1
|
Elbialy A, Sood A, Wang SJ, Wang P, Fadiel A, Parwani AV, Huang S, Shvets G, Putluri N, Li J, Liu X. Unveiling racial disparities in prostate cancer using an integrative genomic and transcriptomic analysis. CELL INSIGHT 2025; 4:100238. [PMID: 40104216 PMCID: PMC11914995 DOI: 10.1016/j.cellin.2025.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 03/20/2025]
Abstract
Prostate cancer exhibits significant racial disparities, with African American (AA) individuals showing ∼64% higher incidence and 2.3 times greater mortality rates compared to their Caucasian (CA) counterparts. Understanding the complex interplay of genetic, environmental, lifestyle, socioeconomic, and healthcare access factors is crucial for developing effective interventions to reduce this disproportionate burden. This study aims to uncover the genetic and transcriptomic differences driving these disparities through a comprehensive analysis using RNA sequencing (RNA-seq) and exome sequencing of prostate cancer tissues from both Black and White patients. Our transcriptomics analysis revealed enhanced activity in pathways linked to immune response and cellular interactions in AA prostate cancer samples, with notable regulation by histone-associated transcription factors (HIST1H1A, HIST1H1D, and HIST1H1B) suggests potential involvement of histone modification mechanisms. Additionally, pseudogenes and long non-coding RNAs (lncRNAs) among the regulated genes indicate non-coding elements' role in these disparities. Exome sequencing identified unique variants in AA patient samples within key genes, including TP73 (tumor suppression), XYLB (metabolism), ALDH4A1 (oxidative stress), PTPRB (cellular signaling), and HLA-DRB5 (immune response). These genetic variations likely contribute to disease progression and therapy response disparities. This study highlights the importance of considering genetic and epigenetic variations in developing tailored therapeutic approaches to improve treatment efficacy and reduce mortality rates across diverse populations.
Collapse
Affiliation(s)
- Abdalla Elbialy
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL, 60637, USA
| | - Akshay Sood
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Urology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Shang-Jui Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Peng Wang
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Medicine, College of Medicine, The Ohio State University, Columbus, OH, 3210, USA
| | - Ahmed Fadiel
- Computational Oncology Unit, The University of Chicago Comprehensive Cancer Center, 900 E 57th Street, KCBD Bldg., STE 4144, Chicago, IL, 60637, USA
| | - Anil V Parwani
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Steven Huang
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, 14850, USA
| | - Gennady Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, 14850, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jenny Li
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Xuefeng Liu
- OSU Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Departments of Pathology, Urology, and Radiation Oncology, College of Medicine, The Ohio StateUniversity, Columbus, OH, 43210, USA
| |
Collapse
|
2
|
Ishii Y, Orr JC, El Mdawar MB, de Pilger DRB, Pearce DR, Lazarus KA, Graham RE, Nikolić MZ, Ketteler R, Carragher NO, Janes SM, Hynds RE. Compound screening in human airway basal cells identifies Wnt pathway activators as potential pro-regenerative therapies. J Cell Sci 2025; 138:jcs263487. [PMID: 40065746 PMCID: PMC12045047 DOI: 10.1242/jcs.263487] [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: 08/13/2024] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Regeneration of the airway epithelium restores barrier function and mucociliary clearance following lung injury and infection. The mechanisms regulating the proliferation and differentiation of tissue-resident airway basal stem cells remain incompletely understood. To identify compounds that promote human airway basal cell proliferation, we performed phenotype-based compound screening of 1429 compounds (from the ENZO and Prestwick Chemical libraries) in 384-well format using primary cells transduced with lentiviral luciferase. A total of 17 pro-proliferative compounds were validated in independent donor cell cultures, including the antiretroviral therapy agent abacavir and several Wnt signalling pathway-activating compounds. The effects of compounds on proliferation were further explored in colony formation and 3D organoid assays. Structurally and functionally related compounds that more potently induced Wnt pathway activation were investigated. One such compound, 1-azakenpaullone, induced Wnt target gene activation and basal cell proliferation in mice. Our results demonstrate the pro-proliferative effect of small-molecule Wnt pathway activators on airway basal cells. These findings contribute to the rationale to develop novel approaches to modulate Wnt signalling during airway epithelial repair.
Collapse
Affiliation(s)
- Yuki Ishii
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Jessica C. Orr
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Marie-Belle El Mdawar
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | | | - David R. Pearce
- UCL Cancer Institute, University College London, London WC1N 6DD, UK
| | - Kyren A. Lazarus
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Rebecca E. Graham
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Marko Z. Nikolić
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Neil O. Carragher
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Sam M. Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Robert E. Hynds
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
- UCL Cancer Institute, University College London, London WC1N 6DD, UK
| |
Collapse
|
3
|
Li C, Zhang X, Cai H, Luo K, Shi B, Chen B, Zeng G, Zheng J, He X. Ferrostatin-1 inhibits tracheal basal cell ferroptosis to facilitate the rapid epithelization of 3D-printed tissue-engineered tracheas. Stem Cell Res Ther 2025; 16:147. [PMID: 40128770 PMCID: PMC11934759 DOI: 10.1186/s13287-025-04263-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Tracheal replacement is a promising approach for treating tracheal defects that are caused by conditions such as stenosis, trauma, or tumors. However, slow postoperative epithelial regeneration often leads to complications, such as infection and granulation tissue formation. Ferroptosis, which is an iron-dependent form of regulated cell death, limits the proliferation of tracheal basal cells (TBCs), which are essential for the epithelialization of tissue-engineered tracheas (TETs). This study explored the potential of ferrostatin-1 (FER-1), which is a ferroptosis inhibitor, to increase TBC proliferation and accelerate the epithelialization of 3D-printed TETs. METHODS TBCs were isolated from rabbit bronchial mucosal tissues and cultured in vitro. Ferroptosis was induced in TBCs at passage 2, as shown by increased reactive oxygen species (ROS) levels, Fe2⁺ accumulation, decreased ATP contents, and mitochondrial damage. TBCs were treated with FER-1 (1 μM) for 48 h to inhibit ferroptosis. The effects on ROS levels, Fe2⁺ levels, ATP contents, and mitochondrial morphology were measured. For in vivo experiments, FER-1-treated TBCs were seeded onto 3D-printed polycaprolactone (PCL) scaffolds, which were implanted into rabbits with tracheal injury. Epithelial regeneration and granulation tissue formation were evaluated 6 months after surgery. RESULTS FER-1 treatment significantly reduced ferroptosis marker levels in vitro; that is, FER-1 treatment decreased ROS and Fe2⁺ accumulation, ameliorated mitochondrial structures, and increased ATP levels. TBC proliferation and viability were increased after ferroptosis inhibition. In vivo, the group that received 3D-printed scaffolds seeded with TBCs exhibited accelerated TET epithelialization and reduced granulation tissue formation compared with the control groups. These results suggest that inhibiting ferroptosis with FER-1 improves TBC function, leading to more efficient tracheal repair. CONCLUSIONS Ferrostatin-1 effectively inhibits ferroptosis in tracheal basal cells, promoting their viability and proliferation. This results in faster epithelialization of tissue-engineered tracheas, offering a promising strategy for improving tracheal reconstruction outcomes and reducing complications such as infection and granulation tissue formation. Future studies are needed to further investigate the molecular mechanisms underlying ferroptosis in TBCs and its potential clinical applications.
Collapse
Affiliation(s)
- Cong Li
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Xiaoyang Zhang
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Haoqi Cai
- Department of Orthopedic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Kai Luo
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Bozhong Shi
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Bo Chen
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Guowei Zeng
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Jinghao Zheng
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
4
|
Huo Y, He S, Chen Y. Lung organoids in COPD: recent advances and future prospects. Respir Res 2025; 26:76. [PMID: 40022099 PMCID: PMC11871743 DOI: 10.1186/s12931-025-03138-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease that is characterized by progressive airflow limitation, a high prevalence, and a high mortality rate. However, the specific mechanisms remain unclear, partly due to the lack of robust data from in vitro experimental models and animal models that do not adequately represent the structure and pathophysiology of the human lung. The recent advancement of lung organoid culture systems has facilitated new avenues for the investigation of COPD. Lung organoids are in vitro models derived from adult stem cells, human pluripotent stem cells, or embryonic stem cells, established through three-dimensional culture. They exhibit a high degree of homology and genetic consistency with human tissues and can better mimic human lungs in terms of function and structure compared to other traditional models. This review will summarise the generation process of lung organoids from different cell sources and their application in COPD research, and provide suggestions for future research directions.
Collapse
Affiliation(s)
- Yajie Huo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shengyang He
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, China.
- Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, China.
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, China.
- Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
5
|
Liu J, Zhang Y, Yu Y. Establishment of nasal and olfactory epithelium organoids for unveiling mechanism of tissue regeneration and pathogenesis of nasal diseases. Cell Mol Life Sci 2025; 82:33. [PMID: 39751829 PMCID: PMC11699091 DOI: 10.1007/s00018-024-05557-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: 07/13/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Organoid is an ideal in vitro model with cellular heterogeneity and genetic stability when passaging. Currently, organoids are exploited as new tools in a variety of preclinical researches and applications for disease modeling, drug screening, host-microbial interactions, and regenerative therapy. Advances have been made in the establishment of nasal and olfactory epithelium organoids that are used to investigate the pathogenesis of smell-related diseases and cellular/molecular mechanism underlying the regeneration of olfactory epithelium. A set of critical genes are identified to function in cell proliferation and neuronal differentiation in olfactory epithelium organoids. Besides, nasal epithelium organoids derived from chronic rhinosinusitis patients have been established to reveal the pathogenesis of this disease, potentially applied in drug responses in individual patient. The present article reviews recent research progresses of nasal and olfactory epithelium organoids in fundamental and preclinical researches, and proposes current advances and potential future direction in the field of organoid research and application.
Collapse
Affiliation(s)
- Jinxia Liu
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
- Olfactory Disorder Diagnosis and Treatment Center, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yunfeng Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yiqun Yu
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.
- Olfactory Disorder Diagnosis and Treatment Center, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China.
- Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai, 200031, China.
| |
Collapse
|
6
|
Zhu Z, Cheng Y, Liu X, Ding W, Liu J, Ling Z, Wu L. Advances in the Development and Application of Human Organoids: Techniques, Applications, and Future Perspectives. Cell Transplant 2025; 34:9636897241303271. [PMID: 39874083 PMCID: PMC11775963 DOI: 10.1177/09636897241303271] [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] [Revised: 10/10/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025] Open
Abstract
Organoids are three-dimensional (3D) cell cultures derived from human pluripotent stem cells or adult stem cells that recapitulate the cellular heterogeneity, structure, and function of human organs. These microstructures are invaluable for biomedical research due to their ability to closely mimic the complexity of native tissues while retaining human genetic material. This fidelity to native organ systems positions organoids as a powerful tool for advancing our understanding of human biology and for enhancing preclinical drug testing. Recent advancements have led to the successful development of a variety of organoid types, reflecting a broad range of human organs and tissues. This progress has expanded their application across several domains, including regenerative medicine, where organoids offer potential for tissue replacement and repair; disease modeling, which allows for the study of disease mechanisms and progression in a controlled environment; drug discovery and evaluation, where organoids provide a more accurate platform for testing drug efficacy and safety; and microecological research, where they contribute to understanding the interactions between microbes and host tissues. This review provides a comprehensive overview of the historical development of organoid technology, highlights the key achievements and ongoing challenges in the field, and discusses the current and emerging applications of organoids in both laboratory research and clinical practice.
Collapse
Affiliation(s)
- Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| |
Collapse
|
7
|
Stack JT, Rayner RE, Nouri R, Suarez CJ, Kim SH, Kanke KL, Vetter TA, Cormet-Boyaka E, Vaidyanathan S. DNA-PKcs inhibition improves sequential gene insertion of the full-length CFTR cDNA in airway stem cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102339. [PMID: 39398224 PMCID: PMC11470261 DOI: 10.1016/j.omtn.2024.102339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Although many people with CF (pwCF) are treated using CFTR modulators, some are non-responsive due to their genotype or other uncharacterized reasons. Autologous airway stem cell therapies, in which the CFTR cDNA has been replaced, may enable a durable therapy for all pwCF. Previously, CRISPR-Cas9 with two AAVs was used to sequentially insert two-halves of the CFTR cDNA and an enrichment cassette into the CFTR locus. However, the editing efficiency was <10% and required enrichment to restore CFTR function. Further improvement in gene insertion may enhance cell therapy production. To improve CFTR cDNA insertion in human airway basal stem cells (ABCs), we evaluated the use of the small molecules AZD7648 and ART558, which inhibit non-homologous end-joining (NHEJ) and micro-homology mediated end-joining (MMEJ). Adding AZD7648 alone improved gene insertion by 2- to 3-fold. Adding both ART558 and AZD7648 improved gene insertion but induced toxicity. ABCs edited in the presence of AZD7648 produced differentiated airway epithelial sheets with restored CFTR function after enrichment. Adding AZD7648 did not increase off-target editing. Further studies are necessary to validate if AZD7648 treatment enriches cells with oncogenic mutations.
Collapse
Affiliation(s)
- Jacob T. Stack
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Rachael E. Rayner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Reza Nouri
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Carlos J. Suarez
- Department of Pathology, Stanford University, Palo Alto, CA 94305, USA
| | - Sun Hee Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Karen L. Kanke
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
| | - Tatyana A. Vetter
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Sriram Vaidyanathan
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Daneshdoust D, He K, Wang QE, Li J, Liu X. Modeling respiratory tract diseases for clinical translation employing conditionally reprogrammed cells. CELL INSIGHT 2024; 3:100201. [PMID: 39391007 PMCID: PMC11462205 DOI: 10.1016/j.cellin.2024.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 10/12/2024]
Abstract
Preclinical models serve as indispensable tools in translational medicine. Specifically, patient-derived models such as patient-derived xenografts (PDX), induced pluripotent stem cells (iPSC), organoids, and recently developed technique of conditional reprogramming (CR) have been employed to reflect the host characteristics of diseases. CR technology involves co-culturing epithelial cells with irradiated Swiss-3T3-J2 mouse fibroblasts (feeder cells) in the presence of a Rho kinase (ROCK) inhibitor, Y-27632. CR technique facilitates the rapid conversion of both normal and malignant cells into a "reprogrammed stem-like" state, marked by robust in vitro proliferation. This is achieved without reliance on exogenous gene expression or viral transfection, while maintaining the genetic profile of the parental cells. So far, CR technology has been used to study biology of diseases, targeted therapies (precision medicine), regenerative medicine, and noninvasive diagnosis and surveillance. Respiratory diseases, ranking as the third leading cause of global mortality, pose a significant burden to healthcare systems worldwide. Given the substantial mortality and morbidity rates of respiratory diseases, efficient and rapid preclinical models are imperative to accurately recapitulate the diverse spectrum of respiratory conditions. In this article, we discuss the applications and future potential of CR technology in modeling various respiratory tract diseases, including lung cancer, respiratory viral infections (such as influenza and Covid-19 and etc.), asthma, cystic fibrosis, respiratory papillomatosis, and upper aerodigestive track tumors. Furthermore, we discuss the potential utility of CR in personalized medicine, regenerative medicine, and clinical translation.
Collapse
Affiliation(s)
- Danyal Daneshdoust
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kai He
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Qi-En Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Radiation Oncology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jenny Li
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Departments of Pathology, Urology, and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| |
Collapse
|
9
|
Ma J, Wu C, Xu J. The Development of Lung Tissue Engineering: From Biomaterials to Multicellular Systems. Adv Healthc Mater 2024; 13:e2401025. [PMID: 39206615 DOI: 10.1002/adhm.202401025] [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: 03/22/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
The challenge of the treatment of end-stage lung disease poses an urgent clinical demand for lung tissue engineering. Over the past few years, various lung tissue-engineered constructs are developed for lung tissue regeneration and respiratory pathology study. In this review, an overview of recent achievements in the field of lung tissue engineering is proposed. The introduction of lung structure and lung injury are stated briefly at first. After that, the lung tissue-engineered constructs are categorized into three types: acellular, monocellular, and multicellular systems. The different bioengineered constructs included in each system that can be applied to the reconstruction of the trachea, airway epithelium, alveoli, and even whole lung are described in detail, followed by the highlight of relevant representative research. Finally, the challenges and future directions of biomaterials, manufacturing technologies, and cells involved in lung tissue engineering are discussed. Overall, this review can provide referable ideas for the realization of functional lung regeneration and permanent lung substitution.
Collapse
Affiliation(s)
- Jingge Ma
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinfu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| |
Collapse
|
10
|
Chen X, Shen J, Jiang X, Pan M, Chang S, Li J, Wang L, Miao M, Feng X, Zhang L, Shu G, Liu W, Xu F, Zhang W, Ding Z, Zong H, Liu W, Li D, Chen B, Shao M, Fei G, Zha X, Fan X. Characterization of dipyridamole as a novel ferroptosis inhibitor and its therapeutic potential in acute respiratory distress syndrome management. Theranostics 2024; 14:6947-6968. [PMID: 39629132 PMCID: PMC11610143 DOI: 10.7150/thno.102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Rationale: Ferroptosis in lung epithelium and endothelium contributes to the pathogenesis of acute respiratory distress syndrome (ARDS), a critical and often fatal condition marked by acute inflammation and elevated pulmonary vascular permeability. Despite this, there are currently no FDA-approved therapeutics specifically targeting ferroptosis for ARDS management. Methods: A screening of 259 FDA-approved drugs was conducted to identify an effective ferroptosis inhibitor in pulmonary epithelial and endothelial cells. The anti-ferroptotic and therapeutic efficacy of this screened drug was rigorously evaluated using two distinct ARDS mouse models (LPS-induced acute lung injury and CLP-induced sepsis) and human airway organoids (hAOs). The regulatory mechanism of this drug on ferroptosis inhibition was investigated via RNA-sequencing, qRT-PCR, western blotting, IF, luciferase reporter assay, chromatin immunoprecipitation assay, limited proteolysis-mass spectrometry assay, cellular thermal shift assay, and drug affinity responsive target stability assay. Furthermore, a proof-of-concept clinical trial was conducted, wherein ARDS patients were administered with the drug as adjunctive therapy. Results: Dipyridamole (DIPY) was identified as a potent inhibitor of ferroptosis in pulmonary epithelial and endothelial cells. DIPY effectively mitigated ferroptosis and pulmonary damage in both mouse models and hAOs, primarily by downregulating heme oxygenase 1 (HMOX1). The transcription factor cAMP responsive element binding protein 1 (CREB1) was identified as a key transactivator of HMOX1, which DIPY effectively downregulated. Mechanistically, DIPY binds to and activates superoxide dismutase 1 (SOD1), which in turn inhibits the CREB1/HMOX1 pathway, thereby suppressing ferroptosis. Notably, the clinical trial further corroborated the therapeutic potential of DIPY in ARDS patients, demonstrating improved outcomes with DIPY adjunctive therapy. Conclusions: These findings provide compelling evidence that DIPY inhibits ferroptosis in pulmonary epithelial and endothelial cells by modulating the SOD1/CREB1/HMOX1 signaling axis and suggest DIPY as a promising therapeutic strategy for ARDS treatment.
Collapse
Affiliation(s)
- Xu Chen
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Jiapan Shen
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Xueqin Jiang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| | - Min Pan
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Shuang Chang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Juanjuan Li
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lei Wang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Manli Miao
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Xiaoxia Feng
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Ling Zhang
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Guoqing Shu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Wenjian Liu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangzhou Xu
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
| | - Wentao Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zhao Ding
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Huaiyuan Zong
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Weiwei Liu
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Dapeng Li
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Biao Chen
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Min Shao
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guanghe Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Geriatric Institute, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
11
|
Orr JC, Laali A, Durrenberger PF, Lazarus KA, El Mdawar MB, Janes SM, Hynds RE. A lentiviral toolkit to monitor airway epithelial cell differentiation using bioluminescence. Am J Physiol Lung Cell Mol Physiol 2024; 327:L587-L599. [PMID: 39137525 PMCID: PMC11482462 DOI: 10.1152/ajplung.00047.2024] [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: 02/09/2024] [Revised: 07/26/2024] [Accepted: 08/11/2024] [Indexed: 08/15/2024] Open
Abstract
Basal cells are adult stem cells in the airway epithelium and regenerate differentiated cell populations, including the mucosecretory and ciliated cells that enact mucociliary clearance. Human basal cells can proliferate and produce differentiated epithelium in vitro. However, studies of airway epithelial differentiation mostly rely on immunohistochemical or immunofluorescence-based staining approaches, meaning that a dynamic approach is lacking, and quantitative data are limited. Here, we use a lentiviral reporter gene approach to transduce primary human basal cells with bioluminescence reporter constructs to monitor airway epithelial differentiation longitudinally. We generated three constructs driven by promoter sequences from the TP63, MUC5AC, and FOXJ1 genes to quantitatively assess basal cell, mucosecretory cell, and ciliated cell abundance, respectively. We validated these constructs by tracking differentiation of basal cells in air-liquid interface and organoid ("bronchosphere") cultures. Transduced cells also responded appropriately to stimulation with interleukin 13 (IL-13; to increase mucosecretory differentiation and mucus production) and IL-6 (to increase ciliated cell differentiation). These constructs represent a new tool for monitoring airway epithelial cell differentiation in primary epithelial and/or induced pluripotent stem cell (iPSC)-derived cell cultures.NEW & NOTEWORTHY Orr et al. generated and validated new lentiviral vectors to monitor the differentiation of airway basal cells, goblet cells, or multiciliated cells using bioluminescence.
Collapse
Affiliation(s)
- Jessica C Orr
- Lungs for Living Research Centre, UCL Respiratory, University College London, 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
| | - Asma Laali
- 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
| | - Pascal F Durrenberger
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Kyren A Lazarus
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Marie-Belle El Mdawar
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Robert E Hynds
- 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
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
12
|
Cheng C, Katoch P, Zhong YP, Higgins CT, Moredock M, Chang MEK, Flory MR, Randell SH, Streeter PR. Identification of a Novel Subset of Human Airway Epithelial Basal Stem Cells. Int J Mol Sci 2024; 25:9863. [PMID: 39337350 PMCID: PMC11432080 DOI: 10.3390/ijms25189863] [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: 08/16/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The basal cell maintains the airway's respiratory epithelium as the putative resident stem cell. Basal cells are known to self-renew and differentiate into airway ciliated and secretory cells. However, it is not clear if every basal cell functions as a stem cell. To address functional heterogeneity amongst the basal cell population, we developed a novel monoclonal antibody, HLO1-6H5, that identifies a subset of KRT5+ (cytokeratin 5) basal cells. We used HLO1-6H5 and other known basal cell-reactive reagents to isolate viable airway subsets from primary human airway epithelium by Fluorescence Activated Cell Sorting. Isolated primary cell subsets were assessed for the stem cell capabilities of self-renewal and differentiation in the bronchosphere assay, which revealed that bipotent stem cells were, at minimum 3-fold enriched in the HLO1-6H5+ cell subset. Crosslinking-mass spectrometry identified the HLO1-6H5 target as a glycosylated TFRC/CD71 (transferrin receptor) proteoform. The HLO1-6H5 antibody provides a valuable new tool for identifying and isolating a subset of primary human airway basal cells that are substantially enriched for bipotent stem/progenitor cells and reveals TFRC as a defining surface marker for this novel cell subset.
Collapse
Affiliation(s)
- Christopher Cheng
- Oregon Stem Cell Center, Papè Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Parul Katoch
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Yong-Ping Zhong
- Oregon Stem Cell Center, Papè Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Claire T. Higgins
- Oregon Stem Cell Center, Papè Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Maria Moredock
- Oregon Stem Cell Center, Papè Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Matthew E. K. Chang
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Mark R. Flory
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Scott H. Randell
- Marsico Lung Institute/Cystic Fibrosis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7248, USA
| | - Philip R. Streeter
- Oregon Stem Cell Center, Papè Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239-3098, USA
| |
Collapse
|
13
|
Hynds RE, Magin CM, Ikonomou L, Aschner Y, Beers MF, Burgess JK, Heise RL, Hume PS, Krasnodembskaya AD, Mei SHJ, Misharin AV, Park JA, Reynolds SD, Tschumperlin DJ, Tanneberger AE, Vaidyanathan S, Waters CM, Zettler PJ, Weiss DJ, Ryan AL. Stem cells, cell therapies, and bioengineering in lung biology and diseases 2023. Am J Physiol Lung Cell Mol Physiol 2024; 327:L327-L340. [PMID: 38772903 PMCID: PMC11442098 DOI: 10.1152/ajplung.00052.2024] [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: 02/14/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/23/2024] Open
Abstract
Repair and regeneration of a diseased lung using stem cells or bioengineered tissues is an exciting therapeutic approach for a variety of lung diseases and critical illnesses. Over the past decade, increasing evidence from preclinical models suggests that mesenchymal stromal cells, which are not normally resident in the lung, can be used to modulate immune responses after injury, but there have been challenges in translating these promising findings to the clinic. In parallel, there has been a surge in bioengineering studies investigating the use of artificial and acellular lung matrices as scaffolds for three-dimensional lung or airway regeneration, with some recent attempts of transplantation in large animal models. The combination of these studies with those involving stem cells, induced pluripotent stem cell derivatives, and/or cell therapies is a promising and rapidly developing research area. These studies have been further paralleled by significant increases in our understanding of the molecular and cellular events by which endogenous lung stem and/or progenitor cells arise during lung development and participate in normal and pathological remodeling after lung injury. For the 2023 Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases Conference, scientific symposia were chosen to reflect the most cutting-edge advances in these fields. Sessions focused on the integration of "omics" technologies with function, the influence of immune cells on regeneration, and the role of the extracellular matrix in regeneration. The necessity for basic science studies to enhance fundamental understanding of lung regeneration and to design innovative translational studies was reinforced throughout the conference.
Collapse
Affiliation(s)
- Robert E Hynds
- Epithelial Cell Biology in ENT Research Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Chelsea M Magin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Laertis Ikonomou
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Michael F Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine and PENN-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Janette K Burgess
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Patrick S Hume
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, United States
| | - Susan D Reynolds
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
| | - Alicia E Tanneberger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Sriram Vaidyanathan
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Christopher M Waters
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Patricia J Zettler
- Moritz College of Law, Drug Enforcement and Policy Center, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont, United States
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
14
|
Ndongo Sonfack DJ, Tanguay Boivin C, Touzel Deschênes L, Maurand T, Maguemoun C, Berthod F, Gros-Louis F, Champagne PO. Bioengineering Human Upper Respiratory Mucosa: A Systematic Review of the State of the Art of Cell Culture Techniques. Bioengineering (Basel) 2024; 11:826. [PMID: 39199784 PMCID: PMC11352167 DOI: 10.3390/bioengineering11080826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND The upper respiratory mucosa plays a crucial role in both the physical integrity and immunological function of the respiratory tract. However, in certain situations such as infections, trauma, or surgery, it might sustain damage. Tissue engineering, a field of regenerative medicine, has found applications in various medical fields including but not limited to plastic surgery, ophthalmology, and urology. However, its application to the respiratory system remains somewhat difficult due to the complex morphology and histology of the upper respiratory tract. To date, a culture protocol for producing a handleable, well-differentiated nasal mucosa has yet to be developed. The objective of this review is to describe the current state of research pertaining to cell culture techniques used for producing autologous healthy human upper respiratory cells and mucosal tissues, as well as describe its clinical applications. METHODS A search of the relevant literature was carried out with no time restriction across Embase, Cochrane, PubMed, and Medline Ovid databases. Keywords related to "respiratory mucosa" and "culture techniques of the human airway" were the focus of the search strategy for this review. The risk of bias in retained studies was assessed using the Joanna Briggs Institute's (JBI) critical appraisal tools for qualitative research. A narrative synthesis of our results was then conducted. RESULTS A total of 33 studies were included in this review, and thirteen of these focused solely on developing a cell culture protocol without further use. The rest of the studies used their own developed protocol for various applications such as cystic fibrosis, pharmacological, and viral research. One study was able to develop a promising model for nasal mucosa that could be employed as a replacement in nasotracheal reconstructive surgery. CONCLUSIONS This systematic review extensively explored the current state of research regarding cell culture techniques for producing tissue-engineered nasal mucosa. Bioengineering the nasal mucosa holds great potential for clinical use. However, further research on mechanical properties is essential, as the comparison of engineered tissues is currently focused on morphology rather than comprehensive mechanical assessments.
Collapse
Affiliation(s)
- Davaine Joel Ndongo Sonfack
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
- LOEX, CHU de Québec—Laval University Research Center, Quebec, QC G1J 5B3, Canada;
- Laval University Neurosurgery Innovation Laboratory (LINUL), Quebec, QC G1J 5B3, Canada
- Department of Neurosurgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Clémence Tanguay Boivin
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
| | - Lydia Touzel Deschênes
- LOEX, CHU de Québec—Laval University Research Center, Quebec, QC G1J 5B3, Canada;
- Laval University Neurosurgery Innovation Laboratory (LINUL), Quebec, QC G1J 5B3, Canada
| | - Thibault Maurand
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
| | - Célina Maguemoun
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
| | - François Berthod
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
- LOEX, CHU de Québec—Laval University Research Center, Quebec, QC G1J 5B3, Canada;
| | - François Gros-Louis
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
- LOEX, CHU de Québec—Laval University Research Center, Quebec, QC G1J 5B3, Canada;
| | - Pierre-Olivier Champagne
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada; (C.T.B.); (T.M.); (C.M.); (F.B.); (F.G.-L.); (P.-O.C.)
- LOEX, CHU de Québec—Laval University Research Center, Quebec, QC G1J 5B3, Canada;
- Laval University Neurosurgery Innovation Laboratory (LINUL), Quebec, QC G1J 5B3, Canada
- Department of Neurosurgery, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
15
|
Stack JT, Rayner RE, Nouri R, Suarez CJ, Kim SH, Kanke KL, Vetter TA, Cormet-Boyaka E, Vaidyanathan S. DNA-PKcs Inhibition Improves Sequential Gene Insertion of the Full-Length CFTR cDNA in Airway Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607571. [PMID: 39185207 PMCID: PMC11343149 DOI: 10.1101/2024.08.12.607571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Although many people with CF (pwCF) are treated using CFTR modulators, some are non-responsive due to their genotype or other uncharacterized reasons. Autologous airway stem cell therapies, in which the CFTR cDNA has been replaced, may enable a durable therapy for all pwCF. Previously, CRISPR-Cas9 with two AAVs was used to sequentially insert two halves of the CFTR cDNA and an enrichment cassette into the CFTR locus. However, the editing efficiency was <10% and required enrichment to restore CFTR function. Further improvement in gene insertion may enhance cell therapy production. To improve CFTR cDNA insertion in human airway basal stem cells (ABCs), we evaluated the use of the small molecules AZD7648 and ART558 which inhibit non-homologous end joining (NHEJ) and micro-homology mediated end joining (MMEJ). Adding AZD7648 alone improved gene insertion by 2-3-fold. Adding both ART558 and AZD7648 improved gene insertion but induced toxicity. ABCs edited in the presence of AZD7648 produced differentiated airway epithelial sheets with restored CFTR function after enrichment. Adding AZD7648 did not increase off-target editing. Further studies are necessary to validate if AZD7648 treatment enriches cells with oncogenic mutations.
Collapse
Affiliation(s)
- Jacob T. Stack
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215
| | - Rachael E. Rayner
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Reza Nouri
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215
| | - Carlos J. Suarez
- Department of Pathology, Stanford University, Palo Alto, CA 94305
| | - Sun Hee Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Karen L. Kanke
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215
| | - Tatyana A. Vetter
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215
| | | | - Sriram Vaidyanathan
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43215
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
16
|
Lau CH, Rouhani MJ, Maughan EF, Orr JC, Kolluri KK, Pearce DR, Haughey EK, Sutton L, Flatau S, Balboa PL, Bageta ML, O'Callaghan C, Smith CM, Janes SM, Hewitt R, Petrof G, Martinez AE, McGrath JA, Butler CR, Hynds RE. Lentiviral expression of wild-type LAMA3A restores cell adhesion in airway basal cells from children with epidermolysis bullosa. Mol Ther 2024; 32:1497-1509. [PMID: 38429928 PMCID: PMC11081864 DOI: 10.1016/j.ymthe.2024.02.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/26/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
The hallmark of epidermolysis bullosa (EB) is fragile attachment of epithelia due to genetic variants in cell adhesion genes. We describe 16 EB patients treated in the ear, nose, and throat department of a tertiary pediatric hospital linked to the United Kingdom's national EB unit between 1992 and 2023. Patients suffered a high degree of morbidity and mortality from laryngotracheal stenosis. Variants in laminin subunit alpha-3 (LAMA3) were found in 10/15 patients where genotype was available. LAMA3 encodes a subunit of the laminin-332 heterotrimeric extracellular matrix protein complex and is expressed by airway epithelial basal stem cells. We investigated the benefit of restoring wild-type LAMA3 expression in primary EB patient-derived basal cell cultures. EB basal cells demonstrated weak adhesion to cell culture substrates, but could otherwise be expanded similarly to non-EB basal cells. In vitro lentiviral overexpression of LAMA3A in EB basal cells enabled them to differentiate in air-liquid interface cultures, producing cilia with normal ciliary beat frequency. Moreover, transduction restored cell adhesion to levels comparable to a non-EB donor culture. These data provide proof of concept for a combined cell and gene therapy approach to treat airway disease in LAMA3-affected EB.
Collapse
Affiliation(s)
- Chun Hang Lau
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, UCL Great Ormond Street Institute of Child Health, University College London, 20c Guilford Street, London WC1N 1DZ, UK
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK; Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Elizabeth F Maughan
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, UCL Great Ormond Street Institute of Child Health, University College London, 20c Guilford Street, London WC1N 1DZ, UK; Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Jessica C Orr
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, UCL Great Ormond Street Institute of Child Health, University College London, 20c Guilford Street, London WC1N 1DZ, UK; Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Krishna K Kolluri
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - David R Pearce
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Elizabeth K Haughey
- Infection, Immunity, and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Liam Sutton
- Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Sam Flatau
- Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Pablo Lopez Balboa
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Maria Laura Bageta
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Christopher O'Callaghan
- Infection, Immunity, and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Claire M Smith
- Infection, Immunity, and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, Division of Medicine, University College London, 5 University Street, London WC1E 6JF, UK
| | - Richard Hewitt
- Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Gabriela Petrof
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - Anna E Martinez
- Department of Dermatology, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, Guy's Hospital, St Thomas Street, London SE1 9RT, UK
| | - Colin R Butler
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, UCL Great Ormond Street Institute of Child Health, University College London, 20c Guilford Street, London WC1N 1DZ, UK; Ear, Nose, and Throat Department, Great Ormond Street Hospital NHS Foundation Trust, London WC1N 3JH, UK.
| | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, UCL Great Ormond Street Institute of Child Health, University College London, 20c Guilford Street, London WC1N 1DZ, UK; UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
17
|
Zhu J, Lu Y, Shan Y, Yuan L, Wu Q, Shen Z, Sun F, Shi H. Global Bibliometric and Visualized Analysis of Tracheal Tissue Engineering Research. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:198-216. [PMID: 37658839 DOI: 10.1089/ten.teb.2023.0129] [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: 09/05/2023]
Abstract
The development of tracheal tissue engineering (TTE) has seen a rapid growth in recent years. The purpose of this study was to investigate the global status, trends, and hotspots of TTE research based on bibliometrics and visualization analysis. Publications related to TTE were retrieved and included in the Web of Science Core Collection. VOSviewer and CiteSpace were used to generate knowledge maps. Six hundred fifty-five publications were identified, and the quantity of the annual publications worldwide was on the increase. International collaboration is a widespread reality. The United States led the world in the field of trachea tissue engineering, whereas University College London was the institution with the greatest contribution. In addition, Biomaterials had a great influence in this field, attracting the largest number of papers. Moreover, the topics of TTE research largely concentrated on the biomechanical scaffold preparation, the vascularization and epithelialization of scaffold, the tracheal cartilage regeneration, and the tissue-engineered tracheal transplantation. And the research on the application of decellularization and 3D printing for the construction of a tissue-engineered trachea was likely to receive more widespread attention in the future. Impact statement In recent years, tracheal tissue engineering (TTE) has experienced rapid growth. In this study, we investigated the worldwide status and trends of TTE research, and revealed the countries, institutions, journals, and authors that had made significant contributions to the field of TTE. Moreover, the possible research hotspots in the future were predicted. According to our research, researchers can gain a better understanding of the trends in this field, and stay informed of the most current research by tracking key journals, institutions, and authors.
Collapse
Affiliation(s)
- Jianwei Zhu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yi Lu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Yibo Shan
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Lei Yuan
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Qiang Wu
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiming Shen
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Fei Sun
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Hongcan Shi
- Clinical Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Rouhani MJ, Janes SM, Kim CF. Epithelial stem and progenitor cells of the upper airway. Cells Dev 2024; 177:203905. [PMID: 38355015 DOI: 10.1016/j.cdev.2024.203905] [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: 12/29/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024]
Abstract
The upper airway acts as a conduit for the passage of air to the respiratory system and is implicated in several chronic diseases. Whilst the cell biology of the distal respiratory system has been described in great detail, less is known about the proximal upper airway. In this review, we describe the relevant anatomy of the upper airway and discuss the literature detailing the identification and roles of the progenitor cells of these regions.
Collapse
Affiliation(s)
- Maral J Rouhani
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Sam M Janes
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Carla F Kim
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Lee RE, Mascenik TM, Major SC, Galiger JR, Bulik-Sullivan E, Siesser PF, Lewis CA, Bear JE, Le Suer JA, Hawkins FJ, Pickles RJ, Randell SH. Viral airway injury promotes cell engraftment in an in vitro model of cystic fibrosis cell therapy. Am J Physiol Lung Cell Mol Physiol 2024; 326:L226-L238. [PMID: 38150545 PMCID: PMC11280688 DOI: 10.1152/ajplung.00421.2022] [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: 12/13/2022] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
Cell therapy is a potential treatment for cystic fibrosis (CF). However, cell engraftment into the airway epithelium is challenging. Here, we model cell engraftment in vitro using the air-liquid interface (ALI) culture system by injuring well-differentiated CF ALI cultures and delivering non-CF cells at the time of peak injury. Engraftment efficiency was quantified by measuring chimerism by droplet digital PCR and functional ion transport in Ussing chambers. Using this model, we found that human bronchial epithelial cells (HBECs) engraft more efficiently when they are cultured by conditionally reprogrammed cell (CRC) culture methods. Cell engraftment into the airway epithelium requires airway injury, but the extent of injury needed is unknown. We compared three injury models and determined that severe injury with partial epithelial denudation facilitates long-term cell engraftment and functional CFTR recovery up to 20% of wildtype function. The airway epithelium promptly regenerates in response to injury, creating competition for space and posing a barrier to effective engraftment. We examined competition dynamics by time-lapse confocal imaging and found that delivered cells accelerate airway regeneration by incorporating into the epithelium. Irradiating the repairing epithelium granted engrafting cells a competitive advantage by diminishing resident stem cell proliferation. Intentionally, causing severe injury to the lungs of people with CF would be dangerous. However, naturally occurring events like viral infection can induce similar epithelial damage with patches of denuded epithelium. We found that viral preconditioning promoted effective engraftment of cells primed for viral resistance.NEW & NOTEWORTHY Cell therapy is a potential treatment for cystic fibrosis (CF). Here, we model cell engraftment by injuring CF air-liquid interface cultures and delivering non-CF cells. Successful engraftment required severe epithelial injury. Intentionally injuring the lungs to this extent would be dangerous. However, naturally occurring events like viral infection induce similar epithelial damage. We found that viral preconditioning promoted the engraftment of cells primed for viral resistance leading to CFTR functional recovery to 20% of the wildtype.
Collapse
Affiliation(s)
- Rhianna E Lee
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Teresa M Mascenik
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Sidra C Major
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jacob R Galiger
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Emily Bulik-Sullivan
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Priscila F Siesser
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Catherine A Lewis
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jake A Le Suer
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, United States
- Department of Medicine, The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Finn J Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, United States
- Department of Medicine, The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Raymond J Pickles
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
20
|
Wu M, Zhang X, Tu Y, Cheng W, Zeng Y. Culture and expansion of murine proximal airway basal stem cells. Stem Cell Res Ther 2024; 15:26. [PMID: 38287366 PMCID: PMC10826159 DOI: 10.1186/s13287-024-03642-2] [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: 08/01/2023] [Accepted: 01/21/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND The stem cell characteristic makes basal cells desirable for ex vivo modeling of airway diseases. However, to date, approaches allowing them extensively in vitro serial expansion and maintaining bona fide stem cell property are still awaiting to be established. This study aims to develop a feeder-free culture system of mouse airway basal stem cells (ABSCs) that sustain their stem cell potential in vitro, providing an experimental basis for further in-depth research and mechanism exploration. METHODS We used ROCK inhibitor Y-27632-containing 3T3-CM, MEF-CM, and RbEF-CM to determine the proper feeder-free culture system that could maintain in vitro stem cell morphology of mouse ABSCs. Immunocytofluorescence was used to identify the basal cell markers of obtained cells. Serial propagation was carried out to observe whether the stem cell morphology and basal cell markers could be preserved in this cultivation system. Next, we examined the in vitro expansion and self-renewal ability by evaluating population doubling time and colony-forming efficiency. Moreover, the differentiation potential was detected by an in vitro differentiation culture and a 3D tracheosphere assay. RESULTS When the mouse ABSCs were cultured using 3T3-CM containing ROCK inhibitor Y-27632 in combination with Matrigel-coated culture dishes, they could stably expand and maintain stem cell-like clones. We confirmed that the obtained clones comprised p63/Krt5 double-positive ABSCs. In continuous passage and maintenance culture, we found that it could be subculture to at least 15 passages in vitro, stably maintaining its stem cell morphology, basal cell markers, and in vitro expansion and self-renewal capabilities. Meanwhile, through in vitro differentiation culture and 3D tracheosphere culture, we found that in addition to maintaining self-renewal, mouse ABSCs could differentiate into other airway epithelial cells such as acetylated tubulin (Act-Tub) + ciliated and MUC5AC + mucus-secreting cells. However, they failed to differentiate into alveoli epithelial cells, including alveolar type I and alveolar type II. CONCLUSION We established an in vitro feeder-free culture system that allows mouse ABSCs to maintain their stem cell characteristics, including self-renewal and airway epithelium differentiation potential, while keeping up in vitro expansion stability.
Collapse
Affiliation(s)
- Meirong Wu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Xiaojing Zhang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yanjuan Tu
- Department of Pathology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Wenzhao Cheng
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, People's Republic of China.
| |
Collapse
|
21
|
Brand M, Ritzmann F, Kattler K, Milasius D, Yao Y, Herr C, Kirsch SH, Müller R, Yildiz D, Bals R, Beisswenger C. Biochemical and transcriptomic evaluation of a 3D lung organoid platform for pre-clinical testing of active substances targeting senescence. Respir Res 2024; 25:3. [PMID: 38172839 PMCID: PMC10765931 DOI: 10.1186/s12931-023-02636-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Chronic lung diseases such as chronic obstructive pulmonary disease and cystic fibrosis are incurable. Epithelial senescence, a state of dysfunctional cell cycle arrest, contributes to the progression of such diseases. Therefore, lung epithelial cells are a valuable target for therapeutic intervention. Here, we present a 3D airway lung organoid platform for the preclinical testing of active substances with regard to senescence, toxicity, and inflammation under standardized conditions in a 96 well format. Senescence was induced with doxorubicin and measured by activity of senescence associated galactosidase. Pharmaceutical compounds such as quercetin antagonized doxorubicin-induced senescence without compromising organoid integrity. Using single cell sequencing, we identified a subset of cells expressing senescence markers which was decreased by quercetin. Doxorubicin induced the expression of detoxification factors specifically in goblet cells independent of quercetin. In conclusion, our platform enables for the analysis of senescence-related processes and will allow the pre-selection of a wide range of compounds (e.g. natural products) in preclinical studies, thus reducing the need for animal testing.
Collapse
Affiliation(s)
- Michelle Brand
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Felix Ritzmann
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123, Saarbrücken, Germany
| | - Kathrin Kattler
- Department of Genetics/Epigenetics, Saarland University, 66123, Saarbrücken, Germany
| | - Deivydas Milasius
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Yiwen Yao
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
| | - Susanne H Kirsch
- Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123, Saarbrücken, Germany
| | - Rolf Müller
- Department of Microbial Natural Products (MINS), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology, PZMS, and Center for Human and Molecular Biology (ZHMB), Saarland University, 66421, Homburg, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123, Saarbrücken, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
22
|
Vazquez-Armendariz AI, Tata PR. Recent advances in lung organoid development and applications in disease modeling. J Clin Invest 2023; 133:e170500. [PMID: 37966116 PMCID: PMC10645385 DOI: 10.1172/jci170500] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Over the last decade, several organoid models have evolved to acquire increasing cellular, structural, and functional complexity. Advanced lung organoid platforms derived from various sources, including adult, fetal, and induced pluripotent stem cells, have now been generated, which more closely mimic the cellular architecture found within the airways and alveoli. In this regard, the establishment of novel protocols with optimized stem cell isolation and culture conditions has given rise to an array of models able to study key cellular and molecular players involved in lung injury and repair. In addition, introduction of other nonepithelial cellular components, such as immune, mesenchymal, and endothelial cells, and employment of novel precision gene editing tools have further broadened the range of applications for these systems by providing a microenvironment and/or phenotype closer to the desired in vivo scenario. Thus, these developments in organoid technology have enhanced our ability to model various aspects of lung biology, including pathogenesis of diseases such as chronic obstructive pulmonary disease, pulmonary fibrosis, cystic fibrosis, and infectious disease and host-microbe interactions, in ways that are often difficult to undertake using only in vivo models. In this Review, we summarize the latest developments in lung organoid technology and their applicability for disease modeling and outline their strengths, drawbacks, and potential avenues for future development.
Collapse
Affiliation(s)
- Ana I. Vazquez-Armendariz
- University of Bonn, Transdisciplinary Research Area Life and Health, Organoid Biology, Life & Medical Sciences Institute, Bonn, Germany
- Department of Medicine V, Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research and Institute for Lung Health, Giessen, Germany
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
23
|
Lee DF, Everest DJ, Cooley W, Chambers MA. Investigation of nasal epithelial cells as a surrogate for bronchial epithelial cells in the research of equine asthma. PLoS One 2023; 18:e0293956. [PMID: 37943759 PMCID: PMC10635438 DOI: 10.1371/journal.pone.0293956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Equine asthma, previously known as Recurrent Airway Obstruction (RAO) or Inflammatory Airway Disease (IAD), is an often-debilitating condition that may severely affect both performance and quality of life. Research is hindered by the low sample numbers of subjects recruited to studies, a consequence in part of the invasive nature of the sampling methods of bronchial brushing and biopsy. We present an alternative method of sampling equine airway epithelial cells, the 'nasal brush method' (NBM). Obtained by light brushing of the ventral meatus whilst the horse is under standing sedation, these cells express the same markers of differentiation as their deeper counterparts. Grown as 3-D spheroids or as air-liquid interface cultures, nasal epithelial cells are responsive to the inflammatory cytokine interleukin-13. This may be attenuated by modulation of the Notch signalling pathway using the gamma-secretase inhibitor Semagecestat; a previously unreported finding that cements the link between equine and human asthma research and strengthens the case for a One Health approach in researching asthma pathophysiology and therapeutic intervention.
Collapse
Affiliation(s)
- Diane Frances Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| | | | - William Cooley
- Animal and Plant Health Agency, Addlestone, Surrey, United Kingdom
| | - Mark Andrew Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, United Kingdom
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| |
Collapse
|
24
|
Nizamoglu M, Joglekar MM, Almeida CR, Larsson Callerfelt AK, Dupin I, Guenat OT, Henrot P, van Os L, Otero J, Elowsson L, Farre R, Burgess JK. Innovative three-dimensional models for understanding mechanisms underlying lung diseases: powerful tools for translational research. Eur Respir Rev 2023; 32:230042. [PMID: 37495250 PMCID: PMC10369168 DOI: 10.1183/16000617.0042-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/04/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic lung diseases result from alteration and/or destruction of lung tissue, inevitably causing decreased breathing capacity and quality of life for patients. While animal models have paved the way for our understanding of pathobiology and the development of therapeutic strategies for disease management, their translational capacity is limited. There is, therefore, a well-recognised need for innovative in vitro models to reflect chronic lung diseases, which will facilitate mechanism investigation and the advancement of new treatment strategies. In the last decades, lungs have been modelled in healthy and diseased conditions using precision-cut lung slices, organoids, extracellular matrix-derived hydrogels and lung-on-chip systems. These three-dimensional models together provide a wide spectrum of applicability and mimicry of the lung microenvironment. While each system has its own limitations, their advantages over traditional two-dimensional culture systems, or even over animal models, increases the value of in vitro models. Generating new and advanced models with increased translational capacity will not only benefit our understanding of the pathobiology of lung diseases but should also shorten the timelines required for discovery and generation of new therapeutics. This article summarises and provides an outline of the European Respiratory Society research seminar "Innovative 3D models for understanding mechanisms underlying lung diseases: powerful tools for translational research", held in Lisbon, Portugal, in April 2022. Current in vitro models developed for recapitulating healthy and diseased lungs are outlined and discussed with respect to the challenges associated with them, efforts to develop best practices for model generation, characterisation and utilisation of models and state-of-the-art translational potential.
Collapse
Affiliation(s)
- Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Mugdha M Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Catarina R Almeida
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | | | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Olivier T Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- Service d'exploration fonctionnelle respiratoire, CHU de Bordeaux, Pessac, France
| | - Lisette van Os
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ramon Farre
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, The Netherlands
| |
Collapse
|
25
|
Tang H, Sun W, Liu X, Gao Q, Chen Y, Xie C, Lin W, Chen J, Wang L, Fan Z, Zhang L, Ren Y, She Y, He Y, Chen C. A bioengineered trachea-like structure improves survival in a rabbit tracheal defect model. Sci Transl Med 2023; 15:eabo4272. [PMID: 37729433 DOI: 10.1126/scitranslmed.abo4272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
A practical strategy for engineering a trachea-like structure that could be used to repair or replace a damaged or injured trachea is an unmet need. Here, we fabricated bioengineered cartilage (BC) rings from three-dimensionally printed fibers of poly(ɛ-caprolactone) (PCL) and rabbit chondrocytes. The extracellular matrix (ECM) secreted by the chondrocytes combined with the PCL fibers formed a "concrete-rebar structure," with ECM deposited along the PCL fibers, forming a grid similar to that of native cartilage. PCL fiber-hydrogel rings were then fabricated and alternately stacked with BC rings on silicone tubes. This trachea-like structure underwent vascularization after heterotopic transplantation into rabbits for 4 weeks. The vascularized bioengineered trachea-like structure was then orthotopically transplanted by end-to-end anastomosis to native rabbit trachea after a segment of trachea had been resected. The bioengineered trachea-like structure displayed mechanical properties similar to native rabbit trachea and transmural angiogenesis between the rings. The 8-week survival rate in transplanted rabbits was 83.3%, and the respiratory rate of these animals was similar to preoperative levels. This bioengineered trachea-like structure may have potential for treating tracheal stenosis and other tracheal injuries.
Collapse
Affiliation(s)
- Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Chaoqi Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Ziwen Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yijiu Ren
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai 200433, China
| |
Collapse
|
26
|
Ma L, Thapa BR, Le Suer JA, Tilston-Lünel A, Herriges MJ, Berical A, Beermann ML, Wang F, Bawa PS, Kohn A, Ysasi AB, Kiyokawa H, Matte TM, Randell SH, Varelas X, Hawkins FJ, Kotton DN. Airway stem cell reconstitution by the transplantation of primary or pluripotent stem cell-derived basal cells. Cell Stem Cell 2023; 30:1199-1216.e7. [PMID: 37625411 PMCID: PMC10528754 DOI: 10.1016/j.stem.2023.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/13/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023]
Abstract
Life-long reconstitution of a tissue's resident stem cell compartment with engrafted cells has the potential to durably replenish organ function. Here, we demonstrate the engraftment of the airway epithelial stem cell compartment via intra-airway transplantation of mouse or human primary and pluripotent stem cell (PSC)-derived airway basal cells (BCs). Murine primary or PSC-derived BCs transplanted into polidocanol-injured syngeneic recipients give rise for at least two years to progeny that stably display the morphologic, molecular, and functional phenotypes of airway epithelia. The engrafted basal-like cells retain extensive self-renewal potential, evident by the capacity to reconstitute the tracheal epithelium through seven generations of secondary transplantation. Using the same approach, human primary or PSC-derived BCs transplanted into NOD scid gamma (NSG) recipient mice similarly display multilineage airway epithelial differentiation in vivo. Our results may provide a step toward potential future syngeneic cell-based therapy for patients with diseases resulting from airway epithelial cell damage or dysfunction.
Collapse
Affiliation(s)
- Liang Ma
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bibek R Thapa
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Department of Biology, Boston University, Boston, MA 02215, USA
| | - Jake A Le Suer
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew Tilston-Lünel
- The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Michael J Herriges
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew Berical
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mary Lou Beermann
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Feiya Wang
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Pushpinder S Bawa
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anat Kohn
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alexandra B Ysasi
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hirofumi Kiyokawa
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Taylor M Matte
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Finn J Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
27
|
Lo Cicero S, Castelli G, Blaconà G, Bruno SM, Sette G, Pigliucci R, Villella VR, Esposito S, Zollo I, Spadaro F, Maria RD, Biffoni M, Cimino G, Amato F, Lucarelli M, Eramo A. L1077P CFTR pathogenic variant function rescue by Elexacaftor-Tezacaftor-Ivacaftor in cystic fibrosis patient-derived air-liquid interface (ALI) cultures and organoids: in vitro guided personalized therapy of non-F508del patients. Respir Res 2023; 24:217. [PMID: 37674160 PMCID: PMC10483775 DOI: 10.1186/s12931-023-02516-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/17/2023] [Indexed: 09/08/2023] Open
Abstract
Cystic fibrosis (CF) is caused by defects of the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR-modulating drugs may overcome specific defects, such as the case of Trikafta, which is a clinically approved triple combination of Elexacaftor, Tezacaftor and Ivacaftor (ETI) that exhibited a strong ability to rescue the function of the most frequent F508del pathogenic variant even in genotypes with the mutated allele in single copy. Nevertheless, most rare genotypes lacking the F508del allele are still not eligible for targeted therapies. Via the innovative approach of using nasal conditionally reprogrammed cell (CRC) cell-based models that mimic patient disease in vitro, which are obtainable from each patient due to the 100% efficiency of the cell culture establishment, we theratyped orphan CFTR mutation L1077P. Protein studies, Forskolin-induced organoid swelling, and Ussing chamber assays congruently proved the L1077P variant function rescue by ETI. Notably, this rescue takes place even in the context of a single-copy L1077P allele, which appears to enhance its expression. Thus, the possibility of single-allele treatment also arises for rare genotypes, with an allele-specific modulation as part of the mechanism. Of note, besides providing indication of drug efficacy with respect to specific CFTR pathogenic variants or genotypes, this approach allows the evaluation of the response of single-patient cells within their genetic background. In this view, our studies support in vitro guided personalized CF therapies also for rare patients who are nearly excluded from clinical trials.
Collapse
Affiliation(s)
- Stefania Lo Cicero
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Blaconà
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Sabina Maria Bruno
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Sette
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Riccardo Pigliucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valeria Rachela Villella
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l, Naples, Italy
| | - Speranza Esposito
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l, Naples, Italy
| | - Immacolata Zollo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l, Naples, Italy
| | - Francesca Spadaro
- Confocal Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario 'A. Gemelli'-IRCCS, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Cimino
- Cystic Fibrosis Reference Center of Lazio Region, AOU Policlinico Umberto I, Rome, Italy
| | - Felice Amato
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l, Naples, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
28
|
Keiser NW, Cant E, Sitaraman S, Shoemark A, Limberis MP. Restoring Ciliary Function: Gene Therapeutics for Primary Ciliary Dyskinesia. Hum Gene Ther 2023; 34:821-835. [PMID: 37624733 DOI: 10.1089/hum.2023.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is a genetic disease characterized by defects in motile cilia, which play an important role in several organ systems. Lung disease is a hallmark of PCD, given the essential role of cilia in airway surface defense. Diagnosis of PCD is complicated due to its reliance on complex tests that are not utilized by every clinic and also its phenotypic overlap with several other respiratory diseases. Nonetheless, PCD is increasingly being recognized as more common than once thought. The disease is genetically complex, with several genes reported to be associated with PCD. There is no cure for PCD, but gene therapy remains a promising therapeutic strategy. In this review, we provide an overview of the clinical symptoms, diagnosis, genetics, and current treatment regimens for PCD. We also describe PCD model systems and discuss the therapeutic potential of different gene therapeutics for targeting the intended cellular target, the ciliated cells of the airway.
Collapse
Affiliation(s)
| | - Erin Cant
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | | |
Collapse
|
29
|
Matkovic Leko I, Schneider RT, Thimraj TA, Schrode N, Beitler D, Liu HY, Beaumont K, Chen YW, Snoeck HW. A distal lung organoid model to study interstitial lung disease, viral infection and human lung development. Nat Protoc 2023; 18:2283-2312. [PMID: 37165073 PMCID: PMC11486529 DOI: 10.1038/s41596-023-00827-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/24/2023] [Indexed: 05/12/2023]
Abstract
Organoids have been an exciting advancement in stem cell research. Here we describe a strategy for directed differentiation of human pluripotent stem cells into distal lung organoids. This protocol recapitulates lung development by sequentially specifying human pluripotent stem cells to definitive endoderm, anterior foregut endoderm, ventral anterior foregut endoderm, lung bud organoids and finally lung organoids. The organoids take ~40 d to generate and can be maintained more than 180 d, while progressively maturing up to a stage consistent with the second trimester of human gestation. They are unique because of their branching morphology, the near absence of non-lung endodermal lineages, presence of mesenchyme and capacity to recapitulate interstitial lung diseases. This protocol can be performed by anyone familiar with cell culture techniques, is conducted in serum-free conditions and does not require lineage-specific reporters or enrichment steps. We also provide a protocol for the generation of single-cell suspensions for single-cell RNA sequencing.
Collapse
Affiliation(s)
- Ivana Matkovic Leko
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Remy T Schneider
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tania A Thimraj
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Nadine Schrode
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Center for Advanced Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Beitler
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Hsiao-Yun Liu
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristin Beaumont
- Department of Genetic and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Center for Advanced Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ya-Wen Chen
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Institute for Airway Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hans-Willem Snoeck
- Columbia Center for Human Development/Center for Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
30
|
Rani AQ, Nurmemet D, Liffick J, Khan A, Mitchell D, Li J, Zhao B, Liu X. Conditional Cell Reprogramming and Air-Liquid Interface Modeling Life Cycle of Oncogenic Viruses (HPV and EBV) in Epithelial Cells and Virus-Associated Human Carcinomas. Viruses 2023; 15:1388. [PMID: 37376685 DOI: 10.3390/v15061388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Several oncogenic viruses are associated with approximately 20% of human cancers. Experimental models are crucial for studying the pathogenicity and biological aspects of oncogenic viruses and their potential mechanisms in tumorigenesis. Current cell models have considerable limitations such as: their low yield, genetic and epigenetic modification, and reduction in tumor heterogeneity during long propagation. Cancer cell lines are limited and not appropriate for studying the viral life cycle, for example, natural viral life cycles of HPV and EBV, and their persistence and latency in epithelial cells are poorly understood, since these processes are highly related to epithelial differentiation. Therefore, there is an urgent need of reliable human physiological cell models to study viral life cycle and cancer initiation. Conditional cell reprogramming (CCR) is a rapid and robust cell culture system, where the cells can be established from minimally invasive or noninvasive specimens and their lineage functions preserved during the long-term culture. These CR cells retain their ability to differentiate at air-liquid interface (ALI). Here, we recapitulated the applications of CR and ALI approaches in modeling host-virus interactions and viral-mediated tumorigenesis.
Collapse
Affiliation(s)
- Abdul Qawee Rani
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Dilber Nurmemet
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Joseph Liffick
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Anam Khan
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Darrion Mitchell
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Jenny Li
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Bo Zhao
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Departments of Pathology, Urology and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
31
|
Dai H, Zheng Q, Shan S, Lei Y, Zhang J, Zhong C, Xu Y, Liu Z, Ren T. A novel type of serum‑free medium for culturing human airway epithelium cells. Exp Ther Med 2023; 25:239. [PMID: 37114176 PMCID: PMC10126810 DOI: 10.3892/etm.2023.11938] [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: 12/01/2022] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
The present study aimed to evaluate the ability of a novel serum-free medium (SFM) to culture human airway epithelium cells (hAECs). hAECs were cultured in the novel SFM as the experimental group in the PneumaCult-Ex medium and Dulbecco's modified eagle medium (DMEM) and fetal bovine serum (FBS) as the control groups. Cell morphology, proliferative capacity, differentiation capacity and expression levels of basal cell markers were assessed accordingly in both culture systems. Optical microscope photos of hAECs were collected for cell morphology assessment. Cell Counting Kit-8 assay was conducted to assess the proliferation ability, and an air-liquid interface (ALI) assay was conducted to assess the differentiation capacity. Markers for proliferating basal and differentiated cells were relatively identified by immunohistochemical and immunofluorescent analysis. The results show that whether grown in the novel SFM or Ex medium, hAECs exhibited similar morphology at every passage, whereas cells could hardly form colonies in the DMEM + FBS group. Cells typically exhibited cobblestone shape, while a proportion of them in the novel SFM at late passage exhibited a larger shape. White vesicles appeared in the cytoplasm of some control cells at the later stage of culture. Basal cell markers (P63+KRT5+KI67+CC10-) for proliferating ability were found in the hAECs cultured by the novel SFM and Ex medium. hAECs at passage 3 cultured in the novel SFM and Ex medium both had the capacity to differentiate into ciliated cells (acetylated tubulin+), goblet cells (MUC5AC+) and club cells (CC10+) in the ALI culture assay. In conclusion, the novel SFM was capable of culturing hAECs. The hAECs cultured by the novel SFM could proliferate and differentiate in vitro. The novel SFM does not change the morphological characteristics or biomarkers of hAECs. The novel SFM has the potential for the amplification of hAECs for scientific research and clinical application.
Collapse
Affiliation(s)
- Haotian Dai
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Qi Zheng
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Shan Shan
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yuqiong Lei
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Jingyuan Zhang
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Cheng Zhong
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yongle Xu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Zeyu Liu
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
- Correspondence to: Professor Tao Ren or Dr Zeyu Liu, Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui, Shanghai 200233, P.R. China
| | - Tao Ren
- Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
- Correspondence to: Professor Tao Ren or Dr Zeyu Liu, Department of Respiratory Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Xuhui, Shanghai 200233, P.R. China
| |
Collapse
|
32
|
Ahmed TA, Eldaly B, Eldosuky S, Elkhenany H, El-Derby AM, Elshazly MF, El-Badri N. The interplay of cells, polymers, and vascularization in three-dimensional lung models and their applications in COVID-19 research and therapy. Stem Cell Res Ther 2023; 14:114. [PMID: 37118810 PMCID: PMC10144893 DOI: 10.1186/s13287-023-03341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Millions of people have been affected ever since the emergence of the corona virus disease of 2019 (COVID-19) outbreak, leading to an urgent need for antiviral drug and vaccine development. Current experimentation on traditional two-dimensional culture (2D) fails to accurately mimic the in vivo microenvironment for the disease, while in vivo animal model testing does not faithfully replicate human COVID-19 infection. Human-based three-dimensional (3D) cell culture models such as spheroids, organoids, and organ-on-a-chip present a promising solution to these challenges. In this report, we review the recent 3D in vitro lung models used in COVID-19 infection and drug screening studies and highlight the most common types of natural and synthetic polymers used to generate 3D lung models.
Collapse
Affiliation(s)
- Toka A Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Bassant Eldaly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Shadwa Eldosuky
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 22785, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Muhamed F Elshazly
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12582, Egypt.
| |
Collapse
|
33
|
Deguchi K, Zambaiti E, De Coppi P. Regenerative medicine: current research and perspective in pediatric surgery. Pediatr Surg Int 2023; 39:167. [PMID: 37014468 PMCID: PMC10073065 DOI: 10.1007/s00383-023-05438-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/05/2023]
Abstract
The field of regenerative medicine, encompassing several disciplines including stem cell biology and tissue engineering, continues to advance with the accumulating research on cell manipulation technologies, gene therapy and new materials. Recent progress in preclinical and clinical studies may transcend the boundaries of regenerative medicine from laboratory research towards clinical reality. However, for the ultimate goal to construct bioengineered transplantable organs, a number of issues still need to be addressed. In particular, engineering of elaborate tissues and organs requires a fine combination of different relevant aspects; not only the repopulation of multiple cell phenotypes in an appropriate distribution but also the adjustment of the host environmental factors such as vascularisation, innervation and immunomodulation. The aim of this review article is to provide an overview of the recent discoveries and development in stem cells and tissue engineering, which are inseparably interconnected. The current status of research on tissue stem cells and bioengineering, and the possibilities for application in specific organs relevant to paediatric surgery have been specifically focused and outlined.
Collapse
Affiliation(s)
- Koichi Deguchi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Elisa Zambaiti
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK
- UOC Chirurgia Pediatrica, Ospedale Infantile Regina Margherita, Turin, Italy
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, London, UK.
- NIHR BRC SNAPS Great Ormond Street Hospitals, London, UK.
- Stem Cells and Regenerative Medicine Section, Faculty of Population Health Sciences, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
34
|
Kasai Y, Morino T, Nakayama T, Yamamoto K, Kojima H. Analysis of the potential of human cultured nasal epithelial cell sheets to differentiate into airway epithelium. FASEB Bioadv 2023; 5:89-100. [PMID: 36876298 PMCID: PMC9983074 DOI: 10.1096/fba.2022-00106] [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: 10/12/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Understanding the expected efficacy and safety of a new regenerative therapy requires analysis of the fate of the transplanted cell graft. We have shown that transplantation of autologous cultured nasal epithelial cell sheets onto the middle ear mucosa can improve middle ear aeration and hearing. However, it remains unknown whether cultured nasal epithelial cell sheets have the potential to gain mucociliary function in the environment of the middle ear because sampling cell sheets after transplantation is challenging. The present study re-cultured cultured nasal epithelial cell sheets in different culture media and evaluated whether the sheets have the potential to differentiate into airway epithelium. Before re-cultivation, cultured nasal epithelial cell sheets fabricated in keratinocyte culture medium (KCM) contained no FOXJ1-positive and acetyl-α-tubulin-positive multiciliated cells or MUC5AC-positive mucus cells. Interestingly, multiciliated cells and mucus cells were observed when the cultured nasal epithelial cell sheets were re-cultured in conditions that promote differentiation of airway epithelium. However, multiciliated cells, mucus cells and CK1-positive keratinized cells were not observed when cultured nasal epithelial cell sheets were re-cultured in conditions that promote epithelial keratinization. These findings support the suggestion that cultured nasal epithelial cell sheets have the ability to differentiate and gain mucociliary function in response to an appropriate environment (possibly including the environment found in the middle ear) but are unable to develop into an epithelial type that differs from its origins.
Collapse
Affiliation(s)
- Yoshiyuki Kasai
- Department of OtorhinolaryngologyThe Jikei University School of MedicineTokyoJapan
| | - Tsunetaro Morino
- Department of OtorhinolaryngologyThe Jikei University School of MedicineTokyoJapan
| | - Tsuguhisa Nakayama
- Department of OtorhinolaryngologyThe Jikei University School of MedicineTokyoJapan
- Department of Otorhinolaryngology, Head and Neck SurgeryDokkyo Medical UniversityTochigiJapan
| | - Kazuhisa Yamamoto
- Department of OtorhinolaryngologyThe Jikei University School of MedicineTokyoJapan
| | - Hiromi Kojima
- Department of OtorhinolaryngologyThe Jikei University School of MedicineTokyoJapan
| |
Collapse
|
35
|
Chernokal B, Gonyea CR, Gleghorn JP. Lung Development in a Dish: Models to Interrogate the Cellular Niche and the Role of Mechanical Forces in Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:29-48. [PMID: 37195525 DOI: 10.1007/978-3-031-26625-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Over the past decade, emphasis has been placed on recapitulating in vitro the architecture and multicellular interactions found in organs in vivo [1, 2]. Whereas traditional reductionist approaches to in vitro models enable teasing apart the precise signaling pathways, cellular interactions, and response to biochemical and biophysical cues, model systems that incorporate higher complexity are needed to ask questions about physiology and morphogenesis at the tissue scale. Significant advancements have been made in establishing in vitro models of lung development to understand cell-fate specification, gene regulatory networks, sexual dimorphism, three-dimensional organization, and how mechanical forces interact to drive lung organogenesis [3-5]. In this chapter, we highlight recent advances in the rapid development of various lung organoids, organ-on-a-chip models, and whole lung ex vivo explant models currently used to dissect the roles of these cellular signals and mechanical cues in lung development and potential avenues for future investigation (Fig. 3.1).
Collapse
Affiliation(s)
- Brea Chernokal
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Cailin R Gonyea
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
36
|
Dean CH, Cheong SS. Simple Models of Lung Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:17-28. [PMID: 37195524 DOI: 10.1007/978-3-031-26625-6_2] [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
Models are essential to further our understanding of lung development and regeneration and to facilitate identification and testing of potential treatments for lung diseases. A wide variety of rodent and human models are available that recapitulate one or more stages of lung development. This chapter describes the existing 'simple' in vitro, in silico and ex vivo models of lung development. We define which stage(s) of development each model recapitulates and highlight their pros and cons.
Collapse
Affiliation(s)
- Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Sek-Shir Cheong
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
37
|
Yang M, Chen J, Chen Y, Lin W, Tang H, Fan Z, Wang L, She Y, Jin F, Zhang L, Sun W, Chen C. Scaffold-Free Tracheal Engineering via a Modular Strategy Based on Cartilage and Epithelium Sheets. Adv Healthc Mater 2023; 12:e2202022. [PMID: 36461102 DOI: 10.1002/adhm.202202022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/11/2022] [Indexed: 12/04/2022]
Abstract
Tracheal defects lead to devastating problems, and practical clinical substitutes that have complex functional structures and can avoid adverse influences from exogenous bioscaffolds are lacking. Herein, a modular strategy for scaffold-free tracheal engineering is developed. A cartilage sheet (Cart-S) prepared by high-density culture is laminated and reshaped to construct a cartilage tube as the main load-bearing structure in which the chondrocytes exhibit a stable phenotype and secreted considerable cartilage-specific matrix, presenting a native-like grid arrangement. To further build a tracheal epithelial barrier, a temperature-sensitive technique is used to construct the monolayer epithelium sheet (Epi-S), in which the airway epithelial cells present integrated tight junctions, good transepithelial electrical resistance, and favorable ciliary differentiation capability. Epi-S can be integrally transferred to inner wall of cartilage tube, forming a scaffold-free complex tracheal substitute (SC-trachea). Interestingly, when Epi-S is attached to the cartilage surface, epithelium-specific gene expression is significantly enhanced. SC-trachea establishes abundant blood supply via heterotopic vascularization and then is pedicle transplanted for tracheal reconstruction, achieving 83.3% survival outcomes in rabbit models. Notably, the scaffold-free engineered trachea simultaneously satisfies sufficient mechanical properties and barrier function due to its matrix-rich cartilage structure and well-differentiated ciliated epithelium, demonstrating great clinical potential for long-segmental tracheal reconstruction.
Collapse
Affiliation(s)
- Minglei Yang
- Department of Cardiothoracic Surgery, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315020, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Ziwen Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Feng Jin
- Shandong Province Chest Hospital, Shandong, 250011, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200092, China
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| |
Collapse
|
38
|
Gautam LK, Harriott NC, Caceres AM, Ryan AL. Basic Science Perspective on Engineering and Modeling the Large Airways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:73-106. [PMID: 37195527 DOI: 10.1007/978-3-031-26625-6_5] [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
The airway epithelium provides a physical and biochemical barrier playing a key role in protecting the lung from infiltration of pathogens and irritants and is, therefore, crucial in maintaining tissue homeostasis and regulating innate immunity. Due to continual inspiration and expiration of air during breathing, the epithelium is exposed to a plethora of environmental insults. When severe or persistent, these insults lead to inflammation and infection. The effectiveness of the epithelium as a barrier is reliant upon its capacity for mucociliary clearance, immune surveillance, and regeneration upon injury. These functions are accomplished by the cells that comprise the airway epithelium and the niche in which they reside. Engineering of new physiological and pathological models of the proximal airways requires the generation of complex structures comprising the surface airway epithelium, submucosal gland epithelium, extracellular matrix, and niche cells, including smooth muscle cells, fibroblasts, and immune cells. This chapter focuses on the structure-function relationships in the airways and the challenges of developing complex engineered models of the human airway.
Collapse
Affiliation(s)
- Lalit K Gautam
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Noa C Harriott
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Adrian M Caceres
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
39
|
Maughan EF, Hynds RE, Pennycuick A, Nigro E, Gowers KH, Denais C, Gómez-López S, Lazarus KA, Orr JC, Pearce DR, Clarke SE, Lee DDH, Woodall MN, Masonou T, Case KM, Teixeira VH, Hartley BE, Hewitt RJ, Al Yaghchi C, Sandhu GS, Birchall MA, O’Callaghan C, Smith CM, De Coppi P, Butler CR, Janes SM. Cell-intrinsic differences between human airway epithelial cells from children and adults. iScience 2022; 25:105409. [PMID: 36388965 PMCID: PMC9664344 DOI: 10.1016/j.isci.2022.105409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/30/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
The airway epithelium is a protective barrier that is maintained by the self-renewal and differentiation of basal stem cells. Increasing age is a principle risk factor for chronic lung diseases, but few studies have explored age-related molecular or functional changes in the airway epithelium. We retrieved epithelial biopsies from histologically normal tracheobronchial sites from pediatric and adult donors and compared their cellular composition and gene expression profile (in laser capture-microdissected whole epithelium, fluorescence-activated cell-sorted basal cells, and basal cells in cell culture). Histologically, pediatric and adult tracheobronchial epithelium was similar in composition. We observed age-associated changes in RNA sequencing studies, including higher interferon-associated gene expression in pediatric epithelium. In cell culture, pediatric cells had higher colony formation ability, sustained in vitro growth, and outcompeted adult cells in a direct competitive proliferation assay. Our results demonstrate cell-intrinsic differences between airway epithelial cells from children and adults in both homeostatic and proliferative states.
Collapse
Affiliation(s)
- Elizabeth F. Maughan
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Robert E. Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Adam Pennycuick
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Ersilia Nigro
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Kate H.C. Gowers
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Celine Denais
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Sandra Gómez-López
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Kyren A. Lazarus
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Jessica C. Orr
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - David R. Pearce
- University College London Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sarah E. Clarke
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | - Dani Do Hyang Lee
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Maximillian N.J. Woodall
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Tereza Masonou
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Katie-Marie Case
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Vitor H. Teixeira
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| | | | | | - Chadwan Al Yaghchi
- The National Centre for Airway Reconstruction, Department of Otolaryngology, Charing Cross Hospital, London W6 8RF, UK
| | - Gurpreet S. Sandhu
- The National Centre for Airway Reconstruction, Department of Otolaryngology, Charing Cross Hospital, London W6 8RF, UK
| | - Martin A. Birchall
- University College London Ear Institute, University College London, London WC1X 8EE, UK
| | - Christopher O’Callaghan
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Claire M. Smith
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1E 1EH, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, University College London Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Colin R. Butler
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
- Tracheal Service, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Sam M. Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E 6JF, UK
| |
Collapse
|
40
|
Abstract
Acute and chronic lung diseases are a leading cause of morbidity and mortality globally. Unfortunately, these diseases are increasing in frequency and we have limited treatment options for severe lung diseases. New therapies are needed that not only treat symptoms or slow disease progression, but also enable the regeneration of functional lung tissue. Both airways and alveoli contain populations of epithelial stem cells with the potential to self-renew and produce differentiated progeny. Understanding the mechanisms that determine the behaviour of these cells, and their interactions with their niches, will allow future generations of respiratory therapies that protect the lungs from disease onset, promote regeneration from endogenous stem cells or enable regeneration through the delivery of exogenous cells. This review summarises progress towards each of these goals, highlighting the challenges and opportunities of developing pro-regenerative (bio)pharmaceutical, gene and cell therapies for respiratory diseases.
Collapse
Affiliation(s)
- Robert E. Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, WC1N 1DZ, UK
- UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| |
Collapse
|
41
|
Lee DF, Thompson CL, Baynes RE, Enomoto H, Smith GW, Chambers MA. Development and evaluation of a bovine lung-on-chip (bLOC) to study bovine respiratory diseases. IN VITRO MODELS 2022; 1:333-346. [PMID: 36660607 PMCID: PMC9383688 DOI: 10.1007/s44164-022-00030-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/22/2023]
Abstract
Purpose Current air-liquid interface (ALI) models of bovine proximal airways have their limitations. They do not simulate blood flow necessary to mimic systemic drug administration, and repeated sampling requires multiple, independent cultures. A bovine lung-on-chip (bLOC) would overcome these limitations, providing a convenient and cost-effective model for pharmacokinetic or pathogenicity studies. Methods Bovine pulmonary arterial endothelial cells seeded into the endothelial channel of an Emulate Lung-Chip were interfaced with bovine bronchial epithelial cells in the epithelial channel. Cells were cultured at ALI for up to 21 days. Differentiation was assessed by mucin quantification, phase-contrast light microscopy and immunofluorescence of cell-specific markers in fixed cultures. Barrier integrity was determined by FITC-labelled dextran 3-5 kDa permeability. To evaluate the model, endothelial-epithelial transport of the antibiotic drug, danofloxacin, was followed using liquid chromatography-mass spectrometry, with the aim of replicating data previously determined in vivo. Results bLOC cultures secreted quantifiable mucins, whilst cilia formation was evident in the epithelial channel. Barrier integrity of the model was demonstrated by resistance to FITC-Dextran 3-5 kDa permeation. Bronchial epithelial and endothelial cell-specific markers were observed. Close to plasma, representative PK data for danofloxacin was observed in the endothelial channel; however, danofloxacin in the epithelial channel was mostly below the limit of quantification. Conclusion A co-culture model of the bovine proximal airway was successfully generated, with potential to replace in vivo experimentation. With further optimisation and characterisation, the bLOC may be suitable to perform drug pharmacokinetic studies for bovine respiratory disease (BRD), and other applications.
Collapse
Affiliation(s)
- Diane F. Lee
- School of Veterinary Medicine, University of Surrey, Guildford, UK
- Now at Sussex Drug Discovery Centre, University of Sussex, Falmer, UK
| | - Clare L. Thompson
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Ronald E. Baynes
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | - Hiroko Enomoto
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | | | - Mark A. Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, UK
| |
Collapse
|
42
|
Hu Z, Chen Y, Gao M, Chi X, He Y, Zhang C, Yang Y, Li Y, Lv Y, Huang Y, Deng X. Novel strategy for primary epithelial cell isolation: Combination of hyaluronidase and collagenase I. Cell Prolif 2022; 56:e13320. [PMID: 35920005 PMCID: PMC9816927 DOI: 10.1111/cpr.13320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Different strategies for epithelial cell isolation significantly affect the viability and physiological properties of primary cells. Trypsin digestion, a conventional method, causes collateral damage owing to its strong digestive potential. To better preserve the physiological properties of epithelial tissues, we aimed to develop a modified method (hyaluronidase and collagenase I combination) for primary cell isolation. METHOD We used conventional and modified methods to compare cell viability, morphology and stemness. Additionally, we investigated the passaging stability of epithelial cells and their capacity for organoid formation. Finally, we compared the two methods for isolating urothelial, oesophageal, lingual, and epidermal epithelial cells. RESULT Gingival epithelial cells obtained using the modified method had higher viability, better morphology and stronger stemness than those obtained using the conventional method. Additionally, primary cells obtained using the modified method were stably passaged. Regarding organoid culture, adopting the modified method led to a significant increase in the growth rate and expression of the stem cell markers cytokeratin (CK)-19 and Ki-67. Furthermore, the modified method outperformed the conventional method for isolating urothelial, epidermal, oesophageal and lingual epithelial cells. CONCLUSION We demonstrated that the combination of hyaluronidase and collagenase I outperformed trypsin in preserving the physiological properties of primary cells and organoid formation. The modified method could be broadly applied to isolate different types of epithelial cells and facilitate studies on organoids and tissue engineering.
Collapse
Affiliation(s)
- Zhewen Hu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Yiming Chen
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Min Gao
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Xiaopei Chi
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Ying He
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Chenguang Zhang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of StomatologySun Yat‐Sen UniversityGuangzhouPeople's Republic of China
| | - Yue Yang
- Department of Prosthodontics, The First Clinical DivisionPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Yuman Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Yan Lv
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of StomatologyCapital Medical UniversityBeijingPeople's Republic of China
| | - Ying Huang
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijingPeople's Republic of China
| |
Collapse
|
43
|
Gao E, Wang P, Chen F, Xu Y, Wang Q, Chen H, Jiang G, Zhou G, Li D, Liu Y, Duan L. Skin-derived epithelial lining facilitates orthotopic tracheal transplantation by protecting the tracheal cartilage and inhibiting granulation hyperplasia. BIOMATERIALS ADVANCES 2022; 139:213037. [PMID: 35882125 DOI: 10.1016/j.bioadv.2022.213037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/28/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
Long-segment tracheal defects caused by tumours, inflammation or trauma can cause serious damage to the quality of life of patients. Although many novel neotracheas have been constructed, the therapeutic effect of orthotopic transplantation was compromised mainly because of the lack of an epithelial lining in those neotracheas. In this study, we aimed to investigate the therapeutic function of skin-derived epithelial lining for orthotopic tracheal transplantation. Strips of auricular cartilage with fixed interval were interrupted sutured on a silicone tube to mimic the cartilage rings of the native trachea. Neotrachea in the with epithelium group retained the unilateral skin as the epithelial lining in the lumen, whereas the neotrachea in the without epithelium group consisted solely of cartilage strips. After revascularized in the sternohyoid muscle, 2-cm-long tracheal defects were made and were reconstructed using these neotracheas. Our results showed that the skin-derived epithelial lining simultaneously protected the engineered tracheal cartilage and inhibited granulation hyperplasia in the tracheal lumen; further, compared with the without epithelium group, the group with epithelium showed a marked improvement in the tracheal lumen patency and the survival rate of rabbits. Our study provides a critical cue for improvements in the repair of tracheal defects via skin-derived epithelial lining and may significantly advance the clinical translation of tissue-engineered trachea.
Collapse
Affiliation(s)
- Erji Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengli Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Feifan Chen
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Plastic Surgery, Weifang Medical College, Weifang, China
| | - Yong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyi Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Plastic Surgery, Weifang Medical College, Weifang, China
| | - Hong Chen
- Department of Hand Surgery, Ningbo Sixth Hospital, Ningbo, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Plastic Surgery, Weifang Medical College, Weifang, China.
| | - Dan Li
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Dermatology, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Liang Duan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
44
|
Lakhdar R, Mumby S, Abubakar-Waziri H, Porter A, Adcock IM, Chung KF. Lung toxicity of particulates and gaseous pollutants using ex-vivo airway epithelial cell culture systems. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 305:119323. [PMID: 35447256 DOI: 10.1016/j.envpol.2022.119323] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Air pollution consists of a multi-faceted mix of gases and ambient particulate matter (PM) with diverse organic and non-organic chemical components that contribute to increasing morbidity and mortality worldwide. In particular, epidemiological and clinical studies indicate that respiratory health is adversely affected by exposure to air pollution by both causing and worsening (exacerbating) diseases such as chronic obstructive pulmonary disease (COPD), asthma, interstitial pulmonary fibrosis and lung cancer. The molecular mechanisms of air pollution-induced pulmonary toxicity have been evaluated with regards to different types of PM of various sizes and concentrations with single and multiple exposures over different time periods. These data provide a plausible interrelationship between cellular toxicity and the activation of multiple biological processes including proinflammatory responses, oxidative stress, mitochondrial oxidative damage, autophagy, apoptosis, cell genotoxicity, cellular senescence and epithelial-mesenchymal transition. However, these molecular changes have been studied predominantly in cell lines rather than in primary bronchial or nasal cells from healthy subjects or those isolated from patients with airways disease. In addition, they have been conducted under different cell culture conditions and generally in submerged culture rather than the more relevant air-liquid interface culture and with a variety of air pollutant exposure protocols. Cell types may respond differentially to pollution delivered as an aerosol rather than being bathed in media containing agglomerations of particles. As a result, the actual pathophysiological pathways activated by different PMs in primary cells from the airways of healthy and asthmatic subjects remains unclear. This review summarises the literature on the different methodologies utilised in studying the impact of submicron-sized pollutants on cells derived from the respiratory tract with an emphasis on data obtained from primary human cell. We highlight the critical underlying molecular mechanisms that may be important in driving disease processes in response to air pollution in vivo.
Collapse
Affiliation(s)
- Ramzi Lakhdar
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Sharon Mumby
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Hisham Abubakar-Waziri
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Alexandra Porter
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Ian M Adcock
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| | - Kian Fan Chung
- National Heart and Lung Institute and *Department of Materials, Imperial College London, London, SW3 6LY, United Kingdom.
| |
Collapse
|
45
|
Chiu MC, Li C, Liu X, Yu Y, Huang J, Wan Z, Xiao D, Chu H, Cai JP, Zhou B, Sit KY, Au WK, Wong KKY, Li G, Chan JFW, To KKW, Chen Z, Jiang S, Clevers H, Yuen KY, Zhou J. A bipotential organoid model of respiratory epithelium recapitulates high infectivity of SARS-CoV-2 Omicron variant. Cell Discov 2022; 8:57. [PMID: 35710786 PMCID: PMC9203776 DOI: 10.1038/s41421-022-00422-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/15/2022] [Indexed: 12/31/2022] Open
Abstract
The airways and alveoli of the human respiratory tract are lined by two distinct types of epithelium, which are the primary targets of respiratory viruses. We previously established long-term expanding human lung epithelial organoids from lung tissues and developed a ‘proximal’ differentiation protocol to generate mucociliary airway organoids. However, a respiratory organoid system with bipotential of the airway and alveolar differentiation remains elusive. Here we defined a ‘distal’ differentiation approach to generate alveolar organoids from the same source for the derivation of airway organoids. The alveolar organoids consisting of type I and type II alveolar epithelial cells (AT1 and AT2, respectively) functionally simulate the alveolar epithelium. AT2 cells maintained in lung organoids serve as progenitor cells from which alveolar organoids derive. Moreover, alveolar organoids sustain a productive SARS-CoV-2 infection, albeit a lower replicative fitness was observed compared to that in airway organoids. We further optimized 2-dimensional (2D) airway organoids. Upon differentiation under a slightly acidic pH, the 2D airway organoids exhibit enhanced viral replication, representing an optimal in vitro correlate of respiratory epithelium for modeling the high infectivity of SARS-CoV-2. Notably, the higher infectivity and replicative fitness of the Omicron variant than an ancestral strain were accurately recapitulated in these optimized airway organoids. In conclusion, we have established a bipotential organoid culture system able to reproducibly expand the entire human respiratory epithelium in vitro for modeling respiratory diseases, including COVID-19.
Collapse
Affiliation(s)
- Man Chun Chiu
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Cun Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaojuan Liu
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yifei Yu
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jingjing Huang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhixin Wan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ding Xiao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hin Chu
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China
| | - Jian-Piao Cai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Biao Zhou
- AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ko-Yung Sit
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, and Queen Mary Hospital, Hong Kong, China
| | - Wing-Kuk Au
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, and Queen Mary Hospital, Hong Kong, China
| | - Kenneth Kak-Yuen Wong
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, and Queen Mary Hospital, Hong Kong, China
| | - Gang Li
- Department of Otolaryngology-Head and Neck Surgery, Precision Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guang dong, China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.,Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kelvin Kai-Wang To
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.,Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhiwei Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China.,AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Center (UMC) Utrecht, Utrecht, the Netherlands.
| | - Kwok Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China. .,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China. .,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China. .,Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jie Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China. .,State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China. .,Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong, China.
| |
Collapse
|
46
|
Tang XY, Wu S, Wang D, Chu C, Hong Y, Tao M, Hu H, Xu M, Guo X, Liu Y. Human organoids in basic research and clinical applications. Signal Transduct Target Ther 2022; 7:168. [PMID: 35610212 PMCID: PMC9127490 DOI: 10.1038/s41392-022-01024-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Organoids are three-dimensional (3D) miniature structures cultured in vitro produced from either human pluripotent stem cells (hPSCs) or adult stem cells (AdSCs) derived from healthy individuals or patients that recapitulate the cellular heterogeneity, structure, and functions of human organs. The advent of human 3D organoid systems is now possible to allow remarkably detailed observation of stem cell morphogens, maintenance and differentiation resemble primary tissues, enhancing the potential to study both human physiology and developmental stage. As they are similar to their original organs and carry human genetic information, organoids derived from patient hold great promise for biomedical research and preclinical drug testing and is currently used for personalized, regenerative medicine, gene repair and transplantation therapy. In recent decades, researchers have succeeded in generating various types of organoids mimicking in vivo organs. Herein, we provide an update on current in vitro differentiation technologies of brain, retinal, kidney, liver, lung, gastrointestinal, cardiac, vascularized and multi-lineage organoids, discuss the differences between PSC- and AdSC-derived organoids, summarize the potential applications of stem cell-derived organoids systems in the laboratory and clinic, and outline the current challenges for the application of organoids, which would deepen the understanding of mechanisms of human development and enhance further utility of organoids in basic research and clinical studies.
Collapse
Affiliation(s)
- Xiao-Yan Tang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Shanshan Wu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Da Wang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Chu Chu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Yuan Hong
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Mengdan Tao
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Hao Hu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Min Xu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China
| | - Xing Guo
- Department of Neurobiology, School of Basic Medical Sciences; Nanjing Medical University, Nanjing, China.
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy; State Key Laboratory of Reproductive Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine; Nanjing Medical University, Nanjing, China.
| |
Collapse
|
47
|
Wu M, Zhang X, Lin Y, Zeng Y. Roles of airway basal stem cells in lung homeostasis and regenerative medicine. Respir Res 2022; 23:122. [PMID: 35562719 PMCID: PMC9102684 DOI: 10.1186/s12931-022-02042-5] [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: 02/14/2022] [Accepted: 05/01/2022] [Indexed: 11/10/2022] Open
Abstract
Airway basal stem cells (BSCs) in the proximal airways are recognized as resident stem cells capable of self-renewing and differentiating to virtually every pseudostratified epithelium cell type under steady-state and after acute injury. In homeostasis, BSCs typically maintain a quiescent state. However, when exposed to acute injuries by either physical insults, chemical damage, or pathogen infection, the remaining BSCs increase their proliferation rate apace within the first 24 h and differentiate to restore lung homeostasis. Given the progenitor property of airway BSCs, it is attractive to research their biological characteristics and how they maintain homeostatic airway structure and respond to injury. In this review, we focus on the roles of BSCs in lung homeostasis and regeneration, detail the research progress in the characteristics of airway BSCs, the cellular and molecular signaling communications involved in BSCs-related airway repair and regeneration, and further discuss the in vitro models for airway BSC propagation and their applications in lung regenerative medicine therapy.
Collapse
Affiliation(s)
- Meirong Wu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian Province, People's Republic of China
| | - Xiaojing Zhang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian Province, People's Republic of China
| | - Yijian Lin
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian Province, People's Republic of China
| | - Yiming Zeng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China. .,Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China. .,Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian Province, People's Republic of China.
| |
Collapse
|
48
|
Xia S, Wu M, Zhou X, Zhang X, Ye L, Zhang K, Kang Y, Liu J, Zhang Y, Wu W, Dong D, Chen H, Li H. Treating intrauterine adhesion using conditionally reprogrammed physiological endometrial epithelial cells. Stem Cell Res Ther 2022; 13:178. [PMID: 35505443 PMCID: PMC9066886 DOI: 10.1186/s13287-022-02860-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/21/2022] [Indexed: 01/18/2023] Open
Abstract
Background There is unmet need for effective therapies of intrauterine adhesions (IUAs) that are common cause of menstrual disturbance and infertility, since current clinical procedures do not improve prognosis for patients with moderate to severe IUA, with a recurrence rate of 23–50%. Stem cell-based therapy has emerged as a therapeutic option with unsolved issues for IUA patients in the past few years. Primary endometrial epithelial cells for cell therapy are largely hampered with the extremely limited proliferation capacity of uterine epithelial cells. This study was to evaluate whether IUA is curable with conditionally reprogrammed (CR) endometrial epithelial cells. Methods Mouse endometrial epithelial cells (MEECs) were isolated from C57BL female mice, and long-term cultures of MEECs were established and maintained with conditional reprogramming (CR) method. DNA damage response analysis, soft agar assay, and matrigel 3D culture were carried out to determine the normal biological characteristics of CR-MEECs. The tissue-specific differentiation potential of MEECs was analyzed with air–liquid interface (ALI) 3D culture, hematoxylin and eosin (H&E) staining, Masson’s trichrome and DAB staining, immunofluorescence assay. IUA mice were constructed and transplanted with CR-MEECs. Repair and mechanisms of MEECs transplantation in IUA mice were measured with qRT-PCR, Masson’s trichrome, and DAB staining. Results We first successfully established long-term cultures of MEECs using CR approach. CR-MEECs maintained a rapid and stable proliferation in this co-culture system. Our data confirmed that CR-MEECs retained normal biological characteristics and endometrium tissue-specific differentiation potential. CR-MEECs also expressed estrogen and progesterone receptors and maintained the exquisite sensitivity to sex hormones in vitro. Most importantly, allogeneic transplantation of CR-MEECs successfully repaired the injured endometrium and significantly improved the pregnancy rate of IUA mice. Conclusions Conditionally reprogrammed physiological endometrial epithelial cells provide a novel strategy in IUA clinics in a personalized or generalized manner and also serve as a physiological model to explore biology of endometrial epithelial cells and mechanisms of IUA.
Collapse
Affiliation(s)
- Siyu Xia
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Ming Wu
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinhao Zhou
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiu Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Lina Ye
- Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China
| | - Kang Zhang
- Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China
| | - Yiyi Kang
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jun Liu
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Yunci Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Wang Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Dirong Dong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Hong Chen
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Hui Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China. .,Wuhan University Shenzhen Institute, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
49
|
Lin L, Wang X, Niu M, Wu Q, Wang H, Zu Y, Wang W. Biomimetic epithelium/endothelium on chips. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
50
|
Tsao CK, Liao KH, Hsiao HY, Liu YH, Wu CT, Cheng MH, Zhong WB. Tracheal reconstruction with pedicled tandem grafts engineered by a radial stretch bioreactor. J Biomater Appl 2022; 37:118-131. [PMID: 35412872 DOI: 10.1177/08853282221082357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The engineering of tracheal substitutes is pivotal in improving tracheal reconstruction. In this study, we aimed to investigate the effects of biomechanical stimulation on tissue engineering tracheal cartilage by mimicking the trachea motion through a novel radial stretching bioreactor, which enables to dynamically change the diameter of the hollow cylindrical implants. Applying our bioreactor, we demonstrated that chondrocytes seeded on the surface of Poly (ε-caprolactone) scaffold respond to mechanical stimulation by improvement of infiltration into implants and upregulation of cartilage-specific genes. Further, the mechanical stimulation enhanced the accumulation of cartilage neo-tissues and cartilage-specific extracellular macromolecules in the muscle flap-remodeled implants and reconstructed trachea. Nevertheless, the invasion of fibrous tissues in the reconstructed trachea was suppressed upon mechanical loading.
Collapse
Affiliation(s)
- Chung-Kan Tsao
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Kuan-Hao Liao
- Division of Reconstructive Microsurgery, Department of Plastic and Reconstructive Surgery, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Hui-Yi Hsiao
- Center for Tissue Engineering, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Yun-Hen Liu
- Division of Thoracic Surgery, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Chieh-Tsai Wu
- Division of Pediatric Neurosurgery, Chang Gung Children's Hospital, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Ming-Huei Cheng
- Center of Lymphedema Microsurgery, Department of Plastic and Reconstructive Surgery, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Wen-Bin Zhong
- Center for Tissue Engineering, 38014Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan.,Center for Biomedical Engineering, College of Engineering, 38014Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|