1
|
Korkmaz L, Alan C, Topal İ, Tayfur M, Bozkurt AS, Gürsul C, Baştuğ O. Can amniotic fluid protect developing fetal lungs against the harmful effects of oxidative stress? Turk J Med Sci 2023; 53:109-120. [PMID: 36945927 PMCID: PMC10387876 DOI: 10.55730/1300-0144.5564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/18/2022] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Preterm births cause fetuses to be born without completing the development of their organs. Due to this undesirable situation, it is the pulmonary tissue which has to be most exposed to harmful effects of extrauterine environment. Early disappearance of the prophylactic and constructive effects of amniotic fluid (AF) on developing tissues, such as pulmonary tissue, facilitates the formation of pulmonary morbidities resulting from oxygen. Setting out from this knowledge, we wanted, in addition to assessing the beneficent effects of AF on pulmonary tissue, to study the importance of AF in morbidities of this tissue thought to originate from oxygen. METHODS In this experimental study, while the study group was made up of the fetuses of pregnant rats exposed to hyperbaric oxygen, (hyperoxic pregnant rat fetuses-HPRF), the control group was formed of the fetuses of the rats pregnant in the usual room setting (normoxic pregnant rat fetuses-NPRF). The pulmonary and hepatic tissues taken from the fetuses of these pregnant rats on the 21st day of their pregnancy were compared biochemically and histologically. For biochemical assessment, total glutathione (tGSH), catalase (CAT), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α) values and for histopathological assessment, apoptosis, alveolar wall count (AWC), vena centralis count (VCC) were included. RESULTS Statistical significance was found in the pulmonary tissue values of tGSH on behalf of NPRF, and MDA on behalf of HPRF (p < 0.05). In liver tissue, statistical significance was detected in tGSH and CAT values in favor of NPRF and in MDA, and TNF-α values in favor of HPRF (p < 0.05). DISCUSSION : Our study has demonstrated that AF protects the pulmonary tissue from the harmful effects of oxygen in the intrauterine period. In addition, our data have suggested that the pulmonary tissue's being deprived of the useful effects of AF owing to premature birth may be an important trigger in the occurrence of the pulmonary morbidities thought to result from oxygen.
Collapse
Affiliation(s)
- Levent Korkmaz
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Cumali Alan
- Department of Pediatrics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - İsmail Topal
- Department of Pediatrics, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Mahir Tayfur
- Department of Pathology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Ali Seydi Bozkurt
- Department of Urology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Cebrail Gürsul
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Osman Baştuğ
- Division of Neonatology, Department of Pediatrics, Kayseri Training and Research Hospital, Kayseri, Turkey
| |
Collapse
|
2
|
Tao X, Mo L, Zeng L. Hyperoxia Induced Bronchopulmonary Dysplasia-Like Inflammation via miR34a-TNIP2-IL-1β Pathway. Front Pediatr 2022; 10:805860. [PMID: 35433535 PMCID: PMC9005975 DOI: 10.3389/fped.2022.805860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
Lung injury induced by oxygen is a key contributor to the pathogenesis of preterm infant bronchopulmonary dysplasia (BPD). To date, there are comprehensive therapeutic strategy for this disease, but the underlying mechanism is still in progress. By using lentivirus, we constructed microRNA34a (miR34a)-overexpressing or knockdown A549 cell lines, and exposure to hyperoxia to mimic oxygen induce lung injury. In this study, we investigated 4 proinflammatory cytokines, interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), angiopoietin-1 (Ang-1), and Cyclooxygenase-2 (COX-2) in the secreted sputum of infants who received mechanical ventilation, and found that IL-1β was substantially elevated in the first week after oxygen therapy and with no significant decrease until the fourth week, while TNF-α, Ang-1, and COX-2 were increased in the first week but decreased quickly in the following weeks. In addition, in vitro assay revealed that hyperoxia significantly increased the expression of miR-34a, which positively regulated the proinflammatory cytokine IL-1β in a time- and concentration-dependent manner in A549 cells. Overexpressing or knockdown miR34 would exacerbate or inhibit production of IL-1β and its upstream NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome signaling pathway. Mechanically, it's found that TNFAIP3 interacting protein 2 (TNIP2), an inhibitor of nuclear factor κB (NF-κB), is a direct target of miR34a, negatively regulated activation of NLRP3 inflammasome and the production of IL-1β. Overexpressing TNIP2 ameliorated hyperoxia-induced production of IL-1β and cell apoptosis. Our findings suggest that TNIP2 may be a potential clinical marker in the diagnosis of BPD.
Collapse
Affiliation(s)
- Xuwei Tao
- Department of Neonatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luxia Mo
- Department of Neonatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingkong Zeng
- Department of Neonatology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Chalphin AV, Lazow SP, Labuz DF, Tracy SA, Kycia I, Zurakowski D, Fauza DO. Transamniotic Stem Cell Therapy for Experimental Congenital Diaphragmatic Hernia: Structural, Transcriptional, and Cell Kinetics Analyses in the Nitrofen Model. Fetal Diagn Ther 2021; 48:381-391. [PMID: 33853064 DOI: 10.1159/000515277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/15/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE We examined select pulmonary effects and donor cell kinetics after transamniotic stem cell therapy (TRASCET) in a model of congenital diaphragmatic hernia (CDH). METHODS Pregnant dams (n = 58) received nitrofen on gestational day 9.5 (E9) to induce fetal CDH. Fetuses (n = 681) were divided into 4 groups: untreated (n = 99) and 3 groups receiving volume-matched intra-amniotic injections on E17 of either saline (n = 142), luciferase-labeled amniotic fluid-derived mesenchymal stem cells (afMSCs; n = 299), or acellular recombinant luciferase (n = 141). Pulmonary morphometry, quantitative gene expression of pulmonary vascular tone mediators, or screening for labeled afMSCs were performed at term (E22). Statistical comparisons were by Mann-Whitney U-test, nested ANOVA, and Wald test. RESULTS TRASCET led to significant downregulation of endothelial nitric oxide synthase and endothelin receptor-A expressions compared to both untreated and saline groups (both p < 0.001). TRASCET also led to a significant decrease in arteriole wall thickness compared to the untreated group (p < 0.001) but not the saline group (p = 0.180). Donor afMSCs were identified in the bone marrow and umbilical cord (p = 0.035 and 0.015, respectively, vs. plain luciferase controls). CONCLUSIONS The effects of TRASCET in experimental CDH appear to be centered on the pulmonary vasculature and to derive from circulating donor cells.
Collapse
Affiliation(s)
- Alexander V Chalphin
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stefanie P Lazow
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel F Labuz
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah A Tracy
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
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: 8.7] [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
|
5
|
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: 10] [Impact Index Per Article: 3.3] [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
|
6
|
Amniotic fluid and breast milk: a rationale for breast milk stem cell therapy in neonatal diseases. Pediatr Surg Int 2020; 36:999-1007. [PMID: 32671487 DOI: 10.1007/s00383-020-04710-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Amniotic fluid and breast milk play important roles in structural development throughout fetal growth and infancy. Given their significance in physical maturation, many studies have investigated the therapeutic and protective roles of amniotic fluid and breast milk in neonatal diseases. Of particular interest to researchers are stem cells found in the two fluids. These stem cells have been investigated due to their ability to self-replicate, differentiate, reduce tissue damage, and their expression of pluripotent markers. While amniotic fluid stem cells have received some attention regarding their ability to treat neonatal diseases, breast milk stem cells have not been investigated to the same extent given the recency of their discovery. The purpose of this review is to compare the functions of amniotic fluid, breast milk, and their stem cells to provide a rationale for the use of breast milk stem cells as a therapy for neonatal diseases. Breast milk stem cells present as an important tool for treating neonatal diseases given their ability to reduce inflammation and tissue damage, as well as their multilineage differentiation potential, easy accessibility, and ability to be used in disease modelling.
Collapse
|
7
|
Signaling Pathways Involved in the Development of Bronchopulmonary Dysplasia and Pulmonary Hypertension. CHILDREN-BASEL 2020; 7:children7080100. [PMID: 32824651 PMCID: PMC7465273 DOI: 10.3390/children7080100] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/31/2022]
Abstract
The alveolar and vascular developmental arrest in the premature infants poses a major problem in the management of these infants. Although, with the current management, the survival rate has improved in these infants, but bronchopulmonary dysplasia (BPD) is a serious complication associated with a high mortality rate. During the neonatal developmental period, these infants are vulnerable to stress. Hypoxia, hyperoxia, and ventilation injury lead to oxidative and inflammatory stress, which induce further damage in the lung alveoli and vasculature. Development of pulmonary hypertension (PH) in infants with BPD worsens the prognosis. Despite considerable progress in the management of premature infants, therapy to prevent BPD is not yet available. Animal experiments have shown deregulation of multiple signaling factors such as transforming growth factorβ (TGFβ), connective tissue growth factor (CTGF), fibroblast growth factor 10 (FGF10), vascular endothelial growth factor (VEGF), caveolin-1, wingless & Int-1 (WNT)/β-catenin, and elastin in the pathogenesis of BPD. This article reviews the signaling pathways entailed in the pathogenesis of BPD associated with PH and the possible management.
Collapse
|
8
|
Sato Y, Ochiai D, Abe Y, Masuda H, Fukutake M, Ikenoue S, Kasuga Y, Shimoda M, Kanai Y, Tanaka M. Prophylactic therapy with human amniotic fluid stem cells improved survival in a rat model of lipopolysaccharide-induced neonatal sepsis through immunomodulation via aggregates with peritoneal macrophages. Stem Cell Res Ther 2020; 11:300. [PMID: 32690106 PMCID: PMC7370504 DOI: 10.1186/s13287-020-01809-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/06/2020] [Accepted: 07/03/2020] [Indexed: 01/15/2023] Open
Abstract
Background Despite recent advances in neonatal care, sepsis remains a leading cause of mortality in neonates. Mesenchymal stem cells derived from various tissues, such as bone marrow, umbilical cord, and adipose tissue, have beneficial effects on adult sepsis. Although human amniotic fluid stem cells (hAFSCs) have mesenchymal stem cell properties, the efficacy of hAFSCs on neonatal sepsis is yet to be elucidated. This study aimed to investigate the therapeutic potential of hAFSCs on neonatal sepsis using a rat model of lipopolysaccharide (LPS)-induced sepsis. Methods hAFSCs were isolated as CD117-positive cells from human amniotic fluid. Three-day-old rat pups were intraperitoneally treated with LPS to mimic neonatal sepsis. hAFSCs were administered either 3 h before or at 0, 3, or 24 h after LPS exposure. Serum inflammatory cytokine levels, gene expression profiles from spleens, and multiple organ damage were analyzed. hAFSC localization was determined in vivo. In vitro LPS stimulation tests were performed using neonatal rat peritoneal macrophages co-cultured with hAFSCs in a cell-cell contact-dependent/independent manner. Immunoregulation in the spleen was determined using a DNA microarray analysis. Results Prophylactic therapy with hAFSCs improved survival in the LPS-treated rats while the hAFSCs transplantation after LPS exposure did not elicit a therapeutic response. Therefore, hAFSC pretreatment was used for all subsequent studies. Inflammatory cytokine levels were elevated after LPS injection, which was attenuated by hAFSC pretreatment. Subsequently, inflammation-induced damages in the brain, lungs, and liver were ameliorated. hAFSCs aggregated with peritoneal macrophages and/or transiently accumulated in the liver, mesentery, and peritoneum. Paracrine factors released by hAFSCs induced M1-M2 macrophage polarization in a cell-cell contact-independent manner. Direct contact between hAFSCs and peritoneal macrophages further enhanced the polarization. Microarray analysis of the spleen showed that hAFSC pretreatment reduced the expression of genes involved in apoptosis and inflammation and subsequently suppressed toll-like receptor 4 signaling pathways. Conclusions Prophylactic therapy with hAFSCs improved survival in a rat model of LPS-induced neonatal sepsis. These effects might be mediated by a phenotypic switch from M1 to M2 in peritoneal macrophages, triggered by hAFSCs in a cell-cell contact-dependent/independent manner and the subsequent immunomodulation of the spleen.
Collapse
Affiliation(s)
- Yu Sato
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Daigo Ochiai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan.
| | - Yushi Abe
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Hirotaka Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Marie Fukutake
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Satoru Ikenoue
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Yoshifumi Kasuga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, 35, Shinanomachi Shinjyukuku, Tokyo, 160-8582, Japan
| |
Collapse
|
9
|
Bonadies L, Zaramella P, Porzionato A, Perilongo G, Muraca M, Baraldi E. Present and Future of Bronchopulmonary Dysplasia. J Clin Med 2020; 9:jcm9051539. [PMID: 32443685 PMCID: PMC7290764 DOI: 10.3390/jcm9051539] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common respiratory disorder among infants born extremely preterm. The pathogenesis of BPD involves multiple prenatal and postnatal mechanisms affecting the development of a very immature lung. Their combined effects alter the lung's morphogenesis, disrupt capillary gas exchange in the alveoli, and lead to the pathological and clinical features of BPD. The disorder is ultimately the result of an aberrant repair response to antenatal and postnatal injuries to the developing lungs. Neonatology has made huge advances in dealing with conditions related to prematurity, but efforts to prevent and treat BPD have so far been only partially effective. Seeing that BPD appears to have a role in the early origin of chronic obstructive pulmonary disease, its prevention is pivotal also in long-term respiratory outcome of these patients. There is currently some evidence to support the use of antenatal glucocorticoids, surfactant therapy, protective noninvasive ventilation, targeted saturations, early caffeine treatment, vitamin A, and fluid restriction, but none of the existing strategies have had any significant impact in reducing the burden of BPD. New areas of research are raising novel therapeutic prospects, however. For instance, early topical (intratracheal or nebulized) steroids seem promising: they might help to limit BPD development without the side effects of systemic steroids. Evidence in favor of stem cell therapy has emerged from several preclinical trials, and from a couple of studies in humans. Mesenchymal stromal/stem cells (MSCs) have revealed a reparatory capability, preventing the progression of BPD in animal models. Administering MSC-conditioned media containing extracellular vesicles (EVs) have also demonstrated a preventive action, without the potential risks associated with unwanted engraftment or the adverse effects of administering cells. In this paper, we explore these emerging treatments and take a look at the revolutionary changes in BPD and neonatology on the horizon.
Collapse
Affiliation(s)
- Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
| | - Patrizia Zaramella
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
| | - Andrea Porzionato
- Human Anatomy Section, Department of Neurosciences, University of Padova, 35128 Padova, Italy;
| | - Giorgio Perilongo
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
| | - Maurizio Muraca
- Institute of Pediatric Research “Città della Speranza”, Stem Cell and Regenerative Medicine Laboratory, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy; (L.B.); (P.Z.)
- Correspondence: ; Tel.: +39-049-821-3560; Fax: +39-049-821-3502
| |
Collapse
|
10
|
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: 20] [Impact Index Per Article: 5.0] [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
|
11
|
Chalphin AV, Tracy SA, Lazow SP, Kycia I, Zurakowski D, Fauza DO. Congenital diaphragmatic hernia as a potential target for transamniotic stem cell therapy. J Pediatr Surg 2020; 55:249-252. [PMID: 31753611 DOI: 10.1016/j.jpedsurg.2019.10.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 10/26/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE We sought to determine whether TRASCET could impact congenital diaphragmatic hernia (CDH). METHODS Twelve pregnant dams received Nitrofen on gestational day 9.5 (E9; term = 22 days) to induce fetal CDH. Fetuses were divided into three groups: untreated (n = 31) and two groups receiving volume-matched intraamniotic injections of either saline (n = 37) or a suspension of 2 × 106 cells/mL of amniotic fluid-derived mesenchymal stem cells (afMSCs; n = 65) on E17. Animals were euthanized at term. Expression of fibroblast growth factor-10 (FGF-10), vascular endothelial growth factor-A (VEGF-A), and surfactant protein-C (SPC) was quantified by qRT-PCR. Statistical analysis was by the Mann-Whitney U test with Bonferroni adjusted criterion (p ≤ 0.01). RESULTS Among survivors with CDH (n = 27/133), the TRASCET group showed significant downregulation of FGF-10 and VEGF-A gene expressions compared to the untreated (p < 0.001 for both) and saline groups (p = 0.005 and p = 0.004, respectively). SPC expression was higher in the TRASCET group compared to the untreated group (p = 0.01), but not the saline group (p = 0.043). Lung laterality had minimal impact on these comparisons. CONCLUSIONS Transamniotic stem cell therapy affects select processes of lung development in experimental congenital diaphragmatic hernia. Further scrutiny into this novel therapy as a potential component of the prenatal management of this disease is warranted. LEVEL OF EVIDENCE N/A (animal and laboratory study).
Collapse
Affiliation(s)
- Alexander V Chalphin
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah A Tracy
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stefanie P Lazow
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Nitkin CR, Rajasingh J, Pisano C, Besner GE, Thébaud B, Sampath V. Stem cell therapy for preventing neonatal diseases in the 21st century: Current understanding and challenges. Pediatr Res 2020; 87:265-276. [PMID: 31086355 PMCID: PMC6854309 DOI: 10.1038/s41390-019-0425-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
Diseases of the preterm newborn such as bronchopulmonary dysplasia, necrotizing enterocolitis, cerebral palsy, and hypoxic-ischemic encephalopathy continue to be major causes of infant mortality and long-term morbidity. Effective therapies for the prevention or treatment for these conditions are still lacking as recent clinical trials have shown modest or no benefit. Stem cell therapy is rapidly emerging as a novel therapeutic tool for several neonatal diseases with encouraging pre-clinical results that hold promise for clinical translation. However, there are a number of unanswered questions and facets to the development of stem cell therapy as a clinical intervention. There is much work to be done to fully elucidate the mechanisms by which stem cell therapy is effective (e.g., anti-inflammatory versus pro-angiogenic), identifying important paracrine mediators, and determining the timing and type of therapy (e.g., cellular versus secretomes), as well as patient characteristics that are ideal. Importantly, the interaction between stem cell therapy and current, standard-of-care interventions is nearly completely unknown. In this review, we will focus predominantly on the use of mesenchymal stromal cells for neonatal diseases, highlighting the promises and challenges in clinical translation towards preventing neonatal diseases in the 21st century.
Collapse
Affiliation(s)
- Christopher R Nitkin
- Division of Neonatology, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Johnson Rajasingh
- Department of Cardiovascular Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, MO, USA
| | - Courtney Pisano
- Department of Pediatric Surgery, Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Gail E Besner
- Department of Pediatric Surgery, Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Bernard Thébaud
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada
| | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children's Mercy Kansas City, Kansas City, MO, USA.
| |
Collapse
|
13
|
Muraca M, Zaramella P, Porzionato A, Baraldi E. Exosome Treatment of Bronchopulmonary Dysplasia: How Pure Should Your Exosome Preparation Be? Am J Respir Crit Care Med 2019; 197:969-970. [PMID: 29160724 DOI: 10.1164/rccm.201709-1851le] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
14
|
Yuan HS, Xiong DQ, Huang F, Cui J, Luo H. MicroRNA-421 inhibition alleviates bronchopulmonary dysplasia in a mouse model via targeting Fgf10. J Cell Biochem 2019; 120:16876-16887. [PMID: 31144392 DOI: 10.1002/jcb.28945] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 11/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common and refractory disease affecting newborn children and infants with alveolar dysplasia and declined pulmonary function. Several microRNAs (miRNAs) have been found to be differentially expressed in BPD progression. This study further explores the role of miR-421 via fibroblast growth factor 10 (Fgf10) in mice with BPD. A mouse model of BPD was established through the induction of hyperoxia, in which the expression pattern of miR-421 and Fgf10 was identified. Furthermore, adenovirus-packed vectors were injected in mice to intervene miR-421 and Fgf10 expression, including miR-421 mimics or inhibitors, and si-Fgf10 to explore the role of miR-421 and Fgf10 in BPD. The target relationship between miR-421 and Fgf10 was investigated. Inflammatory response and cell apoptosis were observed in the mice, with inflammatory cytokines and apoptosis-related factors detected by applying Reverse transcription quantitative polymerase chain reaction, Western blot analysis, and enzyme-linked immunosorbent assay. Fgf10 was confirmed as a target gene of miR-421. Elevated expression of miR-421 was evident, while Fgf10 was poorly expressed in BPD. upregulation of miR-421 and silence of Fgf10 aggravated inflammatory response in lung tissue and promoted lung cell apoptosis in BPD. The aforementioned alterations could be reversed by downregulation of miR-421. Collectively, inhibition of miR-421 can assist in the development of BPD in mice BPD by upregulating Fgf10. Therefore, the present study provides a probable target for the treatment of BPD.
Collapse
Affiliation(s)
- Hua-Shu Yuan
- Department of Oncology, People's Hospital of Taihe County of Jiangxi Province, Taihe, People's Republic of China
| | - Dai-Qun Xiong
- Department of Oncology, The Third Hospital of Nanchang, Nanchang, People's Republic of China
| | - Fang Huang
- Department of Radiotherapy, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jian Cui
- Department of Radiotherapy, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Hui Luo
- The 1st Department of Radiotherapy, Cancer Hospital of Jiangxi Province, Nanchang, People's Republic of China
| |
Collapse
|
15
|
Preventing bronchopulmonary dysplasia: new tools for an old challenge. Pediatr Res 2019; 85:432-441. [PMID: 30464331 DOI: 10.1038/s41390-018-0228-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most prevalent chronic lung disease in infants and presents as a consequence of preterm birth. Due to the lack of effective preventive and treatment strategies, BPD currently represents a major therapeutic challenge that requires continued research efforts at the basic, translational, and clinical levels. However, not all very low birth weight premature babies develop BPD, which suggests that in addition to known gestational age and intrauterine and extrauterine risk factors, other unknown factors must be involved in this disease's development. One of the main goals in BPD research is the early prediction of very low birth weight infants who are at risk of developing BPD in order to initiate the adequate preventive strategies. Other benefits of determining the risk of BPD include providing prognostic information and stratifying infants for clinical trial enrollment. In this article, we describe new opportunities to address BPD's complex pathophysiology by identifying prognostic biomarkers and develop novel, complex in vitro human lung models in order to develop effective therapies. These therapies for protecting the immature lung from injury can be developed by taking advantage of recent scientific progress in -omics, 3D organoids, and regenerative medicine.
Collapse
|
16
|
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: 13.8] [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
|
17
|
Hu Y, Fu J, Xue X. Association of the proliferation of lung fibroblasts with the ERK1/2 signaling pathway in neonatal rats with hyperoxia-induced lung fibrosis. Exp Ther Med 2018; 17:701-708. [PMID: 30651853 PMCID: PMC6307421 DOI: 10.3892/etm.2018.6999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 11/08/2018] [Indexed: 01/02/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common, serious complication occurring in premature infants. Although clinical characteristics and pathologic changes are well described, the pathogenesis of alveolar dysplasia and interstitial fibrosis is less clear. Lung fibroblasts (LFs) are present in the extracellular matrix and serve essential roles during pulmonary epithelial injury and in response to fibrosis development in BPD. The current study investigated hyperoxia-induced proliferation of primary LFs in vitro and mechanisms that may be involved. Newborn rats were exposed to 90% oxygen, while control rats were kept in normal atmosphere. Primary LFs were isolated on postnatal day 3, 7 and 14. Hyperoxia-induced proliferation of LFs isolated on day 7 and 14 by accelerating the cell cycle progression from G1 to S phase. Collagen type I protein secretion and mRNA expression on day 7 and 14 were increased by hyperoxia compared with the controls. Hyperoxia significantly increased the phosphorylation of extracellular signal-regulated kinase (ERK) and significantly increased collagen type I expression compared with the room air control group. The findings indicated that an increase in LF proliferation in response to hyperoxia was associated with ERK1/2 phosphorylation. This mechanism may contribute to over-proliferation of LFs leading to disturbed formation of normal alveoli.
Collapse
Affiliation(s)
- Yu Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
18
|
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: 3.8] [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
|
19
|
Loukogeorgakis SP, De Coppi P. Concise Review: Amniotic Fluid Stem Cells: The Known, the Unknown, and Potential Regenerative Medicine Applications. Stem Cells 2018; 35:1663-1673. [PMID: 28009066 DOI: 10.1002/stem.2553] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 09/07/2016] [Accepted: 10/01/2016] [Indexed: 12/19/2022]
Abstract
The amniotic fluid has been identified as an untapped source of cells with broad potential, which possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. CD117(c-Kit)+ cells selected from amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumors, making them ideal candidates for regenerative medicine applications. Moreover, their ability to engraft in injured organs and modulate immune and repair responses of host tissues, suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases. Although significant questions remain regarding the origin, heterogeneous phenotype, and expansion potential of amniotic fluid stem cells, evidence to date supports their potential role as a valuable stem cell source for the field of regenerative medicine. Stem Cells 2017;35:1663-1673.
Collapse
Affiliation(s)
- Stavros P Loukogeorgakis
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
20
|
LaGrandeur RG, Singhal M, Bany-Mohammed F, Uy C, Koeppel R, Zaldivar F, Haddad F, Nalbandian A, Donovan P, Cooper DM, Aslam M. Pilot feasibility study to detect mesenchymal stem cell biomarkers of bronchopulmonary dysplasia in the tracheal aspirate fluid of preterm infants. J Neonatal Perinatal Med 2018; 11:1-10. [PMID: 29689740 DOI: 10.3233/npm-181722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
OBJECTIVE This study aimed to detect novel mesenchymal stem cell peptides/biomarkers of bronchopulmonary dysplasia (BPD) in the tracheal aspirate fluid (TAF) of preterm infants. STUDY DESIGN Participants included infants less than 32 weeks' gestational age or birth weight under 1500 grams who required endotracheal intubation and mechanical ventilation within first 24 hours of life. TAF sample collection was performed at the time of the first clinically indicated routine suctioning. Standardization curves for human levels of osteopontin (Opn), macrophage colony stimulating factor 1 (Csf1), transforming growth factor beta 1 (TGF-β1), and secretory immunoglobulin A (sIgA) were generated for 15 enrolled participants. RESULTS We demonstrated that stem cell biomarkers are secreted into the TAF of preterm infants and their concentrations can be easily measured during the first week of life. CONCLUSIONS Further studies are warranted to determine a causal relationship between these biomarkers and BPD development and severity.
Collapse
Affiliation(s)
- R G LaGrandeur
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - M Singhal
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - F Bany-Mohammed
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - C Uy
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - R Koeppel
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - F Zaldivar
- Institute for Clinical and Translational Science, University of California Irvine, Irvine, CA, USA
| | - F Haddad
- Institute for Clinical and Translational Science, University of California Irvine, Irvine, CA, USA
| | - A Nalbandian
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| | - P Donovan
- Department of Developmental and Cellular Biology and of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - D M Cooper
- Department of Pediatrics, Division of Pulmonology, University of California Irvine, Irvine, CA, USA
| | - M Aslam
- Department of Pediatrics, Division of Neonatology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
21
|
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: 18.3] [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
|
22
|
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: 1.1] [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
|
23
|
O'Reilly M, Thébaud B. Cell-based therapies for neonatal lung disease. Cell Tissue Res 2016; 367:737-745. [PMID: 27770256 DOI: 10.1007/s00441-016-2517-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/26/2016] [Indexed: 01/06/2023]
Abstract
Preterm birth occurs in approximately 11 % of all births worldwide. Advances in perinatal care have enabled the survival of preterm infants born as early as 23-24 weeks of gestation. However, many are affected by bronchopulmonary dysplasia (BPD)-a common respiratory complication of preterm birth, which has life-long consequences for lung health. Currently, there is no specific treatment for BPD. Recent advances in stem cell research have opened new therapeutic avenues for prevention/repair of lung damage. This review summarizes recent pre-clinical data and early clinical translation of cell-based therapies for BPD.
Collapse
Affiliation(s)
- Megan O'Reilly
- Department of Physiology and Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada, T6G 2E1
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine and Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, Canada, K1H 8L6. .,Division of Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, 401 Smyth Road, Ottawa, ON, Canada, K1H 5B2.
| |
Collapse
|
24
|
Xu Y, Xiang J, Zhao H, Liang H, Huang J, Li Y, Pan J, Zhou H, Zhang X, Wang JH, Liu Z, Wang J. Human amniotic fluid stem cells labeled with up-conversion nanoparticles for imaging-monitored repairing of acute lung injury. Biomaterials 2016; 100:91-100. [PMID: 27244692 DOI: 10.1016/j.biomaterials.2016.05.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 01/11/2023]
Abstract
Human amniotic fluid stem (hAFS) cells have generated a great deal of excitement in cell-based therapies and regenerative medicine. Here, we examined the effect of hAFS cells labeled with dual-polymer-coated UCNP-PEG-PEI nanoparticles in a murine model of acute lung injury (ALI). We observed hAFS cells migration to the lung using highly sensitive in vivo upconversion luminescence (UCL) imaging. We demonstrated that hAFS cells remained viable and retained their ability to differentiate even after UCNP-PEG-PEI labeling. More importantly, hAFS cells displayed remarkable positive effects on ALI-damaged lung tissue repair compared with mouse bone marrow mesenchymal stem cells (mBMSCs), which include recovery of the integrity of alveolar-capillary membrane, attenuation of transepithelial leukocyte and neutrophil migration, and down-regulation of proinflammatory cytokine and chemokine expression. Our work highlights a promising role for imaging-guided hAFS cell-based therapy in ALI.
Collapse
Affiliation(s)
- Yunyun Xu
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Jian Xiang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - He Zhao
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Hansi Liang
- Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province, State Key Laboratory Incubation Base, Soochow University, Suzhou, Jiangsu 215007, China
| | - Jie Huang
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Yan Li
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Jian Pan
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Huiting Zhou
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Xueguang Zhang
- Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province, State Key Laboratory Incubation Base, Soochow University, Suzhou, Jiangsu 215007, China
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jian Wang
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China.
| |
Collapse
|
25
|
Laube M, Stolzing A, Thome UH, Fabian C. Therapeutic potential of mesenchymal stem cells for pulmonary complications associated with preterm birth. Int J Biochem Cell Biol 2016; 74:18-32. [PMID: 26928452 DOI: 10.1016/j.biocel.2016.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
Preterm infants frequently suffer from pulmonary complications resulting in significant morbidity and mortality. Physiological and structural lung immaturity impairs perinatal lung transition to air breathing resulting in respiratory distress. Mechanical ventilation and oxygen supplementation ensure sufficient oxygen supply but enhance inflammatory processes which might lead to the establishment of a chronic lung disease called bronchopulmonary dysplasia (BPD). Current therapeutic options to prevent or treat BPD are limited and have salient side effects, highlighting the need for new therapeutic approaches. Mesenchymal stem cells (MSCs) have demonstrated therapeutic potential in animal models of BPD. This review focuses on MSC-based therapeutic approaches to treat pulmonary complications and critically compares results obtained in BPD models. Thereby bottlenecks in the translational systems are identified that are preventing progress in combating BPD. Notably, current animal models closely resemble the so-called "old" BPD with profound inflammation and injury, whereas clinical improvements shifted disease pathology towards a "new" BPD in which arrest of lung maturation predominates. Future studies need to evaluate the utility of MSC-based therapies in animal models resembling the "new" BPD though promising in vitro evidence suggests that MSCs do possess the potential to stimulate lung maturation. Furthermore, we address the mode-of-action of MSC-based therapies with regard to lung development and inflammation/fibrosis. Their therapeutic efficacy is mainly attributed to an enhancement of regeneration and immunomodulation due to paracrine effects. In addition, we discuss current improvement strategies by genetic modifications or precondition of MSCs to enhance their therapeutic efficacy which could also prove beneficial for BPD therapies.
Collapse
Affiliation(s)
- Mandy Laube
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Alexandra Stolzing
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Loughborough University, Wolfson School of Mechanical and Manufacturing Engineering, Centre for Biological Engineering, Loughborough, UK.
| | - Ulrich H Thome
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Claire Fabian
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
26
|
Porzionato A, Guidolin D, Macchi V, Sarasin G, Grisafi D, Tortorella C, Dedja A, Zaramella P, De Caro R. Fractal analysis of alveolarization in hyperoxia-induced rat models of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2016; 310:L680-8. [DOI: 10.1152/ajplung.00231.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 02/02/2016] [Indexed: 11/22/2022] Open
Abstract
No papers are available about potentiality of fractal analysis in quantitative assessment of alveolarization in bronchopulmonary dysplasia (BPD). Thus, we here performed a comparative analysis between fractal [fractal dimension ( D) and lacunarity] and stereological [mean linear intercept ( Lm), total volume of alveolar air spaces, total number of alveoli, mean alveolar volume, total volume and surface area of alveolar septa, and mean alveolar septal thickness] parameters in experimental hyperoxia-induced models of BPD. At birth, rats were distributed between the following groups: 1) rats raised in ambient air for 2 wk; 2) rats exposed to 60% oxygen for 2 wk; 3) rats raised in normoxia for 6 wk; and 4) rats exposed to 60% hyperoxia for 2 wk and to room air for further 4 wk. Normoxic 6-wk rats showed increased D and decreased lacunarity with respect to normoxic 2-wk rats, together with changes in all stereological parameters except for mean alveolar volume. Hyperoxia-exposed 2-wk rats showed significant changes only in total number of alveoli, mean alveolar volume, and lacunarity with respect to equal-in-age normoxic rats. In the comparison between 6-wk rats, the hyperoxia-exposed group showed decreased D and increased lacunarity, together with changes in all stereological parameters except for septal thickness. Analysis of receiver operating characteristic curves showed a comparable discriminatory power of D, lacunarity, and total number of alveoli; Lmand mean alveolar volume were less discriminative. D and lacunarity did not show significant changes when different segmentation thresholds were applied, suggesting that the fractal approach may be fit to automatic image analysis.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| | - Diego Guidolin
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| | - Veronica Macchi
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| | - Gloria Sarasin
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| | - Davide Grisafi
- Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Cinzia Tortorella
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| | - Arben Dedja
- Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Patrizia Zaramella
- Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Molecular Medicine, University of Padova, Padova, Italy; and
| |
Collapse
|
27
|
Sung DK, Chang YS, Ahn SY, Sung SI, Yoo HS, Choi SJ, Kim SY, Park WS. Optimal Route for Human Umbilical Cord Blood-Derived Mesenchymal Stem Cell Transplantation to Protect Against Neonatal Hyperoxic Lung Injury: Gene Expression Profiles and Histopathology. PLoS One 2015; 10:e0135574. [PMID: 26305093 PMCID: PMC4549285 DOI: 10.1371/journal.pone.0135574] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/23/2015] [Indexed: 11/21/2022] Open
Abstract
The aim of this study was to determine the optimal route of mesenchymal stem cell (MSC) transplantation. To this end, gene expression profiling was performed to compare the effects of intratracheal (IT) versus intravenous (IV) MSC administration. Furthermore, the therapeutic efficacy of each route to protect against neonatal hyperoxic lung injury was also determined. Newborn Sprague-Dawley rats were exposed to hyperoxia (90% oxygen) from birth for 14 days. Human umbilical cord blood-derived MSCs labeling with PKH26 were transplanted through either the IT (5×105) or IV (2×106) route at postnatal day (P) 5. At P14, lungs were harvested for histological, biochemical and microarray analyses. Hyperoxic conditions induced an increase in the mean linear intercept and mean alveolar volume (MAV), indicative of impaired alveolarization. The number of ED-1 positive cells was significantly decreased by both IT and IV transplantations. However, IT administration of MSCs resulted in a greater decrease in MAV and ED-1 positive cells compared to IV administration. Moreover, the number of TUNEL-positive cells was significantly decreased in the IT group, but not in the IV group. Although the IT group received only one fourth of the number of MSCs that the IV group did, a significantly higher number of donor cell-derived red PKH 26 positivity were recovered in the IT group. Hyperoxic conditions induced the up regulation of genes associated with the inflammatory response, such as macrophage inflammatory protein-1 α, tumor necrosis factor-α and inter leukin-6; genes associated with cell death, such as p53 and caspases; and genes associated with fibrosis, such as connective tissue growth factor. In contrast, hyperoxic conditions induced the dwon-regulation of vascular endothelial growth factor and hepatocyte growth factor. These hyperoxia-induced changes in gene expression were decreased in the IT group, but not in the IV group. Thus, local IT MSC transplantation was more effective than systemic IV MSC administration in protecting against neonatal hyperoxic lung injury.
Collapse
Affiliation(s)
- Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Samsung Biomedical Research Institute, 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
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Soo Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Soo Yoon Kim
- Samsung Biomedical Research Institute, 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
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
28
|
Combined iNO and endothelial progenitor cells improve lung alveolar and vascular structure in neonatal rats exposed to prolonged hyperoxia. Pediatr Res 2015; 77:784-92. [PMID: 25742118 DOI: 10.1038/pr.2015.39] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 11/06/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Stem cells or inhaled nitric oxide (iNO) are reported to improve lung structures in bronchopulmonary dysplasia (BPD) models. We hypothesized that combined iNO and transplanted endothelial progenitor cells (EPCs) might restore lung structure in rats after neonatal hyperoxia. METHODS Litters were separated into eight groups: room air, hyperoxia, hyperoxia + iNO, hyperoxia + iNO + L-NAME, hyperoxia + EPCs, hyperoxia + EPCs + L-NAME, hyperoxia + EPCs + iNO, and hyperoxia + EPCs + iNO + L-NAME. Litters were exposed to hyperoxia from the 21st day, then, sacrificed. EPCs were injected on the 21st day. L-NAME was injected daily for 7 d from the 21st day. Serum vascular endothelial growth factor (VEGF), radial alveolar count (RAC), VIII factor, EPCs engraftment, lung VEGF, VEGFR2, endothelial nitric oxide (eNOS) and SDF-1 expression, and NO production were examined. RESULTS Hyperoxia exposure led to air space enlargement, loss of lung capillaries, and low expression of VEGF and eNOS. Transplanted EPCs, when combined with iNO, had significantly increased engraftment in lungs, compared to EPCs alone, upon hyperoxia exposure. There was improvement in alveolarization, microvessel density, and upregulation of VEGF and eNOS proteins in the hyperoxia-exposed EPCs with iNO group, compared to hyperoxia alone. CONCLUSION Combined EPCs and iNO improved lung structures after neonatal hyperoxia. This was associated with the upregulation of VEGF and eNOS expression.
Collapse
|
29
|
Christ GJ, Siriwardane ML, de Coppi P. Engineering muscle tissue for the fetus: getting ready for a strong life. Front Pharmacol 2015; 6:53. [PMID: 25914643 PMCID: PMC4392316 DOI: 10.3389/fphar.2015.00053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/03/2015] [Indexed: 11/17/2022] Open
Abstract
Congenital malformations frequently involve either skeletal, smooth or cardiac tissues. When large parts of those tissues are damaged, the repair of the malformations is challenged by the fact that so much autologous tissue is missing. Current treatments require the use of prostheses or other therapies and are associated with a significant morbidity and mortality. Nonetheless, affected children have generally good survival rates and mostly normal schooling. As such, new therapeutic modalities need to represent significant improvements with clear safety profiles. Regenerative medicine and tissue engineering technologies have the potential to dramatically improve the treatment of any disease or disorder involving a lack of viable tissue. With respect to congenital soft tissue anomalies, the development of, for example, implantable muscle constructs would provide not only the usual desired elasticity and contractile proprieties, but should also be able to grow with the fetus and/or in the postnatal life. Such an approach would eliminate the need for multiple surgeries. However, the more widespread clinical applications of regenerative medicine and tissue engineering technologies require identification of the optimal indications, as well as further elucidation of the precise mechanisms and best methods (cells, scaffolds/biomaterials) for achieving large functional tissue regeneration in those clinical indications. In short, despite some amazing scientific progress, significant safety and efficacy hurdles remain. However, the rapid preclinical advances in the field bode well for future applications. As such, translational researchers and clinicians alike need be informed and prepared to utilize these new techniques for the benefit of their patients, as soon as they are available. To this end, we review herein, the clinical need(s), potential applications, and the relevant preclinical studies that are currently guiding the field toward novel therapeutics.
Collapse
Affiliation(s)
- George J Christ
- Wake Forest Institute for Regenerative Medicine Winston-Salem, NC, USA ; Laboratory of Regenerative Therapeutics, Deptartment of Biomedical Engineering and Orthopaedic Surgery, University of Virginia Charlottesville, VA, USA
| | | | - Paolo de Coppi
- Developmental Biology and Cancer Programme, UCL Institute of Child Health, Great Ormond Street Hospital London, UK
| |
Collapse
|
30
|
Porzionato A, Sfriso MM, Mazzatenta A, Macchi V, De Caro R, Di Giulio C. Effects of hyperoxic exposure on signal transduction pathways in the lung. Respir Physiol Neurobiol 2015; 209:106-14. [DOI: 10.1016/j.resp.2014.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 12/18/2022]
|
31
|
Jeanty C, Kunisaki SM, MacKenzie TC. Novel non-surgical prenatal approaches to treating congenital diaphragmatic hernia. Semin Fetal Neonatal Med 2014; 19:349-56. [PMID: 25456754 DOI: 10.1016/j.siny.2014.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This review focuses on the emerging field of non-surgical in-utero therapies in the management of fetal pulmonary hypoplasia and pulmonary hypertension associated with congenital diaphragmatic hernia (CDH). These experimental approaches include pharmacologic as well as stem-cell-based strategies. Current barriers of non-surgical therapies toward clinical translation are emphasized. As the severity of CDH will likely influence the efficacy of any in-utero therapy, the current status of prenatal imaging and the role of novel biomarkers, especially those related to fetal inflammation, are also reviewed.
Collapse
Affiliation(s)
- Cerine Jeanty
- Department of Surgery, University of California San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine, University of California San Francisco, CA, USA
| | - Shaun M Kunisaki
- Department of Surgery, C.S. Mott Children's Hospital, University of Michigan Health System, Ann Arbor, MI, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine, University of California San Francisco, CA, USA.
| |
Collapse
|
32
|
Vadasz S, Jensen T, Moncada C, Girard E, Zhang F, Blanchette A, Finck C. Second and third trimester amniotic fluid mesenchymal stem cells can repopulate a de-cellularized lung scaffold and express lung markers. J Pediatr Surg 2014; 49:1554-63. [PMID: 25475793 DOI: 10.1016/j.jpedsurg.2014.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND/PURPOSE This study examined the potential of amniotic fluid mesenchymal stem cells (AF-MSCs) to generate lung precursor cells in vitro and on a xenologous three-dimensional de-cellularized lung scaffold. METHODS AF-MSCs were isolated from human amniotic fluid obtained from 17-37 weeks gestation. Lung differentiation was induced on Matrigel or on de-cellularized rat lungs intra-tracheally injected with AF-MSCs by culturing with a modification of small airway growth medium (mSAGM) lacking retinoic acid (RA) and triodothyronine (T3) with addition of fibroblast growth factor-10 (FGF10). Cells and scaffolds were characterized by immunofluorescence and RT-PCR for markers of viability, proliferation, and lung distal airway differentiation (TTF-1(+) and SPC(+)) in the absence of markers of brain (TuJ1(-)) and thyroid (Pax8(-)). RESULTS After culture in mSAGM on either Matrigel or lung scaffolds, there were TTF-1(+)/TuJ1(-)/Pax8(-) cells, indicating a lung precursor phenotype. In addition, SPC(+) cells also evolved suggesting a more mature lung phenotype. CONCLUSIONS We demonstrate that mid- to late-trimester AF-MSCs can be induced to develop into lung precursor cells when cultured on the appropriate extracellular matrix (ECM), making them a viable source for use in cell therapy or development of an ex vivo tissue engineered lung.
Collapse
Affiliation(s)
- Stephanie Vadasz
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Todd Jensen
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Camilo Moncada
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Eric Girard
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Fan Zhang
- Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106
| | - Alex Blanchette
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030
| | - Christine Finck
- Department of Vascular Biology, University of Connecticut Health Center, 263 Farmington Avenue MC3501, Farmington, CT 06030; Department of Surgery, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT 06106.
| |
Collapse
|
33
|
Abstract
Regenerative medicine has recently been established as an emerging interdisciplinary field focused on the repair; replacement or regeneration of cells, tissues and organs. It involves various disciplines, which are focused on different aspects of the regeneration process such as cell biology, gene therapy, bioengineering, material science and pharmacology. In this article, we will outline progress on tissue engineering of specific tissues and organs relevant to paediatric surgery.
Collapse
Affiliation(s)
- Panagiotis Maghsoudlou
- Surgery Unit, Institute of Child Health and Great Ormond Street Hospital, University College London, 30 Guilford St, London WC1N 1EH, UK
| | - Luca Urbani
- Surgery Unit, Institute of Child Health and Great Ormond Street Hospital, University College London, 30 Guilford St, London WC1N 1EH, UK
| | - Paolo De Coppi
- Surgery Unit, Institute of Child Health and Great Ormond Street Hospital, University College London, 30 Guilford St, London WC1N 1EH, UK.
| |
Collapse
|
34
|
Postnatal hyperoxia exposure differentially affects hepatocytes and liver haemopoietic cells in newborn rats. PLoS One 2014; 9:e105005. [PMID: 25115881 PMCID: PMC4130630 DOI: 10.1371/journal.pone.0105005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/17/2014] [Indexed: 11/29/2022] Open
Abstract
Premature newborns are frequently exposed to hyperoxic conditions and experimental data indicate modulation of liver metabolism by hyperoxia in the first postnatal period. Conversely, nothing is known about possible modulation of growth factors and signaling molecules involved in other hyperoxic responses and no data are available about the effects of hyperoxia in postnatal liver haematopoiesis. The aim of the study was to analyse the effects of hyperoxia in the liver tissue (hepatocytes and haemopoietic cells) and to investigate possible changes in the expression of Vascular Endothelial Growth Factor (VEGF), Matrix Metalloproteinase 9 (MMP-9), Hypoxia-Inducible Factor-1α (HIF-1α), endothelial Nitric Oxide Synthase (eNOS), and Nuclear Factor-kB (NF-kB). Experimental design of the study involved exposure of newborn rats to room air (controls), 60% O2 (moderate hyperoxia), or 95% O2 (severe hyperoxia) for the first two postnatal weeks. Immunohistochemical and Western blot analyses were performed. Severe hyperoxia increased hepatocyte apoptosis and MMP-9 expression and decreased VEGF expression. Reduced content in reticular fibers was found in moderate and severe hyperoxia. Some other changes were specifically produced in hepatocytes by moderate hyperoxia, i.e., upregulation of HIF-1α and downregulation of eNOS and NF-kB. Postnatal severe hyperoxia exposure increased liver haemopoiesis and upregulated the expression of VEGF (both moderate and severe hyperoxia) and eNOS (severe hyperoxia) in haemopoietic cells. In conclusion, our study showed different effects of hyperoxia on hepatocytes and haemopoietic cells and differential involvement of the above factors. The involvement of VEGF and eNOS in the liver haemopoietic response to hyperoxia may be hypothesized.
Collapse
|
35
|
Paracrine regulation of fetal lung morphogenesis using human placenta-derived mesenchymal stromal cells. J Surg Res 2014; 190:255-63. [DOI: 10.1016/j.jss.2014.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/24/2014] [Accepted: 04/04/2014] [Indexed: 12/20/2022]
|
36
|
Di Bernardo J, Maiden MM, Hershenson MB, Kunisaki SM. Amniotic fluid derived mesenchymal stromal cells augment fetal lung growth in a nitrofen explant model. J Pediatr Surg 2014; 49:859-64; discussion 864-5. [PMID: 24888823 DOI: 10.1016/j.jpedsurg.2014.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 01/27/2014] [Indexed: 01/18/2023]
Abstract
PURPOSE Recent experimental work suggests the therapeutic role of mesenchymal stromal cells (MSCs) during lung morphogenesis. The purpose of this study was to investigate the potential paracrine effects of amniotic fluid-derived MSCs (AF-MSCs) on fetal lung growth in a nitrofen explant model. METHODS Pregnant Sprague-Dawley dams were gavage fed nitrofen on gestational day 9.5 (E9.5). E14.5 lung explants were subsequently harvested and cultured ex vivo for three days on filter membranes in conditioned media from rat AF-MSCs isolated from control (AF-Ctr) or nitrofen-exposed (AF-Nitro) dams. The lungs were analyzed morphometrically and by quantitative gene expression. RESULTS Although there were no significant differences in total lung surface area among hypoplastic lungs, there were significant increases in terminal budding among E14.5+3 nitrofen explants exposed to AF-Ctr compared to explants exposed to medium alone (58.8±8.4 vs. 39.0±10.0 terminal buds, respectively; p<0.05). In contrast, lungs cultured in AF-Nitro medium failed to augment terminal budding. Nitrofen explants exposed to AF-Ctr showed significant upregulation of surfactant protein C to levels observed in normal fetal lungs. CONCLUSIONS AF-MSCs can augment branching morphogenesis and lung epithelial maturation in a fetal explant model of pulmonary hypoplasia. Cell therapy using donor-derived AF-MSCs may represent a novel strategy for the treatment of fetal congenital diaphragmatic hernia.
Collapse
Affiliation(s)
- Julie Di Bernardo
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Michael M Maiden
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Health System, Ann Arbor, MI, USA
| | - Shaun M Kunisaki
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA.
| |
Collapse
|
37
|
Abstract
Congenital malformations are major causes of disease and death during the first years of life and, most of the time, functional replacement of the missing or damaged organs remains an unmet clinical need. Particularly relevant for the treatment of congenital malformation would be to collect the stem cells at diagnosis, before birth, to be able to intervene during the gestation or in the neonatal period. Human AFSCs (amniotic fluid stem cells), which have characteristics intermediate between those of embryonic and adult stem cells, have been isolated. c-Kit+Lin− cells derived from amniotic fluid display a multilineage haemopoietic potential and they can be easily reprogrammed to a pluripotent status. Although, in the future, we hope to use cells derived from the amniotic fluid, we and others have proved recently that simple organs such as the trachea can be engineered using adult progenitors utilizing decellularized cadaveric matrices. A similar approach could be used in the future for more complex organs such as the muscles, intestines or lungs.
Collapse
|
38
|
The Regenerative Role of the Fetal and Adult Stem Cell Secretome. J Clin Med 2013; 2:302-27. [PMID: 26237150 PMCID: PMC4470151 DOI: 10.3390/jcm2040302] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/17/2013] [Accepted: 11/25/2013] [Indexed: 02/07/2023] Open
Abstract
For a long time, the stem cell regenerative paradigm has been based on the assumption that progenitor cells play a critical role in tissue repair by means of their plasticity and differentiation potential. However, recent works suggest that the mechanism underlying the benefits of stem cell transplantation might relate to a paracrine modulatory effect rather than the replacement of affected cells at the site of injury. Therefore, mounting evidence that stem cells may act as a reservoir of trophic signals released to modulate the surrounding tissue has led to a paradigm shift in regenerative medicine. Attention has been shifted from analysis of the stem cell genome to understanding the stem cell “secretome”, which is represented by the growth factors, cytokines and chemokines produced through paracrine secretion. Insights into paracrine-mediated repair support a new approach in regenerative medicine and the isolation and administration of specific stem cell-derived paracrine factors may represent an extremely promising strategy, introducing paracrine-based therapy as a novel and feasible clinical application. In this review, we will discuss the regenerative potential of fetal and adult stem cells, with particular attention to their secretome.
Collapse
|
39
|
Madurga A, Mizíková I, Ruiz-Camp J, Morty RE. Recent advances in late lung development and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2013; 305:L893-905. [PMID: 24213917 DOI: 10.1152/ajplung.00267.2013] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In contrast to early lung development, a process exemplified by the branching of the developing airways, the later development of the immature lung remains very poorly understood. A key event in late lung development is secondary septation, in which secondary septa arise from primary septa, creating a greater number of alveoli of a smaller size, which dramatically expands the surface area over which gas exchange can take place. Secondary septation, together with architectural changes to the vascular structure of the lung that minimize the distance between the inspired air and the blood, are the objectives of late lung development. The process of late lung development is disturbed in bronchopulmonary dysplasia (BPD), a disease of prematurely born infants in which the structural development of the alveoli is blunted as a consequence of inflammation, volutrauma, and oxygen toxicity. This review aims to highlight notable recent developments in our understanding of late lung development and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Alicia Madurga
- Dept. of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, D-61231 Bad Nauheim, Germany.
| | | | | | | |
Collapse
|
40
|
Porzionato A, Macchi V, Zaramella P, Sarasin G, Grisafi D, Dedja A, Chiandetti L, De Caro R. Effects of postnatal hyperoxia exposure on the rat dentate gyrus and subventricular zone. Brain Struct Funct 2013; 220:229-47. [PMID: 24135771 DOI: 10.1007/s00429-013-0650-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 10/04/2013] [Indexed: 12/27/2022]
Abstract
Premature newborns may be exposed to hyperoxia in the first postnatal period, but clinical and experimental works have raised the question of oxygen toxicity for the developing brain. However, specific analysis of hyperoxia exposure on neurogenesis is still lacking. Thus, the aim of the present study was to evaluate possible changes in the morphometric parameters of the main neurogenic sites in newborn rats exposed to 60 or 95 % oxygen for the first 14 postnatal days. The optical disector, a morphometric method based upon unbiased sampling principles of stereology, was applied to analyse cell densities, total volumes, and total cell numbers of the dentate gyrus (DG) and subventricular zone (SVZ). Apoptosis and proliferation were also studied by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling method and anti-ki67 immunohistochemistry, respectively. Severe hyperoxia increased the percentage of apoptotic cells in the DG. Moderate and severe hyperoxia induced a proliferative response both in the DG and SVZ, but the two neurogenic sites showed different changes in their morphometric parameters. The DG of both the hyperoxic groups showed lower volume and total cell number than that of the normoxic one. Conversely, the SVZ of newborn rats exposed to 95 % hyperoxia showed statistically significant higher volume and total cell number than SVZ of rats raised in normoxia. Our findings indicate that hyperoxia exposure in the first postnatal period affects both the neurogenic areas, although in different ways, i.e. reduction of DG and expansion of SVZ.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Anatomy, Department of Molecular Medicine, University of Padova, Via A Gabelli 65, 35127, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|