1
|
Liu S, Cao H, Wang Z, Zhu J, An X, Zhang L, Song Y. Single-cell transcriptomics reveals extracellular matrix remodeling and collagen dynamics during lactation in sheep mammary gland. Int J Biol Macromol 2025; 312:143669. [PMID: 40319976 DOI: 10.1016/j.ijbiomac.2025.143669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland is a dynamic organ with diverse cell populations that maintain glandular homeostasis, particularly during lactation. However, the cellular architecture and molecular mechanisms underlying lactational remodeling in the sheep mammary gland remain incompletely understood. Given similarities in mammary stromal structure, sheep serve as a valuable model for studying lactational changes relevant to the human breast, which experiences collagen loss and sagging during lactation. Utilizing single-cell transcriptomics (scRNA-seq), we mapped the sheep mammary gland's cellular landscape at postpartum days 60 and 150, identifying seven major cell types, including six distinct epithelial clusters. These clusters revealed differentiation among luminal progenitors, hormone-sensing, and myoepithelial cells across peak and late lactation stages. Transcriptomic analysis highlighted pivotal roles for epithelial integrity and ECM remodeling, with myoepithelial cells centrally involved in these processes. We observed significant collagen remodeling driven by fibroblast-epithelial crosstalk and ECM reorganization during late lactation. Comparative analysis with human mammary epithelial cells showed conserved basal and myoepithelial cell populations, while luminal cells diverged across species. This study provides insights into lactation biology and ECM remodeling, offering a framework to inform future studies on lactational adaptation and its implications for human health.
Collapse
Affiliation(s)
- Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhanhang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
2
|
Khadim A, Kiliaris G, Vazquez-Armendariz AI, Procida-Kowalski T, Glaser D, Bartkuhn M, Malik T, Chu X, Moiseenko A, Kuznetsova I, Ahmadvand N, Lingampally A, Hadzic S, Alexopoulos I, Chen Y, Günther A, Behr J, Neumann J, Schiller HB, Li X, Weissmann N, Braun T, Seeger W, Wygrecka M, Morty RE, Herold S, El Agha E. Myofibroblasts emerge during alveolar regeneration following influenza-virus-induced lung injury. Cell Rep 2025; 44:115248. [PMID: 39903667 DOI: 10.1016/j.celrep.2025.115248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/24/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025] Open
Abstract
Alveolar regeneration requires the coordinated engagement of epithelial stem cells and mesenchymal niche cells to restore the intricate alveolar architecture of the lung. The current paradigm is that certain aspects of lung organogenesis are mimicked during injury repair in the adult stage. Here, we employ a longitudinal single-cell transcriptomic survey to fate map lung mesenchymal cells throughout development and adulthood. We show that myofibroblasts that are reminiscent of developmental alveolar myofibroblasts (AMFs), termed AMF-like cells, are activated during alveolar regeneration following influenza-virus-induced lung injury. Although AMF-like cells share a similar transcriptomic signature with myofibroblasts that are associated with aberrant repair and fibrosis, these cells do not derive from fibroblast growth factor 10-positive alveolar fibroblasts, and their dysregulation is associated with failed alveolar regeneration in humans. Our data emphasize the role played by developmental mechanisms in alveolar regeneration and highlight the context-dependent nature of myofibroblast biology and function during injury repair.
Collapse
Affiliation(s)
- Ali Khadim
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Georgios Kiliaris
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ana Ivonne Vazquez-Armendariz
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; University of Bonn, Transdisciplinary Research Area Life and Health, Organoid Biology, Life & Medical Sciences Institute, 53115 Bonn, Germany
| | - Tara Procida-Kowalski
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - David Glaser
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Tanya Malik
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Xuran Chu
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Alena Moiseenko
- Immunology and Respiratory Department, Boehringer Ingelheim Pharma GmbH, Biberach 88400, Germany
| | - Irina Kuznetsova
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Negah Ahmadvand
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Arun Lingampally
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Stefan Hadzic
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ioannis Alexopoulos
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Yuexin Chen
- Research Unit Precision Regenerative Medicine (PRM), Comprehensive Pneumology Center (CPC), German Center for Lung Research (DZL), 81377 Munich, Germany; Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Andreas Günther
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Jürgen Behr
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Jens Neumann
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center, German Center for Lung Research (DZL), 80336 Munich, Germany
| | - Herbert B Schiller
- Research Unit Precision Regenerative Medicine (PRM), Comprehensive Pneumology Center (CPC), German Center for Lung Research (DZL), 81377 Munich, Germany; Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, 61231 Bad Nauheim, Germany
| | - Werner Seeger
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, 61231 Bad Nauheim, Germany
| | - Malgorzata Wygrecka
- Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Department of Medicine II, Internal Medicine, Pulmonary and Critical Care, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, Heidelberg University Hospital, 69117 Heidelberg, Germany
| | - Susanne Herold
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Elie El Agha
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus Liebig University Giessen, 35392 Giessen, Germany; Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany; Institute for Lung Health (ILH), 35392 Giessen, Germany; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
3
|
Ferrini E, Stellari FF, Franceschi V, Macchi F, Russo L, Murgia A, Grisendi G, Villetti G, Dominici M, Donofrio G. Persistency of Mesenchymal Stromal/Stem Cells in Lungs. Front Cell Dev Biol 2021; 9:709225. [PMID: 34336863 PMCID: PMC8322774 DOI: 10.3389/fcell.2021.709225] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 01/03/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are a fibroblast-like cell population with high regenerative potential that can be isolated from many different tissues. Several data suggest MSCs as a therapeutic tool capable of migrating to a site of injury and guide tissue regeneration mainly through their secretome. Pulmonary first-pass effect occurs during intravenous administration of MSCs, where 50 to 80% of the cells tend to localize in the lungs. This phenomenon has been exploited to study MSC potential therapeutic effects in several preclinical models of lung diseases. Data demonstrated that, regardless of the lung disease severity and the delivery route, MSCs were not able to survive longer than 24 h in the respiratory tract but still surprisingly determined a therapeutic effect. In this work, two different mouse bone marrow-derived mesenchymal stromal/stem cell (mBM-MSC) lines, stably transduced with a third-generation lentiviral vector expressing luciferase and green fluorescent protein reporter genes tracking MSCs in vivo biodistribution and persistency, have been generated. Cells within the engrafted lung were in vivo traced using the high-throughput bioluminescence imaging (BLI) technique, with no invasiveness on animal, minimizing biological variations and costs. In vivo BLI analysis allowed the detection and monitoring of the mBM-MSC clones up to 28 days after implantation independently from the delivery route. This longer persistency than previously observed (24 h) could have a strong impact in terms of pharmacokinetics and pharmacodynamics of MSCs as a therapeutic tool.
Collapse
Affiliation(s)
- Erica Ferrini
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy.,Department of Veterinary Science, University of Parma, Parma, Italy
| | | | | | - Francesca Macchi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Luca Russo
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Alba Murgia
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Hospital of Modena, Modena, Italy.,Scientific and Technological Park of Medicine "Mario Veronesi," Mirandola, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Hospital of Modena, Modena, Italy
| | - Gino Villetti
- Chiesi Farmaceutici S.p.A., Corporate Pre-Clinical R&D, Parma, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Hospital of Modena, Modena, Italy.,Scientific and Technological Park of Medicine "Mario Veronesi," Mirandola, Italy
| | - Gaetano Donofrio
- Department of Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
4
|
Mitsialis SA, Kourembanas S. Stem cell-based therapies for the newborn lung and brain: Possibilities and challenges. Semin Perinatol 2016; 40:138-51. [PMID: 26778234 PMCID: PMC4808378 DOI: 10.1053/j.semperi.2015.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There have been substantial advances in neonatal medical care over the past 2 decades that have resulted in the increased survival of very low birth weight infants, survival that in some centers extends to 22 weeks gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term pulmonary and neurologic consequences. No single therapy has proven to be effective in preventing or treating either developmental lung and brain injuries in preterm infants or the hypoxic-ischemic injury that can be inflicted on the full-term brain as a result of in utero or perinatal complications. Stem cell-based therapies are emerging as a potential paradigm-shifting approach for such complex diseases with multifactorial etiologies, but a great deal of work is still required to understand the role of stem/progenitor cells in normal development and in the repair of injured tissue. This review will summarize the biology of the various stem/progenitor cells, their effects on tissue repair in experimental models of lung and brain injury, the recent advances in our understanding of their mechanism of action, and the challenges that remain to be addressed before their eventual application to clinical care.
Collapse
|
5
|
Gui L, Qian H, Rocco KA, Grecu L, Niklason LE. Efficient intratracheal delivery of airway epithelial cells in mice and pigs. Am J Physiol Lung Cell Mol Physiol 2015; 308:L221-8. [PMID: 25416381 DOI: 10.1152/ajplung.00147.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cellular therapy via direct intratracheal delivery has gained interest as a novel therapeutic strategy for treating various pulmonary diseases including cystic fibrosis lung disease. However, concerns such as insufficient cell engraftment in lungs and lack of large animal model data remain to be resolved. This study aimed to establish a simple method for evaluating cell retention in lungs and to develop reproducible approaches for efficient cell delivery into mouse and pig lungs. Human lung epithelial cells including normal human bronchial/tracheal epithelial (NHBE) cells and human lung epithelial cell line A549 were infected with pSicoR-green fluorescent protein (GFP) lentivirus. GFP-labeled NHBE cells were delivered via a modified intratracheal cell instillation method into the lungs of C57BL/6J mice. Two days following cell delivery, GFP ELISA-based assay revealed a substantial cell-retention efficiency (10.48 ± 2.86%, n = 7) in mouse lungs preinjured with 2% polidocanol. When GFP-labeled A549 cells were transplanted into Yorkshire pig lungs with a tracheal intubation fiberscope, a robust initial cell attachment (22.32% efficiency) was observed at 24 h. In addition, a lentiviral vector was developed to induce the overexpression and apical localization of cystic fibrosis transmembrane conductance regulator (CFTR)-GFP fusion proteins in NHBE cells as a means of ex vivo CFTR gene transfer in nonprogenitor (relatively differentiated) lung epithelial cells. These results have demonstrated the convenience and efficiency of direct delivery of exogenous epithelial cells to lungs in mouse and pig models and provided important background for future preclinical evaluation of intratracheal cell transplantation to treat lung diseases.
Collapse
|
6
|
Sdrimas K, Kourembanas S. MSC microvesicles for the treatment of lung disease: a new paradigm for cell-free therapy. Antioxid Redox Signal 2014; 21:1905-15. [PMID: 24382303 PMCID: PMC4202925 DOI: 10.1089/ars.2013.5784] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Bronchopulmonary dysplasia (BPD), also known as chronic lung disease of infancy, is a major complication of preterm birth that, despite improvements in neonatal respiratory support and perinatal care, remains an important cause of morbidity and mortality, often with severe adverse neurodevelopmental sequelae. Even with major advances in our understanding of the pathogenesis of this disease, BPD remains essentially without adequate treatment. RECENT ADVANCES Cell-based therapies arose as a promising treatment for acute and chronic lung injury in many experimental models of disease. Currently, more than 3000 human clinical trials employing cell therapy for the treatment of diverse diseases, including cardiac, neurologic, immune, and respiratory conditions, are ongoing or completed. Among the treatments, mesenchymal stem cells (MSCs) are the most studied and have been extensively tested in experimental models of BPD, pulmonary hypertension, pulmonary fibrosis, and acute lung injury. CRITICAL ISSUES Despite the promising potential, MSC therapy for human lung disease still remains at an experimental stage and optimal transplantation conditions need to be determined. Although the mechanism of MSC action can be manifold, accumulating evidence suggests a predominant paracrine, immunomodulatory, and cytoprotective effect. FUTURE DIRECTIONS The current review summarizes the effect of MSC treatment in models of lung injury, including BPD, and focuses on the MSC secretome and, specifically, MSC-derived microvesicles as potential key mediators of therapeutic action that can be the focus of future therapies.
Collapse
Affiliation(s)
- Konstantinos Sdrimas
- 1 Division of Newborn Medicine, Boston Children's Hospital , Boston, Massachusetts
| | | |
Collapse
|
7
|
Stessuk T, Ruiz MA, Greco OT, Bilaqui A, Ribeiro-Paes MJDO, Ribeiro-Paes JT. Phase I clinical trial of cell therapy in patients with advanced chronic obstructive pulmonary disease: follow-up of up to 3 years. Rev Bras Hematol Hemoter 2013; 35:352-7. [PMID: 24255620 PMCID: PMC3832317 DOI: 10.5581/1516-8484.20130113] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/26/2013] [Indexed: 01/16/2023] Open
Abstract
Background Chronic obstructive pulmonary disease is a major inflammatory disease of the
airways and an enormous therapeutic challenge. Within the spectrum of chronic
obstructive pulmonary disease, pulmonary emphysema is characterized by the
destruction of the alveolar walls with an increase in the air spaces distal to the
terminal bronchioles but without significant pulmonary fibrosis. Therapeutic
options are limited and palliative since they are unable to promote morphological
and functional regeneration of the alveolar tissue. In this context, new
therapeutic approaches, such as cell therapy with adult stem cells, are being
evaluated. Objective This article aims to describe the follow-up of up to 3 years after the beginning
of a phase I clinical trial and discuss the spirometry parameters achieved by
patients with advanced pulmonary emphysema treated with bone marrow mononuclear
cells. Methods Four patients with advanced pulmonary emphysema were submitted to autologous
infusion of bone marrow mononuclear cells. Follow-ups were performed by spirometry
up to 3 years after the procedure. Results The results showed that autologous cell therapy in patients having chronic
obstructive pulmonary disease is a safe procedure and free of adverse effects.
There was an improvement in laboratory parameters (spirometry) and a slowing down
in the process of pathological degeneration. Also, patients reported improvements
in the clinical condition and quality of life. Conclusions Despite being in the initial stage and in spite of the small sample, the results
of the clinical protocol of cell therapy in advanced pulmonary emphysema as
proposed in this study, open new therapeutic perspectives in chronic obstructive
pulmonary disease. It is worth emphasizing that this study corresponds to the
first study in the literature that reports a change in the natural history of
pulmonary emphysema after the use of cell therapy with a pool of bone marrow
mononuclear cells.
Collapse
|
8
|
Cell therapy with bone marrow mononuclear cells in elastase-induced pulmonary emphysema. Stem Cell Rev Rep 2013; 9:210-8. [PMID: 23242964 DOI: 10.1007/s12015-012-9419-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Emphysema is characterized by destruction of alveolar walls with loss of gas exchange surface and consequent progressive dyspnea. This study aimed to evaluate the efficiency of cell therapy with bone marrow mononuclear cells (BMMC) in an animal model of elastase-induced pulmonary emphysema. Emphysema was induced in C57Bl/J6 female mice by intranasal instillation of elastase. After 21 days, the mice received bone marrow mononuclear cells from EGFP male mice with C57Bl/J6 background. The groups were assessed by comparison and statistically significant differences (p < 0.05) were observed among the groups treated with BMMC and evaluated after 7, 14 and 21 days. Analysis of the mean linear intercept (Lm) values for the different groups allowed to observe that the group treated with BMMC and evaluated after 21 days showed the most significant result. The group that received no treatment showed a statistically significant difference when compared to other groups, except the group treated and evaluated after 21 days, evidencing the efficacy of cell therapy with BMMC in pulmonary emphysema.
Collapse
|
9
|
Weiss DJ, Bertoncello I, Borok Z, Kim C, Panoskaltsis-Mortari A, Reynolds S, Rojas M, Stripp B, Warburton D, Prockop DJ. Stem cells and cell therapies in lung biology and lung diseases. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2011; 8:223-72. [PMID: 21653527 PMCID: PMC3132784 DOI: 10.1513/pats.201012-071dw] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/03/2011] [Indexed: 11/20/2022]
Abstract
The University of Vermont College of Medicine and the Vermont Lung Center, with support of the National Heart, Lung, and Blood Institute (NHLBI), the Alpha-1 Foundation, the American Thoracic Society, the Emory Center for Respiratory Health,the Lymphangioleiomyomatosis (LAM) Treatment Alliance,and the Pulmonary Fibrosis Foundation, convened a workshop,‘‘Stem Cells and Cell Therapies in Lung Biology and Lung Diseases,’’ held July 26-29, 2009 at the University of Vermont,to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of the mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases.
Collapse
Affiliation(s)
- Daniel J Weiss
- Vermont Lung Center, University of Vermont College of Medicine, Burlington, Vermont 05405, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Leblond AL, Naud P, Forest V, Gourden C, Sagan C, Romefort B, Mathieu E, Delorme B, Collin C, Pagès JC, Sensebé L, Pitard B, Lemarchand P. Developing cell therapy techniques for respiratory disease: intratracheal delivery of genetically engineered stem cells in a murine model of airway injury. Hum Gene Ther 2010; 20:1329-43. [PMID: 19606934 DOI: 10.1089/hum.2009.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Interest has increased in the use of exogenous stem cells to optimize lung repair and serve as carriers of a therapeutic gene for genetic airway diseases such as cystic fibrosis. We investigated the survival and engraftment of exogenous stem cells after intratracheal injection, in a murine model of acute epithelial airway injury already used in gene therapy experiments on cystic fibrosis. Embryonic stem cells and mesenchymal stem cells were intratracheally injected 24 hr after 2% polidocanol administration, when epithelial airway injury was maximal. Stem cells were transfected with reporter genes immediately before administration. Reporter gene expression was analyzed in trachea-lungs and bronchoalveolar lavage, using nonfluorescence, quantitative, and sensitive methods. Enzyme-linked immunosorbent assay quantitative results showed that 0.4 to 5.5% of stem cells survived in the injured airway. Importantly, no stem cells survived in healthy airway or in the epithelial lining fluid. Using 5-bromo-4-chloro-3-indolyl-beta-d-galactopyranoside staining, transduced mesenchymal stem cells were detected in injured trachea and bronchi lumen. When the epithelium was spontaneously regenerated, the in vivo amount of engrafted mesenchymal stem cells from cell lines decreased dramatically. No stem cells from primary culture were located within the lungs at 7 days. This study demonstrated the feasibility of intratracheal cell delivery for airway diseases with acute epithelial injury.
Collapse
|
11
|
Sueblinvong V, Weiss DJ. Stem cells and cell therapy approaches in lung biology and diseases. Transl Res 2010; 156:188-205. [PMID: 20801416 PMCID: PMC4201367 DOI: 10.1016/j.trsl.2010.06.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/14/2010] [Accepted: 06/16/2010] [Indexed: 12/19/2022]
Abstract
Cell-based therapies with embryonic or adult stem cells, including induced pluripotent stem cells, have emerged as potential novel approaches for several devastating and otherwise incurable lung diseases, including emphysema, pulmonary fibrosis, pulmonary hypertension, and the acute respiratory distress syndrome. Although initial studies suggested engraftment of exogenously administered stem cells in lung, this is now generally felt to be a rare occurrence of uncertain physiologic significance. However, more recent studies have demonstrated paracrine effects of administered cells, including stimulation of angiogenesis and modulation of local inflammatory and immune responses in mouse lung disease models. Based on these studies and on safety and initial efficacy data from trials of adult stem cells in other diseases, groundbreaking clinical trials of cell-based therapy have been initiated for pulmonary hypertension and for chronic obstructive pulmonary disease. In parallel, the identity and role of endogenous lung progenitor cells in development and in repair from injury and potential contribution as lung cancer stem cells continue to be elucidated. Most recently, novel bioengineering approaches have been applied to develop functional lung tissue ex vivo. Advances in each of these areas will be described in this review with particular reference to animal models.
Collapse
Key Words
- aec, alveolar epithelial cell
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- basc, bronchioalveolar stem cell
- ccsp, clara cell secretory protein
- cf, cystic fibrosis
- cftr, cystic fibrosis transmembrane conductance regulator
- clp, cecal ligation and puncture
- copd, chronic obstructive pulmonary disease
- enos, endothelial nitric oxide synthetase
- epc, endothelial progenitor cell
- esc, embryonic stem cell
- fev1, forced expiratory volume in 1 second
- fvc, forced vital capacity
- gfp, green fluorescent protein
- hsc, hematopoietic stem cell
- ipf, idiopathic pulmonary fibrosis
- kgf, keratinocyte growth factor
- lps, lipopolysaccharide
- mct, monocrotaline
- mhc, major histocompatibility complex
- msc, mesenchymal stromal (stem) cell
- ph, pulmonary hypertension
- pro-spc, pro-surfactant protein c
- sca-1, stem cell antigen-1
Collapse
Affiliation(s)
- Viranuj Sueblinvong
- Division of Pulmonary, Critical Care and Allergy, Department of Medicine, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
12
|
Roszell B, Seaton A, Fong GH, Finck CM. Cell-based therapy improves cell viability and increases airway size in an explant model. Exp Lung Res 2010; 35:501-13. [PMID: 19842834 DOI: 10.1080/01902140902718353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cell-based therapy is a promising treatment option for lung disease, but no studies have demonstrated its benefit in promoting perinatal lung growth. Embryonic day 18 (E18) fetal lungs treated with vascular inhibitors were grown as explant organ cultures to inhibit endothelial growth in the explant cultures. Disruption of pulmonary vasculature decreased explant mean cord length and viability, whereas coculture with fetal pulmonary or predifferentiated embryonic stem cells rescued both parameters. These results demonstrate in a model of perinatal lung growth, exogenous addition of fetal pulmonary cells or differentiated embryonic stem (ES) cells promotes survival and alveolar morphogenesis. These experiments represent the first evidence of the benefits of cell-based therapy for perinatal lung growth.
Collapse
Affiliation(s)
- Blair Roszell
- Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | | | | |
Collapse
|
13
|
Roszell B, Mondrinos MJ, Seaton A, Simons DM, Koutzaki SH, Fong GH, Lelkes PI, Finck CM. Efficient derivation of alveolar type II cells from embryonic stem cells for in vivo application. Tissue Eng Part A 2009; 15:3351-65. [PMID: 19388834 PMCID: PMC2811058 DOI: 10.1089/ten.tea.2008.0664] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 04/23/2009] [Indexed: 11/12/2022] Open
Abstract
In the present study, mouse embryonic stem cells (ESCs) were differentiated into alveolar epithelial type II (AEII) cells for endotracheal injection. These enriched lung-like populations expressed lung epithelial markers SP-A, SP-B, SP-C, and CC10. First we show that rapid differentiation of ESCs requires a dissociated seeding method instead of an embryoid body culture method. We then investigated a two-step differentiation of ESCs into definitive endoderm by activin or A549-conditioned medium as a precursor to lung epithelial cells. When conditioned medium from A549 cells was used to derive endoderm, yield was increased above that of activin alone. Further studies showed that Wnt3a may be one of the secreted factors produced by A549 cells and promotes definitive endoderm differentiation, in part, through suppression of primitive endoderm. Activin and Wnt3a together at appropriate doses with dissociated cell seeding promoted greater endoderm yield than activin alone. Next, fibroblast growth factor 2 was shown to induce a dose-dependent expression of SPC, and these cells contained lamellar bodies characteristic of mature AEII cells from ESC-derived endoderm. Finally, ES-derived lung cells were endotracheally injected into preterm mice with evidence of AEII distribution within the lung parenchyma. This study concludes that a recapitulation of development may enhance derivation of an enriched population of lung-like cells for use in cell-based therapy.
Collapse
Affiliation(s)
- Blair Roszell
- School of Biomedical Engineering, Sciences, and Health Systems, Drexel University, Philadelphia, Pennsylvania
- University of Connecticut Health Center, Farmington, Connecticut
| | - Mark J. Mondrinos
- School of Biomedical Engineering, Sciences, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Ariel Seaton
- University of Connecticut Health Center, Farmington, Connecticut
| | - Donald M. Simons
- School of Biomedical Engineering, Sciences, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Sirma H. Koutzaki
- School of Biomedical Engineering, Sciences, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Guo-Hua Fong
- University of Connecticut Health Center, Farmington, Connecticut
| | - Peter I. Lelkes
- School of Biomedical Engineering, Sciences, and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Christine M. Finck
- University of Connecticut Health Center, Farmington, Connecticut
- Connecticut Children's Medical Center, Hartford, Connecticut
| |
Collapse
|
14
|
van Haaften T, Byrne R, Bonnet S, Rochefort GY, Akabutu J, Bouchentouf M, Rey-Parra GJ, Galipeau J, Haromy A, Eaton F, Chen M, Hashimoto K, Abley D, Korbutt G, Archer SL, Thébaud B. Airway delivery of mesenchymal stem cells prevents arrested alveolar growth in neonatal lung injury in rats. Am J Respir Crit Care Med 2009; 180:1131-42. [PMID: 19713449 DOI: 10.1164/rccm.200902-0179oc] [Citation(s) in RCA: 350] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE Bronchopulmonary dysplasia (BPD) and emphysema are characterized by arrested alveolar development or loss of alveoli; both are significant global health problems and currently lack effective therapy. Bone marrow-derived mesenchymal stem cells (BMSCs) prevent adult lung injury, but their therapeutic potential in neonatal lung disease is unknown. OBJECTIVES We hypothesized that intratracheal delivery of BMSCs would prevent alveolar destruction in experimental BPD. METHODS In vitro, BMSC differentiation and migration were assessed using co-culture assays and a modified Boyden chamber. In vivo, the therapeutic potential of BMSCs was assessed in a chronic hyperoxia-induced model of BPD in newborn rats. MEASUREMENTS AND MAIN RESULTS In vitro, BMSCs developed immunophenotypic and ultrastructural characteristics of type II alveolar epithelial cells (AEC2) (surfactant protein C expression and lamellar bodies) when co-cultured with lung tissue, but not with culture medium alone or liver. Migration assays revealed preferential attraction of BMSCs toward oxygen-damaged lung versus normal lung. In vivo, chronic hyperoxia in newborn rats led to air space enlargement and loss of lung capillaries, and this was associated with a decrease in circulating and resident lung BMSCs. Intratracheal delivery of BMSCs on Postnatal Day 4 improved survival and exercise tolerance while attenuating alveolar and lung vascular injury and pulmonary hypertension. Engrafted BMSCs coexpressed the AEC2-specific marker surfactant protein C. However, engraftment was disproportionately low for cell replacement to account for the therapeutic benefit, suggesting a paracrine-mediated mechanism. In vitro, BMSC-derived conditioned medium prevented O(2)-induced AEC2 apoptosis, accelerated AEC2 wound healing, and enhanced endothelial cord formation. CONCLUSIONS BMSCs prevent arrested alveolar and vascular growth in part through paracrine activity. Stem cell-based therapies may offer new therapeutic avenues for lung diseases that currently lack efficient treatments.
Collapse
Affiliation(s)
- Timothy van Haaften
- Department of Pediatrics, Women and Children Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Xin H, Sun R, Kanehira M, Takahata T, Itoh J, Mizuguchi H, Saijo Y. Intratracheal delivery of CX3CL1-expressing mesenchymal stem cells to multiple lung tumors. Mol Med 2009; 15:321-7. [PMID: 19603106 DOI: 10.2119/molmed.2009.00059] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Accepted: 06/16/2009] [Indexed: 12/16/2022] Open
Abstract
The lung is one of the organs to which cancers from solid tumors frequently metastasize. Multiple tumors in the lung are usually treated by systemic chemotherapy because of the lack of efficient methods of targeting antitumor agents to the lung. Although intratracheal administration is an ideal route for targeting multiple lung tumors, antitumor agents are often harmful to the organ or induce inflammation. Mesenchymal stem cells (MSCs), nonhematopoietic stem cells capable of differentiating into various mesoderm-type cells, have a propensity to migrate to and proliferate in tumor tissues after systemic administration. We intratracheally injected MSCs expressing CX3CL1 (MSC/RGDFKN) into the lung of lung tumor-bearing mice with multiple metastases of C26 or Lewis lung carcinoma (LLC). Antitumor effects were evaluated by counting the number of lung metastases and survival. We demonstrated the tropism of mouse MSCs to lung tumor tissues after intratracheal administration of GFP-positive MSCs. Intratracheal injection of MSC/RGDFKN strongly inhibited growth of lung metastases of C26 or LLC, and thus prolonged survival. Intratracheal injection of MSC/RGDFKN did not induce an inflammatory reaction in the lung. These results suggest that MSCs expressing antitumor agents can be delivered intratracheally into multiple lung tumor tissues without causing inflammation.
Collapse
Affiliation(s)
- Hong Xin
- Department of Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Stem cells and cell therapies in lung biology and lung diseases. Ann Am Thorac Soc 2008; 5:637-67. [PMID: 18625757 DOI: 10.1513/pats.200804-037dw] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
17
|
Fritzell JA, Mao Q, Gundavarapu S, Pasquariello T, Aliotta JM, Ayala A, Padbury JF, De Paepe ME. Fate and effects of adult bone marrow cells in lungs of normoxic and hyperoxic newborn mice. Am J Respir Cell Mol Biol 2008; 40:575-87. [PMID: 18988921 DOI: 10.1165/rcmb.2008-0176oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cell-based therapy in adult lung injury models is associated with highly variable donor cell engraftment and epithelial reconstitution. The role of marrow-derived cell therapy in neonatal lung injury is largely unknown. In this study, we determined the fate and effects of adult bone marrow cells in a model of neonatal lung injury. Wild-type mice placed in a normoxic or hyperoxic (95% O(2)) environment received bone marrow cells from animals expressing green fluorescent protein (GFP) at Postnatal Day (P)5. Controls received vehicle buffer. Lungs were analyzed between Post-Transplantation (TPX) Day 2 and Week 8. The volume of GFP-immunoreactive donor cells, monitored by stereologic volumetry, remained constant between Post-TPX Weeks 1 and 8 and was similar in normoxic and hyperoxia-exposed recipients. Virtually all marrow-derived cells showed colocalization of GFP and the pan-macrophage marker, F4/80, by double immunofluorescence studies. Epithelial transdifferentiation was not seen. Marrow cell administration had adverse effects on somatic growth and alveolarization in normoxic mice, while no effects were discerned in hyperoxia-exposed recipients. Reexposure of marrow-treated animals to hyperoxia at P66 resulted in significant expansion of the donor-derived macrophage population. In conclusion, intranasal administration of unfractionated bone marrow cells to newborn mice does not achieve epithelial reconstitution, but establishes persistent alveolar macrophage chimerism. The predominantly adverse effects of marrow treatment in newborn lungs are likely due to macrophage-associated paracrine effects. While this model and route of cell therapy may not achieve epithelial reconstitution, the role of selected stem cell populations and/or alternate routes of administration for cell-based therapy in injured newborn lungs deserve further investigation.
Collapse
Affiliation(s)
- James A Fritzell
- Women and Infants Hospital, Dept. of Pathology, 101 Dudley Street, Providence, RI 02905, USA
| | | | | | | | | | | | | | | |
Collapse
|