1
|
Moghassemi S, Nikanfar S, Dadashzadeh A, Sousa MJ, Wan Y, Sun F, Colson A, De Windt S, Kwaspen L, Kanbar M, Sobhani K, Yang J, Vlieghe H, Li Y, Debiève F, Wyns C, Amorim CA. The revolutionary role of placental derivatives in biomedical research. Bioact Mater 2025; 49:456-485. [PMID: 40177109 PMCID: PMC11964572 DOI: 10.1016/j.bioactmat.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The human placenta is a transient yet crucial organ that plays a key role in sustaining the relationship between the maternal and fetal organisms. Despite its historical classification as "biowaste," placental tissues have garnered increasing attention since the early 1900s for their significant medical potential, particularly in wound repair and surgical application. As ethical considerations regarding human placental derivatives have largely been assuaged in many countries, they have gained significant attention due to their versatile applications in various biomedical fields, such as biomedical engineering, regenerative medicine, and pharmacology. Moreover, there is a substantial trend toward various animal product substitutions in laboratory research with human placental derivatives, reflecting a broader commitment to advancing ethical and sustainable research methodologies. This review provides a comprehensive examination of the current applications of human placental derivatives, explores the mechanisms behind their therapeutic effects, and outlines the future potential and directions of this rapidly advancing field.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Saba Nikanfar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yuting Wan
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Fengxuan Sun
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sven De Windt
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lena Kwaspen
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Keyvan Sobhani
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Yongqian Li
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
2
|
Figueira RL, Khoshgoo N, Doktor F, Khalaj K, Islam T, Moheimani N, Blundell M, Antounians L, Post M, Zani A. Antenatal Administration of Extracellular Vesicles Derived From Amniotic Fluid Stem Cells Improves Lung Function in Neonatal Rats With Congenital Diaphragmatic Hernia. J Pediatr Surg 2024; 59:1771-1777. [PMID: 38519389 DOI: 10.1016/j.jpedsurg.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND The severity of pulmonary hypoplasia is a main determinant of outcome for babies with congenital diaphragmatic hernia (CDH). Antenatal administration of extracellular vesicles derived from amniotic fluid stem cells (AFSC-EVs) has been shown to rescue morphological features of lung development in the rat nitrofen model of CDH. Herein, we evaluated whether AFSC-EV administration to fetal rats with CDH is associated with neonatal improvement in lung function. METHODS AFSC-EVs were isolated by ultracentrifugation and characterized by size, morphology, and canonical marker expression. At embryonic (E) day 9.5, dams were gavaged with olive oil (control) or nitrofen to induce CDH. At E18.5, fetuses received an intra-amniotic injection of either saline or AFSC-EVs. At E21.5, rats were delivered and subjected to a tracheostomy for mechanical ventilation (flexiVent system). Groups were compared for lung compliance, resistance, Newtonian resistance, tissue damping and elastance. Lungs were evaluated for branching morphogenesis and collagen quantification. RESULTS Compared to healthy control, saline-treated pups with CDH had fewer airspaces, more collagen deposition, and functionally exhibited reduced compliance and increased airway resistance, elastance, and tissue damping. Conversely, AFSC-EV administration resulted in improvement of lung mechanics (compliance, resistance, tissue damping, elastance) as well as lung branching morphogenesis and collagen deposition. CONCLUSIONS Our studies show that the rat nitrofen model reproduces lung function impairment similar to that of human babies with CDH. Antenatal administration of AFSC-EVs improves lung morphology and function in neonatal rats with CDH. LEVEL OF EVIDENCE N/A (animal and laboratory study).
Collapse
Affiliation(s)
- Rebeca L Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Naghmeh Khoshgoo
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tasneem Islam
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nazgol Moheimani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Matisse Blundell
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada; Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Ganji N, Li B, Lee C, Pierro A. Necrotizing enterocolitis: recent advances in treatment with translational potential. Pediatr Surg Int 2023; 39:205. [PMID: 37247104 DOI: 10.1007/s00383-023-05476-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/30/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most prevalent and devastating gastrointestinal disorders in neonates. Despite advances in neonatal care, the incidence and mortality due to NEC remain high, highlighting the need to devise novel treatments for this disease. There have been a number of recent advancements in therapeutic approaches for the treatment of NEC; these involve remote ischemic conditioning (RIC), stem cell therapy, breast milk components (human milk oligosaccharides, exosomes, lactoferrin), fecal microbiota transplantation, and immunotherapy. This review summarizes the most recent advances in NEC treatment currently underway as well as their applicability and associated challenges and limitations, with the aim to provide new insight into the paradigm of care for NEC worldwide.
Collapse
Affiliation(s)
- Niloofar Ganji
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Bo Li
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Carol Lee
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada
| | - Agostino Pierro
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
- Translational Medicine, Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
4
|
Hosseini-Siyanaki MR, Liu S, Dagra A, Reddy R, Reddy A, Carpenter SL, Khan M, Lucke-Wold B. Surgical Management of Myelomeningocele. NEONATAL 2023; 4:08. [PMID: 38179156 PMCID: PMC10766379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
Myelomeningocele (MMC) is one of the most common abnormalities of the central nervous system that causes significant neurological impairment. Traditionally, treatment consisted of postnatal closure with the management of the complications, such as ventricular shunting. MMC is a plausible candidate for in-utero surgery because of the mechanism of neurologic damage that begins with abnormal neurulation and continues throughout gestation. Researchers discussed the benefits of in-utero closure prior to the publication of the prospective randomized multicenter Management of Myelomeningocele Study (MOMS trial). Compared to postnatal repair with maternal complications and prematurity as trade-offs, prenatal repair reduced shunting, reversed hindbrain herniation, and improved neurological function. This article discusses the diagnosis, evaluation, long-term follow-up, surgical options, and innovative treatment for fetal myelomeningocele.
Collapse
Affiliation(s)
| | - Sophie Liu
- Department of Neuroscience, University of Johns Hopkins, Baltimore, MD, USA
| | - Abeer Dagra
- University of Florida, College of Medicine, Gainesville, FL, USA
| | - Ramya Reddy
- University of Florida, College of Medicine, Gainesville, FL, USA
| | - Akshay Reddy
- University of Florida, College of Medicine, Gainesville, FL, USA
| | | | - Majid Khan
- University of Nevada, Reno School of Medicine, Reno, NV, USA
| | | |
Collapse
|
5
|
Rosner M, Hengstschläger M. Amniotic Fluid Stem Cells: What They Are and What They Can Become. Curr Stem Cell Res Ther 2023; 18:7-16. [PMID: 34895127 DOI: 10.2174/1574888x16666211210143640] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/21/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
In the last two decades, fetal amniotic fluid stem cells progressively attracted attention in the context of both basic research and the development of innovative therapeutic concepts. They exhibit broadly multipotent plasticity with the ability to differentiate into cells of all three embryonic germ layers and low immunogenicity. They are convenient to maintain, highly proliferative, genomically stable, non-tumorigenic, perfectly amenable to genetic modifications, and do not raise ethical concerns. However, it is important to note that among the various fetal amniotic fluid cells, only c-Kit+ amniotic fluid stem cells represent a distinct entity showing the full spectrum of these features. Since amniotic fluid additionally contains numerous terminally differentiated cells and progenitor cells with more limited differentiation potentials, it is of highest relevance to always precisely describe the isolation procedure and characteristics of the used amniotic fluid-derived cell type. It is of obvious interest for scientists, clinicians, and patients alike to be able to rely on up-todate and concisely separated pictures of the utilities as well as the limitations of terminally differentiated amniotic fluid cells, amniotic fluid-derived progenitor cells, and c-Kit+ amniotic fluid stem cells, to drive these distinct cellular models towards as many individual clinical applications as possible.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Boehm AK, Hillebrandt KH, Dziodzio T, Krenzien F, Neudecker J, Spuler S, Pratschke J, Sauer IM, Andreas MN. Tissue engineering for the diaphragm and its various therapeutic possibilities – A Systematic Review. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Agnes K Boehm
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| | - Karl H Hillebrandt
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Tomasz Dziodzio
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Felix Krenzien
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Jens Neudecker
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| | - Simone Spuler
- Muscle Research Unit Experimental and Clinical Research Center Charité Universitätsmedizin Berlin and Max‐Delbrück‐Centrum für Molekulare Medizin in der Helmholtz‐Gemeinschaft Lindenberger Weg 80 Berlin 13125 Germany
| | - Johann Pratschke
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Cluster of Excellence Matters of Activity. Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG German Research Foundation) under Germany's Excellence Strategy Berlin EXC 2025 Germany
| | - Igor M Sauer
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Cluster of Excellence Matters of Activity. Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG German Research Foundation) under Germany's Excellence Strategy Berlin EXC 2025 Germany
| | - Marco N Andreas
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| |
Collapse
|
7
|
Sharma S, Jeyaraman M, Muthu S. Role of stem cell therapy in neurosciences. ESSENTIALS OF EVIDENCE-BASED PRACTICE OF NEUROANESTHESIA AND NEUROCRITICAL CARE 2022:163-179. [DOI: 10.1016/b978-0-12-821776-4.00012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
8
|
Stem C, Rodman C, Ramamurthy RM, George S, Meares D, Farland A, Atala A, Doering CB, Spencer HT, Porada CD, Almeida-Porada G. Investigating Optimal Autologous Cellular Platforms for Prenatal or Perinatal Factor VIII Delivery to Treat Hemophilia A. Front Cell Dev Biol 2021; 9:678117. [PMID: 34447745 PMCID: PMC8383113 DOI: 10.3389/fcell.2021.678117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
Patients with the severe form of hemophilia A (HA) present with a severe phenotype, and can suffer from life-threatening, spontaneous hemorrhaging. While prophylactic FVIII infusions have revolutionized the clinical management of HA, this treatment is short-lived, expensive, and it is not available to many A patients worldwide. In the present study, we evaluated a panel of readily available cell types for their suitability as cellular vehicles to deliver long-lasting FVIII replacement following transduction with a retroviral vector encoding a B domain-deleted human F8 transgene. Given the immune hurdles that currently plague factor replacement therapy, we focused our investigation on cell types that we deemed to be most relevant to either prenatal or very early postnatal treatment and that could, ideally, be autologously derived. Our findings identify several promising candidates for use as cell-based FVIII delivery vehicles and lay the groundwork for future mechanistic studies to delineate bottlenecks to efficient production and secretion of FVIII following genetic-modification.
Collapse
Affiliation(s)
- Christopher Stem
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher Rodman
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ritu M. Ramamurthy
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Sunil George
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Diane Meares
- Special Hematology Laboratory, Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andrew Farland
- Special Hematology Laboratory, Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher B. Doering
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, United States
| | - H. Trent Spencer
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Christopher D. Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
9
|
Valiulienė G, Zentelytė A, Beržanskytė E, Navakauskienė R. Metabolic Profile and Neurogenic Potential of Human Amniotic Fluid Stem Cells From Normal vs. Fetus-Affected Gestations. Front Cell Dev Biol 2021; 9:700634. [PMID: 34336852 PMCID: PMC8322743 DOI: 10.3389/fcell.2021.700634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 12/04/2022] Open
Abstract
Human amniotic fluid stem cells (hAFSCs) possess some characteristics with mesenchymal stem cells (MSCs) and embryonic stem cells and have a broader differentiation potential compared to MSCs derived from other sources. Although hAFSCs are widely researched, their analysis mainly involves stem cells (SCs) obtained from normal, fetus-unaffected gestations. However, in clinical settings, knowledge about hAFSCs from normal gestations could be poorly translational, as hAFSCs from healthy and fetus-diseased gestations may differ in their differentiation and metabolic potential. Therefore, a more thorough investigation of hAFSCs derived from pathological gestations would provide researchers with the knowledge about the general characteristics of these cells that could be valuable for further scientific investigations and possible future clinical applicability. The goal of this study was to look into the neurogenic and metabolic potential of hAFSCs derived from diseased fetuses, when gestations were concomitant with polyhydramnios and compare them to hAFSCs derived from normal fetuses. Results demonstrated that these cells are similar in gene expression levels of stemness markers (SOX2, NANOG, LIN28A, etc.). However, they differ in expression of CD13, CD73, CD90, and CD105, as flow cytometry analysis revealed higher expression in hAFSCs from unaffected gestations. Furthermore, hAFSCs from “Normal” and “Pathology” groups were different in oxidative phosphorylation rate, as well as level of ATP and reactive oxygen species production. Although the secretion of neurotrophic factors BDNF and VEGF was of comparable degree, as evaluated with enzyme-linked immunosorbent assay (ELISA) test, hAFSCs from normal gestations were found to be more prone to neurogenic differentiation, compared to hAFSCs from polyhydramnios. Furthermore, hAFSCs from polyhydramnios were distinguished by higher secretion of pro-inflammatory cytokine TNFα, which was significantly downregulated in differentiated cells. Overall, these observations show that hAFSCs from pathological gestations with polyhydramnios differ in metabolic and inflammatory status and also possess lower neurogenic potential compared to hAFSCs from normal gestations. Therefore, further in vitro and in vivo studies are necessary to dissect the potential of hAFSCs from polyhydramnios in stem cell-based therapies. Future studies should also search for strategies that could improve the characteristics of hAFSCs derived from diseased fetuses in order for those cells to be successfully applied for regenerative medicine purposes.
Collapse
Affiliation(s)
- Giedrė Valiulienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aistė Zentelytė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Elizabet Beržanskytė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
10
|
Nuge T, Liu X, Tshai KY, Lim SS, Nordin N, Hoque ME, Liu Z. Accelerated wound closure: Systematic evaluation of cellulose acetate effects on biologically active molecules release from amniotic fluid stem cells. Biotechnol Appl Biochem 2021; 69:906-919. [PMID: 33826152 DOI: 10.1002/bab.2162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/26/2021] [Indexed: 01/07/2023]
Abstract
Despite a lot of intensive research on cell-scaffold interaction, the focus is mainly on the capacity of construct scaffolds to regulate cell mobility, migration, and cytotoxicity. The effect of the scaffold's topographical and material properties on the expression of biologically active compounds from stem cells is not well understood. In this study, the influence of cellulose acetate (CA) on the electrospinnability of gelatin and the roles of gelatin-cellulose acetate (Ge-CA) on modulating the release of biologically active compounds from amniotic fluid stem cells (AFSCs) is emphasized. It was found that the presence of a small amount of CA could provide a better microenvironment that mimics AFSCs' niche. However, a large amount of CA exhibited no significant effect on AFSCs migration and infiltration. Further study on the effect of surface topography and mechanical properties on AFSCs showed that the tailored microenvironment provided by the Ge-CA scaffolds had transduced physical cues to biomolecules released into the culture media. It was found that the AFSCs seeded on electrospun scaffolds with less CA proportions have profound effects on the secretion of metabolic compounds compared to those with higher CA contained and gelatin coating. The enhanced secretion of biologically active molecules by the AFSCs on the electrospun scaffolds was proven by the accelerated wound closure on the injured human dermal fibroblast (HDF) model. The rapid HDF cell migration could be anticipated due to a higher level of paracrine factors in AFSCs media. Our study demonstrates that the fibrous topography and mechanical properties of the scaffold are a key material property that modulates the high expression of biologically active compounds from the AFSCs. The discovery elucidates a new aspect of material functions and scaffolds material-AFSC interaction for regulating biomolecules release to promote tissue regeneration/repair. To the best of our knowledge, this is the first report describing the scaffolds material-AFSC interaction and the efficacy of scratch assays on quantifying the cell migration in response to the AFSCs metabolic products.
Collapse
Affiliation(s)
- Tamrin Nuge
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Xiaoling Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Kim Yeow Tshai
- Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Malaysia
| | - Siew Shee Lim
- Faculty of Science and Engineering, University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, Malaysia
| | - Norshariza Nordin
- Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | - Md Enamul Hoque
- Department of Biomedical Engineering, Military Institute of Science and Technology, Dhaka, Bangladesh
| | - Ziqian Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, Ningbo, China
| |
Collapse
|
11
|
Ochiai D, Abe Y, Fukutake M, Sato Y, Ikenoue S, Kasuga Y, Masuda H, Tanaka M. Cell sheets using human amniotic fluid stem cells reduce tissue fibrosis in murine full-thickness skin wounds. Tissue Cell 2021; 68:101472. [PMID: 33360545 DOI: 10.1016/j.tice.2020.101472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
The use of mesenchymal stem cell sheets is a promising strategy for skin regeneration. The injection of dissociated human amniotic fluid stem cells (hAFSCs) was recently found to accelerate cutaneous wound healing with reduced fibrotic scarring, similar to fetal wound healing. However, the use of hAFSCs in applications of cell sheet technology remains limited. The aim of this study was to determine the in vivo efficacy of in vitro-cultured hAFSC sheets in wound healing. The cell sheets were characterized by immunohistochemistry and RT-qPCR and grafted onto full-thickness wounds in BALB/c mice. The wound size was measured, and re-epithelialization, granulation tissue area, and collagen content of the regenerated wound were analyzed histologically. Although the hAFSC sheet contained abundant extracellular matrix molecules and expressed high levels of anti-fibrotic mediators, its grafting did not affect wound closure or the size of the granulation tissue area. In contrast, the organization of type I collagen bundles in the regenerated wound was markedly reduced, while the levels of type III collagen were increased after implantation of the hAFSC sheet. These results suggest that hAFSC sheets can exert anti-fibrotic properties without delaying wound closure.
Collapse
Affiliation(s)
- Daigo Ochiai
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan.
| | - Yushi Abe
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Marie Fukutake
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Sato
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Ikenoue
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Kasuga
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hirotaka Masuda
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Chusilp S, Lee C, Li B, Lee D, Yamoto M, Ganji N, Vejchapipat P, Pierro A. Human amniotic fluid stem cells attenuate cholangiocyte apoptosis in a bile duct injury model of liver ductal organoids. J Pediatr Surg 2021; 56:11-16. [PMID: 33129508 DOI: 10.1016/j.jpedsurg.2020.09.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 11/17/2022]
Abstract
PURPOSE Biliary atresia (BA) is a fibro-obliterative cholangiopathy that involves both extrahepatic and intrahepatic bile ducts in infants. Cholangiocyte apoptosis has an influence on the fibrogenesis process of bile ducts and the progression of liver fibrosis in BA. Human amniotic fluid stem cells (hAFSCs) are multipotent cells that have ability to inhibit cell apoptosis. We aimed to investigate whether hAFSCs have the potential to attenuate cholangiocyte apoptosis and injury induced fibrogenic response in our ex vivo bile duct injury model of liver ductal organoids. METHODS The anti-apoptotic effect of hAFSCs was tested in the acetaminophen-induced injury model of neonatal mouse liver ductal organoids (AUP #42681) by using direct and indirect co-culture systems. Cell apoptosis and proliferation were evaluated by immunofluorescent staining. Expression of fibrogenic cytokines was analyzed by RT-qPCR. Data were compared using one-way ANOVA with post hoc test. RESULTS In our injury model, liver ductal organoids that were treated with hAFSCs in both direct and indirect co-culture systems had a significantly smaller number of apoptotic cholangiocytes and decreased expression of fibrogenic cytokines, transforming growth factor beta-1 (TGF-β1) and platelet-derived growth factor-BB (PDGF-BB). Moreover, hAFSCs increased cholangiocyte proliferation in injured organoids. CONCLUSION hAFSCs have the ability to protect the organoids from injury by decreasing cholangiocyte apoptosis and promoting cholangiocyte proliferation. This protective ability of hAFSCs leads to inhibition of the fibrogenic response in the injured organoids. hAFSCs have high therapeutic potential to attenuate liver fibrogenesis in cholangiopathic diseases such as BA.
Collapse
Affiliation(s)
- Sinobol Chusilp
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Division of Pediatric Surgery, Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Carol Lee
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Dorothy Lee
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Masaya Yamoto
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Niloofar Ganji
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Paisarn Vejchapipat
- Division of Pediatric Surgery, Department of Surgery, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
13
|
Choudhary P, Gupta A, Singh S. Therapeutic Advancement in Neuronal Transdifferentiation of Mesenchymal Stromal Cells for Neurological Disorders. J Mol Neurosci 2020; 71:889-901. [PMID: 33047251 DOI: 10.1007/s12031-020-01714-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders have become the leading cause of chronic pain and death. Treatments available are not sufficient to help the patients as they only alleviate the symptoms and not the cause. In this regard, stem cells therapy has emerged as an upcoming option for the replacement of dead and damaged neurons. Stem cells, in general, are characterized as cells exhibiting potency properties, i.e., on being subjected to specific conditions they transform into cells of another lineage. Of all the types, mesenchymal stem cells (MSCs) are known for their pluripotent nature without the obstacle of ethical concern surrounding the procurement of other cell types. Although fibroblasts are quite similar to MSCs morphologically, certain markers like CD73, CD 90 are specific to MSCs, making both the cell types distinguishable from each other. This is implemented while procuring MSCs from a plethora of sources like umbilical cord blood, adipose tissue, bone marrow, etc. Among these, bone marrow MSCs are the most widely used type for neural regeneration. Neural regeneration is achieved via transdifferentiation. Several studies have either transplanted the stem cells into rodent models or have carried out transdifferentiation in vitro. The process involves a combination of growth factors, pre-treatment factors, and neuronal differentiation inducing mediums. The results obtained are characterized by neuron-like morphology, expression of markers, along with electrophysical activity in some. Recent attempts involve exploring biomaterials that may mimic the native ECM and therefore can be directly introduced at the site of interest. The review gives a brief description of MSCs, their sources and markers, and the different attempts that have been made towards achieving the goal of differentiating MSCs into neurons.
Collapse
Affiliation(s)
- Princy Choudhary
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Ayushi Gupta
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Sangeeta Singh
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India.
| |
Collapse
|
14
|
Li B, Lee C, O'Connell JS, Antounians L, Ganji N, Alganabi M, Cadete M, Nascimben F, Koike Y, Hock A, Botts SR, Wu RY, Miyake H, Minich A, Maalouf MF, Zani-Ruttenstock E, Chen Y, Johnson-Henry KC, De Coppi P, Eaton S, Maattanen P, Delgado Olguin P, Zani A, Sherman PM, Pierro A. Activation of Wnt signaling by amniotic fluid stem cell-derived extracellular vesicles attenuates intestinal injury in experimental necrotizing enterocolitis. Cell Death Dis 2020; 11:750. [PMID: 32929076 PMCID: PMC7490270 DOI: 10.1038/s41419-020-02964-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
Necrotizing enterocolitis (NEC) is a devastating intestinal disease primarily affecting preterm neonates and causing high morbidity, high mortality, and huge costs for the family and society. The treatment and the outcome of the disease have not changed in recent decades. Emerging evidence has shown that stimulating the Wnt/β-catenin pathway and enhancing intestinal regeneration are beneficial in experimental NEC, and that they could potentially be used as a novel treatment. Amniotic fluid stem cells (AFSC) and AFSC-derived extracellular vesicles (EV) can be used to improve intestinal injury in experimental NEC. However, the mechanisms by which they affect the Wnt/β-catenin pathway and intestinal regeneration are unknown. In our current study, we demonstrated that AFSC and EV attenuate NEC intestinal injury by activating the Wnt signaling pathway. AFSC and EV stimulate intestinal recovery from NEC by increasing cellular proliferation, reducing inflammation and ultimately regenerating a normal intestinal epithelium. EV administration has a rescuing effect on intestinal injury when given during NEC induction; however, it failed to prevent injury when given prior to NEC induction. AFSC-derived EV administration is thus a potential emergent novel treatment strategy for NEC.
Collapse
Affiliation(s)
- Bo Li
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Carol Lee
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Joshua S O'Connell
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Lina Antounians
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Niloofar Ganji
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Mashriq Alganabi
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Marissa Cadete
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Francesca Nascimben
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Yuhki Koike
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Alison Hock
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Steven R Botts
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Richard Y Wu
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Hiromu Miyake
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Adam Minich
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Michael F Maalouf
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Elke Zani-Ruttenstock
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Yong Chen
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | | | - Paolo De Coppi
- UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Pekka Maattanen
- Biology Department, Burman University, Lacombe, AB, T4L 2E5, Canada
| | - Paul Delgado Olguin
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, M5S 3H2, Canada
| | - Augusto Zani
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Philip M Sherman
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1G6, Canada
| | - Agostino Pierro
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
- Department of Surgery, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
15
|
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: 0.2] [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
|
16
|
Biancotti JC, Walker KA, Jiang G, Di Bernardo J, Shea LD, Kunisaki SM. Hydrogel and neural progenitor cell delivery supports organotypic fetal spinal cord development in an ex vivo model of prenatal spina bifida repair. J Tissue Eng 2020; 11:2041731420943833. [PMID: 32782773 PMCID: PMC7383650 DOI: 10.1177/2041731420943833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Studying how the fetal spinal cord regenerates in an ex vivo model of spina bifida repair may provide insights into the development of new tissue engineering treatment strategies to better optimize neurologic function in affected patients. Here, we developed hydrogel surgical patches designed for prenatal repair of myelomeningocele defects and demonstrated viability of both human and rat neural progenitor donor cells within this three-dimensional scaffold microenvironment. We then established an organotypic slice culture model using transverse lumbar spinal cord slices harvested from retinoic acid–exposed fetal rats to study the effect of fibrin hydrogel patches ex vivo. Based on histology, immunohistochemistry, gene expression, and enzyme-linked immunoabsorbent assays, these experiments demonstrate the biocompatibility of fibrin hydrogel patches on the fetal spinal cord and suggest this organotypic slice culture system as a useful platform for evaluating mechanisms of damage and repair in children with neural tube defects.
Collapse
Affiliation(s)
- Juan C Biancotti
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Kendal A Walker
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Guihua Jiang
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Julie Di Bernardo
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Shaun M Kunisaki
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA.,Fetal Program, Johns Hopkins Children's Center, Baltimore, MD, USA
| |
Collapse
|
17
|
Wagner R, Montalva L, Zani A, Keijzer R. Basic and translational science advances in congenital diaphragmatic hernia. Semin Perinatol 2020; 44:151170. [PMID: 31427115 DOI: 10.1053/j.semperi.2019.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Congenital Diaphragmatic Hernia (CDH) is a birth defect that is characterized by lung hypoplasia, pulmonary hypertension and a diaphragmatic defect that allows herniation of abdominal organs into the thoracic cavity. Although widely unknown to the public, it occurs as frequently as cystic fibrosis (1:2500). There is no monogenetic cause, but different animal models revealed various biological processes and epigenetic factors involved in the pathogenesis. However, the pathobiology of CDH is not sufficiently understood and its mortality still ranges between 30 and 50%. Future collaborative initiatives are required to improve our basic knowledge and advance novel strategies to (prenatally) treat the abnormal lung development. This review focusses on the genetic, epigenetic and protein background and the latest advances in basic and translational aspects of CDH research.
Collapse
Affiliation(s)
- Richard Wagner
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology (Adjunct), University of Manitoba and Children's Hospital Research Institute of Manitoba, Biology of Breathing Theme, Winnipeg, Manitoba, Canada; Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Louise Montalva
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada and Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada; Department of Pediatric Surgery, Hospital Robert Debré, Paris, France
| | - Augusto Zani
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada and Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology (Adjunct), University of Manitoba and Children's Hospital Research Institute of Manitoba, Biology of Breathing Theme, Winnipeg, Manitoba, Canada.
| |
Collapse
|
18
|
Rodriguez M, Porada CD, Almeida-Porada G. Mechanistic Insights into Factor VIII Immune Tolerance Induction via Prenatal Cell Therapy in Hemophilia A. CURRENT STEM CELL REPORTS 2019; 5:145-161. [PMID: 32351874 DOI: 10.1007/s40778-019-00165-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Purpose of Review Prenatal stem cell and gene therapy approaches are amongst the few therapies that can promise the birth of a healthy infant with specific known genetic diseases. This review describes fetal immune cell signaling and its potential influence on donor cell engraftment, and summarizes mechanisms of central T cell tolerance to peripherally-acquired antigen in the context of prenatal therapies for Hemophilia A. Recent Findings During early gestation, different subsets of antigen presenting cells take up peripherally-acquired, non-inherited antigens and induce the deletion of antigen-reactive T-cell precursors in the thymus, demonstrating the potential for using prenatal cell and gene therapies to induce central tolerance to FVIII in the context of prenatal diagnosis/therapy of Hemophilia A. Summary Prenatal cell and gene therapies are promising approaches to treat several genetic disorders including Hemophilia A and B. Understanding the mechanisms of how FVIII-specific tolerance is achieved during ontogeny could help develop novel therapies for HA and better approaches to overcome FVIII inhibitors.
Collapse
Affiliation(s)
- Martin Rodriguez
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Graҫa Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
19
|
Abe Y, Ochiai D, Masuda H, Sato Y, Otani T, Fukutake M, Ikenoue S, Miyakoshi K, Okano H, Tanaka M. In Utero Amniotic Fluid Stem Cell Therapy Protects Against Myelomeningocele via Spinal Cord Coverage and Hepatocyte Growth Factor Secretion. Stem Cells Transl Med 2019; 8:1170-1179. [PMID: 31407874 PMCID: PMC6811697 DOI: 10.1002/sctm.19-0002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/02/2019] [Indexed: 12/13/2022] Open
Abstract
Despite the poor prognosis associated with myelomeningocele (MMC), the options for prenatal treatments are still limited. Recently, fetal cellular therapy has become a new option for treating birth defects, although the therapeutic effects and mechanisms associated with such treatments remain unclear. The use of human amniotic fluid stem cells (hAFSCs) is ideal with respect to immunoreactivity and cell propagation. The prenatal diagnosis of MMC during early stages of pregnancy could allow for the ex vivo proliferation and modulation of autologous hAFSCs for use in utero stem cell therapy. Therefore, we investigated the therapeutic effects and mechanisms of hAFSCs‐based treatment for fetal MMC. hAFSCs were isolated as CD117‐positive cells from the amniotic fluid of 15‐ to 17‐week pregnant women who underwent amniocentesis for prenatal diagnosis and consented to this study. Rat dams were exposed to retinoic acid to induce fetal MMC and were subsequently injected with hAFSCs in each amniotic cavity. We measured the exposed area of the spinal cord and hepatocyte growth factor (HGF) levels at the lesion. The exposed spinal area of the hAFSC‐treated group was significantly smaller than that of the control group. Immunohistochemical analysis demonstrated a reduction in neuronal damage such as neurodegeneration and astrogliosis in the hAFSC‐treated group. Additionally, in lesions of the hAFSC‐treated group, HGF expression was upregulated and HGF‐positive hAFSCs were identified, suggesting that these cells migrated to the lesion and secreted HGF to suppress neuronal damage and induce neurogenesis. Therefore, in utero hAFSC therapy could become a novel strategy for fetal MMC. stem cells translational medicine2019;8:1170–1179
Collapse
Affiliation(s)
- Yushi Abe
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daigo Ochiai
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hirotaka Masuda
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Sato
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Toshimitsu Otani
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Marie Fukutake
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Ikenoue
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kei Miyakoshi
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics & Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|