science and Polymer Engineering, University of Akron, Akron, OH 44325, USA
| | - William D Stuart
- Perinatal Institute, Division of Neonatology, Perinatal, and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anusha Sridharan
- Perinatal Institute, Division of Neonatology, Perinatal, and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xuemei Cui
- Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, University of Cincinnati College of Medicine, Cincinnati, OH 45221, USA
| | - Matthew E Siefert
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Koichi Araki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Assem G Ziady
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Donglu Shi
- Materials Science and Engineering Program, Department of Mechanical and Materials Engineering, University of Cincinnati College of Medicine, Cincinnati, OH 45221, USA; Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jeffery A Whitsett
- Perinatal Institute, Division of Neonatology, Perinatal, and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| | - Yutaka Maeda
- Perinatal Institute, Division of Neonatology, Perinatal, and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
5
|
Atak D, Yıldız E, Özkan E, Yousefi M, Özkan A, Yılmaz AB, Kızılırmak AB, Alnajjar IA, Kanar Ç, Caan ZL, Zeybek ŞÜ, Küçükali Cİ, Tüzün E, Gürsoy‐Özdemir Y, Vural A. Longitudinal Investigation of Brain and Spinal Cord Pericytes After Inducible PDGFRβ + Cell Ablation in Adult Mice. J Neurochem 2025; 169:e70035. [PMID: 40066845 PMCID: PMC11894923 DOI: 10.1111/jnc.70035] [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: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
Central nervous system (CNS) pericytes play crucial roles in vascular development and blood-brain barrier maturation during prenatal development, as well as in regulating cerebral blood flow in adults. They have also been implicated in the pathogenesis of numerous neurological disorders. However, the behavior of pericytes in the adult brain after injury remains poorly understood, partly due to limitations in existing pericyte ablation models. To investigate pericyte responses following acute ablation and characterize a novel rodent model for pericyte research, we developed a tamoxifen-inducible PDGFRβ+ cell ablation model by crossing PDGFRβ-P2A-CreERT2 and Rosa26-DTA176 transgenic mouse lines. Using this model, we studied the effects of different tamoxifen doses and conducted histological examinations 15 and 60 days post-injection to assess the impacts of PDGFRβ+ cell ablation in both acute and chronic phases, respectively. Our results demonstrate that a low dose of tamoxifen effectively ablates PDGFRβ+ cells of the CNS in mice without reducing survival or causing significant systemic side effects, such as weight loss. Additionally, we found that the extent of PDGFRβ+ cell depletion varies between the cortex and the spinal cord, as well as between the gray and white matter regions of the spinal cord. Importantly, we observed that both pericyte coverage and numbers increased in the weeks following acute ablation, indicating the regenerative capacity of CNS pericytes in vivo. This study offers a valuable tool for future studies on the role of pericytes in neurological disorders by overcoming the limitations of constitutive pericyte ablation models and providing its longitudinal characterization in the CNS.
Collapse
Affiliation(s)
- Dila Atak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Erdost Yıldız
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Esra Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Mohammadreza Yousefi
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ayşe Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of PhysiologyBakırçay UniversityİzmirTürkiye
| | - Aysu Bilge Yılmaz
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ali Burak Kızılırmak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | | | - Çiçek Kanar
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Zeynep Lal Caan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Şakir Ümit Zeybek
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Cem İsmail Küçükali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Yasemin Gürsoy‐Özdemir
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Atay Vural
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Bridges JP, Vladar EK, Kurche JS, Krivoi A, Stancil IT, Dobrinskikh E, Hu Y, Sasse SK, Lee JS, Blumhagen RZ, Yang IV, Gerber AN, Peljto AL, Evans CM, Redente EF, Riches DW, Schwartz DA. Progressive lung fibrosis: reprogramming a genetically vulnerable bronchoalveolar epithelium. J Clin Invest 2025; 135:e183836. [PMID: 39744946 PMCID: PMC11684817 DOI: 10.1172/jci183836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is etiologically complex, with well-documented genetic and nongenetic origins. In this Review, we speculate that the development of IPF requires two hits: the first establishes a vulnerable bronchoalveolar epithelium, and the second triggers mechanisms that reprogram distal epithelia to initiate and perpetuate a profibrotic phenotype. While vulnerability of the bronchoalveolar epithelia is most often driven by common or rare genetic variants, subsequent injury of the bronchoalveolar epithelia results in persistent changes in cell biology that disrupt tissue homeostasis and activate fibroblasts. The dynamic biology of IPF can best be contextualized etiologically and temporally, including stages of vulnerability, early disease, and persistent and progressive lung fibrosis. These dimensions of IPF highlight critical mechanisms that adversely disrupt epithelial function, activate fibroblasts, and lead to lung remodeling. Together with better recognition of early disease, this conceptual approach should lead to the development of novel therapeutics directed at the etiologic and temporal drivers of lung fibrosis that will ultimately transform the care of patients with IPF from palliative to curative.
Collapse
Affiliation(s)
- James P. Bridges
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eszter K. Vladar
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jonathan S. Kurche
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Andrei Krivoi
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ian T. Stancil
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University, School of Medicine, Stanford, California, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Yan Hu
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Joyce S. Lee
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel Z. Blumhagen
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Ivana V. Yang
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Anna L. Peljto
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christopher M. Evans
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - David W.H. Riches
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David A. Schwartz
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veteran Affairs Medical Center, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
9
|
Khan IS, Molina C, Ren X, Auyeung VC, Cohen M, Tsukui T, Atakilit A, Sheppard D. Impaired myofibroblast proliferation is a central feature of pathologic post-natal alveolar simplification. eLife 2024; 13:RP94425. [PMID: 39660606 PMCID: PMC11634066 DOI: 10.7554/elife.94425] [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] [Indexed: 12/12/2024] Open
Abstract
Premature infants with bronchopulmonary dysplasia (BPD) have impaired alveolar gas exchange due to alveolar simplification and dysmorphic pulmonary vasculature. Advances in clinical care have improved survival for infants with BPD, but the overall incidence of BPD remains unchanged because we lack specific therapies to prevent this disease. Recent work has suggested a role for increased transforming growth factor-beta (TGFβ) signaling and myofibroblast populations in BPD pathogenesis, but the functional significance of each remains unclear. Here, we utilize multiple murine models of alveolar simplification and comparative single-cell RNA sequencing to identify shared mechanisms that could contribute to BPD pathogenesis. Single-cell RNA sequencing reveals a profound loss of myofibroblasts in two models of BPD and identifies gene expression signatures of increased TGFβ signaling, cell cycle arrest, and impaired proliferation in myofibroblasts. Using pharmacologic and genetic approaches, we find no evidence that increased TGFβ signaling in the lung mesenchyme contributes to alveolar simplification. In contrast, this is likely a failed compensatory response, since none of our approaches to inhibit TGFβ signaling protect mice from alveolar simplification due to hyperoxia while several make simplification worse. In contrast, we find that impaired myofibroblast proliferation is a central feature in several murine models of BPD, and we show that inhibiting myofibroblast proliferation is sufficient to cause pathologic alveolar simplification. Our results underscore the importance of impaired myofibroblast proliferation as a central feature of alveolar simplification and suggest that efforts to reverse this process could have therapeutic value in BPD.
Collapse
Affiliation(s)
- Imran S Khan
- Division of Neonatology, Department of Pediatrics, UCSFSan FranciscoUnited States
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
| | - Christopher Molina
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Xin Ren
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Vincent C Auyeung
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
| | - Max Cohen
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Tatsuya Tsukui
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Amha Atakilit
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| | - Dean Sheppard
- Cardiovascular Research Institute, UCSFSan FranciscoUnited States
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSFSan FranciscoUnited States
- Department of Medicine, UCSFSan FranciscoUnited States
| |
Collapse
|
10
|
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
|
11
|
Khan IS, Molina C, Ren X, Auyeung VC, Cohen M, Tsukui T, Atakilit A, Sheppard D. Impaired Myofibroblast Proliferation is a Central Feature of Pathologic Post-Natal Alveolar Simplification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572766. [PMID: 38187712 PMCID: PMC10769348 DOI: 10.1101/2023.12.21.572766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Premature infants with bronchopulmonary dysplasia (BPD) have impaired alveolar gas exchange due to alveolar simplification and dysmorphic pulmonary vasculature. Advances in clinical care have improved survival for infants with BPD, but the overall incidence of BPD remains unchanged because we lack specific therapies to prevent this disease. Recent work has suggested a role for increased transforming growth factor-beta (TGFβ) signaling and myofibroblast populations in BPD pathogenesis, but the functional significance of each remains unclear. Here, we utilize multiple murine models of alveolar simplification and comparative single-cell RNA sequencing to identify shared mechanisms that could contribute to BPD pathogenesis. Single-cell RNA sequencing reveals a profound loss of myofibroblasts in two models of BPD and identifies gene expression signatures of increased TGFβ signaling, cell cycle arrest, and impaired proliferation in myofibroblasts. Using pharmacologic and genetic approaches, we find no evidence that increased TGFβ signaling in the lung mesenchyme contributes to alveolar simplification. In contrast, this is likely a failed compensatory response, since none of our approaches to inhibit TGFb signaling protect mice from alveolar simplification due to hyperoxia while several make simplification worse. In contrast, we find that impaired myofibroblast proliferation is a central feature in several murine models of BPD, and we show that inhibiting myofibroblast proliferation is sufficient to cause pathologic alveolar simplification. Our results underscore the importance of impaired myofibroblast proliferation as a central feature of alveolar simplification and suggest that efforts to reverse this process could have therapeutic value in BPD.
Collapse
Affiliation(s)
- Imran S. Khan
- Division of Neonatology, Department of Pediatrics, UCSF
- Cardiovascular Research Institute, UCSF
| | - Christopher Molina
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Xin Ren
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Vincent C. Auyeung
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Max Cohen
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Tatsuya Tsukui
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Amha Atakilit
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| | - Dean Sheppard
- Cardiovascular Research Institute, UCSF
- Division of Pulmonary, Critical Care, Allergy, and Sleep, UCSF
- Department of Medicine, UCSF
| |
Collapse
|
12
|
Tsukui T, Wolters PJ, Sheppard D. Alveolar fibroblast lineage orchestrates lung inflammation and fibrosis. Nature 2024; 631:627-634. [PMID: 38987592 DOI: 10.1038/s41586-024-07660-1] [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: 08/02/2022] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
Fibroblasts are present throughout the body and function to maintain tissue homeostasis. Recent studies have identified diverse fibroblast subsets in healthy and injured tissues1,2, but the origins and functional roles of injury-induced fibroblast lineages remain unclear. Here we show that lung-specialized alveolar fibroblasts take on multiple molecular states with distinct roles in facilitating responses to fibrotic lung injury. We generate a genetic tool that uniquely targets alveolar fibroblasts to demonstrate their role in providing niches for alveolar stem cells in homeostasis and show that loss of this niche leads to exaggerated responses to acute lung injury. Lineage tracing identifies alveolar fibroblasts as the dominant origin for multiple emergent fibroblast subsets sequentially driven by inflammatory and pro-fibrotic signals after injury. We identify similar, but not completely identical, fibroblast lineages in human pulmonary fibrosis. TGFβ negatively regulates an inflammatory fibroblast subset that emerges early after injury and stimulates the differentiation into fibrotic fibroblasts to elicit intra-alveolar fibrosis. Blocking the induction of fibrotic fibroblasts in the alveolar fibroblast lineage abrogates fibrosis but exacerbates lung inflammation. These results demonstrate the multifaceted roles of the alveolar fibroblast lineage in maintaining normal alveolar homeostasis and orchestrating sequential responses to lung injury.
Collapse
Affiliation(s)
- Tatsuya Tsukui
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Dean Sheppard
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
Zanini F, Che X, Suresh NE, Knutsen C, Klavina P, Xie Y, Domingo-Gonzalez R, Liu M, Kum A, Jones RC, Quake SR, Alvira CM, Cornfield DN. Hyperoxia prevents the dynamic neonatal increases in lung mesenchymal cell diversity. Sci Rep 2024; 14:2033. [PMID: 38263350 PMCID: PMC10805790 DOI: 10.1038/s41598-023-50717-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 12/23/2023] [Indexed: 01/25/2024] Open
Abstract
Rapid expansion of the pulmonary microvasculature through angiogenesis drives alveolarization, the final stage of lung development that occurs postnatally and dramatically increases lung gas-exchange surface area. Disruption of pulmonary angiogenesis induces long-term structural and physiologic lung abnormalities, including bronchopulmonary dysplasia, a disease characterized by compromised alveolarization. Although endothelial cells are primary determinants of pulmonary angiogenesis, mesenchymal cells (MC) play a critical and dual role in angiogenesis and alveolarization. Therefore, we performed single cell transcriptomics and in-situ imaging of the developing lung to profile mesenchymal cells during alveolarization and in the context of lung injury. Specific mesenchymal cell subtypes were present at birth with increasing diversity during alveolarization even while expressing a distinct transcriptomic profile from more mature correlates. Hyperoxia arrested the transcriptomic progression of the MC, revealed differential cell subtype vulnerability with pericytes and myofibroblasts most affected, altered cell to cell communication, and led to the emergence of Acta1 expressing cells. These insights hold the promise of targeted treatment for neonatal lung disease, which remains a major cause of infant morbidity and mortality across the world.
Collapse
Affiliation(s)
- Fabio Zanini
- School of Clinical Medicine, University of New South Wales, Sydney, Australia.
- Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW, Australia.
- Evolution & Ecology Research Centre, University of New South Wales, Sydney, NSW, Australia.
| | - Xibing Che
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nina E Suresh
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Carsten Knutsen
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Paula Klavina
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Yike Xie
- School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Racquel Domingo-Gonzalez
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Min Liu
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Kum
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Jones
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Cristina M Alvira
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David N Cornfield
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
17
|
Hutchison V, Lynch A, Gutierrez-Gamez AM, Chen J. Inducible tricolor reporter mouse for parallel imaging of lysosomes, mitochondria, and microtubules. J Cell Biol 2024; 223:e202305086. [PMID: 37917008 PMCID: PMC10621751 DOI: 10.1083/jcb.202305086] [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: 05/22/2023] [Revised: 09/06/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Cell type-specific use of the same DNA blueprint generates diverse cell types. Such diversity must also be executed via differential deployment of the same subcellular machinery. However, our understanding of the size, distribution, and dynamics of subcellular machinery in native tissues and their connection to cellular diversity remains limited. We generate and characterize an inducible tricolor reporter mouse, dubbed "Kaleidoscope," for simultaneous imaging of lysosomes, mitochondria, and microtubules in any cell type and at a single-cell resolution. The expected subcellular compartments are labeled in culture and in tissues with no impact on cellular and organismal viability. Quantitative and live imaging of the tricolor reporter captures cell type-specific organelle features and kinetics in the lung, as well as their changes after Sendai virus infection. Yap/Taz mutant lung epithelial cells undergo accelerated lamellar body maturation, a subcellular manifestation of their molecular defects. A comprehensive toolbox of reporters for all subcellular structures is expected to transform our understanding of cell biology in tissues.
Collapse
Affiliation(s)
- Vera Hutchison
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Anne Lynch
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
18
|
Riccetti MR, Green J, Taylor TJ, Perl AKT. Prenatal FGFR2 Signaling via PI3K/AKT Specifies the PDGFRA + Myofibroblast. Am J Respir Cell Mol Biol 2024; 70:63-77. [PMID: 37734036 PMCID: PMC10768833 DOI: 10.1165/rcmb.2023-0245oc] [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: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
It is well known that FGFR2 (fibroblast growth factor receptor 2) signaling is critical for proper lung development. Recent studies demonstrate that epithelial FGFR2 signaling during the saccular phase of lung development (sacculation) regulates alveolar type 1 (AT1) and AT2 cell differentiation. During sacculation, PDGFRA (platelet-derived growth factor receptor-α)-positive lung fibroblasts exist as three functional subtypes: contractile myofibroblasts, extracellular matrix-producing matrix fibroblasts, and lipofibroblasts. All three subtypes are required during alveolarization to establish a niche that supports AT2 epithelial cell self-renewal and AT1 epithelial cell differentiation. FGFR2 signaling directs myofibroblast differentiation in PDGFRA+ fibroblasts during alveolar reseptation after pneumonectomy. However, it remains unknown if FGFR2 signaling regulates PDGFRA+ myo-, matrix, or lipofibroblast differentiation during sacculation. In this study, FGFR2 signaling was inhibited by temporal expression of a secreted dominant-negative FGFR2b (dnFGFR2) by AT2 cells from embryonic day (E) 16.5 to E18.5. Fibroblast and epithelial differentiation were analyzed at E18.5 and postnatal days 7 and 21. At all time points, the number of myofibroblasts was reduced and the number of lipo-/matrix fibroblasts was increased. AT2 cells are increased and AT1 cells are reduced postnatally, but not at E18.5. Similarly, in organoids made with PDGFRA+ fibroblasts from dnFGFR2 lungs, increased AT2 cells and reduced AT1 cells were observed. In vitro treatment of primary wild-type E16.5 adherent saccular lung fibroblasts with recombinant dnFGFR2b/c resulted in reduced myofibroblast contraction. Treatment with the PI3K/AKT activator 740 Y-P rescued the lack of myofibroblast differentiation caused by dnFGFR2b/2c. Moreover, treatment with the PI3K/AKT activator 740 Y-P rescued myofibroblast differentiation in E18.5 fibroblasts isolated from dnFGFR2 lungs.
Collapse
Affiliation(s)
- Matthew R. Riccetti
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Jenna Green
- Division of Neonatology and Pulmonary Biology and
| | - Thomas J. Taylor
- Division of Neonatology and Pulmonary Biology and
- College of Arts and Sciences, University of Cincinnati, Cincinnati, Ohio; and
| | - Anne-Karina T. Perl
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
19
|
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
|
20
|
Abstract
Advancing age is the most important risk factor for the development of and mortality from acute and chronic lung diseases, including pneumonia, chronic obstructive pulmonary disease, and pulmonary fibrosis. This risk was manifest during the COVID-19 pandemic, when elderly people were disproportionately affected and died from SARS-CoV-2 pneumonia. However, the recent pandemic also provided lessons on lung resilience. An overwhelming majority of patients with SARS-CoV-2 pneumonia, even those with severe disease, recovered with near-complete restoration of lung architecture and function. These observations are inconsistent with historic views of the lung as a terminally differentiated organ incapable of regeneration. Here, we review emerging hypotheses that explain how the lung repairs itself after injury and why these mechanisms of lung repair fail in some individuals, particularly the elderly.
Collapse
Affiliation(s)
- SeungHye Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- Cell and Developmental Biology, Northwestern University, Chicago, Illinois, USA
| | - Cara J. Gottardi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- Cell and Developmental Biology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
21
|
Ghonim MA, Boyd DF, Flerlage T, Thomas PG. Pulmonary inflammation and fibroblast immunoregulation: from bench to bedside. J Clin Invest 2023; 133:e170499. [PMID: 37655660 PMCID: PMC10471178 DOI: 10.1172/jci170499] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In recent years, there has been an explosion of interest in how fibroblasts initiate, sustain, and resolve inflammation across disease states. Fibroblasts contain heterogeneous subsets with diverse functionality. The phenotypes of these populations vary depending on their spatial distribution within the tissue and the immunopathologic cues contributing to disease progression. In addition to their roles in structurally supporting organs and remodeling tissue, fibroblasts mediate critical interactions with diverse immune cells. These interactions have important implications for defining mechanisms of disease and identifying potential therapeutic targets. Fibroblasts in the respiratory tract, in particular, determine the severity and outcome of numerous acute and chronic lung diseases, including asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and idiopathic pulmonary fibrosis. Here, we review recent studies defining the spatiotemporal identity of the lung-derived fibroblasts and the mechanisms by which these subsets regulate immune responses to insult exposures and highlight past, current, and future therapeutic targets with relevance to fibroblast biology in the context of acute and chronic human respiratory diseases. This perspective highlights the importance of tissue context in defining fibroblast-immune crosstalk and paves the way for identifying therapeutic approaches to benefit patients with acute and chronic pulmonary disorders.
Collapse
Affiliation(s)
- Mohamed A. Ghonim
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - David F. Boyd
- Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
22
|
Han S, Lee M, Shin Y, Giovanni R, Chakrabarty RP, Herrerias MM, Dada LA, Flozak AS, Reyfman PA, Khuder B, Reczek CR, Gao L, Lopéz-Barneo J, Gottardi CJ, Budinger GRS, Chandel NS. Mitochondrial integrated stress response controls lung epithelial cell fate. Nature 2023; 620:890-897. [PMID: 37558881 PMCID: PMC10447247 DOI: 10.1038/s41586-023-06423-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/11/2023] [Indexed: 08/11/2023]
Abstract
Alveolar epithelial type 1 (AT1) cells are necessary to transfer oxygen and carbon dioxide between the blood and air. Alveolar epithelial type 2 (AT2) cells serve as a partially committed stem cell population, producing AT1 cells during postnatal alveolar development and repair after influenza A and SARS-CoV-2 pneumonia1-6. Little is known about the metabolic regulation of the fate of lung epithelial cells. Here we report that deleting the mitochondrial electron transport chain complex I subunit Ndufs2 in lung epithelial cells during mouse gestation led to death during postnatal alveolar development. Affected mice displayed hypertrophic cells with AT2 and AT1 cell features, known as transitional cells. Mammalian mitochondrial complex I, comprising 45 subunits, regenerates NAD+ and pumps protons. Conditional expression of yeast NADH dehydrogenase (NDI1) protein that regenerates NAD+ without proton pumping7,8 was sufficient to correct abnormal alveolar development and avert lethality. Single-cell RNA sequencing revealed enrichment of integrated stress response (ISR) genes in transitional cells. Administering an ISR inhibitor9,10 or NAD+ precursor reduced ISR gene signatures in epithelial cells and partially rescued lethality in the absence of mitochondrial complex I function. Notably, lung epithelial-specific loss of mitochondrial electron transport chain complex II subunit Sdhd, which maintains NAD+ regeneration, did not trigger high ISR activation or lethality. These findings highlight an unanticipated requirement for mitochondrial complex I-dependent NAD+ regeneration in directing cell fate during postnatal alveolar development by preventing pathological ISR induction.
Collapse
Affiliation(s)
- SeungHye Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA.
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Goyang-si, Republic of Korea
| | - Youngjin Shin
- Department of Life Science, Dongguk University-Seoul, Goyang-si, Republic of Korea
| | - Regina Giovanni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Ram P Chakrabarty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Mariana M Herrerias
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Laura A Dada
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Annette S Flozak
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Paul A Reyfman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Basil Khuder
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Colleen R Reczek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - José Lopéz-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Cara J Gottardi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Navdeep S Chandel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA.
- Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
23
|
El Agha E, Thannickal VJ. The lung mesenchyme in development, regeneration, and fibrosis. J Clin Invest 2023; 133:e170498. [PMID: 37463440 DOI: 10.1172/jci170498] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Mesenchymal cells are uniquely located at the interface between the epithelial lining and the stroma, allowing them to act as a signaling hub among diverse cellular compartments of the lung. During embryonic and postnatal lung development, mesenchyme-derived signals instruct epithelial budding, branching morphogenesis, and subsequent structural and functional maturation. Later during adult life, the mesenchyme plays divergent roles wherein its balanced activation promotes epithelial repair after injury while its aberrant activation can lead to pathological remodeling and fibrosis that are associated with multiple chronic pulmonary diseases, including bronchopulmonary dysplasia, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this Review, we discuss the involvement of the lung mesenchyme in various morphogenic, neomorphogenic, and dysmorphogenic aspects of lung biology and health, with special emphasis on lung fibroblast subsets and smooth muscle cells, intercellular communication, and intrinsic mesenchymal mechanisms that drive such physiological and pathophysiological events throughout development, homeostasis, injury repair, regeneration, and aging.
Collapse
Affiliation(s)
- 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, Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| |
Collapse
|
24
|
Hutchison V, Lynch A, Gamez AMG, Chen J. An inducible tricolor reporter mouse for simultaneous imaging of lysosomes, mitochondria and microtubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541817. [PMID: 37293075 PMCID: PMC10245888 DOI: 10.1101/2023.05.22.541817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cell-type-specific use of the same DNA blueprint generates diverse cell types. Such diversity must also be executed via differential deployment of the same subcellular machinery. However, our understanding of the size, distribution, and dynamics of subcellular machinery in native tissues, and their connection to cellular diversity, remain limited. We generate and characterize an inducible tricolor reporter mouse, dubbed "kaleidoscope", for simultaneous imaging of lysosomes, mitochondria and microtubules in any cell type and at a single cell resolution. The expected subcellular compartments are labeled in culture and in tissues with no impact on cellular and organismal viability. Quantitative and live imaging of the tricolor reporter captures cell-type-specific organelle features and kinetics in the lung, as well as their changes after Sendai virus infection. Yap/Taz mutant lung epithelial cells undergo accelerated lamellar body maturation, a subcellular manifestation of their molecular defects. A comprehensive toolbox of reporters for all subcellular structures is expected to transform our understanding of cell biology in tissues.
Collapse
Affiliation(s)
- Vera Hutchison
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anne Lynch
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | - Jichao Chen
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
25
|
Yie TA, Loomis CA, Nowatzky J, Khodadadi-Jamayran A, Lin Z, Cammer M, Barnett C, Mezzano V, Alu M, Novick JA, Munger JS, Kugler MC. Hedgehog and Platelet-derived Growth Factor Signaling Intersect during Postnatal Lung Development. Am J Respir Cell Mol Biol 2023; 68:523-536. [PMID: 36693140 PMCID: PMC10174164 DOI: 10.1165/rcmb.2022-0269oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 01/24/2023] [Indexed: 01/26/2023] Open
Abstract
Normal lung development critically depends on HH (Hedgehog) and PDGF (platelet-derived growth factor) signaling, which coordinate mesenchymal differentiation and proliferation. PDGF signaling is required for postnatal alveolar septum formation by myofibroblasts. Recently, we demonstrated a requirement for HH in postnatal lung development involving alveolar myofibroblast differentiation. Given shared features of HH signaling and PDGF signaling and their impact on this key cell type, we sought to clarify their relationship during murine postnatal lung development. Timed experiments revealed that HH inhibition phenocopies the key lung myofibroblast phenotypes of Pdgfa (platelet-derived growth factor subunit A) and Pdgfra (platelet-derived growth factor receptor alpha) knockouts during secondary alveolar septation. Using a dual signaling reporter, Gli1lZ;PdgfraEGFP, we show that HH and PDGF pathway intermediates are concurrently expressed during alveolar septal myofibroblast accumulation, suggesting pathway convergence in the generation of lung myofibroblasts. Consistent with this hypothesis, HH inhibition reduces Pdgfra expression and diminishes the number of Pdgfra-positive and Pdgfra-lineage cells in postnatal lungs. Bulk RNA sequencing data of Pdgfra-expressing cells from Postnatal Day 8 (P8) lungs show that HH inhibition alters the expression not only of well-established HH targets but also of several putative PDGF target genes. This, together with the presence of Gli-binding sites in PDGF target genes, suggests HH input into PDGF signaling. We identified these HH/PDGF targets in several postnatal lung mesenchymal cell populations, including myofibroblasts, using single-cell transcriptomic analysis. Collectively, our data indicate that HH signaling and PDGF signaling intersect to support myofibroblast/fibroblast function during secondary alveolar septum formation. Moreover, they provide a molecular foundation relevant to perinatal lung diseases associated with impaired alveolarization.
Collapse
Affiliation(s)
- Ting-An Yie
- Division of Pulmonary, Critical Care and Sleep Medicine and
| | | | - Johannes Nowatzky
- Division of Rheumatology, Department of Medicine
- Department of Pathology
| | | | | | | | - Clea Barnett
- Division of Pulmonary, Critical Care and Sleep Medicine and
| | | | | | | | - John S. Munger
- Division of Pulmonary, Critical Care and Sleep Medicine and
- Department of Cell Biology, School of Medicine and Langone Medical Center, New York University, New York, New York
| | | |
Collapse
|
26
|
Ligresti G, Raslan AA, Hong J, Caporarello N, Confalonieri M, Huang SK. Mesenchymal cells in the Lung: Evolving concepts and their role in fibrosis. Gene 2023; 859:147142. [PMID: 36603696 PMCID: PMC10068350 DOI: 10.1016/j.gene.2022.147142] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023]
Abstract
Mesenchymal cells in the lung are crucial during development, but also contribute to the pathogenesis of fibrotic disorders, including idiopathic pulmonary fibrosis (IPF), the most common and deadly form of fibrotic interstitial lung diseases. Originally thought to behave as supporting cells for the lung epithelium and endothelium with a singular function of producing basement membrane, mesenchymal cells encompass a variety of cell types, including resident fibroblasts, lipofibroblasts, myofibroblasts, smooth muscle cells, and pericytes, which all occupy different anatomic locations and exhibit diverse homeostatic functions in the lung. During injury, each of these subtypes demonstrate remarkable plasticity and undergo varying capacity to proliferate and differentiate into activated myofibroblasts. Therefore, these cells secrete high levels of extracellular matrix (ECM) proteins and inflammatory cytokines, which contribute to tissue repair, or in pathologic situations, scarring and fibrosis. Whereas epithelial damage is considered the initial trigger that leads to lung injury, lung mesenchymal cells are recognized as the ultimate effector of fibrosis and attempts to better understand the different functions and actions of each mesenchymal cell subtype will lead to a better understanding of why fibrosis develops and how to better target it for future therapy. This review summarizes current findings related to various lung mesenchymal cells as well as signaling pathways, and their contribution to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Giovanni Ligresti
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US.
| | - Ahmed A Raslan
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Jeongmin Hong
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston MA, US
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, US
| | - Marco Confalonieri
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, US
| |
Collapse
|
27
|
Xia S, Vila Ellis L, Winkley K, Menden H, Mabry SM, Venkatraman A, Louiselle D, Gibson M, Grundberg E, Chen J, Sampath V. Neonatal hyperoxia induces activated pulmonary cellular states and sex-dependent transcriptomic changes in a model of experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L123-L140. [PMID: 36537711 PMCID: PMC9902224 DOI: 10.1152/ajplung.00252.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperoxia disrupts lung development in mice and causes bronchopulmonary dysplasia (BPD) in neonates. To investigate sex-dependent molecular and cellular programming involved in hyperoxia, we surveyed the mouse lung using single cell RNA sequencing (scRNA-seq), and validated our findings in human neonatal lung cells in vitro. Hyperoxia-induced inflammation in alveolar type (AT) 2 cells gave rise to damage-associated transient progenitors (DATPs). It also induced a new subpopulation of AT1 cells with reduced expression of growth factors normally secreted by AT1 cells, but increased mitochondrial gene expression. Female alveolar epithelial cells had less EMT and pulmonary fibrosis signaling in hyperoxia. In the endothelium, expansion of Car4+ EC (Cap2) was seen in hyperoxia along with an emergent subpopulation of Cap2 with repressed VEGF signaling. This regenerative response was increased in females exposed to hyperoxia. Mesenchymal cells had inflammatory signatures in hyperoxia, with a new distal interstitial fibroblast subcluster characterized by repressed lipid biosynthesis and a transcriptomic signature resembling myofibroblasts. Hyperoxia-induced gene expression signatures in human neonatal fibroblasts and alveolar epithelial cells in vitro resembled mouse scRNA-seq data. These findings suggest that neonatal exposure to hyperoxia programs distinct sex-specific stem cell progenitor and cellular reparative responses that underpin lung remodeling in BPD.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Konner Winkley
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Aparna Venkatraman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Daniel Louiselle
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Margaret Gibson
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Elin Grundberg
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| |
Collapse
|