1
|
Palit S, Shrestha AK, Thapa S, L. Grimm S, Coarfa C, Theis F, Simon LM, Shivanna B. Leveraging Integrated RNA Sequencing to Decipher Adrenomedullin's Protective Mechanisms in Experimental Bronchopulmonary Dysplasia. Genes (Basel) 2024; 15:806. [PMID: 38927741 PMCID: PMC11202456 DOI: 10.3390/genes15060806] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly affecting premature infants, with limited therapeutic options and increased long-term consequences. Adrenomedullin (Adm), a proangiogenic peptide hormone, has been found to protect rodents against experimental BPD. This study aims to elucidate the molecular and cellular mechanisms through which Adm influences BPD pathogenesis using a lipopolysaccharide (LPS)-induced model of experimental BPD in mice. Bulk RNA sequencing of Adm-sufficient (wild-type or Adm+/+) and Adm-haplodeficient (Adm+/-) mice lungs, integrated with single-cell RNA sequencing data, revealed distinct gene expression patterns and cell type alterations associated with Adm deficiency and LPS exposure. Notably, computational integration with cell atlas data revealed that Adm-haplodeficient mouse lungs exhibited gene expression signatures characteristic of increased inflammation, natural killer (NK) cell frequency, and decreased endothelial cell and type II pneumocyte frequency. Furthermore, in silico human BPD patient data analysis supported our cell type frequency finding, highlighting elevated NK cells in BPD infants. These results underscore the protective role of Adm in experimental BPD and emphasize that it is a potential therapeutic target for BPD infants with an inflammatory phenotype.
Collapse
Affiliation(s)
- Subarna Palit
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Sandra L. Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabian Theis
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| |
Collapse
|
2
|
Zabihi M, Khadim A, Schäfer TM, Alexopoulos I, Bartkuhn M, El Agha E, Vazquez-Armendariz AI, Herold S. An Optimized Protocol for the Generation of Alveolospheres from Wild-Type Mice. Cells 2024; 13:922. [PMID: 38891054 PMCID: PMC11171706 DOI: 10.3390/cells13110922] [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: 03/04/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Organoid models have become an integral part of the research methodology in the lung field. These systems allow for the study of progenitor and stem cell self-renewal, self-organization, and differentiation. Distinct models of lung organoids mimicking various anatomical regions of mature lungs have emerged in parallel to the increased gain of knowledge regarding epithelial stem and progenitor cell populations and the corresponding mesenchymal cells that populate the in vivo niche. In the distal lung, type 2 alveolar epithelial cells (AEC2s) represent a stem cell population that is engaged in regenerative mechanisms in response to various insults. These cells self-renew and give rise to AEC1s that carry out gas exchange. Multiple experimental protocols allowing the generation of alveolar organoids, or alveolospheres, from murine lungs have been described. Among the drawbacks have been the requirement of transgenic mice allowing the isolation of AEC2s with high viability and purity, and the occasional emergence of bronchiolar and bronchioalveolar organoids. Here, we provide a refined gating strategy and an optimized protocol for the generation of alveolospheres from wild-type mice. Our approach not only overcomes the need for transgenic mice to generate such organoids, but also yields a pure culture of alveolospheres that is devoid of bronchiolar and bronchioalveolar organoids. Our protocol contributes to the standardization of this important research tool.
Collapse
Affiliation(s)
- Mahsa Zabihi
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ali Khadim
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Theresa M. Schäfer
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine, Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany;
| | - Elie El Agha
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ana I. Vazquez-Armendariz
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
- Transdisciplinary Research Area Life and Health, Organoid Biology, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Susanne Herold
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig University Giessen (JLU), 35392 Giessen, Germany; (M.Z.); (A.K.); (T.M.S.); (I.A.)
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| |
Collapse
|
3
|
Kryvenko V, Vadász I. Modeling the Human Alveolar Epithelium: Promises and Challenges. Am J Respir Cell Mol Biol 2024; 70:329-330. [PMID: 38353673 PMCID: PMC11109586 DOI: 10.1165/rcmb.2023-0471ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine Justus Liebig University Giessen, Germany
- Universities of Giessen and Marburg Lung Center Member of the German Center for Lung Research Giessen, Germany
- The Cardio-Pulmonary Institute Giessen, Germany
- Institute for Lung Health Giessen, Germany
| | - István Vadász
- Department of Internal Medicine Justus Liebig University Giessen, Germany
- Universities of Giessen and Marburg Lung Center Member of the German Center for Lung Research Giessen, Germany
- The Cardio-Pulmonary Institute Giessen, Germany
- Institute for Lung Health Giessen, Germany
| |
Collapse
|
4
|
Licciardello M, Traldi C, Cicolini M, Bertana V, Marasso SL, Cocuzza M, Tonda-Turo C, Ciardelli G. A miniaturized multicellular platform to mimic the 3D structure of the alveolar-capillary barrier. Front Bioeng Biotechnol 2024; 12:1346660. [PMID: 38646009 PMCID: PMC11026571 DOI: 10.3389/fbioe.2024.1346660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Several diseases affect the alveoli, and the efficacy of medical treatments and pharmaceutical therapies is hampered by the lack of pre-clinical models able to recreate in vitro the diseases. Microfluidic devices, mimicking the key structural and compositional features of the alveoli, offer several advantages to medium and high-throughput analysis of new candidate therapies. Here, we developed an alveolus-on-a-chip recapitulating the microanatomy of the physiological tissue by including the epithelium, the fibrous interstitial layer and the capillary endothelium. A PDMS device was obtained assembling a top layer and a bottom layer obtained by replica molding. A polycaprolactone/gelatin (PCL-Gel) electrospun membrane was included within the two layers supporting the seeding of 3 cell phenotypes. Epithelial cells were grown on a fibroblast-laden collagen hydrogel located on the top side of the PCL-Gel mats while endothelial cells were seeded on the basolateral side of the membrane. The innovative design of the microfluidic device allows to replicate both cell-cell and cell-extracellular matrix interactions according to the in vivo cell arrangement along with the establishment of physiologically relevant air-liquid interface conditions. Indeed, high cell viability was confirmed for up to 10 days and the formation of a tight endothelial and epithelial barrier was assessed by immunofluorescence assays.
Collapse
Affiliation(s)
- Michela Licciardello
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Cecilia Traldi
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Martina Cicolini
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Valentina Bertana
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Simone Luigi Marasso
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
- CNR-IMEM, National Research Council-Institute of Materials for Electronics and Magnetism, Parma, Italy
| | - Matteo Cocuzza
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Chiara Tonda-Turo
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Gianluca Ciardelli
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
- CNR-IPCF, National Research Council-Institute for Chemical and Physical Processes, Pisa, Italy
| |
Collapse
|
5
|
Basil MC, Alysandratos KD, Kotton DN, Morrisey EE. Lung repair and regeneration: Advanced models and insights into human disease. Cell Stem Cell 2024; 31:439-454. [PMID: 38492572 PMCID: PMC11070171 DOI: 10.1016/j.stem.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
6
|
Watson BE, Miles JA, Moss MA. Human in vitro blood barrier models: architectures and applications. Tissue Barriers 2024; 12:2222628. [PMID: 37339009 PMCID: PMC11042067 DOI: 10.1080/21688370.2023.2222628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Blood barriers serve as key points of transport for essential molecules as well as lines of defense to protect against toxins. In vitro modeling of these barriers is common practice in the study of their physiology and related diseases. This review describes a common method of using an adaptable, low cost, semipermeable, suspended membrane to experimentally model three blood barriers in the human body: the blood-brain barrier (BBB), the gut-blood barrier (GBB), and the air-blood barrier (ABB). The GBB and ABB both protect from the outside environment, while the BBB protects the central nervous system from potential neurotoxic agents in the blood. These barriers share several commonalities, including the formation of tight junctions, polarized cellular monolayers, and circulatory system contact. Cell architectures used to mimic barrier anatomy as well as applications to study function, dysfunction, and response provide an overview of the versatility enabled by these cultural systems.
Collapse
Affiliation(s)
| | - Julia A. Miles
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
| | - Melissa A. Moss
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
- Department of Chemical Engineering, Univ of South Carolina, Columbia, SCUSA
| |
Collapse
|
7
|
Li Y, Prakash YS, Tan Q, Tschumperlin D. Defining signals that promote human alveolar type I differentiation. Am J Physiol Lung Cell Mol Physiol 2024; 326:L409-L418. [PMID: 38349124 PMCID: PMC11281788 DOI: 10.1152/ajplung.00191.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/23/2023] [Accepted: 01/18/2024] [Indexed: 02/29/2024] Open
Abstract
Alveolar type I (ATI) cells cover >95% of the lung's distal surface and facilitate gas exchange through their exceptionally thin shape. ATI cells in vivo are replenished by alveolar type II cell division and differentiation, but a detailed understanding of ATI biology has been hampered by the challenges in direct isolation of these cells due to their fragility and incomplete understanding of the signaling interactions that promote differentiation of ATII to ATI cells. Here, we explored the signals that maintain ATII versus promote ATI fates in three-dimensional (3-D) organoid cultures and developed a human alveolar type I differentiation medium (hATIDM) suitable for generating ATI cells from either mixed distal human lung cells or purified ATII cells. This media adds bone morphogenetic protein 4 (BMP4) and removes epidermal growth factor (EGF), Wnt agonist CHIR99021, and transforming growth factor-beta (TGF-β) inhibitor SB431542 from previously developed alveolar organoid culture media. We demonstrate that BMP4 promotes expression of the ATI marker gene AGER and HOPX, whereas CHIR99021 and SB431542 maintain expression of the ATII marker gene SFTPC. The human ATI spheroids generated with hATIDM express multiple molecular and morphological features reminiscent of human ATI cells. Our results demonstrate that signaling interactions among BMP, TGF-β, and Wnt signaling pathways in alveolar spheroids and distal lung organoids including IPF-organoids coordinate human ATII to ATI differentiation.NEW & NOTEWORTHY Alveolar type I (ATI) epithelial cells perform essential roles in maintaining lung function but have been challenging to study. We explored the signals that promote ATI fate in 3-D organoid cultures generated from either mixed distal human lung cells or purified alveolar type II (ATII) cells. This work fills an important void in our experimental repertoire for studying alveolar epithelial cells and identifies signals that promote human ATII to ATI cell differentiation.
Collapse
Affiliation(s)
- Yong Li
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Qi Tan
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States
| | - Daniel Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
8
|
Tanabe I, Ishimori K, Ishikawa S. Development of an in vitro human alveolar epithelial air-liquid interface model using a small molecule inhibitor cocktail. BMC Mol Cell Biol 2024; 25:9. [PMID: 38500038 PMCID: PMC10946194 DOI: 10.1186/s12860-024-00507-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/11/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND The alveolar epithelium is exposed to numerous stimuli, such as chemicals, viruses, and bacteria that cause a variety of pulmonary diseases through inhalation. Alveolar epithelial cells (AECs) cultured in vitro are a valuable tool for studying the impacts of these stimuli and developing therapies for associated diseases. However, maintaining the proliferative capacity of AECs in vitro is challenging. In this study, we used a cocktail of three small molecule inhibitors to cultivate AECs: Y-27632, A-83-01, and CHIR99021 (YAC). These inhibitors reportedly maintain the proliferative capacity of several types of stem/progenitor cells. RESULTS Primary human AECs cultured in medium containing YAC proliferated for more than 50 days (over nine passages) under submerged conditions. YAC-treated AECs were subsequently cultured at the air-liquid interface (ALI) to promote differentiation. YAC-treated AECs on ALI day 7 formed a monolayer of epithelial tissue with strong expression of the surfactant protein-encoding genes SFTPA1, SFTPB, SFTPC, and SFTPD, which are markers for type II AECs (AECIIs). Immunohistochemical analysis revealed that paraffin sections of YAC-treated AECs on ALI day 7 were mainly composed of cells expressing surfactant protein B and prosurfactant protein C. CONCLUSIONS Our results indicate that YAC-containing medium could be useful for expansion of AECIIs, which are recognized as local stem/progenitor cells, in the alveoli.
Collapse
Affiliation(s)
- Ikuya Tanabe
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan
| | - Kanae Ishimori
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan
| | - Shinkichi Ishikawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| |
Collapse
|
9
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
10
|
Zhang Y, Liu K, He H, Xiao H, Fang Z, Chen X, Li H. Innovative explorations: unveiling the potential of organoids for investigating environmental pollutant exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:16256-16273. [PMID: 38342830 DOI: 10.1007/s11356-024-32256-7] [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: 09/21/2023] [Accepted: 01/25/2024] [Indexed: 02/13/2024]
Abstract
As the economy rapidly develops, chemicals are widely produced and used. This has exacerbated the problems associated with environmental pollution, raising the need for efficient toxicological evaluation techniques to investigate the toxic effects and mechanisms of toxicity of environmental pollutants. The progress in the techniques of cell culture in three dimensions has resulted in the creation of models that are more relevant in terms of biology and physiology. This enables researchers to study organ development, toxicology, and drug screening. Adult stem cells (ASCs) and induced pluripotent stem cells (iPSCs) can be obtained from various mammalian tissues, including cancerous and healthy tissues. Such stem cells exhibit a significant level of tissue memory and ability to self-assemble. When cultivated in 3D in vitro environments, the resulting organoids demonstrate a remarkable capacity to recapitulate the cellular composition and function of organs in vivo. Recently, many tumors' tissue-derived organoids have been widely used in research on tumor pathogenesis, drug development, precision medicine, and other fields, including those derived from colon cancer, cholangiocarcinoma, liver cancer, and gastric cancer. However, the application of organoid models for evaluating the toxicity of environmental pollutants is still in its infancy. This review introduces the characteristics of the toxicity responses of organoid models upon exposure to pollutants from the perspectives of organoid characteristics, tissue types, and their applications in toxicology; discusses the feasibility of using organoid models in evaluating the toxicity of pollutants; and provides a reference for future toxicological studies on environmental pollutants based on organoid models.
Collapse
Affiliation(s)
- Yuanhang Zhang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Kai Liu
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huan He
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China
| | - Hui Xiao
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Zhihong Fang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Xianxian Chen
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huiming Li
- School of Environment, Nanjing Normal University, Nanjing, 210023, China.
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China.
| |
Collapse
|
11
|
Li C, Zhang H, Mo J, Zuo J, Ye L. Caspase-3/GSDME dependent pyroptosis contributes to offspring lung injury induced by gestational PFOS exposure via PERK/ATF4 signaling. Arch Toxicol 2024; 98:207-221. [PMID: 37955688 PMCID: PMC10761489 DOI: 10.1007/s00204-023-03626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is widely used in industry and consumer products. Previous studies have showed that PFOS gestational exposure is associated with offspring lung damage in rat. However, the underlying mechanisms remain poorly understood. In this study, we investigated the role of gasdermin E (GSDME) in lung injury of offspring and its underlying mechanisms using in vivo and in vitro approaches. Pregnant SD rats were exposed to PFOS (1 mg/kg BW/d) between gestational day 12-18, and the lung tissue of the offspring was evaluated on postnatal day 7. PFOS treated animals exhibited alveolar septal thickening and inflammation-related damages, with an increased expression of GSDME in alveolar type II epithelial cells (AECII). Furthermore, in vitro experiments demonstrated that PFOS exposure (with 225 μM and up) upregulated the caspase-3/GSDME signaling pathway in AECII. Also, ultrastructure analysis revealed significant changes in the endoplasmic reticulum (ER) structure in PFOS-induced pyroptotic cells, which is consistent with the ER stress detected in these cells. Additionally, PFOS exposure led to increased expression of ER stress-related proteins, including p-PERK, p-eIF2α, ATF4, and CHOP. Subsequently, using specific inhibitors, we found that the PERK/ATF4 pathway acted as an upstream signal regulating GSDME-dependent pyroptosis. Overall, our findings show that GSDME-dependent pyroptosis plays a crucial role in the lung injury induced by gestational PFOS exposure, and the PERK/ATF4 pathway may function as a possible mediator of this process.
Collapse
Affiliation(s)
- Cong Li
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Huishan Zhang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
- Department of Respiratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200120, China
| | - Jiali Mo
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Jingye Zuo
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Leping Ye
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China.
| |
Collapse
|
12
|
Joo H, Min S, Cho SW. Advanced lung organoids for respiratory system and pulmonary disease modeling. J Tissue Eng 2024; 15:20417314241232502. [PMID: 38406820 PMCID: PMC10894554 DOI: 10.1177/20417314241232502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Amidst the recent coronavirus disease 2019 (COVID-19) pandemic, respiratory system research has made remarkable progress, particularly focusing on infectious diseases. Lung organoid, a miniaturized structure recapitulating lung tissue, has gained global attention because of its advantages over other conventional models such as two-dimensional (2D) cell models and animal models. Nevertheless, lung organoids still face limitations concerning heterogeneity, complexity, and maturity compared to the native lung tissue. To address these limitations, researchers have employed co-culture methods with various cell types including endothelial cells, mesenchymal cells, and immune cells, and incorporated bioengineering platforms such as air-liquid interfaces, microfluidic chips, and functional hydrogels. These advancements have facilitated applications of lung organoids to studies of pulmonary diseases, providing insights into disease mechanisms and potential treatments. This review introduces recent progress in the production methods of lung organoids, strategies for improving maturity, functionality, and complexity of organoids, and their application in disease modeling, including respiratory infection and pulmonary fibrosis.
Collapse
Affiliation(s)
- Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| |
Collapse
|
13
|
Selvarajah B, Platé M, Chambers RC. Pulmonary fibrosis: Emerging diagnostic and therapeutic strategies. Mol Aspects Med 2023; 94:101227. [PMID: 38000335 DOI: 10.1016/j.mam.2023.101227] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023]
Abstract
Fibrosis is the concluding pathological outcome and major cause of morbidity and mortality in a number of common chronic inflammatory, immune-mediated and metabolic diseases. The progressive deposition of a collagen-rich extracellular matrix (ECM) represents the cornerstone of the fibrotic response and culminates in organ failure and premature death. Idiopathic pulmonary fibrosis (IPF) represents the most rapidly progressive and lethal of all fibrotic diseases with a dismal median survival of 3.5 years from diagnosis. Although the approval of the antifibrotic agents, pirfenidone and nintedanib, for the treatment of IPF signalled a watershed moment for the development of anti-fibrotic therapeutics, these agents slow but do not halt disease progression or improve quality of life. There therefore remains a pressing need for the development of effective therapeutic strategies. In this article, we review emerging therapeutic strategies for IPF as well as the pre-clinical and translational approaches that will underpin a greater understanding of the key pathomechanisms involved in order to transform the way we diagnose and treat pulmonary fibrosis.
Collapse
Affiliation(s)
- Brintha Selvarajah
- Oncogenes and Tumour Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Manuela Platé
- Department of Respiratory Medicine (UCL Respiratory), Division of Medicine, University College London, UK
| | - Rachel C Chambers
- Department of Respiratory Medicine (UCL Respiratory), Division of Medicine, University College London, UK.
| |
Collapse
|
14
|
Licciardello M, Sgarminato V, Ciardelli G, Tonda-Turo C. Development of biomimetic co-culture and tri-culture models to mimic the complex structure of the alveolar-capillary barrier. BIOMATERIALS ADVANCES 2023; 154:213620. [PMID: 37690344 DOI: 10.1016/j.bioadv.2023.213620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/29/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Alveoli are the functional area of respiratory system where the gaseous exchanges take place at level of the alveolar-capillary barrier. The development of safe and effective therapeutic approaches for treating lung disease is currently limited due to the lack of realistic preclinical models for their testing and validation. In this work, tissue engineering approaches were exploited to develop a biomimetic platform that provide an appropriate mimicking of the extracellular environment and the multicellular architecture of human alveoli. Here, we propose the implementation of two biomimetic in vitro models to reproduce the features of the main anatomic portions of the physiological alveolar-capillary barrier. First, a co-culture barrier model was obtained by integrating an electrospun polycaprolactone-gelatin (PCL-Gel) membrane in a modified transwell insert (PCL-Gel TW) to mimic the alveolar basement membrane (coded as thin model). Alveolar epithelial (A549) and lung microvascular endothelial (HULEC-5a) cells were cultured on the apical and basolateral side of the PCL-Gel membrane, respectively, under physiologic air-liquid interface (ALI) conditions for 7 days. The ALI condition promoted the expression of type I and type II alveolar epithelial cell markers and the secretion of mucus in A549 cells. Increased cell viability and barrier properties in co-cultures of A549 and HULEC-5a compared to mono-cultures revealed the effectiveness of the model to reproduce in vitro physiological-relevant features of the alveolar-capillary barrier. The second portion of the alveolar-capillary barrier was developed implementing a tri-culture model (coded as thick model) including a type I collagen (COLL) hydrogel formulated to host lung fibroblasts (MRC-5). The thick barrier model was implemented by seeding HULEC-5a on the basolateral side of PCL-Gel TW and then pouring sequentially MRC-5-laden COLL hydrogel and A549 cells on the apical side of the electrospun membrane. The thick model was maintained up to 7 days at ALI and immunofluorescence staining of tight and adherent junctions demonstrated the formation of a tight barrier. Lastly, the ability of models to emulate pathological inflammatory conditions was validated by exposing the apical compartment of the PCL-Gel TW to lipopolysaccharide (LPS). The damage of A549 tight junctions, the increase of barrier permeability and IL-6 pro-inflammatory cytokine release was observed after 48 h exposure to LPS.
Collapse
Affiliation(s)
- Michela Licciardello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy
| | - Viola Sgarminato
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy; CNR-IPCF, National Research Council-Institute for Chemical and Physical Processes, Pisa, Italy
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy.
| |
Collapse
|
15
|
Nashihah AK, Muhammad Firdaus FI, Fauzi MB, Mobarak NN, Lokanathan Y. Role of Biomaterials in the Development of Epithelial Support in 3D In Vitro Airway Epithelium Development: A Systematic Review. Int J Mol Sci 2023; 24:14935. [PMID: 37834382 PMCID: PMC10573735 DOI: 10.3390/ijms241914935] [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: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Respiratory diseases have a major impact on global health. The airway epithelium, which acts as a frontline defence, is one of the most common targets for inhaled allergens, irritants, or micro-organisms to enter the respiratory system. In the tissue engineering field, biomaterials play a crucial role. Due to the continuing high impact of respiratory diseases on society and the emergence of new respiratory viruses, in vitro airway epithelial models with high microphysiological similarities that are also easily adjustable to replicate disease models are urgently needed to better understand those diseases. Thus, the development of biomaterial scaffolds for the airway epithelium is important due to their function as a cell-support device in which cells are seeded in vitro and then are encouraged to lay down a matrix to form the foundations of a tissue for transplantation. Studies conducted in in vitro models are necessary because they accelerate the development of new treatments. Moreover, in comparatively controlled conditions, in vitro models allow for the stimulation of complex interactions between cells, scaffolds, and growth factors. Based on recent studies, the biomaterial scaffolds that have been tested in in vitro models appear to be viable options for repairing the airway epithelium and avoiding any complications. This review discusses the role of biomaterial scaffolds in in vitro airway epithelium models. The effects of scaffold, physicochemical, and mechanical properties in recent studies were also discussed.
Collapse
Affiliation(s)
- Ab Karim Nashihah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Fairuz Izan Muhammad Firdaus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Mh. Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Nadhratun Naiim Mobarak
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia UKM, Bangi 43600, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| |
Collapse
|
16
|
Huang X, Lu Z, Jiang X, Zhang Z, Yan K, Yu G. Single-cell RNA sequencing reveals distinct tumor microenvironment of ground glass nodules and solid nodules in lung adenocarcinoma. Front Cell Dev Biol 2023; 11:1198338. [PMID: 37745301 PMCID: PMC10513029 DOI: 10.3389/fcell.2023.1198338] [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: 04/01/2023] [Accepted: 08/04/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: Lung adenocarcinoma (LUAD) is the most prevalent lung cancer. LUAD presents as ground glass nodules (GGN) and solid nodules (SN) in imaging studies. GGN is an early type of LUAD with good prognosis. However, SN exhibits a more malignant behavior than GGN, including worse pathological staging and tumor prognosis. The mechanism leading to the different malignancy levels of GGN and SN remains elusive. Methods: Three patients with GGN and three patients with SN diagnosed with early LUAD were enrolled. The tumor samples were digested to a single-cell suspension and analyzed using 10× Genomic Single-cell ribonucleic acid sequences (scRNA-seq) techniques. Results: A total of 15,902 cells were obtained and classified into nine major types. The tumor microenvironment (TME) was subsequently described in detail. ScRNA-seq revealed that ribosome-related pathways and cell adhesion played similar but distinct roles in the two groups. SN also had more active cell proliferation, enriched cell cycle regulatory pathways, and severe inflammatory responses. Conclusion: We observed changes in the cellular composition and transcriptomic profile of GGN and SN. The study improved the understanding of the underlying mechanisms of lung carcinogenesis and contributed to lung cancer prevention and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Guiping Yu
- Department of Cardiothoracic Surgery, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, China
| |
Collapse
|
17
|
Carius P, Jungmann A, Bechtel M, Grißmer A, Boese A, Gasparoni G, Salhab A, Seipelt R, Urbschat K, Richter C, Meier C, Bojkova D, Cinatl J, Walter J, Schneider‐Daum N, Lehr C. A Monoclonal Human Alveolar Epithelial Cell Line ("Arlo") with Pronounced Barrier Function for Studying Drug Permeability and Viral Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207301. [PMID: 36748276 PMCID: PMC10015904 DOI: 10.1002/advs.202207301] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Indexed: 06/18/2023]
Abstract
In the development of orally inhaled drug products preclinical animal models regularly fail to predict pharmacological as well as toxicological responses in humans. Models based on human cells and tissues are potential alternatives to animal experimentation allowing for the isolation of essential processes of human biology and making them accessible in vitro. Here, the generation of a novel monoclonal cell line "Arlo," derived from the polyclonal human alveolar epithelium lentivirus immortalized cell line hAELVi via single-cell printing, and its characterization as a model for the human alveolar epithelium as well as a building block for future complex in vitro models is described. "Arlo" is systematically compared in vitro to primary human alveolar epithelial cells (hAEpCs) as well as to the polyclonal hAELVi cell line. "Arlo" cells show enhanced barrier properties with high transepithelial electrical resistance (TEER) of ≈3000 Ω cm2 and a potential difference (PD) of ≈30 mV under air-liquid interface (ALI) conditions, that can be modulated. The cells grow in a polarized monolayer and express genes relevant to barrier integrity as well as homeostasis as is observed in hAEpCs. Successful productive infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a proof-of-principle study offers an additional, attractive application of "Arlo" beyond biopharmaceutical experimentation.
Collapse
Affiliation(s)
- Patrick Carius
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| | - Annemarie Jungmann
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Marco Bechtel
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Alexander Grißmer
- Department of Anatomy and Cellular BiologySaarland UniversityKirrberger StraßeBuilding 6166421Homburg SaarGermany
| | - Annette Boese
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
| | - Gilles Gasparoni
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Abdulrahman Salhab
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Ralf Seipelt
- Section of Thoracic Surgery of the Saar Lung CenterSHG Clinics VölklingenRichardstraße 5‐966333VölklingenGermany
| | - Klaus Urbschat
- Section of Thoracic Surgery of the Saar Lung CenterSHG Clinics VölklingenRichardstraße 5‐966333VölklingenGermany
| | - Clémentine Richter
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| | - Carola Meier
- Department of Anatomy and Cellular BiologySaarland UniversityKirrberger StraßeBuilding 6166421Homburg SaarGermany
| | - Denisa Bojkova
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Jindrich Cinatl
- Institute of Medical VirologyUniversity Hospital FrankfurtPaul‐Ehrlich‐Str. 4060596Frankfurt am MainGermany
| | - Jörn Walter
- Department of Genetics and EpigeneticsSaarland UniversityCampus A2 466123SaarbrückenGermany
| | - Nicole Schneider‐Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
| | - Claus‐Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus E8.166123SaarbrückenGermany
| |
Collapse
|
18
|
Hughes T, Dijkstra KK, Rawlins EL, Hynds RE. Open questions in human lung organoid research. Front Pharmacol 2023; 13:1083017. [PMID: 36712670 PMCID: PMC9880211 DOI: 10.3389/fphar.2022.1083017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Organoids have become a prominent model system in pulmonary research. The ability to establish organoid cultures directly from patient tissue has expanded the repertoire of physiologically relevant preclinical model systems. In addition to their derivation from adult lung stem/progenitor cells, lung organoids can be derived from fetal tissue or induced pluripotent stem cells to fill a critical gap in modelling pulmonary development in vitro. Recent years have seen important progress in the characterisation and refinement of organoid culture systems. Here, we address several open questions in the field, including how closely organoids recapitulate the tissue of origin, how well organoids recapitulate patient cohorts, and how well organoids capture diversity within a patient. We advocate deeper characterisation of models using single cell technologies, generation of more diverse organoid biobanks and further standardisation of culture media.
Collapse
Affiliation(s)
- Tessa Hughes
- Wellcome Trust/CRUK Gurdon Institute and Department Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Krijn K. Dijkstra
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Emma L. Rawlins
- Wellcome Trust/CRUK Gurdon Institute and Department Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Robert E. Hynds
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, Great Ormond Street UCL Institute of Child Health, University College London, London, United Kingdom
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
19
|
Balážová K, Clevers H, Dost AFM. The role of macrophages in non-small cell lung cancer and advancements in 3D co-cultures. eLife 2023; 12:82998. [PMID: 36809334 PMCID: PMC9943070 DOI: 10.7554/elife.82998] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. Traditional therapeutic approaches such as chemotherapy or radiotherapy have provided only a marginal improvement in the treatment of lung carcinomas. Inhibitors targeting specific genetic aberrations present in non-small cell lung cancer (NSCLC), the most common subtype (85%), have improved the prognostic outlook, but due to the complexity of the LC mutational spectrum, only a fraction of patients benefit from these targeted molecular therapies. More recently, the realization that the immune infiltrate surrounding solid tumors can foster tumor-promoting inflammation has led to the development and implementation of anticancer immunotherapies in the clinic. In NSCLC, one of the most abundant leukocyte infiltrates is macrophages. These highly plastic phagocytes, which are part of the cellular repertoire of the innate immunity, can have a pivotal role in early NSCLC establishment, malignant progression, and tumor invasion. Emerging macrophage-targeting therapies have been focused on the re-differentiation of the macrophages toward an antitumorigenic phenotype, depletion of tumor-promoting macrophage subtypes, or combination therapies combining traditional cytotoxic treatments with immunotherapeutic agents. The most extensively used models employed for the exploration of NSCLC biology and therapy have been 2D cell lines and murine models. However, studying cancer immunology requires appropriately complex models. 3D platforms, including organoid models, are quickly advancing powerful tools to study immune cell-epithelial cell interactions within the tumor microenvironment. Co-cultures of immune cells along with NSCLC organoids allow for an in vitro observation of the tumor microenvironment dynamics closely resembling in vivo settings. Ultimately, the implementation of 3D organoid technology into tumor microenvironment-modeling platforms might facilitate the exploration of macrophage-targeted therapies in NSCLC immunotherapeutic research, thus establishing a new frontier in NSCLC treatment.
Collapse
Affiliation(s)
- Katarína Balážová
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| | - Hans Clevers
- Roche Pharma Research and early DevelopmentBaselSwitzerland
| | - Antonella FM Dost
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| |
Collapse
|
20
|
Xuefei Y, Dongyan L, Tianming L, Hejuan Z, Jianhua F. O-linked N-acetylglucosamine affects mitochondrial homeostasis by regulating Parkin-dependent mitophagy in hyperoxia-injured alveolar type II cells injury. Respir Res 2023; 24:16. [PMID: 36647045 PMCID: PMC9841680 DOI: 10.1186/s12931-022-02287-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The level of linked N-acetylglucosamine (O-GlcNAc) has been proved to be a sensor of cell state, but its relationship with hyperoxia-induced alveolar type 2 epithelial cells injure and bronchopulmonary dysplasia (BPD) has not been clarified. In this study, we evaluated if these effects ultimately led to functional damage in hyperoxia-induced alveolar cells. METHODS We treated RLE-6TN cells at 85% hyperoxia for 0, 24 and 48 h with Thiamet G (TG), an OGA inhibitor; OSMI-1 (OS), an OGT inhibitor; or with UDP-GlcNAc, which is involved in synthesis of O-GlcNAc as a donor. The metabolic rerouting, cell viability and apoptosis resulting from the changes in O-GlcNAc glycosyltransferase levels were evaluated in RLE-6TN cells after hyperoxia exposure. We constructed rat Park2 overexpression and knockdown plasmmids for in vitro verification and Co-immunoprecipitation corroborated the binding of Parkin and O-GlcNAc. Finally, we assessed morphological detection in neonatal BPD rats with TG and OS treatment. RESULTS We found a decrease in O-GlcNAc content and levels of its metabolic enzymes in RLE-6TN cells under hyperoxia. However, the inhibition of OGT function with OSMI-1 ameliorated hyperoxia-induced lung epithelial cell injury, enhanced cell metabolism and viability, reduced apoptosis, and accelerated the cell proliferation. Mitochondrial homeostasis was affected by O-GlcNAc and regulated Parkin. CONCLUSION The results revealed that the decreased O-GlcNAc levels and increased O-GlcNAcylation of Parkin might cause hyperoxia-induced alveolar type II cells injurys.
Collapse
Affiliation(s)
- Yu Xuefei
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning 110004 People’s Republic of China
| | - Liu Dongyan
- grid.412467.20000 0004 1806 3501Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning China
| | - Li Tianming
- grid.412467.20000 0004 1806 3501Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Shengjing Hospital of China Medical University, Shenyang, 110004 Liaoning China
| | - Zheng Hejuan
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning 110004 People’s Republic of China
| | - Fu Jianhua
- grid.412467.20000 0004 1806 3501Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning 110004 People’s Republic of China
| |
Collapse
|
21
|
Zamprogno P, Schulte J, Ferrari D, Rechberger K, Sengupta A, van Os L, Weber T, Zeinali S, Geiser T, Guenat OT. Lung-on-a-Chip Models of the Lung Parenchyma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:191-211. [PMID: 37195532 DOI: 10.1007/978-3-031-26625-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Since the publication of the first lung-on-a-chip in 2010, research has made tremendous progress in mimicking the cellular environment of healthy and diseased alveoli. As the first lung-on-a-chip products have recently reached the market, innovative solutions to even better mimic the alveolar barrier are paving the way for the next generation lung-on-chips. The original polymeric membranes made of PDMS are being replaced by hydrogel membranes made of proteins from the lung extracellular matrix, whose chemical and physical properties exceed those of the original membranes. Other aspects of the alveolar environment are replicated, such as the size of the alveoli, their three-dimensional structure, and their arrangement. By tuning the properties of this environment, the phenotype of alveolar cells can be tuned, and the functions of the air-blood barrier can be reproduced, allowing complex biological processes to be mimicked. Lung-on-a-chip technologies also provide the possibility of obtaining biological information that was not possible with conventional in vitro systems. Pulmonary edema leaking through a damaged alveolar barrier and barrier stiffening due to excessive accumulation of extracellular matrix proteins can now be reproduced. Provided that the challenges of this young technology are overcome, there is no doubt that many application areas will benefit greatly.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Jan Schulte
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Karin Rechberger
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Lisette van Os
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Tobias Weber
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Soheila Zeinali
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, 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.
| |
Collapse
|
22
|
Cui T, Wangpaichitr M, Schally AV, Griswold AJ, Vidaurre I, Sha W, Jackson RM. Alveolar epithelial cell growth hormone releasing hormone receptor in alveolar epithelial inflammation. Exp Lung Res 2023; 49:152-164. [PMID: 37584484 DOI: 10.1080/01902148.2023.2246074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
Purpose: Growth hormone-releasing hormone (GHRH) is a 44-amino acid peptide that regulates growth hormone (GH) secretion. We hypothesized that GHRH receptor (GHRH-R) in alveolar type 2 (AT2) cells could modulate pro-inflammatory and possibly subsequent pro-fibrotic effects of lipopolysaccharide (LPS) or cytokines, such that AT2 cells could participate in lung inflammation and fibrosis. Methods: We used human alveolar type 2 (iAT2) epithelial cells derived from induced pluripotent stem cells (iPSC) to investigate how GHRH-R modulates gene and protein expression. We tested iAT2 cells' gene expression in response to LPS or cytokines, seeking whether these mechanisms caused endogenous production of pro-inflammatory molecules or mesenchymal markers. Quantitative real-time PCR (RT-PCR) and Western blotting were used to investigate differential expression of epithelial and mesenchymal markers. Result: Incubation of iAT2 cells with LPS increased expression of IL1-β and TNF-α in addition to mesenchymal genes, including ACTA2, FN1 and COL1A1. Alveolar epithelial cell gene expression due to LPS was significantly inhibited by GHRH-R peptide antagonist MIA-602. Incubation of iAT2 cells with cytokines like those in fibrotic lungs similarly increased expression of genes for IL1-β, TNF-α, TGFβ-1, Wnt5a, smooth muscle actin, fibronectin and collagen. Expression of mesenchymal proteins, such as N-cadherin and vimentin, were also elevated after prolonged exposure to cytokines, confirming epithelial production of pro-inflammatory molecules as an important mechanism that might lead to subsequent fibrosis. Conclusion: iAT2 cells clearly expressed the GHRH-R. Exposure to LPS or cytokines increased iAT2 cell production of pro-inflammatory factors. GHRH-R antagonist MIA-602 inhibited pro-inflammatory gene expression, implicating iAT2 cell GHRH-R signaling in lung inflammation and potentially in fibrosis.
Collapse
Affiliation(s)
- Tengjiao Cui
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Andrew V Schally
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Pathology and Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Anthony J Griswold
- Dr. John T. McDonald Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Wei Sha
- Research Service, Miami VAHS, Miami, Florida, USA
| | - Robert M Jackson
- Research Service, Miami VAHS, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
23
|
Humbert MV, Spalluto CM, Bell J, Blume C, Conforti F, Davies ER, Dean LSN, Elkington P, Haitchi HM, Jackson C, Jones MG, Loxham M, Lucas JS, Morgan H, Polak M, Staples KJ, Swindle EJ, Tezera L, Watson A, Wilkinson TMA. Towards an artificial human lung: modelling organ-like complexity to aid mechanistic understanding. Eur Respir J 2022; 60:2200455. [PMID: 35777774 DOI: 10.1183/13993003.00455-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/11/2022] [Indexed: 11/05/2022]
Abstract
Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.
Collapse
Affiliation(s)
- Maria Victoria Humbert
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Cosma Mirella Spalluto
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- M.V. Humbert and C.M. Spalluto are co-first authors and contributed equally to this work
| | - Joseph Bell
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Cornelia Blume
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Franco Conforti
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Elizabeth R Davies
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Lareb S N Dean
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Paul Elkington
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hans Michael Haitchi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Claire Jackson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Mark G Jones
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Matthew Loxham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Hywel Morgan
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Marta Polak
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Karl J Staples
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Emily J Swindle
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Liku Tezera
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Infection and Immunity, Faculty of Medicine, University College London, London, UK
| | - Alastair Watson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Tom M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
24
|
Demchenko A, Lavrov A, Smirnikhina S. Lung organoids: current strategies for generation and transplantation. Cell Tissue Res 2022; 390:317-333. [PMID: 36178558 PMCID: PMC9522545 DOI: 10.1007/s00441-022-03686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/08/2022] [Indexed: 01/19/2023]
Abstract
Lung diseases occupy a leading position in human morbidity and are the third leading cause of death. Often the chronic forms of these diseases do not respond to therapy, so that lung transplantation is the only treatment option. The development of cellular and biotechnologies offers a new solution-the use of lung organoids for transplantation in such patients. Here, we review types of lung organoids, methods of their production and characterization, and experimental works on transplantation in vivo. These results show the promise of work in this direction. Despite the current problems associated with a low degree of cell engraftment, immune response, and insufficient differentiation, we are confident that organoid transplantation will find it is clinical application.
Collapse
Affiliation(s)
- Anna Demchenko
- Research Centre for Medical Genetics, Laboratory of Genome Editing, Moscow, 115522 Russia
| | - Alexander Lavrov
- Research Centre for Medical Genetics, Laboratory of Genome Editing, Moscow, 115522 Russia
| | - Svetlana Smirnikhina
- Research Centre for Medical Genetics, Laboratory of Genome Editing, Moscow, 115522 Russia
| |
Collapse
|
25
|
Zhao Y, Zhang J, Lu H, Mao Y, Qin J, Wang Y, Wang X, Dai Z, Wang X, Yang Z, Hou L. CARDIOPULMONARY BYPASS-DERIVED PLASMA EXOSOMAL HMGB1 CONTRIBUTES TO ALVEOLAR EPITHELIAL CELL NECROPTOSIS VIA mtDNA/CGAS/STING PATHWAY. Shock 2022; 58:534-541. [PMID: 36516451 DOI: 10.1097/shk.0000000000002006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Our previous study confirmed that cardiopulmonary bypass (CPB) leads to acute lung injury (ALI) via inducing high-mobility group box 1 (HMGB1) release. Recent research showed that HMGB1 promotes pulmonary injury mainly via exosomes transport. Currently, alveolar epithelial cell (AEC) necroptosis has been demonstrated to be involved in ALI. However, it is unknown whether exosomal inflammatory cytokine HMGB1 promotes ALI by inducing AEC necroptosis, and its underlying mechanisms remain elusive. Here, a prospective cohort study was carried out, in which plasma samples from 21 CPB patients were isolated at four specific time points: pre-CPB, 2, 12, and 24 h after initiation of CPB. Plasma exosomes were extracted via ultra-high-speed centrifugation and cocultured with AEC cell line-A549 cells at increasing concentrations of 50, 100, and 150 μg/mL. Then, HMGB1 antagonist-Box A and mtDNA deficiency ethidium bromide (EtBr) were applied to explore the underlying role of exosomal HMGB1 and cytoplasm mitochondrial DNA in AEC. Western blot analysis showed that plasma exosomal HMGB1 expression gradually increased and peaked at 24 h after CPB. Twenty-four-hour treatment of CPB-derived exosomes at 150 μg/mL for 24 h could induce necroptosis by promoting mitochondrial fission and further elevating cytoplasm mtDNA levels in A549 cells, which was successfully blocked by Box A or EtBr. Most importantly, EtBr significantly inhibited cytoplasm mtDNA downstream guanosine monophosphate (GMP)-AMP synthase (cGAS)/stimulator of interferon gene (STING) signal pathway. Collectively, these data demonstrate that CPB-derived plasma exosomal HMGB1 contributes to AEC necroptosis through the mtDNA/cGAS/STING pathway.
Collapse
Affiliation(s)
- Yupeng Zhao
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinyuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huihong Lu
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiliang Mao
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiawen Qin
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yinglin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuebin Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiguang Dai
- Department of Anesthesiology, Shanghai East Hospital Ji'an Hospital, Ji'an City, Jiangxi Province, China
| | - Xiangrui Wang
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Yang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | | |
Collapse
|
26
|
Dye BR, Decker JT, Hein RF, Miller AJ, Huang S, Spence JR, Shea LD. Human Lung Organoid Culture in Alginate With and Without Matrigel to Model Development and Disease. Tissue Eng Part A 2022; 28:893-906. [PMID: 36029210 PMCID: PMC9805886 DOI: 10.1089/ten.tea.2022.0054] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/12/2022] [Indexed: 01/13/2023] Open
Abstract
Human lung organoids (HLOs) are enabling the study of human lung development and disease by modeling native organ tissue structure, cellular composition, and cellular organization. In this report, we demonstrate that HLOs derived from human pluripotent stem cells cultured in alginate, a fully defined nonanimal product substrate, exhibit enhanced cellular differentiation compared with HLOs cultured in the commercially available Matrigel. More specifically, we observed an earlier onset and increase in the number of multiciliated cells, along with mucus producing MUC5AC+ goblet-like cells that were not observed in HLOs cultured in Matrigel. The epithelium in alginate-grown HLOs was organized in a pseudostratified epithelium with airway basal cells lining the basal lamina, but with the apical surface of cells on the exterior of the organoid. We further observed that HLOs cultured in Matrigel exhibited mesenchymal overgrowth that was not present in alginate cultures. The containment of the mesenchyme within HLOs in alginate enabled modeling of key features of idiopathic pulmonary fibrosis (IPF) by treatment with transforming growth factor β (TGFβ). TGFβ treatment resulted in morphological changes including an increase in mesenchymal growth, increased expression of IPF markers, and decreased numbers of alveolar-like cells. This culture system provides a model to study the interaction of the mesenchyme with the epithelium during lung development and diseased states such as IPF.
Collapse
Affiliation(s)
- Briana R. Dye
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph T. Decker
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Renee F.C. Hein
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alyssa J. Miller
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R. Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Zhang L, Luo W, Liu J, Xu M, Peng Q, Zou W, You J, Shu Y, Zhao P, Wagstaff W, Zhao G, Qin K, Haydon RC, Luu HH, Reid RR, Bi Y, Zhao T, He TC, Fu Z. Modeling lung diseases using reversibly immortalized mouse pulmonary alveolar type 2 cells (imPAC2). Cell Biosci 2022; 12:159. [PMID: 36138472 PMCID: PMC9502644 DOI: 10.1186/s13578-022-00894-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/30/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND A healthy alveolar epithelium is critical to the gas exchange function of the lungs. As the major cell type of alveolar epithelium, alveolar type 2 (AT2) cells play a critical role in maintaining pulmonary homeostasis by serving as alveolar progenitors during lung injury, inflammation, and repair. Dysregulation of AT2 cells may lead to the development of acute and chronic lung diseases and cancer. The lack of clinically relevant AT2 cell models hampers our ability to understand pulmonary diseases. Here, we sought to establish reversibly immortalized mouse pulmonary alveolar type 2 cells (imPAC2) and investigate their potential in forming alveolar organoids to model pulmonary diseases. METHODS Primary mouse pulmonary alveolar cells (mPACs) were isolated and immortalized with a retroviral expression of SV40 Large T antigen (LTA). Cell proliferation and survival was assessed by crystal violet staining and WST-1 assays. Marker gene expression was assessed by qPCR, Western blotting, and/or immunostaining. Alveolar organoids were generated by using matrigel. Ad-TGF-β1 was used to transiently express TGF-β1. Stable silencing β-catenin or overexpression of mutant KRAS and TP53 was accomplished by using retroviral vectors. Subcutaneous cell implantations were carried out in athymic nude mice. The retrieved tissue masses were subjected to H & E histologic evaluation. RESULTS We immortalized primary mPACs with SV40 LTA to yield the imPACs that were non-tumorigenic and maintained long-term proliferative activity that was reversible by FLP-mediated removal of SV40 LTA. The EpCAM+ AT2-enriched subpopulation (i.e., imPAC2) was sorted out from the imPACs, and was shown to express AT2 markers and form alveolar organoids. Functionally, silencing β-catenin decreased the expression of AT2 markers in imPAC2 cells, while TGF-β1 induced fibrosis-like response by regulating the expression of epithelial-mesenchymal transition markers in the imPAC2 cells. Lastly, concurrent expression of oncogenic KRAS and mutant TP53 rendered the imPAC2 cells a tumor-like phenotype and activated lung cancer-associated pathways. Collectively, our results suggest that the imPAC2 cells may faithfully represent AT2 populations that can be further explored to model pulmonary diseases.
Collapse
Affiliation(s)
- Linghuan Zhang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Wenping Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
- Laboratory Animal Center, Southwest University, Chongqing, 400715, China
| | - Jiang Liu
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Maozhu Xu
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qi Peng
- University-Town Hospital, Chongqing Medical University, Chongqing, 401331, China
| | - Wenjing Zou
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jingyi You
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yi Shu
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
- Departments of Orthopaedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400046, China
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
- Departments of Orthopaedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400046, China
| | - Kevin Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
- Rosalind Franklin University of Medicine, North Chicago, IL, 60064, USA
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA
| | - Tianyu Zhao
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, the Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC3079, Chicago, IL, 60637, USA.
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA.
| | - Zhou Fu
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, and the Department of Respiratory Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
28
|
Jeong MH, Han H, Lagares D, Im H. Recent Advances in Molecular Diagnosis of Pulmonary Fibrosis for Precision Medicine. ACS Pharmacol Transl Sci 2022; 5:520-538. [PMID: 35983278 PMCID: PMC9379941 DOI: 10.1021/acsptsci.2c00028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis is a serious, progressive lung disease characterized by scarring and stiffening lung tissues, affecting the respiratory system and leading to organ failure. It is a complex disease consisting of alveolar damage, chronic inflammation, and a varying degree of lung fibrosis. Significant challenges with pulmonary fibrosis include the lack of effective means to diagnose the disease at early stages, identify patients at higher risks of progress, and assess disease progression and treatment response. Precision medicine powered by accurate molecular profiling and phenotyping could significantly improve our understanding of the disease's heterogeneity, potential biomarkers for diagnosis and prognosis, and molecular targets for treatment development. This Review discusses various translational model systems, including organoids and lung-on-a-chip systems, biomarkers in single cells and extracellular vesicles, and functional pharmacodynamic markers. We also highlight emerging sensing technologies for molecular characterization of pulmonary fibrosis and biomarker detection.
Collapse
Affiliation(s)
- Mi Ho Jeong
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Hongwei Han
- Department
of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts
General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - David Lagares
- Department
of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts
General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hyungsoon Im
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| |
Collapse
|
29
|
He Y, Yu F, Tian Y, Hu Q, Wang B, Wang L, Hu Y, Tao Y, Chen X, Peng M. Single-Cell RNA Sequencing Unravels Distinct Tumor Microenvironment of Different Components of Lung Adenocarcinoma Featured as Mixed Ground-Glass Opacity. Front Immunol 2022; 13:903513. [PMID: 35874770 PMCID: PMC9299373 DOI: 10.3389/fimmu.2022.903513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022] Open
Abstract
Lung adenocarcinoma featured as mixed ground-glass opacity (mGGO) doubled its volume half of the time in comparison with that featured as pure ground-glass opacity (pGGO). The mechanisms underlying the heterogeneous appearance of mGGO remain elusive. In this study, we macro-dissected the solid (S) components and ground-glass (GG) components of mGGO and performed single-cell sequencing analyses of six paired components from three mGGO patients. A total of 19,391 single-cell profiles were taken into analysis, and the data of each patient were analyzed independently to obtain a common alteration. Cancer cells and macrophages were the dominant cell types in the S and GG components, respectively. Cancer cells in the S components, which showed relatively malignant phenotypes, were likely to originate from both the GG and S components and monitor the surrounding tumor microenvironment (TME) through an intricate cell interaction network. SPP1hi macrophages were enriched in the S components and showed increased activity of chemoattraction, while macrophages in the GG components displayed an active antimicrobial process with a higher stress-induced state. In addition, the CD47–SIRPA axis was demonstrated to be critical in the maintenance of the GG components. Taken together, our study unraveled the alterations of cell components and transcriptomic features between different components in mGGOs.
Collapse
Affiliation(s)
- Yu He
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yi Tian
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Thoracic Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qikang Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bin Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yan Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yongguang Tao
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaofeng Chen
- Department of Anaesthesia, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Muyun Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
30
|
Baptista D, Moreira Teixeira L, Barata D, Tahmasebi Birgani Z, King J, van Riet S, Pasman T, Poot AA, Stamatialis D, Rottier RJ, Hiemstra PS, Carlier A, van Blitterswijk C, Habibović P, Giselbrecht S, Truckenmüller R. 3D Lung-on-Chip Model Based on Biomimetically Microcurved Culture Membranes. ACS Biomater Sci Eng 2022; 8:2684-2699. [PMID: 35502997 PMCID: PMC9198974 DOI: 10.1021/acsbiomaterials.1c01463] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
A comparatively straightforward
approach to accomplish more physiological
realism in organ-on-a-chip (OoC) models is through substrate geometry.
There is increasing evidence that the strongly, microscale curved
surfaces that epithelial or endothelial cells experience when lining
small body lumens, such as the alveoli or blood vessels, impact their
behavior. However, the most commonly used cell culture substrates
for modeling of these human tissue barriers in OoCs, ion track-etched
porous membranes, provide only
flat surfaces. Here, we propose a more realistic culture environment
for alveolar cells based on biomimetically microcurved track-etched
membranes. They recreate the mainly spherical geometry of the cells’
native microenvironment. In this feasibility study, the membranes
were given the shape of hexagonally arrayed hemispherical microwells
by an innovative combination of three-dimensional (3D) microfilm (thermo)forming
and ion track technology. Integrated in microfluidic chips, they separated
a top from a bottom cell culture chamber. The microcurved membranes
were seeded by infusion with primary human alveolar epithelial cells.
Despite the pronounced topology, the cells fully lined the alveoli-like
microwell structures on the membranes’ top side. The confluent
curved epithelial cell monolayers could be cultured successfully at
the air−liquid interface for 14 days. Similarly, the top and
bottom sides of the microcurved membranes were seeded with cells from
the Calu-3 lung epithelial cell line and human lung microvascular
endothelial cells, respectively. Thereby, the latter lined the interalveolar
septum-like interspace between the microwells in a network-type fashion,
as in the natural counterpart. The coculture was maintained for 11
days. The presented 3D lung-on-a-chip model might set the stage for
other (micro)anatomically inspired membrane-based OoCs in the future.
Collapse
Affiliation(s)
- Danielle Baptista
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Liliana Moreira Teixeira
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.,Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - David Barata
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.,Instituto de Medicina Molecular, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Zeinab Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Jasia King
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Thijs Pasman
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - André A Poot
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery/Cell Biology, Erasmus (University) Medical Center Rotterdam - Sophia Children's Hospital, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
31
|
Samperio Ventayol P, Bartfeld S. Immune cell-stem cell interactions in regeneration and repair: who's calling the shots? Development 2022; 149:275251. [DOI: 10.1242/dev.200228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
In November 2021, the Institute for Regenerative Medicine (IRM) and the Institute for Immunology (IFI) at the University of Pennsylvania, USA, joined forces and organized a symposium featuring external speakers as well as locally based scientists to discuss how the immune system influences tissue stem cell biology. As we review here, the presentations highlighted emerging concepts in the field, revealing how tissue-specific immune cell activation can guide stem cells in regeneration and repair.
Collapse
Affiliation(s)
- Pilar Samperio Ventayol
- Medical Biotechnology, Institute for Biotechnology, Technische Universität Berlin, Berlin 13355, Germany
| | - Sina Bartfeld
- Medical Biotechnology, Institute for Biotechnology, Technische Universität Berlin, Berlin 13355, Germany
- Si-M/‘Der Simulierte Mensch’, a Science Framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians Universität Würzburg, Würzburg 97020, Germany
| |
Collapse
|
32
|
Fei L, Sun G, Sun J, Wu D. The effect of N6-methyladenosine (m6A) factors on the development of acute respiratory distress syndrome in the mouse model. Bioengineered 2022; 13:7622-7634. [PMID: 35263199 PMCID: PMC8973778 DOI: 10.1080/21655979.2022.2049473] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) can cause loss of alveolar-capillary membrane integrity and life-threatening immune responses. The underlying molecular mechanisms of ARDS remain unclear. N6-methyladenosine (m6A)-RNA modification plays an important part in many biological processes. However, it is not clear whether ARDS alters RNA methylation in lung tissue. We tried to investigate the changes of m6A-RNA methylation in lung tissues of lipopolysaccharide (LPS)-induced ARDS mice. Lung tissue samples were collected to detect the expression of m6A factors through hematoxylin and eosin (HE) staining, quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR), immunohistochemical analysis and western blot. The overall m6A levels in lung tissue of ARDS in mouse were detected by UPLC-UV-MS. HE staining showed that the degree of the inflammatory response was more severe in the LPS-3 h group. The mRNA expression of YTHDF1, YTHDC1 and IGFBP3 was remarkably up-regulated at, respectively, 6, 6 and 12 h after LPS treatment. The mRNA expression of METTL16, FTO, METTL3, KIAA1429, RBM15, ALKBH5, YTHDF2, YTHDF3, YTHDC2 and IGFBP2 was significantly down-regulated at 24 h after LPS treatment. The protein expression of METTL16 and FTO increased, YTHDC1, IGFBP3 YTHDF1 and YTHDF3 showed a down-regulation trend after LPS induction. Overall m6A-RNA methylation levels were significantly increased at 6 h after LPS induction. In ARDS mice, LPS-induced m6A methylation may be involved in the expression regulation of inflammatory factors and may play important roles in the occurrence and development of lung tissue. It is suggested that m6A modification may be a promising therapeutic target for ARDS.
Collapse
Affiliation(s)
- Liming Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juan Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Dong Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
33
|
He Y, Rofaani E, Huang X, Huang B, Liang F, Wang L, Shi J, Peng J, Chen Y. Generation of Alveolar Epithelium Using Reconstituted Basement Membrane and hiPSC-Derived Organoids. Adv Healthc Mater 2022; 11:e2101972. [PMID: 34935309 DOI: 10.1002/adhm.202101972] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/01/2021] [Indexed: 12/11/2022]
Abstract
In vitro modeling of alveolar epithelium needs to recapitulate features of both cellular and noncellular components of the lung tissues. Herein, a method is presented to generate alveolar epithelium by using human induced pluripotent stem cells (hiPSCs) and reconstituted or artificial basement membrane (ABM). The ABM is obtained by self-assembling type IV collagen and laminin with a monolayer of crosslinked gelatin nanofibers as backbone and a patterned honeycomb microframe for handling. Alveolar organoids are obtained from hiPSCs and then dissociated into single cells. After replating the alveolar cells on the ABM and a short-period incubation under submerged and air-liquid interface culture conditions, an alveolar epithelium is achieved, showing high-level expressions of both alveolar cell-specific proteins and characteristic tight junctions. Besides, endothelial cells derived from the same hiPSCs are cocultured on the backside of the epithelium, forming an air-blood barrier. The method is generic and can potentially be applied to other types of artificial epithelium and endothelium.
Collapse
Affiliation(s)
- Yong He
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Elrade Rofaani
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Xiaochen Huang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Boxin Huang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Feng Liang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Li Wang
- MesoBioTech 231 Rue Saint‐Honoré Paris 75001 France
| | - Jian Shi
- MesoBioTech 231 Rue Saint‐Honoré Paris 75001 France
| | - Juan Peng
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Yong Chen
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| |
Collapse
|
34
|
van Riet S, van Schadewijk A, Khedoe PPSJ, Limpens RWAL, Bárcena M, Stolk J, Hiemstra PS, van der Does AM. Organoid-based Expansion of Patient-Derived Primary Alveolar Type-2 Cells for Establishment of Alveolus Epithelial Lung-Chip Cultures. Am J Physiol Lung Cell Mol Physiol 2022; 322:L526-L538. [PMID: 35137633 PMCID: PMC8957343 DOI: 10.1152/ajplung.00153.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Development of effective treatment strategies for lung tissue destruction as seen in emphysema would greatly benefit from representative human in vitro models of the alveolar compartment. Studying how cellular cross-talk and/or (altered) biomechanical cues affect alveolar epithelial function could provide new insight for tissue repair strategies. Preclinical models of the alveolus ideally combine human primary patient-derived lung cells with advanced cell culture applications such as breathing-related stretch, to reliably represent the alveolar microenvironment. To test the feasibility of such a model, we isolated primary alveolar type-2 cells (AEC2) from patient-derived lung tissues including those from patients with severe emphysema, using magnetic bead-based selection of cells expressing the AEC2 marker HTII-280. We obtained pure alveolar feeder-free organoid cultures using a minimally modified commercial medium. This was confirmed by known AEC2 markers as well as by detection of lamellar bodies using electron microscopy. Following (organoid-based) expansion, cells were seeded on both cell culture inserts and the Chip-S1® Organ-Chip that has a flexible PDMS membrane enabling the application of dynamic stretch. AEC2 cultured for 7 days on inserts or the chip maintained expression of HTII-280, pro-surfactant protein C (SP-C), SP-A and SP-B and zonula occludens-1 (ZO-1) also in the presence of stretch. AEC2 cultured on the chip showed lower expression levels of epithelial-mesenchymal transition-related vimentin expression compared to static cultures on inserts. The combination of a straightforward culture method of patient-derived AEC2 and their application in microfluidic chip cultures, supports successful development of more representative human preclinical models of the (diseased) alveolar compartment.
Collapse
Affiliation(s)
- Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - P Padmini S J Khedoe
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ronald W A L Limpens
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, the Netherlands
| | - Montserrat Bárcena
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, the Netherlands
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne M van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
35
|
Yang JW, Lin YR, Chu YL, Chung JHY, Lu HE, Chen GY. Tissue-level alveolar epithelium model for recapitulating SARS-CoV-2 infection and cellular plasticity. Commun Biol 2022; 5:70. [PMID: 35046486 PMCID: PMC8770515 DOI: 10.1038/s42003-022-03026-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
AbstractPulmonary sequelae following COVID-19 pneumonia have been emerging as a challenge; however, suitable cell sources for studying COVID-19 mechanisms and therapeutics are currently lacking. In this paper, we present a standardized primary alveolar cell culture method for establishing a human alveolar epithelium model that can recapitulate viral infection and cellular plasticity. The alveolar model is infected with a SARS-CoV-2 pseudovirus, and the clinically relevant features of the viral entry into the alveolar type-I/II cells, cytokine production activation, and pulmonary surfactant destruction are reproduced. For this damaged alveolar model, we find that the inhibition of Wnt signaling via XAV939 substantially improves alveolar repair function and prevents subsequent pulmonary fibrosis. Thus, the proposed alveolar cell culture strategy exhibits potential for the identification of pathogenesis and therapeutics in basic and translational research.
Collapse
|
36
|
Generation of macrophage containing alveolar organoids derived from human pluripotent stem cells for pulmonary fibrosis modeling and drug efficacy testing. Cell Biosci 2021; 11:216. [PMID: 34922627 PMCID: PMC8684607 DOI: 10.1186/s13578-021-00721-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/18/2021] [Indexed: 11/10/2022] Open
Abstract
Macrophages are a central immune component in various types of in vitro human organoid systems to recapitulate normal and pathological development. However, to date, generation of human alveolar organoids (AOs) containing macrophages for use as a pulmonary fibrosis (PF) model and drug efficacy evaluation has not been reported. Here, we generated multicellular alveolar organoids (Mac-AOs) containing functional macrophages derived from human pluripotent stem cells based on stepwise direct differentiation by mimicking developmental cues in a temporally controlled manner. Derived Mac-AOs contained the expected range of cell types, including alveolar progenitors, mesenchymal cells, alveolar epithelial cells (type 1 and 2), and macrophages. Treatment with transforming growth factor (TGF-β1) induced inflammation and fibrotic changes in Mac-AOs, offering a PF model for validating the therapeutic potential of new drugs. TGF-β1-induced fibrotic responses and collagen accumulation in these Mac-AOs were effectively ameliorated by treatment with Pirfenidone, Nintedanib, and NP-011 via suppression of extracellular signal-regulated kinase signaling. To the best of our knowledge, this is the first report to provide non-epithelial functional macrophage-containing human AO system, which will better recapitulate the complexity of in vivo alveolar tissues and advance our understanding of the pathogenesis and development of effective therapies for PF.
Collapse
|
37
|
Increased LGR6 Expression Sustains Long-Term Wnt Activation and Acquisition of Senescence in Epithelial Progenitors in Chronic Lung Diseases. Cells 2021; 10:cells10123437. [PMID: 34943945 PMCID: PMC8700573 DOI: 10.3390/cells10123437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/28/2021] [Accepted: 12/03/2021] [Indexed: 01/14/2023] Open
Abstract
Chronic lung diseases (CLDs) represent a set of disorders characterized by the progressive loss of proper lung function. Among severe CLDs, the incidence of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) has grown over the last decades, mainly in the elderly population. Several studies have highlighted an increased expression of senescence-related markers in the resident progenitor cells in COPD and IPF, possibly undermining epithelial integrity and contributing to the progression and the aggravation of both diseases. Recently, the chronic activation of the canonical Wnt/β-catenin pathway was shown to induce cellular senescence. Here, we investigated the localization and the expression of leucin-rich repeat-containing G-protein-coupled receptor 6 (LGR6), a protein that activates and potentiates the canonical Wnt signalling. Through immunohistochemical analyses, we identified a lesion-associated rise in LGR6 levels in abnormal lung epithelial progenitors in COPD and IPF when compared to histologically normal tissues. Moreover, in areas of aberrant regeneration, chronic damage and fibrosis, LGR6-expressing epithelial progenitors displayed a major increase in the expression of senescence-associated markers. Our study suggests the involvement of LGR6 in the chronic activation of the Wnt/β-catenin pathway, mediating the impairment and exhaustion of epithelial progenitors in COPD and IPF.
Collapse
|
38
|
Collins DP, Osborn MJ, Steer CJ. Differentiation of immortalized human multi-lineage progenitor to alveolar type 2-like cells: angiotensin-converting enzyme 2 expression and binding of severe acute respiratory syndrome coronavirus 2 spike and spike 1 proteins. Cytotherapy 2021; 23:1064-1073. [PMID: 34551876 PMCID: PMC8367755 DOI: 10.1016/j.jcyt.2021.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 12/04/2022]
Abstract
Along with the nasal epithelium, the lung epithelium is a portal of entry for sudden acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and many other respiratory viruses. In the case of SARS-CoV-2, the virus surface spike proteins bind to the angiotensin-converting enzyme 2 (ACE-2) receptor to facilitate entry into the respiratory epithelium. Alveolar type 2 (AT2) cells are committed respiratory progenitor cells responsible for the integrity and regeneration of the respiratory epithelium and production of respiratory surfactant proteins. AT2 cells express high levels of surface ACE-2 and thus are a leading target for primary infection by SARS-CoV-2. This study describes a method for directly differentiating telomerase reverse transcriptase-immortalized human cord blood-derived multi-lineage progenitor cells (MLPCs) to AT2-like cells for the purpose of generating an in vitro cellular platform for viral studies. Differentiation was confirmed with the acquisition of AT2 and absence of alveolar type 1 (AT1) specific markers by confocal microscopy. Expression of the ACE-2 receptor was confirmed by immunofluorescence antibody staining, quantitative reverse transcription polymerase chain reaction and binding of biotinylated SARS-CoV-2 spike and spike 1 proteins. The binding of biotinylated spike proteins was specifically blocked by unlabeled spike proteins and neutralizing antibodies. Additionally, it was demonstrated that the spike protein was internalized after binding to the surface membrane of the cells. The authors defined the culture conditions that enabled AT2-like cells to be repeatedly passaged and cryopreserved without further differentiation to AT1. The authors' method provides a stable and renewable source of AT2 cells for respiratory viral binding, blocking and uptake studies.
Collapse
Affiliation(s)
| | - Mark J Osborn
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Clifford J Steer
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
39
|
Lee D, Kim Y, Chung C. Scientific Validation and Clinical Application of Lung Cancer Organoids. Cells 2021; 10:cells10113012. [PMID: 34831235 PMCID: PMC8616085 DOI: 10.3390/cells10113012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/30/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
Lung cancer organoid (LCO) is a novel model of lung cancer that facilitates drug screening. However, the success rate of LCOs varies from 7% to 87%, and the culture medium compositions are markedly different. Airway organoid media can be used for LCO cultures, but this promotes the overgrowth of normal cell organoids especially in LCOs from intrapulmonary lesions. Several modified media are specifically utilized for promoting the cancer cell's growth. For culturing high-purity LCOs, cancer cells from metastatic lesions and malignant effusions are used. Recently, single-cell RNA sequencing has identified previously unknown cell populations in the lungs and lung cancer. This sequencing technology can be used to validate whether the LCO recapitulates the heterogeneity and functional hierarchy of the primary tumor. Several groups have attempted to culture LCOs with mesenchymal cells and immune cells to recapitulate the tumor microenvironment. Disease modeling using LCO provides novel insight into the pathophysiology of lung cancer and enables high-throughput screening for drug discovery and prognosis prediction. An LCO model would help to identify new concepts as a basis for lung cancer targeting by discovering innovative therapeutic targets.
Collapse
Affiliation(s)
- Dahye Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 34134, Korea; (D.L.); (Y.K.)
| | - Yoonjoo Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 34134, Korea; (D.L.); (Y.K.)
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 34134, Korea; (D.L.); (Y.K.)
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Korea
- Correspondence:
| |
Collapse
|
40
|
Liberti DC, Morrisey EE. Organoid models: assessing lung cell fate decisions and disease responses. Trends Mol Med 2021; 27:1159-1174. [PMID: 34674972 DOI: 10.1016/j.molmed.2021.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Organoids can be derived from various cell types in the lung, and they provide a reproducible and tractable model for understanding the complex signals driving cell fate decisions in a regenerative context. In this review, we provide a retrospective account of organoid methodologies and outline new opportunities for optimizing these methods to further explore emerging concepts in lung biology. Moreover, we examine the benefits of integrating organoid assays with in vivo modeling to explore how the various niches and compartments in the respiratory system respond to both acute and chronic lung disease. The strategic implementation and improvement of organoid techniques will provide exciting new opportunities to understand and identify new therapeutic approaches to ameliorate lung disease states.
Collapse
Affiliation(s)
- Derek C Liberti
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Rijsbergen LC, van Dijk LLA, Engel MFM, de Vries RD, de Swart RL. In Vitro Modelling of Respiratory Virus Infections in Human Airway Epithelial Cells - A Systematic Review. Front Immunol 2021; 12:683002. [PMID: 34489934 PMCID: PMC8418200 DOI: 10.3389/fimmu.2021.683002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Respiratory tract infections (RTI) are a major cause of morbidity and mortality in humans. A large number of RTIs is caused by viruses, often resulting in more severe disease in infants, elderly and the immunocompromised. Upon viral infection, most individuals experience common cold-like symptoms associated with an upper RTI. However, in some cases a severe and sometimes life-threatening lower RTI may develop. Reproducible and scalable in vitro culture models that accurately reflect the human respiratory tract are needed to study interactions between respiratory viruses and the host, and to test novel therapeutic interventions. Multiple in vitro respiratory cell culture systems have been described, but the majority of these are based on immortalized cell lines. Although useful for studying certain aspects of viral infections, such monomorphic, unicellular systems fall short in creating an understanding of the processes that occur at an integrated tissue level. Novel in vitro models involving primary human airway epithelial cells and, more recently, human airway organoids, are now in use. In this review, we describe the evolution of in vitro cell culture systems and their characteristics in the context of viral RTIs, starting from advances after immortalized cell cultures to more recently developed organoid systems. Furthermore, we describe how these models are used in studying virus-host interactions, e.g. tropism and receptor studies as well as interactions with the innate immune system. Finally, we provide an outlook for future developments in this field, including co-factors that mimic the microenvironment in the respiratory tract.
Collapse
Affiliation(s)
- Laurine C. Rijsbergen
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Laura L. A. van Dijk
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Maarten F. M. Engel
- Medical Library, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| | - Rik L. de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
42
|
Khedoe PPPSJ, Wu X, Gosens R, Hiemstra PS. Repairing damaged lungs using regenerative therapy. Curr Opin Pharmacol 2021; 59:85-94. [PMID: 34161852 PMCID: PMC9188766 DOI: 10.1016/j.coph.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/14/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022]
Abstract
There is an urgent need for better treatment of lung diseases that are a major cause of morbidity and mortality worldwide. This urgency is illustrated by the current COVID-19 health crisis. Moderate-to-extensive lung injury characterizes several lung diseases, and not only therapies that reduce such lung injury are needed but also those that regenerate lung tissue and repair existing lung injury. At present, such therapies are not available, but as a result of a rapid increase in our understanding of lung development and repair, lung regenerative therapies are on the horizon. Here, we discuss existing targets for treatment, as well as novel strategies for development of pharmacological and cell therapy-based regenerative treatment for a variety of lung diseases and clinical studies. We discuss how both patient-relevant in vitro disease models using innovative culture techniques and other advanced new technologies aid in the development of pulmonary regenerative medicine.
Collapse
Affiliation(s)
| | - Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
43
|
Sun AM, Hoffman T, Luu BQ, Ashammakhi N, Li S. Application of lung microphysiological systems to COVID-19 modeling and drug discovery: a review. Biodes Manuf 2021; 4:757-775. [PMID: 34178414 PMCID: PMC8213042 DOI: 10.1007/s42242-021-00136-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
There is a pressing need for effective therapeutics for coronavirus disease 2019 (COVID-19), the respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. The process of drug development is a costly and meticulously paced process, where progress is often hindered by the failure of initially promising leads. To aid this challenge, in vitro human microphysiological systems need to be refined and adapted for mechanistic studies and drug screening, thereby saving valuable time and resources during a pandemic crisis. The SARS-CoV-2 virus attacks the lung, an organ where the unique three-dimensional (3D) structure of its functional units is critical for proper respiratory function. The in vitro lung models essentially recapitulate the distinct tissue structure and the dynamic mechanical and biological interactions between different cell types. Current model systems include Transwell, organoid and organ-on-a-chip or microphysiological systems (MPSs). We review models that have direct relevance toward modeling the pathology of COVID-19, including the processes of inflammation, edema, coagulation, as well as lung immune function. We also consider the practical issues that may influence the design and fabrication of MPS. The role of lung MPS is addressed in the context of multi-organ models, and it is discussed how high-throughput screening and artificial intelligence can be integrated with lung MPS to accelerate drug development for COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Argus M. Sun
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- UC San Diego Healthcare, UCSD, La Jolla, CA 92037 USA
| | - Tyler Hoffman
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
| | - Bao Q. Luu
- Pulmonary Diseases and Critical Care, Scripps Green Hospital, Scripps Health, La Jolla, CA 92037 USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824 USA
| | - Song Li
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 USA
| |
Collapse
|
44
|
Huang KY, Petretto E. Cross-species integration of single-cell RNA-seq resolved alveolar-epithelial transitional states in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2021; 321:L491-L506. [PMID: 34132117 DOI: 10.1152/ajplung.00594.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Single-cell transcriptomics analyses of the fibrotic lung uncovered two cell states critical to lung injury recovery in the alveolar epithelium-a reparative transitional cell state in the mouse and a disease-specific cell state (KRT5-/KRT17+) in human idiopathic pulmonary fibrosis (IPF). The murine transitional cell state lies between the differentiation from type 2 (AT2) to type 1 pneumocyte (AT1), and the human KRT5-/KRT17+ cell state may arise from the dysregulation of this differentiation process. We review major findings of single-cell transcriptomics analyses of the fibrotic lung and reanalyzed data from seven single-cell RNA sequencing studies of human and murine models of IPF, focusing on the alveolar epithelium. Our comparative and cross-species single-cell transcriptomics analyses allowed us to further delineate the differentiation trajectories from AT2 to AT1 and AT2 to the KRT5-/KRT17+ cell state. We observed AT1 cells in human IPF retain the transcriptional signature of the murine transitional cell state. Using pseudotime analysis, we recapitulated the differentiation trajectories from AT2 to AT1 and from AT2 to KRT5-/KRT17+ cell state in multiple human IPF studies. We further delineated transcriptional programs underlying cell-state transitions and determined the molecular phenotypes at terminal differentiation. We hypothesize that in addition to the reactivation of developmental programs (SOX4, SOX9), senescence (TP63, SOX4) and the Notch pathway (HES1) are predicted to steer intermediate progenitors to the KRT5-/KRT17+ cell state. Our analyses suggest that activation of SMAD3 later in the differentiation process may explain the fibrotic molecular phenotype typical of KRT5-/KRT17+ cells.
Collapse
Affiliation(s)
- Kevin Y Huang
- Program in Cardiovascular and Metabolic Disorders (CVMD) and Center for Computational Biology (CCB), Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Enrico Petretto
- Program in Cardiovascular and Metabolic Disorders (CVMD) and Center for Computational Biology (CCB), Duke-NUS Medical School, Singapore, Republic of Singapore
| |
Collapse
|
45
|
Oglesby IK, Schweikert A, Fox B, Redmond C, Donnelly SC, Hurley K. Lung organoids and other preclinical models of pulmonary fibrosis. QJM 2021; 114:167-173. [PMID: 33484260 DOI: 10.1093/qjmed/hcaa281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 11/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal disease affecting over 100 000 people in Europe with an increasing incidence. Available treatments offer only slowing of disease progression and are poorly tolerated by patients leading to cessation of therapy. Lung transplant remains the only cure. Therefore, alternative treatments are urgently required. The pathology of IPF is complex and poorly understood and thus creates a major obstacle to the discovery of novel treatments. Additionally, preclinical assessment of new treatments currently relies upon animal models where disparities with human lung biology often hamper drug development. At a cellular level, IPF is characterized by persistent and abnormal deposition of extracellular matrix by fibroblasts and alveolar epithelial cell injury which is seen as a key event in initiation of disease progression. In-depth investigation of the role of alveolar epithelial cells in health and disease has been impeded due to difficulties in primary cell isolation and culture ex vivo. Novel strategies employing patient-derived induced pluripotent stem cells engineered to produce type 2 alveolar epithelial cells (iAEC2) cultured as three-dimensional organoids have the potential to overcome these hurdles and inform new effective precision treatments for IPF leading to improved survival and quality of life for patients worldwide.
Collapse
Affiliation(s)
- I K Oglesby
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, D02 H903, Ireland
| | - A Schweikert
- Interfaculty Institute of Biochemistry, Eberhard Karls Universität Tübingen, Geschwister-Scholl-Platz 72074 Tübingen, Germany
| | - B Fox
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
| | - C Redmond
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
| | - S C Donnelly
- Department of Respiratory & Interstitial Lung Disease, Tallaght University Hospital Tallaght, Dublin D24 NR0A, Ireland
- School of Medicine, Trinity College Dublin, The University of Dublin, College Green, Dublin D02 PN40, Ireland
| | - K Hurley
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, D09 YD60, Ireland
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, D02 H903, Ireland
| |
Collapse
|
46
|
Ebisudani T, Sugimoto S, Haga K, Mitsuishi A, Takai-Todaka R, Fujii M, Toshimitsu K, Hamamoto J, Sugihara K, Hishida T, Asamura H, Fukunaga K, Yasuda H, Katayama K, Sato T. Direct derivation of human alveolospheres for SARS-CoV-2 infection modeling and drug screening. Cell Rep 2021; 35:109218. [PMID: 34038715 PMCID: PMC8133488 DOI: 10.1016/j.celrep.2021.109218] [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: 08/07/2020] [Revised: 12/04/2020] [Accepted: 05/13/2021] [Indexed: 12/15/2022] Open
Abstract
Although the main cellular target of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is thought to be alveolar cells, the absence of their tractable culture system precludes the development of a clinically relevant SARS-CoV-2 infection model. Here, we establish an efficient human alveolosphere culture method and sphere-based drug testing platform for SARS-CoV-2. Alveolospheres exhibit indolent growth in a Wnt- and R-spondin-dependent manner. Gene expression, immunofluorescence, and electron microscopy analyses reveal the presence of alveolar cells in alveolospheres. Alveolospheres express ACE2 and allow SARS-CoV-2 to propagate nearly 100,000-fold in 3 days of infection. Whereas lopinavir and nelfinavir, protease inhibitors used for the treatment of human immunodeficiency virus (HIV) infection, have a modest anti-viral effect on SARS-CoV-2, remdesivir, a nucleotide prodrug, shows an anti-viral effect at the concentration comparable with the circulating drug level. These results demonstrate the validity of the alveolosphere culture system for the development of therapeutic agents to combat SARS-CoV-2.
Collapse
Affiliation(s)
- Toshiki Ebisudani
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Shinya Sugimoto
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kei Haga
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Akifumi Mitsuishi
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Reiko Takai-Todaka
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8641, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kohta Toshimitsu
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Junko Hamamoto
- Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kai Sugihara
- Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tomoyuki Hishida
- Division of Thoracic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hisao Asamura
- Division of Thoracic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Koichi Fukunaga
- Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Coronavirus Task Force, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroyuki Yasuda
- Department of Pulmonary Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Kazuhiko Katayama
- Laboratory of Viral Infection I, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control Sciences, Kitasato University, Tokyo 108-8641, Japan; Coronavirus Task Force, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; Coronavirus Task Force, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
47
|
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
|
48
|
Iverson E, Kaler L, Agostino EL, Song D, Duncan GA, Scull MA. Leveraging 3D Model Systems to Understand Viral Interactions with the Respiratory Mucosa. Viruses 2020; 12:E1425. [PMID: 33322395 PMCID: PMC7763686 DOI: 10.3390/v12121425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory viruses remain a significant cause of morbidity and mortality in the human population, underscoring the importance of ongoing basic research into virus-host interactions. However, many critical aspects of infection are difficult, if not impossible, to probe using standard cell lines, 2D culture formats, or even animal models. In vitro systems such as airway epithelial cultures at air-liquid interface, organoids, or 'on-chip' technologies allow interrogation in human cells and recapitulate emergent properties of the airway epithelium-the primary target for respiratory virus infection. While some of these models have been used for over thirty years, ongoing advancements in both culture techniques and analytical tools continue to provide new opportunities to investigate airway epithelial biology and viral infection phenotypes in both normal and diseased host backgrounds. Here we review these models and their application to studying respiratory viruses. Furthermore, given the ability of these systems to recapitulate the extracellular microenvironment, we evaluate their potential to serve as a platform for studies specifically addressing viral interactions at the mucosal surface and detail techniques that can be employed to expand our understanding.
Collapse
Affiliation(s)
- Ethan Iverson
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Logan Kaler
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
| | - Eva L. Agostino
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| | - Daniel Song
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Gregg A. Duncan
- Biophysics Program, University of Maryland, College Park, MD 20742, USA; (L.K.); (G.A.D.)
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA;
| | - Margaret A. Scull
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA; (E.I.); (E.L.A.)
| |
Collapse
|
49
|
Salahudeen AA, Choi SS, Rustagi A, Zhu J, van Unen V, de la O SM, Flynn RA, Margalef-Català M, Santos AJM, Ju J, Batish A, Usui T, Zheng GXY, Edwards CE, Wagar LE, Luca V, Anchang B, Nagendran M, Nguyen K, Hart DJ, Terry JM, Belgrader P, Ziraldo SB, Mikkelsen TS, Harbury PB, Glenn JS, Garcia KC, Davis MM, Baric RS, Sabatti C, Amieva MR, Blish CA, Desai TJ, Kuo CJ. Progenitor identification and SARS-CoV-2 infection in human distal lung organoids. Nature 2020; 588:670-675. [PMID: 33238290 PMCID: PMC8003326 DOI: 10.1038/s41586-020-3014-1] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/18/2020] [Indexed: 12/17/2022]
Abstract
The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange. Three-dimensional in vitro human distal lung culture systems would strongly facilitate the investigation of pathologies such as interstitial lung disease, cancer and coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we describe the development of a long-term feeder-free, chemically defined culture system for distal lung progenitors as organoids derived from single adult human alveolar epithelial type II (AT2) or KRT5+ basal cells. AT2 organoids were able to differentiate into AT1 cells, and basal cell organoids developed lumens lined with differentiated club and ciliated cells. Single-cell analysis of KRT5+ cells in basal organoids revealed a distinct population of ITGA6+ITGB4+ mitotic cells, whose offspring further segregated into a TNFRSF12Ahi subfraction that comprised about ten per cent of KRT5+ basal cells. This subpopulation formed clusters within terminal bronchioles and exhibited enriched clonogenic organoid growth activity. We created distal lung organoids with apical-out polarity to present ACE2 on the exposed external surface, facilitating infection of AT2 and basal cultures with SARS-CoV-2 and identifying club cells as a target population. This long-term, feeder-free culture of human distal lung organoids, coupled with single-cell analysis, identifies functional heterogeneity among basal cells and establishes a facile in vitro organoid model of human distal lung infections, including COVID-19-associated pneumonia.
Collapse
Affiliation(s)
- Ameen A Salahudeen
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Shannon S Choi
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arjun Rustagi
- Division of Infectious Disease and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Junjie Zhu
- Stanford University School of Engineering, Department of Electrical Engineering, Stanford, CA, USA
| | - Vincent van Unen
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean M de la O
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan A Flynn
- Stanford ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Mar Margalef-Català
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - António J M Santos
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jihang Ju
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arpit Batish
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tatsuya Usui
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Caitlin E Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lisa E Wagar
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent Luca
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benedict Anchang
- Division of Biomedical Data Science, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Monica Nagendran
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Khanh Nguyen
- Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel J Hart
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | - Pehr B Harbury
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey S Glenn
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chiara Sabatti
- Division of Biomedical Data Science, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Manuel R Amieva
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Catherine A Blish
- Division of Infectious Disease and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Tushar J Desai
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
50
|
Kim SY, Mongey R, Griffiths M, Hind M, Dean CH. An Ex Vivo Acid Injury and Repair (AIR) Model Using Precision-Cut Lung Slices to Understand Lung Injury and Repair. ACTA ACUST UNITED AC 2020; 10:e85. [PMID: 33217226 DOI: 10.1002/cpmo.85] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Recent advances in cell culture models like air-liquid interface culture and ex vivo models such as organoids have advanced studies of lung biology; however, gaps exist between these models and tools that represent the complexity of the three-dimensional environment of the lung. Precision-cut lung slices (PCLS) mimic the in vivo environment and bridge the gap between in vitro and in vivo models. We have established the acid injury and repair (AIR) model where a spatially restricted area of tissue is injured using drops of HCl combined with Pluronic gel. Injury and repair are assessed by immunofluorescence using robust markers, including Ki67 for cell proliferation and prosurfactant protein C for alveolar type 2/progenitor cells. Importantly, the AIR model enables the study of injury and repair in mouse lung tissue without the need for an initial in vivo injury, and the results are highly reproducible. Here, we present detailed protocols for the generation of PCLS and the AIR model. We also describe methods to analyze and quantify injury in AIR-PCLS by immunostaining with established early repair markers and fluorescence imaging. This novel ex vivo model is a versatile tool for studying lung cell biology in acute lung injury and for semi-high-throughput screening of potential therapeutics. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Generation of precision-cut lung slices Basic Protocol 2: The acid injury and repair model Basic Protocol 3: Analysis of AIR-PCLS: Immunostaining and imaging.
Collapse
Affiliation(s)
- Sally Yunsun Kim
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Róisín Mongey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Mark Griffiths
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Peri-Operative Medicine Department, St Bartholomew's Hospital, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,National Institute for Health Research (NIHR) Respiratory Biomedical Research Unit at the Royal Brompton & Harefield NHS Foundation Trust and Imperial College, London, United Kingdom
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,MRC Harwell Institute, Harwell Campus, Oxfordshire, United Kingdom
| |
Collapse
|