1
|
Chen P, Van Hassel J, Pinezich MR, Diane M, Hudock MR, Kaslow SR, Gavaudan OP, Fung K, Kain ML, Lopez H, Saqi A, Guenthart BA, Hozain AE, Romanov A, Bacchetta M, Vunjak-Novakovic G. Recovery of extracorporeal lungs using cross-circulation with injured recipient swine. J Thorac Cardiovasc Surg 2024; 167:e106-e130. [PMID: 37741314 PMCID: PMC10954590 DOI: 10.1016/j.jtcvs.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVE Lung transplantation remains limited by the shortage of healthy organs. Cross-circulation with a healthy swine recipient provides a durable physiologic environment to recover injured donor lungs. In a clinical application, a recipient awaiting lung transplantation could be placed on cross-circulation to recover damaged donor lungs, enabling eventual transplantation. Our objective was to assess the ability of recipient swine with respiratory compromise to tolerate cross-circulation and support recovery of donor lungs subjected to extended cold ischemia. METHODS Swine donor lungs (n = 6) were stored at 4 °C for 24 hours while recipient swine (n = 6) underwent gastric aspiration injury before cross-circulation. Longitudinal multiscale analyses (blood gas, bronchoscopy, radiography, histopathology, cytokine quantification) were performed to evaluate recipient swine and extracorporeal lungs on cross-circulation. RESULTS Recipient swine lung injury resulted in sustained, impaired oxygenation (arterial oxygen tension/inspired oxygen fraction ratio 205 ± 39 mm Hg vs 454 ± 111 mm Hg at baseline). Radiographic, bronchoscopic, and histologic assessments demonstrated bilateral infiltrates, airway cytokine elevation, and significantly worsened lung injury scores. Recipient swine provided sufficient metabolic support for extracorporeal lungs to demonstrate robust functional improvement (0 hours, arterial oxygen tension/inspired oxygen fraction ratio 138 ± 28.2 mm Hg; 24 hours, 539 ± 156 mm Hg). Multiscale analyses demonstrated improved gross appearance, aeration, and cellular regeneration in extracorporeal lungs by 24 hours. CONCLUSIONS We demonstrate that acutely injured recipient swine tolerate cross-circulation and enable recovery of donor lungs subjected to extended cold storage. This proof-of-concept study supports feasibility of cross-circulation for recipients with isolated lung disease who are candidates for this clinical application.
Collapse
Affiliation(s)
- Panpan Chen
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | - Julie Van Hassel
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | - Meghan R Pinezich
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Mohamed Diane
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Maria R Hudock
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Sarah R Kaslow
- Department of Biomedical Engineering, Columbia University, New York, NY; Department of Surgery, Columbia University Medical Center, New York, NY
| | | | - Kenmond Fung
- Clinical Perfusion, Columbia University Medical Center, New York, NY
| | - Mandy L Kain
- Institute of Comparative Medicine, Columbia University, New York, NY
| | - Hermogenes Lopez
- Clinical Perfusion, Columbia University Medical Center, New York, NY
| | - Anjali Saqi
- Pathology and Cell Biology, Columbia University Medical Center, New York, NY
| | - Brandon A Guenthart
- Department of Cardiothoracic Surgery, Stanford University Medical Center, Stanford, Calif
| | - Ahmed E Hozain
- Department of Surgery, Columbia University Medical Center, New York, NY
| | - Alexander Romanov
- Institute of Comparative Medicine, Columbia University, New York, NY
| | - Matthew Bacchetta
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tenn; Department of Biomedical Engineering, Vanderbilt University, Nashville, Tenn
| | | |
Collapse
|
2
|
Kim SY, McTeague D, Cheong SS, Hind M, Dean CH. Deciphering the impacts of modulating the Wnt-planar cell polarity (PCP) pathway on alveolar repair. Front Cell Dev Biol 2024; 12:1349312. [PMID: 38476262 PMCID: PMC10927798 DOI: 10.3389/fcell.2024.1349312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Many adult lung diseases involve dysregulated lung repair. Deciphering the molecular and cellular mechanisms that govern intrinsic lung repair is essential to develop new treatments to repair/regenerate the lungs. Aberrant Wnt signalling is associated with lung diseases including emphysema, idiopathic pulmonary fibrosis and pulmonary arterial hypertension but how Wnt signalling contributes to these diseases is still unclear. There are several alternative pathways that can be stimulated upon Wnt ligand binding, one of these is the Planar Cell Polarity (PCP) pathway which induces actin cytoskeleton remodelling. Wnt5a is known to stimulate the PCP pathway and this ligand is of particular interest in regenerative lung biology because of its association with lung diseases and its role in the alveolar stem cell niche. To decipher the cellular mechanisms through which Wnt5a and the PCP pathway affect alveolar repair we utilised a 3-D ex-vivo model of lung injury and repair, the AIR model. Our results show that Wnt5a specifically enhances the alveolar epithelial progenitor cell population following injury and surprisingly, this function is attenuated but not abolished in Looptail (Lp) mouse lungs in which the PCP pathway is dysfunctional. However, Lp tracheal epithelial cells show reduced stiffness and Lp alveolar epithelial cells are less migratory than wildtype (WT), indicating that Lp lung epithelial cells have a reduced capacity for repair. These findings provide important mechanistic insight into how Wnt5a and the PCP pathway contribute to lung repair and indicate that these components of Wnt signalling may be viable targets for the development of pro-repair treatments.
Collapse
Affiliation(s)
- Sally Yunsun Kim
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David McTeague
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Sek-Shir Cheong
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hind
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Charlotte H. Dean
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Mishra S, Gandhi D, Tiwari RR, Rajasekaran S. Beneficial role of kaempferol and its derivatives from different plant sources on respiratory diseases in experimental models. Inflammopharmacology 2023; 31:2311-2336. [PMID: 37410224 DOI: 10.1007/s10787-023-01282-1] [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: 01/19/2023] [Accepted: 06/02/2023] [Indexed: 07/07/2023]
Abstract
Respiratory illnesses impose a significant health burden and cause deaths worldwide. Despite many advanced strategies to improve patient outcomes, they are often less effective. There is still considerable room for improvement in the treatment of various respiratory diseases. In recent years, alternative medicinal agents derived from food plants have shown better beneficial effects against a wide variety of disease models, including cancer. In this regard, kaempferol (KMF) and its derivatives are the most commonly found dietary flavonols. They have been found to exhibit protective effects on multiple chronic diseases like diabetes, fibrosis, and so on. A few recent articles have reviewed the pharmacological actions of KMF in cancer, central nervous system diseases, and chronic inflammatory diseases. However, there is no comprehensive review that exists regarding the beneficial effects of KMF and its derivatives on both malignant- and non-malignant respiratory diseases. Many experimental studies reveal that KMF and its derivatives are helpful in managing a wide range of respiratory diseases, including acute lung injury, fibrosis, asthma, cancer, and chronic obstructive pulmonary disease, and their underlying molecular mechanisms. In addition, we also discussed the chemistry and sources, the absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties, methods to enhance bioavailability, as well as our perspective on future research with KMF and its derivatives.
Collapse
Affiliation(s)
- Sehal Mishra
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Deepa Gandhi
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India.
| |
Collapse
|
4
|
Sagar S, Faizan MI, Chaudhary N, Singh V, Singh P, Gheware A, Sharma K, Azmi I, Singh VP, Kharya G, Mabalirajan U, Agrawal A, Ahmad T, Sinha Roy S. Obesity impairs cardiolipin-dependent mitophagy and therapeutic intercellular mitochondrial transfer ability of mesenchymal stem cells. Cell Death Dis 2023; 14:324. [PMID: 37173333 PMCID: PMC10181927 DOI: 10.1038/s41419-023-05810-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
Mesenchymal stem cell (MSC) transplantation alleviates metabolic defects in diseased recipient cells by intercellular mitochondrial transport (IMT). However, the effect of host metabolic conditions on IMT and thereby on the therapeutic efficacy of MSCs has largely remained unexplored. Here we found impaired mitophagy, and reduced IMT in MSCs derived from high-fat diet (HFD)-induced obese mouse (MSC-Ob). MSC-Ob failed to sequester their damaged mitochondria into LC3-dependent autophagosomes due to decrease in mitochondrial cardiolipin content, which we propose as a putative mitophagy receptor for LC3 in MSCs. Functionally, MSC-Ob exhibited diminished potential to rescue mitochondrial dysfunction and cell death in stress-induced airway epithelial cells. Pharmacological modulation of MSCs enhanced cardiolipin-dependent mitophagy and restored their IMT ability to airway epithelial cells. Therapeutically, these modulated MSCs attenuated features of allergic airway inflammation (AAI) in two independent mouse models by restoring healthy IMT. However, unmodulated MSC-Ob failed to do so. Notably, in human (h)MSCs, induced metabolic stress associated impaired cardiolipin-dependent mitophagy was restored upon pharmacological modulation. In summary, we have provided the first comprehensive molecular understanding of impaired mitophagy in obese-derived MSCs and highlight the importance of pharmacological modulation of these cells for therapeutic intervention. A MSCs obtained from (HFD)-induced obese mice (MSC-Ob) show underlying mitochondrial dysfunction with a concomitant decrease in cardiolipin content. These changes prevent LC3-cardiolipin interaction, thereby reducing dysfunctional mitochondria sequestration into LC3-autophagosomes and thus impaired mitophagy. The impaired mitophagy is associated with reduced intercellular mitochondrial transport (IMT) via tunneling nanotubes (TNTs) between MSC-Ob and epithelial cells in co-culture or in vivo. B Pyrroloquinoline quinone (PQQ) modulation in MSC-Ob restores mitochondrial health, cardiolipin content, and thereby sequestration of depolarized mitochondria into the autophagosomes to alleviate impaired mitophagy. Concomitantly, MSC-Ob shows restoration of mitochondrial health upon PQQ treatment (MSC-ObPQQ). During co-culture with epithelial cells or transplantation in vivo into the mice lungs, MSC-ObPQQ restores IMT and prevents epithelial cell death. C Upon transplantation in two independent allergic airway inflammatory mouse models, MSC-Ob failed to rescue the airway inflammation, hyperactivity, metabolic changes in epithelial cells. D PQQ modulated MSCs restored these metabolic defects and restored lung physiology and airway remodeling parameters.
Collapse
Affiliation(s)
- Shakti Sagar
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Md Imam Faizan
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Nisha Chaudhary
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Vandana Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Atish Gheware
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Khushboo Sharma
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Iqbal Azmi
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Vijay Pal Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Gaurav Kharya
- Center for Bone Marrow Transplantation & Cellular Therapy Indraprastha Apollo Hospital, New Delhi, 110076, India
| | | | - Anurag Agrawal
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tanveer Ahmad
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India.
| | - Soumya Sinha Roy
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
5
|
Barzaghini B, Carelli S, Messa L, Rey F, Avanzini MA, Jacchetti E, Maghraby E, Berardo C, Zuccotti G, Raimondi MT, Cereda C, Calcaterra V, Pelizzo G. Bone Marrow Mesenchymal Stem Cells Expanded Inside the Nichoid Micro-Scaffold: a Focus on Anti-Inflammatory Response. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2023:1-12. [PMID: 37363698 PMCID: PMC10027280 DOI: 10.1007/s40883-023-00296-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/08/2023] [Accepted: 02/19/2023] [Indexed: 03/28/2023]
Abstract
Purpose Mesenchymal stem cells (MSCs) represent a promising source for stem cell therapies in numerous diseases, including pediatric respiratory system diseases. Characterized by low immunogenicity, high anti-inflammatory, and immunoregulatory features, MSCs demonstrated an excellent therapeutic profile in numerous in vitro and preclinical models. MSCs reside in a specialized physiologic microenvironment, characterized by a unique combination of biophysical, biochemical, and cellular properties. The exploitation of the 3D micro-scaffold Nichoid, which simulates the native niche, enhanced the anti-inflammatory potential of stem cells through mechanical stimulation only, overcoming the limitation of biochemical and xenogenic growth factors application. Materials and Methods In this work, we expanded pediatric bone marrow MSCs (BM-MSCs) inside the Nichoid and performed a complete cellular characterization with different approaches including viability assays, immunofluorescence analyses, RNA sequencing, and gene expression analysis. Results We demonstrated that BM-MSCs inside the scaffold remain in a stem cell quiescent state mimicking the condition of the in vivo environment. Moreover, the gene expression profile of these cells shows a significant up-regulation of genes involved in immune response when compared with the flat control. Conclusion The significant changes in the expression profile of anti-inflammatory genes could potentiate the therapeutic effect of BM-MSCs, encouraging the possible clinical translation for the treatment of pediatric congenital and acquired pulmonary disorders, including post-COVID lung manifestations. Lay Summary Regenerative medicine is the research field integrating medicine, biology, and biomedical engineering. In this context, stem cells, which are a fundamental cell source able to regenerate tissues and restore damage in the body, are the key component for a regenerative therapeutic approach. When expanded outside the body, stem cells tend to differentiate spontaneously and lose regenerative potential due to external stimuli. For this reason, we exploit the scaffold named Nichoid, which mimics the in vivo cell niche architecture. In this scaffold, mesenchymal stem cells "feel at home" due to the three-dimensional mechanical stimuli, and our findings could be considered as an innovative culture system for the in vitro expansion of stem cells for clinical translation. Future Perspective The increasing demand of safe and effective cell therapies projects our findings toward the possibility of improving cell therapies based on the use of BM-MSCs, particularly for their clinical translation in lung diseases. Graphical Abstract
Collapse
Affiliation(s)
- Bianca Barzaghini
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta,” Politecnico Di Milano, Milan, Italy
| | - Stephana Carelli
- Pediatric Research Center “Romeo Ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
| | - Letizia Messa
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
- Department of Electronic, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Federica Rey
- Pediatric Research Center “Romeo Ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
| | - Maria Antonietta Avanzini
- Immunology and Transplantation Laboratory, Cell Factory, Pediatric Hematology Oncology, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta,” Politecnico Di Milano, Milan, Italy
| | - Erika Maghraby
- Pediatric Research Center “Romeo Ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Clarissa Berardo
- Pediatric Research Center “Romeo Ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Research Center “Romeo Ed Enrica Invernizzi,” Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta,” Politecnico Di Milano, Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
| | - Valeria Calcaterra
- Department of Pediatrics, Buzzi Children’s Hospital, Milan, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Unit, Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Adamič N, Vengust M. Regenerative medicine in lung diseases: A systematic review. Front Vet Sci 2023; 10:1115708. [PMID: 36733636 PMCID: PMC9887049 DOI: 10.3389/fvets.2023.1115708] [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: 12/04/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
Regenerative medicine has opened the door to the exploration of new therapeutic methods for the treatment of various diseases, especially those associated with local or general disregulation of the immune system. In pulmonary diseases, new therapeutic strategies have emerged that are aimed at restoring functional lung tissue rather than alleviating symptoms. These strategies focus on tissue regeneration using stem cells and/or their derivatives or replacement of dysfunctional tissue using biomedical engineering. Animal health can directly benefit from regenerative therapy strategies and also serve as a translational experimental model for human disease. Several clinical trials have been conducted to evaluate the effects of cellular treatment on inflammatory lung disease in animals. Data reported to date show several beneficial effects in ex vivo and in vivo models; however, our understanding of the mechanisms that regenerative therapies exert on diseased tissues remains incomplete.
Collapse
|
7
|
Huang QY, Le Y, Hu H, Wan ZJ, Ning J, Han JL. Experimental research on surface acoustic wave microfluidic atomization for drug delivery. Sci Rep 2022; 12:7930. [PMID: 35562384 PMCID: PMC9106708 DOI: 10.1038/s41598-022-11132-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/14/2022] [Indexed: 11/09/2022] Open
Abstract
This paper demonstrates that surface acoustic wave (SAW) atomization can produce suitable aerosol concentration and size distribution for efficient inhaled lung drug delivery and is a potential atomization device for asthma treatment. Using the SAW device, we present comprehensive experimental results exploring the complexity of the acoustic atomization process and the influence of input power, device frequency, and liquid flow rate on aerosol size distribution. It is hoped that these studies will explain the mechanism of SAW atomization aerosol generation and how they can be controlled. The insights from the high-speed flow visualization studies reveal that it is possible by setting the input power above 4.17 W, thus allowing atomization to occur from a relatively thin film, forming dense, monodisperse aerosols. Moreover, we found that the aerosol droplet size can be effectively changed by adjusting the input power and liquid flow rate to change the film conditions. In this work, we proposed a method to realize drug atomization by a microfluidic channel. A SU-8 flow channel was prepared on the surface of a piezoelectric substrate by photolithography technology. Combined with the silicon dioxide coating process and PDMS process closed microfluidic channel was prepared, and continuous drug atomization was provided to improve the deposition efficiency of drug atomization by microfluidic.
Collapse
Affiliation(s)
- Qing-Yun Huang
- School of Mechanical Engineering and Automation, Harbin Institute of Technology Shenzhen, Guangdong, 518055, China.,Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Ying Le
- Department of Endocrinology and Metabolism, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Hong Hu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology Shenzhen, Guangdong, 518055, China.
| | | | - Jia Ning
- School of Mechanical Engineering and Automation, Harbin Institute of Technology Shenzhen, Guangdong, 518055, China
| | - Jun-Long Han
- School of Mechanical Engineering and Automation, Harbin Institute of Technology Shenzhen, Guangdong, 518055, China
| |
Collapse
|
8
|
Hurskainen M, Cyr-Depauw C, Thébaud B. Insights into the mechanisms of alveolarization - Implications for lung regeneration and cell therapies. Semin Fetal Neonatal Med 2022; 27:101243. [PMID: 33962890 DOI: 10.1016/j.siny.2021.101243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although the lung has extensive regenerative capacity, some diseases affecting the distal lung result in irreversible loss of pulmonary alveoli. Hitherto, treatments are supportive and do not specifically target tissue repair. Regenerative medicine offers prospects to promote lung repair and regeneration. The neonatal lung may be particularly receptive, because of its growth potential, compared to the adult lung. Based on our current understanding of neonatal lung injury, the ideal therapeutic approach includes mitigation of inflammation and fibrosis, and induction of regenerative signals. Cell-based therapies have shown potential to prevent and reverse impaired lung development. Their mechanisms of action suggest effects on both, mitigating the pathophysiological processes and promoting lung growth. Here, we review our current understanding of normal and impaired alveolarization, provide some rationale for the use of cell-based therapies and summarize current evidence for the therapeutic potential of cell-based therapies for pulmonary regeneration in preterm infants.
Collapse
Affiliation(s)
- Maria Hurskainen
- Division of Pediatric Cardiology, New Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Pediatric Research Center, New Children's Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| | - Chanèle Cyr-Depauw
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada; Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
9
|
Damianos A, Xu K, Kalin GT, Kalinichenko VV. Placental tissue stem cells and their role in neonatal diseases. Semin Fetal Neonatal Med 2022; 27:101322. [PMID: 34953760 DOI: 10.1016/j.siny.2021.101322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neonatal diseases such as hypoxic ischemic encephalopathy, diseases of prematurity and congenital disorders carry increased morbidity and mortality. Despite technological advancements, their incidence remains largely unabated. Stem cell (SC) interventions are novel therapies in the neonatal world. In pre-clinical models of neonatal diseases, SC applications have shown encouraging results. SC sources vary, with the bone marrow being the most utilized. However, the ability to harvest bone marrow SCs from neonates is limited. Placental-tissue derived SCs (PTSCs), provide an alternative and highly attractive source. Human placentas, the cornerstone of fetal survival, are abundant with such cells. Comparing to adult pools, PTSCs exhibit increased potency, decreased immunogenicity and stronger anti-inflammatory effects. Several types of PTSCs have been identified, with mesenchymal stem cells being the most utilized population. This review will focus on PTSCs and their pre-clinical and clinical applications in neonatology.
Collapse
Affiliation(s)
- Andreas Damianos
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Kui Xu
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gregory T Kalin
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Feng CY, Bai SY, Li ML, Zhao JY, Sun JM, Bao HJ, Ren Y, Su XM. Adipose-Derived Mesenchymal Stem Cell-Derived Exosomal miR-301a-3p Regulates Airway Smooth Muscle Cells During Asthma by Targeting STAT3. J Asthma Allergy 2022; 15:99-110. [PMID: 35115789 PMCID: PMC8806053 DOI: 10.2147/jaa.s335680] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/13/2021] [Indexed: 11/24/2022] Open
Abstract
Background Asthma is a chronic inflammatory disease featured by inflammation and remodeling of airway. Adipose-derived mesenchymal stem cell (ADSCs)-derived exosomal miRNAs have been suggested as promising therapeutic manners for diseases. Methods ADSCs and airway smooth muscle cells (ASMCs) were isolated from SD rats. Flow cytometry was conducted to detect the surface biomarkers of isolated cells. Exosomes were extracted by sequentially centrifuge method and identified by Western blotting and nanoparticle tracking analysis (NTA). Uptake of exosomes by ASMCs was detected by confocal assay. ASMCs were treated with platelet-derived growth factor-BB (PDGF-BB) to mimic cell remodeling and inflammation. Cell counting 8 (CCK-8), Transwell, and flow cytometry were performed to determine the viability, migration, and apoptosis of ASMCs. Release of inflammatory factors was detected by enzyme-linked immunosorbent assay (ELISA). Levels of RNAs and proteins were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. Interaction between miR-301a-3p and signal transducer and activator of transcription 3 (STAT3) was determined by luciferase reporter gene assay. The effect of Exosomal miR-301a-3p was analyzed in ovalbumin (OVA)-induced asthma mouse model. Results ADSCs-derived exosomes could be effectively internalized by ASMCs. Exosomal miR-301a-3p notably suppressed the PDGF-BB-stimulated proliferation and migration of ASMCs, and enhanced apoptosis, as well as decreased the secretion of inflammatory factors. MiR-301a-3p directly targeted the 3ʹUTR region of STAT3. STAT3 overexpression reversed the suppressive effects of exosomal miR-301a-3p on ASMCs under PDGF-BB stimulation. The expression of miR-301a-3p and STAT3 was negative correlation in specimen from patients with asthma. Exosomal miR-301a-3p inhibited OVA-induced lung injury by targeting STAT3 in mice. Conclusion This study exposed that exosomal miR-301a-3p from ADSCs could effectively alleviate PDGF-BB-stimulated remodeling and inflammation of ASMCs via targeting STAT3, presented ADSCs-derived exosomal miR-301a-3p as a promising therapeutic approach for asthma.
Collapse
Affiliation(s)
- Chen-Ye Feng
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Shi-Yao Bai
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Meng-Lu Li
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Jie-Yu Zhao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Jia-Min Sun
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Hui-Jing Bao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yuan Ren
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Xin-Ming Su
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, Shenyang, Liaoning, People’s Republic of China
- Correspondence: Xin-Ming Su Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, The First Affiliated Hospital of China Medical University; Respiratory Disease Institution of China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, People’s Republic of China Email
| |
Collapse
|
11
|
Rangasamy T, Ghimire L, Jin L, Le J, Periasamy S, Paudel S, Cai S, Jeyaseelan S. Host Defense against Klebsiella pneumoniae Pneumonia Is Augmented by Lung-Derived Mesenchymal Stem Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:1112-1127. [PMID: 34341173 DOI: 10.4049/jimmunol.2000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Klebsiella pneumoniae is a common cause of Gram-negative pneumonia. The spread of antibiotic-resistant and hypervirulent strains has made treatment more challenging. This study sought to determine the immunomodulatory, antibacterial, and therapeutic potential of purified murine stem cell Ag-1+ (Sca-1+) lung mesenchymal stem cells (LMSCs) using in vitro cell culture and an in vivo mouse model of pneumonia caused by K pneumoniae. Sca-1+ LMSCs are plastic adherent, possess colony-forming capacity, express mesenchymal stem cell markers, differentiate into osteogenic and adipogenic lineages in vitro, and exhibit a high proliferative capacity. Further, these Sca-1+ LMSCs are morphologically similar to fibroblasts but differ ultrastructurally. Moreover, Sca-1+ LMSCs have the capacity to inhibit LPS-induced secretion of inflammatory cytokines by bone marrow-derived macrophages and neutrophils in vitro. Sca-1+ LMSCs inhibit the growth of K pneumoniae more potently than do neutrophils. Sca-1+ LMSCs also possess the intrinsic ability to phagocytize and kill K. pneumoniae intracellularly. Whereas the induction of autophagy promotes bacterial replication, inhibition of autophagy enhances the intracellular clearance of K. pneumoniae in Sca-1+ LMSCs during the early time of infection. Adoptive transfer of Sca-1+ LMSCs in K. pneumoniae-infected mice improved survival, reduced inflammatory cells in bronchoalveolar lavage fluid, reduced inflammatory cytokine levels and pathological lesions in the lung, and enhanced bacterial clearance in the lung and in extrapulmonary organs. To our knowledge, these results together illustrate for the first time the protective role of LMSCs in bacterial pneumonia.
Collapse
Affiliation(s)
- Tirumalai Rangasamy
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Laxman Ghimire
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Liliang Jin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - John Le
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sivakumar Periasamy
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Sagar Paudel
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Shanshan Cai
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and
| | - Samithamby Jeyaseelan
- Center for Lung Biology and Disease, Louisiana State University, Baton Rouge, LA; .,Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA; and.,Division of Pulmonary and Critical Care, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
12
|
Yang YK, Li Y, Wang YY, Ruan GP, Tian C, Wang Q, He HY, Zhu GH, Fang D, Wang M, Zhu XQ, Pan XH. The effects of BMMSC treatment on lung tissue degeneration in elderly macaques. Stem Cell Res Ther 2021; 12:156. [PMID: 33648583 PMCID: PMC7923486 DOI: 10.1186/s13287-021-02201-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Age-associated lung tissue degeneration is a risk factor for lung injury and exacerbated lung disease. It is also the main risk factor for chronic lung diseases (such as COPD, idiopathic pulmonary fibrosis, cancer, among others). So, it is particularly important to find new anti-aging treatments. METHODS We systematically screened and evaluated elderly senile multiple organ dysfunction macaque models to determine whether BMMSCs inhibited lung tissue degeneration. RESULTS The average alveolar area, mean linear intercept (MLI), and fibrosis area in the elderly macaque models were significantly larger than in young rhesus monkeys (p < 0.05), while the capillary density around the alveoli was significantly low than in young macaque models (p < 0.05). Intravenous infusion of BMMSCs reduced the degree of pulmonary fibrosis, increased the density of capillaries around the alveoli (p < 0.05), and the number of type II alveolar epithelium in elderly macaques (p < 0.05). In addition, the infusion reduced lung tissue ROS levels, systemic and lung tissue inflammatory levels, and Treg cell ratio in elderly macaque models (p < 0.05). Indirect co-cultivation revealed that BMMSCs suppressed the expression of senescence-associated genes, ROS levels, apoptosis rate of aging type II alveolar epithelial cells (A549 cells), and enhanced their proliferation (p < 0.05). CONCLUSIONS BMMSC treatment inhibited age-associated lung tissue degeneration.
Collapse
Affiliation(s)
- Yu-Kun Yang
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan Province, China
| | - Ye Li
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan Province, China
| | - Yan-Ying Wang
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
| | - Guang-Ping Ruan
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
| | - Chuan Tian
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
| | - Qiang Wang
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China
| | - Huan-Yu He
- Kunming Medical University, Kunming, Yunnan Province, China
| | - Gao-Hong Zhu
- Department of Nuclear Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Dong Fang
- Department of Nuclear Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Mao Wang
- Department of Nuclear Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xiang-Qing Zhu
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China.
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China.
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China.
- Kunming Medical University, Kunming, Yunnan Province, China.
| | - Xing-Hua Pan
- Kunming Key Laboratory of Stem Cell and Regenerative Medicine, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China.
- Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, 920th Hospital of the PLA Joint Logistics Support Force, Kunming, 650032, Yunnan Province, China.
- Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming, Yunnan Province, China.
- Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
13
|
Li L, Dong L, Zhang J, Gao F, Hui J, Yan J. Mesenchymal stem cells with downregulated Hippo signaling attenuate lung injury in mice with lipopolysaccharide‑induced acute respiratory distress syndrome. Int J Mol Med 2018; 43:1241-1252. [PMID: 30628652 PMCID: PMC6365074 DOI: 10.3892/ijmm.2018.4047] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/27/2018] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cell (MSC)-mediated repair of injured alveolar epithelial cells is a promising potential cure for acute respiratory distress syndrome (ARDS); however, the repairing effect of MSCs is limited by poor homing and differentiation. Our previous study revealed that the inhibition of the Hippo signaling pathway promotes the proliferation, migration and differentiation of MSCs in vitro, leading to the hypothesis that MSCs with downregulated Hippo signaling could further ameliorate lipopolysaccharide (LPS)-induced ARDS in vivo. In the current study, mouse bone marrow-derived MSCs (mMSCs) with downregulated Hippo signaling were constructed by shRNA-mediated knockdown of large tumor suppressor kinase 1 (Lats1) and were intratracheally administered to LPS-induced mouse models of ARDS. The inhibition of Hippo signaling increased the retention of mMSC in ARDS lung tissue and their differentiation toward alveolar type II epithelial cells. Furthermore, mMSCs with downregulated Hippo signaling led to a decreased lung wet weight/body weight ratio, decreased total protein and albumin concentrations in bronchoalveolar lavage fluid, decreased levels of proinflammatory factors and increased levels of anti-inflammatory factors. Finally, mMSCs with downregulated Hippo signaling improved pathological changes and decreased pulmonary fibrosis in lungs of mice with ARDS. These results suggest that the inhibition of the Hippo signaling pathway in mouse mMSCs by knockdown of Lats1 could further improve the protective effects of mMSCs against epithelial damage and the therapeutic potential of mMSCs on mice with ARDS.
Collapse
Affiliation(s)
- Lang Li
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Liang Dong
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jiangqian Zhang
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Fei Gao
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jiaojie Hui
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jie Yan
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
14
|
Conese M, Beccia E, Carbone A, Castellani S, Di Gioia S, Corti F, Angiolillo A, Colombo C. The role of stem cells in cystic fibrosis disease modeling and drug discovery. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1549480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Massimo Conese
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elisa Beccia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Chronobiology Unit, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo (FG), Italy
| | - Stefano Castellani
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Fabiola Corti
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Carla Colombo
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
15
|
López-Giraldo A, Cruz T, Molins L, Guirao Á, Saco A, Cuerpo S, Ramirez J, Agustí Á, Faner R. Characterization, localization and comparison of c-Kit+ lung cells in never smokers and smokers with and without COPD. BMC Pulm Med 2018; 18:123. [PMID: 30064386 PMCID: PMC6066937 DOI: 10.1186/s12890-018-0688-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/10/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND c-Kit + lung stem cells have been described in the human healthy lung. Their potential relation with smoking and/or chronic obstructive pulmonary disease (COPD) is unknown. METHODS We characterized and compared c-Kit+ cells in lung tissue of 12 never smokers (NS), 15 smokers with normal spirometry (S) and 44 COPD patients who required lung resectional surgery. Flow cytometry (FACS) was used to characterize c-Kit+ cells in fresh lung tissue disaggregates, and immunofluorescence (IF) for further characterization and to determine their location in OCT- embedded lung tissue. RESULTS We identified 4 c-Kit+ cell populations, with similar proportions in NS, S and COPD: (1) By FACS, c-Kithigh/CD45+ cells (4.03 ± 2.97% (NS), 3.96 ± 5.30% (S), and 5.20 ± 3.44% (COPD)). By IF, these cells were tryptase+ (hence, mast cells) and located around the airways; (2) By IF, c-Kitlow/CD45+/triptase- (0.07 ± 0.06 (NS), 0.03 ± 0.02 (S), and 0.06 ± 0.07 (COPD) cells/field), which likely correspond to innate lymphoid cells; (3) By FACS, c-Kitlow/CD45-/CD34+ (0.95 ± 0.84% (NS), 1.14 ± 0.94% (S) and 0.95 ± 1.38% (COPD)). By IF these cells were c-Kitlow/CD45-/CD31+, suggesting an endothelial lineage, and were predominantly located in the alveolar wall; and, (4) by FACS, an infrequent c-Kitlow/CD45-/CD34- population (0.09 ± 0.14% (NS), 0.08 ± 0.09% (S) and 0.08 ± 0.11% (COPD)) compatible with a putative lung stem cell population. Yet, IF failed to detect them and we could not isolate or grow them, thus questioning the existence of c-Kit+ lung stem-cells. CONCLUSIONS The adult human lung contains a mixture of c-Kit+ cells, unlikely to be lung stem cells, which are independent of smoking status and/or presence of COPD.
Collapse
Affiliation(s)
- Alejandra López-Giraldo
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Tamara Cruz
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Laureano Molins
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ángela Guirao
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Adela Saco
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Sandra Cuerpo
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Ramirez
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Álvar Agustí
- Respiratory Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Faner
- Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. .,CIBER Enfermedades Respiratorias(CIBERES), Instituto de Salud Carlos III, Madrid, Spain. .,, Barcelona, Spain.
| |
Collapse
|
16
|
Esmaili Gourvarchin Galeh H, Meysam Abtahi Froushani S, Afzale Ahangaran N, Hadai SN. Effects of Educated Monocytes with Xenogeneic Mesenchymal Stem Cell-Derived Conditioned Medium in a Mouse Model of Chronic Asthma. Immunol Invest 2018; 47:504-520. [PMID: 29671652 DOI: 10.1080/08820139.2018.1458108] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND This study was conducted to determine the potential of the monocytes educated with rat bone marrow mesenchymal stem cell-derived conditioned medium (MCM) in ameliorating animal model of asthma. METHODS Chronic asthma was induced in the BALB/c mice using ovalbumin (OVA) sensitization. The monocytes were isolated from blood of mice and supplemented with 50% MCM or negative control media. After 24 h, the cells were designated as "non-educated or educated". Fourteen weeks after the onset of the study, animals were treated with educated or non-educated monocytes twice with a 1-week interval. RESULTS The educated monocytes showed a reduction in the potential production of the respiratory burst and nitric oxide and the secretion of IL-12 and IL-4 compared to non-educated monocytes. Conversely, these monocytes exhibited a significant increase in the production of IL-10 and TGF-?. Also, the levels of CD68+/CD206+ cells significantly increased in the population of educated monocytes. More importantly, the severity of histopathological lesions, NF-?B p65 mRNA level in lung tissues, total serum IgE and the total cell count, as well as the eosinophil count in the bronchoalveolar lavage fluid, were significantly decreased in OVA-inhaled mice treated with educated monocytes compared to OVA-sensitized group receiving non-educated monocytes. With no advantage in up-regulation of Foxp3 Treg cells, the treatment with educated monocytes reduced the secretion of IL-5 and IL-13 by splenocytes of asthma mice more than splenocytes of the asthma mice treated with non-educated monocytes. CONCLUSION The educated monocytes with MCM may be as a promising strategy for cell-based therapies of asthma.
Collapse
Affiliation(s)
| | | | | | - Siamak Naji Hadai
- b Department of Pathology, Faculty of Medicine , Urmia University of Medical Science , Urmia , Iran
| |
Collapse
|
17
|
Broekman W, Khedoe PPSJ, Schepers K, Roelofs H, Stolk J, Hiemstra PS. Mesenchymal stromal cells: a novel therapy for the treatment of chronic obstructive pulmonary disease? Thorax 2018; 73:565-574. [PMID: 29653970 PMCID: PMC5969341 DOI: 10.1136/thoraxjnl-2017-210672] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
COPD is characterised by tissue destruction and inflammation. Given the lack of curative treatments and the progressive nature of the disease, new treatments for COPD are highly relevant. In vitro cell culture and animal studies have demonstrated that mesenchymal stromal cells (MSCs) have the capacity to modify immune responses and to enhance tissue repair. These properties of MSCs provided a rationale to investigate their potential for treatment of a variety of diseases, including COPD. Preclinical models support the hypothesis that MSCs may have clinical efficacy in COPD. However, although clinical trials have demonstrated the safety of MSC treatment, thus far they have not provided evidence for MSC efficacy in the treatment of COPD. In this review, we discuss the rationale for MSC-based cell therapy in COPD, the main findings from in vitro and in vivo preclinical COPD model studies, clinical trials in patients with COPD and directions for further research.
Collapse
Affiliation(s)
- Winifred Broekman
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Padmini P S J Khedoe
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen Schepers
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Helene Roelofs
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
18
|
Sun Z, Li F, Zhou X, Chung KF, Wang W, Wang J. Stem cell therapies for chronic obstructive pulmonary disease: current status of pre-clinical studies and clinical trials. J Thorac Dis 2018; 10:1084-1098. [PMID: 29607186 DOI: 10.21037/jtd.2018.01.46] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease that has a major impact worldwide. The currently-available drugs mainly focus on relieving the symptoms of COPD patients. Novel regenerative therapeutic approaches have been investigated with the aim of repairing or replacing the injured functional structures of the respiratory system. We summarized the progress made by regenerative therapies for COPD by analyzing results from both pre-clinical studies and completed clinical trials. These approaches include the application of exogenous stem cells or small molecules to stimulate the regeneration by endogenous lung stem/progenitor cells. Exogenous mesenchymal stem cells (MSCs) have been reported to repair the structure and improve the function of the injured respiratory system in COPD models. However, the studies that used MSCs in patients with moderate-to-severe COPD patients did not lead to clear respiratory functional improvements. Exogenous human lung stem cells applied to cryo-injured (CI) lungs of mice have been shown to organize into human-like pulmonary structures, indicating a new property of stem cells that is potentially capable of curing COPD patients. Small molecules like retinoic acid has been shown to lead to regeneration and repair of the damaged lung structures in COPD mouse models probably by activation of endogenous lung stem/progenitor cells. However, retinoic acid or agonists of retinoic acid receptor administered to moderate or severe COPD patients did not improve the density and function of the damaged lung. These novel regenerative approaches have failed in preliminary clinical trials, possibly due to the advanced severity of the disease. Further work should be done to develop the current regenerative approaches for curing patients at different stages of COPD. We suggest that some modifications of the approach in the clinical studies may lead to more successful outcomes of regenerative therapy for COPD.
Collapse
Affiliation(s)
- Zhongwei Sun
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Feng Li
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Wen Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Jialun Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| |
Collapse
|
19
|
Xu T, Zhang Y, Chang P, Gong S, Shao L, Dong L. Mesenchymal stem cell-based therapy for radiation-induced lung injury. Stem Cell Res Ther 2018; 9:18. [PMID: 29386045 PMCID: PMC5793340 DOI: 10.1186/s13287-018-0776-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Since radiotherapy is widely used in managing thoracic tumors, physicians have begun to realize that radiation-induced lung injury (RILI) seriously limits the effects of radiotherapy. Unfortunately, there are still no effective methods for controlling RILI. Over the last few decades numerous studies have reported the beneficial effects of mesenchymal stem cells (MSCs) on tissue repair and regeneration. MSCs can not only differentiate into lung alveolar epithelial cells and secrete anti-inflammatory factors, but they also deliver some vehicles for gene therapy in repairing the injured lung, which provides new ideas for managing RILI. Thus, many scientists have attempted to manage RILI using MSC-based therapy. However, as a novel therapy MSCs still face various limitations. Herein, we shed light on the current understanding of MSC-based therapy for RILI, including the feasibility, molecular mechanisms, animal studies, and clinical research of MSC-based therapy for RILI. We also present an overview of RILI and MSCs.
Collapse
Affiliation(s)
- Tiankai Xu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Pengyu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Shouliang Gong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.,Key Laboratory of Radiobiology, Ministry of health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lihong Shao
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Lihua Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
20
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
21
|
Conese M, Beccia E, Castellani S, Di Gioia S, Colombo C, Angiolillo A, Carbone A. The long and winding road: stem cells for cystic fibrosis. Expert Opin Biol Ther 2017; 18:281-292. [PMID: 29216777 DOI: 10.1080/14712598.2018.1413087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic syndrome with a high mortality rate due to severe lung disease. Despite having several drugs targeting specific mutated CFTR proteins already in clinical trials, new therapies, based on stem cells, are also emerging to treat those patients. AREAS COVERED The authors review the main sources of stem cells, including embryonic stem cells (ESCs), induced-pluripotent stem cells (iPSCs), gestational stem cells, and adult stem cells, such as mesenchymal stem cells (MSCs) in the context of CF. Furthermore, they describe the main animal and human models of lung physiology and pathology, involved in the optimization of these stem cell-applied therapies in CF. EXPERT OPINION ESCs and iPSCs are emerging sources for disease modeling and drug discovery purposes. The allogeneic transplant of healthy MSCs, that acts independently to specific mutations, is under intense scrutiny due to their secretory, immunomodulatory, anti-inflammatory and anti-bacterial properties. The main challenge for future developments will be to get exogenous stem cells into the appropriate lung location, where they can regenerate endogenous stem cells and act as inflammatory modulators. The clinical application of stem cells for the treatment of CF certainly warrants further insight into pre-clinical models, including large animals, organoids, decellularized organs and lung bioengineering.
Collapse
Affiliation(s)
- Massimo Conese
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Elisa Beccia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy.,b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Stefano Castellani
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Sante Di Gioia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Carla Colombo
- c Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation , University of Milan , Milan , Italy
| | - Antonella Angiolillo
- b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Annalucia Carbone
- d Division of Internal Medicine and Chronobiology Unit , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo (FG) , Italy
| |
Collapse
|
22
|
Payne C, Dolan EB, O'Sullivan J, Cryan SA, Kelly HM. A methylcellulose and collagen based temperature responsive hydrogel promotes encapsulated stem cell viability and proliferation in vitro. Drug Deliv Transl Res 2017; 7:132-146. [PMID: 27924469 DOI: 10.1007/s13346-016-0347-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
With the number of stem cell-based therapies emerging on the increase, the need for novel and efficient delivery technologies to enable therapies to remain in damaged tissue and exert their therapeutic benefit for extended periods, has become a key requirement for their translation. Hydrogels, and in particular, thermoresponsive hydrogels, have the potential to act as such delivery systems. Thermoresponsive hydrogels, which are polymer solutions that transform into a gel upon a temperature increase, have a number of applications in the biomedical field due to their tendency to maintain a liquid state at room temperature, thereby enabling minimally invasive administration and a subsequent ability to form a robust gel upon heating to physiological temperature. However, various hurdles must be overcome to increase the clinical translation of hydrogels as a stem cell delivery system, with barriers including their low tensile strength and their inadequate support of cell viability and attachment. In order to address these issues, a methylcellulose based hydrogel was formulated in combination with collagen and beta glycerophosphate, and key development issues such as injectability and sterilisation processes were examined. The polymer solution underwent thermogelation at ~36 °C as determined by rheological analysis, and when gelled, was sufficiently robust to resist significant disintegration in the presence of phosphate buffered saline (PBS) while concomitantly allowing for diffusion of methylene blue dye solution into the gel. We demonstrate that human mesenchymal stem cells (hMSCs) encapsulated within the gel remained viable and showed raised levels of dsDNA at increasing time points, an indication of cell proliferation. Mechanical testing showed the "injectability", i.e. force required for delivery of the polymer solution through devices such as a syringe, needle or catheter. Sterilisation of the freeze-dried polymer wafer via gamma irradiation showed no adverse effects on the formed hydrogel characteristics. Taken together, these results indicate the potential of this gel as a clinically translatable delivery system for stem cells and therapeutic molecules in vivo.
Collapse
Affiliation(s)
- Christina Payne
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Eimear B Dolan
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Janice O'Sullivan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin 2, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Helena M Kelly
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland. .,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
23
|
Kim J, Guenthart B, O'Neill JD, Dorrello NV, Bacchetta M, Vunjak-Novakovic G. Controlled delivery and minimally invasive imaging of stem cells in the lung. Sci Rep 2017; 7:13082. [PMID: 29026127 PMCID: PMC5638808 DOI: 10.1038/s41598-017-13280-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/19/2017] [Indexed: 12/11/2022] Open
Abstract
Intratracheal delivery of stem cells into injured or diseased lungs can provide a variety of therapeutic and immunomodulatory effects for the treatment of acute lung injury and chronic lung disease. While the efficacy of this approach depends on delivering the proper cell dosage into the target region of the airway, tracking and analysis of the cells have been challenging, largely due to the limited understanding of cell transport and lack of suitable cell monitoring techniques. We report on the transport and deposition of intratracheally delivered stem cells as well as strategies to modulate the number of cells (e.g., dose), topographic distribution, and region-specific delivery in small (rodent) and large (porcine and human) lungs. We also developed minimally invasive imaging techniques for real-time monitoring of intratracheally delivered cells. We propose that this approach can facilitate the implementation of patient-specific cells and lead to enhanced clinical outcomes in the treatment of lung disease with cell-based therapies.
Collapse
Affiliation(s)
- Jinho Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - John D O'Neill
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - N Valerio Dorrello
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.,Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA. .,Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
24
|
Kim SY, Burgess JK, Wang Y, Kable EP, Weiss DJ, Chan HK, Chrzanowski W. Atomized Human Amniotic Mesenchymal Stromal Cells for Direct Delivery to the Airway for Treatment of Lung Injury. J Aerosol Med Pulm Drug Deliv 2016; 29:514-524. [DOI: 10.1089/jamp.2016.1289] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Sally Yunsun Kim
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| | - Janette K. Burgess
- Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, Australia
| | - Yiwei Wang
- ANZAC Research Institute, The University of Sydney, Concord, Australia
| | - Eleanor P.W. Kable
- Australian Centre for Microscopy & Microanalysis, The University of Sydney, Sydney, Australia
| | - Daniel J. Weiss
- College of Medicine, University of Vermont College of Medicine, Burlington, Vermont
| | - Hak-Kim Chan
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| | - Wojciech Chrzanowski
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
- Australian Institute of Nanoscale Science and Technology, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
25
|
Tan Q, Choi KM, Sicard D, Tschumperlin DJ. Human airway organoid engineering as a step toward lung regeneration and disease modeling. Biomaterials 2016; 113:118-132. [PMID: 27815996 DOI: 10.1016/j.biomaterials.2016.10.046] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/14/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Organoids represent both a potentially powerful tool for the study cell-cell interactions within tissue-like environments, and a platform for tissue regenerative approaches. The development of lung tissue-like organoids from human adult-derived cells has not previously been reported. Here we combined human adult primary bronchial epithelial cells, lung fibroblasts, and lung microvascular endothelial cells in supportive 3D culture conditions to generate airway organoids. We demonstrate that randomly-seeded mixed cell populations undergo rapid condensation and self-organization into discrete epithelial and endothelial structures that are mechanically robust and stable during long term culture. After condensation airway organoids generate invasive multicellular tubular structures that recapitulate limited aspects of branching morphogenesis, and require actomyosin-mediated force generation and YAP/TAZ activation. Despite the proximal source of primary epithelium used in the airway organoids, discrete areas of both proximal and distal epithelial markers were observed over time in culture, demonstrating remarkable epithelial plasticity within the context of organoid cultures. Airway organoids also exhibited complex multicellular responses to a prototypical fibrogenic stimulus (TGF-β1) in culture, and limited capacity to undergo continued maturation and engraftment after ectopic implantation under the murine kidney capsule. These results demonstrate that the airway organoid system developed here represents a novel tool for the study of disease-relevant cell-cell interactions, and establishes this platform as a first step toward cell-based therapy for chronic lung diseases based on de novo engineering of implantable airway tissues.
Collapse
Affiliation(s)
- Qi Tan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kyoung Moo Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
26
|
Hegab AE, Betsuyaku T. Lung Stem Cells and Their Use for Patient Care: Are We There Yet? ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-33270-3_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
27
|
Urbanek K, De Angelis A, Spaziano G, Piegari E, Matteis M, Cappetta D, Esposito G, Russo R, Tartaglione G, De Palma R, Rossi F, D’Agostino B. Intratracheal Administration of Mesenchymal Stem Cells Modulates Tachykinin System, Suppresses Airway Remodeling and Reduces Airway Hyperresponsiveness in an Animal Model. PLoS One 2016; 11:e0158746. [PMID: 27434719 PMCID: PMC4951036 DOI: 10.1371/journal.pone.0158746] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/21/2016] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The need for new options for chronic lung diseases promotes the research on stem cells for lung repair. Bone marrow-derived mesenchymal stem cells (MSCs) can modulate lung inflammation, but the data on cellular processes involved in early airway remodeling and the potential involvement of neuropeptides are scarce. OBJECTIVES To elucidate the mechanisms by which local administration of MSCs interferes with pathophysiological features of airway hyperresponsiveness in an animal model. METHODS GFP-tagged mouse MSCs were intratracheally delivered in the ovalbumin mouse model with subsequent functional tests, the analysis of cytokine levels, neuropeptide expression and histological evaluation of MSCs fate and airway pathology. Additionally, MSCs were exposed to pro-inflammatory factors in vitro. RESULTS Functional improvement was observed after MSC administration. Although MSCs did not adopt lung cell phenotypes, cell therapy positively affected airway remodeling reducing the hyperplastic phase of the gain in bronchial smooth muscle mass, decreasing the proliferation of epithelium in which mucus metaplasia was also lowered. Decrease of interleukin-4, interleukin-5, interleukin-13 and increase of interleukin-10 in bronchoalveolar lavage was also observed. Exposed to pro-inflammatory cytokines, MSCs upregulated indoleamine 2,3-dioxygenase. Moreover, asthma-related in vivo upregulation of pro-inflammatory neurokinin 1 and neurokinin 2 receptors was counteracted by MSCs that also determined a partial restoration of VIP, a neuropeptide with anti-inflammatory properties. CONCLUSION Intratracheally administered MSCs positively modulate airway remodeling, reduce inflammation and improve function, demonstrating their ability to promote tissue homeostasis in the course of experimental allergic asthma. Because of a limited tissue retention, the functional impact of MSCs may be attributed to their immunomodulatory response combined with the interference of neuropeptide system activation and tissue remodeling.
Collapse
MESH Headings
- Animals
- Bronchoalveolar Lavage Fluid/chemistry
- Bronchoalveolar Lavage Fluid/immunology
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-13/genetics
- Interleukin-13/immunology
- Interleukin-4/genetics
- Interleukin-4/immunology
- Interleukin-5/genetics
- Interleukin-5/immunology
- Intubation, Intratracheal
- Lung/immunology
- Lung/pathology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/immunology
- Mice
- Mice, Inbred BALB C
- Ovalbumin
- Receptors, Neurokinin-1/genetics
- Receptors, Neurokinin-1/immunology
- Receptors, Neurokinin-2/genetics
- Receptors, Neurokinin-2/immunology
- Respiratory Hypersensitivity/chemically induced
- Respiratory Hypersensitivity/immunology
- Respiratory Hypersensitivity/pathology
- Respiratory Hypersensitivity/therapy
Collapse
Affiliation(s)
- Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
- * E-mail: (AA); (BA)
| | - Giuseppe Spaziano
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Maria Matteis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Gioia Tartaglione
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Second University of Naples, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Bruno D’Agostino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
- * E-mail: (AA); (BA)
| |
Collapse
|
28
|
Smirnova NF, Schamberger AC, Nayakanti S, Hatz R, Behr J, Eickelberg O. Detection and quantification of epithelial progenitor cell populations in human healthy and IPF lungs. Respir Res 2016; 17:83. [PMID: 27423691 PMCID: PMC4947297 DOI: 10.1186/s12931-016-0404-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/10/2016] [Indexed: 12/17/2022] Open
Abstract
Background In the human lung, epithelial progenitor cells in the airways give rise to the differentiated pseudostratified airway epithelium. In mice, emerging evidence confers a progenitor function to cytokeratin 5 (KRT5+) or cytokeratin 14 (KRT14+)-positive basal cells of the airway epithelium. Little is known, however, about the distribution of progenitor subpopulations in the human lung, particularly about aberrant epithelial differentiation in lung disease, such as idiopathic pulmonary fibrosis (IPF). Methods Here, we used multi-color immunofluorescence analysis to detect and quantify the distribution of airway epithelial progenitor subpopulations in human lungs obtained from healthy donors or IPF patients. Results In lungs from both, healthy donors and IPF patients, we detected KRT5+KRT14-, KRT5-KRT14+ and KRT5+KRT14+ populations in the proximal airways. KRT14+ cells, however, were absent in the distal airways of healthy lungs. In IPF, we detected a dramatic increase in the amount of KRT5+ cells and the emergence of a frequent KRT5+KRT14+ epithelial population, in particular in distal airways and alveolar regions. While the KRT14- progenitor population exhibited signs of proper epithelial differentiation, as evidenced by co-staining with pro-SPC, aquaporin 5, CC10, or MUC5B, the KRT14+ cell population did not co-stain with bronchial/alveolar differentiation markers in IPF. Conclusions We provide, for the first time, a quantitative profile of the distribution of epithelial progenitor populations in human lungs. We show compelling evidence for dysregulation and aberrant differentiation of these populations in IPF.
Collapse
Affiliation(s)
- N F Smirnova
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - A C Schamberger
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - S Nayakanti
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - R Hatz
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine, Viszeral, Transplantations, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Munich, Germany.,Asklepios Fachkliniken München-Gauting, Munich, Germany.,German Center of Lung Research (DZL), Hannover, Germany
| | - J Behr
- Asklepios Fachkliniken München-Gauting, Munich, Germany.,Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany.,German Center of Lung Research (DZL), Hannover, Germany
| | - O Eickelberg
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany. .,German Center of Lung Research (DZL), Hannover, Germany. .,Comprehensive Pneumology Center (CPC), Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, D-81377, Germany.
| |
Collapse
|
29
|
Vandenplas S, Willems M, Witten PE, Hansen T, Fjelldal PG, Huysseune A. Epithelial Label-Retaining Cells Are Absent during Tooth Cycling in Salmo salar and Polypterus senegalus. PLoS One 2016; 11:e0152870. [PMID: 27049953 PMCID: PMC4822771 DOI: 10.1371/journal.pone.0152870] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 03/21/2016] [Indexed: 01/17/2023] Open
Abstract
The Atlantic salmon (Salmo salar) and African bichir (Polypterus senegalus) are both actinopterygian fish species that continuously replace their teeth without the involvement of a successional dental lamina. Instead, they share the presence of a middle dental epithelium: an epithelial tier enclosed by inner and outer dental epithelium. It has been hypothesized that this tier could functionally substitute for a successional dental lamina and might be a potential niche to house epithelial stem cells involved in tooth cycling. Therefore, in this study we performed a BrdU pulse chase experiment on both species to (1) determine the localization and extent of proliferating cells in the dental epithelial layers, (2) describe cell dynamics and (3) investigate if label-retaining cells are present, suggestive for the putative presence of stem cells. Cells proliferate in the middle dental epithelium, outer dental epithelium and cervical loop at the lingual side of the dental organ to form a new tooth germ. Using long chase times, both in S. salar (eight weeks) and P. senegalus (eight weeks and twelve weeks), we could not reveal the presence of label-retaining cells in the dental organ. Immunostaining of P. senegalus dental organs for the transcription factor Sox2, often used as a stem cell marker, labelled cells in the zone of outer dental epithelium which grades into the oral epithelium (ODE transition zone) and the inner dental epithelium of a successor only. The location of Sox2 distribution does not provide evidence for epithelial stem cells in the dental organ and, more specifically, in the middle dental epithelium. Comparison of S. salar and P. senegalus reveals shared traits in tooth cycling and thus advances our understanding of the developmental mechanism that ensures lifelong replacement.
Collapse
Affiliation(s)
- Sam Vandenplas
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| | - Maxime Willems
- Pharmaceutical technology, Ghent University, Ghent, Belgium
| | - P. Eckhard Witten
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| | - Tom Hansen
- Institute of Marine Research (IMR), Matre Research Station, Matredal, Norway
| | - Per Gunnar Fjelldal
- Institute of Marine Research (IMR), Matre Research Station, Matredal, Norway
| | - Ann Huysseune
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
30
|
Neonatal Type II Alveolar Epithelial Cell Transplant Facilitates Lung Reparation in Piglets With Acute Lung Injury and Extracorporeal Life Support. Pediatr Crit Care Med 2016; 17:e182-92. [PMID: 26890195 DOI: 10.1097/pcc.0000000000000667] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Type II alveolar epithelial cells have potential for lung growth and reparation. Extracorporeal membrane oxygenation is used as life support for lung impairment resulting from acute respiratory distress syndrome. We hypothesized that intratracheal transplantation of isogeneic primary type II alveolar epithelial cells in combination with extracorporeal membrane oxygenation may facilitate lung reparation for acute lung injury (ALI). DESIGN A randomized, controlled experiment. SETTING An animal laboratory in a university pediatric center. SUBJECTS Twenty-eight 4- to 6-week young piglets, weighing 7-8 kg. INTERVENTIONS Type II alveolar epithelial cells from neonatal male piglet lungs were isolated, purified, cultured, and labeled with chemical stain PKH26. After 3-6 hours of induction of ALI by IV endotoxin and mechanical ventilation (MV), young female piglets were allocated to five groups (n = 5): ALI-MV, ALI treated with MV; ALI-EC, ALI treated with both MV and venovenous extracorporeal membrane oxygenation; ALI-EC-T, ALI-EC protocol plus intratracheal type II alveolar epithelial cell transplant; CON-MV, healthy animals treated with MV; and CON-EC-T, healthy animals treated with venovenous extracorporeal membrane oxygenation. After 24 hours, animals were weaned from treatment for recovery in the ensuing 14 days, with their lungs assessed for injury and reparation. MEASUREMENTS AND MAIN RESULTS Lung injury for animals in ALI-MV was moderate to severe, whereas much milder injuries in ALI-EC-T and ALI-EC were found. More PKH26-labeled type II alveolar epithelial cells were detected by fluorescence in the lungs of ALI-EC-T than in CON-EC-T as further verified by the expression of messenger RNA of sex-determining region of Y chromosome. Electromicroscopically intact type II alveolar epithelial cells and prominent lattice-like tubular myelin were also found in ALI-EC-T and CON-MV but not in ALI-EC. The hydroxyproline level in lung tissue was significantly lower in ALI-EC-T than in ALI-EC and ALI-MV, with most of the lung histopathologic and pathobiologic manifestations in favor of ALI-EC-T. CONCLUSIONS The preliminary data suggested that type II alveolar epithelial cell transplant facilitated lung reparation for ALI in this model.
Collapse
|
31
|
Hirano T, Kikuchi T, Tode N, Santoso A, Yamada M, Mitsuhashi Y, Komatsu R, Kawabe T, Tanimoto T, Ishii N, Tanaka Y, Nishimura H, Nukiwa T, Watanabe A, Ichinose M. OX40 ligand newly expressed on bronchiolar progenitors mediates influenza infection and further exacerbates pneumonia. EMBO Mol Med 2016; 8:422-36. [PMID: 26976612 PMCID: PMC4818750 DOI: 10.15252/emmm.201506154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/11/2016] [Accepted: 02/16/2016] [Indexed: 12/20/2022] Open
Abstract
Influenza virus epidemics potentially cause pneumonia, which is responsible for much of the mortality due to the excessive immune responses. The role of costimulatory OX40-OX40 ligand (OX40L) interactions has been explored in the non-infectious pathology of influenza pneumonia. Here, we describe a critical contribution of OX40L to infectious pathology, with OX40L deficiency, but not OX40 deficiency, resulting in decreased susceptibility to influenza viral infection. Upon infection, bronchiolar progenitors increase in number for repairing the influenza-damaged epithelia. The OX40L expression is induced on the progenitors for the antiviral immunity during the infectious process. However, these defense-like host responses lead to more extensive infection owing to the induced OX40L with α-2,6 sialic acid modification, which augments the interaction with the viral hemagglutinin. In fact, the specific antibody against the sialylated site of OX40L exhibited therapeutic potency in mitigating the OX40L-mediated susceptibility to influenza. Our data illustrate that the influenza-induced expression of OX40L on bronchiolar progenitors has pathogenic value to develop a novel therapeutic approach against influenza.
Collapse
Affiliation(s)
- Taizou Hirano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Tode
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Arif Santoso
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshiya Mitsuhashi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Riyo Komatsu
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Tanimoto
- Kanonji Institute, The Research Foundation for Microbial Diseases of Osaka University, Kanonji, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuetsu Tanaka
- Department of Immunology, Graduate School of Medicine University of the Ryukyus, Okinawa, Japan
| | - Hidekazu Nishimura
- Virus Research Center, Sendai Medical Center National Hospital Organization, Sendai, Japan
| | - Toshihiro Nukiwa
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Watanabe
- Research Division for Development of Anti-Infective Agents, Institute of Development, Aging and Cancer Tohoku University, Sendai, Japan
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
32
|
Vascular Wall-Resident Multipotent Stem Cells of Mesenchymal Nature within the Process of Vascular Remodeling: Cellular Basis, Clinical Relevance, and Implications for Stem Cell Therapy. Stem Cells Int 2016; 2016:1905846. [PMID: 26880936 PMCID: PMC4736960 DOI: 10.1155/2016/1905846] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/18/2015] [Indexed: 12/13/2022] Open
Abstract
Until some years ago, the bone marrow and the endothelial cell compartment lining the vessel lumen (subendothelial space) were thought to be the only sources providing vascular progenitor cells. Now, the vessel wall, in particular, the vascular adventitia, has been established as a niche for different types of stem and progenitor cells with the capacity to differentiate into both vascular and nonvascular cells. Herein, vascular wall-resident multipotent stem cells of mesenchymal nature (VW-MPSCs) have gained importance because of their large range of differentiation in combination with their distribution throughout the postnatal organism which is related to their existence in the adventitial niche, respectively. In general, mesenchymal stem cells, also designated as mesenchymal stromal cells (MSCs), contribute to the maintenance of organ integrity by their ability to replace defunct cells or secrete cytokines locally and thus support repair and healing processes of the affected tissues. This review will focus on the central role of VW-MPSCs within vascular reconstructing processes (vascular remodeling) which are absolute prerequisite to preserve the sensitive relationship between resilience and stability of the vessel wall. Further, a particular advantage for the therapeutic application of VW-MPSCs for improving vascular function or preventing vascular damage will be discussed.
Collapse
|
33
|
Alhasan L, Qi A, Rezk AR, Yeo LY, Chan PPY. Assessment of the potential of a high frequency acoustomicrofluidic nebulisation platform for inhaled stem cell therapy. Integr Biol (Camb) 2016; 8:12-20. [DOI: 10.1039/c5ib00206k] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This study demonstrates the use of a novel high frequency acoustic nebulisation platform as an effective aerosolisation technique for inhaled mesenchymal stem cell (MSC) therapy.
Collapse
Affiliation(s)
- Layla Alhasan
- Department of Biotechnology & Biological Science
- RMIT University
- Melbourne
- Australia
- Micro/Nanophysics Research Laboratory
| | - Aisha Qi
- Micro/Nanophysics Research Laboratory
- RMIT University
- Melbourne
- Australia
| | - Amgad R. Rezk
- Micro/Nanophysics Research Laboratory
- RMIT University
- Melbourne
- Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research Laboratory
- RMIT University
- Melbourne
- Australia
| | - Peggy P. Y. Chan
- Micro/Nanophysics Research Laboratory
- RMIT University
- Melbourne
- Australia
- Department of Biomedical Engineering
| |
Collapse
|
34
|
Hypoxia-Induced Epithelial-Mesenchymal Transition Is Involved in Bleomycin-Induced Lung Fibrosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:232791. [PMID: 26819949 PMCID: PMC4706863 DOI: 10.1155/2015/232791] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a severe disease that contributes to the morbidity and mortality of a number of lung diseases. However, the molecular and cellular mechanisms leading to lung fibrosis are poorly understood. This study investigated the roles of epithelial-mesenchymal transition (EMT) and the associated molecular mechanisms in bleomycin-induced lung fibrosis. The bleomycin-induced fibrosis animal model was established by intratracheal injection of a single dose of bleomycin. Protein expression was measured by Western blot, immunohistochemistry, and immunofluorescence. Typical lesions of lung fibrosis were observed 1 week after bleomycin injection. A progressive increase in MMP-2, S100A4, α-SMA, HIF-1α, ZEB1, CD44, phospho-p44/42 (p-p44/42), and phospho-p38 MAPK (p-p38) protein levels as well as activation of EMT was observed in the lung tissues of bleomycin mice. Hypoxia increased HIF-1α and ZEB1 expression and activated EMT in H358 cells. Also, continuous incubation of cells under mild hypoxic conditions increased CD44, p-p44/42, and p-p38 protein levels in H358 cells, which correlated with the increase in S100A4 expression. In conclusion, bleomycin induces progressive lung fibrosis, which may be associated with activation of EMT. The fibrosis-induced hypoxia may further activate EMT in distal alveoli through a hypoxia-HIF-1α-ZEB1 pathway and promote the differentiation of lung epithelial cells into fibroblasts through phosphorylation of p38 MAPK and Erk1/2 proteins.
Collapse
|
35
|
Syndecan 4 Mediates Nrf2-dependent Expansion of Bronchiolar Progenitors That Protect Against Lung Inflammation. Mol Ther 2015; 24:41-52. [PMID: 26307669 PMCID: PMC4754542 DOI: 10.1038/mt.2015.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 08/11/2015] [Indexed: 12/16/2022] Open
Abstract
The use of lung progenitors for regenerative medicine appears promising, but their biology is not fully understood. Here, we found anti-inflammatory attributes in bronchiolar progenitors that were sorted as a multipotent subset of mouse club cells and found to express secretory leukocyte protease inhibitor (SLPI). Notably, the impaired expression of SLPI in mice increased the number of bronchiolar progenitors and decreased the lung inflammation. We determined a transcriptional profile for the bronchiolar progenitors of Slpi-deficient mice and identified syndecan 4, whose expression was markedly elevated as compared to that of wild-type mice. Systemic administration of recombinant syndecan 4 protein caused a substantial increase in the number of bronchiolar progenitors with concomitant attenuation of both airway and alveolar inflammation. The syndecan 4 administration also resulted in activation of the Keap1-Nrf2 antioxidant pathway in lung cells, which is critically involved in the therapeutic responses to the syndecan 4 treatment. Moreover, in 3D culture, the presence of syndecan 4 induced differentiated club cells to undergo Nrf2-dependent transition into bronchiolar progenitors. Our observations reveal that differentiative switches between bronchiolar progenitors and club cells are under the Nrf2-mediated control of SLPI and syndecan 4, suggesting the possibility of new therapeutic approaches in inflammatory lung diseases.
Collapse
|
36
|
Erythropoietin promotes the protective properties of transplanted endothelial progenitor cells against acute lung injury via PI3K/Akt pathway. Shock 2015; 42:327-36. [PMID: 25051281 DOI: 10.1097/shk.0000000000000216] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Accumulating evidences have indicted the participation and repairing effects of endothelial progenitor cells (EPCs) on acute lung injury (ALI). Researchers have also revealed that erythropoietin (EPO) may exert multiple effects on EPCs including proliferative and adhesive properties. The present study was designed to investigate whether EPO can promote the healing efficiency of transplanted EPCs against ALI and the potential mechanism. METHODS Endothelial progenitor cells were derived from bone marrow mononuclear cells of BALB/c mice. In our in vitro studies, we tested the proproliferative and antiapoptotic effects of EPO on cultured EPCs. In our in vivo studies, we induced BALB/c mice ALI model by intratracheal instillation of lipopolysaccharide and treated with/without intravenous injection of enhanced green fluorescent protein-EPCs harvested from enhanced green fluorescent protein-BALB/c mice. Mice that received EPC transplantation either underwent EPO administration or not. The effects of EPO and EPC treatment on promoting pulmonary endothelial repair, decreasing pulmonary capillary permeability, alleviating pulmonary inflammation, improving gas exchange, and promoting pulmonary vessel angiogenesis were tested. The potential mechanisms that mediate EPO functions on EPC were also investigated. RESULTS Our studies revealed a significant antiapoptotic property of EPO on cultured EPCs as well as its promotion on EPC proliferation. We also found the severity of ALI was reduced by EPC therapy, and the protective effects of EPCs were highly enhanced when combined with EPO administration. However, all these stimulating effects of EPO on EPCs were consisted with the expression of phospho-Akt and were abrogated by PI3K inhibitors. CONCLUSIONS Transplanted EPCs directly incorporated into the injured pulmonary vessels and maintain the integrity of pulmonary endothelium. Erythropoietin improved the survival and proliferation of transplanted EPCs and recruited them to the injured sites to exert their repairing functions. PI3K/Akt pathway mediated EPO's functions on EPCs. The combination of EPO and EPC treatment may be a promising cell-based therapy for ALI patients.
Collapse
|
37
|
|
38
|
van der Laarse A, Cobbaert CM, Umar S. Stem and progenitor cell therapy for pulmonary arterial hypertension: effects on the right ventricle (2013 Grover Conference Series). Pulm Circ 2015; 5:73-80. [PMID: 25992272 DOI: 10.1086/679701] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/25/2014] [Indexed: 12/12/2022] Open
Abstract
In experimental animals and in patients with pulmonary arterial hypertension (PAH), a wide spectrum of structural and functional conditions is known that may be responsible for the switch of a state of "compensated" right ventricular (RV) hypertrophy to a state of RV failure. In recent years, therapy with differentiated cells, endothelial progenitor cells, and mesenchymal stem cells has been shown to cause partial or complete reversal of pathological characteristics of PAH. The therapeutic effects of stem or progenitor cell therapy are considered to be (1) paracrine effects from stem or progenitor cells that had engrafted in the myocardium (or elsewhere), by compounds that have anti-inflammatory, antiapoptotic, and proangiogenic actions and (2) unloading effects on the right ventricle due to stem or progenitor cell-induced decrease in pulmonary vascular resistance and decrease in pulmonary artery pressure.
Collapse
Affiliation(s)
- Arnoud van der Laarse
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands ; Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, California, USA
| |
Collapse
|
39
|
Lowenthal J, Sugarman J. Ethics and policy issues for stem cell research and pulmonary medicine. Chest 2015; 147:824-834. [PMID: 25732448 DOI: 10.1378/chest.14-1696] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Stem cell research and related initiatives in regenerative medicine, cell-based therapy, and tissue engineering have generated considerable scientific and public interest. Researchers are applying stem cell technologies to chest medicine in a variety of ways: using stem cells as models for drug discovery, testing stem cell-based therapies for conditions as diverse as COPD and cystic fibrosis, and producing functional lung and tracheal tissue for physiologic modeling and potential transplantation. Although significant scientific obstacles remain, it is likely that stem cell-based regenerative medicine will have a significant clinical impact in chest medicine. However, stem cell research has also generated substantial controversy, posing a variety of ethical and regulatory challenges for research and clinical practice. Some of the most prominent ethical questions related to the use of stem cell technologies in chest medicine include (1) implications for donors, (2) scientific prerequisites for clinical testing and use, (3) stem cell tourism, (4) innovation and clinical use of emerging stem cell-based interventions, (5) responsible translation of stem cell-based therapies to clinical use, and (6) appropriate and equitable access to emerging therapies. Having a sense of these issues should help to put emerging scientific advances into appropriate context and to ensure the responsible clinical translation of promising therapeutics.
Collapse
Affiliation(s)
- Justin Lowenthal
- School of Medicine, Medical Scientist Training Program, Johns Hopkins University, Baltimore, MD
| | - Jeremy Sugarman
- Berman Institute of Bioethics, Department of Medicine, Department of Health Policy and Management, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
40
|
|
41
|
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
|
42
|
Emura M, Aufderheide M, Mohr U. Target cell types with stem/progenitor function to isolate for in vitro reconstruction of human bronchiolar epithelia. ACTA ACUST UNITED AC 2015; 67:81-8. [DOI: 10.1016/j.etp.2014.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/19/2022]
|
43
|
Abstract
Airway diseases including COPD (chronic obstructive pulmonary disease), cystic fibrosis and lung cancer are leading causes of worldwide morbidity and mortality, with annual healthcare costs of billions of pounds. True regeneration of damaged airways offers the possibility of restoring lung function and protecting against airway transformation. Recently, advances in tissue engineering have allowed the development of cadaveric and biosynthetic airway grafts. Although these have produced encouraging results, the ability to achieve long-term functional airway regeneration remains a major challenge. To promote regeneration, exogenously delivered stem and progenitor cells are being trialled as cellular therapies. Unfortunately, current evidence suggests that only small numbers of exogenously delivered stem cells engraft within lungs, thereby limiting their utility for airway repair. In other organ systems, magnetic targeting has shown promise for improving long-term robust cell engraftment. This technique involves in vitro cell expansion, magnetic actuation and magnetically guided cell engraftment to sites of tissue damage. In the present paper, we discuss the utility of coupling stem cell-mediated cellular therapy with magnetic targeting for improving airway regeneration.
Collapse
|
44
|
Mammoto T, Mammoto A. Implantation of fibrin gel on mouse lung to study lung-specific angiogenesis. J Vis Exp 2014:52012. [PMID: 25548859 PMCID: PMC4396947 DOI: 10.3791/52012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent significant advances in stem cell research and bioengineering techniques have made great progress in utilizing biomaterials to regenerate and repair damage in simple tissues in the orthopedic and periodontal fields. However, attempts to regenerate the structures and functions of more complex three-dimensional (3D) organs such as lungs have not been very successful because the biological processes of organ regeneration have not been well explored. It is becoming clear that angiogenesis, the formation of new blood vessels, plays key roles in organ regeneration. Newly formed vasculatures not only deliver oxygen, nutrients and various cell components that are required for organ regeneration but also provide instructive signals to the regenerating local tissues. Therefore, to successfully regenerate lungs in an adult, it is necessary to recapitulate the lung-specific microenvironments in which angiogenesis drives regeneration of local lung tissues. Although conventional in vivo angiogenesis assays, such as subcutaneous implantation of extracellular matrix (ECM)-rich hydrogels (e.g., fibrin or collagen gels or Matrigel - ECM protein mixture secreted by Engelbreth-Holm-Swarm mouse sarcoma cells), are extensively utilized to explore the general mechanisms of angiogenesis, lung-specific angiogenesis has not been well characterized because methods for orthotopic implantation of biomaterials in the lung have not been well established. The goal of this protocol is to introduce a unique method to implant fibrin gel on the lung surface of living adult mouse, allowing for the successful recapitulation of host lung-derived angiogenesis inside the gel. This approach enables researchers to explore the mechanisms by which the lung-specific microenvironment controls angiogenesis and alveolar regeneration in both normal and pathological conditions. Since implanted biomaterials release and supply physical and chemical signals to adjacent lung tissues, implantation of these biomaterials on diseased lung can potentially normalize the adjacent diseased tissues, enabling researchers to develop new therapeutic approaches for various types of lung diseases.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School;
| | - Akiko Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School;
| |
Collapse
|
45
|
Shen Q, Chen B, Xiao Z, Zhao L, Xu X, Wan X, Jin M, Dai J, Dai H. Paracrine factors from mesenchymal stem cells attenuate epithelial injury and lung fibrosis. Mol Med Rep 2014; 11:2831-7. [PMID: 25514921 DOI: 10.3892/mmr.2014.3092] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/26/2014] [Indexed: 12/30/2022] Open
Abstract
Paracrine factors are currently considered to be the major mechanism through which mesenchymal stem cells (MSCs) exert their actions. The aim of this study was to investigate the protective effects of conditioned medium (CM) from bone marrow mesenchymal stem cells (MSC) on bleomycin (BLM)‑induced lung injury and fibrosis, both in vitro and in vivo. A549 human non‑small cell lung cancer epithelial cells were cultured in serum‑free medium, or MSC‑CM, both with or without BLM. The protective effects of MSC‑CM was determined by MTT assay to assess cell viability and Annexin V‑PE to assess apoptosis. Rats were intratracheally injected with MSC‑CM, saline, or conditioned medium from fibroblasts on day 0 and day 3 after intratracheal administration of BLM, and were sacrificed on day 28. Lung injury and fibrosis were assessed by histological assessment, Ashcroft score, and hydroxyproline assay; lung cell apoptosis was detected using terminal deoxynucleotidyl transferase dUTP nick end labeling assay. In comparison to the control group (0.17±0.01), 8 and 16% MSC‑CM had a significant stimulatory effect on A549 cellular proliferation (0.24±0.03 and 0.24±0.04, respectively, P<0.01). A549 cells cultured with MSC‑CM were protected from BLM‑induced apoptosis, 23.43±3.76% vs. 38.06±4.32%; (P<0.05). In the BLM‑challenged rats, MSC‑CM was shown to protect against lung fibrosis in terms of lung inflammation, fibrotic scores, collagen deposition, and cell apoptosis. This data suggests that MSCs are capable of protecting against lung injury and fibrosis both in vitro and in vivo through a paracrine anti‑inflammatory mechanism. MSC‑CM may provide a novel approach for the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Qinqin Shen
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Bing Chen
- Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Zhifeng Xiao
- Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Lifen Zhao
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xuefeng Xu
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xuan Wan
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Mulan Jin
- Department of Pathology Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Jianwu Dai
- Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Huaping Dai
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao‑Yang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
46
|
Abstract
In addition to its established contribution to innate immunity, recent studies have suggested novel roles for the complement system in the development of various lung diseases. Several studies have demonstrated that complement may serve as a key link between innate and adaptive immunity in a variety of pulmonary conditions. However, the specific contributions of complement to lung diseases based on innate and adaptive immunity are just beginning to emerge. Elucidating the role of complement-mediated immune regulation in these diseases will help to identify new targets for therapeutic interventions.
Collapse
|
47
|
Suen CM, Mei SHJ, Kugathasan L, Stewart DJ. Targeted delivery of genes to endothelial cells and cell- and gene-based therapy in pulmonary vascular diseases. Compr Physiol 2014; 3:1749-79. [PMID: 24265244 DOI: 10.1002/cphy.c120034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease that, despite significant advances in medical therapies over the last several decades, continues to have an extremely poor prognosis. Gene therapy is a method to deliver therapeutic genes to replace defective or mutant genes or supplement existing cellular processes to modify disease. Over the last few decades, several viral and nonviral methods of gene therapy have been developed for preclinical PAH studies with varying degrees of efficacy. However, these gene delivery methods face challenges of immunogenicity, low transduction rates, and nonspecific targeting which have limited their translation to clinical studies. More recently, the emergence of regenerative approaches using stem and progenitor cells such as endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs) have offered a new approach to gene therapy. Cell-based gene therapy is an approach that augments the therapeutic potential of EPCs and MSCs and may deliver on the promise of reversal of established PAH. These new regenerative approaches have shown tremendous potential in preclinical studies; however, large, rigorously designed clinical studies will be necessary to evaluate clinical efficacy and safety.
Collapse
Affiliation(s)
- Colin M Suen
- Sprott Centre for Stem Cell Research, The Ottawa Hospital Research Institute and University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Pimton P, Lecht S, Stabler CT, Johannes G, Schulman ES, Lelkes PI. Hypoxia enhances differentiation of mouse embryonic stem cells into definitive endoderm and distal lung cells. Stem Cells Dev 2014; 24:663-76. [PMID: 25226206 DOI: 10.1089/scd.2014.0343] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We investigated the effects of hypoxia on spontaneous (SP)- and activin A (AA)-induced definitive endoderm (DE) differentiation of mouse embryonic stem cells (mESCs) and their subsequent differentiation into distal pulmonary epithelial cells. SP differentiation for 6 days of mESCs toward endoderm at hypoxia of 1% O2, but not at 3% or 21% (normoxia), increased the expression of Sox17 and Foxa2 by 31- and 63-fold above maintenance culture, respectively. Treatment of mESCs with 20 ng/mL AA for 6 days under hypoxia further increased the expression of DE marker genes Sox17, Foxa2, and Cxcr4 by 501-, 1,483-, and 126-fold above maintenance cultures, respectively. Transient exposure to hypoxia, as short as 24 h, was sufficient to enhance AA-induced endoderm formation. The involvement of hypoxia-inducible factor (HIF)-1α and reactive oxygen species (ROS) in the AA-induced endoderm enrichment was assessed using HIF-1α(-/-) mESCs and the ROS scavenger N-acetylcysteine (NAC). Under SP conditions, HIF-1α(-/-) mESCs failed to increase the expression of endodermal marker genes but rather shifted toward ectoderm. Hypoxia induced only a marginal potentiation of AA-induced endoderm differentiation in HIF-1α(-/-) mESCs. Treatment of mESCs with AA and NAC led to a dose-dependent decrease in Sox17 and Foxa2 expression. In addition, the duration of exposure to hypoxia in the course of a recently reported lung differentiation protocol resulted in differentially enhanced expression of distal lung epithelial cell marker genes aquaporin 5 (Aqp5), surfactant protein C (Sftpc), and secretoglobin 1a1 (Scgb1a1) for alveolar epithelium type I, type II, and club cells, respectively. Our study is the first to show the effects of in vitro hypoxia on efficient formation of DE and lung lineages. We suggest that the extent of hypoxia and careful timing may be important components of in vitro differentiation bioprocesses for the differential generation of distal lung epithelial cells from pluripotent progenitors.
Collapse
Affiliation(s)
- Pimchanok Pimton
- 1 Department of Biology, School of Science, Walailak University , Nakhon Si Thammarat, Thailand
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Regenerative medicine may enable replacement of damaged or diseased tissues. But its clinical success will require deeper understanding of the basic biology of the stem cell niche and coordination between stem cell biologists and those in other fields.
Collapse
|
50
|
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
|