1
|
Kortekaas RK, Geillinger-Kästle KE, Fuentes-Mateos R, Schönleber A, van der Koog L, Elferink RAB, Al-Alyan N, Burgess JK, Gosens R. The soluble factor milieu in idiopathic pulmonary fibrosis dysregulates epithelial differentiation. FASEB J 2024; 38:e70077. [PMID: 39370849 DOI: 10.1096/fj.202302405rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
In idiopathic pulmonary fibrosis (IPF), epithelial abnormalities are present including bronchiolization and alveolar cell dysfunction. We hypothesized that the IPF microenvironment disrupts normal epithelial growth and differentiation. We mimicked the soluble factors within an IPF microenvironment using an IPF cocktail (IPFc), composed of nine factors which are increased in IPF lungs (CCL2, IL-1β, IL-4, IL-8, IL-13, IL-33, TGF-β, TNFα, and TSLP). Using IPFc, we asked whether the soluble factor milieu in IPF affects epithelial growth and differentiation and how IPFc compares to TGF-β alone. Epithelial growth and differentiation were studied using mouse lung organoids (primary Epcam+ epithelial cells co-cultured with CCL206 fibroblasts). Organoids exposed to IPFc and TGF-β were re-sorted into epithelial and fibroblast fractions and subjected to RNA sequencing. IPFc did not affect the number of organoids formed. However, pro-surfactant protein C expression was decreased. On these parameters, TGF-β alone had similar effects. However, RNA sequencing of re-sorted organoids revealed that IPFc and TGF-β had distinct effects on both epithelial cells and fibroblasts. IPFc upregulated goblet cell markers, whereas these were inhibited by TGF-β. Although both IPFc and TGF-β increased extracellular matrix gene expression, only TGF-β increased myofibroblast markers. VEGF-C and Wnt signaling were among the most differentially regulated signaling pathways by IPFc versus TGF-β. Interestingly, Wnt pathway activation rescued Sftpc downregulation induced by IPFc. In conclusion, IPFc alters epithelial differentiation in a way that is distinct from TGF-β. Alterations in Wnt signaling contribute to these effects. IPFc may be a more comprehensive representation of the soluble factor microenvironment in IPF.
Collapse
Affiliation(s)
- Rosa K Kortekaas
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kerstin E Geillinger-Kästle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Rocío Fuentes-Mateos
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anita Schönleber
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Luke van der Koog
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Robin A B Elferink
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nakaa Al-Alyan
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Janette K Burgess
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
2
|
Wang J, Wang R, Li Y, Huang J, Liu Y, Wang J, Xian P, Zhang Y, Yang Y, Zhang H, Li J. Lipolysis engages CD36 to promote ZBP1-mediated necroptosis-impairing lung regeneration in COPD. Cell Rep Med 2024; 5:101732. [PMID: 39255796 DOI: 10.1016/j.xcrm.2024.101732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 06/05/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Lung parenchyma destruction represents a severe condition commonly found in chronic obstructive pulmonary disease (COPD), a leading cause of morbidity and mortality worldwide. Promoting lung regeneration is crucial for achieving clinical improvement. However, no therapeutic drugs are approved to improve the regeneration capacity due to incomplete understanding of the underlying pathogenic mechanisms. Here, we identify a positive feedback loop formed between adipose triglyceride lipase (ATGL)-mediated lipolysis and overexpression of CD36 specific to lung epithelial cells, contributing to disease progression. Genetic deletion of CD36 in lung epithelial cells and pharmacological inhibition of either ATGL or CD36 effectively reduce COPD pathogenesis and promote lung regeneration in mice. Mechanistically, disruption of the ATGL-CD36 loop rescues Z-DNA binding protein 1 (ZBP1)-induced cell necroptosis and restores WNT/β-catenin signaling. Thus, we uncover a crosstalk between lipolysis and lung epithelial cells, suggesting the regenerative potential for therapeutic intervention by targeting the ATGL-CD36-ZBP1 axis in COPD.
Collapse
Affiliation(s)
- Jiazhen Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ru Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yicun Li
- Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jiahui Huang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiayi Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Xian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuanhang Zhang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanmei Yang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haojian Zhang
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
3
|
Thangam T, Parthasarathy K, Supraja K, Haribalaji V, Sounderrajan V, Rao SS, Jayaraj S. Lung Organoids: Systematic Review of Recent Advancements and its Future Perspectives. Tissue Eng Regen Med 2024; 21:653-671. [PMID: 38466362 PMCID: PMC11187038 DOI: 10.1007/s13770-024-00628-2] [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: 07/25/2023] [Revised: 01/06/2024] [Accepted: 01/23/2024] [Indexed: 03/13/2024] Open
Abstract
Organoids are essentially an in vitro (lab-grown) three-dimensional tissue culture system model that meticulously replicates the structure and physiology of human organs. A few of the present applications of organoids are in the basic biological research area, molecular medicine and pharmaceutical drug testing. Organoids are crucial in connecting the gap between animal models and human clinical trials during the drug discovery process, which significantly lowers the time duration and cost associated with each stage of testing. Likewise, they can be used to understand cell-to-cell interactions, a crucial aspect of tissue biology and regeneration, and to model disease pathogenesis at various stages of the disease. Lung organoids can be utilized to explore numerous pathophysiological activities of a lung since they share similarities with its function. Researchers have been trying to recreate the complex nature of the lung by developing various "Lung organoids" models.This article is a systematic review of various developments of lung organoids and their potential progenitors. It also covers the in-depth applications of lung organoids for the advancement of translational research. The review discusses the methodologies to establish different types of lung organoids for studying the regenerative capability of the respiratory system and comprehending various respiratory diseases.Respiratory diseases are among the most common worldwide, and the growing burden must be addressed instantaneously. Lung organoids along with diverse bio-engineering tools and technologies will serve as a novel model for studying the pathophysiology of various respiratory diseases and for drug screening purposes.
Collapse
Affiliation(s)
- T Thangam
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Krupakar Parthasarathy
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India.
| | - K Supraja
- Medway Hospitals, No 2/26, 1st Main Road, Kodambakkam, Chennai, Tamil Nadu, 600024, India
| | - V Haribalaji
- VivagenDx, No. 28, Venkateswara Nagar, 100 Feet Bypass Road, Velachery, Chennai, Tamil Nadu, 600042, India
| | - Vignesh Sounderrajan
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Sudhanarayani S Rao
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Sakthivel Jayaraj
- Centre for Drug Discovery and Development, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| |
Collapse
|
4
|
Zuo X, Wang X, Ma T, Chen S, Cao P, Cheng H, Yang N, Han X, Gao W, Liu X, Sun Y. TNFRSF19 within the 13q12.12 Risk Locus Functions as a Lung Cancer Suppressor by Binding Wnt3a to Inhibit Wnt/β-Catenin Signaling. Mol Cancer Res 2024; 22:227-239. [PMID: 38047807 DOI: 10.1158/1541-7786.mcr-23-0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 10/12/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Cancer risk loci provide special clues for uncovering pathogenesis of cancers. The TNFRSF19 gene located within the 13q12.12 lung cancer risk locus encodes TNF receptor superfamily member 19 (TNFRSF19) protein and has been proved to be a key target gene of a lung tissue-specific tumor suppressive enhancer, but its functional role in lung cancer pathogenesis remains to be elucidated. Here we showed that the TNFRSF19 gene could protect human bronchial epithelial Beas-2B cells from pulmonary carcinogen nicotine-derived nitrosamine ketone (NNK)-induced malignant transformation. Knockout of the TNFRSF19 significantly increased NNK-induced colony formation rate on soft agar. Moreover, TNFRSF19 expression was significantly reduced in lung cancer tissues and cell lines. Restoration of TNFRSF19 expression in A549 lung cancer cell line dramatically suppressed the tumor formation in xenograft mouse model. Interestingly, the TNFRSF19 protein that is an orphan membrane receptor could compete with LRP6 to bind Wnt3a, thereby inhibiting the Wnt/β-catenin signaling pathway that is required for NNK-induced malignant transformation as indicated by protein pulldown, site mutation, and fluorescence energy resonance transfer experiments. Knockout of the TNFRSF19 enhanced LRP6-Wnt3a interaction, promoting β-catenin nucleus translocation and the downstream target gene expression, and thus sensitized the cells to NNK carcinogen. In conclusion, our study demonstrated that the TNFRSF19 inhibited lung cancer carcinogenesis by competing with LRP6 to combine with Wnt3a to inhibit the Wnt/β-catenin signaling pathway. IMPLICATIONS These findings revealed a novel anti-lung cancer mechanism, highlighting the special significance of TNFRSF19 gene within the 13q12.12 risk locus in lung cancer pathogenesis.
Collapse
Affiliation(s)
- Xianglin Zuo
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Xuchun Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Tingzheng Ma
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Shuhan Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Pingping Cao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - He Cheng
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Nan Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
| | - Wei Gao
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Xiaoyu Liu
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
| | - Yujie Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, P.R. China
- Department of Cell Biology, Nanjing Medical University, Nanjing, P.R. China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
5
|
Song S, van Dijk F, Vasse GF, Liu Q, Gosselink IF, Weltjens E, Remels AHV, de Jager MH, Bos S, Li C, Stoeger T, Rehberg M, Kutschke D, van Eck GWA, Wu X, Willems SH, Boom DHA, Kooter IM, Spierings D, Wardenaar R, Cole M, Nawijn MC, Salvati A, Gosens R, Melgert BN. Inhalable Textile Microplastic Fibers Impair Airway Epithelial Differentiation. Am J Respir Crit Care Med 2024; 209:427-443. [PMID: 37971785 DOI: 10.1164/rccm.202211-2099oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Rationale: Microplastics are a pressing global concern, and inhalation of microplastic fibers has been associated with interstitial and bronchial inflammation in flock workers. However, how microplastic fibers affect the lungs is unknown. Objectives: Our aim was to assess the effects of 12 × 31 μm nylon 6,6 (nylon) and 15 × 52 μm polyethylene terephthalate (polyester) textile microplastic fibers on lung epithelial growth and differentiation. Methods: We used human and murine alveolar and airway-type organoids as well as air-liquid interface cultures derived from primary lung epithelial progenitor cells and incubated these with either nylon or polyester fibers or nylon leachate. In addition, mice received one dose of nylon fibers or nylon leachate, and, 7 days later, organoid-forming capacity of isolated epithelial cells was investigated. Measurements and Main Results: We observed that nylon microfibers, more than polyester, inhibited developing airway organoids and not established ones. This effect was mediated by components leaching from nylon. Epithelial cells isolated from mice exposed to nylon fibers or leachate also formed fewer airway organoids, suggesting long-lasting effects of nylon components on epithelial cells. Part of these effects was recapitulated in human air-liquid interface cultures. Transcriptomic analysis revealed upregulation of Hoxa5 after exposure to nylon fibers. Inhibiting Hoxa5 during nylon exposure restored airway organoid formation, confirming Hoxa5's pivotal role in the effects of nylon. Conclusions: These results suggest that components leaching from nylon 6,6 may especially harm developing airways and/or airways undergoing repair, and we strongly encourage characterization in more detail of both the hazard of and the exposure to microplastic fibers.
Collapse
Affiliation(s)
- Shanshan Song
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| | - Fransien van Dijk
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| | - Gwenda F Vasse
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| | - Qiongliang Liu
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, German Center for Lung Research (DZL), Munich, Germany
| | - Irene F Gosselink
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Ellen Weltjens
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Alex H V Remels
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | | | | | - Chenxi Li
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, German Center for Lung Research (DZL), Munich, Germany
| | - Tobias Stoeger
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, German Center for Lung Research (DZL), Munich, Germany
| | - Markus Rehberg
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, German Center for Lung Research (DZL), Munich, Germany
| | - David Kutschke
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, German Center for Lung Research (DZL), Munich, Germany
| | | | - Xinhui Wu
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| | | | - Devin H A Boom
- The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, the Netherlands; and
| | - Ingeborg M Kooter
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
- The Netherlands Organization for Applied Scientific Research (TNO), Utrecht, the Netherlands; and
| | | | - René Wardenaar
- European Research Institute for the Biology of Ageing, and
| | - Matthew Cole
- Plymouth Marine Laboratory, Plymouth, United Kingdom
| | - Martijn C Nawijn
- Groningen Research Institute for Asthma and COPD
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anna Salvati
- Department of Nanomedicine & Drug Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| | - Barbro N Melgert
- Department of Molecular Pharmacology
- Groningen Research Institute for Asthma and COPD
| |
Collapse
|
6
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
7
|
Tesfaigzi Y, Curtis JL, Petrache I, Polverino F, Kheradmand F, Adcock IM, Rennard SI. Does Chronic Obstructive Pulmonary Disease Originate from Different Cell Types? Am J Respir Cell Mol Biol 2023; 69:500-507. [PMID: 37584669 PMCID: PMC10633838 DOI: 10.1165/rcmb.2023-0175ps] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/16/2023] [Indexed: 08/17/2023] Open
Abstract
The onset of chronic obstructive pulmonary disease (COPD) is heterogeneous, and current approaches to define distinct disease phenotypes are lacking. In addition to clinical methodologies, subtyping COPD has also been challenged by the reliance on human lung samples from late-stage diseases. Different COPD phenotypes may be initiated from the susceptibility of different cell types to cigarette smoke, environmental pollution, and infections at early stages that ultimately converge at later stages in airway remodeling and destruction of the alveoli when the disease is diagnosed. This perspective provides discussion points on how studies to date define different cell types of the lung that can initiate COPD pathogenesis, focusing on the susceptibility of macrophages, T and B cells, mast cells, dendritic cells, endothelial cells, and airway epithelial cells. Additional cell types, including fibroblasts, smooth muscle cells, neuronal cells, and other rare cell types not covered here, may also play a role in orchestrating COPD. Here, we discuss current knowledge gaps, such as which cell types drive distinct disease phenotypes and/or stages of the disease and which cells are primarily affected by the genetic variants identified by whole genome-wide association studies. Applying new technologies that interrogate the functional role of a specific cell type or a combination of cell types as well as single-cell transcriptomics and proteomic approaches are creating new opportunities to understand and clarify the pathophysiology and thereby the clinical heterogeneity of COPD.
Collapse
Affiliation(s)
- Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey L. Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Irina Petrache
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
- University of Colorado, Denver, Colorado
| | - Francesca Polverino
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Farrah Kheradmand
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Ian M. Adcock
- Department of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Stephen I. Rennard
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
8
|
Sui J, Boatz JC, Shi J, Hu Q, Li X, Zhang Y, Königshoff M, Kliment CR. Loss of ANT1 Increases Fibrosis and Epithelial Cell Senescence in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2023; 69:556-569. [PMID: 37487137 PMCID: PMC10633847 DOI: 10.1165/rcmb.2022-0315oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 07/24/2023] [Indexed: 07/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by progressive lung scarring and remodeling. Although treatments exist that slow disease progression, IPF is irreversible, and there is no cure. Cellular senescence, a major hallmark of aging, has been implicated in IPF pathogenesis, and mitochondrial dysfunction is increasingly recognized as a driver of senescence. Adenine nucleotide translocases (ANTs) are abundant mitochondrial ATP-ADP transporters critical for regulating cell fate and maintaining mitochondrial function. We sought to determine how alterations in ANTs influence cellular senescence in pulmonary fibrosis. We found that SLC25A4 (solute carrier family 25 member 4) (ANT1) and SLC25A5 (ANT2) expression is reduced in the lungs of patients with IPF, particularly within alveolar type II (AT2) cells, by single-cell RNA sequencing and tissue staining. Loss of ANT1 by siRNA in lung epithelial cells resulted in increased senescence markers such as β-galactosidase and p21, with a reduction in the ratio of nicotinamide adenine dinucleotide to reduced nicotinamide adenine dinucleotide. Bleomycin-treated ANT1 knockdown cells also had increased senescence markers compared with bleomycin-treated control cells. Loss of ANT1 in AT2 cells resulted in a reduction in alveolar organoid growth, with an increase in p21 by staining. Global loss of ANT1 resulted in worse lung fibrosis and increased senescence in the bleomycin- and asbestos-induced mouse models of pulmonary fibrosis. In summary, loss of ANT1 contributes to IPF pathogenesis through mitochondrial dysfunction, increased senescence, and decreased regenerative capacity of AT2 cells, resulting in enhanced lung fibrosis. Modulation of ANTs presents a new therapeutic avenue that may alter cellular senescence pathways and limit pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin Sui
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer C Boatz
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian Shi
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qianjiang Hu
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoyun Li
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yingze Zhang
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie Königshoff
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Corrine R Kliment
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
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: 1.0] [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
|
Stolz D, Matera MG, Rogliani P, van den Berge M, Papakonstantinou E, Gosens R, Singh D, Hanania N, Cazzola M, Maitland-van der Zee AH, Fregonese L, Mathioudakis AG, Vestbo J, Rukhadze M, Page CP. Current and future developments in the pharmacology of asthma and COPD: ERS seminar, Naples 2022. Breathe (Sheff) 2023; 19:220267. [PMID: 37377851 PMCID: PMC10292790 DOI: 10.1183/20734735.0267-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/28/2023] [Indexed: 06/29/2023] Open
Abstract
Pharmacological management of airway obstructive diseases is a fast-evolving field. Several advances in unravelling disease mechanisms as well as intracellular and molecular pathways of drug action have been accomplished. While the clinical translation and implementation of in vitro results to the bedside remains challenging, advances in comprehending the mechanisms of respiratory medication are expected to assist clinicians and scientists in identifying meaningful read-outs and designing clinical studies. This European Respiratory Society Research Seminar, held in Naples, Italy, 5-6 May 2022, focused on current and future developments of the drugs used to treat asthma and COPD; on mechanisms of drug action, steroid resistance, comorbidities and drug interactions; on prognostic and therapeutic biomarkers; on developing novel drug targets based on tissue remodelling and regeneration; and on pharmacogenomics and emerging biosimilars. Related European Medicines Agency regulations are also discussed, as well as the seminar's position on the above aspects.
Collapse
Affiliation(s)
- Daiana Stolz
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, and Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD, and Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | | | | | - Alexander G. Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Maia Rukhadze
- Center of Allergy and Immunology, Teaching University Geomedi LLC, Tbilisi, Georgia
| | - Clive P. Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
12
|
Gao L, Sun Y, Zhang X, Ma D, Xie A, Wang E, Cheng L, Liu S. Wnt3a-Loaded Extracellular Vesicles Promote Alveolar Epithelial Regeneration after Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206606. [PMID: 37072558 PMCID: PMC10288279 DOI: 10.1002/advs.202206606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Compromised regeneration resulting from the deactivation of Wnt/β-catenin signaling contributes to the progression of chronic obstructive pulmonary disease (COPD) with limited therapeutic options. Extracellular cytokine-induced Wnt-based signaling provides an alternative option for COPD treatment. However, the hydrophobic nature of Wnt proteins limits their purification and use. This study devises a strategy to deliver the membrane-bound wingless-type MMTV integration site family, member 3A (Wnt3a) over a long distance by anchoring it to the surface of extracellular vesicles (EVs). The newly engineered Wnt3aWG EVs are generated by co-expressing Wnt3a with two genes encoding the membrane protein, WLS, and an engineered glypican, GPC6ΔGPI -C1C2. The bioactivity of Wnt3aWG EVs is validated using a TOPFlash assay and a mesoderm differentiation model of human pluripotent stem cells. Wnt3aWG EVs activate Wnt signaling and promote cell growth following human alveolar epithelial cell injury. In an elastase-induced emphysema model, impaired pulmonary function and enlarged airspace are greatly restored by the intravenous delivery of Wnt3aWG EVs. Single-cell RNA sequencing-based analyses further highlight that Wnt3aWG EV-activated regenerative programs are responsible for its beneficial effects. These findings suggest that EV-based Wnt3a delivery represents a novel therapeutic strategy for lung repair and regeneration after injury.
Collapse
Affiliation(s)
- Lei Gao
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Yongping Sun
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Xinye Zhang
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Ding Ma
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - An Xie
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Enyu Wang
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Linzhao Cheng
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Senquan Liu
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| |
Collapse
|
13
|
Liu M, Huo Y, Cheng Y. Mechanistic Regulation of Wnt Pathway-Related Progression of Chronic Obstructive Pulmonary Disease Airway Lesions. Int J Chron Obstruct Pulmon Dis 2023; 18:871-880. [PMID: 37215745 PMCID: PMC10198175 DOI: 10.2147/copd.s391487] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic disease associated with inflammation and structural changes in the airways and lungs, resulting from a combination of genetic and environmental factors. This interaction highlights significant genes in early life, particularly those involved in lung development, such as the Wnt signaling pathway. The Wnt signaling pathway plays an important role in cell homeostasis, and its abnormal activation can lead to the occurrence of related diseases such as asthma, COPD, and lung cancer. Due to the fact that the Wnt pathway is mechanically sensitive, abnormal activation of the Wnt pathway by mechanical stress contributes to the progression of chronic diseases. But in the context of COPD, it has received little attention. In this review, we aim to summarize the important current evidence on mechanical stress through the Wnt pathway in airway inflammation and structural changes in COPD and to provide potential targets for COPD treatment strategies.
Collapse
Affiliation(s)
- Minrong Liu
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510630, People’s Republic of China
| | - Yating Huo
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510630, People’s Republic of China
| | - Yuanxiong Cheng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510630, People’s Republic of China
| |
Collapse
|
14
|
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 DOI: 10.1165/rcmb.2022-0209oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [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
|
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: 1.0] [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
|
Sun L, Zhang M, Jiang J, Liu W, Zhao W, Li F. Neutrophil extracellular traps promote bronchopulmonary dysplasia-like injury in neonatal mice via the WNT/β-catenin pathway. Front Cell Infect Microbiol 2023; 13:1126516. [PMID: 37180448 PMCID: PMC10174450 DOI: 10.3389/fcimb.2023.1126516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is one of the most common and severe chronic diseases in preterm infants. Premature infants are susceptible to BPD due to immature lungs and adverse perinatal episodes of infection, hyperoxia, and mechanical ventilation. Methods Neutrophils are the first line of host defence, and the release of neutrophil extracellular traps (NETs) is an important strategy to immobilize and kill invading microorganisms. This study examined whether NETs were associated with BPD in preterm infants and contributed to hyperoxia-induced lung injury in neonatal mice via the WNT/β-catenin pathway. Results In this study, we found that preterm infants with BPD had higher levels of NETs in their tracheal aspirates than those without BPD. Neonatal mice treated with NETs after birth exhibited BPD-like changes in their lungs. Furthermore, the levels of Aquaporin 5 (AQP5) and surfactant-associated protein C (SPC), which represent alveolar differentiation and development, were significantly lower than those in the controls. The WNT/β-catenin pathway is one of the most well-known signalling pathways involved in lung growth. We found that the expression of the target genes c-MYC, cyclin D, and vascular endothelial growth factor (VEGF) and the important proteins WNT3a and β-catenin significantly decreased. Moreover, heparin, which is a NET inhibitor, attenuated changes in gene and protein expression, thereby attenuating BPD-like changes. Discussion This finding indicates that NETs are associated with BPD and can induce BPD-like changes in neonatal mice via the WNT/β-catenin pathway.
Collapse
Affiliation(s)
- Liujuan Sun
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
| | - Meiyu Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
| | - Jin Jiang
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
| | - Wanjiao Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
| | - Wenhao Zhao
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
| | - Fang Li
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatal Diagnosis and Treatment Centre Children’s Hospital of Chongqing Medical University, ChongQing, China
- National Clinical Research Center for Child Health and Disorders, ChongQing, China
- China International Science and Technology Cooperation base of Child Development and Critical Disorders, ChongQing, China
- Chongqing Key Laboratory of Pediatrics, ChongQing, China
- *Correspondence: Fang Li,
| |
Collapse
|
17
|
Song Y, Wang Z, Jiang J, Piao Y, Bai Q, Piao Q, Li L, Xu C, Liu H, Piao H, Li L, Yan G. miR-181-5p attenuates neutrophilic inflammation in asthma by targeting DEK. Int Immunopharmacol 2022; 112:109243. [PMID: 36115279 DOI: 10.1016/j.intimp.2022.109243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022]
Abstract
We investigated the regulatory role of miR-181b-5p in neutrophilic asthma and its mechanisms by targeting DEK. DEK, matrix metalloproteinase (MMP)-2, and MMP-9 were overexpressed and the miR-181b-5p was decreased in mice with neutrophilic asthma. DEK was a direct target of miR-181b-5p. In mouse model, miR-181b-5p agomir had an inhibitory effect on airway inflammation and remodeling. miR-181b-5p inhibited DEK/p-GSK-3βSer9/β-catenin/MMP-9 pathway activation by regulating Wnt ligands in BEAS-2B and 16HBE cells. The ability of supernatants from human bronchial epithelial cells (hBECs) co-stimulated with CXCL8 (IL-8) and miR-181b-5p to induce NETs was weaker than that of IL-8 alone. Moreover, DEK overexpression led to excessive mitochondrial dysfunction, including DRP1 up-regulation, p-DRP1ser637 and MFN2 down-regulation, mitochondrial membrane potential loss, excessive mtROS generation and mitochondrial incompleteness. Interestingly, all these phenotypes were rescued by Wnt inhibitor DKK-1 and miR-181b-5p agomir. Additionally, inhibition of DRP1 with Mdivi-1 decreased MMP-9 on BEAS-2B cells. Overall, miR-181b-5p could attenuate neutrophilic asthma through inhibition of NETs release, DEK/p-GSK-3βSer9/β-catenin/MMP-9 pathway, DEK/Wnt/DRP1/MMP-9 and mitochondria damage. It may become a new therapeutic target for neutrophilic asthma.
Collapse
Affiliation(s)
- Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Yihua Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Intensive Care Unit, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China
| | - Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Qinji Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Hanye Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, Jilin, PR China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, Jilin, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, Jilin, PR China.
| |
Collapse
|
18
|
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
|
19
|
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: 13] [Impact Index Per Article: 6.5] [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
|
20
|
Lin CR, Bahmed K, Kosmider B. Impaired Alveolar Re-Epithelialization in Pulmonary Emphysema. Cells 2022; 11:2055. [PMID: 35805139 PMCID: PMC9265977 DOI: 10.3390/cells11132055] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/24/2023] Open
Abstract
Alveolar type II (ATII) cells are progenitors in alveoli and can repair the alveolar epithelium after injury. They are intertwined with the microenvironment for alveolar epithelial cell homeostasis and re-epithelialization. A variety of ATII cell niches, transcription factors, mediators, and signaling pathways constitute a specific environment to regulate ATII cell function. Particularly, WNT/β-catenin, YAP/TAZ, NOTCH, TGF-β, and P53 signaling pathways are dynamically involved in ATII cell proliferation and differentiation, although there are still plenty of unknowns regarding the mechanism. However, an imbalance of alveolar cell death and proliferation was observed in patients with pulmonary emphysema, contributing to alveolar wall destruction and impaired gas exchange. Cigarette smoking causes oxidative stress and is the primary cause of this disease development. Aberrant inflammatory and oxidative stress responses result in loss of cell homeostasis and ATII cell dysfunction in emphysema. Here, we discuss the current understanding of alveolar re-epithelialization and altered reparative responses in the pathophysiology of this disease. Current therapeutics and emerging treatments, including cell therapies in clinical trials, are addressed as well.
Collapse
Affiliation(s)
- Chih-Ru Lin
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
| | - Karim Bahmed
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA 19140, USA;
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA;
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
21
|
Wang K, Yu Y, Han R, Wang X, Zhao Y, Tang H, Li G. [Establishment of a culture system for human nasal mucosa organoids with controllable differentiation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:868-877. [PMID: 35790437 DOI: 10.12122/j.issn.1673-4254.2022.06.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish a culture system for human nasal mucosal organoids with controllable differentiation to reproduce the structure and function of the source tissue through staged expansion-differentiation culture. METHODS Fresh samples of surgically resected middle turbinate and nasal polyp tissues were collected, from which the nasal mucosa epithelial cells were isolated by enzymatic digestion and filtration for continuous culture at the air-liquid interface for expansion (EO group) or staged culture for expansion and differentiation (DO group). Immunohistochemical staining was used to characterize the structure, cellular composition and ciliary function of nasal mucosal organoids in the two groups. The secretion function of the differentiated nasal mucosal organoids in DO group was evaluated using PAS staining. RESULTS Both of the two organoid culture systems yielded vacuolar or solid spherical 3D organoids, and their diameters increased progressively with time. On day 16 of culture, more vacuolar organoids occurred in DO group, while more solid spherical organoids were seen in EO group, and the proportion of vacuoles was significantly greater in DO group than in EO group [(54.67±13.26)% vs (21.67±8.57)%, P < 0.05]. Short tandem repeat (STR) test of the nasal mucosal organoids and the source tissue showed a 100% match between them. On day 21 of culture, scanning and transmission electron microscopy of the nasal mucosal organoids identified ultrastructure of cilia in DO group and short villi structure in most of the organoids in EO group. Immunohistochemical staining showed positivity for P63 (basal cells), β-tubulin (ciliated columnar cells), and MUC5AC (goblet cells) in the organoids. Compared with those in EO group, the organoids in DO group showed significantly greater percentages of ciliated cells [(7.95±1.81)% vs (27.04±5.91)%, P < 0.05] and goblet cells [(14.46±0.93)% vs (39.85±5.43)%, P < 0.05) with a similar percentage of basal cells [(56.91±14.12)% vs (53.42±15.77)%, P > 0.05]. The differentiated nasal mucosal organoids in DO group were positively stained for glycogen. CONCLUSION The staged expansion-differentiation culture method allows more stable and prolonged growth of the cultured cells in vitro to produce organoids with controllable differentiation closely resembling the morphological structure and functions (ciliary function and secretory function) of the source tissue.
Collapse
Affiliation(s)
- K Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Yu
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - R Han
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Zhao
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Tang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - G Li
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
22
|
Gao F, Li C, Danopoulos S, Al Alam D, Peinado N, Webster S, Borok Z, Kohbodi GA, Bellusci S, Minoo P. Hedgehog-responsive PDGFRa(+) fibroblasts maintain a unique pool of alveolar epithelial progenitor cells during alveologenesis. Cell Rep 2022; 39:110608. [PMID: 35385750 PMCID: PMC9199394 DOI: 10.1016/j.celrep.2022.110608] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/06/2021] [Accepted: 03/11/2022] [Indexed: 01/08/2023] Open
Abstract
The lung alveolus is lined with alveolar type 1 (AT1) and type 2 (AT2) epithelial cells. During alveologenesis, increasing demand associated with expanding alveolar numbers is met by proliferating progenitor AT2s (pAT2). Little information exists regarding the identity of this population and their niche microenvironment. We show that during alveologenesis, Hedgehog-responsive PDGFRa(+) progenitors (also known as SCMFs) are a source of secreted trophic molecules that maintain a unique pAT2 population. SCMFs are in turn maintained by TGFβ signaling. Compound inactivation of Alk5 TβR2 in SCMFs reduced their numbers and depleted the pAT2 pool without impacting differentiation of daughter cells. In lungs of preterm infants who died with bronchopulmonary dysplasia, PDGFRa is reduced and the number of proliferative AT2s is diminished, indicating that an evolutionarily conserved mechanism governs pAT2 behavior during alveologenesis. SCMFs are a transient cell population, active only during alveologenesis, making them a unique stage-specific niche mesodermal cell type in mammalian organs.
Collapse
Affiliation(s)
- Feng Gao
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Changgong Li
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Neil Peinado
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Sha Webster
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Zea Borok
- Hastings Center for Pulmonary Research, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego School of Medicine, San Diego, CA 92093, USA
| | - GoleNaz Adeli Kohbodi
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Saverio Bellusci
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA; Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), 35390 Giessen, Germany
| | - Parviz Minoo
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA; Hastings Center for Pulmonary Research, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
23
|
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: 8.0] [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
|
24
|
Raslan AA, Oh YJ, Jin YR, Yoon JK. R-Spondin2, a Positive Canonical WNT Signaling Regulator, Controls the Expansion and Differentiation of Distal Lung Epithelial Stem/Progenitor Cells in Mice. Int J Mol Sci 2022; 23:ijms23063089. [PMID: 35328508 PMCID: PMC8954098 DOI: 10.3390/ijms23063089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
The lungs have a remarkable ability to regenerate damaged tissues caused by acute injury. Many lung diseases, especially chronic lung diseases, are associated with a reduced or disrupted regeneration potential of the lungs. Therefore, understanding the underlying mechanisms of the regenerative capacity of the lungs offers the potential to identify novel therapeutic targets for these diseases. R-spondin2, a co-activator of WNT/β-catenin signaling, plays an important role in embryonic murine lung development. However, the role of Rspo2 in adult lung homeostasis and regeneration remains unknown. The aim of this study is to determine Rspo2 function in distal lung stem/progenitor cells and adult lung regeneration. In this study, we found that robust Rspo2 expression was detected in different epithelial cells, including airway club cells and alveolar type 2 (AT2) cells in the adult lungs. However, Rspo2 expression significantly decreased during the first week after naphthalene-induced airway injury and was restored by day 14 post-injury. In ex vivo 3D organoid culture, recombinant RSPO2 promoted the colony formation and differentiation of both club and AT2 cells through the activation of canonical WNT signaling. In contrast, Rspo2 ablation in club and AT2 cells significantly disrupted their expansion capacity in the ex vivo 3D organoid culture. Furthermore, mice lacking Rspo2 showed significant defects in airway regeneration after naphthalene-induced injury. Our results strongly suggest that RSPO2 plays a key role in the adult lung epithelial stem/progenitor cells during homeostasis and regeneration, and therefore, it may be a potential therapeutic target for chronic lung diseases with reduced regenerative capability.
Collapse
Affiliation(s)
- Ahmed A. Raslan
- Department of Integrated Biomedical Science, Graduate School, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
| | - Youn Jeong Oh
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
| | - Yong Ri Jin
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Jeong Kyo Yoon
- Department of Integrated Biomedical Science, Graduate School, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, 25 Bongjeong-ro, Dongnam-gu, Cheonan 31151, Korea;
- Correspondence:
| |
Collapse
|
25
|
Jain S, Durugkar S, Saha P, Gokhale SB, Naidu VGM, Sharma P. Effects of intranasal azithromycin on features of cigarette smoke-induced lung inflammation. Eur J Pharmacol 2022; 915:174467. [PMID: 34478690 DOI: 10.1016/j.ejphar.2021.174467] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/04/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Airflow limitation in chronic obstructive pulmonary disease (COPD) is the result of exaggerated airway fibrosis and obliteration of the small airways due to persistent inflammation, and an impaired anti-oxidant response. EMT has been implicated as an active signalling process in cigarette smoke (CS)-induced lung pathology, and macrolide Azithromycin (AZT) use has gained interest in treating COPD. Here, we tested effectiveness of intra-nasal AZT alone and in combination with dexamethasone (DEX) on CS-induced acute lung inflammation. Human alveolar epithelial cells (A549) were treated with CS extract (CSE) for 48 h, and male Balb/c mice were exposed to CS (3 cigarettes-3 times/day) for 4 days. The effects of AZT alone (0.25 and 1.25 μM, in vitro; 0.5 and 5 mg/kg, in vivo) or in combination with DEX (1 μM, in vitro; 1 mg/kg, in vivo) on CS-induced cellular cytotoxicity, oxidative stress, inflammation, and lung function were assessed. AZT alone and in combination with DEX significantly inhibited the CS (E)-induced expression of mesenchymal protein markers and the regulatory protein β-catenin. Furthermore, AZT by itself or in combination with DEX significantly suppressed CS-induced expression of the proinflammtory cytokines TNFα, IL1β and IL6 and prevented pNFkB. Mechanistically, AZT restored the CS-induced reduction in anti-oxidant transcription factor NRF2 and upregulated HDAC2 levels, thereby repressing inflammatory gene expression. Beneficial effects of AZT functionally translated in improved lung mechanics in vivo. Further preclinical and clinical studies are warranted to fully establish and validate the therapeutic efficacy of AZT as a mono- or combination therapy for the treatment of COPD.
Collapse
Affiliation(s)
- Siddhi Jain
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Sneha Durugkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Pritam Saha
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India
| | - Sharad B Gokhale
- Department of Civil Engineering, Indian Institute of Technology Guwahati, North Amingaon, Guwahati, Assam, 781039, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Sila Village, Changsari, Guwahati, Assam, 781101, India.
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
26
|
The NOTCH3 Downstream Target HEYL Is Required for Efficient Human Airway Basal Cell Differentiation. Cells 2021; 10:cells10113215. [PMID: 34831437 PMCID: PMC8620267 DOI: 10.3390/cells10113215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Basal cells (BCs) are stem/progenitor cells of the mucociliary airway epithelium, and their differentiation is orchestrated by the NOTCH signaling pathway. NOTCH3 receptor signaling regulates BC to club cell differentiation; however, the downstream responses that regulate this process are unknown. Overexpression of the active NOTCH3 intracellular domain (NICD3) in primary human bronchial epithelial cells (HBECs) on in vitro air–liquid interface culture promoted club cell differentiation. Bulk RNA-seq analysis identified 692 NICD3-responsive genes, including the classical NOTCH target HEYL, which increased in response to NICD3 and positively correlated with SCGB1A1 (club cell marker) expression. siRNA knockdown of HEYL decreased tight junction formation and cell proliferation. Further, HEYL knockdown reduced club, goblet and ciliated cell differentiation. In addition, we observed decreased expression of HEYL in HBECs from donors with chronic obstructive pulmonary disease (COPD) vs. normal donors which correlates with the impaired differentiation capacity of COPD cells. Finally, overexpression of HEYL in COPD HBECs promoted differentiation into club, goblet and ciliated cells, suggesting the impaired capacity of COPD cells to generate a normal airway epithelium is a reversible phenotype that can be regulated by HEYL. Overall, our data identify the NOTCH3 downstream target HEYL as a key regulator of airway epithelial differentiation.
Collapse
|
27
|
Liu T, Gonzalez De Los Santos F, Hirsch M, Wu Z, Phan SH. Noncanonical Wnt Signaling Promotes Myofibroblast Differentiation in Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 65:489-499. [PMID: 34107237 PMCID: PMC8641847 DOI: 10.1165/rcmb.2020-0499oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 06/08/2021] [Indexed: 11/24/2022] Open
Abstract
The Wnt/β-catenin pathway initiates a signaling cascade that is critical in cell differentiation and the normal development of multiple organ systems. The reactivation of this pathway has been documented in experimental and human idiopathic pulmonary fibrosis, wherein Wnt/β-catenin activation has been implicated in epithelial-cell repair. Furthermore, the canonical ligand Wnt3a is known to induce myofibroblast differentiation; however, the role of noncanonical Wnt ligands remains unclear. This study showed significantly higher levels of Wnt11 expression in cells from both patients with idiopathic pulmonary fibrosis and bleomycin-treated mice, as well as in TGFβ-treated mouse lung fibroblasts. Moreover, Wnt11 induced myofibroblast differentiation as manifested by increased α-SMA (ACTA2) expression, which was similar to that induced by canonical Wnt3a/β-catenin signaling. Further investigation revealed that Wnt11 induction of α-SMA was associated with the activation of JNK (c-Jun N-terminal kinase)/c-Jun signaling and was inhibited by a JNK inhibitor. The potential importance of this signaling pathway was supported by in vivo evidence showing significantly increased levels of Wnt11 and activated JNK in the lungs of mice with bleomycin-induced pulmonary fibrosis. Interestingly, fibroblasts did not express canonical Wnt3a, but treatment of these cells with exogenous Wnt3a induced endogenous Wnt11 and Wnt5a, resulting in repression of the Wnt3a/β-catenin target gene Axin2. These findings suggested that the noncanonical Wnt induction of myofibroblast differentiation mediated by the JNK/c-Jun pathway might play a significant role in pulmonary fibrosis, in addition to or in synergy with canonical Wnt3a/β-catenin signaling. Moreover, Wnt3a activation of noncanonical Wnt signaling might trigger a switch from canonical to noncanonical Wnt signaling to induce myofibroblast differentiation.
Collapse
Affiliation(s)
| | | | - Mitchell Hirsch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
28
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2021. [DOI: 10.1002/sct3.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
29
|
Lin X, Li Y, Gong L, Yun JH, Xu S, Tesfaigzi Y, Qiao D, Zhou X. Tempo-spatial regulation of the Wnt pathway by FAM13A modulates the stemness of alveolar epithelial progenitors. EBioMedicine 2021; 69:103463. [PMID: 34224973 PMCID: PMC8264115 DOI: 10.1016/j.ebiom.2021.103463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Family with Sequence Similarity 13, Member A (FAM13A) gene has been consistently associated with COPD by Genome-wide association studies (GWAS). Our previous study demonstrated that FAM13A was mainly expressed in the lung epithelial progenitors including Club cells and alveolar type II epithelial (ATII) cells. Fam13a-/- mice were resistant to cigarette smoke (CS)-induced emphysema through promoting β-catenin/Wnt activation. Given the important roles of β-catenin/Wnt activation in alveolar regeneration during injury, it is unclear when and where FAM13A regulates the Wnt pathway, the requisite pathway for alveolar epithelial repair, in vivo during CS exposure in lung epithelial progenitors. METHODS Fam13a+/+ or Fam13a-/- mice were crossed with TCF/Lef:H2B-GFP Wnt-signaling reporter mouse line to indicate β-catenin/Wnt-activated cells labeled with GFP followed by acute (1 month) or chronic (7 months) CS exposure. Fluorescence-activated flow cytometry analysis, immunofluorescence and organoid culture system were performed to identify the β-catenin/Wnt-activated cells in Fam13a+/+ or Fam13a-/- mice exposed to CS. Fam13a;SftpcCreERT2;Rosa26RmTmG mouse line, where GFP labels ATII cells, was generated for alveolar organoid culture followed by analyses of organoid number, immunofluorescence and gene expression. Single cell RNA-seq data from COPD ever smokers and nonsmoker control lungs were further analyzed. FINDINGS We found that FAM13A-deficiency significantly increased Wnt activation mainly in lung epithelial cells. Consistently, after long-term CS exposure in vivo, FAM13A deficiency bestows alveolar epithelial progenitor cells with enhanced proliferation and differentiation in the ex vivo organoid model. Importantly, expression of FAM13A is significantly increased in human COPD-derived ATII cells compared to healthy ATII cells as suggested by single cell RNA-sequencing data. INTERPRETATION Our findings suggest that FAM13A-deficiency promotes the Wnt pathway-mediated ATII cell repair/regeneration, and thereby possibly mitigating CS-induced alveolar destruction. FUND: This project is funded by the National Institutes of Health of United States of America (NIH) grants R01HL127200, R01HL137927, R01HL148667 and R01HL147148 (XZ).
Collapse
Affiliation(s)
- Xin Lin
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yujun Li
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Lu Gong
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeong H Yun
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; The Division of Pulmonary and Critical Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Shuang Xu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yohannes Tesfaigzi
- The Division of Pulmonary and Critical Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dandi Qiao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Bai S, Ye R, Wang C, Sun P, Wang D, Yue Y, Wang H, Wu S, Yu M, Xi S, Zhao L. Identification of Proteomic Signatures in Chronic Obstructive Pulmonary Disease Emphysematous Phenotype. Front Mol Biosci 2021; 8:650604. [PMID: 34277700 PMCID: PMC8280333 DOI: 10.3389/fmolb.2021.650604] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly heterogeneous disease. Emphysematous phenotype is the most common and critical phenotype, which is characterized by progressive lung destruction and poor prognosis. However, the underlying mechanism of this structural damage has not been completely elucidated. A total of 12 patients with COPD emphysematous phenotype (COPD-E) and nine patients with COPD non-emphysematous phenotype (COPD-NE) were enrolled to determine differences in differential abundant protein (DAP) expression between both groups. Quantitative tandem mass tag–based proteomics was performed on lung tissue samples of all patients. A total of 29 and 15 lung tissue samples from patients in COPD-E and COPD-NE groups, respectively, were used as the validation cohort to verify the proteomic analysis results using western blotting. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted for DAPs. A total of 4,343 proteins were identified, of which 25 were upregulated and 11 were downregulated in the COPD-E group. GO and KEGG analyses showed that wound repair and retinol metabolism–related pathways play an essential role in the molecular mechanism of COPD emphysematous phenotype. Three proteins, namely, KRT17, DHRS9, and FMO3, were selected for validation. While KRT17 and DHRS9 were highly expressed in the lung tissue samples of the COPD-E group, FMO3 expression was not significantly different between both groups. In conclusion, KRT17 and DHRS9 are highly expressed in the lung tissue of patients with COPD emphysematous phenotype. Therefore, these proteins might involve in wound healing and retinol metabolism in patients with emphysematous phenotype and can be used as phenotype-specific markers.
Collapse
Affiliation(s)
- Shuang Bai
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Ye
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cuihong Wang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Pengbo Sun
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Di Wang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yong Yue
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huiying Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Si Wu
- Department of Biobank, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Yu
- Department of Biobank, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuhua Xi
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang, China
| | - Li Zhao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Tam A, Leclair P, Li LV, Yang CX, Li X, Witzigmann D, Kulkarni JA, Hackett TL, Dorscheid DR, Singhera GK, Hogg JC, Cullis PR, Sin DD, Lim CJ. FAM13A as potential therapeutic target in modulating TGF-β-induced airway tissue remodeling in COPD. Am J Physiol Lung Cell Mol Physiol 2021; 321:L377-L391. [PMID: 34105356 DOI: 10.1152/ajplung.00477.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genome-wide association studies have shown that a gene variant in the Family with sequence similarity 13, member A (FAM13A) is strongly associated with reduced lung function and the appearance of respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD). A key player in smoking-induced tissue injury and airway remodeling is the transforming growth factor-β1 (TGF-β1). To determine the role of FAM13A in TGF-β1 signaling, FAM13A-/- airway epithelial cells were generated using CRISPR-Cas9, whereas overexpression of FAM13A was achieved using lipid nanoparticles. Wild-type (WT) and FAM13A-/- cells were treated with TGF-β1, followed by gene and/or protein expression analyses. FAM13A-/- cells augmented TGF-β1-induced increase in collagen type 1 (COL1A1), matrix metalloproteinase 2 (MMP2), expression compared with WT cells. This effect was mediated by an increase in β-catenin (CTNNB1) expression in FAM13A-/- cells compared with WT cells after TGF-β1 treatment. FAM13A overexpression was partially protective from TGF-β1-induced COL1A1 expression. Finally, we showed that airway epithelial-specific FAM13A protein expression is significantly increased in patients with severe COPD compared with control nonsmokers, and negatively correlated with lung function. In contrast, β-catenin (CTNNB1), which has previously been linked to be regulated by FAM13A, is decreased in the airway epithelium of smokers with COPD compared with non-COPD subjects. Together, our data showed that FAM13A may be protective from TGF-β1-induced fibrotic response in the airway epithelium via sequestering CTNNB1 from its regulation on downstream targets. Therapeutic increase in FAM13A expression in the airway epithelium of smokers at risk for COPD, and those with mild COPD, may reduce the extent of airway tissue remodeling.
Collapse
Affiliation(s)
- Anthony Tam
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Pascal Leclair
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ling Vicky Li
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chen X Yang
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Xuan Li
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Jayesh A Kulkarni
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Delbert R Dorscheid
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Gurpreet K Singhera
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - James C Hogg
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Costa R, Wagner DE, Doryab A, De Santis MM, Schorpp K, Rothenaigner I, Lehmann M, Baarsma HA, Liu X, Schmid O, Campillos M, Yildirim AÖ, Hadian K, Königshoff M. A drug screen with approved compounds identifies amlexanox as a novel Wnt/β-catenin activator inducing lung epithelial organoid formation. Br J Pharmacol 2021; 178:4026-4041. [PMID: 34089180 PMCID: PMC8965750 DOI: 10.1111/bph.15581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 05/04/2021] [Accepted: 05/19/2021] [Indexed: 11/30/2022] Open
Abstract
Background and Purpose: Emphysema is an incurable disease characterized by loss of lung tissue leading to impaired gas exchange. Wnt/β-catenin signalling is reduced in emphysema, and exogenous activation of the pathway in experimental models in vivo and in human ex vivo lung tissue improves lung function and structure. We sought to identify a pharmaceutical able to activate Wnt/β-catenin signalling and assess its potential to activate lung epithelial cells and repair. Experimental Approach: We screened 1216 human-approved compounds for Wnt/β-catenin signalling activation using luciferase reporter cells and selected candidates based on their computationally predicted protein targets. We further performed confirmatory luciferase reporter and metabolic activity assays. Finally, we studied the regenerative potential in murine adult epithelial cell-derived lung organoids and in vivo using a murine elastase-induced emphysema model. Key Results: The primary screen identified 16 compounds that significantly induced Wnt/β-catenin-dependent luciferase activity. Selected compounds activated Wnt/β-catenin signalling without inducing cell toxicity or proliferation. Two compounds were able to promote organoid formation, which was reversed by pharmacological Wnt/β-catenin inhibition, confirming the Wnt/β-catenin-dependent mechanism of action. Amlexanox was used for in vivo evaluation, and preventive treatment resulted in improved lung function and structure in emphysematous mouse lungs. Moreover, gene expression of Hgf, an important alveolar repair marker, was increased, whereas disease marker Eln was decreased, indicating that amlexanox induces proregenerative signalling in emphysema. Conclusion and Implications: Using a drug screen based on Wnt/β-catenin activity, organoid assays and a murine emphysema model, amlexanox was identified as a novel potential therapeutic agent for emphysema.
Collapse
Affiliation(s)
- Rita Costa
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Virology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Darcy E Wagner
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Experimental Medical Sciences, Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Stem Cell Centre, Lund University, Lund, Sweden
| | - Ali Doryab
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Pulmonary Aerosol Delivery, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Martina M De Santis
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Experimental Medical Sciences, Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Stem Cell Centre, Lund University, Lund, Sweden
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ina Rothenaigner
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Hoeke A Baarsma
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xueping Liu
- Institute of Structural Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Otmar Schmid
- Pulmonary Aerosol Delivery, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Monica Campillos
- Institute of Structural Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ali Önder Yildirim
- Immunopathology of COPD, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Melanie Königshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
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: 32] [Impact Index Per Article: 10.7] [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
|
34
|
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: 4.0] [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
|
35
|
Conlon TM, John-Schuster G, Heide D, Pfister D, Lehmann M, Hu Y, Ertüz Z, Lopez MA, Ansari M, Strunz M, Mayr C, Angelidis I, Ciminieri C, Costa R, Kohlhepp MS, Guillot A, Günes G, Jeridi A, Funk MC, Beroshvili G, Prokosch S, Hetzer J, Verleden SE, Alsafadi H, Lindner M, Burgstaller G, Becker L, Irmler M, Dudek M, Janzen J, Goffin E, Gosens R, Knolle P, Pirotte B, Stoeger T, Beckers J, Wagner D, Singh I, Theis FJ, de Angelis MH, O'Connor T, Tacke F, Boutros M, Dejardin E, Eickelberg O, Schiller HB, Königshoff M, Heikenwalder M, Yildirim AÖ. Inhibition of LTβR signalling activates WNT-induced regeneration in lung. Nature 2020; 588:151-156. [PMID: 33149305 PMCID: PMC7718297 DOI: 10.1038/s41586-020-2882-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 08/19/2020] [Indexed: 01/11/2023]
Abstract
Lymphotoxin β-receptor (LTβR) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures1,2, which are associated with severe chronic inflammatory diseases that span several organ systems3-6. How LTβR signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LTβR blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LTβR ligands in adaptive and innate immune cells, enhanced non-canonical NF-κB signalling, and enriched LTβR target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LTβR signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LTβR signalling dampened epithelial non-canonical activation of NF-κB, reduced TGFβ signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/β-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LTβR signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures1 and inhibition of apoptosis with tissue-regenerative strategies.
Collapse
Affiliation(s)
- Thomas M Conlon
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Gerrit John-Schuster
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Danijela Heide
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Dominik Pfister
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Mareike Lehmann
- Comprehensive Pneumology Center (CPC), Lung Repair and Regeneration Research Unit, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| | - Zeynep Ertüz
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Martin A Lopez
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, Liège, Belgium
| | - Meshal Ansari
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
- Institute of Computional Biology (ICB), Helmholtz Zentrum München, Neuherberg, Germany
| | - Maximilian Strunz
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Christoph Mayr
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Ilias Angelidis
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen, The Netherlands
| | - Rita Costa
- Comprehensive Pneumology Center (CPC), Lung Repair and Regeneration Research Unit, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marlene Sophia Kohlhepp
- Department of Hepatology & Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Gizem Günes
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Aicha Jeridi
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Maja C Funk
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Heidelberg, Germany
| | - Giorgi Beroshvili
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Sandra Prokosch
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Jenny Hetzer
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | | | - Hani Alsafadi
- Comprehensive Pneumology Center (CPC), Lung Repair and Regeneration Research Unit, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Michael Lindner
- Asklepios Fachkliniken Munich-Gauting, Member of the German Center for Lung Research (DZL), Munich, Germany
- Translational Lung Research and CPC-M bioArchive, Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michael Dudek
- Institute of Molecular Immunology & Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Janzen
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
- Emmy Noether Research Group Epigenetic Machineries and Cancer, Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eric Goffin
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen, The Netherlands
| | - Percy Knolle
- Institute of Molecular Immunology & Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernard Pirotte
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Experimental Genetics, Technische Universität München, Freising, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Darcy Wagner
- Comprehensive Pneumology Center (CPC), Lung Repair and Regeneration Research Unit, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Indrabahadur Singh
- Emmy Noether Research Group Epigenetic Machineries and Cancer, Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian J Theis
- Institute of Computional Biology (ICB), Helmholtz Zentrum München, Neuherberg, Germany
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Laboratory of Medicinal Chemistry, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
- Experimental Genetics, Technische Universität München, Freising, Germany
| | - Tracy O'Connor
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signaling and Functional Genomics, Heidelberg, Germany
- Medical Faculty Mannheim & BioQuant, Heidelberg University, Heidelberg, Germany
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, Liège, Belgium
| | - Oliver Eickelberg
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| | - Herbert B Schiller
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center (CPC), Lung Repair and Regeneration Research Unit, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, USA
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Heidelberg, Germany.
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC), Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany.
| |
Collapse
|
36
|
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: 2.3] [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
|
37
|
Wang ZN, Tang XX. New Perspectives on the Aberrant Alveolar Repair of Idiopathic Pulmonary Fibrosis. Front Cell Dev Biol 2020; 8:580026. [PMID: 33117807 PMCID: PMC7561442 DOI: 10.3389/fcell.2020.580026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease of unknown etiology and high mortality. Current therapeutic strategies have limited efficacy and the prognosis remains poor. Based on the histological observations of IPF lung tissues and experimental studies using lung fibrosis animal models, it is gradually accepted that impaired epithelial regeneration after lung injury is a critical mechanism underlying the pathogenesis of pulmonary fibrosis. The central role of AEC2 in this process has been well-elucidated, while the contribution of other lung progenitor/stem cells is less discussed. Recently, increasing studies have identified several non-AEC2 epithelial progenitor/stem cells with great plasticity to transform into mature AECs and reconstitute alveolar epithelium after lung injury. However, why these cells do not function as alternate stem cells to regenerate alveolar epithelium in IPF is still unknown. In this review, we discuss the contribution of lung epithelial progenitor/stem cells in the aberrant alveolar regeneration, and provide a novel perspective on the mechanism of IPF pathogenesis, in which non-AEC2 progenitors may play an essential role.
Collapse
Affiliation(s)
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|