1
|
Le NT, Dunleavy MW, Kumar RD, Zhou W, Bhatia SS, El-Hashash AH. Cellular therapies for idiopathic pulmonary fibrosis: current progress and future prospects. AMERICAN JOURNAL OF STEM CELLS 2024; 13:191-211. [PMID: 39308764 PMCID: PMC11411253 DOI: 10.62347/daks5508] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/17/2024] [Indexed: 09/25/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial, fibrotic lung disease characterized by progressive damage. Lung tissues with IPF are replaced by fibrotic tissues with increased collagen deposition, modified extracellular matrix, all which overall damages the alveoli. These changes eventually impede the gas exchange function of the alveoli, and eventually leads to fatal respiratory failure of the lung. Investigations have been conducted to further understand IPF's pathogenesis, and significant progress in understanding its development has been made. Additionally, two therapeutic treatments, Nintedanib and Pirfenidone, have been approved and are currently used in medical applications. Moreover, cell-based treatments have recently come to the forefront of developing disease therapeutics and are the focus of many current studies. Furthermore, a sizable body of research encompassing basic, pre-clinical, and even clinical trials have all been amassed in recent years and hold a great potential for more widespread applications in patient care. Herein, this article reviews the progress in understanding the pathogenesis and pathophysiology of IPF. Additionally, different cell types used in IPF therapy were reviewed, including alveolar epithelial cells (AECs), circulating endothelial progenitors (EPCs), mixed lung epithelial cells, different types of stem cells, and endogenous lung tissue-specific stem cells. Finally, we discussed the contemporary trials that employ or explore cell-based therapy for IPF.
Collapse
Affiliation(s)
- Nicholas T Le
- Biology Department, Texas A&M University College Station, TX, USA
| | | | - Rebecca D Kumar
- Biology Department, Texas A&M University College Station, TX, USA
| | - William Zhou
- The University of Texas at Austin Austin, TX, USA
| | | | | |
Collapse
|
2
|
Goecke T, Ius F, Ruhparwar A, Martin U. Unlocking the Future: Pluripotent Stem Cell-Based Lung Repair. Cells 2024; 13:635. [PMID: 38607074 PMCID: PMC11012168 DOI: 10.3390/cells13070635] [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: 01/05/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
The human respiratory system is susceptible to a variety of diseases, ranging from chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis to acute respiratory distress syndrome (ARDS). Today, lung diseases represent one of the major challenges to the health care sector and represent one of the leading causes of death worldwide. Current treatment options often focus on managing symptoms rather than addressing the underlying cause of the disease. The limitations of conventional therapies highlight the urgent clinical need for innovative solutions capable of repairing damaged lung tissue at a fundamental level. Pluripotent stem cell technologies have now reached clinical maturity and hold immense potential to revolutionize the landscape of lung repair and regenerative medicine. Meanwhile, human embryonic (HESCs) and human-induced pluripotent stem cells (hiPSCs) can be coaxed to differentiate into lung-specific cell types such as bronchial and alveolar epithelial cells, or pulmonary endothelial cells. This holds the promise of regenerating damaged lung tissue and restoring normal respiratory function. While methods for targeted genetic engineering of hPSCs and lung cell differentiation have substantially advanced, the required GMP-grade clinical-scale production technologies as well as the development of suitable preclinical animal models and cell application strategies are less advanced. This review provides an overview of current perspectives on PSC-based therapies for lung repair, explores key advances, and envisions future directions in this dynamic field.
Collapse
Affiliation(s)
- Tobias Goecke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Fabio Ius
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Arjang Ruhparwar
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Lower Saxony Center for Biomedical Engineering, Implant Research and Development /Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (F.I.); (A.R.)
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
3
|
Vats A, Chaturvedi P. The Regenerative Power of Stem Cells: Treating Bleomycin-Induced Lung Fibrosis. Stem Cells Cloning 2023; 16:43-59. [PMID: 37719787 PMCID: PMC10505024 DOI: 10.2147/sccaa.s419474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease with no known cure, characterized by the formation of scar tissue in the lungs, leading to respiratory failure. Although the exact cause of IPF remains unclear, the condition is thought to result from a combination of genetic and environmental factors. One of the most widely used animal models to study IPF is the bleomycin-induced lung injury model in mice. In this model, the administration of the chemotherapeutic agent bleomycin causes pulmonary inflammation and fibrosis, which closely mimics the pathological features of human IPF. Numerous recent investigations have explored the functions of various categories of stem cells in the healing process of lung injury induced by bleomycin in mice, documenting the beneficial effects and challenges of this approach. Differentiation of stem cells into various cell types and their ability to modulate tissue microenvironment is an emerging aspect of the regenerative therapies. This review article aims to provide a comprehensive overview of the role of stem cells in repairing bleomycin-induced lung injury. It delves into the mechanisms through which various types of stem cells, including mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, and lung resident stem cells, exert their therapeutic effects in this specific model. We have also discussed the unique set of intermediate markers and signaling factors that can influence the proliferation and differentiation of alveolar epithelial cells both during lung repair and homeostasis. Finally, we highlight the challenges and opportunities associated with translating stem cell therapy to the clinic for IPF patients. The novelty and implications of this review extend beyond the understanding of the potential of stem cells in treating IPF to the broader field of regenerative medicine. We believe that the review paves the way for further advancements in stem cell therapies, offering hope for patients suffering from this debilitating and currently incurable disease.
Collapse
Affiliation(s)
- Amrita Vats
- Department of Pharmacology and Regenerative Medicine, University of Illinois, Chicago, IL, 60612, USA
| | - Pankaj Chaturvedi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
4
|
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
|
5
|
Abstract
Pulmonary fibrosis (PF) is a chronic and relentlessly progressive interstitial lung disease in which the accumulation of fibroblasts and extracellular matrix (ECM) induces the destruction of normal alveolar structures, ultimately leading to respiratory failure. Patients with advanced PF are unable to perform physical labor and often have concomitant cough and dyspnea, which markedly impair their quality of life. However, there is a paucity of available pharmacological therapies, and to date, lung transplantation remains the only possible treatment for patients suffering from end-stage PF; moreover, the complexity of transplantation surgery and the paucity of donors greatly restrict the application of this treatment. Therefore, there is a pressing need for alternative therapeutic strategies for this complex disease. Due to their capacity for pluripotency and paracrine actions, stem cells are promising therapeutic agents for the treatment of interstitial lung disease, and an extensive body of literature supports the therapeutic efficacy of stem cells in lung fibrosis. Although stem cell transplantation may play an important role in the treatment of PF, some key issues, such as safety and therapeutic efficacy, remain to be resolved. In this review, we summarize recent preclinical and clinical studies on the stem cell-mediated regeneration of fibrotic lungs and present an analysis of concerning issues related to stem cell therapy to guide therapeutic development for this complex disease.
Collapse
|
6
|
Abstract
The lung is the primary site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced immunopathology whereby the virus enters the host cells by binding to angiotensin-converting enzyme 2 (ACE2). Sophisticated regeneration and repair programs exist in the lungs to replenish injured cell populations. However, known resident stem/progenitor cells have been demonstrated to express ACE2, raising a substantial concern regarding the long-term consequences of impaired lung regeneration after SARS-CoV-2 infection. Moreover, clinical treatments may also affect lung repair from antiviral drug candidates to mechanical ventilation. In this review, we highlight how SARS-CoV-2 disrupts a program that governs lung homeostasis. We also summarize the current efforts of targeted therapy and supportive treatments for COVID-19 patients. In addition, we discuss the pros and cons of cell therapy with mesenchymal stem cells or resident lung epithelial stem/progenitor cells in preventing post-acute sequelae of COVID-19. We propose that, in addition to symptomatic treatments being developed and applied in the clinic, targeting lung regeneration is also essential to restore lung homeostasis in COVID-19 patients.
Collapse
Affiliation(s)
- Fuxiaonan Zhao
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - Qingwen Ma
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - Qing Yue
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical School, Tianjin Medical University, Tianjin, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin Haihe Hospital, Tianjin, China
- Department of Basic Medicine, Haihe Hospital, Tianjin University, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| |
Collapse
|
7
|
Basil MC, Katzen J, Engler AE, Guo M, Herriges MJ, Kathiriya JJ, Windmueller R, Ysasi AB, Zacharias WJ, Chapman HA, Kotton DN, Rock JR, Snoeck HW, Vunjak-Novakovic G, Whitsett JA, Morrisey EE. The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future. Cell Stem Cell 2021; 26:482-502. [PMID: 32243808 PMCID: PMC7128675 DOI: 10.1016/j.stem.2020.03.009] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The respiratory system, which includes the trachea, airways, and distal alveoli, is a complex multi-cellular organ that intimately links with the cardiovascular system to accomplish gas exchange. In this review and as members of the NIH/NHLBI-supported Progenitor Cell Translational Consortium, we discuss key aspects of lung repair and regeneration. We focus on the cellular compositions within functional niches, cell-cell signaling in homeostatic health, the responses to injury, and new methods to study lung repair and regeneration. We also provide future directions for an improved understanding of the cell biology of the respiratory system, as well as new therapeutic avenues.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeremy Katzen
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anna E Engler
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Minzhe Guo
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael J Herriges
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jaymin J Kathiriya
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Rebecca Windmueller
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra B Ysasi
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - William J Zacharias
- Division of Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Hal A Chapman
- Division of Pulmonary Medicine, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Jason R Rock
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Hans-Willem Snoeck
- Center for Human Development, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Gordana Vunjak-Novakovic
- Departments of Biomedical Engineering and Medicine, Columbia University, New York, NY 10027, USA
| | - Jeffrey A Whitsett
- Center for Regenerative Medicine of Boston University and Boston Medical Center, The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02215, USA
| | - Edward E Morrisey
- Department of Medicine, Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
8
|
Rothbauer M, Bachmann BE, Eilenberger C, Kratz SR, Spitz S, Höll G, Ertl P. A Decade of Organs-on-a-Chip Emulating Human Physiology at the Microscale: A Critical Status Report on Progress in Toxicology and Pharmacology. MICROMACHINES 2021; 12:470. [PMID: 33919242 PMCID: PMC8143089 DOI: 10.3390/mi12050470] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022]
Abstract
Organ-on-a-chip technology has the potential to accelerate pharmaceutical drug development, improve the clinical translation of basic research, and provide personalized intervention strategies. In the last decade, big pharma has engaged in many academic research cooperations to develop organ-on-a-chip systems for future drug discoveries. Although most organ-on-a-chip systems present proof-of-concept studies, miniaturized organ systems still need to demonstrate translational relevance and predictive power in clinical and pharmaceutical settings. This review explores whether microfluidic technology succeeded in paving the way for developing physiologically relevant human in vitro models for pharmacology and toxicology in biomedical research within the last decade. Individual organ-on-a-chip systems are discussed, focusing on relevant applications and highlighting their ability to tackle current challenges in pharmacological research.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18-22, 1090 Vienna, Austria
| | - Barbara E.M. Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Allgemeine Unfallversicherungsanstalt (AUVA) Research Centre, Donaueschingenstraße 13, 1200 Vienna, Austria
| | - Christoph Eilenberger
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Sebastian R.A. Kratz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Drug Delivery and 3R-Models Group, Buchmann Institute for Molecular Life Sciences & Institute for Pharmaceutical Technology, Goethe University Frankfurt Am Main, 60438 Frankfurt, Germany
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gregor Höll
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
9
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
10
|
Mahfouzi SH, Amoabediny G, Safiabadi Tali SH. Advances in bioreactors for lung bioengineering: From scalable cell culture to tissue growth monitoring. Biotechnol Bioeng 2021; 118:2142-2167. [PMID: 33629350 DOI: 10.1002/bit.27728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Lung bioengineering has emerged to resolve the current lung transplantation limitations and risks, including the shortage of donor organs and the high rejection rate of transplanted lungs. One of the most critical elements of lung bioengineering is bioreactors. Bioreactors with different applications have been developed in the last decade for lung bioengineering approaches, aiming to produce functional reproducible tissue constructs. Here, the current status and advances made in the development and application of bioreactors for bioengineering lungs are comprehensively reviewed. First, bioreactor design criteria are explained, followed by a discussion on using bioreactors as a culture system for scalable expansion and proliferation of lung cells, such as producing epithelial cells from induced pluripotent stem cells (iPSCs). Next, bioreactor systems facilitating and improving decellularization and recellularization of lung tissues are discussed, highlighting the studies that developed bioreactors for producing engineered human-sized lungs. Then, monitoring bioreactors are reviewed, showing their ability to evaluate and optimize the culture conditions for maturing engineered lung tissues, followed by an explanation on the ability of ex vivo lung perfusion systems for reconditioning the lungs before transplantation. After that, lung cancer studies simplified by bioreactors are discussed, showing the potentials of bioreactors in lung disease modeling. Finally, other platforms with the potential of facilitating lung bioengineering are described, including the in vivo bioreactors and lung-on-a-chip models. In the end, concluding remarks and future directions are put forward to accelerate lung bioengineering using bioreactors.
Collapse
Affiliation(s)
- Seyed Hossein Mahfouzi
- Department of Biomedical Engineering, The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| | - Ghassem Amoabediny
- Department of Biomedical Engineering, The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran.,Department of Biotechnology and Pharmaceutical Engineering, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed Hamid Safiabadi Tali
- Department of Biomedical Engineering, The Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
11
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
12
|
Varma R, Soleas JP, Waddell TK, Karoubi G, McGuigan AP. Current strategies and opportunities to manufacture cells for modeling human lungs. Adv Drug Deliv Rev 2020; 161-162:90-109. [PMID: 32835746 PMCID: PMC7442933 DOI: 10.1016/j.addr.2020.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/17/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Chronic lung diseases remain major healthcare burdens, for which the only curative treatment is lung transplantation. In vitro human models are promising platforms for identifying and testing novel compounds to potentially decrease this burden. Directed differentiation of pluripotent stem cells is an important strategy to generate lung cells to create such models. Current lung directed differentiation protocols are limited as they do not 1) recapitulate the diversity of respiratory epithelium, 2) generate consistent or sufficient cell numbers for drug discovery platforms, and 3) establish the histologic tissue-level organization critical for modeling lung function. In this review, we describe how lung development has formed the basis for directed differentiation protocols, and discuss the utility of available protocols for lung epithelial cell generation and drug development. We further highlight tissue engineering strategies for manipulating biophysical signals during directed differentiation such that future protocols can recapitulate both chemical and physical cues present during lung development.
Collapse
Affiliation(s)
- Ratna Varma
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Latner Thoracic Surgery Research Laboratories, Toronto General Hospital, 101 College St., Toronto, ON M5G 1L7, Canada
| | - John P Soleas
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Latner Thoracic Surgery Research Laboratories, Toronto General Hospital, 101 College St., Toronto, ON M5G 1L7, Canada
| | - Thomas K Waddell
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Latner Thoracic Surgery Research Laboratories, Toronto General Hospital, 101 College St., Toronto, ON M5G 1L7, Canada; Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Golnaz Karoubi
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital, 101 College St., Toronto, ON M5G 1L7, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON M5S 3G8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| | - Alison P McGuigan
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON M5S 3E5, Canada.
| |
Collapse
|
13
|
Kızılkurtlu AA, Polat T, Aydın GB, Akpek A. Lung on a Chip for Drug Screening and Design. Curr Pharm Des 2019; 24:5386-5396. [PMID: 30734673 DOI: 10.2174/1381612825666190208122204] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 12/23/2022]
Abstract
Lung-on-a-chip is a micro device that combines the techniques of bioengineering, microbiology, polymer science and microfluidics disciplines in order to mimic physicochemical features and microenvironments, multicellular constructions, cell-cell interfaces of a human lung. Specifically, most novel lung on a chip designs consist of two micro-channeled outer parts, flexible and porous Polydimethylsiloxane (PDMS) membrane to create separation of air-blood chamber and subsidiary vacuum channels which enable stretching of the PDMS membrane to mimic movement mechanisms of the lung. Therefore, studies aim to emulate both tissue and organ functionality since it shall be creating great potential for advancing the studies about drug discovery, disease etiology and organ physiology compared with 2D (two dimensional) and 3D (three dimensional) cell culture models and current organoids. In this study, history of researches on lung anatomy and physiology, techniques of recreating lung functionality such as cell cultures in 2D and 3D models, organoids were covered and finally most advanced and recent state of the art technology product lung-on-a-chips' construction steps, advantages compared with other techniques, usage in lung modeling and diseases, present and future offers were analyzed in detail.
Collapse
Affiliation(s)
| | - Tuğçe Polat
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey
| | - Gül Banu Aydın
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400, Kocaeli, Turkey.,Sabanci University Nanotechnology Research and Application Center, Sabancı University, 34956 Tuzla Istanbul, Turkey
| |
Collapse
|
14
|
Lu Q, El-Hashash AHK. Cell-based therapy for idiopathic pulmonary fibrosis. Stem Cell Investig 2019; 6:22. [PMID: 31559309 PMCID: PMC6737434 DOI: 10.21037/sci.2019.06.09] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an example of interstitial lung diseases that is characterized by chronic, progressive, and fibrotic lung injuries. During lung fibrosis, normal healthy lung tissues are replaced by remarkably destroyed alveolar architecture and altered extracellular cell matrix. These changes eventually cause severe disruption of the tightly-controlled gas exchange process and reduction of lung compliance that ultimately lead to both respiratory failure and death. In the last decade, progress has been made toward understanding the pathogenesis of pulmonary fibrosis, and two novel disease-modifying therapies were approved. However, finding more effective treatments for pulmonary fibrosis is still a challenge, with its incidence continues to increase globally, which is associated with significantly high mortality, morbidity and economical healthcare burden. Different stem cell types have recently emerged as a promising therapy for human diseases, including lung fibrosis, with numerous studies on the identification, characterization, proliferation and differentiation of stem cells. A large body of both basic and pre-clinical research on stem cells has been recently translated to patient care worldwide. Herein, we review recent advances in our understanding of the pathophysiology of IPF, and types of cells used in IPF cell-based therapies, including alveolar and mixed lung epithelial cells, different stem cell types (MSCs, ADSCs, IPSCs…etc.), endogenous lung tissue-specific stem cells, and circulating endothelial progenitors (EPCs). We also discuss recent studies on the applications of these cells in IPF therapy and their delivery routes, effective doses for cell therapy, and timing of delivery. Finally, we discuss attractive recent and current clinical trials conducted on cell-based therapy for IPF.
Collapse
Affiliation(s)
- Qi Lu
- The University of Edinburgh-Zhejiang International campus (UoE-ZJU Institute), Haining, China
- Centre of Stem Cell and Regenerative Medicine Schools of Medicine & Basic Medicine, Hangzhou, China
| | - Ahmed H. K. El-Hashash
- The University of Edinburgh-Zhejiang International campus (UoE-ZJU Institute), Haining, China
- Centre of Stem Cell and Regenerative Medicine Schools of Medicine & Basic Medicine, Hangzhou, China
| |
Collapse
|
15
|
Wismayer K, Mehrban N, Bowen J, Birchall M. Improving cellular migration in tissue-engineered laryngeal scaffolds. J Laryngol Otol 2019; 133:135-148. [PMID: 30898188 DOI: 10.1017/s0022215119000082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To modify the non-porous surface membrane of a tissue-engineered laryngeal scaffold to allow effective cell entry. METHODS The mechanical properties, surface topography and chemistry of polyhedral oligomeric silsesquioxane poly(carbonate-urea) urethane were characterised. A laser technique introduced surface perforations. Micro computed tomography generated porosity data. Scaffolds were seeded with cells, investigated histologically and proliferation studied. Incubation and time effects were assessed. RESULTS Laser cutting perforated the polymer, connecting the substructure with the ex-scaffold environment and increasing porosity (porous, non-perforated = 87.9 per cent; porous, laser-perforated at intensities 3 = 96.4 per cent and 6 = 89.5 per cent). Cellular studies confirmed improved cell viability. Histology showed cells adherent to the scaffold surface and cells within perforations, and indicated that cells migrated into the scaffolds. After 15 days of incubation, scanning electron microscopy revealed an 11 per cent reduction in pore diameter, correlating with a decrease in Young's modulus. CONCLUSION Introducing surface perforations presents a viable method of improving polyhedral oligomeric silsesquioxane poly(carbonate-urea) urethane as a tissue-engineered scaffold.
Collapse
Affiliation(s)
- K Wismayer
- Division of Surgery,Ear Institute,University College London,UK
| | - N Mehrban
- Division of Surgery,Ear Institute,University College London,UK
| | - J Bowen
- School of Engineering and Innovation,Open University,Milton Keynes,UK
| | - M Birchall
- Ear Institute,University College London,UK
| |
Collapse
|
16
|
Nejad-Moghaddam A, Tahmasbpour E, Sohrabiyan M, Jafari H, Ghanei M. Stem cells therapy: a review on approaches that can be used for treatment of respiratory failures in sulfur mustard-injured patients. Immunopharmacol Immunotoxicol 2018; 40:359-367. [PMID: 30488735 DOI: 10.1080/08923973.2018.1510961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/06/2018] [Indexed: 12/28/2022]
Abstract
Sulfur mustard (SM) is a toxic agent which causes severe abnormalities in an airway system such as necrosis and inflammation, oxidative stress, chronic bronchitis, shortness of breath, and chronic obstructive pulmonary disease. Although possible mechanisms of SM toxicity have been extensively considered, there is still need to find an appropriate clinical treatment to decrease chronic lung injuries caused by SM. Due to extensive progresses and achievement in tissue repairing through stem cells therapy, the importance of cell therapy for the treatment of lung injuries has been increased. However, several factors such as types of stem cells, necessary conditions for growth and proliferation of stem cells, and their homing into the target tissues are considered as the most important problems in this issue. Mesenchymal stem cells (MSCs) are a class of multipotent stem cells with proliferative and self-renewal capacity which are able to differentiate into different cell lines such as lung epithelial cells. They have a potential repairing and immune modulatory properties which make them as a good candidate for the regeneration of bronchioles tract in SM-exposed patients. Unlike chemical drugs, the differentiation and high-level safety properties of MSCs can be considered as a new strategy for the treatment of SM-injured patients with pulmonary complications. This review aims to consider the therapeutic effects of MSCs in the treatment of SM-induced pulmonary injuries in both animals and humans.
Collapse
Affiliation(s)
- Amir Nejad-Moghaddam
- a Marine Medicine Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Eisa Tahmasbpour
- b Laboratory of Regenerative Medicine & Biomedical Innovations , Pasteur Institute of Iran , Tehran , Iran
| | - Milad Sohrabiyan
- c Chemical Injuries Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Hosein Jafari
- a Marine Medicine Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Mostafa Ghanei
- c Chemical Injuries Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
17
|
Serrano-Mollar A. Cell Therapy in Idiopathic Pulmonary Fibrosis †. Med Sci (Basel) 2018; 6:medsci6030064. [PMID: 30104544 PMCID: PMC6164035 DOI: 10.3390/medsci6030064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal disease with no effective or curative treatment options. In recent decades, cell-based therapies using stem cells or lung progenitor cells to regenerate lung tissue have experienced rapid growth in both preclinical animal models and translational clinical studies. In this review, the current knowledge of these cell therapies is summarized. Although further investigations are required, these studies indicate that cell therapies are a promising therapeutic approach for the treatment of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Anna Serrano-Mollar
- Departamento de Patología Experimental, Instituto de Investigaciones Biomédicas de Barcelona IIBB-CSIC-IDIBAPS, Rosselló, 161, 08036 Barcelona, Spain.
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
18
|
Conese M, Beccia E, Castellani S, Di Gioia S, Colombo C, Angiolillo A, Carbone A. The long and winding road: stem cells for cystic fibrosis. Expert Opin Biol Ther 2017; 18:281-292. [PMID: 29216777 DOI: 10.1080/14712598.2018.1413087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic syndrome with a high mortality rate due to severe lung disease. Despite having several drugs targeting specific mutated CFTR proteins already in clinical trials, new therapies, based on stem cells, are also emerging to treat those patients. AREAS COVERED The authors review the main sources of stem cells, including embryonic stem cells (ESCs), induced-pluripotent stem cells (iPSCs), gestational stem cells, and adult stem cells, such as mesenchymal stem cells (MSCs) in the context of CF. Furthermore, they describe the main animal and human models of lung physiology and pathology, involved in the optimization of these stem cell-applied therapies in CF. EXPERT OPINION ESCs and iPSCs are emerging sources for disease modeling and drug discovery purposes. The allogeneic transplant of healthy MSCs, that acts independently to specific mutations, is under intense scrutiny due to their secretory, immunomodulatory, anti-inflammatory and anti-bacterial properties. The main challenge for future developments will be to get exogenous stem cells into the appropriate lung location, where they can regenerate endogenous stem cells and act as inflammatory modulators. The clinical application of stem cells for the treatment of CF certainly warrants further insight into pre-clinical models, including large animals, organoids, decellularized organs and lung bioengineering.
Collapse
Affiliation(s)
- Massimo Conese
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Elisa Beccia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy.,b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Stefano Castellani
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Sante Di Gioia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Carla Colombo
- c Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation , University of Milan , Milan , Italy
| | - Antonella Angiolillo
- b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Annalucia Carbone
- d Division of Internal Medicine and Chronobiology Unit , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo (FG) , Italy
| |
Collapse
|
19
|
Hawkins F, Kramer P, Jacob A, Driver I, Thomas DC, McCauley KB, Skvir N, Crane AM, Kurmann AA, Hollenberg AN, Nguyen S, Wong BG, Khalil AS, Huang SX, Guttentag S, Rock JR, Shannon JM, Davis BR, Kotton DN. Prospective isolation of NKX2-1-expressing human lung progenitors derived from pluripotent stem cells. J Clin Invest 2017; 127:2277-2294. [PMID: 28463226 DOI: 10.1172/jci89950] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1-expressing (NKX2-1+) precursor cells. However, this hypothesis has not been formally tested owing to an inability to purify or track these progenitors for detailed characterization. Here we have engineered and developmentally differentiated NKX2-1GFP reporter pluripotent stem cells (PSCs) in vitro to generate and isolate human primordial lung progenitors that express NKX2-1 but are initially devoid of differentiated lung lineage markers. After sorting to purity, these primordial lung progenitors exhibited lung epithelial maturation. In the absence of mesenchymal coculture support, this NKX2-1+ population was able to generate epithelial-only spheroids in defined 3D cultures. Alternatively, when recombined with fetal mouse lung mesenchyme, the cells recapitulated epithelial-mesenchymal developing lung interactions. We imaged these progenitors in real time and performed time-series global transcriptomic profiling and single-cell RNA sequencing as they moved through the earliest moments of lung lineage specification. The profiles indicated that evolutionarily conserved, stage-dependent gene signatures of early lung development are expressed in primordial human lung progenitors and revealed a CD47hiCD26lo cell surface phenotype that allows their prospective isolation from untargeted, patient-specific PSCs for further in vitro differentiation and future applications in regenerative medicine.
Collapse
Affiliation(s)
- Finn Hawkins
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Philipp Kramer
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Anjali Jacob
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ian Driver
- Department of Anatomy, UCSF, San Francisco, California, USA
| | | | - Katherine B McCauley
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Ana M Crane
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Anita A Kurmann
- Center for Regenerative Medicine, and.,Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Brandon G Wong
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA
| | - Ahmad S Khalil
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Sarah Xl Huang
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA.,Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA
| | - Susan Guttentag
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital, Vanderbilt University, Nashville, Tennessee, USA
| | - Jason R Rock
- Department of Anatomy, UCSF, San Francisco, California, USA
| | - John M Shannon
- Division of Pulmonary Biology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Brian R Davis
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, and.,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Camarero-Espinosa S, Cooper-White J. Tailoring biomaterial scaffolds for osteochondral repair. Int J Pharm 2017; 523:476-489. [DOI: 10.1016/j.ijpharm.2016.10.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022]
|
21
|
|
22
|
Law JX, Liau LL, Aminuddin BS, Ruszymah BHI. Tissue-engineered trachea: A review. Int J Pediatr Otorhinolaryngol 2016; 91:55-63. [PMID: 27863642 DOI: 10.1016/j.ijporl.2016.10.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/09/2016] [Accepted: 10/12/2016] [Indexed: 01/23/2023]
Abstract
Tracheal replacement is performed after resection of a portion of the trachea that was impossible to reconnect via direct anastomosis. A tissue-engineered trachea is one of the available options that offer many advantages compared to other types of graft. Fabrication of a functional tissue-engineered trachea for grafting is very challenging, as it is a complex organ with important components, including cartilage, epithelium and vasculature. A number of studies have been reported on the preparation of a graftable trachea. A laterally rigid but longitudinally flexible hollow cylindrical scaffold which supports cartilage and epithelial tissue formation is the key element. The scaffold can be prepared via decellularization of an allograft or fabricated using biodegradable or non-biodegradable biomaterials. Commonly, the scaffold is seeded with chondrocytes and epithelial cells at the outer and luminal surfaces, respectively, to hasten tissue formation and improve functionality. To date, several clinical trials of tracheal replacement with tissue-engineered trachea have been performed. This article reviews the formation of cartilage tissue, epithelium and neovascularization of tissue-engineered trachea, together with the obstacles, possible solutions and future. Furthermore, the role of the bioreactor for in vitro tracheal graft formation and recently reported clinical applications of tracheal graft were also discussed. Generally, although encouraging results have been achieved, however, some obstacles remain to be resolved before the tissue-engineered trachea can be widely used in clinical settings.
Collapse
Affiliation(s)
- Jia Xian Law
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Ling Ling Liau
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Cheras, Kuala Lumpur, Malaysia
| | - Bin Saim Aminuddin
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Cheras, Kuala Lumpur, Malaysia; Ear, Nose & Throat Consultant Clinic, Ampang Puteri Specialist Hospital, 68000, Ampang, Selangor, Malaysia
| | - Bt Hj Idrus Ruszymah
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Cheras, Kuala Lumpur, Malaysia; Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, 56000, Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
23
|
Nonaka PN, Uriarte JJ, Campillo N, Oliveira VR, Navajas D, Farré R. Lung bioengineering: physical stimuli and stem/progenitor cell biology interplay towards biofabricating a functional organ. Respir Res 2016; 17:161. [PMID: 27894293 PMCID: PMC5126992 DOI: 10.1186/s12931-016-0477-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/22/2016] [Indexed: 01/18/2023] Open
Abstract
A current approach to obtain bioengineered lungs as a future alternative for transplantation is based on seeding stem cells on decellularized lung scaffolds. A fundamental question to be solved in this approach is how to drive stem cell differentiation onto the different lung cell phenotypes. Whereas the use of soluble factors as agents to modulate the fate of stem cells was established from an early stage of the research with this type of cells, it took longer to recognize that the physical microenvironment locally sensed by stem cells (e.g. substrate stiffness, 3D architecture, cyclic stretch, shear stress, air-liquid interface, oxygenation gradient) also contributes to their differentiation. The potential role played by physical stimuli would be particularly relevant in lung bioengineering since cells within the organ are physiologically subjected to two main stimuli required to facilitate efficient gas exchange: air ventilation and blood perfusion across the organ. The present review focuses on describing how the cell mechanical microenvironment can modulate stem cell differentiation and how these stimuli could be incorporated into lung bioreactors for optimizing organ bioengineering.
Collapse
Affiliation(s)
- Paula N Nonaka
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Juan J Uriarte
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Noelia Campillo
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Vinicius R Oliveira
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.,CIBER Enfermedades Respiratorias, Madrid, Spain.,Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain. .,CIBER Enfermedades Respiratorias, Madrid, Spain. .,Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain.
| |
Collapse
|
24
|
Konar D, Devarasetty M, Yildiz DV, Atala A, Murphy SV. Lung-On-A-Chip Technologies for Disease Modeling and Drug Development. Biomed Eng Comput Biol 2016; 7:17-27. [PMID: 27127414 PMCID: PMC4839966 DOI: 10.4137/becb.s34252] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 01/13/2023] Open
Abstract
Animal and two-dimensional cell culture models have had a profound impact on not only lung research but also medical research at large, despite inherent flaws and differences when compared with in vivo and clinical observations. Three-dimensional (3D) tissue models are a natural progression and extension of existing techniques that seek to plug the gaps and mitigate the drawbacks of two-dimensional and animal technologies. In this review, we describe the transition of historic models to contemporary 3D cell and organoid models, the varieties of current 3D cell and tissue culture modalities, the common methods for imaging these models, and finally, the applications of these models and imaging techniques to lung research.
Collapse
Affiliation(s)
- Dipasri Konar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Didem V Yildiz
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
25
|
Ikeda M, Imaizumi M, Yoshie S, Otsuki K, Miyake M, Hazama A, Wada I, Omori K. Regeneration of tracheal epithelium using mouse induced pluripotent stem cells. Acta Otolaryngol 2016; 136:373-8. [PMID: 26755348 DOI: 10.3109/00016489.2015.1121548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Conclusion The findings demonstrated the potential use of induced pluripotent stem cells for regeneration of tracheal epithelium. Objective Autologous tissue implantation techniques using skin or cartilage are often applied in cases of tracheal defects with laryngeal inflammatory lesions and malignant tumor invasion. However, these techniques are invasive with an unstable clinical outcome. The purpose of this study was to investigate regeneration in a tracheal defect site of nude rats after implantation of ciliated epithelium that was differentiated from induced pluripotent stem cells. Method Embryoid bodies were formed from mouse induced pluripotent stem cells. They were cultured with growth factors for 5 days, and then cultured at the air-liquid interface. The degree of differentiation achieved prior to implantation was determined by histological findings and the results of real-time polymerase chain reaction. Embryoid bodies including ciliated epithelium were embedded into collagen gel that served as an artificial scaffold, and then implanted into nude rats, creating an 'air-liquid interface model'. Histological evaluation was performed 7 days after implantation. Results The ciliated epithelial structure survived on the lumen side of regenerated tissue. It was demonstrated histologically that the structure was composed of ciliated epithelial cells.
Collapse
Affiliation(s)
- Masakazu Ikeda
- a Department of Otolaryngology , Fukushima Medical University , Fukushima , Japan
| | - Mitsuyoshi Imaizumi
- a Department of Otolaryngology , Fukushima Medical University , Fukushima , Japan
| | - Susumu Yoshie
- a Department of Otolaryngology , Fukushima Medical University , Fukushima , Japan
| | - Koshi Otsuki
- a Department of Otolaryngology , Fukushima Medical University , Fukushima , Japan
| | - Masao Miyake
- b Department of Cellular and Integrative Physiology , Fukushima Medical University , Fukushima , Japan
| | - Akihiro Hazama
- b Department of Cellular and Integrative Physiology , Fukushima Medical University , Fukushima , Japan
| | - Ikuo Wada
- c Department of Cell Science, Institute of Biomedical Sciences , Fukushima Medical University , Fukushima , Japan
| | - Koichi Omori
- a Department of Otolaryngology , Fukushima Medical University , Fukushima , Japan
| |
Collapse
|
26
|
Saksena R, Gao C, Wicox M, de Mel A. Tubular organ epithelialisation. J Tissue Eng 2016; 7:2041731416683950. [PMID: 28228931 PMCID: PMC5308438 DOI: 10.1177/2041731416683950] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 11/21/2016] [Indexed: 12/11/2022] Open
Abstract
Hollow, tubular organs including oesophagus, trachea, stomach, intestine, bladder and urethra may require repair or replacement due to disease. Current treatment is considered an unmet clinical need, and tissue engineering strategies aim to overcome these by fabricating synthetic constructs as tissue replacements. Smart, functionalised synthetic materials can act as a scaffold base of an organ and multiple cell types, including stem cells can be used to repopulate these scaffolds to replace or repair the damaged or diseased organs. Epithelial cells have not yet completely shown to have efficacious cell-scaffold interactions or good functionality in artificial organs, thus limiting the success of tissue-engineered grafts. Epithelial cells play an essential part of respective organs to maintain their function. Without successful epithelialisation, hollow organs are liable to stenosis, collapse, extensive fibrosis and infection that limit patency. It is clear that the source of cells and physicochemical properties of scaffolds determine the successful epithelialisation. This article presents a review of tissue engineering studies on oesophagus, trachea, stomach, small intestine, bladder and urethral constructs conducted to actualise epithelialised grafts.
Collapse
Affiliation(s)
- Rhea Saksena
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Chuanyu Gao
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Mathew Wicox
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Achala de Mel
- Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
27
|
Ghadiri M, Young PM, Traini D. Cell-based therapies for the treatment of idiopathic pulmonary fibrosis (IPF) disease. Expert Opin Biol Ther 2015; 16:375-87. [PMID: 26593230 DOI: 10.1517/14712598.2016.1124085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION During the last few decades, cell-based therapies have shown great potential to treat patients with lung diseases. It has been proposed that the administration of cells into an injured lung could be considered as a therapeutic method to repair and replace lost lung tissue. Using this method, transplanted cells with the ability to proliferate and differentiate into alveolar cells, have been suggested as a therapeutic strategy for IPF treatment. AREAS COVERED In this review, the latest investigations using various types of cells for IPF therapy have been presented. The cells studied for cell-based therapies in IPF are lung alveolar epithelial cells, lung resident stem cells and exogenous adult stem cells such as MSCs. EXPERT OPINION After many years of investigation, the use of cell-based therapies to treat IPF is still at the experimental phase. Problems include bioethical issues, safety of cell transplantation, routes of delivery and the dose and timing of administration. Further investigations are necessary to establish the best strategy for using cell-based therapies effectively for the treatment of IPF.
Collapse
Affiliation(s)
- Maliheh Ghadiri
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Paul M Young
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Daniela Traini
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| |
Collapse
|
28
|
Yoshie S, Imaizumi M, Nakamura R, Otsuki K, Ikeda M, Nomoto Y, Wada I, Omori K. Generation of airway epithelial cells with native characteristics from mouse induced pluripotent stem cells. Cell Tissue Res 2015; 364:319-30. [DOI: 10.1007/s00441-015-2304-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 09/28/2015] [Indexed: 02/02/2023]
|
29
|
Mou H, Brazauskas K, Rajagopal J. Personalized medicine for cystic fibrosis: establishing human model systems. Pediatr Pulmonol 2015; 50 Suppl 40:S14-23. [PMID: 26335952 DOI: 10.1002/ppul.23233] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/26/2015] [Indexed: 12/16/2022]
Abstract
With over 1,500 identifiable mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that result in distinct functional and phenotypical abnormalities, it is virtually impossible to perform randomized clinical trials to identify the best therapeutics for all patients. Therefore, a personalized medicine approach is essential. The only way to realistically accomplish this is through the development of improved in vitro human model systems. The lack of a readily available and infinite supply of human CFTR-expressing airway epithelial cells is a key bottleneck. We propose that a concerted two-pronged approach is necessary for patient-specific cystic fibrosis research to continue to prosper and realize its potential: (1) more effective culture and differentiation conditions for growing primary human airway and nasal epithelial cells and (2) the development of collective protocols for efficiently differentiating disease- and patient-specific induced pluripotent stem cells (iPSC) into pure populations of adult epithelial cells. Ultimately, we need a personalized human model system for cystic fibrosis with the capacity for uncomplicated bankability, widespread availability, and universal applicability for patient-specific disease modeling, novel pharmacotherapy investigation and screening, and readily executable genetic modification.
Collapse
Affiliation(s)
- Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Karissa Brazauskas
- Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Massachusetts
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Pulmonary Critical Care Unit, Internal Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
30
|
Zhang H, Fu W, Xu Z. Re-epithelialization: a key element in tracheal tissue engineering. Regen Med 2015; 10:1005-23. [PMID: 26388452 DOI: 10.2217/rme.15.68] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Trachea-tissue engineering is a thriving new field in regenerative medicine that is reaching maturity and yielding numerous promising results. In view of the crucial role that the epithelium plays in the trachea, re-epithelialization of tracheal substitutes has gradually emerged as the focus of studies in tissue-engineered trachea. Recent progress in our understanding of stem cell biology, growth factor interactions and transplantation immunobiology offer the prospect of optimization of a tissue-engineered tracheal epithelium. In addition, advances in cell culture technology and successful applications of clinical transplantation are opening up new avenues for the construction of a tissue-engineered tracheal epithelium. Therefore, this review summarizes current advances, unresolved obstacles and future directions in the reconstruction of a tissue-engineered tracheal epithelium.
Collapse
Affiliation(s)
- Hengyi Zhang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Zhiwei Xu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| |
Collapse
|
31
|
Fox E, Shojaie S, Wang J, Tseu I, Ackerley C, Bilodeau M, Post M. Three-dimensional culture and FGF signaling drive differentiation of murine pluripotent cells to distal lung epithelial cells. Stem Cells Dev 2015; 24:21-35. [PMID: 25079436 DOI: 10.1089/scd.2014.0227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Reciprocal signaling between the lung mesenchyme and epithelium is crucial for differentiation and branching morphogenesis. We hypothesized that the combination of signaling pathways comprising early epithelial-mesenchymal interactions and a 3D spatial environment are necessary for an efficient induction of embryonic and induced pluripotent stem cells (ESCs and iPSCs) into a lung cell phenotype with hallmarks of the distal niche. Aggregating early, but not late, embryonic lung mesenchyme with endoderm-induced mouse ESCs and iPSCs for 6 days resulted in organization into tubular structures and differentiation of the tubular lining cells to an NKX2-1(+)/SOX2(-)/SOX9(+)/proSFTPC(+) lineage. Over 80% of the endoderm-induced cells committed to an NKX2-1(+) lineage. Electron microscopy analysis demonstrated numerous multivesicular bodies and glycogen deposits in the tubular lining cells, characteristic features of type II epithelial cell progenitors. Using soluble FGFR2 receptor antagonists, we demonstrate that reciprocal fibroblast growth factor (FGF) 2, 7, and 10 signaling is essential for differentiation of endoderm-induced cells to an NKX2-1(+)/proSFTPC(+) phenotype within 3D aggregates. Only FGF2 was able to commit endoderm-induced cells in monolayer cultures to an NKX2-1(+) lineage, however with a significant lower efficiency (∼16%) than seen with mesenchyme. Thus, while FGF2 signaling alone can induce a primed population of ESCs and iPSCs, the cells do not differentiate to distal lung epithelial progenitors with the same efficiency and level of maturity that is achieved when the complex tissue and 3D environment of the developing lung is more accurately recapitulated.
Collapse
Affiliation(s)
- Emily Fox
- 1 Physiology and Experimental Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children , Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
32
|
Wong AP, Chin S, Xia S, Garner J, Bear CE, Rossant J. Efficient generation of functional CFTR-expressing airway epithelial cells from human pluripotent stem cells. Nat Protoc 2015; 10:363-81. [DOI: 10.1038/nprot.2015.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
33
|
Katsirntaki K, Mauritz C, Olmer R, Schmeckebier S, Sgodda M, Puppe V, Eggenschwiler R, Duerr J, Schubert SC, Schmiedl A, Ochs M, Cantz T, Salwig I, Szibor M, Braun T, Rathert C, Martens A, Mall MA, Martin U. Bronchoalveolar sublineage specification of pluripotent stem cells: effect of dexamethasone plus cAMP-elevating agents and keratinocyte growth factor. Tissue Eng Part A 2014; 21:669-82. [PMID: 25316003 DOI: 10.1089/ten.tea.2014.0097] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Respiratory progenitors can be efficiently generated from pluripotent stem cells (PSCs). However, further targeted differentiation into bronchoalveolar sublineages is still in its infancy, and distinct specifying effects of key differentiation factors are not well explored. Focusing on airway epithelial Clara cell generation, we analyzed the effect of the glucocorticoid dexamethasone plus cAMP-elevating agents (DCI) on the differentiation of murine embryonic and induced pluripotent stem cells (iPSCs) into bronchoalveolar epithelial lineages, and whether keratinocyte growth factor (KGF) might further influence lineage decisions. We demonstrate that DCI strongly induce expression of the Clara cell marker Clara cell secretory protein (CCSP). While KGF synergistically supports the inducing effect of DCI on alveolar markers with increased expression of surfactant protein (SP)-C and SP-B, an inhibitory effect on CCSP expression was shown. In contrast, neither KGF nor DCI seem to have an inducing effect on ciliated cell markers. Furthermore, the use of iPSCs from transgenic mice with CCSP promoter-dependent lacZ expression or a knockin of a YFP reporter cassette in the CCSP locus enabled detection of derivatives with Clara cell typical features. Collectively, DCI was shown to support bronchoalveolar specification of mouse PSCs, in particular Clara-like cells, and KGF to inhibit bronchial epithelial differentiation. The targeted in vitro generation of Clara cells with their important function in airway protection and regeneration will enable the evaluation of innovative cellular therapies in animal models of lung diseases.
Collapse
Affiliation(s)
- Katherina Katsirntaki
- 1 Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School , Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Weiss DJ. Concise review: current status of stem cells and regenerative medicine in lung biology and diseases. Stem Cells 2014; 32:16-25. [PMID: 23959715 DOI: 10.1002/stem.1506] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 07/24/2013] [Indexed: 12/29/2022]
Abstract
Lung diseases remain a significant and devastating cause of morbidity and mortality worldwide. In contrast to many other major diseases, lung diseases notably chronic obstructive pulmonary diseases (COPDs), including both asthma and emphysema, are increasing in prevalence and COPD is expected to become the third leading cause of disease mortality worldwide by 2020. New therapeutic options are desperately needed. A rapidly growing number of investigations of stem cells and cell therapies in lung biology and diseases as well as in ex vivo lung bioengineering have offered exciting new avenues for advancing knowledge of lung biology as well as providing novel potential therapeutic approaches for lung diseases. These initial observations have led to a growing exploration of endothelial progenitor cells and mesenchymal stem (stromal) cells in clinical trials of pulmonary hypertension and COPD with other clinical investigations planned. Ex vivo bioengineering of the trachea, larynx, diaphragm, and the lung itself with both biosynthetic constructs as well as decellularized tissues have been used to explore engineering both airway and vascular systems of the lung. Lung is thus a ripe organ for a variety of cell therapy and regenerative medicine approaches. Current state-of-the-art progress for each of the above areas will be presented as will discussion of current considerations for cell therapy-based clinical trials in lung diseases.
Collapse
Affiliation(s)
- Daniel J Weiss
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, USA
| |
Collapse
|
35
|
Vadasz S, Jensen T, Moncada C, Girard E, Zhang F, Blanchette A, Finck C. Second and third trimester amniotic fluid mesenchymal stem cells can repopulate a de-cellularized lung scaffold and express lung markers. J Pediatr Surg 2014; 49:1554-63. [PMID: 25475793 DOI: 10.1016/j.jpedsurg.2014.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND/PURPOSE This study examined the potential of amniotic fluid mesenchymal stem cells (AF-MSCs) to generate lung precursor cells in vitro and on a xenologous three-dimensional de-cellularized lung scaffold. METHODS AF-MSCs were isolated from human amniotic fluid obtained from 17-37 weeks gestation. Lung differentiation was induced on Matrigel or on de-cellularized rat lungs intra-tracheally injected with AF-MSCs by culturing with a modification of small airway growth medium (mSAGM) lacking retinoic acid (RA) and triodothyronine (T3) with addition of fibroblast growth factor-10 (FGF10). Cells and scaffolds were characterized by immunofluorescence and RT-PCR for markers of viability, proliferation, and lung distal airway differentiation (TTF-1(+) and SPC(+)) in the absence of markers of brain (TuJ1(-)) and thyroid (Pax8(-)). RESULTS After culture in mSAGM on either Matrigel or lung scaffolds, there were TTF-1(+)/TuJ1(-)/Pax8(-) cells, indicating a lung precursor phenotype. In addition, SPC(+) cells also evolved suggesting a more mature lung phenotype. CONCLUSIONS We demonstrate that mid- to late-trimester AF-MSCs can be induced to develop into lung precursor cells when cultured on the appropriate extracellular matrix (ECM), making them a viable source for use in cell therapy or development of an ex vivo tissue engineered lung.
Collapse
Affiliation(s)
- Stephanie Vadasz
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Todd Jensen
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Camilo Moncada
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Eric Girard
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Fan Zhang
- Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Alex Blanchette
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Christine Finck
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106.
| |
Collapse
|
36
|
Kajbafzadeh AM, Sabetkish N, Sabetkish S, Tavangar SM, Hossein Beigi RS, Talebi MA, Akbarzadeh A, Nikfarjam L. Lung tissue engineering and preservation of alveolar microstructure using a novel casting method. Biotech Histochem 2014; 90:111-23. [PMID: 25268847 DOI: 10.3109/10520295.2014.957724] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We used a rat model to decellularize and seed alveolar cells on a three-dimensional lung scaffold to preserve alveolar microarchitecture. We verified the preservation of terminal respiratory structure by casting and by scanning electron microscopy (SEM) of the casts after decellularization. Whole lungs were obtained from 12 healthy Sprague-Dawley rats, cannulated through the trachea under sterile conditions, and decellularized using a detergent-based method. Casting of both natural and decellularized lungs was performed to verify preservation of the inner microstructure of scaffolds for further cell seeding. Alveolar cell seeding was performed using green fluorescent protein (GFP) lung cells and non-GFP lung cells, and a peristaltic pump. We assessed cell seeding using histological and immunohistochemical staining, and enzymatic evaluation. All cellular components were removed completely from the scaffolds, and histological staining and SEM of casts were used to verify the preservation of tissue structure. Tensile tests verified conservation of biomechanical properties. The hydroxyproline content of decellularized lungs was similar to native lung. Histological and immunohistochemical evaluations showed effective cell seeding on decellularized matrices. Enzymatic measurement of trypsin and alpha 1 antitrypsin suggested the potential functional properties of the regenerated lungs. Casts produced by our method have satisfactory geometrical properties for further cell seeding of lung scaffolds. Preservation of micro-architecture and terminal alveoli that was confirmed by SEM of lung casts increases the probability of an effective cell seeding process.
Collapse
Affiliation(s)
- A-M Kajbafzadeh
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences , Tehran , Iran
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Nichols JE, Niles JA, Vega SP, Argueta LB, Eastaway A, Cortiella J. Modeling the lung: Design and development of tissue engineered macro- and micro-physiologic lung models for research use. Exp Biol Med (Maywood) 2014; 239:1135-69. [PMID: 24962174 DOI: 10.1177/1535370214536679] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Respiratory tract specific cell populations, or tissue engineered in vitro grown human lung, have the potential to be used as research tools to mimic physiology, toxicology, pathology, as well as infectious diseases responses of cells or tissues. Studies related to respiratory tract pathogenesis or drug toxicity testing in the past made use of basic systems where single cell populations were exposed to test agents followed by evaluations of simple cellular responses. Although these simple single-cell-type systems provided good basic information related to cellular responses, much more can be learned from cells grown in fabricated microenvironments which mimic in vivo conditions in specialized microfabricated chambers or by human tissue engineered three-dimensional (3D) models which allow for more natural interactions between cells. Recent advances in microengineering technology, microfluidics, and tissue engineering have provided a new approach to the development of 2D and 3D cell culture models which enable production of more robust human in vitro respiratory tract models. Complex models containing multiple cell phenotypes also provide a more reasonable approximation of what occurs in vivo without the confounding elements in the dynamic in vivo environment. The goal of engineering good 3D human models is the formation of physiologically functional respiratory tissue surrogates which can be used as pathogenesis models or in the case of 2D screening systems for drug therapy evaluation as well as human toxicity testing. We hope that this manuscript will serve as a guide for development of future respiratory tract model systems as well as a review of conventional models.
Collapse
Affiliation(s)
- Joan E Nichols
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Diseases, Galveston, TX 77555-0435, USA University of Texas Medical Branch, Department of Microbiology and Immunology, Galveston, TX 77555-0435, USA University of Texas Medical Branch, School of Medicine, Galveston, TX 77555-0435, USA
| | - Jean A Niles
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Diseases, Galveston, TX 77555-0435, USA
| | - Stephanie P Vega
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Diseases, Galveston, TX 77555-0435, USA University of Texas Medical Branch, Department of Microbiology and Immunology, Galveston, TX 77555-0435, USA
| | - Lissenya B Argueta
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Diseases, Galveston, TX 77555-0435, USA University of Texas Medical Branch, Department of Microbiology and Immunology, Galveston, TX 77555-0435, USA
| | - Adriene Eastaway
- University of Texas Medical Branch, Department of Internal Medicine, Division of Infectious Diseases, Galveston, TX 77555-0435, USA University of Texas Medical Branch, School of Medicine, Galveston, TX 77555-0435, USA
| | - Joaquin Cortiella
- University of Texas Medical Branch, Department of Anesthesiology, Galveston, TX 77555-0435, USA
| |
Collapse
|
38
|
Quan Y, Wang D. Clinical potentials of human pluripotent stem cells in lung diseases. Clin Transl Med 2014; 3:15. [PMID: 24995122 PMCID: PMC4072658 DOI: 10.1186/2001-1326-3-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/13/2014] [Indexed: 11/10/2022] Open
Abstract
Lung possesses very limited regenerative capacity. Failure to maintain homeostasis of lung epithelial cell populations has been implicated in the development of many life-threatening pulmonary diseases leading to substantial morbidity and mortality worldwide, and currently there is no known cure for these end-stage pulmonary diseases. Embryonic stem cells (ESCs) and somatic cell-derived induced pluripotent stem cells (iPSCs) possess unlimited self-renewal capacity and great potential to differentiate to various cell types of three embryonic germ layers (ectodermal, mesodermal, and endodermal). Therapeutic use of human ESC/iPSC-derived lung progenitor cells for regeneration of injured or diseased lungs will have an enormous clinical impact. This article provides an overview of recent advances in research on pluripotent stem cells in lung tissue regeneration and discusses technical challenges that must be overcome for their clinical applications in the future.
Collapse
Affiliation(s)
- Yuan Quan
- The Brown Foundation Institute of Molecular Medicine for the prevention of Human Diseases, University of Texas Medical School at Houston, 1825 Pressler Street/IMM 437D, Houston, TX 77030, USA
| | - Dachun Wang
- The Brown Foundation Institute of Molecular Medicine for the prevention of Human Diseases, University of Texas Medical School at Houston, 1825 Pressler Street/IMM 437D, Houston, TX 77030, USA
| |
Collapse
|
39
|
Calle EA, Ghaedi M, Sundaram S, Sivarapatna A, Tseng MK, Niklason LE. Strategies for whole lung tissue engineering. IEEE Trans Biomed Eng 2014; 61:1482-96. [PMID: 24691527 PMCID: PMC4126648 DOI: 10.1109/tbme.2014.2314261] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent work has demonstrated the feasibility of using decellularized lung extracellular matrix scaffolds to support the engineering of functional lung tissue in vitro. Rendered acellular through the use of detergents and other reagents, the scaffolds are mounted in organ-specific bioreactors where cells in the scaffold are provided with nutrients and appropriate mechanical stimuli such as ventilation and perfusion. Though initial studies are encouraging, a great deal remains to be done to advance the field and transition from rodent lungs to whole human tissue engineered lungs. To do so, a variety of hurdles must be overcome. In particular, a reliable source of human-sized scaffolds, as well as a method of terminal sterilization of scaffolds, must be identified. Continued research in lung cell and developmental biology will hopefully help identify the number and types of cells that will be required to regenerate functional lung tissue. Finally, bioreactor designs must be improved in order to provide more precise ventilation stimuli and vascular perfusion in order to avoid injury to or death of the cells cultivated within the scaffold. Ultimately, the success of efforts to engineer a functional lung in vitro will critically depend on the ability to create a fully endothelialized vascular network that provides sufficient barrier function and alveolar-capillary surface area to exchange gas at rates compatible with healthy lung function.
Collapse
Affiliation(s)
- Elizabeth A. Calle
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Mahboobe Ghaedi
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Sumati Sundaram
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Amogh Sivarapatna
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Michelle K. Tseng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Laura E. Niklason
- Department of Anesthesia and Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| |
Collapse
|
40
|
Zhou Q, Ye X, Sun R, Matsumoto Y, Moriyama M, Asano Y, Ajioka Y, Saijo Y. Differentiation of mouse induced pluripotent stem cells into alveolar epithelial cells in vitro for use in vivo. Stem Cells Transl Med 2014; 3:675-85. [PMID: 24763685 DOI: 10.5966/sctm.2013-0142] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Alveolar epithelial cells (AECs) differentiated from induced pluripotent stem cells (iPSCs) represent new opportunities in lung tissue engineering and cell therapy. In this study, we modified a two-step protocol for embryonic stem cells that resulted in a yield of ∼9% surfactant protein C (SPC)(+) alveolar epithelial type II (AEC II) cells from mouse iPSCs in a 12-day period. The differentiated iPSCs showed morphological characteristics similar to those of AEC II cells. When differentiated iPSCs were seeded and cultured in a decellularized mouse lung scaffold, the cells reformed an alveolar structure and expressed SPC or T1α protein (markers of AEC II or AEC I cells, respectively). Finally, the differentiated iPSCs were instilled intratracheally into a bleomycin-induced mouse acute lung injury model. The transplanted cells integrated into the lung alveolar structure and expressed SPC and T1α. Significantly reduced lung inflammation and decreased collagen deposition were observed following differentiated iPSC transplantation. In conclusion, we report a simple and rapid protocol for in vitro differentiation of mouse iPSCs into AECs. Differentiated iPSCs show potential for regenerating three-dimensional alveolar lung structure and can be used to abrogate lung injury.
Collapse
Affiliation(s)
- Qiliang Zhou
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Xulu Ye
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Ruowen Sun
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshifumi Matsumoto
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Masato Moriyama
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshiya Asano
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoichi Ajioka
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuo Saijo
- Department of Medical Oncology and Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Pediatric Hematology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China; Department of Neuroanatomy, Cell Biology, and Histology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
41
|
Bertoncello I, McQualter JL. Endogenous lung stem cells: what is their potential for use in regenerative medicine? Expert Rev Respir Med 2014; 4:349-62. [DOI: 10.1586/ers.10.21] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
42
|
The Potential of Wharton’s Jelly Derived Mesenchymal Stem Cells in Treating Patients with Cystic Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 833:23-9. [DOI: 10.1007/5584_2014_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
43
|
Nichols JE, Niles JA, Vega SP, Cortiella J. Novel in vitro respiratory models to study lung development, physiology, pathology and toxicology. Stem Cell Res Ther 2013; 4 Suppl 1:S7. [PMID: 24565038 PMCID: PMC4028843 DOI: 10.1186/scrt368] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Detailed studies of lung pathology in patients during the course of development of acute lung injury or respiratory distress are limited, and in the past information related to lung-specific responses has been derived from the study of lungs from patients who died at autopsy or from animal models. Development of good in vitro human tissue models would help to bridge the gap in our current knowledge of lung responses and provide a better understanding of lung development, physiology and pathology. In vitro models of simple one-cell or two-cell culture systems as well as complex multicellular lung analogs that reproduce defined components of specific human lung responses have already been realized. A benefit of current in vitro lung models is that hypotheses generated from review of data from human or animal disease studies can be tested directly in engineered human tissue models. Results of studies done using simple in vitro lung systems or more complex three-dimensional models have already been used to examine cell-based responses, physiologic functions, pathologic changes and even drug toxicity or drug responses. In the future we will create models with specific genetic profiles to test the importance of single gene products or pathways of significance. Recent development of microfluidics-based models that support high-throughput screening will allow early-stage toxicity testing in human systems and faster development of new and innovative medical products. Model design in the future will also allow for evaluation of multiple organ systems at once, providing a more holistic or whole-body approach to understanding human physiology and responses.
Collapse
|
44
|
Wang CY, Chiou GY, Chien Y, Wu CC, Wu TC, Lo WT, Chen SJ, Chiou SH, Peng HJ, Huang CF. Induced pluripotent stem cells without c-Myc reduce airway responsiveness and allergic reaction in sensitized mice. Transplantation 2013; 96:958-965. [PMID: 23989473 DOI: 10.1097/tp.0b013e3182a53ef7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Allergic disorders have increased substantially in recent years. Asthma is characterized by airway damage and remodeling. Reprogramming induced pluripotent stem cells (iPSCs) from adult somatic cells transfected by Oct-4/Sox-2/Klf-4, but not c-Myc, has shown the potential of embryonic-like cells. These cells have potential for multilineage differentiation and provide a resource for stem cell-based utility. However, the therapeutic potential of iPSCs without c-Myc (iPSC-w/o-c-Myc) in allergic diseases and airway hyperresponsiveness has not been investigated. The aim of this study was to evaluate the therapeutic effect of iPSC-w/o-c-Myc transplantation in a murine asthma model. METHODS BALB/c mice were sensitized with alum-adsorbed ovalbumin (OVA) and then challenged with aerosolized OVA. Phosphate-buffered saline or iPSC-w/o-c-Myc was then intravenously injected after inhalation. Serum allergen-specific antibody levels, airway hyperresponsiveness, cytokine levels in spleen cells and bronchoalveolar lavage fluid (BALF), and cellular distribution in BALF were then examined. RESULTS Treatment with iPSC-w/o-c-Myc effectively suppressed both Th1 and Th2 antibody responses, which was characterized by reduction in serum allergen-specific IgE, IgG, IgG1, and IgG2a levels as well as in interleukin-5 and interferon-γ levels in BALF and in OVA-incubated splenocytes. Meanwhile, regulatory cytokine, interleukin-10, was enhanced. Transplantation of iPSC-w/o-c-Myc also significantly attenuated cellular infiltration in BALF and allergic airway hyperresponsiveness. However, no tumor formation was observed 6 months after transplantation. CONCLUSIONS Administration of iPSC-w/o-c-Myc not only inhibited Th1 inflammatory responses but also had therapeutic effects on systemic allergic responses and airway hyperresponsiveness. iPSC-w/o-c-Myc transplantation may be a potential modality for treating allergic reactions and bronchial asthma.
Collapse
Affiliation(s)
- Chien-Ying Wang
- 1 Department of Medical Research & Education, Taipei Veterans General Hospital, Taipei, Taiwan. 2 School of Medicine, National Yang-Ming University, Taipei, Taiwan. 3 Department of Medicine, Tri-Service General Hospital, Taipei, Taiwan. 4 Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan. 5 Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. 6 Address correspondence to: Ching-Feng Huang, M.D., Ph.D., Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Cheng-Gong Road, Neihu District, Taipei 114, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Adam D, Perotin JM, Lebargy F, Birembaut P, Deslée G, Coraux C. [Regeneration of airway epithelium]. Rev Mal Respir 2013; 31:300-11. [PMID: 24750950 DOI: 10.1016/j.rmr.2013.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 10/04/2013] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Epithelial regeneration is a complex process. It can lead to the remodeling of the airway epithelium as in asthma, COPD or cystic fibrosis. BACKGROUND The development of in vivo and in vitro models has allowed the analysis of remodeling mechanisms and showed the role of components of extracellular matrix, proteases, cytokines and growth factors. Airway epithelial progenitors and stems cells have been studied in these models. However, their identification remains difficult. CONCLUSION Identification and characterization of airway epithelial progenitor/stem-cells, and a better knowledge of the regeneration process may allow the development of new therapeutic strategies for airway epithelial reconstitution.
Collapse
Affiliation(s)
- D Adam
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| | - J-M Perotin
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - F Lebargy
- Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - P Birembaut
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Laboratoire d'histologie Pol Bouin, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| | - G Deslée
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France; Service des maladies respiratoires, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France.
| | - C Coraux
- Inserm UMRS 903, CHU de Reims, 45, rue Cognacq-Jay, 51092 Reims, France
| |
Collapse
|
46
|
Otsuki K, Imaizumi M, Nomoto Y, Nomoto M, Wada I, Miyake M, Omori K. Effective embryoid body formation from induced pluripotent stem cells for regeneration of respiratory epithelium. Laryngoscope 2013; 124:E8-14. [PMID: 23686377 DOI: 10.1002/lary.24201] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/04/2013] [Accepted: 04/04/2013] [Indexed: 01/13/2023]
Abstract
OBJECTIVES/HYPOTHESIS We have previously demonstrated the potential use of induced pluripotent stem (iPS) cells for regeneration of respiratory epithelium by culturing embryoid bodies (EBs). The aim of the present study was to determine the most effective conditions for EB formation from iPS cells for regeneration of respiratory epithelium. STUDY DESIGN Experimental study. METHODS iPS cells cultured on a gelatin-coated dish were seeded on low-attachment plates for generating EBs. Under several conditions including the air-liquid interface (ALI) method, with varying cell numbers and suspension times, EBs were transferred to a gelatin-coated dish supplemented with growth factors. The shape, size, aggregation, and adhesion of EBs for iPS cell differentiation were evaluated, and the cultured tissue was histologically examined. RESULTS EBs appropriate for differentiation were observed using 1,000 cells after 5 days of suspension culture. Respiratory epithelium-like tissue was histologically observed. The ciliary epithelium was confirmed immunohistologically. CONCLUSIONS Based on the varying suspension times and cell numbers with the ALI method, this study presented effective conditions for EB formation from iPS cells for regeneration of respiratory epithelium.
Collapse
Affiliation(s)
- Koshi Otsuki
- Department of Otolaryngology, Fukushima Medical University, Fukushima, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Vrana NE, Lavalle P, Dokmeci MR, Dehghani F, Ghaemmaghami AM, Khademhosseini A. Engineering functional epithelium for regenerative medicine and in vitro organ models: a review. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:529-43. [PMID: 23705900 DOI: 10.1089/ten.teb.2012.0603] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent advances in the fields of microfabrication, biomaterials, and tissue engineering have provided new opportunities for developing biomimetic and functional tissues with potential applications in disease modeling, drug discovery, and replacing damaged tissues. An intact epithelium plays an indispensable role in the functionality of several organs such as the trachea, esophagus, and cornea. Furthermore, the integrity of the epithelial barrier and its degree of differentiation would define the level of success in tissue engineering of other organs such as the bladder and the skin. In this review, we focus on the challenges and requirements associated with engineering of epithelial layers in different tissues. Functional epithelial layers can be achieved by methods such as cell sheets, cell homing, and in situ epithelialization. However, for organs composed of several tissues, other important factors such as (1) in vivo epithelial cell migration, (2) multicell-type differentiation within the epithelium, and (3) epithelial cell interactions with the underlying mesenchymal cells should also be considered. Recent successful clinical trials in tissue engineering of the trachea have highlighted the importance of a functional epithelium for long-term success and survival of tissue replacements. Hence, using the trachea as a model tissue in clinical use, we describe the optimal structure of an artificial epithelium as well as challenges of obtaining a fully functional epithelium in macroscale. One of the possible remedies to address such challenges is the use of bottom-up fabrication methods to obtain a functional epithelium. Modular approaches for the generation of functional epithelial layers are reviewed and other emerging applications of microscale epithelial tissue models for studying epithelial/mesenchymal interactions in healthy and diseased (e.g., cancer) tissues are described. These models can elucidate the epithelial/mesenchymal tissue interactions at the microscale and provide the necessary tools for the next generation of multicellular engineered tissues and organ-on-a-chip systems.
Collapse
Affiliation(s)
- Nihal E Vrana
- 1 Institut National de la Santé et de la Recherche Médicale , INSERM, UMR-S 1121, "Biomatériaux et Bioingénierie," Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
48
|
Kokubun K, Pankajakshan D, Kim MJ, Agrawal DK. Differentiation of porcine mesenchymal stem cells into epithelial cells as a potential therapeutic application to facilitate epithelial regeneration. J Tissue Eng Regen Med 2013; 10:E73-83. [PMID: 23696537 DOI: 10.1002/term.1758] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 02/01/2013] [Accepted: 03/25/2013] [Indexed: 02/06/2023]
Abstract
Epithelial denudation is one of the characteristics of chronic asthma. To restore its functions, the airway epithelium has to rapidly repair the injuries and regenerate its structure and integrity. Mesenchymal stem cells (MSCs) have the ability to differentiate into many cell lineages. However, the differentiation of MSCs into epithelial cells has not been fully studied. Here, we examined the differentiation of MSCs into epithelial cells using three different media compositions with various growth supplementations. The MSCs were isolated from porcine bone marrow by density gradient centrifugation. The isolated MSCs were CD11(-) CD34(-) CD45(-) CD44(+) CD90(+) and CD105(+) by immunostaining and flow cytometry. MSCs were stimulated with EpiGRO (Millipore), BEpiCM (ScienCell) and AECGM (PromoCell) media for 5 and 10 days, and epithelial differentiation was assessed by qPCR (keratin 14, 18 and EpCAM), fluorometry (cytokeratin 7-8, cytokeratin 14-15-16-19 and EpCAM), western blot analysis (pancytokeratin, EpCAM) and flow cytometry (cytokeratin 7-8, cytokeratin 14-15-16-19 and EpCAM). The functional marker MUC1 was also assessed after 10 days of air-liquid interface (ALI) culture in optimized media. Cells cultured in BEpiCM containing fibroblast growth factor and prostaglandin E2 showed the highest expression of the epithelial markers: CK7-8 (85.90%); CK-14-15-16-19 (10.14%); and EpCAM (64.61%). The cells also expressed functional marker MUC1 after ALI culture. The differentiated MSCs when cultured in BEpiCM medium ex vivo in a bioreactor on a decellularized trachea for 10 days retained the epithelial-like phenotype. In conclusion, porcine bone marrow-derived MSCs demonstrate commitment to the epithelial lineage and might be a potential therapy for facilitating the repair of denuded airway epithelium.
Collapse
Affiliation(s)
- Kelsey Kokubun
- Center for Clinical and Translational Science and Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Divya Pankajakshan
- Center for Clinical and Translational Science and Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Min-Jung Kim
- Center for Clinical and Translational Science and Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Center for Clinical and Translational Science and Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
49
|
Conese M, Carbone A, Castellani S, Di Gioia S. Paracrine effects and heterogeneity of marrow-derived stem/progenitor cells: relevance for the treatment of respiratory diseases. Cells Tissues Organs 2013; 197:445-73. [PMID: 23652321 DOI: 10.1159/000348831] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2013] [Indexed: 11/19/2022] Open
Abstract
Stem cell-based treatment may represent a hope for the treatment of acute lung injury and pulmonary fibrosis, and other chronic lung diseases, such as cystic fibrosis, asthma and chronic obstructive pulmonary disease (COPD). It is well established in preclinical models that bone marrow-derived stem and progenitor cells exert beneficial effects on inflammation, immune responses and repairing of damage in virtually all lung-borne diseases. While it was initially thought that the positive outcome was due to a direct engraftment of these cells into the lung as endothelial and epithelial cells, paracrine factors are now considered the main mechanism through which stem and progenitor cells exert their therapeutic effect. This knowledge has led to the clinical use of marrow cells in pulmonary hypertension with endothelial progenitor cells (EPCs) and in COPD with mesenchymal stromal (stem) cells (MSCs). Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells, MSCs, EPCs and fibrocytes, encompass a wide array of cell subsets with different capacities of engraftment and injured tissue-regenerating potential. The characterization/isolation of the stem cell subpopulations represents a major challenge to improve the efficacy of transplantation protocols used in regenerative medicine and applied to lung disorders.
Collapse
Affiliation(s)
- Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | | | | | | |
Collapse
|
50
|
He X, Fu W, Zheng J. Cell sources for trachea tissue engineering: past, present and future. Regen Med 2013; 7:851-63. [PMID: 23164084 DOI: 10.2217/rme.12.96] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Trachea tissue engineering has been one of the most promising approaches to providing a potential clinical application for the treatment of long-segment tracheal stenosis. The sources of the cells are particularly important as the primary factor for tissue engineering. The use of appropriate cells seeded onto scaffolds holds huge promise as a means of engineering the trachea. Furthermore, appropriate cells would accelerate the regeneration of the tissue even without scaffolds. Besides autologous mature cells, various stem cells, including bone marrow-derived mesenchymal stem cells, adipose tissue-derived stem cells, umbilical cord blood-derived mesenchymal stem cells, amniotic fluid stem cells, embryonic stem cells and induced pluripotent stem cells, have received extensive attention in the field of trachea tissue engineering. Therefore, this article reviews the progress on different cell sources for engineering tracheal cartilage and epithelium, which can lead to a better selection and strategy for engineering the trachea.
Collapse
Affiliation(s)
- Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dong Fang Road, Shanghai 200127, China
| | | | | |
Collapse
|