1
|
Li S, Lu Q, Lu J, Song X, Gu Y, Duan X, Jiang W, Gu G, Zheng M, Xie L, Fang M. IRF1-RIG-I signaling defects in the aged alveolar epithelial cells may contribute to decreased pulmonary antiviral immune responses. Mech Ageing Dev 2025; 224:112037. [PMID: 39874992 DOI: 10.1016/j.mad.2025.112037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Alveolar epithelial cells (AECs) are the primary targets of many pathogens and play an important role in sensing viruses and regulating immunity. Yet, little is known about the antiviral responses in the aged AECs. METHODS The responses of young or aged AECs after viral infection were analyzed using methods such as flow cytometry, quantitative real-time PCR, Western blot detection, and transwell chemotaxis assay. Deep sequencing and KEGG analysis were used to identify key pathways and genes associated with aged AECs, followed by functional analysis. RESULTS The retinoic acid-inducible gene I (RIG-I) signaling is defective in aged AECs after influenza A virus (IAV) infection. The interferon regulatory factor 1 (IRF1) binds the promoter of RIG-I gene Ddx58 to activate its expression. The regulation of IRF1 is also defective in AECs from aged mice. Fewer NK cells, monocytes, and T cells are recruited by the cell supernatant from PR8-infected aged AECs. Importantly, IRF1-RIG-I signaling is also impaired in the AECs of elderly people after IAV infection. CONCLUSION Ageing impairs IRF1-RIG-I signaling in AECs, and the defective responses in AECs may contribute to reduced immune cell recruitment and activation in aged individuals after pulmonary viral infection.
Collapse
Affiliation(s)
- Shan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qianqian Lu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China; Henan Key Laboratory of Synthetic Biology and Biomanufacturing, Henan University, Kaifeng, Henan Province 475004, China
| | - Jiao Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xiaotong Song
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China; Henan Key Laboratory of Synthetic Biology and Biomanufacturing, Henan University, Kaifeng, Henan Province 475004, China
| | - Yang Gu
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Guanglei Gu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China; Henan Key Laboratory of Synthetic Biology and Biomanufacturing, Henan University, Kaifeng, Henan Province 475004, China
| | - Mengli Zheng
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China; School of Life Sciences, Henan University, Kaifeng, Henan Province, China; Henan Key Laboratory of Synthetic Biology and Biomanufacturing, Henan University, Kaifeng, Henan Province 475004, China.
| |
Collapse
|
2
|
Huo Y, He S, Chen Y. Lung organoids in COPD: recent advances and future prospects. Respir Res 2025; 26:76. [PMID: 40022099 PMCID: PMC11871743 DOI: 10.1186/s12931-025-03138-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease that is characterized by progressive airflow limitation, a high prevalence, and a high mortality rate. However, the specific mechanisms remain unclear, partly due to the lack of robust data from in vitro experimental models and animal models that do not adequately represent the structure and pathophysiology of the human lung. The recent advancement of lung organoid culture systems has facilitated new avenues for the investigation of COPD. Lung organoids are in vitro models derived from adult stem cells, human pluripotent stem cells, or embryonic stem cells, established through three-dimensional culture. They exhibit a high degree of homology and genetic consistency with human tissues and can better mimic human lungs in terms of function and structure compared to other traditional models. This review will summarise the generation process of lung organoids from different cell sources and their application in COPD research, and provide suggestions for future research directions.
Collapse
Affiliation(s)
- Yajie Huo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shengyang He
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, China.
- Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, China.
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, China.
- Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
3
|
Li D, Kortekaas RK, Douglas KBI, Douwenga W, Eisel ULM, Melgert BN, Gosens R, Schmidt M. TNF signaling mediates lipopolysaccharide-induced lung epithelial progenitor cell responses in mouse lung organoids. Biomed Pharmacother 2024; 181:117704. [PMID: 39581145 DOI: 10.1016/j.biopha.2024.117704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024] Open
Abstract
Bacterial respiratory infections are a major global health concern, often leading to lung injury and triggering lung repair mechanisms. Endogenous epithelial progenitor cells are crucial in this repair, yet the mechanisms remain poorly understood. This study investigates the response of lung epithelial progenitor cells to injury induced by lipopolysaccharide (LPS), a component of gram-negative bacteria, focusing on their regulation during lung repair. Lung epithelial cells (CD31-CD45-Epcam+) from wild-type and tumor necrosis factor (TNF) receptor 1/2 knock-out mice were co-cultured with wild-type fibroblasts. Organoid numbers and size were measured after 14 days of exposure to 100 ng/mL LPS. Immunofluorescence was used to assess differentiation (after 14 days), RNA sequencing analyzed gene expression changes (after 72 hours), and MTS assay assessed proliferative effects of LPS on individual cell types (after 24 hours). LPS treatment increased the number and size of wild-type lung organoids and promoted alveolar differentiation, indicated by more SPC+ organoids. RNA sequencing revealed upregulation of inflammatory and fibrosis-related markers, including Cxcl3, Cxcl5, Ccl20, Mmp13, and Il33, and enrichment of TNF-α signaling and epithelial-mesenchymal transition pathways. TNF receptor 1 deficiency inhibited LPS-induced progenitor cell activation and organoid growth. In conclusion, LPS enhances lung epithelial progenitor cell proliferation and differentiation via TNF receptor 1 signaling, highlighting potential therapeutic targets for bacterial lung injury.
Collapse
MESH Headings
- Animals
- Lipopolysaccharides/pharmacology
- Organoids/drug effects
- Organoids/metabolism
- Lung/pathology
- Lung/drug effects
- Lung/metabolism
- Mice
- Stem Cells/drug effects
- Stem Cells/metabolism
- Signal Transduction/drug effects
- Mice, Knockout
- Tumor Necrosis Factor-alpha/metabolism
- Mice, Inbred C57BL
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Cell Differentiation/drug effects
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Epithelial-Mesenchymal Transition/drug effects
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Dan Li
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rosa K Kortekaas
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Kelly B I Douglas
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Wanda Douwenga
- Department of Molecular Neurobiology and Neuroimmunology, Groningen Institute of Evolutionary Life Science, University of Groningen, Groningen, Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology and Neuroimmunology, Groningen Institute of Evolutionary Life Science, University of Groningen, Groningen, Netherlands
| | - Barbro N Melgert
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands; Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
4
|
Lehmann M, Krishnan R, Sucre J, Kulkarni HS, Pineda RH, Anderson C, Banovich NE, Behrsing HP, Dean CH, Haak A, Gosens R, Kaminski N, Zagorska A, Koziol-White C, Metcalf JP, Kim YH, Loebel C, Neptune E, Noel A, Raghu G, Sewald K, Sharma A, Suki B, Sperling A, Tatler A, Turner S, Rosas IO, van Ry P, Wille T, Randell SH, Pryhuber G, Rojas M, Bourke J, Königshoff M. Precision Cut Lung Slices: Emerging Tools for Preclinical and Translational Lung Research. An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 72:16-31. [PMID: 39499861 PMCID: PMC11707673 DOI: 10.1165/rcmb.2024-0479st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
The urgent need for effective treatments for acute and chronic lung diseases underscores the significance of developing innovative preclinical human research tools. The 2023 ATS Workshop on Precision Cut Lung Slices (PCLS) brought together 35 experts to discuss and address the role of human tissue-derived PCLS as a unique tool for target and drug discovery and validation in pulmonary medicine. With increasing interest and usage, along with advancements in methods and technology, there is a growing need for consensus on PCLS methodology and readouts. The current document recommends standard reporting criteria and emphasizes the requirement for careful collection and integration of clinical metadata. We further discuss current clinically relevant readouts that can be applied to PCLS and highlight recent developments and future steps for implementing novel technologies for PCLS modeling and analysis. The collection and correlation of clinical metadata and multiomic analysis will further advent the integration of this preclinical platform into patient endotyping and the development of tailored therapies for lung disease patients.
Collapse
Affiliation(s)
- Mareike Lehmann
- Philipps University Marburg, Institute for Lung Research, Marburg, Germany
- Helmholtz Center Munich, Institute for Lung Health and Immunity, Munich, Germany;
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Emergency Medicine, Boston, United States
| | - Jennifer Sucre
- Vanderbilt University Medical Center, Pediatrics, Nashville, Tennessee, United States
| | - Hrishikesh S Kulkarni
- Washington University in Saint Louis, Division of Pulmonary and Critical Care Medicine, Saint Louis, Missouri, United States
| | - Ricardo H Pineda
- University of Pittsburgh, Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | - Holger P Behrsing
- Institute for In Vitro Sciences Inc, Gaithersburg, Maryland, United States
| | - Charlotte H Dean
- Imperial College, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Haak
- Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Reinoud Gosens
- University of Groningen, Molecular Pharmacology, Groningen, Netherlands
| | - Naftali Kaminski
- Yale School of Medicine , Pulmonary, Critical Care and Sleep Mediine , New Haven, Connecticut, United States
| | - Anna Zagorska
- Gilead Sciences Inc, Foster City, California, United States
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey, United States
| | - Jordan P Metcalf
- The University of Oklahoma Health Sciences Center, Medicine, Oklahoma City, Oklahoma, United States
| | - Yong Ho Kim
- U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States
| | | | - Enid Neptune
- Johns Hopkins, Medicine/Pulmonary and Critical Care, Baltimore, Maryland, United States
| | - Alexandra Noel
- Louisiana State University, Baton Rouge, Louisiana, United States
| | - Ganesh Raghu
- University of Washington Medical Center, Division of Pulmonary and Critical Care Medicine, Seattle, Washington, United States
| | | | - Ashish Sharma
- University of Florida, Gainesville, Florida, United States
| | - Bela Suki
- Boston University, Biomedical Engineering, Boston, Massachusetts, United States
| | - Anne Sperling
- University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Amanda Tatler
- University of Nottingham, Respiratory Medicine , Nottingham, United Kingdom of Great Britain and Northern Ireland
| | - Scott Turner
- Pliant Therapeutics, South San Francisco, California, United States
| | - Ivan O Rosas
- Brigham and Women's Hospital, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Pam van Ry
- Brigham Young University, Chemistry and Biochemistry, Provo, Utah, United States
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Bundeswehr Medical Academy, Germany, Munich, Germany
| | - Scott H Randell
- University of North Carolina, Department of Cell Biology & Physiology, Chapel Hill, North Carolina, United States
| | - Gloria Pryhuber
- University of Rochester, Pediatrics, Rochester, New York, United States
| | - Mauricio Rojas
- Ohio State University, Columbus, OH, Pulmonary, Critical Care and Sleep Medicine, College of Medicine, , Columbus, Ohio, United States
| | - Jane Bourke
- Monash University, Department of Pharmacology, Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Melanie Königshoff
- University of Pittsburgh, Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
Bailey-Downs LC, Sherlock LG, Crossley MN, Rivera Negron A, Pierce PT, Wang S, Zhong H, Carter C, Burge K, Eckert JV, Rogers LK, Vitiello PF, Tipple TE. Selenium Deficiency Exacerbates Hyperoxia-Induced Lung Injury in Newborn C3H/HeN Mice. Antioxidants (Basel) 2024; 13:391. [PMID: 38671839 PMCID: PMC11047402 DOI: 10.3390/antiox13040391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Extremely preterm infants are often treated with supraphysiological oxygen, which contributes to the development of bronchopulmonary dysplasia (BPD). These same infants exhibit compromised antioxidant capacities due in part to selenium (Se) deficiency. Se is essential for basal and inducible antioxidant responses. The present study utilized a perinatal Se deficiency (SeD) mouse model to identify the combined effects of newborn hyperoxia exposure and SeD on alveolarization and antioxidant responses, including the identification of affected developmental pathways. Se-sufficient (SeS) and SeD C3H/HeN breeding pairs were generated, and pups were exposed to room air or 85% O2 from birth to 14 d. Survival, antioxidant protein expression, and RNA seq analyses were performed. Greater than 40% mortality was observed in hyperoxia-exposed SeD pups. Surviving SeD pups had greater lung growth deficits than hyperoxia-exposed SeS pups. Gpx2 and 4 protein and Gpx activity were significantly decreased in SeD pups. Nrf2-regulated proteins, Nqo1 and Gclc were increased in SeD pups exposed to hyperoxia. RNA seq revealed significant decreases in the Wnt/β-catenin and Notch pathways. Se is a biologically relevant modulator of perinatal lung development and antioxidant responses, especially in the context of hyperoxia exposure. The RNA seq analyses suggest pathways essential for normal lung development are dysregulated by Se deficiency.
Collapse
Affiliation(s)
- Lora C. Bailey-Downs
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Laura G. Sherlock
- University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Michaela N. Crossley
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Aristides Rivera Negron
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Paul T. Pierce
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Shirley Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Hua Zhong
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Cynthia Carter
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Kathryn Burge
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Jeffrey V. Eckert
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Lynette K. Rogers
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Peter F. Vitiello
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
| | - Trent E. Tipple
- University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.C.B.-D.); (S.W.); (H.Z.); (C.C.); (K.B.); (L.K.R.); (P.F.V.)
- Oklahoma Children’s Hospital OU Health, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Obata T, Mizoguchi S, Greaney AM, Adams T, Yuan Y, Edelstein S, Leiby KL, Rivero R, Wang N, Kim H, Yang J, Schupp JC, Stitelman D, Tsuchiya T, Levchenko A, Kaminski N, Niklason LE, Brickman Raredon MS. Organ Boundary Circuits Regulate Sox9+ Alveolar Tuft Cells During Post-Pneumonectomy Lung Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574469. [PMID: 38260691 PMCID: PMC10802449 DOI: 10.1101/2024.01.07.574469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Tissue homeostasis is controlled by cellular circuits governing cell growth, organization, and differentation. In this study we identify previously undescribed cell-to-cell communication that mediates information flow from mechanosensitive pleural mesothelial cells to alveolar-resident stem-like tuft cells in the lung. We find mesothelial cells to express a combination of mechanotransduction genes and lineage-restricted ligands which makes them uniquely capable of responding to tissue tension and producing paracrine cues acting on parenchymal populations. In parallel, we describe a large population of stem-like alveolar tuft cells that express the endodermal stem cell markers Sox9 and Lgr5 and a receptor profile making them uniquely sensitive to cues produced by pleural Mesothelium. We hypothesized that crosstalk from mesothelial cells to alveolar tuft cells might be central to the regulation of post-penumonectomy lung regeneration. Following pneumonectomy, we find that mesothelial cells display radically altered phenotype and ligand expression, in a pattern that closely tracks with parenchymal epithelial proliferation and alveolar tissue growth. During an initial pro-inflammatory stage of tissue regeneration, Mesothelium promotes epithelial proliferation via WNT ligand secretion, orchestrates an increase in microvascular permeability, and encourages immune extravasation via chemokine secretion. This stage is followed first by a tissue remodeling period, characterized by angiogenesis and BMP pathway sensitization, and then a stable return to homeostasis. Coupled with key changes in parenchymal structure and matrix production, the cumulative effect is a now larger organ including newly-grown, fully-functional tissue parenchyma. This study paints Mesothelial cells as a key orchestrating cell type that defines the boundary of the lung and exerts critical influence over the tissue-level signaling state regulating resident stem cell populations. The cellular circuits unearthed here suggest that human lung regeneration might be inducible through well-engineered approaches targeting the induction of tissue regeneration and safe return to homeostasis.
Collapse
Affiliation(s)
- Tomohiro Obata
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Satoshi Mizoguchi
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Allison M. Greaney
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of technology, Cambridge, MA, 02139
| | - Taylor Adams
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Yifan Yuan
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Sophie Edelstein
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Katherine L. Leiby
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Rachel Rivero
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Nuoya Wang
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Haram Kim
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Junchen Yang
- Computational Biology and Biomedical Informatics, Yale University, New Haven, CT, 06511, USA
| | - Jonas C. Schupp
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Respiratory Medicine, Hanover Medical School, Hanover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hanover, Germany
| | - David Stitelman
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Tomoshi Tsuchiya
- Department of Thoracic Surgery, University of Toyama, Toyama, 9300194, Japan
| | - Andre Levchenko
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Systems Biology Institute, Yale University, New Haven, CT, 06511, USA
- Department of Physics, Yale University, New Haven, CT, 06511, USA
| | - Naftali Kaminski
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Laura E. Niklason
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Humacyte, Inc., Durham, North Carolina
| | - Micha Sam Brickman Raredon
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, 06511, USA
- Pulmonary, Critical Care, & Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, CT, 06511, USA
| |
Collapse
|
7
|
Liu X, Zhang L, Zhu B, Liu Y, Li L, Hou J, Qian M, Zheng N, Zeng Y, Chen C, Goel A, Wang X. Role of GSDM family members in airway epithelial cells of lung diseases: a systematic and comprehensive transcriptomic analysis. Cell Biol Toxicol 2023; 39:2743-2760. [PMID: 37462807 DOI: 10.1007/s10565-023-09799-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/05/2023] [Indexed: 12/03/2023]
Abstract
Gasdermin (GSDM) family, the key executioners of pyroptosis, play crucial roles in anti-pathogen and anti-tumor immunities, although little is known about the expression of GSDM in lung diseases at single-cell resolution, especially in lung epithelial cells. We comprehensively investigated the transcriptomic profiles of GSDM members in various lung tissues from healthy subjects or patients with different lung diseases at single cell level, e.g., chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), lung adenocarcinoma (LUAD), or systemic sclerosis (SSC). The expression of GSDM members varied among pulmonary cell types (immune cells, structural cells, and especially epithelial cells) and even across lung diseases. Regarding disease-associated specificities, we found that GSDMC or GSDMD altered significantly in ciliated epithelia of COPD or LUAD, GSDMD in mucous, club, and basal cells of LUAD and GSDMC in mucous epithelia of para-tumor tissue, as compared with the corresponding epithelia of other diseases. The phenomic specificity of GSDM in lung cancer subtypes was noticed by comparing with 15 non-pulmonary cancers and para-cancer samples. GSDM family gene expression changes were also observed in different lung epithelial cell lines (e.g., HBE, A549, H1299, SPC-1, or H460) in responses to external challenges, including lipopolysaccharide (LPS), lysophosphatidylcholine (lysoPC), cigarette smoking extract (CSE), cholesterol, and AR2 inhibitor at various doses or durations. GSDMA is rarely expressed in those cell lines, while GSDMB and GSDMC are significantly upregulated in human lung epithelia. Our data indicated that the heterogeneity of GSDM member expression exists at different cells, pathologic conditions, challenges, probably dependent upon cell biological phenomes, functions, and behaviors, upon cellular responses to external changes, and the nature and severity of lung disease. Thus, the deep exploration of GSDM phenomes may provide new insights into understanding the single-cell roles in the tissue, regulatory roles of the GSDM family in the pathogenesis, and potential values of biomarker identification and development.
Collapse
Affiliation(s)
- Xuanqi Liu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
- Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai, China
| | - Linlin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Bijun Zhu
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
- Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai, China
| | - Yifei Liu
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Liyang Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Jiayun Hou
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
| | - Mengjia Qian
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China
| | - Nannan Zheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Yiming Zeng
- Center of Molecular Diagnosis and Therapy, The Second Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Chengshui Chen
- Quzhou Hospital of Wenzhou Medical University, Quzhou, Zhejiang Province, China.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, China.
- Shanghai Institute of Clinical Bioinformatics, Shanghai, China.
- Shanghai Engineering Research for AI Technology for Cardiopulmonary Diseases, Shanghai, China.
| |
Collapse
|
8
|
Kapellos TS, Conlon TM, Yildirim AÖ, Lehmann M. The impact of the immune system on lung injury and regeneration in COPD. Eur Respir J 2023; 62:2300589. [PMID: 37652569 DOI: 10.1183/13993003.00589-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
COPD is a devastating respiratory condition that manifests via persistent inflammation, emphysema development and small airway remodelling. Lung regeneration is defined as the ability of the lung to repair itself after injury by the proliferation and differentiation of progenitor cell populations, and becomes impaired in the COPD lung as a consequence of cell intrinsic epithelial stem cell defects and signals from the micro-environment. Although the loss of structural integrity and lung regenerative capacity are critical for disease progression, our understanding of the cellular players and molecular pathways that hamper regeneration in COPD remains limited. Intriguingly, despite being a key driver of COPD pathogenesis, the role of the immune system in regulating lung regenerative mechanisms is understudied. In this review, we summarise recent evidence on the contribution of immune cells to lung injury and regeneration. We focus on four main axes: 1) the mechanisms via which myeloid cells cause alveolar degradation; 2) the formation of tertiary lymphoid structures and the production of autoreactive antibodies; 3) the consequences of inefficient apoptotic cell removal; and 4) the effects of innate and adaptive immune cell signalling on alveolar epithelial proliferation and differentiation. We finally provide insight on how recent technological advances in omics technologies and human ex vivo lung models can delineate immune cell-epithelium cross-talk and expedite precision pro-regenerative approaches toward reprogramming the alveolar immune niche to treat COPD.
Collapse
Affiliation(s)
- Theodore S Kapellos
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, University Hospital, Ludwig Maximilians University of Munich, Munich, Germany
| | - Mareike Lehmann
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute for Lung Research, Philipps University of Marburg, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Germany
| |
Collapse
|
9
|
Nizamoglu M, Joglekar MM, Almeida CR, Larsson Callerfelt AK, Dupin I, Guenat OT, Henrot P, van Os L, Otero J, Elowsson L, Farre R, Burgess JK. Innovative three-dimensional models for understanding mechanisms underlying lung diseases: powerful tools for translational research. Eur Respir Rev 2023; 32:230042. [PMID: 37495250 PMCID: PMC10369168 DOI: 10.1183/16000617.0042-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/04/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic lung diseases result from alteration and/or destruction of lung tissue, inevitably causing decreased breathing capacity and quality of life for patients. While animal models have paved the way for our understanding of pathobiology and the development of therapeutic strategies for disease management, their translational capacity is limited. There is, therefore, a well-recognised need for innovative in vitro models to reflect chronic lung diseases, which will facilitate mechanism investigation and the advancement of new treatment strategies. In the last decades, lungs have been modelled in healthy and diseased conditions using precision-cut lung slices, organoids, extracellular matrix-derived hydrogels and lung-on-chip systems. These three-dimensional models together provide a wide spectrum of applicability and mimicry of the lung microenvironment. While each system has its own limitations, their advantages over traditional two-dimensional culture systems, or even over animal models, increases the value of in vitro models. Generating new and advanced models with increased translational capacity will not only benefit our understanding of the pathobiology of lung diseases but should also shorten the timelines required for discovery and generation of new therapeutics. This article summarises and provides an outline of the European Respiratory Society research seminar "Innovative 3D models for understanding mechanisms underlying lung diseases: powerful tools for translational research", held in Lisbon, Portugal, in April 2022. Current in vitro models developed for recapitulating healthy and diseased lungs are outlined and discussed with respect to the challenges associated with them, efforts to develop best practices for model generation, characterisation and utilisation of models and state-of-the-art translational potential.
Collapse
Affiliation(s)
- Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Mugdha M Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Catarina R Almeida
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | | | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Olivier T Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- Service d'exploration fonctionnelle respiratoire, CHU de Bordeaux, Pessac, France
| | - Lisette van Os
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ramon Farre
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, The Netherlands
| |
Collapse
|
10
|
Khedoe PPSJ, van Schadewijk WAAM, Schwiening M, Ng-Blichfeldt JP, Marciniak SJ, Stolk J, Gosens R, Hiemstra PS. Cigarette smoke restricts the ability of mesenchymal cells to support lung epithelial organoid formation. Front Cell Dev Biol 2023; 11:1165581. [PMID: 37795260 PMCID: PMC10546195 DOI: 10.3389/fcell.2023.1165581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Adequate lung epithelial repair relies on supportive interactions within the epithelial niche, including interactions with WNT-responsive fibroblasts. In fibroblasts from patients with chronic obstructive pulmonary disease (COPD) or upon in vitro cigarette smoke exposure, Wnt/β-catenin signalling is distorted, which may affect interactions between epithelial cells and fibroblasts resulting in inadequate lung repair. We hypothesized that cigarette smoke (CS), the main risk factor for COPD, interferes with Wnt/β-catenin signalling in fibroblasts through induction of cellular stress responses, including oxidative- and endoplasmic reticulum (ER) stress, and thereby alters epithelial repair support potential. Therefore, we assessed the effect of CS-exposure and the ER stress inducer Thapsigargin (Tg) on Wnt/β-catenin signalling activation in MRC-5 fibroblasts, and on their ability to support lung epithelial organoid formation. Exposure of MRC-5 cells for 15 min with 5 AU/mL CS extract (CSE), and subsequent 6 h incubation induced oxidative stress (HMOX1). Whereas stimulation with 100 nM Tg increased markers of both the integrated stress response (ISR - GADD34/PPP1R15A, CHOP) and the unfolded protein response (UPR - XBP1spl, GADD34/PPP1R15A, CHOP and HSPA5/BIP), CSE only induced GADD34/PPP1R15A expression. Strikingly, although treatment of MRC-5 cells with the Wnt activator CHIR99021 upregulated the Wnt/β-catenin target gene AXIN2, this response was diminished upon CSE or Tg pre-exposure, which was confirmed using a Wnt-reporter. Furthermore, pre-exposure of MRC-5 cells to CSE or Tg, restricted their ability to support organoid formation upon co-culture with murine pulmonary EpCam+ cells in Matrigel at day 14. This restriction was alleviated by pre-treatment with CHIR99021. We conclude that exposure of MRC-5 cells to CSE increases oxidative stress, GADD34/PPP1R15A expression and impairs their ability to support organoid formation. This inhibitory effect may be restored by activating the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- P. P. S. J. Khedoe
- Department of Pulmonology, Leiden University Medical Centre, Leiden, Netherlands
| | | | - M. Schwiening
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - J. P. Ng-Blichfeldt
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - S. J. Marciniak
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - J. Stolk
- Department of Pulmonology, Leiden University Medical Centre, Leiden, Netherlands
| | - R. Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - P. S. Hiemstra
- Department of Pulmonology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
11
|
Wisman M, Nizamoglu M, Noordhoek JA, Timens W, Burgess JK, Heijink IH. Dysregulated cross-talk between alveolar epithelial cells and stromal cells in idiopathic pulmonary fibrosis reduces epithelial regenerative capacity. Front Med (Lausanne) 2023; 10:1182368. [PMID: 37621459 PMCID: PMC10446880 DOI: 10.3389/fmed.2023.1182368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
In idiopathic pulmonary fibrosis (IPF) constant epithelial micro-injury and aberrant interactions within the stromal micro-environment lead to abnormal alveolar repair and fibrosis. We hypothesized that alveolar epithelial regenerative responses in IPF are impaired due to disturbed crosstalk between epithelial cells and their stromal niche. We established organoid cultures from unfractionated suspensions and isolated EpCAM+ cells from distal lung tissue of patients with and without IPF. We observed significantly more organoids being formed from unfractionated suspensions compared to isolated EpCAM+ cell cultures, indicating the presence of supportive cells in the unfractionated suspensions. Importantly, lower organoid numbers were observed in unfractionated cultures from IPF lungs compared to non-IPF lungs. This difference was not found when comparing organoid formation from isolated EpCAM+ cells alone between IPF and non-IPF groups, suggesting that crosstalk between the supportive population and epithelial cells is impaired in lungs from IPF patients. Additionally, organoids grown from IPF lung-derived cells were larger in size compared to those from non-IPF lungs in both unfractionated and EpCAM+ cultures, indicating an intrinsic abnormality in epithelial progenitors from IPF lungs. Together, our observations suggest that dysregulated crosstalk between alveolar progenitor cells and the stromal niche affects the regenerative capacity, potentially contributing to alveolar impairment in IPF.
Collapse
Affiliation(s)
- Marissa Wisman
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Jacobien A. Noordhoek
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
| | - Janette K. Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, Netherlands
| |
Collapse
|
12
|
Reza AA, Kohram F, Reza HA, Kalin TR, Kannan PS, Zacharias WJ, Kalinichenko VV. FOXF1 Regulates Alveolar Epithelial Morphogenesis through Transcriptional Activation of Mesenchymal WNT5A. Am J Respir Cell Mol Biol 2023; 68:430-443. [PMID: 36542853 PMCID: PMC10112422 DOI: 10.1165/rcmb.2022-0191oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Mutations in the FOXF1 (forkhead box F1) gene, encoding the mesenchymal FOX (forkhead box) transcription factor, are linked to alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a severe congenital disorder associated with the loss of alveolar capillaries and lung hypoplasia. Although proangiogenic functions of FOXF1 have been extensively studied, the role of FOXF1 in mesenchymal-epithelial signaling during lung development remains uncharacterized. Herein, we used murine lung organoids to demonstrate that the S52F FOXF1 mutation (found in patients with ACDMPV) stimulates canonical WNT/β-catenin signaling in type 2 alveolar epithelial cells (AEC2s), leading to increased proliferation of AEC2s and decreased differentiation of AEC2s into type 1 alveolar epithelial cells (AEC1s). Alveolar organoids containing Foxf1WT/S52F lung fibroblasts and wild-type epithelial cells grew faster on Matrigel and exhibited AEC2 hyperplasia. AEC2 hyperplasia and loss of AEC1s were found in the lungs of Foxf1WT/S52F embryos, a mouse model of ACDMPV. Activation of canonical WNT/β-catenin signaling in AEC2s of lung organoids and Foxf1WT/S52F mice was associated with decreased expression of noncanonical WNT5A (Wnt family member 5A) ligand in lung fibroblasts. Mechanistically, FOXF1 directly activates the Wnt5a gene transcription through an evolutionarily conserved +6320/+6326 region located in the first intron of the Wnt5a gene. Site-directed mutagenesis of the +6320/+6326 region prevented the transcriptional activation of the Wnt5a enhancer by FOXF1. Treatment with exogenous WNT5A ligand inhibited the effects of the S52F FOXF1 mutation on canonical WNT/β-catenin signaling in alveolar organoids, preventing aberrant AEC2 expansion and restoring differentiation of AEC1s. Activation of either FOXF1 or WNT5A may provide an attractive strategy to improve lung function in patients with ACDMPV.
Collapse
Affiliation(s)
| | | | | | | | - Paranthaman S. Kannan
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio; and
| | - William J. Zacharias
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio; and
| | - Vladimir V. Kalinichenko
- Center for Lung Regeneration Medicine
- Division of Developmental Biology, and
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio; and
| |
Collapse
|
13
|
Ciminieri C, Woest ME, Reynaert NL, Heijink IH, Wardenaar R, Spierings DCJ, Brandsma CA, Königshoff M, Gosens R. IL-1β Induces a Proinflammatory Fibroblast Microenvironment that Impairs Lung Progenitors' Function. Am J Respir Cell Mol Biol 2023; 68:444-455. [PMID: 36608844 PMCID: PMC12042164 DOI: 10.1165/rcmb.2022-0209oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 01/06/2023] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a persistent inflammatory state in the lungs and defective tissue repair. Although the inflammatory response in patients with COPD is well characterized and known to be exaggerated during exacerbations, its contribution to lung injury and abnormal repair is still unclear. In this study, we aimed to investigate how the inflammatory microenvironment affects the epithelial progenitors and their supporting mesenchymal niche cells involved in tissue repair of the distal lung. We focused on IL-1β, a key inflammatory mediator that is increased during exacerbations of COPD, and used an organoid model of lung epithelial cells and fibroblasts to assess the effect of IL-1β treatment on these cells' transcriptome and secreted factors. Whereas direct treatment of the lung organoids with IL-1β promoted organoid growth, this switched toward inhibition when it was added as fibroblast pretreatment followed by organoid treatment. We then investigated the IL-1β-driven mechanisms in the fibroblasts and found an inflammatory response related to (C-X-C motif) ligand (CXCL) chemokines; we confirmed that these chemokines were responsible for the impaired organoid growth and found that targeting their C-X-C chemokine receptors 1/2 (CXCR1/2) receptors or the IL-1β intracellular signaling reduced the proinflammatory response and restored organoid growth. These data demonstrate that IL-1β alters the fibroblasts' state by promoting a distinct inflammatory response, switching their supportive function on epithelial progenitors toward an inhibitory one in an organoid assay. These results imply that chronic inflammation functions as a shift toward inhibition of repair, thereby contributing to chronic inflammatory diseases like COPD.
Collapse
Affiliation(s)
- Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| | - Manon E Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
- Aquilo BV, Groningen, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht University Medical Center, Maastricht, The Netherlands; and
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
- Groningen Department of Pulmonary Diseases, and
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
| | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| |
Collapse
|
14
|
Dada LA, Welch LC, Magnani ND, Ren Z, Han H, Brazee PL, Celli D, Flozak AS, Weng A, Herrerias MM, Kryvenko V, Vadász I, Runyan CE, Abdala-Valencia H, Shigemura M, Casalino-Matsuda SM, Misharin AV, Budinger GS, Gottardi CJ, Sznajder JI. Hypercapnia alters stroma-derived Wnt production to limit β-catenin signaling and proliferation in AT2 cells. JCI Insight 2023; 8:e159331. [PMID: 36626234 PMCID: PMC9977495 DOI: 10.1172/jci.insight.159331] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Persistent symptoms and radiographic abnormalities suggestive of failed lung repair are among the most common symptoms in patients with COVID-19 after hospital discharge. In mechanically ventilated patients with acute respiratory distress syndrome (ARDS) secondary to SARS-CoV-2 pneumonia, low tidal volumes to reduce ventilator-induced lung injury necessarily elevate blood CO2 levels, often leading to hypercapnia. The role of hypercapnia on lung repair after injury is not completely understood. Here - using a mouse model of hypercapnia exposure, cell lineage tracing, spatial transcriptomics, and 3D cultures - we show that hypercapnia limits β-catenin signaling in alveolar type II (AT2) cells, leading to their reduced proliferative capacity. Hypercapnia alters expression of major Wnts in PDGFRα+ fibroblasts from those maintaining AT2 progenitor activity toward those that antagonize β-catenin signaling, thereby limiting progenitor function. Constitutive activation of β-catenin signaling in AT2 cells or treatment of organoid cultures with recombinant WNT3A protein bypasses the inhibitory effects of hypercapnia. Inhibition of AT2 proliferation in patients with hypercapnia may contribute to impaired lung repair after injury, preventing sealing of the epithelial barrier and increasing lung flooding, ventilator dependency, and mortality.
Collapse
Affiliation(s)
- Laura A. Dada
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lynn C. Welch
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Natalia D. Magnani
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ziyou Ren
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hyebin Han
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Patricia L. Brazee
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Diego Celli
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Annette S. Flozak
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anthea Weng
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mariana Maciel Herrerias
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Vitalii Kryvenko
- Justus Liebig University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Department of Internal Medicine, Giessen, Germany
- The Cardio-Pulmonary Institute, Giessen, Germany
| | - István Vadász
- Justus Liebig University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Department of Internal Medicine, Giessen, Germany
- The Cardio-Pulmonary Institute, Giessen, Germany
| | - Constance E. Runyan
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Masahiko Shigemura
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Alexander V. Misharin
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cara J. Gottardi
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jacob I. Sznajder
- Pulmonary and Critical Care Medicine, Northwestern Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Liu D, Xu C, Jiang L, Zhu X. Pulmonary endogenous progenitor stem cell subpopulation: Physiology, pathogenesis, and progress. JOURNAL OF INTENSIVE MEDICINE 2023; 3:38-51. [PMID: 36789358 PMCID: PMC9924023 DOI: 10.1016/j.jointm.2022.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/09/2022] [Accepted: 08/13/2022] [Indexed: 06/18/2023]
Abstract
Lungs are structurally and functionally complex organs consisting of diverse cell types from the proximal to distal axis. They have direct contact with the external environment and are constantly at risk of various injuries. Capable to proliferate and differentiate, pulmonary endogenous progenitor stem cells contribute to the maintenance of lung structure and function both under homeostasis and following injuries. Discovering candidate pulmonary endogenous progenitor stem cell types and underlying regenerative mechanisms provide insights into therapeutic strategy development for lung diseases. In this review, we reveal their compositions, roles in lung disease pathogenesis and injury repair, and the underlying mechanisms. We further underline the advanced progress in research approach and potential therapy for lung regeneration. We also demonstrate the feasibility and prospects of pulmonary endogenous stem cell transplantation for lung disease treatment.
Collapse
Affiliation(s)
- Di Liu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Chufan Xu
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, 800 Xiangyin Road, Shanghai 200433, China
| |
Collapse
|
16
|
Asadi Jozani K, Kouthouridis S, Hirota JA, Zhang B. Next generation preclinical models of lung development, physiology and disease. CAN J CHEM ENG 2022. [DOI: 10.1002/cjce.24581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Kimia Asadi Jozani
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
| | - Sonya Kouthouridis
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| | - Jeremy Alexander Hirota
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Medicine, Division of Respirology McMaster University Hamilton Ontario Canada
- Firestone Institute for Respiratory Health St. Joseph’s Hospital, Hamilton Ontario Canada
| | - Boyang Zhang
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| |
Collapse
|
17
|
Li C, Peinado N, Smith SM, Zhou J, Gao F, Kohbodi G, Zhou B, Thornton ME, Grubbs BH, Lee MK, Bellusci S, Borok Z, Chen YW, Minoo P. Wnt5a Promotes AT1 and Represses AT2 Lineage-Specific Gene Expression in a Cell-Context-Dependent Manner. Stem Cells 2022; 40:691-703. [PMID: 35429397 PMCID: PMC9332903 DOI: 10.1093/stmcls/sxac031] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022]
Abstract
Lung maturation is not limited to proper structural development but also includes differentiation and functionality of various highly specialized alveolar cell types. Alveolar type 1 (AT1s) cells occupy nearly 95% of the alveolar surface and are critical for establishing efficient gas exchange in the mature lung. AT1 cells arise from progenitors specified during the embryonic stage as well as alveolar epithelial progenitors expressing surfactant protein C (Sftpcpos cells) during postnatal and adult stages. Previously, we found that Wnt5a, a non-canonical Wnt ligand, is required for differentiation of AT1 cells during the saccular phase of lung development. To further investigate the role of Wnt5a in AT1 cell differentiation, we generated and characterized a conditional Wnt5a gain-of-function mouse model. Neonatal Wnt5a gain-of-function disrupted alveologenesis through inhibition of cell proliferation. In this setting Wnt5a downregulated β-catenin-dependent canonical Wnt signaling, repressed AT2 (anti-AT2) and promoted AT1 (pro-AT1) lineage-specific gene expression. In addition, we identified 2 subpopulations of Sftpchigh and Sftpclow alveolar epithelial cells. In Sftpclow cells, Wnt5a exhibits pro-AT1 and anti-AT2 effects, concurrent with inhibition of canonical Wnt signaling. Interestingly, in the Sftpchigh subpopulation, although increasing AT1 lineage-specific gene expression, Wnt5a gain-of-function did not change AT2 gene expression, nor inhibit canonical Wnt signaling. Using primary epithelial cells isolated from human fetal lungs, we demonstrate that this property of Wnt5a is evolutionarily conserved. Wnt5a therefore serves as a selective regulator that ensures proper AT1/AT2 balance in the developing lung.
Collapse
Affiliation(s)
- Changgong Li
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Neil Peinado
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Susan M Smith
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Jing Zhou
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Feng Gao
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - GoleNaz Kohbodi
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Beiyun Zhou
- Hastings Center for Pulmonary Research, USC Keck School of Medicine, Los Angeles, CA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matthew E Thornton
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Brendan H Grubbs
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Matt K Lee
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
| | - Saverio Bellusci
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
- Cardio Pulmonary Institute, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University ofCalifornia San Diego, CA, USA
| | - Ya-Wen Chen
- Hastings Center for Pulmonary Research, USC Keck School of Medicine, Los Angeles, CA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, USC Keck School of Medicine, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Parviz Minoo
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center, USC Keck School of Medicine and Children’s HospitalLos Angeles, CA, USA
- Hastings Center for Pulmonary Research, USC Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
18
|
Wu X, Ciminieri C, Bos IST, Woest ME, D'Ambrosi A, Wardenaar R, Spierings DCJ, Königshoff M, Schmidt M, Kistemaker LEM, Gosens R. Diesel exhaust particles distort lung epithelial progenitors and their fibroblast niche. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119292. [PMID: 35439594 PMCID: PMC11251497 DOI: 10.1016/j.envpol.2022.119292] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 06/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by inflammation and impaired tissue regeneration, and is reported as the fourth leading cause of death worldwide by the Centers for Disease Control and Prevention (CDC). Environmental pollution and specifically motor vehicle emissions are known to play a role in the pathogenesis of COPD, but little is still known about the molecular mechanisms that are altered following diesel exhaust particles (DEP) exposure. Here we used lung organoids derived from co-culture of alveolar epithelial progenitors and fibroblasts to investigate the effect of DEP on the epithelial-mesenchymal signaling niche in the distal lung, which is essential for tissue repair. We found that DEP treatment impaired the number as well as the average diameter of both airway and alveolar type of lung organoids. Bulk RNA-sequencing of re-sorted epithelial cells and fibroblasts following organoid co-culture shows that the Nrf2 pathway, which regulates antioxidants' activity, was upregulated in both cell populations in response to DEP; and WNT/β-catenin signaling, which is essential to promote epithelial repair, was downregulated in DEP-exposed epithelial cells. We show that pharmacological treatment with anti-oxidant agents such as N-acetyl cysteine (NAC) or Mitoquinone mesylate (MitoQ) reversed the effect of DEP on organoids growth. Additionally, a WNT/β-catenin activator (CHIR99021) successfully restored WNT signaling and promoted organoid growth upon DEP exposure. We propose that targeting oxidative stress and specific signaling pathways affected by DEP in the distal lung may represent a strategy to restore tissue repair in COPD.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Manon E Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands
| | - Angela D'Ambrosi
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - René Wardenaar
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713AV, Groningen, the Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713AV, Groningen, the Netherlands
| | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Martina Schmidt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Aquilo BV, Antonius Deusinglaan 1, 9713AV, Groningen, the Netherlands.
| |
Collapse
|
19
|
Zhang T, Zhang C, Zhang J, Lin J, Song D, Zhang P, Liu Y, Chen L, Zhang L. Cadmium impairs zebrafish swim bladder development via ROS mediated inhibition of the Wnt / Hedgehog pathway. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 247:106180. [PMID: 35490551 DOI: 10.1016/j.aquatox.2022.106180] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
The posterior swim bladder is an important organ in teleost fishes, that primarily maintains buoyancy and motility for swimming and survival. In this study, we examined the molecular mechanisms of the toxicity of cadmium (Cd) on the early development of the swim bladder in zebrafish. Embryonic Cd exposure resulted in the non-inflation of the swim bladder when the ambient Cd concentration was greater than or equal to 0.25 mg/L. Cd disturbed surfactant lipid distribution and inhibited the formation of all three tissue layers in the swim bladder. Additionally, excessive Cd down-regulated Wnt (fzd3, nkd1, fzd7 and axin2) and Hedgehog (ihh, shh, ptc1 and ptc2) signaling pathways. Conversely, Wnt signaling activation partially neutralized Cd-induced swim bladder developmental defects. Moreover, ROS scavenger reduced Glutathione (GSH) effectively recovered Cd induced defects in swim bladder and Wnt/Hedgehog signaling. Taken together, our results first revealed that Cd caused swim bladder developmental defects via ROS-mediated inhibition of the Wnt and Hedgehog pathways. These results herein provide important data for future toxicological studies and risk assessments of Cd.
Collapse
Affiliation(s)
- Ting Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Canchuan Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Jin Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, School of life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jiangtian Lin
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Dongdong Song
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Peng Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Yang Liu
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Lizhao Chen
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; University of Chinese Academy of Science, Beijing 100049, China
| | - Li Zhang
- Key Laboratory of Tropical Marine Bio-resourcesand Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China; Institution of South China Sea Ecology and Environmental Engineering, Chinese Academy of Sciences, Guangzhou 510301, China.
| |
Collapse
|
20
|
Baarsma HA, Van der Veen CHTJ, Lobee D, Mones N, Oosterhout E, Cattani-Cavalieri I, Schmidt M. Epithelial 3D-spheroids as a tool to study air pollutant-induced lung pathology. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:185-190. [PMID: 35227934 DOI: 10.1016/j.slasd.2022.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
Cigarette smoke (CS) and air pollutants (AP) activate pathological processes in bronchial epithelial cells resulting in lung function decline which severely impacts human health. Knowledge about the molecular mechanism(s) by which CS and AP induce pathology is limited. Our previous studies in 2D cultures of human bronchial epithelial (BEAS-2B) cells showed that CS exposure activates transforming growth factor-β1 (TGF-β1) release and signaling. Furthermore, CS exposure reduced the expression of E-cadherin, which was prevented by applying a TGF-β1 neutralizing antibody. Exposure of BEAS-2B cells cultured in 2D to diesel exhaust particles (DEP) increased TGF-β1 protein expression and reduced the expression of epithelial cell markers, whereas mesenchymal markers are upregulated. Conventional 2D cell culture may, however, not fully reflect the physiology of bronchial epithelial cells in vivo. To simulate the in vivo situation more closely we cultured the bronchial epithelial cells in a 3D environment in the current study. Treatment of epithelial spheroids with TGF-β resulted in reduced E-cadherin and increased collagen I expression, indicating the activation of epithelial-to-mesenchymal transition (EMT). Similarly, exposure of spheroids to DEP induced and EMT-like phenotype. Collectively, our data indicate AP induces an EMT-like phenotype of BEAS-2B cells in 3D spheroid cultures. This opens new avenues for drug development for the treatment of lung diseases induced by AP. The 3D spheroid cell culture is a novel, innovative and physiologically relevant model for culturing a variety of cells. It is a versatile tool for both high-throughput studies and for identifying molecular mechanisms involved in bronchial epithelial cell (patho)physiology.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland.
| | - Christina H T J Van der Veen
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland
| | - Danique Lobee
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Nienke Mones
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Emily Oosterhout
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland
| |
Collapse
|
21
|
Wu X, Bos IST, Conlon TM, Ansari M, Verschut V, van der Koog L, Verkleij LA, D’Ambrosi A, Matveyenko A, Schiller HB, Königshoff M, Schmidt M, Kistemaker LEM, Yildirim AÖ, Gosens R. A transcriptomics-guided drug target discovery strategy identifies receptor ligands for lung regeneration. SCIENCE ADVANCES 2022; 8:eabj9949. [PMID: 35319981 PMCID: PMC8942365 DOI: 10.1126/sciadv.abj9949] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 12/15/2021] [Indexed: 05/05/2023]
Abstract
Currently, there is no pharmacological treatment targeting defective tissue repair in chronic disease. Here, we used a transcriptomics-guided drug target discovery strategy using gene signatures of smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke, identifying druggable targets expressed in alveolar epithelial progenitors, of which we screened the function in lung organoids. We found several drug targets with regenerative potential, of which EP and IP prostanoid receptor ligands had the most profound therapeutic potential in restoring cigarette smoke-induced defects in alveolar epithelial progenitors in vitro and in vivo. Mechanistically, we found, using single-cell RNA sequencing analysis, that circadian clock and cell cycle/apoptosis signaling pathways were differentially expressed in alveolar epithelial progenitor cells in patients with COPD and in a relevant model of COPD, which was prevented by prostaglandin E2 or prostacyclin mimetics. We conclude that specific targeting of EP and IP receptors offers therapeutic potential for injury to repair in COPD.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - I. Sophie T. Bos
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Thomas M. Conlon
- Institute of Lung Biology and Disease (ILBD)/Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Meshal Ansari
- Institute of Lung Biology and Disease (ILBD)/Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Vicky Verschut
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Aquilo BV, Groningen, Netherlands
| | - Luke van der Koog
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lars A. Verkleij
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Angela D’Ambrosi
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Herbert B. Schiller
- Institute of Lung Biology and Disease (ILBD)/Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | - Martina Schmidt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Loes E. M. Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Aquilo BV, Groningen, Netherlands
| | - Ali Önder Yildirim
- Institute of Lung Biology and Disease (ILBD)/Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Zhou B, Stueve TR, Mihalakakos EA, Miao L, Mullen D, Wang Y, Liu Y, Luo J, Tran E, Siegmund KD, Lynch SK, Ryan AL, Offringa IA, Borok Z, Marconett CN. Comprehensive epigenomic profiling of human alveolar epithelial differentiation identifies key epigenetic states and transcription factor co-regulatory networks for maintenance of distal lung identity. BMC Genomics 2021; 22:906. [PMID: 34922464 PMCID: PMC8684104 DOI: 10.1186/s12864-021-08152-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/05/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Disruption of alveolar epithelial cell (AEC) differentiation is implicated in distal lung diseases such as chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung adenocarcinoma that impact morbidity and mortality worldwide. Elucidating underlying disease pathogenesis requires a mechanistic molecular understanding of AEC differentiation. Previous studies have focused on changes of individual transcription factors, and to date no study has comprehensively characterized the dynamic, global epigenomic alterations that facilitate this critical differentiation process in humans. RESULTS We comprehensively profiled the epigenomic states of human AECs during type 2 to type 1-like cell differentiation, including the methylome and chromatin functional domains, and integrated this with transcriptome-wide RNA expression data. Enhancer regions were drastically altered during AEC differentiation. Transcription factor binding analysis within enhancer regions revealed diverse interactive networks with enrichment for many transcription factors, including NKX2-1 and FOXA family members, as well as transcription factors with less well characterized roles in AEC differentiation, such as members of the MEF2, TEAD, and AP1 families. Additionally, associations among transcription factors changed during differentiation, implicating a complex network of heterotrimeric complex switching in driving differentiation. Integration of AEC enhancer states with the catalog of enhancer elements in the Roadmap Epigenomics Mapping Consortium and Encyclopedia of DNA Elements (ENCODE) revealed that AECs have similar epigenomic structures to other profiled epithelial cell types, including human mammary epithelial cells (HMECs), with NKX2-1 serving as a distinguishing feature of distal lung differentiation. CONCLUSIONS Enhancer regions are hotspots of epigenomic alteration that regulate AEC differentiation. Furthermore, the differentiation process is regulated by dynamic networks of transcription factors acting in concert, rather than individually. These findings provide a roadmap for understanding the relationship between disruption of the epigenetic state during AEC differentiation and development of lung diseases that may be therapeutically amenable.
Collapse
Affiliation(s)
- B Zhou
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - T R Stueve
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - E A Mihalakakos
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - L Miao
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - D Mullen
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - Y Wang
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - Y Liu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - J Luo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - E Tran
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - K D Siegmund
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - S K Lynch
- Department of Engineering, Test Manufacturing Group, MAXIM Integrated Products, Sunnyvale, CA, 95134, USA
| | - A L Ryan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - I A Offringa
- Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - Z Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - C N Marconett
- Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, 90089, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
23
|
Paracrine Regulation of Alveolar Epithelial Damage and Repair Responses by Human Lung-Resident Mesenchymal Stromal Cells. Cells 2021; 10:cells10112860. [PMID: 34831082 PMCID: PMC8616441 DOI: 10.3390/cells10112860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/17/2021] [Accepted: 10/21/2021] [Indexed: 11/17/2022] Open
Abstract
COPD is characterized by irreversible lung tissue damage. We hypothesized that lung-derived mesenchymal stromal cells (LMSCs) reduce alveolar epithelial damage via paracrine processes, and may thus be suitable for cell-based strategies in COPD. We aimed to assess whether COPD-derived LMSCs display abnormalities. LMSCs were isolated from lung tissue of severe COPD patients and non-COPD controls. Effects of LMSC conditioned-medium (CM) on H2O2-induced, electric field- and scratch-injury were studied in A549 and NCI-H441 epithelial cells. In organoid models, LMSCs were co-cultured with NCI-H441 or primary lung cells. Organoid number, size and expression of alveolar type II markers were assessed. Pre-treatment with LMSC-CM significantly attenuated oxidative stress-induced necrosis and accelerated wound repair in A549. Co-culture with LMSCs supported organoid formation in NCI-H441 and primary epithelial cells, resulting in significantly larger organoids with lower type II-marker positivity in the presence of COPD-derived versus control LMSCs. Similar abnormalities developed in organoids from COPD compared to control-derived lung cells, with significantly larger organoids. Collectively, this indicates that LMSCs' secretome attenuates alveolar epithelial injury and supports epithelial repair. Additionally, LMSCs promote generation of alveolar organoids, with abnormalities in the supportive effects of COPD-derived LMCS, reflective of impaired regenerative responses of COPD distal lung cells.
Collapse
|
24
|
Perkins RS, Suthon S, Miranda-Carboni GA, Krum SA. WNT5B in cellular signaling pathways. Semin Cell Dev Biol 2021; 125:11-16. [PMID: 34635443 DOI: 10.1016/j.semcdb.2021.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
The Wnt signaling ligand WNT5B is implicated in various developmental pathways, both in normal and pathological physiology. Most of the research on WNT5B has been associated with expression analysis and disease states, leaving the signaling pathways underexplored. Here, we review the current understandings of WNT5B's regulation of signal transduction, from receptors to downstream mediators and transcription factors. We also describe its roles in β-catenin-dependent and β-catenin-independent (Planar Cell Polarity and Wnt/Ca2+) Wnt signaling.
Collapse
Affiliation(s)
- Rachel S Perkins
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sarocha Suthon
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gustavo A Miranda-Carboni
- Department of Medicine, Division of Hematology and Oncology, University of Tennessee Health Science Center, Memphis, TN, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
25
|
Pouwels SD, Hesse L, Wu X, Allam VSRR, van Oldeniel D, Bhiekharie LJ, Phipps S, Oliver BG, Gosens R, Sukkar MB, Heijink IH. LL-37 and HMGB1 induce alveolar damage and reduce lung tissue regeneration via RAGE. Am J Physiol Lung Cell Mol Physiol 2021; 321:L641-L652. [PMID: 34405719 DOI: 10.1152/ajplung.00138.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The receptor for advanced glycation end-products (RAGE) has been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, it is still unknown whether RAGE directly contributes to alveolar epithelial damage and abnormal repair responses. We hypothesize that RAGE activation not only induces lung tissue damage but also hampers alveolar epithelial repair responses. The effects of the RAGE ligands LL-37 and HMGB1 were examined on airway inflammation and alveolar tissue damage in wild-type and RAGE-deficient mice and on lung damage and repair responses using murine precision cut lung slices (PCLS) and organoids. In addition, their effects were studied on the repair response of human alveolar epithelial A549 cells, using siRNA knockdown of RAGE and treatment with the RAGE inhibitor FPS-ZM1. We observed that intranasal installation of LL-37 and HMGB1 induces RAGE-dependent inflammation and severe alveolar tissue damage in mice within 6 h, with stronger effects in a mouse strain susceptible for emphysema compared with a nonsusceptible strain. In PCLS, RAGE inhibition reduced the recovery from elastase-induced alveolar tissue damage. In organoids, RAGE ligands reduced the organoid-forming efficiency and epithelial differentiation into pneumocyte-organoids. Finally, in A549 cells, we confirmed the role of RAGE in impaired repair responses upon exposure to LL-37. Together, our data indicate that activation of RAGE by its ligands LL-37 and HMGB1 induces acute lung tissue damage and that this impedes alveolar epithelial repair, illustrating the therapeutic potential of RAGE inhibitors for lung tissue repair in emphysema.
Collapse
Affiliation(s)
- Simon D Pouwels
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Laura Hesse
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Xinhui Wu
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Venkata Sita Rama Raju Allam
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Daan van Oldeniel
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Linsey J Bhiekharie
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Brian G Oliver
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Maria B Sukkar
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Irene H Heijink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
26
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
27
|
Organoids in modelling infectious diseases. Drug Discov Today 2021; 27:223-233. [PMID: 34418577 DOI: 10.1016/j.drudis.2021.08.005] [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: 12/03/2020] [Revised: 05/14/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
Approaches based on animal and two-dimensional (2D) cell culture models cannot ensure reliable results in modeling novel pathogens or in drug testing in the short term; therefore, there is rising interest in platforms such as organoids. To develop a toolbox that can be used successfully to overcome current issues in modeling various infections, it is essential to provide a framework of recent achievements in applying organoids. Organoids have been used to study viruses, bacteria, and protists that cause, for example, respiratory, gastrointestinal, and liver diseases. Their future as models of infection will be associated with improvements in system complexity, including abilities to model tissue structure, a dynamic microenvironment, and coinfection. Teaser. Organoids are a flexible tool for modelling viral, bacterial and protist infections. They can provide fast and reliable information on the biology of pathogens and in drug screening, and thus have become essential in combatting emerging infectious diseases.
Collapse
|
28
|
Suthon S, Perkins RS, Bryja V, Miranda-Carboni GA, Krum SA. WNT5B in Physiology and Disease. Front Cell Dev Biol 2021; 9:667581. [PMID: 34017835 PMCID: PMC8129536 DOI: 10.3389/fcell.2021.667581] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
WNT5B, a member of the WNT family of proteins that is closely related to WNT5A, is required for cell migration, cell proliferation, or cell differentiation in many cell types. WNT5B signals through the non-canonical β-catenin-independent signaling pathway and often functions as an antagonist of canonical WNT signaling. Although WNT5B has a high amino acid identity with WNT5A and is often assumed to have similar activities, WNT5B often exhibits unique expression patterns and functions. Here, we describe the distinct effects and mechanisms of WNT5B on development, bone, adipose tissue, cardiac tissue, the nervous system, the mammary gland, the lung and hematopoietic cells, compared to WNT5A. We also highlight aberrances in non-canonical WNT5B signaling contributing to diseases such as osteoarthritis, osteoporosis, obesity, type 2 diabetes mellitus, neuropathology, and chronic diseases associated with aging, as well as various cancers.
Collapse
Affiliation(s)
- Sarocha Suthon
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Rachel S Perkins
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
- Department of Cytokinetics, Institute of Biophysics, Czech Academy of Sciences, Brno, Czechia
| | - Gustavo A Miranda-Carboni
- Division of Hematology and Oncology, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Susan A Krum
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, TN, United States
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
29
|
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
|
30
|
Wu X, Verschut V, Woest ME, Ng-Blichfeldt JP, Matias A, Villetti G, Accetta A, Facchinetti F, Gosens R, Kistemaker LEM. Rho-Kinase 1/2 Inhibition Prevents Transforming Growth Factor-β-Induced Effects on Pulmonary Remodeling and Repair. Front Pharmacol 2021; 11:609509. [PMID: 33551810 PMCID: PMC7855981 DOI: 10.3389/fphar.2020.609509] [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: 09/23/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor (TGF)-β-induced myofibroblast transformation and alterations in mesenchymal-epithelial interactions contribute to chronic lung diseases such as chronic obstructive pulmonary disease (COPD), asthma and pulmonary fibrosis. Rho-associated coiled-coil-forming protein kinase (ROCK) consists as two isoforms, ROCK1 and ROCK2, and both are playing critical roles in many cellular responses to injury. In this study, we aimed to elucidate the differential role of ROCK isoforms on TGF-β signaling in lung fibrosis and repair. For this purpose, we tested the effect of a non-selective ROCK 1 and 2 inhibitor (compound 31) and a selective ROCK2 inhibitor (compound A11) in inhibiting TGF-β-induced remodeling in lung fibroblasts and slices; and dysfunctional epithelial-progenitor interactions in lung organoids. Here, we demonstrated that the inhibition of ROCK1/2 with compound 31 represses TGF-β-driven actin remodeling as well as extracellular matrix deposition in lung fibroblasts and PCLS, whereas selective ROCK2 inhibition with compound A11 did not. Furthermore, the TGF-β induced inhibition of organoid formation was functionally restored in a concentration-dependent manner by both dual ROCK 1 and 2 inhibition and selective ROCK2 inhibition. We conclude that dual pharmacological inhibition of ROCK 1 and 2 counteracts TGF-β induced effects on remodeling and alveolar epithelial progenitor function, suggesting this to be a promising therapeutic approach for respiratory diseases associated with fibrosis and defective lung repair.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Manon E. Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- AQUILO BV, Groningen, Netherlands
| | - John-Poul Ng-Blichfeldt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ana Matias
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gino Villetti
- Corporate Pre-Clinical R and D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | - Alessandro Accetta
- Corporate Pre-Clinical R and D, Chiesi Farmaceutici S.p.A., Parma, Italy
| | | | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Loes E. M. Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- AQUILO BV, Groningen, Netherlands
| |
Collapse
|
31
|
Hu Y, Ciminieri C, Hu Q, Lehmann M, Königshoff M, Gosens R. WNT Signalling in Lung Physiology and Pathology. Handb Exp Pharmacol 2021; 269:305-336. [PMID: 34463851 DOI: 10.1007/164_2021_521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany. .,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
32
|
Brügger MD, Valenta T, Fazilaty H, Hausmann G, Basler K. Distinct populations of crypt-associated fibroblasts act as signaling hubs to control colon homeostasis. PLoS Biol 2020; 18:e3001032. [PMID: 33306673 PMCID: PMC7758045 DOI: 10.1371/journal.pbio.3001032] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/23/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022] Open
Abstract
Despite recent progress in recognizing the importance of mesenchymal cells for the homeostasis of the intestinal system, the current picture of how these cells communicate with the associated epithelial layer remains unclear. To describe the relevant cell populations in an unbiased manner, we carried out a single-cell transcriptome analysis of the adult murine colon, producing a high-quality atlas of matched colonic epithelium and mesenchyme. We identify two crypt-associated colonic fibroblast populations that are demarcated by different strengths of platelet-derived growth factor receptor A (Pdgfra) expression. Crypt-bottom fibroblasts (CBFs), close to the intestinal stem cells, express low levels of Pdgfra and secrete canonical Wnt ligands, Wnt potentiators, and bone morphogenetic protein (Bmp) inhibitors. Crypt-top fibroblasts (CTFs) exhibit high Pdgfra levels and secrete noncanonical Wnts and Bmp ligands. While the Pdgfralow cells maintain intestinal stem cell proliferation, the Pdgfrahigh cells induce differentiation of the epithelial cells. Our findings enhance our understanding of the crosstalk between various colonic epithelial cells and their associated mesenchymal signaling hubs along the crypt axis-placing differential Pdgfra expression levels in the spotlight of intestinal fibroblast identity.
Collapse
Affiliation(s)
| | - Tomas Valenta
- Department of Molecular Life Sciences, University of Zurich, Switzerland
- Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Hassan Fazilaty
- Department of Molecular Life Sciences, University of Zurich, Switzerland
| | - George Hausmann
- Department of Molecular Life Sciences, University of Zurich, Switzerland
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zurich, Switzerland
| |
Collapse
|
33
|
Cheong SS, Akram KM, Matellan C, Kim SY, Gaboriau DCA, Hind M, del Río Hernández AE, Griffiths M, Dean CH. The Planar Polarity Component VANGL2 Is a Key Regulator of Mechanosignaling. Front Cell Dev Biol 2020; 8:577201. [PMID: 33195213 PMCID: PMC7658195 DOI: 10.3389/fcell.2020.577201] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/08/2020] [Indexed: 12/02/2022] Open
Abstract
VANGL2 is a component of the planar cell polarity (PCP) pathway, which regulates tissue polarity and patterning. The Vangl2 Lp mutation causes lung branching defects due to dysfunctional actomyosin-driven morphogenesis. Since the actomyosin network regulates cell mechanics, we speculated that mechanosignaling could be impaired when VANGL2 is disrupted. Here, we used live-imaging of precision-cut lung slices (PCLS) from Vangl2 Lp/+ mice to determine that alveologenesis is attenuated as a result of impaired epithelial cell migration. Vangl2 Lp/+ tracheal epithelial cells (TECs) and alveolar epithelial cells (AECs) exhibited highly disrupted actomyosin networks and focal adhesions (FAs). Functional assessment of cellular forces confirmed impaired traction force generation in Vangl2 Lp/+ TECs. YAP signaling in Vangl2 Lp airway epithelium was reduced, consistent with a role for VANGL2 in mechanotransduction. Furthermore, activation of RhoA signaling restored actomyosin organization in Vangl2 Lp/+ , confirming RhoA as an effector of VANGL2. This study identifies a pivotal role for VANGL2 in mechanosignaling, which underlies the key role of the PCP pathway in tissue morphogenesis.
Collapse
Affiliation(s)
- Sek-Shir Cheong
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Khondoker M. Akram
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Sally Yunsun Kim
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David C. A. Gaboriau
- Facility for Imaging by Light Microscopy, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- National Institute for Health Research, Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mark Griffiths
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Peri-Operative Medicine Department, St Bartholomew’s Hospital, London, United Kingdom
| | - Charlotte H. Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| |
Collapse
|
34
|
Melo-Narváez MC, Stegmayr J, Wagner DE, Lehmann M. Lung regeneration: implications of the diseased niche and ageing. Eur Respir Rev 2020; 29:29/157/200222. [DOI: 10.1183/16000617.0222-2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Most chronic and acute lung diseases have no cure, leaving lung transplantation as the only option. Recent work has improved our understanding of the endogenous regenerative capacity of the lung and has helped identification of different progenitor cell populations, as well as exploration into inducing endogenous regeneration through pharmaceutical or biological therapies. Additionally, alternative approaches that aim at replacing lung progenitor cells and their progeny through cell therapy, or whole lung tissue through bioengineering approaches, have gained increasing attention. Although impressive progress has been made, efforts at regenerating functional lung tissue are still ineffective. Chronic and acute lung diseases are most prevalent in the elderly and alterations in progenitor cells with ageing, along with an increased inflammatory milieu, present major roadblocks for regeneration. Multiple cellular mechanisms, such as cellular senescence and mitochondrial dysfunction, are aberrantly regulated in the aged and diseased lung, which impairs regeneration. Existing as well as new human in vitro models are being developed, improved and adapted in order to study potential mechanisms of lung regeneration in different contexts. This review summarises recent advances in understanding endogenous as well as exogenous regeneration and the development of in vitro models for studying regenerative mechanisms.
Collapse
|
35
|
Canonical WNT pathway is activated in the airway epithelium in chronic obstructive pulmonary disease. EBioMedicine 2020; 61:103034. [PMID: 33045470 PMCID: PMC7559244 DOI: 10.1016/j.ebiom.2020.103034] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a devastating lung disease, mainly due to cigarette smoking, which represents the third cause of mortality worldwide. The mechanisms driving its epithelial salient features remain largely elusive. We aimed to evaluate the activation and the role of the canonical, β-catenin-dependant WNT pathway in the airway epithelium from COPD patients. METHODS The WNT/β-catenin pathway was first assessed by WNT-targeted RNA sequencing of the air/liquid interface-reconstituted bronchial epithelium from COPD and control patients. Airway expression of total and active β-catenin was assessed in lung sections, as well as WNT components in laser-microdissected airway epithelium. Finally, we evaluated the role of WNT at the bronchial epithelial level by modulating the pathway in the reconstituted COPD epithelium. FINDINGS We show that the WNT/β-catenin pathway is upregulated in the COPD airway epithelium as compared with that of non-smokers and control smokers, in targeted RNA-sequencing of in vitro reconstituted airway epithelium, and in situ in lung tissue and laser-microdissected epithelium. Extrinsic activation of this pathway in COPD-derived airway epithelium inhibited epithelial differentiation, polarity and barrier function, and induced TGF-β-related epithelial-to-mesenchymal transition (EMT). Conversely, canonical WNT inhibition increased ciliated cell numbers, epithelial polarity and barrier function, whilst inhibiting EMT, thus reversing COPD features. INTERPRETATION In conclusion, the aberrant reactivation of the canonical WNT pathway in the adult airway epithelium recapitulates the diseased phenotype observed in COPD patients, suggesting that this pathway or its downstream effectors could represent a future therapeutic target. FUNDING This study was supported by the Fondation Mont-Godinne, the FNRS and the WELBIO.
Collapse
|
36
|
Raslan AA, Yoon JK. WNT Signaling in Lung Repair and Regeneration. Mol Cells 2020; 43:774-783. [PMID: 32807748 PMCID: PMC7528681 DOI: 10.14348/molcells.2020.0059] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/06/2020] [Accepted: 07/23/2020] [Indexed: 01/05/2023] Open
Abstract
The lung has a vital function in gas exchange between the blood and the external atmosphere. It also has a critical role in the immune defense against external pathogens and environmental factors. While the lung is classified as a relatively quiescent organ with little homeostatic turnover, it shows robust regenerative capacity in response to injury, mediated by the resident stem/progenitor cells. During regeneration, regionally distinct epithelial cell populations with specific functions are generated from several different types of stem/progenitor cells localized within four histologically distinguished regions: trachea, bronchi, bronchioles, and alveoli. WNT signaling is one of the key signaling pathways involved in regulating many types of stem/progenitor cells in various organs. In addition to its developmental role in the embryonic and fetal lung, WNT signaling is critical for lung homeostasis and regeneration. In this minireview, we summarize and discuss recent advances in the understanding of the role of WNT signaling in lung regeneration with an emphasis on stem/progenitor cells.
Collapse
Affiliation(s)
- Ahmed A. Raslan
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 35, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 35, Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Korea
| |
Collapse
|
37
|
Osei ET, Hackett TL. Epithelial-mesenchymal crosstalk in COPD: An update from in vitro model studies. Int J Biochem Cell Biol 2020; 125:105775. [PMID: 32473924 DOI: 10.1016/j.biocel.2020.105775] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/31/2022]
Abstract
Chronic Obstructive Pulmonary disease (COPD) involves airway inflammation and remodeling leading to small airways disease and emphysema, which results in irreversible airflow obstruction. During lung development, reciprocal interactions between the endoderm and mesoderm (epithelial-mesenchymal trophic unit (EMTU)) are essential for morphogenetic cues that direct cell proliferation, differentiation, and extracellular (ECM) production. In COPD, a significant number of the inflammation and remodeling mediators resemble those released during lung development, which has led to the hypothesis that aberrant activation of the EMTU may occur in the disease. Studies assessing lung epithelial and fibroblast function in COPD, have been primarily focused on monoculture studies. To capture the in vivo environment of the human lung and aid in the understanding of mechanisms and mediators involved in abnormal epithelial-fibroblast communication in COPD, complex co-culture models are required. In this review, we describe the studies that have used co-culture models to assess epithelial-fibroblast interactions and their role in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Emmanuel T Osei
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Lehmann M, Hu Q, Hu Y, Hafner K, Costa R, van den Berg A, Königshoff M. Chronic WNT/β-catenin signaling induces cellular senescence in lung epithelial cells. Cell Signal 2020; 70:109588. [PMID: 32109549 DOI: 10.1016/j.cellsig.2020.109588] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 12/22/2022]
Abstract
The rapid expansion of the elderly population has led to the recent epidemic of age-related diseases, including increased incidence and mortality of chronic lung diseases, such as Idiopathic Pulmonary Fibrosis (IPF). Cellular senescence is a major hallmark of aging and has a higher occurrence in IPF. The lung epithelium represents a major site of tissue injury, cellular senescence and aberrant activity of developmental pathways such as the WNT/β-catenin pathway in IPF. The potential impact of WNT/β-catenin signaling on alveolar epithelial senescence in general as well as in IPF, however, remains elusive. Here, we characterized alveolar epithelial cells of aged mice and assessed the contribution of chronic WNT/β-catenin signaling on alveolar epithelial type (AT) II cell senescence. Whole lungs from old (16-24 months) versus young (3 months) mice had relatively less epithelial (EpCAM+) but more inflammatory (CD45+) cells, as assessed by flow cytometry. Compared to young ATII cells, old ATII cells showed decreased expression of the ATII cell marker Surfactant Protein C along with increased expression of the ATI cell marker Hopx, accompanied by increased WNT/β-catenin activity. Notably, when placed in an organoid assay, old ATII cells exhibited decreased progenitor cell potential. Chronic canonical WNT/β-catenin activation for up to 7 days in primary ATII cells as well as alveolar epithelial cell lines induced a robust cellular senescence, whereas the non-canonical ligand WNT5A was not able to induce cellular senescence. Moreover, chronic WNT3A treatment of precision-cut lung slices (PCLS) further confirmed ATII cell senescence. Simultaneously, chronic but not acute WNT/β-catenin activation induced a profibrotic state with increased expression of the impaired ATII cell marker Keratin 8. These results suggest that chronic WNT/β-catenin activity in the IPF lung contributes to increased ATII cell senescence and reprogramming. In the fibrotic environment, WNT/β-catenin signaling thus might lead to further progenitor cell dysfunction and impaired lung repair.
Collapse
Affiliation(s)
- Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany; Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO 80045, USA.
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany
| | - Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Kathrin Hafner
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany
| | - Rita Costa
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany
| | - Anastasia van den Berg
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Member of the German Center of Lung Research (DZL), Munich 81377, Germany; Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
39
|
WNT5a-ROR Signaling Is Essential for Alveologenesis. Cells 2020; 9:cells9020384. [PMID: 32046118 PMCID: PMC7072327 DOI: 10.3390/cells9020384] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/14/2022] Open
Abstract
WNT5a is a mainly “non-canonical” WNT ligand whose dysregulation is observed in lung diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and asthma. Germline deletion of Wnt5a disrupts embryonic lung development. However, the temporal-specific function of WNT5a remains unknown. In this study, we generated a conditional loss-of-function mouse model (Wnt5aCAG) and examined the specific role of Wnt5a during the saccular and alveolar phases of lung development. The lack of Wnt5a in the saccular phase blocked distal airway expansion and attenuated differentiation of endothelial and alveolar epithelial type I (AT1) cells and myofibroblasts. Postnatal Wnt5a inactivation disrupted alveologenesis, producing a phenotype resembling human bronchopulmonary dysplasia (BPD). Mutant lungs showed hypoalveolization, but endothelial and epithelial differentiation was unaffected. The major impact of Wnt5a inactivation on alveologenesis was on myofibroblast differentiation and migration, with reduced expression of key regulatory genes. These findings were validated in vitro using isolated lung fibroblasts. Conditional inactivation of the WNT5a receptors Ror1 and Ror2 in alveolar myofibroblasts recapitulated the Wnt5aCAG phenotype, demonstrating that myofibroblast defects are the major cause of arrested alveologenesis in Wnt5aCAG lungs. Finally, we show that WNT5a is reduced in human BPD lung samples, indicating the clinical relevance and potential role for WNT5a in pathogenesis of BPD.
Collapse
|
40
|
Fang L, Sun Q, Roth M. Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma. Int J Mol Sci 2020; 21:ijms21030757. [PMID: 31979396 PMCID: PMC7037330 DOI: 10.3390/ijms21030757] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Asthma increases worldwide without any definite reason and patient numbers double every 10 years. Drugs used for asthma therapy relax the muscles and reduce inflammation, but none of them inhibited airway wall remodeling in clinical studies. Airway wall remodeling can either be induced through pro-inflammatory cytokines released by immune cells, or direct binding of IgE to smooth muscle cells, or non-immunological stimuli. Increasing evidence suggests that airway wall remodeling is initiated early in life by epigenetic events that lead to cell type specific pathologies, and modulate the interaction between epithelial and sub-epithelial cells. Animal models are only available for remodeling in allergic asthma, but none for non-allergic asthma. In human asthma, the mechanisms leading to airway wall remodeling are not well understood. In order to improve the understanding of this asthma pathology, the definition of “remodeling” needs to be better specified as it summarizes a wide range of tissue structural changes. Second, it needs to be assessed if specific remodeling patterns occur in specific asthma pheno- or endo-types. Third, the interaction of the immune cells with tissue forming cells needs to be assessed in both directions; e.g., do immune cells always stimulate tissue cells or are inflamed tissue cells calling immune cells to the rescue? This review aims to provide an overview on immunologic and non-immunologic mechanisms controlling airway wall remodeling in asthma.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
| | - Qinzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Michael Roth
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-2337
| |
Collapse
|