51
|
Wang G, Wen B, Deng Z, Zhang Y, Kolesnichenko OA, Ustiyan V, Pradhan A, Kalin TV, Kalinichenko VV. Endothelial progenitor cells stimulate neonatal lung angiogenesis through FOXF1-mediated activation of BMP9/ACVRL1 signaling. Nat Commun 2022; 13:2080. [PMID: 35440116 PMCID: PMC9019054 DOI: 10.1038/s41467-022-29746-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/28/2022] [Indexed: 01/07/2023] Open
Abstract
Pulmonary endothelial progenitor cells (EPCs) are critical for neonatal lung angiogenesis and represent a subset of general capillary cells (gCAPs). Molecular mechanisms through which EPCs stimulate lung angiogenesis are unknown. Herein, we used single-cell RNA sequencing to identify the BMP9/ACVRL1/SMAD1 pathway signature in pulmonary EPCs. BMP9 receptor, ACVRL1, and its downstream target genes were inhibited in EPCs from Foxf1WT/S52F mutant mice, a model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Expression of ACVRL1 and its targets were reduced in lungs of ACDMPV subjects. Inhibition of FOXF1 transcription factor reduced BMP9/ACVRL1 signaling and decreased angiogenesis in vitro. FOXF1 synergized with ETS transcription factor FLI1 to activate ACVRL1 promoter. Nanoparticle-mediated silencing of ACVRL1 in newborn mice decreased neonatal lung angiogenesis and alveolarization. Treatment with BMP9 restored lung angiogenesis and alveolarization in ACVRL1-deficient and Foxf1WT/S52F mice. Altogether, EPCs promote neonatal lung angiogenesis and alveolarization through FOXF1-mediated activation of BMP9/ACVRL1 signaling.
Collapse
Affiliation(s)
- Guolun Wang
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bingqiang Wen
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zicheng Deng
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| | - Yufang Zhang
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Olena A Kolesnichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vladimir Ustiyan
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Arun Pradhan
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
52
|
Bai H, Si L, Jiang A, Belgur C, Zhai Y, Plebani R, Oh CY, Rodas M, Patil A, Nurani A, Gilpin SE, Powers RK, Goyal G, Prantil-Baun R, Ingber DE. Mechanical control of innate immune responses against viral infection revealed in a human lung alveolus chip. Nat Commun 2022; 13:1928. [PMID: 35396513 PMCID: PMC8993817 DOI: 10.1038/s41467-022-29562-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 03/23/2022] [Indexed: 12/24/2022] Open
Abstract
Mechanical breathing motions have a fundamental function in lung development and disease, but little is known about how they contribute to host innate immunity. Here we use a human lung alveolus chip that experiences cyclic breathing-like deformations to investigate whether physical forces influence innate immune responses to viral infection. Influenza H3N2 infection of mechanically active chips induces a cascade of host responses including increased lung permeability, apoptosis, cell regeneration, cytokines production, and recruitment of circulating immune cells. Comparison with static chips reveals that breathing motions suppress viral replication by activating protective innate immune responses in epithelial and endothelial cells, which are mediated in part through activation of the mechanosensitive ion channel TRPV4 and signaling via receptor for advanced glycation end products (RAGE). RAGE inhibitors suppress cytokines induction, while TRPV4 inhibition attenuates both inflammation and viral burden, in infected chips with breathing motions. Therefore, TRPV4 and RAGE may serve as new targets for therapeutic intervention in patients infected with influenza and other potential pandemic viruses that cause life-threatening lung inflammation.
Collapse
Affiliation(s)
- Haiqing Bai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Amanda Jiang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Chaitra Belgur
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Yunhao Zhai
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Roberto Plebani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, 66023, Italy
| | - Crystal Yuri Oh
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Melissa Rodas
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Aditya Patil
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Atiq Nurani
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Sarah E Gilpin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA.
| |
Collapse
|
53
|
Toth A, Steinmeyer S, Kannan P, Gray J, Jackson CM, Mukherjee S, Demmert M, Sheak JR, Benson D, Kitzmiller J, Wayman JA, Presicce P, Cates C, Rubin R, Chetal K, Du Y, Miao Y, Gu M, Guo M, Kalinichenko VV, Kallapur SG, Miraldi ER, Xu Y, Swarr D, Lewkowich I, Salomonis N, Miller L, Sucre JS, Whitsett JA, Chougnet CA, Jobe AH, Deshmukh H, Zacharias WJ. Inflammatory blockade prevents injury to the developing pulmonary gas exchange surface in preterm primates. Sci Transl Med 2022; 14:eabl8574. [PMID: 35353543 PMCID: PMC9082785 DOI: 10.1126/scitranslmed.abl8574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1β and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.
Collapse
Affiliation(s)
- Andrea Toth
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Shelby Steinmeyer
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Paranthaman Kannan
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Jerilyn Gray
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Department of Pediatrics, Division of Allergy and Immunology, University of Rochester, Rochester, NY USA
| | - Shibabrata Mukherjee
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Martin Demmert
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, Institute for Systemic Inflammation Research, University of Lϋbeck, Lϋbeck, Germany
| | - Joshua R. Sheak
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Daniel Benson
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Joseph Kitzmiller
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Joseph A. Wayman
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA USA
| | - Christopher Cates
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Rhea Rubin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Yina Du
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Yifei Miao
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Mingxia Gu
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Minzhe Guo
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Vladimir V. Kalinichenko
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA USA
| | - Emily R. Miraldi
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Yan Xu
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Daniel Swarr
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Ian Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Nathan Salomonis
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Lisa Miller
- California National Primate Research Center, University of California Davis, Davis, CA USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA USA
| | - Jennifer S. Sucre
- Division of Neonatology, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN USA
| | - Jeffrey A. Whitsett
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Alan H. Jobe
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Hitesh Deshmukh
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - William J. Zacharias
- Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
54
|
Leibel SL, Tseu I, Zhou A, Hodges A, Yin J, Bilodeau C, Goltsis O, Post M. Metabolomic profiling of human pluripotent stem cell differentiation into lung progenitors. iScience 2022; 25:103797. [PMID: 35198866 PMCID: PMC8850758 DOI: 10.1016/j.isci.2022.103797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/02/2021] [Accepted: 01/17/2022] [Indexed: 11/29/2022] Open
Abstract
Metabolism is vital to cellular function and tissue homeostasis during human lung development. In utero, embryonic pluripotent stem cells undergo endodermal differentiation toward a lung progenitor cell fate that can be mimicked in vitro using induced human pluripotent stem cells (hiPSCs) to study genetic mutations. To identify differences between wild-type and surfactant protein B (SFTPB)-deficient cell lines during endoderm specification toward lung, we used an untargeted metabolomics approach to evaluate the developmental changes in metabolites. We found that the metabolites most enriched during the differentiation from pluripotent stem cell to lung progenitor cell, regardless of cell line, were sphingomyelins and phosphatidylcholines, two important lipid classes in lung development. The SFTPB mutation had no metabolic impact on early endodermal lung development. The identified metabolite signatures during lung progenitor cell differentiation may be utilized as biomarkers for normal embryonic lung development.
Collapse
Affiliation(s)
- Sandra L Leibel
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92037, USA.,Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Irene Tseu
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Anson Zhou
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Andrew Hodges
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jun Yin
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Claudia Bilodeau
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Olivia Goltsis
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| |
Collapse
|
55
|
Xie T, Lynn H, Parks WC, Stripp B, Chen P, Jiang D, Noble PW. Abnormal respiratory progenitors in fibrotic lung injury. Stem Cell Res Ther 2022; 13:64. [PMID: 35130980 PMCID: PMC8822870 DOI: 10.1186/s13287-022-02737-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/18/2022] [Indexed: 12/19/2022] Open
Abstract
Recent advances in single-cell RNA sequencing (scRNA-seq) and epithelium lineage labeling have yielded identification of multiple abnormal epithelial progenitor populations during alveolar type 2 (ATII) cell differentiation into alveolar type 1 (ATI) cells during regenerative lung post-fibrotic injury. These abnormal cells include basaloid/basal-like cells, ATII transition cells, and persistent epithelial progenitors (PEPs). These cells occurred and accumulated during the regeneration of distal airway and alveoli in response to both chronic and acute pulmonary injury. Among the alveolar epithelial progenitors, PEPs express a distinct Krt8+ phenotype that is rarely found in intact alveoli. However, post-injury, the Krt8+ phenotype is seen in dysplastic epithelial cells. Fully understanding the characteristics and functions of these newly found, injury-induced abnormal behavioral epithelial progenitors and the signaling pathways regulating their phenotype could potentially point the way to unique therapeutic targets for fibrosing lung diseases. This review summarizes recent advances in understanding these epithelial progenitors as they relate to uncovering regenerative mechanisms.
Collapse
Affiliation(s)
- Ting Xie
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Heather Lynn
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William C Parks
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barry Stripp
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peter Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dianhua Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul W Noble
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
56
|
Satora L, Gawlikowski T, Tański A, Formicki K. Quest for breathing: proliferation of alveolar type 1 cells. Histochem Cell Biol 2022; 157:393-401. [PMID: 35050380 PMCID: PMC9001204 DOI: 10.1007/s00418-022-02073-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/22/2022]
Abstract
There is much evidence that the vertebrate lung originated from a progenitor structure which was present in bony fish. However, critical basic elements for the evolution of breathing in tetrapods, such as the central rhythm generator sensitive to CO2/pH and the pulmonary surfactant, were present in the lungless primitive vertebrate. This suggests that the evolution of air breathing in all vertebrates may have evolved through exaptations. It appears that the capability for proliferation of alveolar type 1 (AT1) cells is the "critical factor" which rendered possible the most radical subsequent innovation-the possibility of air breathing. "Epithelial remodeling," which consists in proliferation of alveolar cells-the structural basis for gas diffusion-observed in the alimentary tract of the gut-breathing fishes (GBF) has great potential for application in biomedical research. Such a process probably led to the gradual evolutionary development of lungs in terrestrial vertebrates. Research on the cellular and molecular mechanisms controlling proliferation of squamous epithelial cells in the GBF should contribute to explaining the regeneration-associated phenomena that occur in mammal lungs, and especially to the understanding of signal pathways which govern the process.
Collapse
Affiliation(s)
- Leszek Satora
- Department of Hydrobiology, Ichthyology and Biotechnology of Reproduction, West Pomeranian University of Technology in Szczecin, ul. Kazimierza Królewicza 4, 71-550, Szczecin, Poland.
| | - Tomasz Gawlikowski
- Department of Pharmacology, Clinical Pharmacology and Clinical Toxicology, Andrzej Frycz Modrzewski Krakow University, ul. G. Herlinga-Grudzińskiego 1, 30-705, Kraków, Poland
| | - Adam Tański
- Department of Hydrobiology, Ichthyology and Biotechnology of Reproduction, West Pomeranian University of Technology in Szczecin, ul. Kazimierza Królewicza 4, 71-550, Szczecin, Poland
| | - Krzysztof Formicki
- Department of Hydrobiology, Ichthyology and Biotechnology of Reproduction, West Pomeranian University of Technology in Szczecin, ul. Kazimierza Królewicza 4, 71-550, Szczecin, Poland
| |
Collapse
|
57
|
Sun X, Perl AK, Li R, Bell SM, Sajti E, Kalinichenko VV, Kalin TV, Misra RS, Deshmukh H, Clair G, Kyle J, Crotty Alexander LE, Masso-Silva JA, Kitzmiller JA, Wikenheiser-Brokamp KA, Deutsch G, Guo M, Du Y, Morley MP, Valdez MJ, Yu HV, Jin K, Bardes EE, Zepp JA, Neithamer T, Basil MC, Zacharias WJ, Verheyden J, Young R, Bandyopadhyay G, Lin S, Ansong C, Adkins J, Salomonis N, Aronow BJ, Xu Y, Pryhuber G, Whitsett J, Morrisey EE. A census of the lung: CellCards from LungMAP. Dev Cell 2022; 57:112-145.e2. [PMID: 34936882 PMCID: PMC9202574 DOI: 10.1016/j.devcel.2021.11.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023]
Abstract
The human lung plays vital roles in respiration, host defense, and basic physiology. Recent technological advancements such as single-cell RNA sequencing and genetic lineage tracing have revealed novel cell types and enriched functional properties of existing cell types in lung. The time has come to take a new census. Initiated by members of the NHLBI-funded LungMAP Consortium and aided by experts in the lung biology community, we synthesized current data into a comprehensive and practical cellular census of the lung. Identities of cell types in the normal lung are captured in individual cell cards with delineation of function, markers, developmental lineages, heterogeneity, regenerative potential, disease links, and key experimental tools. This publication will serve as the starting point of a live, up-to-date guide for lung research at https://www.lungmap.net/cell-cards/. We hope that Lung CellCards will promote the community-wide effort to establish, maintain, and restore respiratory health.
Collapse
Affiliation(s)
- Xin Sun
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Anne-Karina Perl
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Rongbo Li
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sheila M Bell
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Eniko Sajti
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Tanya V Kalin
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Ravi S Misra
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hitesh Deshmukh
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jennifer Kyle
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Laura E Crotty Alexander
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jorge A Masso-Silva
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph A Kitzmiller
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gail Deutsch
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratories, Seattle Children's Hospital, OC.8.720, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA
| | - Minzhe Guo
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Yina Du
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Michael P Morley
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Valdez
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haoze V Yu
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kang Jin
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eric E Bardes
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jarod A Zepp
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Terren Neithamer
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William J Zacharias
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Internal Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Jamie Verheyden
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Randee Young
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Gautam Bandyopadhyay
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sara Lin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles Ansong
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bruce J Aronow
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yan Xu
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gloria Pryhuber
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeff Whitsett
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Edward E Morrisey
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
58
|
Jin R, Gao Q, Yin C, Zou M, Lu K, Liu W, Zhu Y, Zhang M, Cheng R. The CD146-HIF-1α axis regulates epithelial cell migration and alveolar maturation in a mouse model of bronchopulmonary dysplasia. J Transl Med 2022; 102:794-804. [PMID: 35306530 PMCID: PMC9309096 DOI: 10.1038/s41374-022-00773-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 11/09/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common challenge in preterm neonates. Retardation of alveolar development characterizes the pulmonary pathology in BPD. In the present study, we explored the roles of the CD146-HIF-1α axis in BPD. We demonstrated that the levels of reactive oxygen species (ROS) and soluble CD146 (sCD1146) were increased in the peripheral blood of preterm neonates with BPD. In alveolar epithelial cells, hyperoxia promoted the expression of HIF-1α and CD146, which reinforced each other. In a mouse model of BPD, by exposing pups to 65% hyperoxia, HIF-1α and CD146 were increased in the pulmonary tissues. Mechanistically, CD146 hindered the migration of alveolar epithelial cells; in contrast, movement was significantly enhanced in CD146-knockout alveolar epithelial cells. As expected, CD146-knockout ameliorated alveolarization and improved BPD disease severity. Taken together, our findings imply that the CD146-HIF-1α axis contributes to alveolarization and that CD146 may be a novel candidate in BPD therapy.
Collapse
Affiliation(s)
- Rui Jin
- grid.452511.6Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing, China ,Department of Neonatal Medical Center, Lianyungang Maternal and Child Health Hospital, Lianyungang, China
| | - Qianqian Gao
- grid.452511.6Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Chunyu Yin
- grid.452511.6Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Mengjia Zou
- grid.452511.6Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Keyu Lu
- grid.452511.6Department of Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Liu
- grid.89957.3a0000 0000 9255 8984Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yuting Zhu
- Department of Neonatology, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Mingshun Zhang
- Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China. .,NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China.
| | - Rui Cheng
- Department of Neonatal Medical Center, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
59
|
Khattar D, Fernandes S, Snowball J, Guo M, Gillen MC, Jain SS, Sinner D, Zacharias W, Swarr DT. PI3K signaling specifies proximal-distal fate by driving a developmental gene regulatory network in SOX9+ mouse lung progenitors. eLife 2022; 11:67954. [PMID: 35976093 PMCID: PMC9427112 DOI: 10.7554/elife.67954] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
The tips of the developing respiratory buds are home to important progenitor cells marked by the expression of SOX9 and ID2. Early in embryonic development (prior to E13.5), SOX9+progenitors are multipotent, generating both airway and alveolar epithelium, but are selective progenitors of alveolar epithelial cells later in development. Transcription factors, including Sox9, Etv5, Irx, Mycn, and Foxp1/2 interact in complex gene regulatory networks to control proliferation and differentiation of SOX9+progenitors. Molecular mechanisms by which these transcription factors and other signaling pathways control chromatin state to establish and maintain cell-type identity are not well-defined. Herein, we analyze paired gene expression (RNA-Seq) and chromatin accessibility (ATAC-Seq) data from SOX9+ epithelial progenitor cells (EPCs) during embryonic development in Mus musculus. Widespread changes in chromatin accessibility were observed between E11.5 and E16.5, particularly at distal cis-regulatory elements (e.g. enhancers). Gene regulatory network (GRN) inference identified a common SOX9+ progenitor GRN, implicating phosphoinositide 3-kinase (PI3K) signaling in the developmental regulation of SOX9+ progenitor cells. Consistent with this model, conditional ablation of PI3K signaling in the developing lung epithelium in mouse resulted in an expansion of the SOX9+ EPC population and impaired airway epithelial cell differentiation. These data demonstrate that PI3K signaling is required for epithelial patterning during lung organogenesis, and emphasize the combinatorial power of paired RNA and ATAC seq in defining regulatory networks in development.
Collapse
Affiliation(s)
- Divya Khattar
- Department of Pediatrics, University of CincinnatiCincinnatiUnited States
| | - Sharlene Fernandes
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States
| | - John Snowball
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States
| | - Minzhe Guo
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States
| | - Matthew C Gillen
- Department of Pediatrics, University of CincinnatiCincinnatiUnited States
| | - Suchi Singh Jain
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Wake Forest UniversityWinston-SalemUnited States
| | - Debora Sinner
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States,Department of Pediatrics, University of CincinnatiCincinnatiUnited States
| | - William Zacharias
- Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States,Department of Medicine, University of CincinnatiCincinnatiUnited States
| | - Daniel T Swarr
- Department of Pediatrics, University of CincinnatiCincinnatiUnited States,Perinatal Institute, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical CenterWinston-SalemUnited States,Department of Pediatrics, University of CincinnatiCincinnatiUnited States
| |
Collapse
|
60
|
Stanton AE, Goodwin K, Sundarakrishnan A, Jaslove JM, Gleghorn JP, Pavlovich AL, Nelson CM. Negative Transpulmonary Pressure Disrupts Airway Morphogenesis by Suppressing Fgf10. Front Cell Dev Biol 2021; 9:725785. [PMID: 34926440 PMCID: PMC8673560 DOI: 10.3389/fcell.2021.725785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Mechanical forces are increasingly recognized as important determinants of cell and tissue phenotype and also appear to play a critical role in organ development. During the fetal stages of lung morphogenesis, the pressure of the fluid within the lumen of the airways is higher than that within the chest cavity, resulting in a positive transpulmonary pressure. Several congenital defects decrease or reverse transpulmonary pressure across the developing airways and are associated with a reduced number of branches and a correspondingly underdeveloped lung that is insufficient for gas exchange after birth. The small size of the early pseudoglandular stage lung and its relative inaccessibility in utero have precluded experimental investigation of the effects of transpulmonary pressure on early branching morphogenesis. Here, we present a simple culture model to explore the effects of negative transpulmonary pressure on development of the embryonic airways. We found that negative transpulmonary pressure decreases branching, and that it does so in part by altering the expression of fibroblast growth factor 10 (Fgf10). The morphogenesis of lungs maintained under negative transpulmonary pressure can be rescued by supplementing the culture medium with exogenous FGF10. These data suggest that Fgf10 expression is regulated by mechanical stress in the developing airways. Understanding the mechanical signaling pathways that connect transpulmonary pressure to FGF10 can lead to the establishment of novel non-surgical approaches for ameliorating congenital lung defects.
Collapse
Affiliation(s)
- Alice E Stanton
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Katharine Goodwin
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Aswin Sundarakrishnan
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Jacob M Jaslove
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Jason P Gleghorn
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Amira L Pavlovich
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Celeste M Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, United States.,Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
61
|
Chen D, Sun J, Zhu J, Ding X, Lan T, Wang X, Wu W, Ou Z, Zhu L, Ding P, Wang H, Luo L, Xiang R, Wang X, Qiu J, Wang S, Li H, Chai C, Liang L, An F, Zhang L, Han L, Zhu Y, Wang F, Yuan Y, Wu W, Sun C, Lu H, Wu J, Sun X, Zhang S, Sahu SK, Liu P, Xia J, Zhang L, Chen H, Fang D, Zeng Y, Wu Y, Cui Z, He Q, Jiang S, Ma X, Feng W, Xu Y, Li F, Liu Z, Chen L, Chen F, Jin X, Qiu W, Wang T, Li Y, Xing X, Yang H, Xu Y, Hua Y, Liu Y, Liu H, Xu X. Single cell atlas for 11 non-model mammals, reptiles and birds. Nat Commun 2021; 12:7083. [PMID: 34873160 PMCID: PMC8648889 DOI: 10.1038/s41467-021-27162-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/18/2021] [Indexed: 01/08/2023] Open
Abstract
The availability of viral entry factors is a prerequisite for the cross-species transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Large-scale single-cell screening of animal cells could reveal the expression patterns of viral entry genes in different hosts. However, such exploration for SARS-CoV-2 remains limited. Here, we perform single-nucleus RNA sequencing for 11 non-model species, including pets (cat, dog, hamster, and lizard), livestock (goat and rabbit), poultry (duck and pigeon), and wildlife (pangolin, tiger, and deer), and investigated the co-expression of ACE2 and TMPRSS2. Furthermore, cross-species analysis of the lung cell atlas of the studied mammals, reptiles, and birds reveals core developmental programs, critical connectomes, and conserved regulatory circuits among these evolutionarily distant species. Overall, our work provides a compendium of gene expression profiles for non-model animals, which could be employed to identify potential SARS-CoV-2 target cells and putative zoonotic reservoirs.
Collapse
Affiliation(s)
| | - Jian Sun
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jiacheng Zhu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiangning Ding
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianming Lan
- BGI-Shenzhen, Shenzhen, 518083, China
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Xiran Wang
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | | | - Zhihua Ou
- BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Peiwen Ding
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haoyu Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lihua Luo
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rong Xiang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoling Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaying Qiu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyou Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haimeng Li
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaochao Chai
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Langchao Liang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuyu An
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou, 510520, China
| | - Le Zhang
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
| | - Lei Han
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
| | - Yixin Zhu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | | | - Wendi Wu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Chengcheng Sun
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haorong Lu
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120, China
- Shenzhen Key Laboratory of Environmental Microbial Genomics and Application, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jihong Wu
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | - Xinghuai Sun
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | - Shenghai Zhang
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | | | - Ping Liu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Jun Xia
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Lijing Zhang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haixia Chen
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yuying Zeng
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiquan Wu
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1868, USA
| | - Zehua Cui
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qian He
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | | | - Xiaoyan Ma
- Department of Biochemistry, University of Cambridge, Cambridge, CB21QW, UK
| | | | - Yan Xu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Fang Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lei Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tianjiao Wang
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yang Li
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiumei Xing
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083, China
- Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI-Shenzhen, Shenzhen, 518120, China
| | - Yanchun Xu
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
- College of Wildlife and Protected Areas, Northeast Forestry University, No. 26, Hexing Road, Xiangfang District, Harbin, 150040, China
| | - Yan Hua
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou, 510520, China.
| | - Yahong Liu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| | - Huan Liu
- BGI-Shenzhen, Shenzhen, 518083, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, 518083, China.
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, 518083, Shenzhen, China.
| |
Collapse
|
62
|
The NOTCH3 Downstream Target HEYL Is Required for Efficient Human Airway Basal Cell Differentiation. Cells 2021; 10:cells10113215. [PMID: 34831437 PMCID: PMC8620267 DOI: 10.3390/cells10113215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Basal cells (BCs) are stem/progenitor cells of the mucociliary airway epithelium, and their differentiation is orchestrated by the NOTCH signaling pathway. NOTCH3 receptor signaling regulates BC to club cell differentiation; however, the downstream responses that regulate this process are unknown. Overexpression of the active NOTCH3 intracellular domain (NICD3) in primary human bronchial epithelial cells (HBECs) on in vitro air–liquid interface culture promoted club cell differentiation. Bulk RNA-seq analysis identified 692 NICD3-responsive genes, including the classical NOTCH target HEYL, which increased in response to NICD3 and positively correlated with SCGB1A1 (club cell marker) expression. siRNA knockdown of HEYL decreased tight junction formation and cell proliferation. Further, HEYL knockdown reduced club, goblet and ciliated cell differentiation. In addition, we observed decreased expression of HEYL in HBECs from donors with chronic obstructive pulmonary disease (COPD) vs. normal donors which correlates with the impaired differentiation capacity of COPD cells. Finally, overexpression of HEYL in COPD HBECs promoted differentiation into club, goblet and ciliated cells, suggesting the impaired capacity of COPD cells to generate a normal airway epithelium is a reversible phenotype that can be regulated by HEYL. Overall, our data identify the NOTCH3 downstream target HEYL as a key regulator of airway epithelial differentiation.
Collapse
|
63
|
Vindin HJ, Oliver BG, Weiss AS. Elastin in healthy and diseased lung. Curr Opin Biotechnol 2021; 74:15-20. [PMID: 34781101 DOI: 10.1016/j.copbio.2021.10.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 01/05/2023]
Abstract
Elastic fibers are an essential part of the pulmonary extracellular matrix (ECM). Intact elastin is required for normal function and its damage contributes profoundly to the etiology and pathology of lung disease. This highlights the need for novel lung-specific imaging methodology that enables high-resolution 3D visualization of the ECM. We consider elastin's involvement in chronic respiratory disease and examine recent methods for imaging and modeling of the lung in the context of advances in lung tissue engineering for research and clinical application.
Collapse
Affiliation(s)
- Howard J Vindin
- Charles Perkins Centre, The University of Sydney, Sydney 2006, NSW, Australia; School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia; The Woolcock Institute, The University of Sydney, Sydney 2006, NSW, Australia
| | - Brian Gg Oliver
- The Woolcock Institute, The University of Sydney, Sydney 2006, NSW, Australia
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, Sydney 2006, NSW, Australia; School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia; Sydney Nano Institute, The University of Sydney, 2006 Sydney, NSW, Australia.
| |
Collapse
|
64
|
Börner K, Teichmann SA, Quardokus EM, Gee JC, Browne K, Osumi-Sutherland D, Herr BW, Bueckle A, Paul H, Haniffa M, Jardine L, Bernard A, Ding SL, Miller JA, Lin S, Halushka MK, Boppana A, Longacre TA, Hickey J, Lin Y, Valerius MT, He Y, Pryhuber G, Sun X, Jorgensen M, Radtke AJ, Wasserfall C, Ginty F, Ho J, Sunshine J, Beuschel RT, Brusko M, Lee S, Malhotra R, Jain S, Weber G. Anatomical structures, cell types and biomarkers of the Human Reference Atlas. Nat Cell Biol 2021; 23:1117-1128. [PMID: 34750582 PMCID: PMC10079270 DOI: 10.1038/s41556-021-00788-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 09/29/2021] [Indexed: 02/05/2023]
Abstract
The Human Reference Atlas (HRA) aims to map all of the cells of the human body to advance biomedical research and clinical practice. This Perspective presents collaborative work by members of 16 international consortia on two essential and interlinked parts of the HRA: (1) three-dimensional representations of anatomy that are linked to (2) tables that name and interlink major anatomical structures, cell types, plus biomarkers (ASCT+B). We discuss four examples that demonstrate the practical utility of the HRA.
Collapse
Affiliation(s)
- Katy Börner
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ellen M Quardokus
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - James C Gee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen Browne
- Department of Health and Human Services, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Osumi-Sutherland
- European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Bruce W Herr
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Andreas Bueckle
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Hrishikesh Paul
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | - Shin Lin
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Avinash Boppana
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Teri A Longacre
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - John Hickey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yiing Lin
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | - M Todd Valerius
- Harvard Institute of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yongqun He
- Department of Microbiology and Immunology, and Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Xin Sun
- Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Marda Jorgensen
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Andrea J Radtke
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Clive Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Fiona Ginty
- Biology and Applied Physics, General Electric Research, Niskayuna, NY, USA
| | - Jonhan Ho
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joel Sunshine
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rebecca T Beuschel
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Maigan Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Sujin Lee
- Division of Vascular Surgery and Endovascular Therapy, Massachusetts General Hospital, Boston, MA, USA
| | - Rajeev Malhotra
- Harvard Institute of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Vascular Surgery and Endovascular Therapy, Massachusetts General Hospital, Boston, MA, USA
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Griffin Weber
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
65
|
Liberti DC, Morrisey EE. Organoid models: assessing lung cell fate decisions and disease responses. Trends Mol Med 2021; 27:1159-1174. [PMID: 34674972 DOI: 10.1016/j.molmed.2021.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Organoids can be derived from various cell types in the lung, and they provide a reproducible and tractable model for understanding the complex signals driving cell fate decisions in a regenerative context. In this review, we provide a retrospective account of organoid methodologies and outline new opportunities for optimizing these methods to further explore emerging concepts in lung biology. Moreover, we examine the benefits of integrating organoid assays with in vivo modeling to explore how the various niches and compartments in the respiratory system respond to both acute and chronic lung disease. The strategic implementation and improvement of organoid techniques will provide exciting new opportunities to understand and identify new therapeutic approaches to ameliorate lung disease states.
Collapse
Affiliation(s)
- Derek C Liberti
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
66
|
Taglauer ES, Fernandez-Gonzalez A, Willis GR, Reis M, Yeung V, Liu X, Prince LS, Mitsialis SA, Kourembanas S. Antenatal Mesenchymal Stromal Cell Extracellular Vesicle Therapy Prevents Preeclamptic Lung Injury in Mice. Am J Respir Cell Mol Biol 2021; 66:86-95. [PMID: 34614384 DOI: 10.1165/rcmb.2021-0307oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In preeclamptic pregnancies, a variety of intrauterine alterations lead to abnormal placentation, release of inflammatory/antiangiogenic factors, and subsequent fetal growth restriction with significant potential to cause a primary insult to the developing fetal lung. Thus, modulation of the maternal intrauterine environment may be a key therapeutic avenue to prevent preeclampsia-associated developmental lung injury. A biologic therapy of interest are mesenchymal stromal cell-derived extracellular vesicles (MEx), which we have previously shown to ameliorate preeclamptic physiology through intrauterine immunomodulation. To evaluate the therapeutic potential of MEx to improve developmental lung injury in experimental preeclampsia. Using the heme oxygenase-1 null mouse (Hmox1-/-) model, preeclamptic pregnant dams were administered intravenous antenatal MEx treatment during each week of pregnancy followed by analysis of fetal and postnatal lung tissues, amniotic fluid protein profiles and lung explant/amniotic fluid co-cultures in comparison with control and untreated preeclamptic pregnancies. We first identified that a preeclamptic intrauterine environment had a significant adverse impact on fetal lung development including alterations in fetal lung developmental gene profiles in addition to postnatal alveolar and bronchial changes. Amniotic fluid proteomic analysis and fetal lung explant/amniotic fluid co-cultures further demonstrated that maternally administered MEx altered the expression of multiple inflammatory mediators in the preeclamptic intrauterine compartment resulting in normalization of fetal lung branching morphogenesis and developmental gene expression. Our evaluation of fetal and postnatal parameters overall suggests that antenatal MEx treatment may provide a highly valuable preventative therapeutic modality for amelioration of lung development in preeclamptic disease.
Collapse
Affiliation(s)
- Elizabeth S Taglauer
- Harvard Medical School, 1811, Boston Children's Hospital, Boston, Massachusetts, United States
| | | | - Gareth R Willis
- Children's Hospital Boston, 1862, Boston, Massachusetts, United States
| | - Monica Reis
- Boston Children's Hospital, Department of Medicine, Division of Newborn Medicine, Boston, Massachusetts, United States.,Harvard Medical School, 1811, Department of Pediatrics, Boston, Massachusetts, United States
| | - Vincent Yeung
- Children's Hospital Boston, 1862, Boston, Massachusetts, United States.,Harvard Medical School, 1811, Boston, Massachusetts, United States
| | - Xianlan Liu
- Boston Children's Hospital, Division of Newborn Medicine, Boston, Massachusetts, United States
| | - Lawrence S Prince
- Stanford University School of Medicine, 10624, Pediatrics, Stanford, California, United States.,Lucile Salter Packard Children's Hospital at Stanford, 24349, Palo Alto, California, United States
| | - S Alex Mitsialis
- Boston Children's Hospital, 1862, Pediatrics, Boston, Massachusetts, United States.,Harvard Medical School, 1811, Pediatics, Boston, Massachusetts, United States
| | - Stella Kourembanas
- Harvard Medical School, 1811, Boston Children's Hospital, Boston, Massachusetts, United States;
| |
Collapse
|
67
|
Gschwend J, Sherman SP, Ridder F, Feng X, Liang HE, Locksley RM, Becher B, Schneider C. Alveolar macrophages rely on GM-CSF from alveolar epithelial type 2 cells before and after birth. J Exp Med 2021; 218:e20210745. [PMID: 34431978 PMCID: PMC8404471 DOI: 10.1084/jem.20210745] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/27/2021] [Accepted: 08/04/2021] [Indexed: 12/31/2022] Open
Abstract
Programs defining tissue-resident macrophage identity depend on local environmental cues. For alveolar macrophages (AMs), these signals are provided by immune and nonimmune cells and include GM-CSF (CSF2). However, evidence to functionally link components of this intercellular cross talk remains scarce. We thus developed new transgenic mice to profile pulmonary GM-CSF expression, which we detected in both immune cells, including group 2 innate lymphoid cells and γδ T cells, as well as AT2s. AMs were unaffected by constitutive deletion of hematopoietic Csf2 and basophil depletion. Instead, AT2 lineage-specific constitutive and inducible Csf2 deletion revealed the nonredundant function of AT2-derived GM-CSF in instructing AM fate, establishing the postnatal AM compartment, and maintaining AMs in adult lungs. This AT2-AM relationship begins during embryogenesis, where nascent AT2s timely induce GM-CSF expression to support the proliferation and differentiation of fetal monocytes contemporaneously seeding the tissue, and persists into adulthood, when epithelial GM-CSF remains restricted to AT2s.
Collapse
Affiliation(s)
- Julia Gschwend
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Frederike Ridder
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Xiaogang Feng
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Hong-Erh Liang
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Richard M. Locksley
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Department of Microbiology & Immunology, University of California San Francisco, San Francisco, CA
- Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
68
|
Gokey JJ, Snowball J, Sridharan A, Sudha P, Kitzmiller JA, Xu Y, Whitsett JA. YAP regulates alveolar epithelial cell differentiation and AGER via NFIB/KLF5/NKX2-1. iScience 2021; 24:102967. [PMID: 34466790 PMCID: PMC8383002 DOI: 10.1016/j.isci.2021.102967] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/26/2021] [Accepted: 08/06/2021] [Indexed: 01/04/2023] Open
Abstract
Ventilation is dependent upon pulmonary alveoli lined by two major epithelial cell types, alveolar type-1 (AT1) and 2 (AT2) cells. AT1 cells mediate gas exchange while AT2 cells synthesize and secrete pulmonary surfactants and serve as progenitor cells which repair the alveoli. We developed transgenic mice in which YAP was activated or deleted to determine its roles in alveolar epithelial cell differentiation. Postnatal YAP activation increased epithelial cell proliferation, increased AT1 cell numbers, and caused indeterminate differentiation of subsets of alveolar cells expressing atypical genes normally restricted to airway epithelial cells. YAP deletion increased expression of genes associated with mature AT2 cells. YAP activation enhanced DNA accessibility in promoters of transcription factors and motif enrichment analysis predicted target genes associated with alveolar cell differentiation. YAP participated with KLF5, NFIB, and NKX2-1 to regulate AGER. YAP plays a central role in a transcriptional network that regulates alveolar epithelial differentiation.
Collapse
Affiliation(s)
- Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John Snowball
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Anusha Sridharan
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Parvathi Sudha
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joseph A. Kitzmiller
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
| | - Yan Xu
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jeffrey A. Whitsett
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Perinatal Institute, Cincinnati, OH 45229, USA
- The Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
69
|
Xu X, Nie Y, Wang W, Ullah I, Tung WT, Ma N, Lendlein A. Generation of 2.5D lung bud organoids from human induced pluripotent stem cells. Clin Hemorheol Microcirc 2021; 79:217-230. [PMID: 34487028 DOI: 10.3233/ch-219111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are a promising cell source to generate the patient-specific lung organoid given their superior differentiation potential. However, the current 3D cell culture approach is tedious and time-consuming with a low success rate and high batch-to-batch variability. Here, we explored the establishment of lung bud organoids by systematically adjusting the initial confluence levels and homogeneity of cell distribution. The efficiency of single cell seeding and clump seeding was compared. Instead of the traditional 3D culture, we established a 2.5D organoid culture to enable the direct monitoring of the internal structure via microscopy. It was found that the cell confluence and distribution prior to induction were two key parameters, which strongly affected hiPSC differentiation trajectories. Lung bud organoids with positive expression of NKX 2.1, in a single-cell seeding group with homogeneously distributed hiPSCs at 70% confluence (SC_70%_hom) or a clump seeding group with heterogeneously distributed cells at 90% confluence (CL_90%_het), can be observed as early as 9 days post induction. These results suggest that a successful lung bud organoid formation with single-cell seeding of hiPSCs requires a moderate confluence and homogeneous distribution of cells, while high confluence would be a prominent factor to promote the lung organoid formation when seeding hiPSCs as clumps. 2.5D organoids generated with defined culture conditions could become a simple, efficient, and valuable tool facilitating drug screening, disease modeling and personalized medicine.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Yan Nie
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Weiwei Wang
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Imran Ullah
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Wing Tai Tung
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Nan Ma
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Hereon, Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| |
Collapse
|
70
|
Affiliation(s)
- Christian Martin
- University Hospital Aachen, RWTH, Aachen, Institute of Pharmacology and Toxicology, Aachen, Germany;
| |
Collapse
|
71
|
Goldsteen PA, Yoseif C, Dolga AM, Gosens R. Human pluripotent stem cells for the modelling and treatment of respiratory diseases. Eur Respir Rev 2021; 30:30/161/210042. [PMID: 34348980 PMCID: PMC9488746 DOI: 10.1183/16000617.0042-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
Respiratory diseases are among the leading causes of morbidity and mortality worldwide, representing a major unmet medical need. New chemical entities rarely make it into the clinic to treat respiratory diseases, which is partially due to a lack of adequate predictive disease models and the limited availability of human lung tissues to model respiratory disease. Human pluripotent stem cells (hPSCs) may help fill this gap by serving as a scalable human in vitro model. In addition, human in vitro models of rare genetic mutations can be generated using hPSCs. hPSC-derived epithelial cells and organoids have already shown great potential for the understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments. These potentials can also be applied to other hPSC-derived lung cell types in the future. In this review, we will discuss how hPSCs have brought, and may continue to bring, major changes to the field of respiratory diseases by understanding the molecular mechanisms of the pathology and by finding efficient therapeutics. Human pluripotent stem cells may help to develop animal-free, fully human in vitro models to advance our understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments.https://bit.ly/3cahaqz
Collapse
Affiliation(s)
- Pien A Goldsteen
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands .,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Christina Yoseif
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
72
|
Wang G, Wen B, Ren X, Li E, Zhang Y, Guo M, Xu Y, Whitsett JA, Kalin TV, Kalinichenko VV. Generation of Pulmonary Endothelial Progenitor Cells for Cell-based Therapy Using Interspecies Mouse-Rat Chimeras. Am J Respir Crit Care Med 2021; 204:326-338. [PMID: 33705684 DOI: 10.1164/rccm.202003-0758oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although pulmonary endothelial progenitor cells (EPCs) hold promise for cell-based therapies for neonatal pulmonary disorders, whether EPCs can be derived from pluripotent embryonic stem cells (ESCs) or induced pluripotent stem cells remains unknown.Objectives: To investigate the heterogeneity of pulmonary EPCs and derive functional EPCs from pluripotent ESCs.Methods: Single-cell RNA sequencing of neonatal human and mouse lung was used to identify the heterogeneity of pulmonary EPCs. CRISPR/Cas9 gene editing was used to genetically label and purify mouse pulmonary EPCs. Functional properties of the EPCs were assessed after cell transplantation into neonatal mice with S52F Foxf1 mutation, a mouse model of alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Interspecies mouse-rat chimeras were produced through blastocyst complementation to generate EPCs from pluripotent ESCs for cell therapy in ACDMPV mice.Measurements and Main Results: We identified a unique population of EPCs, FOXF1+cKIT+ EPCs, as a subset of recently described general capillary cells (gCAPs) expressing SMAD7, ZBTB20, NFIA, and DLL4 but lacking mature arterial, venous, and lymphatic markers. FOXF1+cKIT+ gCAPs are reduced in ACDMPV, and their transcriptomic signature is conserved in mouse and human lungs. After cell transplantation into the neonatal circulation of ACDMPV mice, FOXF1+cKIT+ gCAPs engraft into the pulmonary vasculature, stimulate angiogenesis, improve oxygenation, and prevent alveolar simplification. FOXF1+cKIT+ gCAPs, produced from ESCs in interspecies chimeras, are fully competent to stimulate neonatal lung angiogenesis and alveolarization in ACDMPV mice.Conclusions: Cell-based therapy using donor or ESC/induced pluripotent stem cell-derived FOXF1+cKIT+ endothelial progenitors may be considered for treatment of human ACDMPV.
Collapse
Affiliation(s)
| | | | | | - Enhong Li
- Center for Lung Regenerative Medicine
| | | | | | - Yan Xu
- Division of Pulmonary Biology, and
| | | | | | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine.,Division of Pulmonary Biology, and.,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| |
Collapse
|
73
|
Karolak JA, Gambin T, Szafranski P, Maywald RL, Popek E, Heaney JD, Stankiewicz P. Perturbation of semaphorin and VEGF signaling in ACDMPV lungs due to FOXF1 deficiency. Respir Res 2021; 22:212. [PMID: 34315444 PMCID: PMC8314029 DOI: 10.1186/s12931-021-01797-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare lethal congenital lung disorder in neonates characterized by severe progressive respiratory failure and refractory pulmonary hypertension, resulting from underdevelopment of the peripheral pulmonary tree. Causative heterozygous single nucleotide variants (SNVs) or copy-number variant (CNV) deletions involving FOXF1 or its distant lung-specific enhancer on chromosome 16q24.1 have been identified in 80-90% of ACDMPV patients. FOXF1 maps closely to and regulates the oppositely oriented FENDRR, with which it also shares regulatory elements. METHODS To better understand the transcriptional networks downstream of FOXF1 that are relevant for lung organogenesis, using RNA-seq, we have examined lung transcriptomes in 12 histopathologically verified ACDMPV patients with or without pathogenic variants in the FOXF1 locus and analyzed gene expression profile in FENDRR-depleted fetal lung fibroblasts, IMR-90. RESULTS RNA-seq analyses in ACDMPV neonates revealed changes in the expression of several genes, including semaphorins (SEMAs), neuropilin 1 (NRP1), and plexins (PLXNs), essential for both epithelial branching and vascular patterning. In addition, we have found deregulation of the vascular endothelial growth factor (VEGF) signaling that also controls pulmonary vasculogenesis and a lung-specific endothelial gene TMEM100 known to be essential in vascular morphogenesis. Interestingly, we have observed a substantial difference in gene expression profiles between the ACDMPV samples with different types of FOXF1 defect. Moreover, partial overlap between transcriptome profiles of ACDMPV lungs with FOXF1 SNVs and FENDRR-depleted IMR-90 cells suggests contribution of FENDRR to ACDMPV etiology. CONCLUSIONS Our transcriptomic data imply potential crosstalk between several lung developmental pathways, including interactions between FOXF1-SHH and SEMA-NRP or VEGF/VEGFR2 signaling, and provide further insight into complexity of lung organogenesis in humans.
Collapse
Affiliation(s)
- Justyna A Karolak
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-781, Poznań, Poland
| | - Tomasz Gambin
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.,Institute of Computer Science, Warsaw University of Technology, 00-665, Warsaw, Poland
| | - Przemyslaw Szafranski
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Rebecca L Maywald
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Edwina Popek
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Rm ABBR-R809, Houston, TX, 77030, USA.
| |
Collapse
|
74
|
Kolesnichenko OA, Whitsett JA, Kalin TV, Kalinichenko VV. Therapeutic Potential of Endothelial Progenitor Cells in Pulmonary Diseases. Am J Respir Cell Mol Biol 2021; 65:473-488. [PMID: 34293272 DOI: 10.1165/rcmb.2021-0152tr] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Compromised alveolar development and pulmonary vascular remodeling are hallmarks of pediatric lung diseases such as bronchopulmonary dysplasia (BPD) and alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Although advances in surfactant therapy, corticosteroids, and anti-inflammatory drugs have improved clinical management of preterm infants, still those who suffer with severe vascular complications lack viable treatment options. Paucity of the alveolar capillary network in ACDMPV causes respiratory distress and leads to mortality in a vast majority of ACDMPV infants. The discovery of endothelial progenitor cells (EPCs) in 1997 brought forth the paradigm of postnatal vasculogenesis and hope for promoting vascularization in fragile patient populations, such as those with BPD and ACDMPV. The identification of diverse EPC populations, both hematopoietic and nonhematopoietic in origin, provided a need to identify progenitor cell selective markers which are linked to progenitor properties needed to develop cell-based therapies. Focusing to the future potential of EPCs for regenerative medicine, this review will discuss various aspects of EPC biology, beginning with the identification of hematopoietic, nonhematopoietic, and tissue-resident EPC populations. We will review knowledge related to cell surface markers, signature gene expression, key transcriptional regulators, and will explore the translational potential of EPCs for cell-based therapy for BPD and ACDMPV. The ability to produce pulmonary EPCs from patient-derived induced pluripotent stem cells (iPSCs) in vitro, holds promise for restoring vascular growth and function in the lungs of patients with pediatric pulmonary disorders.
Collapse
Affiliation(s)
- Olena A Kolesnichenko
- Cincinnati Children's Hospital Medical Center, 2518, Cincinnati, Ohio, United States
| | - Jeffrey A Whitsett
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Tanya V Kalin
- Cincinnati Children\'s Hospital Medical Center, 2518, Pediatrics, Cincinnati, Ohio, United States
| | - Vladimir V Kalinichenko
- Cincinnati Children's Hospital Medical Center, Pediatrics, Division of Pulmonary Biology, Cincinnati, Ohio, United States;
| |
Collapse
|
75
|
Fernandes-Silva H, Alves MG, Araújo-Silva H, Silva AM, Correia-Pinto J, Oliveira PF, Moura RS. Lung branching morphogenesis is accompanied by temporal metabolic changes towards a glycolytic preference. Cell Biosci 2021; 11:134. [PMID: 34274010 PMCID: PMC8285861 DOI: 10.1186/s13578-021-00654-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
Background Lung branching morphogenesis is characterized by epithelial-mesenchymal interactions that ultimately define the airway conducting system. Throughout this process, energy and structural macromolecules are necessary to sustain the high proliferative rates. The extensive knowledge of the molecular mechanisms underlying pulmonary development contrasts with the lack of data regarding the embryonic lung metabolic requirements. Here, we studied the metabolic profile associated with the early stages of chicken pulmonary branching. Methods In this study, we used an ex vivo lung explant culture system and analyzed the consumption/production of extracellular metabolic intermediates associated with glucose catabolism (alanine, lactate, and acetate) by 1H-NMR spectroscopy in the culture medium. Then, we characterized the transcript levels of metabolite membrane transporters (glut1, glut3, glut8, mct1, mct3, mct4, and mct8) and glycolytic enzymes (hk1, hk2, pfk1, ldha, ldhb, pdha, and pdhb) by qPCR. ldha and ldhb mRNA spatial localization was determined by in situ hybridization. Proliferation was analyzed by directly assessing DNA synthesis using an EdU-based assay. Additionally, we performed western blot to analyze LDHA and LDHT protein levels. Finally, we used a Clark-Type Electrode to assess the lung explant's respiratory capacity. Results Glucose consumption decreases, whereas alanine, lactate, and acetate production progressively increase as branching morphogenesis proceeds. mRNA analysis revealed variations in the expression levels of key enzymes and transporters from the glycolytic pathway. ldha and ldhb displayed a compartment-specific expression pattern that resembles proximal–distal markers. In addition, high proliferation levels were detected at active branching sites. LDH protein expression levels suggest that LDHB may account for the progressive rise in lactate. Concurrently, there is a stable oxygen consumption rate throughout branching morphogenesis. Conclusions This report describes the temporal metabolic changes that accompany the early stages of chicken lung branching morphogenesis. Overall, the embryonic chicken lung seems to shift to a glycolytic lactate-based metabolism as pulmonary branching occurs. Moreover, this metabolic rewiring might play a crucial role during lung development. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00654-w.
Collapse
Affiliation(s)
- Hugo Fernandes-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,PhDOC PhD Program, ICVS/3B's, School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Marco G Alves
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313, Porto, Portugal.,Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - Henrique Araújo-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Ana M Silva
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Department of Pediatric Surgery, Hospital of Braga, 4710-243, Braga, Portugal
| | - Pedro F Oliveira
- QOPNA &, LAQV, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rute S Moura
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| |
Collapse
|
76
|
Grimm SL, Dong X, Zhang Y, Carisey AF, Arnold AP, Moorthy B, Coarfa C, Lingappan K. Effect of sex chromosomes versus hormones in neonatal lung injury. JCI Insight 2021; 6:e146863. [PMID: 34061778 PMCID: PMC8410054 DOI: 10.1172/jci.insight.146863] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
The main mechanisms underlying sexually dimorphic outcomes in neonatal lung injury are unknown. We tested the hypothesis that hormone- or sex chromosome–mediated mechanisms interact with hyperoxia exposure to impact injury and repair in the neonatal lung. To distinguish sex differences caused by gonadal hormones versus sex chromosome complement (XX versus XY), we used the Four Core Genotypes (FCG) mice and exposed them to hyperoxia (95% FiO2, P1–P4: saccular stage) or room air. This model generates XX and XY mice that each have either testes (with Sry, XXM, or XYM) or ovaries (without Sry, XXF, or XYF). Lung alveolarization and vascular development were more severely impacted in XYM and XYF compared with XXF and XXM mice. Cell cycle–related pathways were enriched in the gonadal or chromosomal females, while muscle-related pathways were enriched in the gonadal males, and immune-response–related pathways were enriched in chromosomal males. Female gene signatures showed a negative correlation with human patients who developed bronchopulmonary dysplasia (BPD) or needed oxygen therapy at 28 days. These results demonstrate that chromosomal sex — and not gonadal sex — impacted the response to neonatal hyperoxia exposure. The female sex chromosomal complement was protective and could mediate sex-specific differences in the neonatal lung injury.
Collapse
Affiliation(s)
- Sandra L Grimm
- Molecular and Cellular Biology Department.,Center for Precision Environmental Health, and
| | - Xiaoyu Dong
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Yuhao Zhang
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Alexandre F Carisey
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Arthur P Arnold
- Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Molecular and Cellular Biology Department.,Center for Precision Environmental Health, and.,Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Krithika Lingappan
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
77
|
Li E, Ustiyan V, Wen B, Kalin GT, Whitsett JA, Kalin TV, Kalinichenko VV. Blastocyst complementation reveals that NKX2-1 establishes the proximal-peripheral boundary of the airway epithelium. Dev Dyn 2021; 250:1001-1020. [PMID: 33428297 DOI: 10.1002/dvdy.298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Distinct boundaries between the proximal conducting airways and more peripheral-bronchial regions of the lung are established early in foregut embryogenesis, demarcated in part by the distribution of SOX family and NKX2-1 transcription factors along the cephalo-caudal axis of the lung. We used blastocyst complementation to identify the role of NKX2-1 in the formation of the proximal-peripheral boundary of the airways in mouse chimeric embryos. RESULTS While Nkx2-1-/- mouse embryos form primordial tracheal cysts, peripheral pulmonary structures are entirely lacking in Nkx2-1-/- mice. Complementation of Nkx2-1-/- embryos with NKX2-1-sufficient embryonic stem cells (ESCs) enabled the formation of all tissue components of the peripheral lung but did not enhance ESC colonization of the most proximal regions of the airways. In chimeric mice, a precise boundary was formed between NKX2-1-deficient basal cells co-expressing SOX2 and SOX9 in large airways and ESC-derived NKX2-1+ SOX9+ epithelial cells of smaller airways. NKX2-1-sufficient ESCs were able to selectively complement peripheral, rather than most proximal regions of the airways. ESC complementation did not prevent ectopic expression of SOX9 but restored β-catenin signaling in Nkx2-1-/- basal cells of large airways. CONCLUSIONS NKX2-1 and β-catenin function in an epithelial cell-autonomous manner to establish the proximal-peripheral boundary along developing airways.
Collapse
Affiliation(s)
- Enhong Li
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
| | - Vladimir Ustiyan
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
| | - Bingqiang Wen
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
| | - Gregory T Kalin
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
78
|
Abstract
Bronchopulmonary dysplasia (BPD) is a form of chronic lung disease that occurs in preterm infants, usually those receiving substantial respiratory support with either mechanical ventilation or supplementation with oxygen. The pathogenesis of BPD is multifactorial, and the clinical phenotype is variable. BPD is associated with substantial mortality and short- and long-term morbidity. The incidence of BPD has remained stable or increased, as advances in neonatal care have led to improved survival of more extremely preterm infants. Extensive basic science, translational, and clinical research focusing on BPD has improved the current understanding of the factors that contribute to BPD pathogenesis. However, despite a better understanding of its pathophysiology, BPD continues to be challenging to prevent and manage adequately. The current review aims to provide a clinically useful synopsis of evidence on the prevention and management of BPD in preterm infants.
Collapse
|
79
|
Khedoe PPPSJ, Wu X, Gosens R, Hiemstra PS. Repairing damaged lungs using regenerative therapy. Curr Opin Pharmacol 2021; 59:85-94. [PMID: 34161852 PMCID: PMC9188766 DOI: 10.1016/j.coph.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/14/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022]
Abstract
There is an urgent need for better treatment of lung diseases that are a major cause of morbidity and mortality worldwide. This urgency is illustrated by the current COVID-19 health crisis. Moderate-to-extensive lung injury characterizes several lung diseases, and not only therapies that reduce such lung injury are needed but also those that regenerate lung tissue and repair existing lung injury. At present, such therapies are not available, but as a result of a rapid increase in our understanding of lung development and repair, lung regenerative therapies are on the horizon. Here, we discuss existing targets for treatment, as well as novel strategies for development of pharmacological and cell therapy-based regenerative treatment for a variety of lung diseases and clinical studies. We discuss how both patient-relevant in vitro disease models using innovative culture techniques and other advanced new technologies aid in the development of pulmonary regenerative medicine.
Collapse
Affiliation(s)
| | - Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
80
|
Sun AM, Hoffman T, Luu BQ, Ashammakhi N, Li S. Application of lung microphysiological systems to COVID-19 modeling and drug discovery: a review. Biodes Manuf 2021; 4:757-775. [PMID: 34178414 PMCID: PMC8213042 DOI: 10.1007/s42242-021-00136-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
There is a pressing need for effective therapeutics for coronavirus disease 2019 (COVID-19), the respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. The process of drug development is a costly and meticulously paced process, where progress is often hindered by the failure of initially promising leads. To aid this challenge, in vitro human microphysiological systems need to be refined and adapted for mechanistic studies and drug screening, thereby saving valuable time and resources during a pandemic crisis. The SARS-CoV-2 virus attacks the lung, an organ where the unique three-dimensional (3D) structure of its functional units is critical for proper respiratory function. The in vitro lung models essentially recapitulate the distinct tissue structure and the dynamic mechanical and biological interactions between different cell types. Current model systems include Transwell, organoid and organ-on-a-chip or microphysiological systems (MPSs). We review models that have direct relevance toward modeling the pathology of COVID-19, including the processes of inflammation, edema, coagulation, as well as lung immune function. We also consider the practical issues that may influence the design and fabrication of MPS. The role of lung MPS is addressed in the context of multi-organ models, and it is discussed how high-throughput screening and artificial intelligence can be integrated with lung MPS to accelerate drug development for COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Argus M. Sun
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- UC San Diego Healthcare, UCSD, La Jolla, CA 92037 USA
| | - Tyler Hoffman
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
| | - Bao Q. Luu
- Pulmonary Diseases and Critical Care, Scripps Green Hospital, Scripps Health, La Jolla, CA 92037 USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824 USA
| | - Song Li
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 USA
| |
Collapse
|
81
|
Sun F, Wang G, Pradhan A, Xu K, Gomez-Arroyo J, Zhang Y, Kalin GT, Deng Z, Vagnozzi RJ, He H, Dunn AW, Wang Y, York AJ, Hegde RS, Woods JC, Kalin TV, Molkentin JD, Kalinichenko VV. Nanoparticle Delivery of STAT3 Alleviates Pulmonary Hypertension in a Mouse Model of Alveolar Capillary Dysplasia. Circulation 2021; 144:539-555. [PMID: 34111939 DOI: 10.1161/circulationaha.121.053980] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a common complication in patients with alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a severe congenital disorder associated with mutations in the FOXF1 gene. Although the loss of alveolar microvasculature causes PH in patients with ACDMPV, it is unknown whether increasing neonatal lung angiogenesis could prevent PH and right ventricular (RV) hypertrophy. METHODS We used echocardiography, RV catheterization, immunostaining, and biochemical methods to examine lung and heart remodeling and RV output in Foxf1WT/S52F mice carrying the S52F Foxf1 mutation (identified in patients with ACDMPV). The ability of Foxf1WT/S52F mutant embryonic stem cells to differentiate into respiratory cell lineages in vivo was examined using blastocyst complementation. Intravascular delivery of nanoparticles with a nonintegrating Stat3 expression vector was used to improve neonatal pulmonary angiogenesis in Foxf1WT/S52F mice and determine its effects on PH and RV hypertrophy. RESULTS Foxf1WT/S52F mice developed PH and RV hypertrophy after birth. The severity of PH in Foxf1WT/S52F mice directly correlated with mortality, low body weight, pulmonary artery muscularization, and increased collagen deposition in the lung tissue. Increased fibrotic remodeling was found in human ACDMPV lungs. Mouse embryonic stem cells carrying the S52F Foxf1 mutation were used to produce chimeras through blastocyst complementation and to demonstrate that Foxf1WT/S52F embryonic stem cells have a propensity to differentiate into pulmonary myofibroblasts. Intravascular delivery of nanoparticles carrying Stat3 cDNA protected Foxf1WT/S52F mice from RV hypertrophy and PH, improved survival, and decreased fibrotic lung remodeling. CONCLUSIONS Nanoparticle therapies increasing neonatal pulmonary angiogenesis may be considered to prevent PH in ACDMPV.
Collapse
Affiliation(s)
- Fei Sun
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Guolun Wang
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Arun Pradhan
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Kui Xu
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Jose Gomez-Arroyo
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Department of Internal Medicine, Section of Pulmonary and Critical Care (J.G.-A.), University of Cincinnati, OH
| | - Yufang Zhang
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Gregory T Kalin
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Division of Pulmonary Biology (G.T.K., H.H., T.V.K., J.D.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Zicheng Deng
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- The Materials Science and Engineering Program, College of Engineering and Applied Science (Z.D., A.W.D.), University of Cincinnati, OH
| | - Ronald J Vagnozzi
- Division of Molecular Cardiovascular Biology, Heart Institute (R.J.V., A.J.Y., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Hua He
- Division of Pulmonary Biology (G.T.K., H.H., T.V.K., J.D.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Andrew W Dunn
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- The Materials Science and Engineering Program, College of Engineering and Applied Science (Z.D., A.W.D.), University of Cincinnati, OH
| | - Yuhua Wang
- Division of Developmental Biology (Y.W., R.S.H., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Allen J York
- Division of Molecular Cardiovascular Biology, Heart Institute (R.J.V., A.J.Y., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Rashmi S Hegde
- Division of Developmental Biology (Y.W., R.S.H., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Department of Pediatrics (R.S.H., J.C.W., T.V.K., J.S.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Jason C Woods
- Department of Pediatrics (R.S.H., J.C.W., T.V.K., J.S.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Center for Pulmonary Imaging Research, Division of Pulmonary Medicine (J.C.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Tanya V Kalin
- Division of Pulmonary Biology (G.T.K., H.H., T.V.K., J.D.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Department of Pediatrics (R.S.H., J.C.W., T.V.K., J.S.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| | - Jeffery D Molkentin
- Division of Pulmonary Biology (G.T.K., H.H., T.V.K., J.D.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Division of Molecular Cardiovascular Biology, Heart Institute (R.J.V., A.J.Y., J.D.M.), Cincinnati Children's Hospital Medical Center, OH
- Department of Pediatrics (R.S.H., J.C.W., T.V.K., J.S.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Howard Hughes Medical Institute (J.D.M.), Cincinnati Children's Hospital Medical Center, OH
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute (F.S., G.W., A.P., K.X., J.G.-A., Y.Z., G.T.K., Z.D., A.W.D., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Division of Pulmonary Biology (G.T.K., H.H., T.V.K., J.D.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Division of Developmental Biology (Y.W., R.S.H., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
- Department of Pediatrics (R.S.H., J.C.W., T.V.K., J.S.M., V.V.K.), Cincinnati Children's Hospital Medical Center, OH
| |
Collapse
|
82
|
Archer F, Bobet-Erny A, Gomes M. State of the art on lung organoids in mammals. Vet Res 2021; 52:77. [PMID: 34078444 PMCID: PMC8170649 DOI: 10.1186/s13567-021-00946-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
The number and severity of diseases affecting lung development and adult respiratory function have stimulated great interest in developing new in vitro models to study lung in different species. Recent breakthroughs in 3-dimensional (3D) organoid cultures have led to new physiological in vitro models that better mimic the lung than conventional 2D cultures. Lung organoids simulate multiple aspects of the real organ, making them promising and useful models for studying organ development, function and disease (infection, cancer, genetic disease). Due to their dynamics in culture, they can serve as a sustainable source of functional cells (biobanking) and be manipulated genetically. Given the differences between species regarding developmental kinetics, the maturation of the lung at birth, the distribution of the different cell populations along the respiratory tract and species barriers for infectious diseases, there is a need for species-specific lung models capable of mimicking mammal lungs as they are of great interest for animal health and production, following the One Health approach. This paper reviews the latest developments in the growing field of lung organoids.
Collapse
Affiliation(s)
- Fabienne Archer
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France.
| | - Alexandra Bobet-Erny
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| | - Maryline Gomes
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| |
Collapse
|
83
|
Mechanical plasticity of collagen directs branch elongation in human mammary gland organoids. Nat Commun 2021; 12:2759. [PMID: 33980857 PMCID: PMC8115695 DOI: 10.1038/s41467-021-22988-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
Epithelial branch elongation is a central developmental process during branching morphogenesis in diverse organs. This fundamental growth process into large arborized epithelial networks is accompanied by structural reorganization of the surrounding extracellular matrix (ECM), well beyond its mechanical linear response regime. Here, we report that epithelial ductal elongation within human mammary organoid branches relies on the non-linear and plastic mechanical response of the surrounding collagen. Specifically, we demonstrate that collective back-and-forth motion of cells within the branches generates tension that is strong enough to induce a plastic reorganization of the surrounding collagen network which results in the formation of mechanically stable collagen cages. Such matrix encasing in turn directs further tension generation, branch outgrowth and plastic deformation of the matrix. The identified mechanical tension equilibrium sets a framework to understand how mechanical cues can direct ductal branch elongation. Mammary organoid growth from single primary human cells rely on distinct morphogenetic processes. Here, the authors observe by live cell imaging the importance of the plastic mechanical response of the extracellular matrix and cell migration for the underlying arborized structure formation process.
Collapse
|
84
|
Liberti DC, Kremp MM, Liberti WA, Penkala IJ, Li S, Zhou S, Morrisey EE. Alveolar epithelial cell fate is maintained in a spatially restricted manner to promote lung regeneration after acute injury. Cell Rep 2021; 35:109092. [PMID: 33979629 PMCID: PMC8220578 DOI: 10.1016/j.celrep.2021.109092] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/23/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
Alveolar epithelial type 2 (AT2) cells integrate signals from multiple molecular pathways to proliferate and differentiate to drive regeneration of the lung alveolus. Utilizing in vivo genetic and ex vivo organoid models, we investigated the role of Fgfr2 signaling in AT2 cells across the lifespan and during adult regeneration after influenza infection. We show that, although dispensable for adult homeostasis, Fgfr2 restricts AT2 cell fate during postnatal lung development. Using an unbiased computational imaging approach, we demonstrate that Fgfr2 promotes AT2 cell proliferation and restrains differentiation in actively regenerating areas after injury. Organoid assays reveal that Fgfr2-deficient AT2 cells remain competent to respond to multiple parallel proliferative inputs. Moreover, genetic blockade of AT2 cell cytokinesis demonstrates that cell division and differentiation are uncoupled during alveolar regeneration. These data reveal that Fgfr2 maintains AT2 cell fate, balancing proliferation and differentiation during lung alveolar regeneration.
Collapse
Affiliation(s)
- Derek C Liberti
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Madison M Kremp
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William A Liberti
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ian J Penkala
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shanru Li
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Zhou
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
85
|
Brouns I, Verckist L, Pintelon I, Timmermans JP, Adriaensen D. Pulmonary Sensory Receptors. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2021; 233:1-65. [PMID: 33950466 DOI: 10.1007/978-3-030-65817-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Inge Brouns
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium.
| | - Line Verckist
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| | - Dirk Adriaensen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerpen (Wilrijk), Belgium
| |
Collapse
|
86
|
Gajjala PR, Madala SK. Notch3: A New Culprit in Fibrotic Lung Disease. Am J Respir Cell Mol Biol 2021; 64:403-404. [PMID: 33591242 PMCID: PMC8008798 DOI: 10.1165/rcmb.2021-0024ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Prathibha R Gajjala
- Department of Pediatrics University of Cincinnati Cincinnati, Ohio and.,Division of Pulmonary Medicine Cincinnati Children's Hospital Medical Center Cincinnati, Ohio
| | - Satish K Madala
- Department of Pediatrics University of Cincinnati Cincinnati, Ohio and.,Division of Pulmonary Medicine Cincinnati Children's Hospital Medical Center Cincinnati, Ohio
| |
Collapse
|
87
|
Zhang L, Zhu J, Wang H, Xia J, Liu P, Chen F, Jiang H, Miao Q, Wu W, Zhang L, Luo L, Jiang X, Bai Y, Sun C, Chen D, Zhang X. A high-resolution cell atlas of the domestic pig lung and an online platform for exploring lung single-cell data. J Genet Genomics 2021; 48:411-425. [PMID: 34144929 DOI: 10.1016/j.jgg.2021.03.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 12/28/2022]
Abstract
The genetically engineered pig is regarded as an optimal source of organ transplantation for humans and an excellent model for human disease research, given its comparable physiology to human beings. A myriad of single-cell RNA sequencing (scRNA-seq) data on humans has been reported, but such data on pigs are scarce. Here, we apply scRNA-seq technology to study the cellular heterogeneity of 3-month-old pig lungs, generating the single-cell atlas of 13,580 cells covering 16 major cell types. Based on these data, we systematically characterize the similarities and differences in the cellular cross-talk and expression patterns of respiratory virus receptors in each cell type of pig lungs compared with human lungs. Furthermore, we analyze pig lung xenotransplantation barriers and reported the cell-type expression patterns of 10 genes associated with pig-to-human immunobiological incompatibility and coagulation dysregulation. We also investigate the conserved transcription factors (TFs) and their candidate target genes and constructed five conserved TF regulatory networks in the main cell types shared by pig and human lungs. Finally, we present a comprehensive and openly accessible online platform, ScdbLung. Our scRNA-seq atlas of the domestic pig lung and ScdbLung database can guide pig lung research and clinical applicability.
Collapse
Affiliation(s)
- Lijing Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Jiacheng Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Haoyu Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Jun Xia
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Ping Liu
- MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Fang Chen
- MGI, BGI-Shenzhen, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Hui Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Qiuling Miao
- Department of Pathology, Shenzhen Children's Hospital, Shenzhen 518038, China
| | - Weiying Wu
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310031, China
| | - Lingli Zhang
- Department of Pathophysiology, School of Basic Medicine, Guilin Medical University, Guilin 541199, China
| | - Lihua Luo
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Yong Bai
- BGI-Shenzhen, Shenzhen 518083, China
| | - Chengcheng Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | | | - Xingliang Zhang
- Institute of Pediatrics, Department of Pediatric Surgery, Shenzhen Children's Hospital, Shenzhen 518038, China; Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
88
|
Basil MC, Katzen J, Engler AE, Guo M, Herriges MJ, Kathiriya JJ, Windmueller R, Ysasi AB, Zacharias WJ, Chapman HA, Kotton DN, Rock JR, Snoeck HW, Vunjak-Novakovic G, Whitsett JA, Morrisey EE. The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future. Cell Stem Cell 2021; 26:482-502. [PMID: 32243808 PMCID: PMC7128675 DOI: 10.1016/j.stem.2020.03.009] [Citation(s) in RCA: 248] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The respiratory system, which includes the trachea, airways, and distal alveoli, is a complex multi-cellular organ that intimately links with the cardiovascular system to accomplish gas exchange. In this review and as members of the NIH/NHLBI-supported Progenitor Cell Translational Consortium, we discuss key aspects of lung repair and regeneration. We focus on the cellular compositions within functional niches, cell-cell signaling in homeostatic health, the responses to injury, and new methods to study lung repair and regeneration. We also provide future directions for an improved understanding of the cell biology of the respiratory system, as well as new therapeutic avenues.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeremy Katzen
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anna E Engler
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Minzhe Guo
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael J Herriges
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jaymin J Kathiriya
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Rebecca Windmueller
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra B Ysasi
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - William J Zacharias
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Hal A Chapman
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jason R Rock
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Hans-Willem Snoeck
- Center for Human Development, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Gordana Vunjak-Novakovic
- Departments of Biomedical Engineering and Medicine, Columbia University, New York, NY 10027, USA
| | - Jeffrey A Whitsett
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Edward E Morrisey
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
89
|
Bodas M, Moore AR, Subramaniyan B, Georgescu C, Wren JD, Freeman WM, Brown BR, Metcalf JP, Walters MS. Cigarette Smoke Activates NOTCH3 to Promote Goblet Cell Differentiation in Human Airway Epithelial Cells. Am J Respir Cell Mol Biol 2021; 64:426-440. [PMID: 33444514 PMCID: PMC8008804 DOI: 10.1165/rcmb.2020-0302oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death in the United States and is primarily caused by cigarette smoking. Increased numbers of mucus-producing secretory ("goblet") cells, defined as goblet cell metaplasia or hyperplasia (GCMH), contributes significantly to COPD pathophysiology. The objective of this study was to determine whether NOTCH signaling regulates goblet cell differentiation in response to cigarette smoke. Primary human bronchial epithelial cells (HBECs) from nonsmokers and smokers with COPD were differentiated in vitro on air-liquid interface and exposed to cigarette smoke extract (CSE) for 7 days. NOTCH signaling activity was modulated using 1) the NOTCH/γ-secretase inhibitor dibenzazepine (DBZ), 2) lentiviral overexpression of the NICD3 (NOTCH3-intracellular domain), or 3) NOTCH3-specific siRNA. Cell differentiation and response to CSE were evaluated by quantitative PCR, Western blotting, immunostaining, and RNA sequencing. We found that CSE exposure of nonsmoker airway epithelium induced goblet cell differentiation characteristic of GCMH. Treatment with DBZ suppressed CSE-dependent induction of goblet cell differentiation. Furthermore, CSE induced NOTCH3 activation, as revealed by increased NOTCH3 nuclear localization and elevated NICD3 protein levels. Overexpression of NICD3 increased the expression of goblet cell-associated genes SPDEF and MUC5AC, whereas NOTCH3 knockdown suppressed CSE-mediated induction of SPDEF and MUC5AC. Finally, CSE exposure of COPD airway epithelium induced goblet cell differentiation in a NOTCH3-dependent manner. These results identify NOTCH3 activation as one of the important mechanisms by which cigarette smoke induces goblet cell differentiation, thus providing a novel potential strategy to control GCMH-related pathologies in smokers and patients with COPD.
Collapse
Affiliation(s)
- Manish Bodas
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Andrew R. Moore
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Bharathiraja Subramaniyan
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Constantin Georgescu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Jonathan D. Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Willard M. Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Brent R. Brown
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Jordan P. Metcalf
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| | - Matthew S. Walters
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and
| |
Collapse
|
90
|
He H, Snowball J, Sun F, Na CL, Whitsett JA. IGF1R controls mechanosignaling in myofibroblasts required for pulmonary alveologenesis. JCI Insight 2021; 6:144863. [PMID: 33591952 PMCID: PMC8026181 DOI: 10.1172/jci.insight.144863] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/10/2021] [Indexed: 11/17/2022] Open
Abstract
Ventilation throughout life is dependent on the formation of pulmonary alveoli, which create an extensive surface area in which the close apposition of respiratory epithelium and endothelial cells of the pulmonary microvascular enables efficient gas exchange. Morphogenesis of the alveoli initiates at late gestation in humans and the early postnatal period in the mouse. Alveolar septation is directed by complex signaling interactions among multiple cell types. Here, we demonstrate that IGF1 receptor gene (Igf1r) expression by a subset of pulmonary fibroblasts is required for normal alveologenesis in mice. Postnatal deletion of Igf1r caused alveolar simplification, disrupting alveolar elastin networks and extracellular matrix without altering myofibroblast differentiation or proliferation. Moreover, loss of Igf1r impaired contractile properties of lung myofibroblasts and inhibited myosin light chain (MLC) phosphorylation and mechanotransductive nuclear YAP activity. Activation of p-AKT, p-MLC, and nuclear YAP in myofibroblasts was dependent on Igf1r. Pharmacologic activation of AKT enhanced MLC phosphorylation, increased YAP activation, and ameliorated alveolar simplification in vivo. IGF1R controls mechanosignaling in myofibroblasts required for lung alveologenesis.
Collapse
Affiliation(s)
- Hua He
- Division of Pulmonary Biology and
| | | | - Fei Sun
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | |
Collapse
|
91
|
Saxena A, Walters MS, Shieh JH, Shen LB, Gomi K, Downey RJ, Crystal RG, Moore MAS. Extracellular vesicles from human airway basal cells respond to cigarette smoke extract and affect vascular endothelial cells. Sci Rep 2021; 11:6104. [PMID: 33731767 PMCID: PMC7969738 DOI: 10.1038/s41598-021-85534-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
The human airway epithelium lining the bronchial tree contains basal cells that proliferate, differentiate, and communicate with other components of their microenvironment. One method that cells use for intercellular communication involves the secretion of exosomes and other extracellular vesicles (EVs). We isolated exosome-enriched EVs that were produced from an immortalized human airway basal cell line (BCi-NS1.1) and found that their secretion is increased by exposure to cigarette smoke extract, suggesting that this stress stimulates release of EVs which could affect signaling to other cells. We have previously shown that primary human airway basal cells secrete vascular endothelial growth factor A (VEGFA) which can activate MAPK signaling cascades in endothelial cells via VEGF receptor-2 (VEGFR2). Here, we show that exposure of endothelial cells to exosome-enriched airway basal cell EVs promotes the survival of these cells and that this effect also involves VEGFR2 activation and is, at least in part, mediated by VEGFA present in the EVs. These observations demonstrate that EVs are involved in the intercellular signaling between airway basal cells and the endothelium which we previously reported. The downstream signaling pathways involved may be distinct and specific to the EVs, however, as increased phosphorylation of Akt, STAT3, p44/42 MAPK, and p38 MAPK was not seen following exposure of endothelial cells to airway basal cell EVs.
Collapse
Affiliation(s)
- Ashish Saxena
- Department of Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA.
| | - Matthew S Walters
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jae-Hung Shieh
- Department of Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ling-Bo Shen
- Department of Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kazunori Gomi
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Robert J Downey
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Malcolm A S Moore
- Department of Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
92
|
TP63 basal cells are indispensable during endoderm differentiation into proximal airway cells on acellular lung scaffolds. NPJ Regen Med 2021; 6:12. [PMID: 33674599 PMCID: PMC7935966 DOI: 10.1038/s41536-021-00124-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/01/2021] [Indexed: 12/24/2022] Open
Abstract
The use of decellularized whole-organ scaffolds for bioengineering of organs is a promising avenue to circumvent the shortage of donor organs for transplantation. However, recellularization of acellular scaffolds from multicellular organs like the lung with a variety of different cell types remains a challenge. Multipotent cells could be an ideal cell source for recellularization. Here we investigated the hierarchical differentiation process of multipotent ES-derived endoderm cells into proximal airway epithelial cells on acellular lung scaffolds. The first cells to emerge on the scaffolds were TP63+ cells, followed by TP63+/KRT5+ basal cells, and finally multi-ciliated and secretory airway epithelial cells. TP63+/KRT5+ basal cells on the scaffolds simultaneously expressed KRT14, like basal cells involved in airway repair after injury. Removal of TP63 by CRISPR/Cas9 in the ES cells halted basal and airway cell differentiation on the scaffolds. These findings suggest that differentiation of ES-derived endoderm cells into airway cells on decellularized lung scaffolds proceeds via TP63+ basal cell progenitors and tracks a regenerative repair pathway. Understanding the process of differentiation is key for choosing the cell source for repopulation of a decellularized organ scaffold. Our data support the use of airway basal cells for repopulating the airway side of an acellular lung scaffold.
Collapse
|
93
|
Deng Z, Kalin GT, Shi D, Kalinichenko VV. Nanoparticle Delivery Systems with Cell-Specific Targeting for Pulmonary Diseases. Am J Respir Cell Mol Biol 2021; 64:292-307. [PMID: 33095997 PMCID: PMC7909340 DOI: 10.1165/rcmb.2020-0306tr] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Respiratory disorders are among the most important medical problems threatening human life. The conventional therapeutics for respiratory disorders are hindered by insufficient drug concentrations at pathological lesions, lack of cell-specific targeting, and various biobarriers in the conducting airways and alveoli. To address these critical issues, various nanoparticle delivery systems have been developed to serve as carriers of specific drugs, DNA expression vectors, and RNAs. The unique properties of nanoparticles, including controlled size and distribution, surface functional groups, high payload capacity, and drug release triggering capabilities, are tailored to specific requirements in drug/gene delivery to overcome major delivery barriers in pulmonary diseases. To avoid off-target effects and improve therapeutic efficacy, nanoparticles with high cell-targeting specificity are essential for successful nanoparticle therapies. Furthermore, low toxicity and high degradability of the nanoparticles are among the most important requirements in the nanoparticle designs. In this review, we provide the most up-to-date research and clinical outcomes in nanoparticle therapies for pulmonary diseases. We also address the current critical issues in key areas of pulmonary cell targeting, biosafety and compatibility, and molecular mechanisms for selective cellular uptake.
Collapse
Affiliation(s)
- Zicheng Deng
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, Ohio; and
- Center for Lung Regenerative Medicine
- Division of Pulmonary Biology, and
| | - Gregory T Kalin
- Center for Lung Regenerative Medicine
- Division of Pulmonary Biology, and
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, Ohio; and
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine
- Division of Pulmonary Biology, and
- Department of Pediatrics, College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
94
|
Wen B, Li E, Ustiyan V, Wang G, Guo M, Na CL, Kalin GT, Galvan V, Xu Y, Weaver TE, Kalin TV, Whitsett JA, Kalinichenko VV. In Vivo Generation of Lung and Thyroid Tissues from Embryonic Stem Cells Using Blastocyst Complementation. Am J Respir Crit Care Med 2021; 203:471-483. [PMID: 32877203 PMCID: PMC7885842 DOI: 10.1164/rccm.201909-1836oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 09/02/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: The regeneration and replacement of lung cells or tissues from induced pluripotent stem cell- or embryonic stem cell-derived cells represent future therapies for life-threatening pulmonary disorders but are limited by technical challenges to produce highly differentiated cells able to maintain lung function. Functional lung tissue-containing airways, alveoli, vasculature, and stroma have never been produced via directed differentiation of embryonic stem cells (ESCs) or induced pluripotent stem cells. We sought to produce all tissue components of the lung from bronchi to alveoli by embryo complementation.Objectives: To determine whether ESCs are capable of generating lung tissue in Nkx2-1-/- mouse embryos with lung agenesis.Methods: Blastocyst complementation was used to produce chimeras from normal mouse ESCs and Nkx2-1-/- embryos, which lack pulmonary tissues. Nkx2-1-/- chimeras were examined using immunostaining, transmission electronic microscopy, fluorescence-activated cell sorter analysis, and single-cell RNA sequencing.Measurements and Main Results: Although peripheral pulmonary and thyroid tissues are entirely lacking in Nkx2-1 gene-deleted embryos, pulmonary and thyroid structures in Nkx2-1-/- chimeras were restored after ESC complementation. Respiratory epithelial cell lineages in restored lungs of Nkx2-1-/- chimeras were derived almost entirely from ESCs, whereas endothelial, immune, and stromal cells were mosaic. ESC-derived cells from multiple respiratory cell lineages were highly differentiated and indistinguishable from endogenous cells based on morphology, ultrastructure, gene expression signatures, and cell surface proteins used to identify cell types by fluorescence-activated cell sorter.Conclusions: Lung and thyroid tissues were generated in vivo from ESCs by blastocyst complementation. Nkx2-1-/- chimeras can be used as "bioreactors" for in vivo differentiation and functional studies of ESC-derived progenitor cells.
Collapse
Affiliation(s)
- Bingqiang Wen
- Center for Lung Regenerative Medicine, Perinatal Institute
| | - Enhong Li
- Center for Lung Regenerative Medicine, Perinatal Institute
| | | | - Guolun Wang
- Center for Lung Regenerative Medicine, Perinatal Institute
| | - Minzhe Guo
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
| | | | | | - Veronica Galvan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; and
| | - Yan Xu
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
| | - Timothy E. Weaver
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
| | - Tanya V. Kalin
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
| | - Jeffrey A. Whitsett
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
- Department of Cellular and Integrative Physiology and The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Vladimir V. Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute
- Division of Pulmonary Biology, and
- Division of Developmental Biology, Cincinnati Children’s Research Foundation, Cincinnati, Ohio
- Department of Cellular and Integrative Physiology and The Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
95
|
Kong J, Wen S, Cao W, Yue P, Xu X, Zhang Y, Luo L, Chen T, Li L, Wang F, Tao J, Zhou G, Luo S, Liu A, Bao F. Lung organoids, useful tools for investigating epithelial repair after lung injury. Stem Cell Res Ther 2021; 12:95. [PMID: 33516265 PMCID: PMC7846910 DOI: 10.1186/s13287-021-02172-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/17/2021] [Indexed: 02/07/2023] Open
Abstract
Organoids are derived from stem cells or organ-specific progenitors. They display structures and functions consistent with organs in vivo. Multiple types of organoids, including lung organoids, can be generated. Organoids are applied widely in development, disease modelling, regenerative medicine, and other multiple aspects. Various human pulmonary diseases caused by several factors can be induced and lead to different degrees of lung epithelial injury. Epithelial repair involves the participation of multiple cells and signalling pathways. Lung organoids provide an excellent platform to model injury to and repair of lungs. Here, we review the recent methods of cultivating lung organoids, applications of lung organoids in epithelial repair after injury, and understanding the mechanisms of epithelial repair investigated using lung organoids. By using lung organoids, we can discover the regulatory mechanisms related to the repair of lung epithelia. This strategy could provide new insights for more effective management of lung diseases and the development of new drugs.
Collapse
Affiliation(s)
- Jing Kong
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China.,The School of Medicine, Kunming University, Kunming, 650214, China
| | - Shiyuan Wen
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Wenjing Cao
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Peng Yue
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xin Xu
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Yu Zhang
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Lisha Luo
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Taigui Chen
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Lianbao Li
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Feng Wang
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Jian Tao
- The School of Medicine, Kunming University, Kunming, 650214, China
| | - Guozhong Zhou
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Suyi Luo
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China
| | - Aihua Liu
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming, Kunming Medical University, Kunming, 650030, China.
| | - Fukai Bao
- The Institute for Tropical Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, 650500, Yunnan, China. .,Department of Microbiology and Immunology, Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
96
|
Lee DD, Park SJ, Zborek KL, Schwarz MA. A shift from glycolytic and fatty acid derivatives toward one-carbon metabolites in the developing lung during transitions of the early postnatal period. Am J Physiol Lung Cell Mol Physiol 2021; 320:L640-L659. [PMID: 33502935 DOI: 10.1152/ajplung.00417.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
During postnatal lung development, metabolic changes that coincide with stages of alveolar formation are poorly understood. Responding to developmental and environmental factors, metabolic changes can be rapidly and adaptively altered. The objective of the present study was to determine biological and technical determinants of metabolic changes during postnatal lung development. Over 118 metabolic features were identified by liquid chromatography with tandem mass spectrometry (LC-MS/MS, Sciex QTRAP 5500 Triple Quadrupole). Biological determinants of metabolic changes were the transition from the postnatal saccular to alveolar stages and exposure to 85% hyperoxia, an environmental insult. Technical determinants of metabolic identification were brevity and temperature of harvesting, both of which improved metabolic preservation within samples. Multivariate statistical analyses revealed the transition between stages of lung development as the period of major metabolic alteration. Of three distinctive groups that clustered by age, the saccular stage was identified by its enrichment of both glycolytic and fatty acid derivatives. The critical transition between stages of development were denoted by changes in amino acid derivatives. Of the amino acid derivatives that significantly changed, a majority were linked to metabolites of the one-carbon metabolic pathway. The enrichment of one-carbon metabolites was independent of age and environmental insult. Temperature was also found to significantly influence the metabolic levels within the postmortem sampled lung, which underscored the importance of methodology. Collectively, these data support not only distinctive stages of metabolic change but also highlight amino acid metabolism, in particular one-carbon metabolites as metabolic signatures of the early postnatal lung.
Collapse
Affiliation(s)
- Daniel D Lee
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sang Jun Park
- Department of Preprofessional Studies, University of Notre Dame, South Bend, Indiana
| | - Kirsten L Zborek
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
97
|
Abstract
The lungs are constantly exposed to the external environment and are therefore vulnerable to insults that can cause infection and injury. Maintaining the integrity and barrier function of the lung epithelium requires complex interactions of multiple cell lineages. Elucidating the cellular players and their regulation mechanisms provides fundamental information to deepen understanding about the responses and contributions of lung stem cells. This Review focuses on advances in our understanding of mammalian alveolar epithelial stem cell subpopulations and discusses insights about the regeneration-specific cell status of alveolar epithelial stem cells. We also consider how these advances can inform our understanding of post-injury lung repair processes and lung diseases.
Collapse
Affiliation(s)
- Huijuan Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
98
|
Whitsett JA, Jobe AH. Commentary on the Truncated Splice Variant of the GM-CSF Receptor Beta-Chain in Peripheral Blood Serves as Severity Biomarker of Respiratory Failure in Newborns. Neonatology 2021; 118:194-197. [PMID: 33744877 PMCID: PMC8178199 DOI: 10.1159/000514639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Jeffrey A Whitsett
- Department of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA,
| | - Alan H Jobe
- Department of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
99
|
Functional Exploration of the Pulmonary NEB ME. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2021; 233:31-67. [PMID: 33950469 DOI: 10.1007/978-3-030-65817-5_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
100
|
Pérez-Bravo D, Myti D, Mižíková I, Pfeffer T, Surate Solaligue DE, Nardiello C, Vadász I, Herold S, Seeger W, Ahlbrecht K, Morty RE. A comparison of airway pressures for inflation fixation of developing mouse lungs for stereological analyses. Histochem Cell Biol 2020; 155:203-214. [PMID: 33372249 PMCID: PMC7910376 DOI: 10.1007/s00418-020-01951-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 12/02/2022]
Abstract
The morphometric analysis of lung structure using the principles of stereology has emerged as a powerful tool to describe the structural changes in lung architecture that accompany the development of lung disease that is experimentally modelled in adult mice. These stereological principles are now being applied to the study of the evolution of the lung architecture over the course of prenatal and postnatal lung development in mouse neonates and adolescents. The immature lung is structurally and functionally distinct from the adult lung, and has a smaller volume than does the adult lung. These differences have raised concerns about whether the inflation fixation of neonatal mouse lungs with the airway pressure (Paw) used for the inflation fixation of adult mouse lungs may cause distortion of the neonatal mouse lung structure, leading to the generation of artefacts in subsequent analyses. The objective of this study was to examine the impact of a Paw of 10, 20 and 30 cmH2O on the estimation of lung volumes and stereologically assessed parameters that describe the lung structure in developing mouse lungs. The data presented demonstrate that low Paw (10 cmH2O) leads to heterogeneity in the unfolding of alveolar structures within the lungs, and that high Paw (30 cmH2O) leads to an overestimation of the lung volume, and thus, affects the estimation of volume-dependent parameters, such as total alveoli number and gas-exchange surface area. Thus, these data support the use of a Paw of 20 cmH2O for inflation fixation in morphometric studies on neonatal mouse lungs.
Collapse
Affiliation(s)
- David Pérez-Bravo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany
| | - Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, 501 Smyth (Box 511), Ottawa, ON, 1H 8L6, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Tilman Pfeffer
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Centre for Paediatric and Adolescent Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Our Lady's Hospital, MoathillCo. Meath, Navan, C15 RK7Y, Ireland
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Cardio Pulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Cardio Pulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany.,Cardio Pulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University Giessen, Aulweg 130, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany. .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Aulweg 123, 35394, Giessen, Germany. .,Cardio Pulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany.
| |
Collapse
|