1
|
Wan R, Liu Y, Yan J, Lin J. Cell therapy: A beacon of hope in the battle against pulmonary fibrosis. FASEB J 2025; 39:e70356. [PMID: 39873972 DOI: 10.1096/fj.202402790r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterized by abnormal activation of myofibroblasts and pathological remodeling of the extracellular matrix, with a poor prognosis and limited treatment options. Lung transplantation is currently the only approach that can extend the life expectancy of patients; however, its applicability is severely restricted due to donor shortages and patient-specific limitations. Therefore, the search for novel therapeutic strategies is imperative. In recent years, stem cells have shown great promise in the field of regenerative medicine due to their self-renewal capacity and multidirectional differentiation potential, and a growing body of literature supports the efficacy of stem cell therapy in PF treatment. This paper systematically summarizes the research progress of various stem cell types in the treatment of PF. Furthermore, it discusses the primary methods and clinical outcomes of stem cell therapy in PF, based on both preclinical and clinical data. Finally, the current challenges and key factors to consider in stem cell therapy for PF are objectively analyzed, and future directions for improving this therapy are proposed, providing new insights and references for the clinical treatment of PF patients.
Collapse
Affiliation(s)
- Ruyan Wan
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jingwen Yan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Lai S, Guo Z. Stem cell therapies for chronic obstructive pulmonary disease: mesenchymal stem cells as a promising treatment option. Stem Cell Res Ther 2024; 15:312. [PMID: 39300523 DOI: 10.1186/s13287-024-03940-9] [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: 04/28/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Chronic obstructive pulmonary disease(COPD) is an inflammatory disease characterized by the progressive and irreversible structural and functional damage of lung tissue. Although COPD is a significant global disease burden, the available treatments only ameliorate the symptoms, but cannot reverse lung damage. Researchers in regenerative medicine have examined the use of stem cell transplantation for treatment of COPD and other diseases because these cells have the potential for unlimited self-renewal and the ability to undergo directed differentiation. Stem cells are typically classified as embryonic stem cells, induced pluripotent stem cells, and adult stem cells (which includes mesenchymal stem cells [MSCs]), each with its own advantages and disadvantages regarding applications in regenerative medicine. Although the heterogeneity and susceptibility to senescence of MSCs make them require careful consideration for clinical applications. However, the low tumourigenicity and minimal ethical concerns of MSCs make them appear to be excellent candidates. This review summarizes the characteristics of various stem cell types and describes their therapeutic potential in the treatment of COPD, with a particular emphasis on MSCs. We aim to facilitate subsequent in-depth research and preclinical applications of MSCs by providing a comprehensive overview.
Collapse
Affiliation(s)
- Sumei Lai
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Zhifeng Guo
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
3
|
Guo S, Wang D. Novel insights into the potential applications of stem cells in pulmonary hypertension therapy. Respir Res 2024; 25:237. [PMID: 38849894 PMCID: PMC11162078 DOI: 10.1186/s12931-024-02865-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Pulmonary hypertension (PH) refers to a group of deadly lung diseases characterized by vascular lesions in the microvasculature and a progressive increase in pulmonary vascular resistance. The prevalence of PH has increased over time. Currently, the treatment options available for PH patients have limited efficacy, and none of them can fundamentally reverse pulmonary vascular remodeling. Stem cells represent an ideal seed with proven efficacy in clinical studies focusing on liver, cardiovascular, and nerve diseases. Since the potential therapeutic effect of mesenchymal stem cells (MSCs) on PH was first reported in 2006, many studies have demonstrated the efficacy of stem cells in PH animal models and suggested that stem cells can help slow the deterioration of lung tissue. Existing PH treatment studies basically focus on the paracrine action of stem cells, including protein regulation, exosome pathway, and cell signaling; however, the specific mechanisms have not yet been clarified. Apoptotic and afunctional pulmonary microvascular endothelial cells (PMVECs) and alveolar epithelial cells (AECs) are two fundamental promoters of PH although they have not been extensively studied by researchers. This review mainly focuses on the supportive communication and interaction between PMVECs and AECs as well as the potential restorative effect of stem cells on their injury. In the future, more studies are needed to prove these effects and explore more radical cures for PH.
Collapse
Affiliation(s)
- Sijia Guo
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Dachun Wang
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- The Brown Foundation Institute of Molecular Medicine for the prevention of Human Diseases, University of Texas Medical School at Houston, Houston, TX, USA
| |
Collapse
|
4
|
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
|
5
|
Kitamura T, Misu M, Yoshikawa M, Ouji Y. Differentiation of embryonic stem cells into lung-like cells using lung-derived matrix sheets. Biochem Biophys Res Commun 2023; 686:149197. [PMID: 37924668 DOI: 10.1016/j.bbrc.2023.149197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/17/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Various extracellular matrix (ECM) in the lungs regulate tissue development and homeostasis, as well as provide support for cell structures. However, few studies regarding the effects of lung cell differentiation using lung-derived ECM (LM) alone have been reported. The present study investigated the capability of lung-derived matrix sheets (LMSs) to induce lung cell differentiation using mouse embryonic stem (ES) cells. Expressions of lung-related cell markers were significantly upregulated in ES-derived embryoid bodies (EBs) cultured on an LMS for two weeks. Moreover, immunohistochemical analysis of EBs grown on LMSs revealed differentiation of various lung-related cells. These results suggest that an LMS can be used to promote differentiation of stem cells into lung cells.
Collapse
Affiliation(s)
- Tomotaka Kitamura
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Masayasu Misu
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Masahide Yoshikawa
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Yukiteru Ouji
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
6
|
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
|
7
|
Kotasová H, Capandová M, Pelková V, Dumková J, Koledová Z, Remšík J, Souček K, Garlíková Z, Sedláková V, Rabata A, Vaňhara P, Moráň L, Pečinka L, Porokh V, Kučírek M, Streit L, Havel J, Hampl A. Expandable Lung Epithelium Differentiated from Human Embryonic Stem Cells. Tissue Eng Regen Med 2022; 19:1033-1050. [PMID: 35670910 PMCID: PMC9478014 DOI: 10.1007/s13770-022-00458-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The progenitors to lung airway epithelium that are capable of long-term propagation may represent an attractive source of cells for cell-based therapies, disease modeling, toxicity testing, and others. Principally, there are two main options for obtaining lung epithelial progenitors: (i) direct isolation of endogenous progenitors from human lungs and (ii) in vitro differentiation from some other cell type. The prime candidates for the second approach are pluripotent stem cells, which may provide autologous and/or allogeneic cell resource in clinically relevant quality and quantity. METHODS By exploiting the differentiation potential of human embryonic stem cells (hESC), here we derived expandable lung epithelium (ELEP) and established culture conditions for their long-term propagation (more than 6 months) in a monolayer culture without a need of 3D culture conditions and/or cell sorting steps, which minimizes potential variability of the outcome. RESULTS These hESC-derived ELEP express NK2 Homeobox 1 (NKX2.1), a marker of early lung epithelial lineage, display properties of cells in early stages of surfactant production and are able to differentiate to cells exhibitting molecular and morphological characteristics of both respiratory epithelium of airway and alveolar regions. CONCLUSION Expandable lung epithelium thus offer a stable, convenient, easily scalable and high-yielding cell source for applications in biomedicine.
Collapse
Affiliation(s)
- Hana Kotasová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Michaela Capandová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Vendula Pelková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Jana Dumková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Zuzana Koledová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Ján Remšík
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Institute of Biophysics, The Czech Academy of Sciences, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Current Address: Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Karel Souček
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Institute of Biophysics, The Czech Academy of Sciences, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Zuzana Garlíková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Veronika Sedláková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Anas Rabata
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Petr Vaňhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lukáš Moráň
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Lukáš Pečinka
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Volodymyr Porokh
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Martin Kučírek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Libor Streit
- Department of Plastic and Cosmetic Surgery, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Plastic and Cosmetic Surgery, St. Anne's Faculty Hospital, Brno, Czech Republic
| | - Josef Havel
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| |
Collapse
|
8
|
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
|
9
|
Wiśniewska J, Sadowska A, Wójtowicz A, Słyszewska M, Szóstek-Mioduchowska A. Perspective on Stem Cell Therapy in Organ Fibrosis: Animal Models and Human Studies. Life (Basel) 2021; 11:life11101068. [PMID: 34685439 PMCID: PMC8538998 DOI: 10.3390/life11101068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components that result from the disruption of regulatory processes responsible for ECM synthesis, deposition, and remodeling. Fibrosis develops in response to a trigger or injury and can occur in nearly all organs of the body. Thus, fibrosis leads to severe pathological conditions that disrupt organ architecture and cause loss of function. It has been estimated that severe fibrotic disorders are responsible for up to one-third of deaths worldwide. Although intensive research on the development of new strategies for fibrosis treatment has been carried out, therapeutic approaches remain limited. Since stem cells, especially mesenchymal stem cells (MSCs), show remarkable self-renewal, differentiation, and immunomodulatory capacity, they have been intensively tested in preclinical studies and clinical trials as a potential tool to slow down the progression of fibrosis and improve the quality of life of patients with fibrotic disorders. In this review, we summarize in vitro studies, preclinical studies performed on animal models of human fibrotic diseases, and recent clinical trials on the efficacy of allogeneic and autologous stem cell applications in severe types of fibrosis that develop in lungs, liver, heart, kidney, uterus, and skin. Although the results of the studies seem to be encouraging, there are many aspects of cell-based therapy, including the cell source, dose, administration route and frequency, timing of delivery, and long-term safety, that remain open areas for future investigation. We also discuss the contemporary status, challenges, and future perspectives of stem cell transplantation for therapeutic options in fibrotic diseases as well as we present recent patents for stem cell-based therapies in organ fibrosis.
Collapse
|
10
|
Tong Y, Zuo J, Yue D. Application Prospects of Mesenchymal Stem Cell Therapy for Bronchopulmonary Dysplasia and the Challenges Encountered. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9983664. [PMID: 33997051 PMCID: PMC8110410 DOI: 10.1155/2021/9983664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature babies, especially affecting those with very low or extremely low birth weights. Survivors experience adverse lung and neurological defects including cognitive dysfunction. This impacts the prognosis of children with BPD and may result in developmental delays. The currently available options for the treatment of BPD are limited owing to low efficacy or several side effects; therefore, there is a lack of effective treatments for BPD. The treatment for BPD must help in the repair of damaged lung tissue and promote further growth of the lung tissue. In recent years, the emergence of stem cell therapy, especially mesenchymal stem cell (MSC) therapy, has improved the treatment of BPD to a great extent. This article briefly reviews the advantages, research progress, and challenges faced with the use of MSCs in the treatment of BPD. Stem cell therapy is beneficial as it repairs damaged tissues by reducing inflammation, fibrosis, and by acting against oxidative stress damage. Experimental trials have also proven that MSCs provide a promising avenue for BPD treatment. However, there are challenges such as the possibility of MSCs contributing to tumorous growths, the presence of heterogeneous cell populations resulting in variable efficacy, and the ethical considerations regarding the use of this treatment in humans. Therefore, more research must be conducted to determine whether MSC therapy can be approved as a treatment option for BPD.
Collapse
Affiliation(s)
- Yajie Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Jingye Zuo
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| |
Collapse
|
11
|
Kawakita N, Toba H, Miyoshi K, Sakamoto S, Matsumoto D, Takashima M, Aoyama M, Inoue S, Morimoto M, Nishino T, Takizawa H, Tangoku A. Bronchioalveolar stem cells derived from mouse-induced pluripotent stem cells promote airway epithelium regeneration. Stem Cell Res Ther 2020; 11:430. [PMID: 33008488 PMCID: PMC7531137 DOI: 10.1186/s13287-020-01946-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/20/2020] [Indexed: 12/20/2022] Open
Abstract
Background Bronchioalveolar stem cells (BASCs) located at the bronchioalveolar-duct junction (BADJ) are stem cells residing in alveoli and terminal bronchioles that can self-renew and differentiate into alveolar type (AT)-1 cells, AT-2 cells, club cells, and ciliated cells. Following terminal-bronchiole injury, BASCs increase in number and promote repair. However, whether BASCs can be differentiated from mouse-induced pluripotent stem cells (iPSCs) remains unreported, and the therapeutic potential of such cells is unclear. We therefore sought to differentiate BASCs from iPSCs and examine their potential for use in the treatment of epithelial injury in terminal bronchioles. Methods BASCs were induced using a modified protocol for differentiating mouse iPSCs into AT-2 cells. Differentiated iPSCs were intratracheally transplanted into naphthalene-treated mice. The engraftment of BASCs into the BADJ and their subsequent ability to promote repair of injury to the airway epithelium were evaluated. Results Flow cytometric analysis revealed that BASCs represented ~ 7% of the cells obtained. Additionally, ultrastructural analysis of these iPSC-derived BASCs via transmission electron microscopy showed that the cells containing secretory granules harboured microvilli, as well as small and immature lamellar body-like structures. When the differentiated iPSCs were intratracheally transplanted in naphthalene-induced airway epithelium injury, transplanted BASCs were found to be engrafted in the BADJ epithelium and alveolar spaces for 14 days after transplantation and to maintain the BASC phenotype. Notably, repair of the terminal-bronchiole epithelium was markedly promoted after transplantation of the differentiated iPSCs. Conclusions Mouse iPSCs could be differentiated in vitro into cells that display a similar phenotype to BASCs. Given that the differentiated iPSCs promoted epithelial repair in the mouse model of naphthalene-induced airway epithelium injury, this method may serve as a basis for the development of treatments for terminal-bronchiole/alveolar-region disorders.
Collapse
Affiliation(s)
- Naoya Kawakita
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Hiroaki Toba
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Keiko Miyoshi
- Department of Molecular Biology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Shinichi Sakamoto
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Daisuke Matsumoto
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Mika Takashima
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Mariko Aoyama
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Seiya Inoue
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masami Morimoto
- Department of Breast Surgery, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Takeshi Nishino
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Hiromitsu Takizawa
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Akira Tangoku
- Department of Thoracic and Endocrine Surgery and Oncology, Institute of Biomedical Sciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
12
|
Li Y, Wu Q, Sun X, Shen J, Chen H. Organoids as a Powerful Model for Respiratory Diseases. Stem Cells Int 2020; 2020:5847876. [PMID: 32256609 PMCID: PMC7086445 DOI: 10.1155/2020/5847876] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/12/2020] [Accepted: 02/27/2020] [Indexed: 02/05/2023] Open
Abstract
Insults to the alveoli usually lead to inefficient gas exchange or even respiratory failure, which is difficult to model in animal studies. Over the past decade, stem cell-derived self-organizing three-dimensional organoids have emerged as a new avenue to recapitulate respiratory diseases in a dish. Alveolar organoids have improved our understanding of the mechanisms underlying tissue homeostasis and pathological alterations in alveoli. From this perspective, we review the state-of-the-art technology on establishing alveolar organoids from endogenous lung epithelial stem/progenitor cells or pluripotent stem cells, as well as the use of alveolar organoids for the study of respiratory diseases, including idiopathic pulmonary fibrosis, tuberculosis infection, and respiratory virus infection. We also discuss challenges that need to be overcome for future application of alveolar organoids in individualized medicine.
Collapse
Affiliation(s)
- Yu Li
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
| | - Qi Wu
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Xin Sun
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
| | - Jun Shen
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| | - Huaiyong Chen
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, China
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China
| |
Collapse
|
13
|
Mokhber Dezfouli MR, Sadeghian Chaleshtori S, Moradmand A, Basiri M, Baharvand H, Tahamtani Y. Hydrocortisone Promotes Differentiation of Mouse Embryonic Stem Cell-Derived Definitive Endoderm toward Lung Alveolar Epithelial Cells. CELL JOURNAL 2018; 20:469-476. [PMID: 30123992 PMCID: PMC6099149 DOI: 10.22074/cellj.2019.5521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/09/2017] [Indexed: 12/12/2022]
Abstract
Objective The ability to generate lung alveolar epithelial type II (ATII) cells from pluripotent stem cells (PSCs) enables the study of lung development, regenerative medicine, and modeling of lung diseases. The establishment of defined, scalable differentiation methods is a step toward this goal. This study intends to investigate the competency of small molecule induced mouse embryonic stem cell-derived definitive endoderm (mESC-DE) cells towards ATII cells. Materials and Methods In this experimental study, we designed a two-step differentiation protocol. mESC line Royan B20 (RB20) was induced to differentiate into DE (6 days) and then into ATII cells (9 days) by using an adherent culture method. To induce differentiation, we treated the mESCs for 6 days in serum-free differentiation (SFD) media and induced them with 200 nM small molecule inducer of definitive endoderm 2 (IDE2). For days 7-15 (9 days) of induction, we treated the resultant DE cells with new differentiation media comprised of 100 ng/ml fibroblast growth factor (FGF2) (group F), 0.5 μg/ml hydrocortisone (group H), and A549 conditioned medium (A549 CM) (group CM) in SFD media. Seven different combinations of factors were tested to assess the efficiencies of these factors to promote differentiation. The expressions of DE- and ATII-specific markers were investigated during each differentiation step. Results Although both F and H (alone and in combination) promoted differentiation through ATII-like cells, the highest percentage of surfactant protein C (SP-C) expressing cells (~37%) were produced in DE-like cells treated by F+H+CM. Ultrastructural analyses also confirmed the presence of lamellar bodies (LB) in the ATII-like cells. Conclusion These results suggest that hydrocortisone can be a promoting factor in alveolar fate differentiation of IDE2-induced mESC-DE cells. These cells have potential for drug screening and cell-replacement therapies.
Collapse
Affiliation(s)
- Mohammad Reza Mokhber Dezfouli
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran. Electronic Address:
| | - Sirous Sadeghian Chaleshtori
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Azadeh Moradmand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell. Electronic Address:
| |
Collapse
|
14
|
Kawai N, Ouji Y, Sakagami M, Tojo T, Sawabata N, Yoshikawa M, Taniguchi S. Induction of lung-like cells from mouse embryonic stem cells by decellularized lung matrix. Biochem Biophys Rep 2018; 15:33-38. [PMID: 29942870 PMCID: PMC6010970 DOI: 10.1016/j.bbrep.2018.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/01/2023] Open
Abstract
Decellularization of tissues is a recently developed technique mostly used to provide a 3-dimensional matrix structure of the original organ, including decellularized lung tissues for lung transplantation. Based on the results of the present study, we propose new utilization of decellularized tissues as inducers of stem cell differentiation. Decellularized lung matrix (L-Mat) samples were prepared from mouse lungs by SDS treatment, then the effects of L-Mat on differentiation of ES cells into lung cells were investigated. ES cell derived-embryoid bodies (EBs) were transplanted into L-Mat samples and cultured for 2 weeks. At the end of the culture, expressions of lung cell-related markers, such as TTF-1 and SP-C (alveolar type II cells), AQP5 (alveolar type I cells), and CC10 (club cells), were detected in EB outgrowths in L-Mat, while those were not found in EB outgrowths attached to the dish. Our results demonstrated that L-Mat has an ability to induce differentiation of ES cells into lung-like cells. Differentiation of ES cells by decellularized lung matrix (L-Mat) was investigated. L-Mat induced differentiation of various lung cell-like cells from ES cells. L-Mat plays an important role for inducing differentiation of lung cells.
Collapse
Affiliation(s)
- Norikazu Kawai
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Yukiteru Ouji
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
- Correspondence to: Department of Pathogen, Infection and Immunity, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan.
| | - Masaharu Sakagami
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Takashi Tojo
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Noriyoshi Sawabata
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Masahide Yoshikawa
- Department of Pathogen, Infection and Immunity, Nara Medical University, Kashihara, Nara, Japan
| | - Shigeki Taniguchi
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
15
|
Ghaedi M, Le AV, Hatachi G, Beloiartsev A, Rocco K, Sivarapatna A, Mendez JJ, Baevova P, Dyal RN, Leiby KL, White ES, Niklason LE. Bioengineered lungs generated from human iPSCs-derived epithelial cells on native extracellular matrix. J Tissue Eng Regen Med 2017; 12:e1623-e1635. [PMID: 29024475 DOI: 10.1002/term.2589] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/28/2017] [Accepted: 10/03/2017] [Indexed: 01/05/2023]
Abstract
The development of an alternative source for donor lungs would change the paradigm of lung transplantation. Recent studies have demonstrated the potential feasibility of using decellularized lungs as scaffolds for lung tissue regeneration and subsequent implantation. However, finding a reliable cell source and the ability to scale up for recellularization of the lung scaffold still remain significant challenges. To explore the possibility of regeneration of human lung tissue from stem cells in vitro, populations of lung progenitor cells were generated from human iPSCs. To explore the feasibility of producing engineered lungs from stem cells, we repopulated decellularized human lung and rat lungs with iPSC-derived epithelial progenitor cells. The iPSCs-derived epithelial progenitor cells lined the decellularized human lung and expressed most of the epithelial markers when were cultured in a lung bioreactor system. In decellularized rat lungs, these human-derived cells attach and proliferate in a manner similar to what was observed in the decellularized human lung. Our results suggest that repopulation of lung matrix with iPSC-derived lung epithelial cells may be a viable strategy for human lung regeneration and represents an important early step toward translation of this technology.
Collapse
Affiliation(s)
- Mahboobe Ghaedi
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Andrew V Le
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Go Hatachi
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Arkadi Beloiartsev
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Kevin Rocco
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Amogh Sivarapatna
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Julio J Mendez
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Pavlina Baevova
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Rachel N Dyal
- Internal Medicine, Pulmonary and Critical Care, University of Michigan, Ann Arbor, MI, USA
| | - Katie L Leiby
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Eric S White
- Internal Medicine, Pulmonary and Critical Care, University of Michigan, Ann Arbor, MI, USA
| | - Laura E Niklason
- Department of Anesthesiology, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
16
|
Vats A, Tolley NS, Bishop AE, Polak JM. Embryonic Stem Cells and Tissue Engineering: Delivering Stem Cells to the Clinic. J R Soc Med 2017; 98:346-50. [PMID: 16055897 PMCID: PMC1181832 DOI: 10.1177/014107680509800804] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- A Vats
- Tissue Engineering and Regenerative Medicine Centre, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, 369 Fulham Road, London SW10 9NH, UK.
| | | | | | | |
Collapse
|
17
|
Azargoon A, Negahdari B. Lung regeneration using amniotic fluid mesenchymal stem cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:447-451. [PMID: 28675062 DOI: 10.1080/21691401.2017.1337023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Respiratory diseases, such as chronic obstructive pulmonary disease (COPD), pulmonary hypertension and lung fibrosis, are yet a major challenge in the world and they result in irreversible structural lung damage. Lung transplantation as the only therapeutic option face some major challenges like graft rejection and cancer, arising as a result of immunosuppression. A low survival rate faced by lung transplantation patients is presently limited to approximately 5 years. Lungs shortage therefore calls for a mechanism that would increase the availability of suitable organs for transplantation. In this review, we give an update on the use of amniotic fluid mesenchymal stem cells (AFMSCs) as an optimal source for lungs scaffold re-cellularization, due to their limitless accessibility and possibility for proliferation and differentiation. Further studies will be required in tissue engineering (TE) and regenerative medicine (RM), especially shifting our focus towards AFMSCs as a cell source for this regeneration.
Collapse
Affiliation(s)
- Alireza Azargoon
- a Department of Internal Medicine , Lorestan University of Medical Sciences , Khoramabad , Iran
| | - Babak Negahdari
- b Department of Medical Biotechnology, School of advanced Technologies in Medicine, Tehran , University of Medical sciences , Tehran , Iran
| |
Collapse
|
18
|
Singh VK, Saini A, Kalsan M, Kumar N, Chandra R. Describing the Stem Cell Potency: The Various Methods of Functional Assessment and In silico Diagnostics. Front Cell Dev Biol 2016; 4:134. [PMID: 27921030 PMCID: PMC5118841 DOI: 10.3389/fcell.2016.00134] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Stem cells are defined by their capabilities to self-renew and give rise to various types of differentiated cells depending on their potency. They are classified as pluripotent, multipotent, and unipotent as demonstrated through their potential to generate the variety of cell lineages. While pluripotent stem cells may give rise to all types of cells in an organism, Multipotent and Unipotent stem cells remain restricted to the particular tissue or lineages. The potency of these stem cells can be defined by using a number of functional assays along with the evaluation of various molecular markers. These molecular markers include diagnosis of transcriptional, epigenetic, and metabolic states of stem cells. Many reports are defining the particular set of different functional assays, and molecular marker used to demonstrate the developmental states and functional capacities of stem cells. The careful evaluation of all these methods could help in generating standard identifying procedures/markers for them.
Collapse
Affiliation(s)
- Vimal K Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Manisha Kalsan
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Neeraj Kumar
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi Delhi, India
| |
Collapse
|
19
|
Schilders KAA, Eenjes E, van Riet S, Poot AA, Stamatialis D, Truckenmüller R, Hiemstra PS, Rottier RJ. Regeneration of the lung: Lung stem cells and the development of lung mimicking devices. Respir Res 2016; 17:44. [PMID: 27107715 PMCID: PMC4842297 DOI: 10.1186/s12931-016-0358-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/25/2016] [Indexed: 01/07/2023] Open
Abstract
Inspired by the increasing burden of lung associated diseases in society and an growing demand to accommodate patients, great efforts by the scientific community produce an increasing stream of data that are focused on delineating the basic principles of lung development and growth, as well as understanding the biomechanical properties to build artificial lung devices. In addition, the continuing efforts to better define the disease origin, progression and pathology by basic scientists and clinicians contributes to insights in the basic principles of lung biology. However, the use of different model systems, experimental approaches and readout systems may generate somewhat conflicting or contradictory results. In an effort to summarize the latest developments in the lung epithelial stem cell biology, we provide an overview of the current status of the field. We first describe the different stem cells, or progenitor cells, residing in the homeostatic lung. Next, we focus on the plasticity of the different cell types upon several injury-induced activation or repair models, and highlight the regenerative capacity of lung cells. Lastly, we summarize the generation of lung mimics, such as air-liquid interface cultures, organoids and lung on a chip, that are required to test emerging hypotheses. Moreover, the increasing collaboration between distinct specializations will contribute to the eventual development of an artificial lung device capable of assisting reduced lung function and capacity in human patients.
Collapse
Affiliation(s)
- Kim A A Schilders
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Evelien Eenjes
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - André A Poot
- Department of Biomaterials Science and Technology, University of Twente, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, P.O Box 217, 7500 AE, Enschede, The Netherlands
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, University of Twente, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, P.O Box 217, 7500 AE, Enschede, The Netherlands
| | - Roman Truckenmüller
- Department of Complex Tissue Regeneration, Maastricht University, Faculty of Health, Medicine and Life Sciences, MERLN Institute for Technology-Inspired Regenerative Medicine, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
20
|
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
|
21
|
Sehic A, Utheim ØA, Ommundsen K, Utheim TP. Pre-Clinical Cell-Based Therapy for Limbal Stem Cell Deficiency. J Funct Biomater 2015; 6:863-88. [PMID: 26343740 PMCID: PMC4598682 DOI: 10.3390/jfb6030863] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/10/2015] [Accepted: 08/21/2015] [Indexed: 12/13/2022] Open
Abstract
The cornea is essential for normal vision by maintaining transparency for light transmission. Limbal stem cells, which reside in the corneal periphery, contribute to the homeostasis of the corneal epithelium. Any damage or disease affecting the function of these cells may result in limbal stem cell deficiency (LSCD). The condition may result in both severe pain and blindness. Transplantation of ex vivo cultured cells onto the cornea is most often an effective therapeutic strategy for LSCD. The use of ex vivo cultured limbal epithelial cells (LEC), oral mucosal epithelial cells, and conjunctival epithelial cells to treat LSCD has been explored in humans. The present review focuses on the current state of knowledge of the many other cell-based therapies of LSCD that have so far exclusively been explored in animal models as there is currently no consensus on the best cell type for treating LSCD. Major findings of all these studies with special emphasis on substrates for culture and transplantation are systematically presented and discussed. Among the many potential cell types that still have not been used clinically, we conclude that two easily accessible autologous sources, epidermal stem cells and hair follicle-derived stem cells, are particularly strong candidates for future clinical trials.
Collapse
Affiliation(s)
- Amer Sehic
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Sognsvannsveien 10, Oslo 0372, Norway.
| | - Øygunn Aass Utheim
- Department of Ophthalmology, Oslo University Hospital, Kirkeveien 166, Oslo 0407, Norway.
| | - Kristoffer Ommundsen
- Department of Medical Biochemistry, Oslo University Hospital, Kirkeveien 166, Oslo 0407, Norway.
| | - Tor Paaske Utheim
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Sognsvannsveien 10, Oslo 0372, Norway.
- Department of Medical Biochemistry, Oslo University Hospital, Kirkeveien 166, Oslo 0407, Norway.
| |
Collapse
|
22
|
Ghaedi M, Niklason LE, Williams J. Development of Lung Epithelium from Induced Pluripotent Stem Cells. CURRENT TRANSPLANTATION REPORTS 2015; 2:81-89. [PMID: 26052480 PMCID: PMC4452199 DOI: 10.1007/s40472-014-0039-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Considerable progress has been made in the field of in vitro development of alveolar epithelium from induced pluripotent stem cells. Patient specific derived alveolar cells could potentially populate tissue engineered lungs, provide a cell source for drug testing or function as a model for research into lung diseases. Induced to pluripotency through a variety of techniques, stem cells can be differentiated to alveolar epithelium through exposure to a variety of different culture conditions and growth media. The ultimate success of differentiated cells for translational medicine applications will depend on further advances in the understanding of the human lung developmental pathway, and successful application to in vitro culture. In this review will focus the major signaling pathways and molecules in lung development and the existing protocol for directed different ion of iPSC and hESC to cells resembling respiratory epithelium in vitro.
Collapse
Affiliation(s)
- Mahboobe Ghaedi
- Departments of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut, 06520, USA
| | - Laura E. Niklason
- Departments of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut, 06520, USA
| | - Jordana Williams
- Departments of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut, 06520, USA
| |
Collapse
|
23
|
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]
|
24
|
Wu B, Wang C, Hei F, Long C, Chen M, Yang S, Yu J, Ju Z. The differentiation of rat-induced pluripotent stem cells into alveolar type II epithelial cells with a three-step induction protocol. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2014.1001435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
25
|
Katona RL. De novo formed satellite DNA-based mammalian artificial chromosomes and their possible applications. Chromosome Res 2015; 23:143-57. [DOI: 10.1007/s10577-014-9458-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
26
|
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
|
27
|
Cerrada A, de la Torre P, Grande J, Haller T, Flores AI, Pérez-Gil J. Human decidua-derived mesenchymal stem cells differentiate into functional alveolar type II-like cells that synthesize and secrete pulmonary surfactant complexes. PLoS One 2014; 9:e110195. [PMID: 25333871 PMCID: PMC4198213 DOI: 10.1371/journal.pone.0110195] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 09/18/2014] [Indexed: 02/05/2023] Open
Abstract
Lung alveolar type II (ATII) cells are specialized in the synthesis and secretion of pulmonary surfactant, a lipid-protein complex that reduces surface tension to minimize the work of breathing. Surfactant synthesis, assembly and secretion are closely regulated and its impairment is associated with severe respiratory disorders. At present, well-established ATII cell culture models are not available. In this work, Decidua-derived Mesenchymal Stem Cells (DMSCs) have been differentiated into Alveolar Type II- Like Cells (ATII-LCs), which display membranous cytoplasmic organelles resembling lamellar bodies, the organelles involved in surfactant storage and secretion by native ATII cells, and accumulate disaturated phospholipid species, a surfactant hallmark. Expression of characteristic ATII cells markers was demonstrated in ATII-LCs at gene and protein level. Mimicking the response of ATII cells to secretagogues, ATII-LCs were able to exocytose lipid-rich assemblies, which displayed highly surface active capabilities, including faster interfacial adsorption kinetics than standard native surfactant, even in the presence of inhibitory agents. ATII-LCs could constitute a highly useful ex vivo model for the study of surfactant biogenesis and the mechanisms involved in protein processing and lipid trafficking, as well as the packing and storage of surfactant complexes.
Collapse
Affiliation(s)
- Alejandro Cerrada
- Departmento de Bioquímica y Biología Molecular, Facultad de Biología, Universidad Complutense, Madrid, Spain
| | - Paz de la Torre
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Jesús Grande
- Departmento de Obstetricia y Ginecología, Hospital 12 de Octubre, Madrid, Spain
| | - Thomas Haller
- Department of Physiology, Innsbruck Medical University, Innsbruck, Austria
| | - Ana I. Flores
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Jesús Pérez-Gil
- Departmento de Bioquímica y Biología Molecular, Facultad de Biología, Universidad Complutense, Madrid, Spain
| |
Collapse
|
28
|
Lecht S, Gerstenhaber JA, Stabler CT, Pimton P, Karamil S, Marcinkiewicz C, Schulman ES, Lelkes PI. Heterogeneous Mixed-Lineage Differentiation of Mouse Embryonic Stem Cells Induced by Conditioned Media from A549 Cells. Stem Cells Dev 2014; 23:1923-36. [DOI: 10.1089/scd.2014.0042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Shimon Lecht
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Jonathan A. Gerstenhaber
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Collin T. Stabler
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Pimchanok Pimton
- Department of Biology, School of Science, Walailak University, Thammarat, Thailand
| | - Seda Karamil
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Edward S. Schulman
- Division of Pulmonary, Critical Care and Sleep Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Peter I. Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Garreta E, Melo E, Navajas D, Farré R. Low oxygen tension enhances the generation of lung progenitor cells from mouse embryonic and induced pluripotent stem cells. Physiol Rep 2014; 2:2/7/e12075. [PMID: 25347858 PMCID: PMC4187564 DOI: 10.14814/phy2.12075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Whole-organ decellularization technology has emerged as a new alternative for the fabrication of bioartificial lungs. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) are potentially useful for recellularization since they can be directed to express phenotypic marker genes of lung epithelial cells. Normal pulmonary development takes place in a low oxygen environment ranging from 1 to 5%. By contrast, in vitro ESC and iPSC differentiation protocols are usually carried out at room-air oxygen tension. Here, we sought to determine the role played by oxygen tension on the derivation of Nkx2.1+ lung/thyroid progenitor cells from mouse ESC and iPSC. A step-wise differentiation protocol was used to generate Nkx2.1+ lung/thyroid progenitors under 20% and 5% oxygen tension. On day 12, gene expression analysis revealed that Nkx2.1 and Foxa2 (endodermal and early lung epithelial cell marker) were significantly upregulated at 5% oxygen tension in ESC and iPSC differentiated cultures compared to 20% oxygen conditions. In addition, quantification of Foxa2+Nkx2.1+Pax8- cells corresponding to the lung field, with exclusion of the potential thyroid fate identified by Pax8 expression, confirmed that the low physiologic oxygen tension exerted a significant positive effect on early pulmonary differentiation of ESC and iPSC. In conclusion, we found that 5% oxygen tension enhanced the derivation of lung progenitors from mouse ESC and iPSC compared to 20% room-air oxygen tension.
Collapse
Affiliation(s)
- Elena Garreta
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain Centre de Medicina Regenerativa de Barcelona (CMRB), Parc de Recerca Biomèdica de Barcelona (PRBB), Dr. Aiguader88 7ª Planta, Barcelona, 08003, Spain
| | - Esther Melo
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain F. Hoffmann-La Roche, AG, NORD DTABldg. 69/331, Basel, CH-4070, Switzerland
| | - Daniel Navajas
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut de Bioenginyeria de Catalunya, Barcelona, Spain
| | - Ramon Farré
- Facultat de Medicina, Unitat de Biofísica i Bioenginyeria, Universitat de Barcelona, Barcelona, Spain CIBER de Enfermedades Respiratorias, Madrid, Spain Institut Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
Ghaedi M, Mendez JJ, Bove PF, Sivarapatna A, Raredon MSB, Niklason LE. Alveolar epithelial differentiation of human induced pluripotent stem cells in a rotating bioreactor. Biomaterials 2014; 35:699-710. [PMID: 24144903 PMCID: PMC3897000 DOI: 10.1016/j.biomaterials.2013.10.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/02/2013] [Indexed: 12/13/2022]
Abstract
Traditional stem cell differentiation protocols make use of a variety of cytokines including growth factors (GFs) and inhibitors in an effort to provide appropriate signals for tissue specific differentiation. In this study, iPSC-derived type II pneumocytes (iPSC-ATII) as well as native isolated human type II pneumocytes (hATII) were differentiated toward a type I phenotype using a unique air-liquid interface (ALI) system that relies on a rotating apparatus that mimics in vivo respiratory conditions. A relatively homogenous population of alveolar type II-like cells from iPSC was first generated (iPSC-ATII cells), which had phenotypic properties similar to mature human alveolar type II cells. iPSC-ATII cells were then cultured in a specially designed rotating culture apparatus. The effectiveness of the ALI bioreactor was compared with the effectiveness of small molecule-based differentiation of type II pneumocytes toward type 1 pneumocytes. The dynamics of differentiation were examined by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), flow cytometry and immunocytochemistry. iPSC-ATII and hATII cells cultured in the ALI bioreactor had higher levels of type I markers, including aquaporin-5(AQ5), caveolin-1, and T1α, at both the RNA and protein levels as compared with the flask-grown iPSC-ATII and hATII that had been treated with small molecules to induce differentiation. In summary, this study demonstrates that a rotating bioreactor culture system that provides an air-liquid interface is a potent inducer of type I epithelial differentiation for both iPS-ATII cells and hATII cells, and provides a method for large-scale production of alveolar epithelium for tissue engineering and drug discovery.
Collapse
Affiliation(s)
- Mahboobe Ghaedi
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Julio J. Mendez
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Peter F. Bove
- Cystic Fibrosis/Pulmonary Research Treatment Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Amogh Sivarapatna
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Micha Sam B. Raredon
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Laura E. Niklason
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
34
|
Hazeltine LB, Selekman JA, Palecek SP. Engineering the human pluripotent stem cell microenvironment to direct cell fate. Biotechnol Adv 2013; 31:1002-19. [PMID: 23510904 PMCID: PMC3758782 DOI: 10.1016/j.biotechadv.2013.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 02/20/2013] [Accepted: 03/11/2013] [Indexed: 01/31/2023]
Abstract
Human pluripotent stem cells (hPSCs), including both embryonic stem cells and induced pluripotent stem cells, offer a potential cell source for research, drug screening, and regenerative medicine applications due to their unique ability to self-renew or differentiate to any somatic cell type. Before the full potential of hPSCs can be realized, robust protocols must be developed to direct their fate. Cell fate decisions are based on components of the surrounding microenvironment, including soluble factors, substrate or extracellular matrix, cell-cell interactions, mechanical forces, and 2D or 3D architecture. Depending on their spatio-temporal context, these components can signal hPSCs to either self-renew or differentiate to cell types of the ectoderm, mesoderm, or endoderm. Researchers working at the interface of engineering and biology have identified various factors which can affect hPSC fate, often based on lessons from embryonic development, and they have utilized this information to design in vitro niches which can reproducibly direct hPSC fate. This review highlights culture systems that have been engineered to promote self-renewal or differentiation of hPSCs, with a focus on studies that have elucidated the contributions of specific microenvironmental cues in the context of those culture systems. We propose the use of microsystem technologies for high-throughput screening of spatial-temporal presentation of cues, as this has been demonstrated to be a powerful approach for differentiating hPSCs to desired cell types.
Collapse
Affiliation(s)
| | | | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison 1415 Engineering Drive, Madison, WI 53706 USA
| |
Collapse
|
35
|
Sun H, Quan Y, Yan Q, Peng X, Mao Z, Wetsel RA, Wang D. Isolation and characterization of alveolar epithelial type II cells derived from mouse embryonic stem cells. Tissue Eng Part C Methods 2013; 20:464-72. [PMID: 24102479 DOI: 10.1089/ten.tec.2013.0415] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The use of embryonic stem cells (ESCs) to regenerate distal lung epithelia damaged by injuries or diseases requires development of safe and efficient methodologies that direct ESC differentiation into transplantable distal lung epithelial progenitors. Time-consuming culture procedure and low differentiation efficiency are major problems that are associated with conventional differentiation approaches via embryoid body formation. The use of a growth factor cocktail or a lung-specific cell-conditioned medium to enrich definitive endoderm for efficient differentiation of mouse ESCs (mESC) into alveolar epithelial progenitor type II cells (ATIICs) has been reported, but not yet successful for generating a homogenous population of ATIICs for tissue regeneration purpose, and it remains unclear whether or not those mESC-derived ATIICs possess normal biological functions. Here, we report a novel method using a genetically modified mESC line harboring an ATIIC-specific neomycin(R) transgene in Rosa 26 locus. We showed that ATIICs can be efficiently differentiated from mESCs as early as day 7 by culturing them directly on Matrigel-coated plates in DMEM containing 15% knockout serum replacement. With this culture condition, the genetically modified mESCs can be selectively differentiated into a homogenous population (>99%) of ATIICs. Importantly, the mESC-derived ATIICs (mESC-ATIICs) exhibited typical lamellar bodies and expressed surfactant protein A, B, and C as normal control ATIICs. When cultured with an air-liquid-interface culture system in Small Airway Epithelial Cell Growth Medium, the mESC-ATIICs can be induced to secrete surfactant proteins after being treated with dibutyryl cAMP+dexamethasone. These mESC-ATIICs can synthesize and secrete surfactant lipid in response to secretagogue, demonstrating active surfactant metabolism in mESC-ATIICs as that seen in normal control ATIICs. In addition, we demonstrated that the selected mESC-ATIICs can be maintained on Matrigel-coated plates for at least 4 days with robust proliferative capacity. When cultured in DMEM medium containing 10% FBS, mESC-ATIICs spontaneously differentiated into alveolar epithelial type I cells. Collectively, these data demonstrate that the genetically modified mESCs can be selectively differentiated into a homogenous population of functional ATIICs, providing a reliable cell source to explore their therapeutic potential in lung tissue regeneration.
Collapse
Affiliation(s)
- Huanhuan Sun
- 1 Research Center for Immunology and Autoimmune Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Medical School at Houston , Houston, Texas
| | | | | | | | | | | | | |
Collapse
|
36
|
Ghaedi M, Calle EA, Mendez JJ, Gard AL, Balestrini J, Booth A, Bove PF, Gui L, White ES, Niklason LE. Human iPS cell-derived alveolar epithelium repopulates lung extracellular matrix. J Clin Invest 2013; 123:4950-62. [PMID: 24135142 DOI: 10.1172/jci68793] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 08/15/2013] [Indexed: 01/11/2023] Open
Abstract
The use of induced pluripotent stem cells (iPSCs) has been postulated to be the most effective strategy for developing patient-specific respiratory epithelial cells, which may be valuable for lung-related cell therapy and lung tissue engineering. We generated a relatively homogeneous population of alveolar epithelial type II (AETII) and type I (AETI) cells from human iPSCs that had phenotypic properties similar to those of mature human AETII and AETI cells. We used these cells to explore whether lung tissue can be regenerated in vitro. Consistent with an AETII phenotype, we found that up to 97% of cells were positive for surfactant protein C, 95% for mucin-1, 93% for surfactant protein B, and 89% for the epithelial marker CD54. Additionally, exposing induced AETII to a Wnt/β-catenin inhibitor (IWR-1) changed the iPSC-AETII-like phenotype to a predominantly AETI-like phenotype. We found that of induced AET1 cells, more than 90% were positive for type I markers, T1α, and caveolin-1. Acellular lung matrices were prepared from whole rat or human adult lungs treated with decellularization reagents, followed by seeding these matrices with alveolar cells derived from human iPSCs. Under appropriate culture conditions, these progenitor cells adhered to and proliferated within the 3D lung tissue scaffold and displayed markers of differentiated pulmonary epithelium.
Collapse
|
37
|
Long-term research of stem cells in monocrotaline-induced pulmonary arterial hypertension. Clin Exp Med 2013; 14:439-46. [DOI: 10.1007/s10238-013-0256-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 10/26/2022]
|
38
|
Wong AP, Rossant J. Generation of Lung Epithelium from Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2013; 1:137-145. [PMID: 23662247 PMCID: PMC3646155 DOI: 10.1007/s40139-013-0016-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The understanding of key processes and signaling mechanisms in lung development has been mainly demonstrated through gain and loss of function studies in mice, while human lung development remains largely unexplored due to inaccessibility. Several recent reports have exploited the identification of key signaling mechanisms that regulate lineage commitment and restriction in mouse lung development, to direct differentiation of both mouse and human pluripotent stem cells towards lung epithelial cells. In this review, we discuss the recent advances in the generation of respiratory epithelia from pluripotent stem cells and the potential of these engineered cells for novel scientific discoveries in lung diseases and future translation into regenerative therapies.
Collapse
Affiliation(s)
- Amy P. Wong
- Program in Developmental & Stem Cell Biology, Hospital for Sick Children, Toronto, ON M5G 1L7 Canada
| | - Janet Rossant
- Program in Developmental & Stem Cell Biology, Hospital for Sick Children, Toronto, ON M5G 1L7 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8 Canada
- Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8 Canada
| |
Collapse
|
39
|
Schmeckebier S, Mauritz C, Katsirntaki K, Sgodda M, Puppe V, Duerr J, Schubert SC, Schmiedl A, Lin Q, Paleček J, Draeger G, Ochs M, Zenke M, Cantz T, Mall MA, Martin U. Keratinocyte growth factor and dexamethasone plus elevated cAMP levels synergistically support pluripotent stem cell differentiation into alveolar epithelial type II cells. Tissue Eng Part A 2013; 19:938-51. [PMID: 23176317 DOI: 10.1089/ten.tea.2012.0066] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Alveolar epithelial type II (ATII)-like cells can be generated from murine embryonic stem cells (ESCs), although to date, no robust protocols applying specific differentiation factors are established. We hypothesized that the keratinocyte growth factor (KGF), an important mediator of lung organogenesis and primary ATII cell maturation and proliferation, together with dexamethasone, 8-bromoadenosine-cAMP, and isobutylmethylxanthine (DCI), which induce maturation of primary fetal ATII cells, also support the alveolar differentiation of murine ESCs. Here we demonstrate that the above stimuli synergistically potentiate the alveolar differentiation of ESCs as indicated by increased expression of the surfactant proteins (SP-) C and SP-B. This effect is most profound if KGF is supplied not only in the late stage, but at least also during the intermediate stage of differentiation. Our results indicate that KGF most likely does not enhance the generation of (mes)endodermal or NK2 homeobox 1 (Nkx2.1) expressing progenitor cells but rather, supported by DCI, accelerates further differentiation/maturation of respiratory progeny in the intermediate phase and maturation/proliferation of emerging ATII cells in the late stage of differentiation. Ultrastructural analyses confirmed the presence of ATII-like cells with intracellular composite and lamellar bodies. Finally, induced pluripotent stem cells (iPSCs) were generated from transgenic mice with ATII cell-specific lacZ reporter expression. Again, KGF and DCI synergistically increased SP-C and SP-B expression in iPSC cultures, and lacZ expressing ATII-like cells developed. In conclusion, ATII cell-specific reporter expression enabled the first reliable proof for the generation of murine iPSC-derived ATII cells. In addition, we have shown KGF and DCI to synergistically support the generation of ATII-like cells from ESCs and iPSCs. Combined application of these factors will facilitate more efficient generation of stem cell-derived ATII cells for future basic research and potential therapeutic application.
Collapse
Affiliation(s)
- Sabrina Schmeckebier
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Angelini DJ, Dorsey RM, Willis KL, Hong C, Moyer RA, Oyler J, Jensen NS, Salem H. Chemical warfare agent and biological toxin-induced pulmonary toxicity: could stem cells provide potential therapies? Inhal Toxicol 2013; 25:37-62. [DOI: 10.3109/08958378.2012.750406] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
41
|
Nichols JE, Niles JA, Cortiella J. Design and development of tissue engineered lung: Progress and challenges. Organogenesis 2012; 5:57-61. [PMID: 19794900 DOI: 10.4161/org.5.2.8564] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Accepted: 03/27/2009] [Indexed: 11/19/2022] Open
Abstract
Before we can realize our long term goal of engineering lung tissue worthy of clinical applications, advances in the identification and utilization of cell sources, development of standardized procedures for differentiation of cells, production of matrix tailored to meet the needs of the lung and design of methods or techniques of applying the engineered tissues into the injured lung environment will need to occur. Design of better biomaterials with the capacity to guide stem cell behavior and facilitate lung lineage choice as well as seamlessly integrate with living lung tissue will be achieved through advances in the development of decellularized matrices and new understandings related to the influence of extracellular matrix on cell behavior and function. We have strong hopes that recent developments in the engineering of conducting airway from decellularized trachea will lead to similar breakthroughs in the engineering of distal lung components in the future.
Collapse
|
42
|
Vosdoganes P, Lim R, Moss TJM, Wallace EM. Cell therapy: a novel treatment approach for bronchopulmonary dysplasia. Pediatrics 2012; 130:727-37. [PMID: 22945412 DOI: 10.1542/peds.2011-2576] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a major cause of substantial lifelong morbidity in preterm infants. Despite a better understanding of the pathophysiology of BPD and significant research effort into its management, there remains today no effective treatment. Cell-based therapy is a novel approach that offers much promise in the prevention and treatment of BPD. Recent research supports a therapeutic role for cell transplantation in the management of a variety of acute and chronic adult and childhood lung diseases, with potential of such therapy to reduce inflammation and prevent acute lung injury. However, considerable uncertainties remain regarding cell therapies before they can be established as safe and effective clinical treatments for BPD. This review summarizes the current literature investigating cell therapies in lung disease, with particular focus on the various types of cells available and their specific properties in the context of a future therapy for BPD.
Collapse
Affiliation(s)
- Patricia Vosdoganes
- Ritchie Centre, Department of Obstetrics and Gynaecology, Monash Medical Centre, 246 Clayton Rd, Clayton, VIC, Australia 3168
| | | | | | | |
Collapse
|
43
|
Notara M, Hernandez D, Mason C, Daniels JT. Characterization of the phenotype and functionality of corneal epithelial cells derived from mouse embryonic stem cells. Regen Med 2012; 7:167-78. [PMID: 22397607 DOI: 10.2217/rme.11.117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIMS To investigate the optimum conditions for the differentiation of a mouse embryonic stem cell line towards corneal epithelial cell fate. MATERIALS & METHODS The effect of conditioned media from both metabolically active (to produce lineage A) and growth-arrested limbal fibroblasts (lineage G) were compared with basal media (lineage N) in terms of morphology and marker expression, assessed by immunocytochemistry and reverse transcription PCR. Cultures were transplanted into a porcine ex vivo model to investigate their ability for wound healing and cornea repair. RESULTS Lineage N exhibited cobblestone morphology and expressed CK12 and p63α, while OCT4 and SSEA1 were downregulated. Post-transplantation, these cells were able to multilayer and heal after wounding while maintaining marker expression. CONCLUSION Lineages with corneal epithelial-like characteristics, which are derived from embryonic stem cells, have potential for use in the study of corneal wound healing and therapy.
Collapse
Affiliation(s)
- Maria Notara
- Department of Ocular Biology & Therapeutics, UCL Institute of Ophthalmology, London, UK.
| | | | | | | |
Collapse
|
44
|
CD166(pos) subpopulation from differentiated human ES and iPS cells support repair of acute lung injury. Mol Ther 2012; 20:2335-46. [PMID: 22968480 DOI: 10.1038/mt.2012.182] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Previous efforts to derive lung progenitor cells from human embryonic stem (hES) cells using embryoid body formation or stromal feeder cocultures had been limited by low efficiencies. Here, we report a step-wise differentiation method to drive both hES and induced pluripotent stem (iPS) cells toward the lung lineage. Our data demonstrated a 30% efficiency in generating lung epithelial cells (LECs) that expresses various distal lung markers. Further enrichment of lung progenitor cells using a stem cell marker, CD166 before transplantation into bleomycin-injured NOD/SCID mice resulted in enhanced survivability of mice and improved lung pulmonary functions. Immunohistochemistry of lung sections from surviving mice further confirmed the specific engraftment of transplanted cells in the damaged lung. These cells were shown to express surfactant protein C, a specific marker for distal lung progenitor in the alveoli. Our study has therefore demonstrated the proof-of-concept of using iPS cells for the repair of acute lung injury, demonstrating the potential usefulness of using patient's own iPS cells to prevent immune rejection which arise from allogenic transplantation.
Collapse
|
45
|
Bajpai VK, Andreadis ST. Stem cell sources for vascular tissue engineering and regeneration. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:405-25. [PMID: 22571595 DOI: 10.1089/ten.teb.2011.0264] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review focuses on the stem cell sources with the potential to be used in vascular tissue engineering and to promote vascular regeneration. The first clinical studies using tissue-engineered vascular grafts are already under way, supporting the potential of this technology in the treatment of cardiovascular and other diseases. Despite progress in engineering biomaterials with the appropriate mechanical properties and biological cues as well as bioreactors for generating the correct tissue microenvironment, the source of cells that make up the vascular tissues remains a major challenge for tissue engineers and physicians. Mature cells from the tissue of origin may be difficult to obtain and suffer from limited proliferative capacity, which may further decline as a function of donor age. On the other hand, multipotent and pluripotent stem cells have great potential to provide large numbers of autologous cells with a great differentiation capacity. Here, we discuss the adult multipotent as well as embryonic and induced pluripotent stem cells, their differentiation potential toward vascular lineages, and their use in engineering functional and implantable vascular tissues. We also discuss the associated challenges that need to be addressed in order to facilitate the transition of this technology from the bench to the bedside.
Collapse
Affiliation(s)
- Vivek K Bajpai
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, New York 14260-4200, USA
| | | |
Collapse
|
46
|
Bone marrow mesenchymal stem cells can differentiate into type II alveolar epithelial cells in vitro. Cell Biol Int 2012; 35:1261-6. [PMID: 21542803 DOI: 10.1042/cbi20110026] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, we demonstrate that BMSCs (bone marrow mesenchymal stem cells) can be successfully differentiated into type II alveolar epithelial cells in vitro under mimic pulmonary microenvironment. BMSCs were co-cultured with MRC-5 cells in modified SAGM (small airway growth medium). The BMSC-derived type II alveolar epithelial cells morphologically resemble human lung epithelial cells. They began to appear after 10 days in co-culture and became morphologically dominant after day 15. Correspondingly, SPC (surfactant protein C), a specific functional marker of human type II alveolar epithelial cells, was detected in differentiated cells by RT-PCR (reverse transcription-PCR) analysis after day 15. Immunostaining analysis revealed the present of scattered SPC-positive cells with a differentiation efficiency of 2.43-4.21%. Our study further showed that the SPC gene expression level in differentiated cells was related to the ratio of BMSCs to MRC-5 cells and the components of modified SAGM.
Collapse
|
47
|
Longmire TA, Ikonomou L, Hawkins F, Christodoulou C, Cao Y, Jean JC, Kwok LW, Mou H, Rajagopal J, Shen SS, Dowton AA, Serra M, Weiss DJ, Green MD, Snoeck HW, Ramirez MI, Kotton DN. Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell Stem Cell 2012; 10:398-411. [PMID: 22482505 PMCID: PMC3322392 DOI: 10.1016/j.stem.2012.01.019] [Citation(s) in RCA: 292] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 12/18/2011] [Accepted: 01/25/2012] [Indexed: 11/17/2022]
Abstract
Two populations of Nkx2-1(+) progenitors in the developing foregut endoderm give rise to the entire postnatal lung and thyroid epithelium, but little is known about these cells because they are difficult to isolate in a pure form. We demonstrate here the purification and directed differentiation of primordial lung and thyroid progenitors derived from mouse embryonic stem cells (ESCs). Inhibition of TGFβ and BMP signaling, followed by combinatorial stimulation of BMP and FGF signaling, can specify these cells efficiently from definitive endodermal precursors. When derived using Nkx2-1(GFP) knockin reporter ESCs, these progenitors can be purified for expansion in culture and have a transcriptome that overlaps with developing lung epithelium. Upon induction, they can express a broad repertoire of markers indicative of lung and thyroid lineages and can recellularize a 3D lung tissue scaffold. Thus, we have derived a pure population of progenitors able to recapitulate the developmental milestones of lung/thyroid development.
Collapse
Affiliation(s)
- Tyler A. Longmire
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Laertis Ikonomou
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Finn Hawkins
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Constantina Christodoulou
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Yuxia Cao
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - JC Jean
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Letty W. Kwok
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Hongmei Mou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA02114, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA02114, USA
| | - Steven S. Shen
- Section of Computational Biomedicine, and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USAw
- Center for Health Informatics and Bioinformatics, Department of Biochemistry and Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Anne A. Dowton
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Maria Serra
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Daniel J. Weiss
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, VT 05405
| | - Michael D. Green
- Mount Sinai School of Medicine, Department of Oncological Science, New York, NY 10029, USA
| | - Hans-Willem Snoeck
- Mount Sinai School of Medicine, Department of Oncological Science, New York, NY 10029, USA
| | - Maria I. Ramirez
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Darrell N. Kotton
- Boston University Pulmonary Center, Boston, Massachusetts 02118, USA
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
48
|
Siti-Ismail N, Samadikuchaksaraei A, Bishop AE, Polak JM, Mantalaris A. Development of a Novel Three-Dimensional, Automatable and Integrated Bioprocess for the Differentiation of Embryonic Stem Cells into Pulmonary Alveolar Cells in a Rotating Vessel Bioreactor System. Tissue Eng Part C Methods 2012; 18:263-72. [DOI: 10.1089/ten.tec.2011.0299] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Norhayati Siti-Ismail
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, United Kingdom
- Stem Cells and Regenerative Medicine, Department of Experimental Medicine and Toxicology, Imperial College London, London, United Kingdom
| | - Ali Samadikuchaksaraei
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Anne E. Bishop
- Stem Cells and Regenerative Medicine, Department of Experimental Medicine and Toxicology, Imperial College London, London, United Kingdom
| | - Julia M. Polak
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
49
|
Human embryonic stem cells differentiated to lung lineage-specific cells ameliorate pulmonary fibrosis in a xenograft transplant mouse model. PLoS One 2012; 7:e33165. [PMID: 22470441 PMCID: PMC3314647 DOI: 10.1371/journal.pone.0033165] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 02/10/2012] [Indexed: 11/19/2022] Open
Abstract
Background Our aim was to differentiate human (h) embryonic stem (ES) cells into lung epithelial lineage-specific cells [i.e., alveolar epithelial type I (AEI) and type II (AEII) cells and Clara cells] as the first step in the development of cell-based strategies to repair lung injury in the bleomycin mouse model of idiopathic pulmonary fibrosis (IPF). A heterogeneous population of non-ciliated lung lineage-specific cells was derived by a novel method of embryoid body (EB) differentiation. This differentiated human cell population was used to modulate the profibrotic phenotype in transplanted animals. Methodology and Principal Findings Omission or inclusion of one or more components in the differentiation medium skewed differentiation of H7 hES cells into varying proportions of AEI, AEII, and Clara cells. ICG-001, a small molecule inhibitor of Wnt/β-catenin/Creb-binding protein (CBP) transcription, changed marker expression of the differentiated ES cells from an AEII-like phenotype to a predominantly AEI-like phenotype. The differentiated cells were used in xenograft transplantation studies in bleomycin-treated Rag2γC−/− mice. Human cells were detected in lungs of the transplanted groups receiving differentiated ES cells treated with or without ICG-001. The increased lung collagen content found in bleomycin-treated mice receiving saline was significantly reduced by transplantation with the lung-lineage specific epithelial cells differentiated from ES cells. A significant increase in progenitor number was observed in the airways of bleomycin-treated mice after transplantation of differentiated hES cells. Conclusions This study indicates that ES cell-based therapy may be a powerful novel approach to ameliorate lung fibrosis.
Collapse
|
50
|
Novel interventional approaches for ALI/ARDS: cell-based gene therapy. Mediators Inflamm 2011; 2011:560194. [PMID: 21785528 PMCID: PMC3139183 DOI: 10.1155/2011/560194] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 05/09/2011] [Accepted: 05/22/2011] [Indexed: 12/21/2022] Open
Abstract
Acute lung injury (ALI) and its more severe
form, acute respiratory distress syndrome (ARDS),
continue to be a major cause of morbidity and
mortality in critically ill patients. The present
therapeutic strategies for ALI/ARDS including
supportive care, pharmacological treatments, and
ventilator support are still controversial. More
scientists are focusing on therapies involving
stem cells, which have self-renewing capabilities
and differentiate into multiple cell lineages,
and, genomics therapy which has the potential to
upregulate expression of anti-inflammatory
mediators. Recently, the combination of cell and
gene therapy which has been demonstrated to
provide additive benefit has opened up a new
chapter in therapeutic strategy and provides a
basis for the development of an innovative
approach for the prevention and treatment of
ALI/ARDS.
Collapse
|