1
|
Chang YS, Yang M, Ahn SY, Sung SI, Park WS. Improving the future of clinical trials and translation of mesenchymal stromal cell therapies for neonatal disorders. Stem Cells Transl Med 2024; 13:941-948. [PMID: 39120439 PMCID: PMC11465171 DOI: 10.1093/stcltm/szae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/23/2024] [Indexed: 08/10/2024] Open
Abstract
Despite recent advances in neonatal intensive care medicine, neonatal disorders such as (bronchopulmonary dysplasia [BPD], intraventricular hemorrhage [IVH], and hypoxic ischemic encephalopathy [HIE]) remain major causes of death and morbidity in survivors, with few effective treatments being available. Recent preclinical studies have demonstrated the pleiotropic host injury-responsive paracrine protective effects of cell therapy especially with mesenchymal stromal cells (MSCs) against BPD, IVH, and HIE. These findings suggest that MSCs therapy might emerge as a novel therapeutic modality for these currently devastating neonatal disorders with complex multifactorial etiologies. Although early-phase clinical trials suggest their safety and feasibility, their clinical therapeutic benefits have not yet been proven. Therefore, based on currently available preclinical research and clinical trial data, we focus on critical issues that need to be addressed for future successful clinical trials and eventual clinical translation such as selecting the right patient and optimal cell type, route, dose, and timing of MSCs therapy for neonatal disorders such as BPD, HIE, and IVH.
Collapse
Affiliation(s)
- Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Misun Yang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Gangnam Cha Hospital, Cha University, Seoul, Korea
| |
Collapse
|
2
|
Purcell E, Shah J, Powell C, Nguyen T, Zhou L, McDonald CA, Allison BJ, Malhotra A. Umbilical cord blood-derived therapy for preterm lung injury: a systematic review and meta-analysis. Stem Cells Transl Med 2024; 13:606-624. [PMID: 38819251 PMCID: PMC11227974 DOI: 10.1093/stcltm/szae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/14/2024] [Indexed: 06/01/2024] Open
Abstract
INTRODUCTION Lung injuries, such as bronchopulmonary dysplasia (BPD), remain a major complication of preterm birth, with limited therapeutic options. One potential emerging therapy is umbilical cord blood (UCB)-derived therapy. OBJECTIVES To systematically assess the safety and efficacy of UCB-derived therapy for preterm lung injury in preclinical and clinical studies. METHODS A systematic search of MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and WHO International Trials Registry Platform was performed. A meta-analysis was conducted with Review Manager (5.4.1) using a random effects model. Data was expressed as standardized mean difference (SMD) for preclinical data and pooled relative risk (RR) for clinical data, with 95% confidence intervals (CI). Potential effect modifiers were investigated via subgroup analysis. Certainty of evidence was assessed using the GRADE system. RESULTS Twenty-three preclinical studies and six clinical studies met eligibility criteria. Statistically significant improvements were seen across several preclinical outcomes, including alveolarization (SMD, 1.32, 95%CI [0.99, 1.65]), angiogenesis (SMD, 1.53, 95%CI [0.87, 2.18]), and anti-inflammatory cytokines (SMD, 1.68, 95%CI [1.03, 2.34]). In clinical studies, 103 preterm infants have received UCB-derived therapy for preterm lung injury and no significant difference was observed in the development of BPD (RR, 0.93, 95%CI [0.73, 1.18]). Across both preclinical and clinical studies, administration of UCB-derived therapy appeared safe. Certainty of evidence was assessed as "low." CONCLUSIONS Administration of UCB-derived therapy was associated with statistically significant improvements across several lung injury markers in preclinical studies. Early clinical studies demonstrated the administration of UCB-derived therapy as safe and feasible but lacked data regarding efficacy.
Collapse
Affiliation(s)
- Elisha Purcell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Jainam Shah
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Cameron Powell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Timothy Nguyen
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| |
Collapse
|
3
|
Ahn SY, Chang YS, Park WS. Stem cells for neonatal brain injury - Lessons from the bench. Semin Perinatol 2023; 47:151726. [PMID: 37003920 DOI: 10.1016/j.semperi.2023.151726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Neonatal brain injury resulting from various intractable disorders including intraventricular hemorrhage and hypoxic ischemic encephalopathy still remains a major cause of mortality and morbidities with few effective treatments. Recent preclinical research results showing the pleiotropic neuroprotective effects of stem cell therapy, specifically mesenchymal stem cells (MSCs), suggest that MSCs transplantation might be a promising new therapeutic modality for neuroprotection against the currently intractable and devastating neonatal brain injury with complex multifactorial etiology. This review summarizes recent advances in preclinical stem cell research for treating neonatal brain injury with a focus on the important issues including the mechanism of neuroprotection, and determining the ideal cell source, route, timing and dose of MSCs transplantation.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, South Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAHIST), Samsung Medical Center, Seoul 06351, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, South Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAHIST), Samsung Medical Center, Seoul 06351, South Korea.
| |
Collapse
|
4
|
Koivunotko E, Snirvi J, Merivaara A, Harjumäki R, Rautiainen S, Kelloniemi M, Kuismanen K, Miettinen S, Yliperttula M, Koivuniemi R. Angiogenic Potential of Human Adipose-Derived Mesenchymal Stromal Cells in Nanofibrillated Cellulose Hydrogel. Biomedicines 2022; 10:2584. [PMID: 36289846 PMCID: PMC9599553 DOI: 10.3390/biomedicines10102584] [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: 07/11/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASCs) hold great potential for cellular therapies by having immunomodulatory behavior and tissue regenerative properties. Due to the capability of ASCs to differentiate into endothelial cells (ECs) and other angiogenic cell types, such as pericytes, ASCs are a highly valuable source for stimulating angiogenesis. However, cellular therapies in tissue engineering have faced challenges in poor survival of the cells after transplantation, which is why a protective biomaterial scaffold is required. In this work, we studied the potential of nanofibrillated cellulose (NFC) hydrogel to be utilized as a suitable matrix for three-dimensional (3D) cell culturing of human-derived ASCs (hASCs) and studied their angiogenic properties and differentiation potential in ECs and pericytes. In addition, we tested the effect of hASC-conditioned medium and stimulation with angiopoietin-1 (Ang-1) on human umbilical vein endothelial cells (HUVECs) to induce blood vessel-type tube formation in NFC hydrogel. The hASCs were successfully 3D cell cultured in NFC hydrogel as they formed spheroids and had high cell viability with angiogenic features. Most importantly, they showed angiogenic potential by having pericyte-like characteristics when differentiated in EC medium, and their conditioned medium improved HUVEC viability and tube formation, which recalls the active paracrine properties. This study recommends NFC hydrogel for future use as an animal-free biomaterial scaffold for hASCs in therapeutic angiogenesis and other cell therapy purposes.
Collapse
Affiliation(s)
- Elle Koivunotko
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Jasmi Snirvi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Arto Merivaara
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Swarna Rautiainen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, 33520 Tampere, Finland
| | - Kirsi Kuismanen
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33520 Tampere, Finland
| | - Susanna Miettinen
- Faculty of Medicine and Health Technologies, University of Tampere, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Raili Koivuniemi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| |
Collapse
|
5
|
Sharma M, Bellio MA, Benny M, Kulandavelu S, Chen P, Janjindamai C, Han C, Chang L, Sterling S, Williams K, Damianos A, Batlahally S, Kelly K, Aguilar-Caballero D, Zambrano R, Chen S, Huang J, Wu S, Hare JM, Schmidt A, Khan A, Young K. Mesenchymal Stem Cell-derived Extracellular Vesicles Prevent Experimental Bronchopulmonary Dysplasia Complicated By Pulmonary Hypertension. Stem Cells Transl Med 2022; 11:828-840. [PMID: 35758326 PMCID: PMC9397655 DOI: 10.1093/stcltm/szac041] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 04/18/2022] [Indexed: 11/12/2022] Open
Abstract
Mesenchymal stem cell (MSC) extracellular vesicles (EVs) have beneficial effects in preclinical bronchopulmonary dysplasia and pulmonary hypertension (BPD-PH) models. The optimal source, dosing, route, and duration of effects are however unknown. The objectives of this study were to (a) compare the efficacy of GMP-grade EVs obtained from Wharton’s Jelly MSCs (WJ-MSCs) and bone marrow (BM-MSCs), (b) determine the optimal dosing and route of administration, (c) evaluate its long-term effects, and (d) determine how MSC EVs alter the lung transcriptome. Newborn rats exposed to normoxia or hyperoxia (85% O2) from postnatal day (P)1-P14 were given (a) intra-tracheal (IT) BM or WJ-MSC EVs or placebo, (b) varying doses of IT WJ-MSC EVs, or (c) IT or intravenous (IV) WJ-MSC EVs on P3. Rats were evaluated at P14 or 3 months. Early administration of IT BM-MSC or WJ-MSC EVs had similar beneficial effects on lung structure and PH in hyperoxia-exposed rats. WJ-MSC EVs however had superior effects on cardiac remodeling. Low, medium, and high dose WJ-MSC EVs had similar cardiopulmonary regenerative effects. IT and IV WJ-MSC EVs similarly improved vascular density and reduced PH in hyperoxic rats. Gene-set enrichment analysis of transcripts differentially expressed in WJ-MSC EV-treated rats showed that induced transcripts were associated with angiogenesis. Long-term studies demonstrated that a single early MSC EV dose has pulmonary vascular protective effects 3 months after administration. Together, our findings have significant translational implications as it provides critical insight into the optimal source, dosing, route, mechanisms of action, and duration of effects of MSC-EVs for BPD-PH.
Collapse
Affiliation(s)
- Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael A Bellio
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Chawisa Janjindamai
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Chenxu Han
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Liming Chang
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shanique Sterling
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin Williams
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andreas Damianos
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sunil Batlahally
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kaitlyn Kelly
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniela Aguilar-Caballero
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shaoyi Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karen Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.,Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
6
|
Benny M, Courchia B, Shrager S, Sharma M, Chen P, Duara J, Valasaki K, Bellio MA, Damianos A, Huang J, Zambrano R, Schmidt A, Wu S, Velazquez OC, Hare JM, Khan A, Young KC. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:189-199. [PMID: 35298658 PMCID: PMC8929420 DOI: 10.1093/stcltm/szab011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/17/2021] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a life-threatening condition in preterm infants with few effective therapies. Mesenchymal stem or stromal cells (MSCs) are a promising therapeutic strategy for BPD. The ideal MSC source for BPD prevention is however unknown. The objective of this study was to compare the regenerative effects of MSC obtained from bone marrow (BM) and umbilical cord tissue (UCT) in an experimental BPD model. In vitro, UCT-MSC demonstrated greater proliferation and expression of anti-inflammatory cytokines as compared to BM-MSC. Lung epithelial cells incubated with UCT-MSC conditioned media (CM) had better-wound healing following scratch injury. UCT-MSC CM and BM-MSC CM had similar pro-angiogenic effects on hyperoxia-exposed pulmonary microvascular endothelial cells. In vivo, newborn rats exposed to normoxia or hyperoxia (85% O2) from postnatal day (P) 1 to 21 were given intra-tracheal (IT) BM or UCT-MSC (1 × 106 cells/50 μL), or placebo (PL) on P3. Hyperoxia PL-treated rats had marked alveolar simplification, reduced lung vascular density, pulmonary vascular remodeling, and lung inflammation. In contrast, administration of both BM-MSC and UCT-MSC significantly improved alveolar structure, lung angiogenesis, pulmonary vascular remodeling, and lung inflammation. UCT-MSC hyperoxia-exposed rats however had greater improvement in some morphometric measures of alveolarization and less lung macrophage infiltration as compared to the BM-MSC-treated group. Together, these findings suggest that BM-MSC and UCT-MSC have significant lung regenerative effects in experimental BPD but UCT-MSC suppresses lung macrophage infiltration and promotes lung epithelial cell healing to a greater degree.
Collapse
Affiliation(s)
- Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Benjamin Courchia
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sebastian Shrager
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joanne Duara
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Krystalenia Valasaki
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael A Bellio
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andreas Damianos
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
- Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omaida C Velazquez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aisha Khan
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karen C Young
- Corresponding author: Karen C. Young, MD, Batchelor Children’s Research Institute, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, RM-345, Miami, FL 33136, USA. Tel: 305-243-4531;
| |
Collapse
|
7
|
Bagno LL, Salerno AG, Balkan W, Hare JM. Mechanism of Action of Mesenchymal Stem Cells (MSCs): impact of delivery method. Expert Opin Biol Ther 2021; 22:449-463. [PMID: 34882517 DOI: 10.1080/14712598.2022.2016695] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs; AKA mesenchymal stem cells) stimulate healing and reduce inflammation. Promising therapeutic responses are seen in many late-phase clinical trials, but others have not satisfied their primary endpoints, making translation of MSCs into clinical practice difficult. These inconsistencies may be related to the route of MSC delivery, lack of product optimization, or varying background therapies received in clinical trials over time. AREAS COVERED Here we discuss the different routes of MSC delivery, highlighting the proposed mechanism(s) of therapeutic action as well as potential safety concerns. PubMed search criteria used: MSC plus: local administration; routes of administration; delivery methods; mechanism of action; therapy in different diseases. EXPERT OPINION Direct injection of MSCs using a controlled local delivery approach appears to have benefits in certain disease states, but further studies are required to make definitive conclusions regarding the superiority of one delivery method over another.
Collapse
Affiliation(s)
- Luiza L Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| |
Collapse
|
8
|
Kadota T, Fujita Y, Araya J, Watanabe N, Fujimoto S, Kawamoto H, Minagawa S, Hara H, Ohtsuka T, Yamamoto Y, Kuwano K, Ochiya T. Human bronchial epithelial cell-derived extracellular vesicle therapy for pulmonary fibrosis via inhibition of TGF-β-WNT crosstalk. J Extracell Vesicles 2021; 10:e12124. [PMID: 34377373 PMCID: PMC8329991 DOI: 10.1002/jev2.12124] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 06/16/2021] [Accepted: 07/04/2021] [Indexed: 01/02/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by devastating and progressive lung parenchymal fibrosis, resulting in poor patient prognosis. An aberrant recapitulation of developmental lung gene expression, including genes for transforming growth factor (TGF)-β and WNT, has been widely implicated in the pathogenic IPF wound healing process that results from repetitive alveolar epithelial injury. Extracellular vesicles (EVs) have been shown to carry bioactive molecules and to be involved in various physiological and pathological processes. Here, we demonstrate that, by attenuating WNT signalling, human bronchial epithelial cell-derived EVs (HBEC EVs) inhibit TGF-β mediated induction of both myofibroblast differentiation and lung epithelial cellular senescence. This effect of HBEC EVs is more pronounced than that observed with mesenchymal stem cell-derived EVs. Mechanistically, the HBEC EV microRNA (miRNA) cargo is primarily responsible for attenuating both myofibroblast differentiation and cellular senescence. This attenuation occurs via inhibition of canonical and non-canonical WNT signalling pathways. Among enriched miRNA species present in HBEC EVs, miR-16, miR-26a, miR-26b, miR-141, miR-148a, and miR-200a are mechanistically involved in reducing WNT5A and WNT10B expression in LFs, and in reducing WNT3A, WNT5A, and WNT10B expression in HBECs. Mouse models utilizing intratracheal administration of EVs demonstrate efficient attenuation of bleomycin-induced lung fibrosis development accompanied by reduced expression of both β-catenin and markers of cellular senescence. These findings indicate that EVs derived from normal resident lung HBECs may possess anti-fibrotic properties. They further suggest that, via miRNA-mediated inhibition of TGF-β-WNT crosstalk, HBEC EVs administration can be a promising anti-fibrotic modality of treatment for IPF.
Collapse
Affiliation(s)
- Tsukasa Kadota
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Yu Fujita
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
- Department of Translational Research for ExosomesThe Jikei University School of MedicineTokyoJapan
| | - Jun Araya
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Naoaki Watanabe
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
- Division of Cellular SignalingNational Cancer Center Research InstituteTokyoJapan
| | - Shota Fujimoto
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Hironori Kawamoto
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Shunsuke Minagawa
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Hiromichi Hara
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Takashi Ohtsuka
- Division of Thoracic SurgeryDepartment of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Yusuke Yamamoto
- Division of Cellular SignalingNational Cancer Center Research InstituteTokyoJapan
| | - Kazuyoshi Kuwano
- Division of Respiratory DiseasesDepartment of Internal MedicineThe Jikei University School of MedicineTokyoJapan
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineInstitute of Medical ScienceTokyo Medical UniversityTokyoJapan
| |
Collapse
|
9
|
Tong Y, Zuo J, Yue D. Application Prospects of Mesenchymal Stem Cell Therapy for Bronchopulmonary Dysplasia and the Challenges Encountered. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9983664. [PMID: 33997051 PMCID: PMC8110410 DOI: 10.1155/2021/9983664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature babies, especially affecting those with very low or extremely low birth weights. Survivors experience adverse lung and neurological defects including cognitive dysfunction. This impacts the prognosis of children with BPD and may result in developmental delays. The currently available options for the treatment of BPD are limited owing to low efficacy or several side effects; therefore, there is a lack of effective treatments for BPD. The treatment for BPD must help in the repair of damaged lung tissue and promote further growth of the lung tissue. In recent years, the emergence of stem cell therapy, especially mesenchymal stem cell (MSC) therapy, has improved the treatment of BPD to a great extent. This article briefly reviews the advantages, research progress, and challenges faced with the use of MSCs in the treatment of BPD. Stem cell therapy is beneficial as it repairs damaged tissues by reducing inflammation, fibrosis, and by acting against oxidative stress damage. Experimental trials have also proven that MSCs provide a promising avenue for BPD treatment. However, there are challenges such as the possibility of MSCs contributing to tumorous growths, the presence of heterogeneous cell populations resulting in variable efficacy, and the ethical considerations regarding the use of this treatment in humans. Therefore, more research must be conducted to determine whether MSC therapy can be approved as a treatment option for BPD.
Collapse
Affiliation(s)
- Yajie Tong
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Jingye Zuo
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 Liaoning, China
| |
Collapse
|
10
|
Ahn SY, Chang YS, Lee MH, Sung SI, Lee BS, Kim KS, Kim AR, Park WS. Stem cells for bronchopulmonary dysplasia in preterm infants: A randomized controlled phase II trial. Stem Cells Transl Med 2021; 10:1129-1137. [PMID: 33876883 PMCID: PMC8284779 DOI: 10.1002/sctm.20-0330] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
We previously demonstrated the safety and feasibility of mesenchymal stem cell (MSC) transplantation for bronchopulmonary dysplasia (BPD) in preterm infants in a phase I clinical trial. We thus investigated the therapeutic efficacy of MSCs for BPD in premature infants. A phase II double-blind, randomized, placebo-controlled clinical trial was conducted on preterm infants at 23 to 28 gestational weeks (GW) receiving mechanical ventilator support with respiratory deterioration between postnatal days 5 and 14. Infants were stratified by 23 to 24 GW and 25 to 28 GW and randomly allocated (1:1) to receive stem cells (1 × 107 cells/kg, n = 33) or placebo (n = 33). Although the inflammatory cytokines in the tracheal aspirate fluid were significantly reduced with MSCs, the primary outcome of death or severe/moderate BPD in the control group (18/33, 55%) was not significantly improved with MSC transplantation (17/33, 52%). In the subgroup analysis, the secondary outcome of severe BPD was significantly improved from 53% (8/15) to 19% (3/16) with MSC transplantation in the 23 to 24 GW group but not in the 25 to 28 GW subgroup. In summary, although MSC transplantation might be safe and feasible, this small study was underpowered to detect its therapeutic efficacy in preterm infants at 23 to 28 GW. Accordingly, we are now conducting an additional larger and controlled phase II clinical trial focusing on infants at 23 to 24 GW (NCT03392467). ClinicalTrials.gov identifier: NCT01828957.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Myung Hee Lee
- Statistics and Data Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byong Sop Lee
- Department of Pediatrics, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Ki Soo Kim
- Department of Pediatrics, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Ai-Rhan Kim
- Department of Pediatrics, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Ahn SY, Park WS, Sung SI, Chang YS. Mesenchymal stem cell therapy for intractable neonatal disorders. Pediatr Neonatol 2021; 62 Suppl 1:S16-S21. [PMID: 33485822 DOI: 10.1016/j.pedneo.2020.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/30/2020] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has emerged as a new promising therapeutic strategy for the treatment of intractable and devastating neonatal disorders with complex multifactorial etiologies, including bronchopulmonary dysplasia (BPD), intraventricular hemorrhage (IVH), and hypoxic-ischemic encephalopathy (HIE). In response to inflammatory and noxious environments, MSCs secrete various paracrine factors that perform several reparative functions, including exerting anti-inflammatory, anti-oxidative, anti-apoptotic, and anti-fibrotic effects, to enhance the regeneration of damaged cells and tissues. In this review, we summarize recent advances in stem cell research focusing on the use of MSCs in the prevention and treatment of newborn BPD, IVH and HIE, with particular emphasis on preclinical and clinical data.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
12
|
Porzionato A, Zaramella P, Dedja A, Guidolin D, Bonadies L, Macchi V, Pozzobon M, Jurga M, Perilongo G, De Caro R, Baraldi E, Muraca M. Intratracheal administration of mesenchymal stem cell-derived extracellular vesicles reduces lung injuries in a chronic rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2021; 320:L688-L704. [PMID: 33502939 DOI: 10.1152/ajplung.00148.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Arben Dedja
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Diego Guidolin
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Michela Pozzobon
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Marcin Jurga
- The Cell Factory BVBA (Esperite NV), Niel, Belgium
| | - Giorgio Perilongo
- Institute of Pediatric Research, Padua, Italy.,Pediatric Clinic, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Padua, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women's and Children's Health, University of Padova, Padua, Italy.,Institute of Pediatric Research, Padua, Italy
| | - Maurizio Muraca
- Institute of Pediatric Research, Padua, Italy.,Stem Cell and Regenerative Medicine Laboratory, Department of Women's and Children's Health, University of Padova, Padua, Italy
| |
Collapse
|
13
|
MSC Based Therapies to Prevent or Treat BPD-A Narrative Review on Advances and Ongoing Challenges. Int J Mol Sci 2021; 22:ijms22031138. [PMID: 33498887 PMCID: PMC7865378 DOI: 10.3390/ijms22031138] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains one of the most devastating consequences of preterm birth resulting in life-long restrictions in lung function. Distorted lung development is caused by its inflammatory response which is mainly provoked by mechanical ventilation, oxygen toxicity and bacterial infections. Dysfunction of resident lung mesenchymal stem cells (MSC) represents one key hallmark that drives BPD pathology. Despite all progress in the understanding of pathomechanisms, therapeutics to prevent or treat BPD are to date restricted to a few drugs. The limited therapeutic efficacy of established drugs can be explained by the fact that they fail to concurrently tackle the broad spectrum of disease driving mechanisms and by the huge overlap between distorted signal pathways of lung development and inflammation. The great enthusiasm about MSC based therapies as novel therapeutic for BPD arises from the capacity to inhibit inflammation while simultaneously promoting lung development and repair. Preclinical studies, mainly performed in rodents, raise hopes that there will be finally a broadly acting, efficient therapy at hand to prevent or treat BPD. Our narrative review gives a comprehensive overview on preclinical achievements, results from first early phase clinical studies and challenges to a successful translation into the clinical setting.
Collapse
|
14
|
You J, Zhou O, Liu J, Zou W, Zhang L, Tian D, Dai J, Luo Z, Liu E, Fu Z, Zou L. Human Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Alleviate Lung Injury in Rat Model of Bronchopulmonary Dysplasia by Affecting Cell Survival and Angiogenesis. Stem Cells Dev 2020; 29:1520-1532. [PMID: 33040709 DOI: 10.1089/scd.2020.0156] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a serious chronic lung disease in premature newborns, with high morbidity and mortality rates. Mesenchymal stem cell (MSC) transplantation has developed into a promising approach to alleviate BPD. Small extracellular vesicles, which are an important therapeutic component of MSCs, have been reported to be effective in a mouse model of BPD. However, the affected cell types and detailed underlying mechanisms are unclear. In this study, we found that human umbilical cord mesenchymal stem cell-derived small extracellular vesicles (hucMSC-sEVs) were successfully absorbed by lung tissue after intratracheal administration, and remained in the lungs for at least 72 h. The results showed that hucMSC-sEVs restored alveolar structure and lung function, and ameliorated pulmonary hypertension in a rat model of BPD. The number of Ki-67-positive lung cells were improved, while the number of TUNEL-positive lung cells were reduced in our hucMSC-sEV-treated BPD model. Additionally, SP-C staining (a marker of type II alveolar epithelial cells, TIIAECs) and CD31 staining (a marker of pulmonary vascular endothelial cells, PVECs) were both increased in a hyperoxia-induced BPD model treated with hucMSC-sEVs. In vitro, under hyperoxic conditions, the tube-like structure formation was improved in human umbilical vein endothelial cells, and the proliferation was increased and the apoptosis was attenuated in MLE-12 cells treated with hucMSC-sEVs. Furthermore, we observed downregulated expression of PTEN and cleaved-caspase3, and upregulated expression of p-Akt and vascular endothelial growth factor-A in our hucMSC-sEV-treated BPD model. In conclusion, hucMSC-sEVs improved alveolarization and angiogenesis in a rat BPD model by protecting TIIAECs and PVECs, which were associated with the PTEN/Akt signaling pathway.
Collapse
Affiliation(s)
- Jingyi You
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Ou Zhou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Jiang Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Wenjing Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Linghuan Zhang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Daiyin Tian
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Jihong Dai
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Zhengxiu Luo
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Enmei Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Zhou Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China
| | - Lin Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, China.,Department of Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
De Paepe ME, Wong T, Chu S, Mao Q. Stromal cell-derived factor-1 (SDF-1) expression in very preterm human lungs: potential relevance for stem cell therapy for bronchopulmonary dysplasia. Exp Lung Res 2020; 46:146-156. [PMID: 32281423 DOI: 10.1080/01902148.2020.1751899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: The axis formed by CXC chemokine receptor 4 (CXCR4), expressed on mesenchymal stromal cells (MSCs), and stromal cell-derived factor-1 (SDF-1), expressed in recipient organs, is a critical mediator of MSC migration in non-pulmonary injury models. The role and regulation of SDF-1 expression in preterm lungs, of potential relevance for MSC-based cell therapy for bronchopulmonary dysplasia (BPD), is unknown. The aim of this study was to determine the spatiotemporal pattern of CXCR4/SDF-1 expression in lungs of extremely preterm infants at risk for BPD.Methods: Postmortem lung samples were collected from ventilated extremely preterm infants who died between 23 and 29 wks ("short-term ventilated") or between 36 and 39 wks ("long-term ventilated") corrected postmenstrual age. Results were compared with age-matched infants who had lived <12 h or stillborn infants ("early" and "late" controls). CXCR4 and SDF-1 expression was studied by immunohistochemistry, immunofluorescence/confocal microscopy, and qRT-PCR analysis.Results: Compared with age-matched controls without antenatal infection, lungs of early control infants with evidence of intrauterine infection/inflammation showed significant upregulation of SDF-1 expression, localized to the respiratory epithelium, and of CXCR4 expression, localized to stromal cells. Similarly, pulmonary SDF-1 mRNA levels were significantly higher in long-term ventilated ex-premature infants with established BPD than in age-matched controls. The pulmonary vasculature was devoid of SDF-1 expression at all time points. Endogenous CXCR4-positive stromal cells were preferentially localized along the basal aspect of SDF-1-positive bronchial and respiratory epithelial cells, suggestive of functionality of the CXCR4/SDF-1 axis.Conclusions: Incipient and established neonatal lung injury is associated with upregulation of SDF-1 expression, restricted to the respiratory epithelium. Knowledge of the clinical associations, time-course and localization of pulmonary SDF-1 expression may guide decisions about the optimal timing and delivery route of MSC-based cell therapy for BPD.
Collapse
Affiliation(s)
- Monique E De Paepe
- Department of Pathology and Laboratory Medicine, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Talia Wong
- Department of Pathology and Laboratory Medicine, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Sharon Chu
- Department of Pathology and Laboratory Medicine, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Quanfu Mao
- Department of Pathology and Laboratory Medicine, Women and Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Clinical trials of mesenchymal stem/stromal cell (MSC) therapy for bronchopulmonary dysplasia (BPD) are underway. A thorough understanding of the preclinical work that underpins these trials is critical for neonatal practitioners to properly evaluate them. RECENT FINDINGS Significant progress has been made in understanding that MSCs have anti-inflammatory and proangiogenic effects, and that these can be mediated by the noncellular exosome fraction of MSCs. SUMMARY In rodent hyperoxia models of BPD, MSCs have a proangiogenic effect mediated largely by vascular endothelial growth factor and shift the balance of endogenous lung cells from a proinflammatory to a prohealing phenotype. MSC-derived exosomes can recapitulate these effects.
Collapse
|
17
|
Behnke J, Kremer S, Shahzad T, Chao CM, Böttcher-Friebertshäuser E, Morty RE, Bellusci S, Ehrhardt H. MSC Based Therapies-New Perspectives for the Injured Lung. J Clin Med 2020; 9:jcm9030682. [PMID: 32138309 PMCID: PMC7141210 DOI: 10.3390/jcm9030682] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases pose a tremendous global burden. At least one in four people suffer from severe pulmonary sequelae over the course of a lifetime. Despite substantial improvements in therapeutic interventions, persistent alleviation of clinical symptoms cannot be offered to most patients affected to date. Despite broad discrepancies in origins and pathomechanisms, the important disease entities all have in common the pulmonary inflammatory response which is central to lung injury and structural abnormalities. Mesenchymal stem cells (MSC) attract particular attention due to their broadly acting anti-inflammatory and regenerative properties. Plenty of preclinical studies provided congruent and convincing evidence that MSC have the therapeutic potential to alleviate lung injuries across ages. These include the disease entities bronchopulmonary dysplasia, asthma and the different forms of acute lung injury and chronic pulmonary diseases in adulthood. While clinical trials are so far restricted to pioneering trials on safety and feasibility, preclinical results point out possibilities to boost the therapeutic efficacy of MSC application and to take advantage of the MSC secretome. The presented review summarizes the most recent advances and highlights joint mechanisms of MSC action across disease entities which provide the basis to timely tackle this global disease burden.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Sarah Kremer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Cho-Ming Chao
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), German Center for Lung Research (DZL), Aulweg 130, 35392 Giessen, Germany;
| | | | - Rory E. Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Ludwigstrasse 43, 61231 Bad Nauheim, Germany;
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), German Center for Lung Research (DZL), Aulweg 130, 35392 Giessen, Germany;
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
- Correspondence: ; Tel.: +49-985-43400; Fax: +49-985-43419
| |
Collapse
|
18
|
Moreira A, Winter C, Joy J, Winter L, Jones M, Noronha M, Porter M, Quim K, Corral A, Alayli Y, Seno T, Mustafa S, Hornsby P, Ahuja S. Intranasal delivery of human umbilical cord Wharton's jelly mesenchymal stromal cells restores lung alveolarization and vascularization in experimental bronchopulmonary dysplasia. Stem Cells Transl Med 2020; 9:221-234. [PMID: 31774626 PMCID: PMC6988765 DOI: 10.1002/sctm.18-0273] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating lung condition that develops in premature newborns exposed to prolonged mechanical ventilation and supplemental oxygen. Significant morbidity and mortality are associated with this costly disease and effective therapies are limited. Mesenchymal stem/stromal cells (MSCs) are multipotent cells that can repair injured tissue by secreting paracrine factors known to restore the function and integrity of injured lung epithelium and endothelium. Most preclinical studies showing therapeutic efficacy of MSCs for BPD are administered either intratracheally or intravenously. The purpose of this study was to examine the feasibility and effectiveness of human cord tissue-derived MSC administration given via the intranasal route. Human umbilical cord tissue MSCs were isolated, characterized, and given intranasally (500 000 cells per 20 μL) to a hyperoxia-induced rat model of BPD. Lung alveolarization, vascularization, and pulmonary vascular remodeling were restored in animals receiving MSC treatment. Gene and protein analysis suggest the beneficial effects of MSCs were attributed, in part, to a concerted effort targeting angiogenesis, immunomodulation, wound healing, and cell survival. These findings are clinically significant, as neonates who develop BPD have altered alveolar development, decreased pulmonary vascularization and chronic inflammation, all resulting in impaired tissue healing. Our study is the first to report the intranasal delivery of umbilical cord Wharton's jelly MSCs in experimental BPD is feasible, noninvasive, and an effective route that may bear clinical applicability.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Caitlyn Winter
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Jooby Joy
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Lauryn Winter
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Maxwell Jones
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Michelle Noronha
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Melissa Porter
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Kayla Quim
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Alexis Corral
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Yasmeen Alayli
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Tyrelle Seno
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Shamimunisa Mustafa
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Peter Hornsby
- Department of Pediatrics, Cellular and Integrative PhysiologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| | - Sunil Ahuja
- Microbiology and ImmunologyUniversity of Texas Health Science Center San Antonio (UTHSCSA)San AntonioTexas
| |
Collapse
|
19
|
Augustine S, Cheng W, Avey MT, Chan ML, Lingappa SMC, Hutton B, Thébaud B. Are all stem cells equal? Systematic review, evidence map, and meta-analyses of preclinical stem cell-based therapies for bronchopulmonary dysplasia. Stem Cells Transl Med 2020; 9:158-168. [PMID: 31746123 PMCID: PMC6988768 DOI: 10.1002/sctm.19-0193] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/10/2019] [Indexed: 12/25/2022] Open
Abstract
Regenerative stem cell-based therapies for bronchopulmonary dysplasia (BPD), the most common preterm birth complication, demonstrate promise in animals. Failure to objectively appraise available preclinical data and identify knowledge gaps could jeopardize clinical translation. We performed a systematic review and network meta-analysis (NMA) of preclinical studies testing cell-based therapies in experimental neonatal lung injury. Fifty-three studies assessing 15 different cell-based therapies were identified: 35 studied the effects of mesenchymal stromal cells (MSCs) almost exclusively in hyperoxic rodent models of BPD. Exploratory NMAs, for select outcomes, suggest that MSCs are the most effective therapy. Although a broad range of promising cell-based therapies has been assessed, few head-to-head comparisons and unclear risk of bias exists. Successful clinical translation of cell-based therapies demands robust preclinical experimental design with appropriately blinded, randomized, and statistically powered studies, based on biological plausibility for a given cell product, in standardized models and endpoints with transparent reporting.
Collapse
Affiliation(s)
- Sajit Augustine
- Division of NeonatologyWindsor Regional HospitalWindsorOntarioCanada
- Department of Pediatrics, Schulich Medicine & DentistryWestern UniversityLondonOntarioCanada
| | - Wei Cheng
- Ottawa Hospital Research InstituteOttawaOntarioCanada
| | | | - Monica L. Chan
- Department of NeonatologyChildren's Hospital of Eastern OntarioOttawaOntarioCanada
| | | | - Brian Hutton
- Ottawa Hospital Research InstituteOttawaOntarioCanada
- School of Epidemiology, Public Health and Preventive Medicine, Faculty of Medicine, University of OttawaOttawaOntarioCanada
| | - Bernard Thébaud
- Ottawa Hospital Research InstituteOttawaOntarioCanada
- Department of NeonatologyChildren's Hospital of Eastern OntarioOttawaOntarioCanada
- Department of PediatricsChildren's Hospital of Eastern Ontario Research Institute, University of OttawaOttawaOntarioCanada
| |
Collapse
|
20
|
Bao L, Shi Y. [Mesenchymal stem cell transplantation in the treatment of bronchopulmonary dysplasia: opportunities and challenges]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:619-623. [PMID: 31315757 PMCID: PMC7389107 DOI: 10.7499/j.issn.1008-8830.2019.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/06/2019] [Indexed: 06/10/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in neonates especially in preterm infants. It is also the main reason leading to a poor prognosis. The prognosis of the neonates with BPD is unsatisfactory with current treatment strategies. Recent clinical trails have found that mesenchymal stem cell (MSC) transplantation might be effective and promising for treatment of BPD in neonates. This article outlines the characteristics of MSC and the potential mechanisms of MSC transplantation for BPD in vivo, and the safety and feasibility of MSC transplantation in BPD neonates, as well as the challenges in clinical trials on MSC transplantation for treatment of BPD.
Collapse
Affiliation(s)
- Lei Bao
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China.
| | | |
Collapse
|
21
|
Mesenchymal Stem/Stromal Cell Therapy for Bronchopulmonary Dysplasia in the Neonatal Intensive Care Unit. CURRENT PEDIATRICS REPORTS 2019. [DOI: 10.1007/s40124-019-00198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
22
|
Moreira A, Alayli Y, Balgi S, Winter C, Kahlenberg S, Mustafa S, Hornsby P. Upcycling umbilical cords: bridging regenerative medicine with neonatology. J Matern Fetal Neonatal Med 2019; 32:1378-1387. [PMID: 29132234 PMCID: PMC6175672 DOI: 10.1080/14767058.2017.1405387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 11/11/2017] [Accepted: 11/12/2017] [Indexed: 02/06/2023]
Abstract
Preterm birth is a major health concern that affects 10% of all worldwide deliveries. Many preterm infants are discharged from the hospital with morbidities that lead to an increased risk for neurodevelopmental impairment, recurrent hospitalizations, and life-long conditions. Unfortunately, the treatment of these conditions is palliative rather than curative, which calls for novel and innovative strategies. Progress in regenerative medicine has offered therapeutic options for many of these conditions. Specifically, human umbilical cord mesenchymal stem cells (MSCs) and cord blood (UCB) cells have shown promise in treating adult-onset diseases. Unlike bone-marrow and embryonic derived stem cells, umbilical cord-derived cells are easily and humanely obtained, have low immunogenicity, and offer the potential of autologous therapy. While there are several studies to uphold the efficacy of umbilical cord MSCs in adult therapies, there remains an unmet need for the investigation of its use in treating neonates. The purpose of this review is to provide a summary of current information on the potential therapeutic benefits and clinical applicability of umbilical cord MSCs and UCB cells. Promising preclinical studies have now led to a research movement that is focusing on cell-based therapies for preterm infants.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Yasmeen Alayli
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Saloni Balgi
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Caitlyn Winter
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Samuel Kahlenberg
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Shamimunisa Mustafa
- Department of Pediatrics, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Peter Hornsby
- Department of Cellular and Integrative Physiology, University of Texas Health-San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| |
Collapse
|
23
|
Porzionato A, Zaramella P, Dedja A, Guidolin D, Van Wemmel K, Macchi V, Jurga M, Perilongo G, De Caro R, Baraldi E, Muraca M. Intratracheal administration of clinical-grade mesenchymal stem cell-derived extracellular vesicles reduces lung injury in a rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 316:L6-L19. [DOI: 10.1152/ajplung.00109.2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) prevent the onset of bronchopulmonary dysplasia (BPD) in animal models, an effect that seems to be mediated by their secreted extracellular vesicles (EVs). The aim of this study was to compare the protective effects of intratracheally (IT) administered MSCs versus MSC-EVs in a hyperoxia-induced rat model of BPD. At birth, rats were distributed as follows: animals raised in ambient air for 2 wk ( n = 10), and animals exposed to 60% oxygen for 2 wk and treated with IT-administered physiological solution ( n = 10), MSCs ( n = 10), or MSC-EVs ( n = 10) on postnatal days 3, 7, and 10. The sham-treated hyperoxia-exposed animals showed reductions in total surface area of alveolar air spaces, and total number of alveoli ( Nalv), and an increased mean alveolar volume (Valv). EVs prompted a significant increase in Nalv ( P < 0.01) and a significant decrease in Valv ( P < 0.05) compared with sham-treated animals, whereas MSCs only significantly improved Nalv ( P < 0.05). Small pulmonary vessels of the sham-treated hyperoxia-exposed rats also showed an increase in medial thickness, which only EVs succeeded in preventing significantly ( P < 0.05). In conclusion, both EVs and MSCs reduce hyperoxia-induced damage, with EVs obtaining better results in terms of alveolarization and lung vascularization parameters. This suggests that IT-administered EVs could be an effective approach to BPD treatment.
Collapse
Affiliation(s)
- Andrea Porzionato
- Human Anatomy Section, Department of Neurosciences, University of Padova, Padua, Italy
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, Padua, Italy
| | - Arben Dedja
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padua, Italy
| | - Diego Guidolin
- Human Anatomy Section, Department of Neurosciences, University of Padova, Padua, Italy
| | | | - Veronica Macchi
- Human Anatomy Section, Department of Neurosciences, University of Padova, Padua, Italy
| | - Marcin Jurga
- The Cell Factory BVBA (Esperite NV), Niel, Belgium
| | - Giorgio Perilongo
- Pediatric Clinic, Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Institute of Pediatric Research, “Città della Speranza,” Padua, Italy
| | - Raffaele De Caro
- Human Anatomy Section, Department of Neurosciences, University of Padova, Padua, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Institute of Pediatric Research, “Città della Speranza,” Padua, Italy
| | - Maurizio Muraca
- Institute of Pediatric Research, “Città della Speranza,” Padua, Italy
- Stem Cell and Regenerative Medicine Laboratory, Department of Women’s and Children’s Health, University of Padova, Padua, Italy
| |
Collapse
|
24
|
Ko HR, Ahn SY, Chang YS, Hwang I, Yun T, Sung DK, Sung SI, Park WS, Ahn JY. Human UCB-MSCs treatment upon intraventricular hemorrhage contributes to attenuate hippocampal neuron loss and circuit damage through BDNF-CREB signaling. Stem Cell Res Ther 2018; 9:326. [PMID: 30463591 PMCID: PMC6249960 DOI: 10.1186/s13287-018-1052-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/15/2022] Open
Abstract
Background Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) have been shown to prevent brain damage and improve neurocognition following intraventricular hemorrhage (IVH). However, the molecular mechanisms underlying the effects of hUCB-MSCs are still elusive. Thus, as the hippocampus is essential for learning, memory, and cognitive functions and is intimately involved in the ventricular system, making it a potential site of IVH-induced injury, we determined the molecular basis of the effects of hUCB-derived MSCs on hippocampal neurogenesis and the recovery of hippocampal neural circuits after IVH in a rodent model. Methods We inflicted severe IVH injury on postnatal day 4 (P4) in rats. After confirmation of successful induction of IVH using MRI (P5), intracerebroventricular administration of MSCs (ICV-MSC) was performed at 2 days post-injury (P6). For hippocampal synaptic determination, a rat entorhinal-hippocampus (EH) organotypic slice co-culture (OSC) was performed using day 3 post-IVH brains (P7) with or without ICV-MSCs. A similar strategy of experiments was applied to those rats receiving hUCB-MSC transfected with BDNF-Si-RNA for knockdown of BDNF or scrambled siRNA controls after IVH. The molecular mechanism of the MSCs effects on neurogenesis and the attenuation of neuron death was determined by evaluation of BDNF-TrkB-Akt-CREB signaling axis. Results We showed that treatment with hUCB-MSCs attenuated neuronal loss and promoted neurogenesis in the hippocampus, an area highly vulnerable to IVH-induced brain injury. hUCB-MSCs activate BDNF-TrkB receptor signaling, eliciting intracellular activation of Akt and/or Erk and subsequent phosphorylation of CREB, which is responsible for promoting rat BDNF transcription. In addition to the beneficial effects of neuroprotection and neurogenesis, hUCB-MSCs also contribute to the restoration of impaired synaptic circuits in the hippocampus and improve neurocognitive functions in IVH-injured neonatal rat through BDNF-TrkB-CREB signaling axis activation. Conclusions Our data suggest that hUCB-MSCs possess therapeutic potential for treating neuronal loss and neurocognitive dysfunction in IVH through the activation of intracellular TrkB-CREB signaling that is invoked by hUCB-MSC-secreted BDNF. Electronic supplementary material The online version of this article (10.1186/s13287-018-1052-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyo Rim Ko
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Taegwan Yun
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, South Korea. .,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea. .,Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwonro, Gangnam-gu, Seoul, 06351, South Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea. .,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
25
|
Kim YE, Park WS, Ahn SY, Sung DK, Chang YS. Intratracheal transplantation of mesenchymal stem cells attenuates hyperoxia-induced lung injury by down-regulating, but not direct inhibiting formyl peptide receptor 1 in the newborn mice. PLoS One 2018; 13:e0206311. [PMID: 30356317 PMCID: PMC6200259 DOI: 10.1371/journal.pone.0206311] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022] Open
Abstract
Formyl peptide receptor 1 (FPR1) has been shown to be a key regulator of inflammation. However, its role in bronchopulmonary dysplasia (BPD) has not been delineated yet. We investigated whether FPR1 plays a pivotal role in regulating lung inflammation and injuries, and whether intratracheally transplanted mesenchymal stem cells (MSCs) attenuate hyperoxic lung inflammation and injuries by down-regulating FPR1. Newborn wild type (WT) or FPR1 knockout (FPR1-/-) C57/BL6 mice were randomly exposed to 80% oxygen or room air for 14 days. At postnatal day (P) 5, 2×105 MSCs were intratracheally transplanted. At P14, mice were sacrificed for histopathological and morphometric analyses. Hyperoxia significantly increased lung neutrophils, macrophages, and TUNEL-positive cells, while impairing alveolarization and angiogenesis, along with a significant increase in FPR1 mRNA levels in WT mice. The hyperoxia-induced lung inflammation and lung injuries were significantly attenuated, with the reduced mRNA level of FPR1, in WT mice with MSC transplantation and in FPR1-/- mice, irrespective of MSCs transplantation. However, only MSC transplantation, but not the FPR1 knockout, significantly attenuated the hyperoxia-induced increase in TUNEL-positive cells. Our findings indicate that FPR1 play a critical role in regulating lung inflammation and injuries in BPD, and MSCs attenuate hyperoxic lung inflammation and injuries, but not apoptosis, with down regulating, but not direct inhibiting FPR1.
Collapse
Affiliation(s)
- Young Eun Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Korea
| | - Won Soon Park
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Korea
- Department of Pediatrics, Samsung Medical Center, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Korea
- Department of Pediatrics, Samsung Medical Center, Seoul, Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun Sil Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Korea
- Department of Pediatrics, Samsung Medical Center, Seoul, Korea
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
26
|
Off-label mesenchymal stromal cell treatment in two infants with severe bronchopulmonary dysplasia: clinical course and biomarkers profile. Cytotherapy 2018; 20:1337-1344. [PMID: 30327248 DOI: 10.1016/j.jcyt.2018.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the most prevalent sequelae of premature birth, for which therapeutic options are currently limited. Mesenchymal stromal cells (MSCs) are a potential therapy for prevention or reversal of BPD. SERIES OF CASES We report on two infants with severe BPD in whom off-label treatment with repeated intravenous doses of allogeneic bone marrow-derived MSCs were administered. We analyzed the temporal profile of serum and tracheal cytokines and growth factors as well as safety, tolerability and clinical response. The administration of repeated intravenous doses of MSCs in two human babies with severe and advanced BPD was feasible and safe and was associated with a decrease of pro-inflammatory molecules and lung injury biomarkers. Both patients were at very advanced stages of BPD with very severe lung fibrosis and did not survive the disease. CONCLUSIONS MSCs are a promising therapy for BPD, but they should be administered in early stages of the disease.
Collapse
|
27
|
Bjørge IM, Kim SY, Mano JF, Kalionis B, Chrzanowski W. Extracellular vesicles, exosomes and shedding vesicles in regenerative medicine - a new paradigm for tissue repair. Biomater Sci 2018; 6:60-78. [PMID: 29184934 DOI: 10.1039/c7bm00479f] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration by stem cells is driven by the paracrine activity of shedding vesicles and exosomes, which deliver specific cargoes to the recipient cells. Proteins, RNA, cytokines and subsequent gene expression, orchestrate the regeneration process by improving the microenvironment to promote cell survival, controlling inflammation, repairing injury and enhancing the healing process. The action of microRNA is widely accepted as an essential driver of the regenerative process through its impact on multiple downstream biological pathways, and its ability to regulate the host immune response. Here, we present an overview of the recent potential uses of exosomes for regenerative medicine and tissue engineering. We also highlight the differences in composition between shedding vesicles and exosomes that depend on the various types of stem cells from which they are derived. The conditions that affect the production of exosomes in different cell types are deliberated. This review also presents the current status of candidate exosomal microRNAs for potential therapeutic use in regenerative medicine, and in applications involving widely studied organs and tissues such as heart, lung, cartilage and bone.
Collapse
Affiliation(s)
- I M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | | | | | | | | |
Collapse
|
28
|
The Potentials and Caveats of Mesenchymal Stromal Cell-Based Therapies in the Preterm Infant. Stem Cells Int 2018; 2018:9652897. [PMID: 29765429 PMCID: PMC5911321 DOI: 10.1155/2018/9652897] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/04/2018] [Indexed: 02/06/2023] Open
Abstract
Preponderance of proinflammatory signals is a characteristic feature of all acute and resulting long-term morbidities of the preterm infant. The proinflammatory actions are best characterized for bronchopulmonary dysplasia (BPD) which is the chronic lung disease of the preterm infant with lifelong restrictions of pulmonary function and severe consequences for psychomotor development and quality of life. Besides BPD, the immature brain, eye, and gut are also exposed to inflammatory injuries provoked by infection, mechanical ventilation, and oxygen toxicity. Despite the tremendous progress in the understanding of disease pathologies, therapeutic interventions with proven efficiency remain restricted to a few drug therapies with restricted therapeutic benefit, partially considerable side effects, and missing option of applicability to the inflamed brain. The therapeutic potential of mesenchymal stromal cells (MSCs)—also known as mesenchymal stem cells—has attracted much attention during the recent years due to their anti-inflammatory activities and their secretion of growth and development-promoting factors. Based on a molecular understanding, this review summarizes the positive actions of exogenous umbilical cord-derived MSCs on the immature lung and brain and the therapeutic potential of reprogramming resident MSCs. The pathomechanistic understanding of MSC actions from the animal model is complemented by the promising results from the first phase I clinical trials testing allogenic MSC transplantation from umbilical cord blood. Despite all the enthusiasm towards this new therapeutic option, the caveats and outstanding issues have to be critically evaluated before a broad introduction of MSC-based therapies.
Collapse
|
29
|
Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatr Res 2018; 83:214-222. [PMID: 28972960 DOI: 10.1038/pr.2017.249] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation represents the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies, including bronchopulmonary dysplasia, hypoxic ischemic encephalopathy, and intraventricular hemorrhage. Absent engraftment and direct differentiation of transplanted MSCs, and the "hit-and-run" therapeutic effects of these MSCs suggest that their pleiotropic protection might be attributable to paracrine activity via the secretion of various biologic factors rather than to regenerative activity. The transplanted MSCs, therefore, exert their therapeutic effects not by acting as "stem cells," but rather by acting as "paracrine factors factory." The MSCs sense the microenvironment of the injury site and secrete various paracrine factors that serve several reparative functions, including antiapoptotic, anti-inflammatory, antioxidative, antifibrotic, and/or antibacterial effects in response to environmental cues to enhance regeneration of the damaged tissue. Therefore, the therapeutic efficacy of MSCs might be dependent on their paracrine potency. In this review, we focus on recent investigations that elucidate the specifically regulated paracrine mechanisms of MSCs by injury type and discuss potential strategies to enhance paracrine potency, and thus therapeutic efficacy, of transplanted MSCs, including determining the appropriate source and preconditioning strategy for MSCs and the route and timing of their administration.
Collapse
|
30
|
Ee MT, Thébaud B. The Therapeutic Potential of Stem Cells for Bronchopulmonary Dysplasia: "It's About Time" or "Not so Fast" ? Curr Pediatr Rev 2018; 14:227-238. [PMID: 30205800 PMCID: PMC6416190 DOI: 10.2174/1573396314666180911100503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/09/2018] [Accepted: 09/10/2018] [Indexed: 12/23/2022]
Abstract
OBJECTIVE While the survival of extremely premature infants has improved over the past decades, the rate of complications - especially for bronchopulmonary dysplasia (BPD) - remains unacceptably high. Over the past 50 years, no safe therapy has had a substantial impact on the incidence and severity of BPD. METHODS This may stem from the multifactorial disease pathogenesis and the increasing lung immaturity. Mesenchymal Stromal Cells (MSCs) display pleiotropic effects and show promising results in neonatal rodents in preventing or rescuing lung injury without adverse effects. Early phase clinical trials are now underway to determine the safety and efficacy of this therapy in extremely premature infants. RESULTS AND CONCLUSION This review summarizes our current knowledge about MSCs, their mechanism of action and the results of preclinical studies that provide the rationale for early phase clinical trials and discuss remaining gaps in our knowledge.
Collapse
Affiliation(s)
- Mong Tieng Ee
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON, Canada
| | - Bernard Thébaud
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON, Canada.,Sinclair Centre for Regenerative Medicine, Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
31
|
Liem NT, Anh TL, Thai TTH, Anh BV. Bone Marrow Mononuclear Cells Transplantation in Treatment of Established Bronchopulmonary Dysplasia: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2017; 18:1090-1094. [PMID: 29021519 PMCID: PMC5652889 DOI: 10.12659/ajcr.905244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Patient: Male, newborn Final Diagnosis: Bronchopulmonary displasia Symptoms: Difficult to breath • patient could not wean from oxygen/premature Medication: — Clinical Procedure: Bone marrow mononuclear cells transplantation Specialty: Pulmonology
Collapse
Affiliation(s)
- Nguyen Thanh Liem
- Department of Stem Cell and Immune Cell, inmec Research Institute of Stem Cells and Gene Technology, Hanoi, Vietnam
| | - Tran Lien Anh
- Department of Neonatology, Vinmec International Hospital, Hanoi, Vietnam
| | - Trieu T Hong Thai
- Department of Neonatology, Vinmec International Hospital, Hanoi, Vietnam
| | - Bui Viet Anh
- Department of Stem Cell and Immune Cell, Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| |
Collapse
|
32
|
Stromal derived factor-1 mediates the lung regenerative effects of mesenchymal stem cells in a rodent model of bronchopulmonary dysplasia. Respir Res 2017; 18:137. [PMID: 28701189 PMCID: PMC5506612 DOI: 10.1186/s12931-017-0620-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/03/2017] [Indexed: 02/08/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) attenuate lung injury in experimental models of bronchopulmonary dysplasia (BPD). Stromal derived factor-1 (SDF-1), a chemokine secreted by MSCs, modulates angiogenesis and stem cell recruitment. Here we tested the hypothesis that SDF-1 mediates MSC protective effects in experimental BPD by modulating angiogenesis. Methods SDF-1 was knocked down in MSCs using lentiviral vectors carrying anti-SDF-1 short hairpin RNA (MSC-SDF KD). Non-silencing short hairpin RNA was used as control (MSC-NS control). Newborn rats exposed to normoxia or hyperoxia (FiO2 = 0.85) for 3 weeks, were randomly assigned to receive a single intra-tracheal injection (IT) of MSC-NS control or MSC-SDF KD (1 × 106 cells/50 μl) or placebo on postnatal day 7. The degree of alveolarization, lung angiogenesis, inflammation, and pulmonary hypertension (PH) were assessed at postnatal day 21. Results Administration of IT MSC-NS control improved lung alveolarization, angiogenesis and inflammation, and attenuated PH in newborn rats with hyperoxia-induced lung injury (HILI). In contrast, knockdown of SDF-1 in MSCs significantly reduced their beneficial effects on alveolarization, angiogenesis, inflammation and PH. Conclusions The therapeutic benefits of MSCs in neonatal HILI are in part mediated by SDF-1, through anti-inflammatory and angiogenesis promoting mechanisms. Therapies directly targeting this chemokine may provide a novel strategy for the treatment of BPD.
Collapse
|
33
|
Ahn SY, Chang YS, Kim JH, Sung SI, Park WS. Two-Year Follow-Up Outcomes of Premature Infants Enrolled in the Phase I Trial of Mesenchymal Stem Cells Transplantation for Bronchopulmonary Dysplasia. J Pediatr 2017; 185:49-54.e2. [PMID: 28341525 DOI: 10.1016/j.jpeds.2017.02.061] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/30/2016] [Accepted: 02/22/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To determine the long-term safety and outcomes of mesenchymal stem cells (MSCs) for bronchopulmonary dysplasia in premature infants enrolled in a previous phase I clinical trial up to 2 years of corrected age (CA). STUDY DESIGN We assessed serious adverse events, somatic growth, and respiratory and neurodevelopmental outcomes at visit 1 (4-6 months of CA), visit 2 (8-12 months of CA), and visit 3 (18-24 months of CA) in a prospective longitudinal follow-up study up to 2 years' CA of infants who received MSCs (MSC group). We compared these data with those from a historical case-matched comparison group. RESULTS One of 9 infants in the MSC group died of Enterobacter cloacae sepsis at 6 months of CA, the remaining 8 infants survived without any transplantation-related adverse outcomes, including tumorigenicity. No infant in the MSC group was discharged with home supplemental oxygen compared with 22% in the comparison group. The average rehospitalization rate in the MSC group was 1.4/patient because of respiratory infections during 2 years of follow-up. The mean body weight of the MSC group at visit 3 was significantly higher compared with that of the comparison group. No infant in the MSC group was diagnosed with cerebral palsy, blindness, or developmental delay; in the comparison group, 1 infant was diagnosed with cerebral palsy and 1 with developmental delay. CONCLUSIONS Intratracheal transplantation of MSCs in preterm infants appears to be safe, with no adverse respiratory, growth, and neurodevelopmental effects at 2 years' CA. TRIAL REGISTRATION ClinicalTrials.gov: NCT01632475.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji Hye Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Republic of Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, Republic of Korea; Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Lesage F, Jimenez J, Toelen J, Deprest J. Preclinical evaluation of cell-based strategies to prevent or treat bronchopulmonary dysplasia in animal models: a systematic review. J Matern Fetal Neonatal Med 2017; 31:958-966. [PMID: 28277906 DOI: 10.1080/14767058.2017.1301927] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common complication of extreme prematurity as no effective treatment is available to date. This calls for the exploration of new therapeutic options like cell therapy, which is already effective for various human (lung) disorders. We systematically searched the MEDLINE, Embase, and Web of Science databases from the earliest date till January 2017 and included original studies on the perinatal use of cell-based therapies (i.e. cells and/or cell-derivatives) to treat BDP in animal models. Fourth publications describing 47 interventions were retrieved. Newborn mice/rats raised in a hyperoxic environment were studied in most interventions. Different cell types - either intact cells or their conditioned medium - were administered, but bone marrow and umbilical cord blood derived mesenchymal stem cells were most prevalent. All studies reported positive effects on outcome parameters including alveolar and vascular morphometry, lung function, and inflammation. Cell homing to the lungs was demonstrated in some studies, but the therapeutic effects seemed to be mostly mediated via paracrine modulation of inflammation, fibrosis and angiogenesis. CONCLUSION Multiple rat/mouse studies show promise for cell therapy for BPD. Yet careful study of action mechanisms and side effects in large animal models is imperative before clinical translation can be achieved.
Collapse
Affiliation(s)
- Flore Lesage
- a Department of Development and Regeneration, Group Biomedical Sciences , KU Leuven , Leuven , Belgium
| | - Julio Jimenez
- a Department of Development and Regeneration, Group Biomedical Sciences , KU Leuven , Leuven , Belgium.,b Department of Obstetrics and Gynaecology , Clínica Alemana Universidad del Desarrollo , Santiago , Chile
| | - Jaan Toelen
- a Department of Development and Regeneration, Group Biomedical Sciences , KU Leuven , Leuven , Belgium.,c Department of Pediatrics , University Hospitals Leuven , Leuven , Belgium
| | - Jan Deprest
- a Department of Development and Regeneration, Group Biomedical Sciences , KU Leuven , Leuven , Belgium.,d Research Department of Maternal Fetal Medicine , UCL Institute for Women's Health (IWH), University College London , London , United Kingdom
| |
Collapse
|
35
|
Abstract
Despite recent advances in neonatal medicine, neonatal disorders, such as bronchopulmonary dysplasia and intraventricular hemorrhage in preterm neonates and hypoxic ischemic encephalopathy in term neonates, remain major causes of mortality and morbidities. Promising preclinical research results suggest that stem cell therapies represent the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies. This review focuses primarily on the potential role of stem cell therapy in the above mentioned neonatal disorders, highlighting the results of human clinical trials and the challenges that remain to be addressed for their safe and successful translation into clinical care of newborn infants.
Collapse
Affiliation(s)
- Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sein Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
36
|
Park WS, Ahn SY, Sung SI, Ahn JY, Chang YS. Mesenchymal Stem Cells: The Magic Cure for Intraventricular Hemorrhage? Cell Transplant 2016; 26:439-448. [PMID: 27938484 DOI: 10.3727/096368916x694193] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Severe intraventricular hemorrhage (IVH) remains a major cause of mortality and long-term neurologic morbidities in premature infants, despite recent advances in neonatal intensive care medicine. Several preclinical studies have demonstrated the beneficial effects of mesenchymal stem cell (MSC) transplantation in attenuating brain injuries resulting from severe IVH. Because there currently exists no effective intervention for severe IVH, the therapeutic potential of MSC transplantation in this intractable and devastating disease is creating excitement in this field. This review summarizes recent progress in stem cell research for treating neonatal brain injury due to severe IVH, with a particular focus on preclinical data concerning important issues, such as mechanism of protective action and determining optimal source, route, timing, and dose of MSC transplantation, and on the translation of these preclinical study results to a clinical trial.
Collapse
|
37
|
Intratracheal transplantation of mesenchymal stem cells simultaneously attenuates both lung and brain injuries in hyperoxic newborn rats. Pediatr Res 2016; 80:415-24. [PMID: 27064241 DOI: 10.1038/pr.2016.88] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/20/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia is an independent risk factor for adverse neurodevelopmental outcomes in premature infants. We investigated whether attenuation of hyperoxic lung injury with intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells (MSCs) could simultaneously mitigate brain damage in neonatal rats. METHODS Newborn Sprague-Dawley rats were exposed to hyperoxia or normoxia conditions for 14 d. MSCs (5 × 10(5) cells) were transplanted intratracheally at postnatal day (P) 5. At P14, lungs and brains were harvested for histological and biochemical analyses. RESULTS Hyperoxic lung injuries, such as impaired alveolarization evident from increased mean linear intercept (MLI) and elevated inflammatory cytokine levels were significantly alleviated with MSC transplantation. Hyperoxia decreased brain weight, increased brain cell death, and induced hypomyelination. MSC transplantation significantly ameliorated hyperoxia-induced increased terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells in the dentate gyrus and reduced myelin basic protein. In correlation analyses, brain weight and myelin basic protein (MBP) were significantly inversely correlated with lung MLI and inflammatory cytokines, while TUNEL-positive brain cell number showed a significant positive correlation with lung MLI. CONCLUSION Despite no significant improvement in short-term neurofunctional outcome, intratracheal transplantation of MSCs simultaneously attenuated hyperoxic lung and brain injuries in neonatal rats, with the extent of such attenuation being closely linked in the two tissues.
Collapse
|
38
|
Qiang Y, Liang G, Yu L. Human amniotic mesenchymal stem cells alleviate lung injury induced by ischemia and reperfusion after cardiopulmonary bypass in dogs. J Transl Med 2016; 96:537-46. [PMID: 26927516 DOI: 10.1038/labinvest.2016.37] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/06/2016] [Accepted: 01/18/2016] [Indexed: 12/20/2022] Open
Abstract
Transplantation of mesenchymal stem cells may inhibit pathological immune processes contributing to ischemia/reperfusion (I/R) injury. This study aimed to assess the capacity of human amniotic MSC (hAMSCs) to ameliorate I/R injury in a dog model of cardiopulmonary bypass (CPB). Dissociated hAMSCs were cultured ex vivo, and their immunophenotypes were assessed by flow cytometry and immunohistochemistry. A dog model of CPB was established by surgical blockage of the aorta for 1 h. Dogs either underwent mock surgery (Sham group), CPB (model group), or CPB, followed by femoral injection of 2 × 10(7) hAMSCs (n=6). Anti-human nuclei staining revealed hAMSCs in the lungs 3 h after surgery. Oxygen index (OI) and respiratory index (RI) of arterial blood were measured using a biochemical analyzer. Venous blood TNF-α, IL-8, MMP-9, and IL-10 concentrations were measured by ELISA. Pathological changes in the lung were assessed by light microscopy. Third-generation-cultured hAMSCs expressed high levels of CD29, CD44, CD49D, CD73, and CD166 levels, but low CD34 or CD45 amounts and their cytoplasm contained Vimentin. In CPB model animals, OI was elevated and RI reduced; TNF-α, IL-8, and MMP-9 levels were elevated, and IL-10 levels reduced within 3h (P<0.05), but hAMSC transplantation significantly ameliorated these changes (P<0.05). Pathological changes observed in the hAMSC group were significantly less severe than those in the CPB group. In conclusion, hAMSC transplantation can downregulate proinflammatory factors and reduce MMP-9 levels, whereas upregulating the anti-inflammatory molecule IL-10, thus reducing I/R lung injury in a dog model of CPB.
Collapse
Affiliation(s)
- Yong Qiang
- Department of Cardiothoracic Surgery, Nanjing General Hospital of Nanjing Military Command, Nanjing, Jiangsu Province, China
| | - Guiyou Liang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Limei Yu
- Key Laboratory of Cell Engineering in Guizhou Province, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
39
|
Equal distribution of mesenchymal stem cells after hepatic ischemia-reperfusion injury. J Surg Res 2016; 203:360-7. [PMID: 27363644 DOI: 10.1016/j.jss.2016.03.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/15/2016] [Accepted: 03/24/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND Liver ischemia-reperfusion (I/R) injury is one of the major causes of hepatocellular injury-related mortality and morbidity after liver transplantation. Mesenchymal stem cells (MSCs) have been shown to reduce liver I/R injury and improve regeneration. The purpose of the present study was to investigate the difference in the distribution of systemically delivered MSCs in the recipient's liver between the ischemic injury area and nonischemic area. MATERIAL AND METHODS Fishers' rats (7-8 week of age) were used as donors of MSCs and recipients. Bone marrow-derived MSCs were isolated from the donor's femur. Before systemic administration, MSCs were labeled with the fluorescent dye PKH26. The rats were divided into four groups: (1) I/R injury + MSC group, (2) MSC only, without I/R injury, (3) I/R injury + saline group, and (4) the Sham group. I/R injury was performed by clamping the inflow vascular structures of the left and middle lobes of the recipient's liver for 60 min. The right lobe was considered as a nonischemic part. Subsequently, 1.5 × 10(6) of MSCs or saline (NaCl, 0.9%) was administrated via the rat's tail vein. Thereafter, the rats were killed after days one, three, or seven for the analyses. RESULTS A fluorescent microscopy assay for labeled MSCs showed positive cells in both ischemic and nonischemic parts of the recipient's liver. The number of cells was significantly higher in the I/R injury + MSC group compared with the only MSC, without I/R injury group. Immunohistochemical staining showed that there was no significant difference in the proliferation of Ki-67-positive cells between the I/R + MSCs and I/R + saline groups. In addition, the serum transaminase levels were not different between the I/R + MSCs and I/R + saline groups. CONCLUSIONS After partial liver I/R injury, transplanted MSCs migrate equally to the ischemic and nonischemic parts of the recipient's liver. Considering the unique ability of the liver to regenerate, both parts of the liver presumably receive signals for regeneration.
Collapse
|
40
|
Mitsialis SA, Kourembanas S. Stem cell-based therapies for the newborn lung and brain: Possibilities and challenges. Semin Perinatol 2016; 40:138-51. [PMID: 26778234 PMCID: PMC4808378 DOI: 10.1053/j.semperi.2015.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There have been substantial advances in neonatal medical care over the past 2 decades that have resulted in the increased survival of very low birth weight infants, survival that in some centers extends to 22 weeks gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term pulmonary and neurologic consequences. No single therapy has proven to be effective in preventing or treating either developmental lung and brain injuries in preterm infants or the hypoxic-ischemic injury that can be inflicted on the full-term brain as a result of in utero or perinatal complications. Stem cell-based therapies are emerging as a potential paradigm-shifting approach for such complex diseases with multifactorial etiologies, but a great deal of work is still required to understand the role of stem/progenitor cells in normal development and in the repair of injured tissue. This review will summarize the biology of the various stem/progenitor cells, their effects on tissue repair in experimental models of lung and brain injury, the recent advances in our understanding of their mechanism of action, and the challenges that remain to be addressed before their eventual application to clinical care.
Collapse
|
41
|
Ghadiri M, Young PM, Traini D. Cell-based therapies for the treatment of idiopathic pulmonary fibrosis (IPF) disease. Expert Opin Biol Ther 2015; 16:375-87. [PMID: 26593230 DOI: 10.1517/14712598.2016.1124085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION During the last few decades, cell-based therapies have shown great potential to treat patients with lung diseases. It has been proposed that the administration of cells into an injured lung could be considered as a therapeutic method to repair and replace lost lung tissue. Using this method, transplanted cells with the ability to proliferate and differentiate into alveolar cells, have been suggested as a therapeutic strategy for IPF treatment. AREAS COVERED In this review, the latest investigations using various types of cells for IPF therapy have been presented. The cells studied for cell-based therapies in IPF are lung alveolar epithelial cells, lung resident stem cells and exogenous adult stem cells such as MSCs. EXPERT OPINION After many years of investigation, the use of cell-based therapies to treat IPF is still at the experimental phase. Problems include bioethical issues, safety of cell transplantation, routes of delivery and the dose and timing of administration. Further investigations are necessary to establish the best strategy for using cell-based therapies effectively for the treatment of IPF.
Collapse
Affiliation(s)
- Maliheh Ghadiri
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Paul M Young
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Daniela Traini
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| |
Collapse
|