1
|
Dallatana A, Cremonesi L, Pezzini F, Fontana G, Innamorati G, Giacomello L. The Placenta as a Source of Human Material for Neuronal Repair. Biomedicines 2024; 12:1567. [PMID: 39062139 PMCID: PMC11275125 DOI: 10.3390/biomedicines12071567] [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: 06/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Stem cell therapy has the potential to meet unsolved problems in tissue repair and regeneration, particularly in the neural tissues. However, an optimal source has not yet been found. Growing evidence indicates that positive effects produced in vivo by mesenchymal stem cells (MSCs) can be due not only to their plasticity but also to secreted molecules including extracellular vesicles (EVs) and the extracellular matrix (ECM). Trophic effects produced by MSCs may reveal the key to developing effective tissue-repair strategies, including approaches based on brain implants or other implantable neural electrodes. In this sense, MSCs will become increasingly valuable and needed in the future. The placenta is a temporary organ devoted to protecting and supporting the fetus. At the same time, the placenta represents an abundant and extremely convenient source of MSCs. Nonetheless, placenta-derived MSCs (P-MSCs) remain understudied as compared to MSCs isolated from other sources. This review outlines the limited literature describing the neuroregenerative effects of P-MSC-derived biomaterials and advocates for exploiting the potential of this untapped source for human regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | - Giulio Innamorati
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy; (A.D.); (L.C.); (F.P.); (G.F.); (L.G.)
| | | |
Collapse
|
2
|
Kitawi R, Ledger S, Kelleher AD, Ahlenstiel CL. Advances in HIV Gene Therapy. Int J Mol Sci 2024; 25:2771. [PMID: 38474018 DOI: 10.3390/ijms25052771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Early gene therapy studies held great promise for the cure of heritable diseases, but the occurrence of various genotoxic events led to a pause in clinical trials and a more guarded approach to progress. Recent advances in genetic engineering technologies have reignited interest, leading to the approval of the first gene therapy product targeting genetic mutations in 2017. Gene therapy (GT) can be delivered either in vivo or ex vivo. An ex vivo approach to gene therapy is advantageous, as it allows for the characterization of the gene-modified cells and the selection of desired properties before patient administration. Autologous cells can also be used during this process which eliminates the possibility of immune rejection. This review highlights the various stages of ex vivo gene therapy, current research developments that have increased the efficiency and safety of this process, and a comprehensive summary of Human Immunodeficiency Virus (HIV) gene therapy studies, the majority of which have employed the ex vivo approach.
Collapse
Affiliation(s)
- Rose Kitawi
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Scott Ledger
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anthony D Kelleher
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- St. Vincent's Hospital, Darlinghurst, NSW 2010, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Chantelle L Ahlenstiel
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
- UNSW RNA Institute, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
3
|
Jaime-Rodríguez M, Cadena-Hernández AL, Rosales-Valencia LD, Padilla-Sánchez JM, Chavez-Santoscoy RA. Are genetic drift and stem cell adherence in laboratory culture issues for cultivated meat production? Front Nutr 2023; 10:1189664. [PMID: 37701376 PMCID: PMC10493286 DOI: 10.3389/fnut.2023.1189664] [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: 03/19/2023] [Accepted: 08/11/2023] [Indexed: 09/14/2023] Open
Abstract
Mesenchymal stem cell-based cultivated meat is a promising solution to the ecological and ethical problems posed by traditional meat production, since it exhibits a protein content and composition that is more comparable to original meat proteins than any other source of cultivated meat products, including plants, bacteria, and fungi. Nonetheless, the nature and laboratory behavior of mesenchymal stem cells pose two significant challenges for large-scale production: genetic drift and adherent growth in culture. Culture conditions used in the laboratory expose the cells to a selective pressure that causes genetic drift, which may give rise to oncogene activation and the loss of "stemness." This is why genetic and functional analysis of the cells during culture is required to determine the maximum number of passages within the laboratory where no significant mutations or loss of function are detected. Moreover, the adherent growth of mesenchymal stem cells can be an obstacle for their large-scale production since volume to surface ratio is limited for high volume containers. Multi-tray systems, roller bottles, and microcarriers have been proposed as potential solutions to scale-up the production of adherent cells required for cultivated meat. The most promising solutions for the safety problems and large-scale obstacles for cultivated meat production are the determination of a limit number of passages based on a genetic analysis and the use of microcarriers from edible materials to maximize the volume to surface proportion and decrease the downstream operations needed for cultivated meat production.
Collapse
|
4
|
Yasmin IA, Dharmarajan A, Warrier S. iPSC-Derived Glioblastoma Cells Have Enhanced Stemness Wnt/β-Catenin Activity Which Is Negatively Regulated by Wnt Antagonist sFRP4. Cancers (Basel) 2023; 15:3622. [PMID: 37509281 PMCID: PMC10377620 DOI: 10.3390/cancers15143622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Growing evidence indicates that cancer stem cells (CSCs) endow the tumor with stem-like properties. Recently, induced pluripotent stem cells (iPSCs) have gained increased attention because of their easy derivation and availability and their potential to differentiate into any cell type. A CSC model derived from iPSCs of human origin would help understand the driving force of tumor initiation and early progression. We report the efficient generation of feeder-free SSEA4, TRA-1-60 and TRA-1-81 positive iPSCs from amniotic membrane-derived mesenchymal stem cells (AMMSCs), which successfully differentiated into three germ layers. We then developed human iPSC-derived glioblastoma multiforme (GBM) model using conditioned media (CM) from U87MG cell line and CSCs derived from U87MG, which confer iPSCs with GBM and GSC-like phenotypes within five days. Both cell types overexpress MGMT and GLI2, but only GSCs overexpress CD133, CD44, ABCG2 and ABCC2. We also observed overexpression of LEF1 and β-catenin in both cell types. Down-regulation of Wnt antagonist secreted frizzled-related protein 4 (sFRP4) in GBM and GSCs, indicating activation of the Wnt/β-catenin pathway, which could be involved in the conversion of iPSCs to CSCs. From future perspectives, our study will help in the creation of a rapid cell-based platform for understanding the complexity of GBM.
Collapse
Affiliation(s)
- Ishmat Ara Yasmin
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600 116, India
- School of Human Sciences, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600 116, India
- Cuor Stem Cellutions Pvt Ltd., Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| |
Collapse
|
5
|
Teoh PL, Mohd Akhir H, Abdul Ajak W, Hiew VV. Human Mesenchymal Stromal Cells Derived from Perinatal Tissues: Sources, Characteristics and Isolation Methods. Malays J Med Sci 2023; 30:55-68. [PMID: 37102047 PMCID: PMC10125235 DOI: 10.21315/mjms2023.30.2.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/22/2022] [Indexed: 04/28/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) derived from perinatal tissues have become indispensable sources for clinical applications due to their superior properties, ease of accessibility and minimal ethical concerns. MSCs isolated from different placenta (PL) and umbilical cord (UC) compartments exhibit great potential for stem cell-based therapies. However, their biological activities could vary due to tissue origins and differences in differentiation potentials. This review provides an overview of MSCs derived from various compartments of perinatal tissues, their characteristics and current isolation methods. Factors affecting the yield and purity of MSCs are also discussed as they are important to ensure consistent and unlimited supply for regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Peik Lin Teoh
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| | | | - Warda Abdul Ajak
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| | - Vun Vun Hiew
- Biotechnology Research Institute, Universiti Malaysia Sabah, Sabah, Malaysia
| |
Collapse
|
6
|
Najafi-Ghalehlou N, Feizkhah A, Mobayen M, Pourmohammadi-Bejarpasi Z, Shekarchi S, Roushandeh AM, Roudkenar MH. Plumping up a Cushion of Human Biowaste in Regenerative Medicine: Novel Insights into a State-of-the-Art Reserve Arsenal. Stem Cell Rev Rep 2022; 18:2709-2739. [PMID: 35505177 PMCID: PMC9064122 DOI: 10.1007/s12015-022-10383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/03/2022]
Abstract
Major breakthroughs and disruptive methods in disease treatment today owe their thanks to our inch by inch developing conception of the infinitive aspects of medicine since the very beginning, among which, the role of the regenerative medicine can on no account be denied, a branch of medicine dedicated to either repairing or replacing the injured or diseased cells, organs, and tissues. A novel means to accomplish such a quest is what is being called "medical biowaste", a large assortment of biological samples produced during a surgery session or as a result of physiological conditions and biological activities. The current paper accentuating several of a number of promising sources of biowaste together with their plausible applications in routine clinical practices and the confronting challenges aims at inspiring research on the existing gap between clinical and basic science to further extend our knowledge and understanding concerning the potential applications of medical biowaste.
Collapse
Affiliation(s)
- Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Feizkhah
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Mobayen
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Pourmohammadi-Bejarpasi
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shekarchi
- Anatomical Sciences Department, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mehryar Habibi Roudkenar
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
7
|
Yudintceva N, Mikhailova N, Fedorov V, Samochernych K, Vinogradova T, Muraviov A, Shevtsov M. Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice. Bioengineering (Basel) 2022; 9:662. [PMID: 36354573 PMCID: PMC9687734 DOI: 10.3390/bioengineering9110662] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are attractive in various fields of regenerative medicine due to their therapeutic potential and complex unique properties. Basic stem cell research and the global COVID-19 pandemic have given impetus to the development of cell therapy for infectious diseases. The aim of this review was to systematize scientific data on the applications of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) in the combined treatment of infectious diseases. Application of MSCs and MSC-EVs in the treatment of infectious diseases has immunomodulatory, anti-inflammatory, and antibacterial effects, and also promotes the restoration of the epithelium and stimulates tissue regeneration. The use of MSC-EVs is a promising cell-free treatment strategy that allows solving the problems associated with the safety of cell therapy and increasing its effectiveness. In this review, experimental data and clinical trials based on MSCs and MSC-EVs for the treatment of infectious diseases are presented. MSCs and MSC-EVs can be a promising tool for the treatment of various infectious diseases, particularly in combination with antiviral drugs. Employment of MSC-derived EVs represents a more promising strategy for cell-free treatment, demonstrating a high therapeutic potential in preclinical studies.
Collapse
Affiliation(s)
- Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Natalia Mikhailova
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Alexandr Muraviov
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| |
Collapse
|
8
|
Chetty S, Yarani R, Swaminathan G, Primavera R, Regmi S, Rai S, Zhong J, Ganguly A, Thakor AS. Umbilical cord mesenchymal stromal cells-from bench to bedside. Front Cell Dev Biol 2022; 10:1006295. [PMID: 36313578 PMCID: PMC9597686 DOI: 10.3389/fcell.2022.1006295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/27/2022] [Indexed: 11/27/2022] Open
Abstract
In recent years, mesenchymal stromal cells (MSCs) have generated a lot of attention due to their paracrine and immuno-modulatory properties. mesenchymal stromal cells derived from the umbilical cord (UC) are becoming increasingly recognized as having increased therapeutic potential when compared to mesenchymal stromal cells from other sources. The purpose of this review is to provide an overview of the various compartments of umbilical cord tissue from which mesenchymal stromal cells can be isolated, the differences and similarities with respect to their regenerative and immuno-modulatory properties, as well as the single cell transcriptomic profiles of in vitro expanded and freshly isolated umbilical cord-mesenchymal stromal cells. In addition, we discuss the therapeutic potential and biodistribution of umbilical cord-mesenchymal stromal cells following systemic administration while providing an overview of pre-clinical and clinical trials involving umbilical cord-mesenchymal stromal cells and their associated secretome and extracellular vesicles (EVs). The clinical applications of umbilical cord-mesenchymal stromal cells are also discussed, especially in relation to obstacles and potential solutions for their effective translation from bench to bedside.
Collapse
Affiliation(s)
- Shashank Chetty
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Reza Yarani
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Ganesh Swaminathan
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Rosita Primavera
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Shobha Regmi
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Sravanthi Rai
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Jim Zhong
- Department of Diagnostic and Interventional Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Abantika Ganguly
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| | - Avnesh S Thakor
- Interventional Radiology Innovation at Stanford (IRIS), Stanford University, Department of Radiology, Palo Alto, CA, United States
| |
Collapse
|
9
|
Heat-Induced Proteotoxic Stress Response in Placenta-Derived Stem Cells (PDSCs) Is Mediated through HSPA1A and HSPA1B with a Potential Higher Role for HSPA1B. Curr Issues Mol Biol 2022; 44:4748-4768. [PMID: 36286039 PMCID: PMC9600182 DOI: 10.3390/cimb44100324] [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: 08/17/2022] [Revised: 09/28/2022] [Accepted: 10/07/2022] [Indexed: 11/18/2022] Open
Abstract
Placenta-derived stem cells (PDSCs), due to unique traits such as mesenchymal and embryonic characteristics and the absence of ethical constraints, are in a clinically and therapeutically advantageous position. To aid in stemness maintenance, counter pathophysiological stresses, and withstand post-differentiation challenges, stem cells require elevated protein synthesis and consequently augmented proteostasis. Stem cells exhibit source-specific proteostasis traits, making it imperative to study them individually from different sources. These studies have implications for understanding stem cell biology and exploitation in the augmentation of therapeutic applications. Here, we aim to identify the primary determinants of proteotoxic stress response in PDSCs. We generated heat-induced dose-responsive proteotoxic stress models of three stem cell types: placental origin cells, the placenta-derived mesenchymal stem cells (pMSCs), maternal origin cells, the decidua parietalis mesenchymal stem cells (DPMSCs), and the maternal–fetal interface cells, decidua basalis mesenchymal stem cells (DBMSCs), and measured stress induction through biochemical and cell proliferation assays. RT-PCR array analysis of 84 genes involved in protein folding and protein quality control led to the identification of Hsp70 members HSPA1A and HSPA1B as the prominent ones among 17 significantly expressed genes and with further analysis at the protein level through Western blotting. A kinetic analysis of HSPA1A and HSPA1B gene and protein expression allowed a time series evaluation of stress response. As identified by protein expression, an active stress response is in play even at 24 h. More prominent differences in expression between the two homologs are detected at the translational level, alluding to a potential higher requirement for HSPA1B during proteotoxic stress response in PDSCs.
Collapse
|
10
|
Low-dose telomerase is required for the expansion and migration of placental mesenchymal stem cells. Biochem Biophys Res Commun 2022; 636:40-47. [DOI: 10.1016/j.bbrc.2022.10.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022]
|
11
|
Pipino C, Bernabé-García Á, Cappellacci I, Stelling-Férez J, Di Tomo P, Santalucia M, Navalón C, Pandolfi A, Nicolás FJ. Effect of the Human Amniotic Membrane on the Umbilical Vein Endothelial Cells of Gestational Diabetic Mothers: New Insight on Inflammation and Angiogenesis. Front Bioeng Biotechnol 2022; 10:854845. [PMID: 35866032 PMCID: PMC9294233 DOI: 10.3389/fbioe.2022.854845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/11/2022] [Indexed: 01/18/2023] Open
Abstract
One of the most relevant diabetes complications is impaired wound healing, mainly characterized by reduced peripheral blood flow and diminished neovascularization together with increased inflammation and oxidative stress. Unfortunately, effective therapies are currently lacking. Recently, the amniotic membrane (AM) has shown promising results in wound management. Here, the potential role of AM on endothelial cells isolated from the umbilical cord vein of gestational diabetes-affected women (GD-HUVECs), has been investigated. Indeed, GD-HUVECs in vivo exposed to chronic hyperglycemia during pregnancy compared to control cells (C-HUVECs) have shown molecular modifications of cellular homeostasis ultimately impacting oxidative and nitro-oxidative stress, inflammatory phenotype, nitric oxide (NO) synthesis, and bioavailability, thus representing a useful model for studying the mechanisms potentially supporting the role of AM in chronic non-healing wounds. In this study, the anti-inflammatory properties of AM have been assessed using a monocyte–endothelium interaction assay in cells pre-stimulated with tumor necrosis factor-α (TNF-α) and through vascular adhesion molecule expression and membrane exposure, together with the AM impact on the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-kB) pathway and NO bioavailability. Moreover, GD-HUVEC migration and tube formation ability were evaluated in the presence of AM. The results showed that AM significantly reduced TNF-α-stimulated monocyte–endothelium interaction and the membrane exposure of the endothelial vascular and intracellular adhesion molecules (VCAM-1 and ICAM-1, respectively) in both C- and GD-HUVECs. Strikingly, AM treatment significantly improved vessel formation in GD-HUVECs and cell migration in both C- and GD-HUVECs. These collective results suggest that AM positively affects various critical pathways in inflammation and angiogenesis, thus providing further validation for ongoing clinical trials in diabetic foot ulcers.
Collapse
Affiliation(s)
- Caterina Pipino
- Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), Department of Medical, Oral and Biotechnological Sciences, University G. D’Annunzio Chieti-Pescara, StemTeCh Group, Chieti, Italy
- *Correspondence: Caterina Pipino, ; Francisco José Nicolás,
| | - Ángel Bernabé-García
- Regeneration, Molecular Oncology and TGFß, IMIB-Arrixaca, Hospital Clínico Universitario Virgen de La Arrixaca, Murcia, Spain
| | - Ilaria Cappellacci
- Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), Department of Medical, Oral and Biotechnological Sciences, University G. D’Annunzio Chieti-Pescara, StemTeCh Group, Chieti, Italy
| | - Javier Stelling-Férez
- Regeneration, Molecular Oncology and TGFß, IMIB-Arrixaca, Hospital Clínico Universitario Virgen de La Arrixaca, Murcia, Spain
- Department of Nutrition and Food Technology, UCAM, Murcia, Spain
| | - Pamela Di Tomo
- Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), Department of Medical, Oral and Biotechnological Sciences, University G. D’Annunzio Chieti-Pescara, StemTeCh Group, Chieti, Italy
| | - Manuela Santalucia
- Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), Department of Medical, Oral and Biotechnological Sciences, University G. D’Annunzio Chieti-Pescara, StemTeCh Group, Chieti, Italy
| | - Carlos Navalón
- Regeneration, Molecular Oncology and TGFß, IMIB-Arrixaca, Hospital Clínico Universitario Virgen de La Arrixaca, Murcia, Spain
| | - Assunta Pandolfi
- Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), Department of Medical, Oral and Biotechnological Sciences, University G. D’Annunzio Chieti-Pescara, StemTeCh Group, Chieti, Italy
| | - Francisco José Nicolás
- Regeneration, Molecular Oncology and TGFß, IMIB-Arrixaca, Hospital Clínico Universitario Virgen de La Arrixaca, Murcia, Spain
- *Correspondence: Caterina Pipino, ; Francisco José Nicolás,
| |
Collapse
|
12
|
Moonshi SS, Adelnia H, Wu Y, Ta HT. Placenta‐Derived Mesenchymal Stem Cells for Treatment of Diseases: A Clinically Relevant Source. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200054] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shehzahdi S. Moonshi
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hossein Adelnia
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| | - Yuao Wu
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Bioscience Discipline School of Environment and Science Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| |
Collapse
|
13
|
Avilla-Royo E, Ochsenbein-Kölble N, Vonzun L, Ehrbar M. Biomaterial-based treatments for the prevention of preterm birth after iatrogenic rupture of the fetal membranes. Biomater Sci 2022; 10:3695-3715. [DOI: 10.1039/d2bm00401a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Minimally invasive interventions to ameliorate or correct fetal abnormalities are becoming a clinical reality. However, the iatrogenic premature preterm rupture of the fetal membranes (FMs) (iPPROM), which may result in...
Collapse
|
14
|
Skowron-Kandzia K, Tomsia M, Koryciak-Komarska H, Plewka D, Wieczorek P, Czekaj P. Gene Expression in Amnion-Derived Cells Cultured on Recombinant Laminin 332-A Preliminary Study. Front Med (Lausanne) 2021; 8:719899. [PMID: 34859000 PMCID: PMC8631290 DOI: 10.3389/fmed.2021.719899] [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: 06/03/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Abstract
Human amniotic cells (hAC) exhibit characteristics of undifferentiated cells and immunomodulatory properties. Recognition of the relationship between amniotic cells and components of the extracellular matrix is an important condition for their ex vivo preparation and further successful clinical application in regenerative medicine and transplantology. Laminin 332 (LN-332), as a natural component of the basement membrane of amniotic epithelial cells and a ligand for integrin receptors, may strongly influence the phenotype and fate of amniotic cells. We investigated the impact of recombinant LN-332 on hAC viability and expression of markers for pluripotency, early differentiation, adhesion, and immunomodulatory properties. During 14 days of culture, hAC were quantified and qualified by light microscopy, immunohistochemistry, immunocytochemistry, and flow cytometry. Gene expression was assessed with real-time polymerase chain reaction (RT-PCR) arrays and compared with differentiated cells originated from the three germ layers. LN-332 caused an over 2-fold increase in the total number of hAC, accompanied by a 75% reduction of SSEA-4-positive cells and an increase in HLA-ABC-positive cells. In particular, we observed that the presence of laminin 332 in the medium of a short-time culture modifies the effect of culture duration on hAC, enhancing time-dependent inhibition of expression of certain genes, including pluripotency and differentiation markers, laminin 332 subunits (which may be part of self-regulation of LN-332 synthesis by amniotic cells), and integrins. The changes observed in hAC were more distinct with respect to differentiated mesenchymal cells, resulting in more comparable phenotypes than those represented by differentiated endo- and ectodermal cells. We concluded that laminin 332 present in the culture medium influences to a certain extent proliferation, adhesion, and differentiation of amniotic cells in culture.
Collapse
Affiliation(s)
- Katarzyna Skowron-Kandzia
- Students Scientific Society, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marcin Tomsia
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Halina Koryciak-Komarska
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Danuta Plewka
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Patrycja Wieczorek
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
15
|
Khan K, Makhoul G, Yu B, Jalani G, Derish I, Rutman AK, Cerruti M, Schwertani A, Cecere R. Amniotic stromal stem cell-loaded hydrogel repairs cardiac tissue in infarcted rat hearts via paracrine mediators. J Tissue Eng Regen Med 2021; 16:110-127. [PMID: 34726328 DOI: 10.1002/term.3262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/18/2021] [Accepted: 10/19/2021] [Indexed: 11/07/2022]
Abstract
The use of stem cells to repair the heart after a myocardial infarction (MI) remains promising, yet clinical trials over the past 20 years suggest that cells fail to integrate into the native tissue, resulting in limited improvements in cardiac function. Here, we demonstrate the cardioprotective potential of a composite inserting human amniotic stromal mesenchymal stem cells (ASMCs) in a chitosan and hyaluronic acid (C/HA) based hydrogel in a rat MI model. Mechanical characterization of the C/HA platform indicated a swift elastic conversion at 40°C and a rapid sol-gel transition time at 37°C. Cell viability assay presented active and proliferating AMSCs in the C/HA. The ASMCs + C/HA injected composite significantly increased left ventricular ejection fraction, fractional shortening, and neovessel formation. The encapsulated AMSCs were abundantly detected in the infarcted myocardium 6 weeks post-administration and co-expressed cardiac proteins and notably proliferative markers. Proteomic profiling revealed that extracellular vesicles released from hypoxia preconditioned ASMCs contained proteins involved in cytoprotection, angiogenesis, cardiac differentiation and non-canonical Wnt-signaling. Independent activation of non-canonical Wnt-signaling pathways in ASMCs induced cardiogenesis. Despite a low injected cellular density at baseline, the encapsulated AMSCs were abundantly retained and increased cardiac function. Furthermore, the C/HA hydrogel provided an active milieu for the AMSCs to proliferate, co-express cardiac proteins, and induce new vessel formation. Hence, this novel composite of AMSCs + C/HA scaffold is a conceivable candidate that could restore cardiac function and reduce remodeling.
Collapse
Affiliation(s)
- Kashif Khan
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Georges Makhoul
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Bin Yu
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Ghulam Jalani
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Ida Derish
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alissa K Rutman
- Human Islet Transplant Laboratory, Department of Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Marta Cerruti
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Adel Schwertani
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada
| | - Renzo Cecere
- Divisions of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, Quebec, Canada.,The Royal Victoria Hospital Montreal, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Han J, Li G, Hou M, Ng J, Kwon MY, Xiong K, Liang X, Taglauer E, Shi Y, Mitsialis SA, Kourembanas S, El-Chemaly S, Lederer JA, Rosas IO, Perrella MA, Liu X. Intratracheal transplantation of trophoblast stem cells attenuates acute lung injury in mice. Stem Cell Res Ther 2021; 12:487. [PMID: 34461993 PMCID: PMC8404310 DOI: 10.1186/s13287-021-02550-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a common lung disorder that affects millions of people every year. The infiltration of inflammatory cells into the lungs and death of the alveolar epithelial cells are key factors to trigger a pathological cascade. Trophoblast stem cells (TSCs) are immune privileged, and demonstrate the capability of self-renewal and multipotency with differentiation into three germ layers. We hypothesized that intratracheal transplantation of TSCs may alleviate ALI. METHODS ALI was induced by intratracheal delivery of bleomycin (BLM) in mice. After exposure to BLM, pre-labeled TSCs or fibroblasts (FBs) were intratracheally administered into the lungs. Analyses of the lungs were performed for inflammatory infiltrates, cell apoptosis, and engraftment of TSCs. Pro-inflammatory cytokines/chemokines of lung tissue and in bronchoalveolar lavage fluid (BALF) were also assessed. RESULTS The lungs displayed a reduction in cellularity, with decreased CD45+ cells, and less thickening of the alveolar walls in ALI mice that received TSCs compared with ALI mice receiving PBS or FBs. TSCs decreased infiltration of neutrophils and macrophages, and the expression of interleukin (IL) 6, monocyte chemoattractant protein-1 (MCP-1) and keratinocyte-derived chemokine (KC) in the injured lungs. The levels of inflammatory cytokines in BALF, particularly IL-6, were decreased in ALI mice receiving TSCs, compared to ALI mice that received PBS or FBs. TSCs also significantly reduced BLM-induced apoptosis of alveolar epithelial cells in vitro and in vivo. Transplanted TSCs integrated into the alveolar walls and expressed aquaporin 5 and prosurfactant protein C, markers for alveolar epithelial type I and II cells, respectively. CONCLUSION Intratracheal transplantation of TSCs into the lungs of mice after acute exposure to BLM reduced pulmonary inflammation and cell death. Furthermore, TSCs engrafted into the alveolar walls to form alveolar epithelial type I and II cells. These data support the use of TSCs for the treatment of ALI.
Collapse
Affiliation(s)
- Junwen Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Gu Li
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Minmin Hou
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Julie Ng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Min-Young Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Kevin Xiong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Xiaoliang Liang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, 77024, USA
| | - Elizabeth Taglauer
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - S Alex Mitsialis
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Stella Kourembanas
- Department of Pediatrics, Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, 77024, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Gorodetsky R, Aicher WK. Allogenic Use of Human Placenta-Derived Stromal Cells as a Highly Active Subtype of Mesenchymal Stromal Cells for Cell-Based Therapies. Int J Mol Sci 2021; 22:5302. [PMID: 34069909 PMCID: PMC8157571 DOI: 10.3390/ijms22105302] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
The application of mesenchymal stromal cells (MSCs) from different sources, including bone marrow (BM, bmMSCs), adipose tissue (atMSCs), and human term placenta (hPSCs) has been proposed for various clinical purposes. Accumulated evidence suggests that the activity of the different MSCs is indirect and associated with paracrine release of pro-regenerative and anti-inflammatory factors. A major limitation of bmMSCs-based treatment for autologous application is the limited yield of cells harvested from BM and the invasiveness of the procedure. Similar effects of autologous and allogeneic MSCs isolated from various other tissues were reported. The easily available fresh human placenta seems to represent a preferred source for harvesting abundant numbers of human hPSCs for allogenic use. Cells derived from the neonate tissues of the placenta (f-hPSC) can undergo extended expansion with a low risk of senescence. The low expression of HLA class I and II on f-hPSCs reduces the risk of rejection in allogeneic or xenogeneic applications in normal immunocompetent hosts. The main advantage of hPSCs-based therapies seems to lie in the secretion of a wide range of pro-regenerative and anti-inflammatory factors. This renders hPSCs as a very competent cell for therapy in humans or animal models. This review summarizes the therapeutic potential of allogeneic applications of f-hPSCs, with reference to their indirect pro-regenerative and anti-inflammatory effects and discusses clinical feasibility studies.
Collapse
Affiliation(s)
- Raphael Gorodetsky
- Biotechnology and Radiobiology Laboratory, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Wilhelm K. Aicher
- Center of Medical Research, Department of Urology at UKT, Eberhard-Karls-University, 72076 Tuebingen, Germany
| |
Collapse
|
18
|
Chia WK, Cheah FC, Abdul Aziz NH, Kampan NC, Shuib S, Khong TY, Tan GC, Wong YP. A Review of Placenta and Umbilical Cord-Derived Stem Cells and the Immunomodulatory Basis of Their Therapeutic Potential in Bronchopulmonary Dysplasia. Front Pediatr 2021; 9:615508. [PMID: 33791258 PMCID: PMC8006350 DOI: 10.3389/fped.2021.615508] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating lung disorder of preterm infants as a result of an aberrant reparative response following exposures to various antenatal and postnatal insults. Despite sophisticated medical treatment in this modern era, the incidence of BPD remains unabated. The current strategies to prevent and treat BPD have met with limited success. The emergence of stem cell therapy may be a potential breakthrough in mitigating this complex chronic lung disorder. Over the last two decades, the human placenta and umbilical cord have gained increasing attention as a highly potential source of stem cells. Placenta-derived stem cells (PDSCs) and umbilical cord-derived stem cells (UCDSCs) display several advantages such as immune tolerance and are generally devoid of ethical constraints, in addition to their stemness qualities. They possess the characteristics of both embryonic and mesenchymal stromal/stem cells. Recently, there are many preclinical studies investigating the use of these cells as therapeutic agents in neonatal disease models for clinical applications. In this review, we describe the preclinical and clinical studies using PDSCs and UCDSCs as treatment in animal models of BPD. The source of these stem cells, routes of administration, and effects on immunomodulation, inflammation and regeneration in the injured lung are also discussed. Lastly, a brief description summarized the completed and ongoing clinical trials using PDSCs and UCDSCs as therapeutic agents in preventing or treating BPD. Due to the complexity of BPD, the development of a safe and efficient therapeutic agent remains a major challenge to both clinicians and researchers.
Collapse
Affiliation(s)
- Wai Kit Chia
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Fook Choe Cheah
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Haslinda Abdul Aziz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Salwati Shuib
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yee Khong
- Department of Pathology, SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Iacobazzi D, Rapetto F, Albertario A, Swim MM, Narayan S, Skeffington K, Salih T, Alvino VV, Madeddu P, Ghorbel MT, Caputo M. Wharton's Jelly-Mesenchymal Stem Cell-Engineered Conduit for Pediatric Translation in Heart Defect. Tissue Eng Part A 2021; 27:201-213. [PMID: 32571164 DOI: 10.1089/ten.tea.2020.0088] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The materials available for the right ventricular outflow tract (RVOT) reconstruction in patients with tetralogy of fallot (TOF)/pulmonary atresia come with the severe limitation of long-term degeneration and lack of growth potential, causing right ventricular dysfunction, aneurysm formation, and arrhythmias, thus necessitating several high-risk reoperations throughout patients' lives. In this study, we evaluated the capacity of mesenchymal stem cells (MSCs) derived from the Wharton's Jelly (WJ-MSCs), the gelatinous inner portion of the umbilical cord, to grow and recellularize an extracellular matrix (ECM) graft in our optimized xeno-free, good manufacturing practice-compliant culture system. WJ-MSCs were phenotypically and functionally characterized by flow cytometry and multilineage differentiation capacity, respectively. The typical MSC immunophenotype and functional characteristics were retained in our xeno-free culture system, as well as the capacity to grow and engraft onto a naturally occurring scaffold. WJ-MSCs, from both human and swine source, showed excellent capacity to recellularize ECM graft producing a living cell-seeded construct. In addition, we have provided an in vivo proof of concept of feasibility of the cellularized conduit, engineered with swine WJ-MSCs, to be used in a novel porcine model of main pulmonary artery reconstruction, where it showed good integration within the host tissue. Our study indicates that the addition of WJ-MSCs to the ECM scaffold can upgrade the material, converting it into a living tissue, with the potential to grow, repair, and remodel the RVOT. These results could potentially represent a paradigm shift in pediatric cardiac intervention toward new modalities for effective and personalized surgical restoration of pulmonary artery and RVOT function in TOF/pulmonary atresia patients. Impact Statement The materials available for pulmonary artery reconstruction in pediatric patients with Congenital Heart Defect come with the limitation of long-term degeneration and lack of growth, thus necessitating several reoperations. Here, we describe a novel approach combining perinatal stem cells and naturally occurring scaffold to create a living tissue engineered conduit that showed good growth potential in a pulmonary artery reconstruction porcine model. We envision this approach is of great interest and relevance in tissue engineering field applied to cardiovascular regenerative medicine, as it may open up new avenues for correction of congenital cardiac defects, with remarkable medical and social benefits.
Collapse
Affiliation(s)
- Dominga Iacobazzi
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Filippo Rapetto
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Ambra Albertario
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Megan M Swim
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Srinivas Narayan
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Katie Skeffington
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Tasneem Salih
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Vincenza Valeria Alvino
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Mohamed T Ghorbel
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, United Kingdom
| |
Collapse
|
20
|
Prokopyuk V, Karpenko V, Shevchenko M, Safonov R, Pasieshvili N, Lazurenko V, Prokopyuk O. Experience in Clinical Application of Cryopreserved Placental Derivatives: Cells, Tissue, Membranes, Extract, and Cord Blood Serum. INNOVATIVE BIOSYSTEMS AND BIOENGINEERING 2020. [DOI: 10.20535/ibb.2020.4.3.215215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
21
|
Borzou B, Mehrabani D, Zare S, Zamani-Pereshkaft M, Acker JP. The Effect of Age and Type of Media on Growth Kinetics of Human Amniotic Fluid Stem Cells. Biopreserv Biobank 2020; 18:389-394. [PMID: 32799559 DOI: 10.1089/bio.2019.0103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: This study compared growth kinetics of human amniotic fluid stem cells (hAFSCs) in different maternal age groups and two different media of AmnioMAX and Dulbecco's modified Eagle's medium (DMEM). Materials and Methods: Three milliliters of amniotic fluid (AF) was provided from 16 pregnant women who were referred for amniocentesis from 16 to 18 weeks of gestation. Mothers were divided to 20-29 (n = 5), 30-39 (n = 5) and 40-49 (n = 6) years old age groups. AF was immediately centrifuged and the cell pellet was cultured. Cells were characterized morphologically, by flow cytometry and for osteogenic and adipogenic inductions. Population doubling time (PDT) and growth kinetics were determined. AFSCs cultured in AmnioMAX were compared in various age groups. A comparison of growth kinetics of AFSCs cultured in AmnioMAX and DMEM from 40 to 49 years old pregnant women was undertaken. Results: AFSCs were adherent to culture flasks and were spindle shape, and positive for osteogenic and adipogenic inductions and for expression of CD73, CD90 and CD105 markers, but negative for CD34 and CD45. PDT among 20-29, 30-39, and 40-49 years old women was 30.9, 38.3, and 43.9 hours, respectively showing a higher cell proliferation in younger ages. When comparing AmnioMAX and DMEM, PDT was 53 and 96.9 hours, respectively denoting to a higher cell proliferation in AmnioMAX. Conclusions: Higher proliferation and plasticity of hAFSCs were noted in AmnioMAX and in younger mothers' samples. These findings can be added to the literature and open a new avenue in regenerative medicine, when hAFSCs are targeted for cell therapy purposes.
Collapse
Affiliation(s)
- Bahareh Borzou
- Department of Biochemistry, Science and Research Branch, Islamic Azad University, Shiraz, Iran.,Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Shahrokh Zare
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Jason P Acker
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada.,Centre for Innovation, Canadian Blood Services, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Mayo JN, Kauer SD, Brumley MR, Bearden SE. Pericytes improve locomotor recovery after spinal cord injury in male and female neonatal rats. Microcirculation 2020; 27:e12646. [PMID: 32608116 DOI: 10.1111/micc.12646] [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] [Received: 04/23/2020] [Revised: 06/06/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE It is not known how activation of the hypoxia-inducible factor (HIF) pathway in pericytes, cells of the microvascular wall, influences new capillary growth. We tested the hypothesis that HIF-activated pericytes promote angiogenesis in a neonatal model of spinal cord injury (SCI). METHODS Human placental pericytes stimulated with cobalt chloride and naïve pericytes were injected into the site of a thoracic hemi-section of the spinal cord in rat pups on postnatal day three (P3). Hindlimb motor recovery and Doppler blood flow perfusion at the site of transection were measured on P10. Immunohistochemistry was used to visualize vessel and neurofilament density for quantification. RESULTS Injection of HIF-activated pericytes resulted in greater vascular density in males but did not result in improved motor function for males or females. Injection of non-HIF-activated pericytes resulted improved motor function recovery in both sexes (males, 2.722 ± 0.31-fold score improvement; females, 3.824 ± 0.58-fold score improvement, P < .05) but produced no significant changes in vessel density. CONCLUSIONS HIF-activated pericytes promote vascular density in males post-SCI. Acute delivery of non-HIF-activated pericytes at the site of injury can improve motor recovery post-SCI.
Collapse
Affiliation(s)
- Jamie N Mayo
- IDEAS 2.0 Centre of Innovation, VA Salt Lake City Health Care System, Salt Lake City, UT, USA.,Division of Epidemiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.,Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Sierra D Kauer
- Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Michele R Brumley
- Department of Psychology, Idaho State University, Pocatello, ID, USA
| | - Shawn E Bearden
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
23
|
Repeated Freezing Procedures Preserve Structural and Functional Properties of Amniotic Membrane for Application in Ophthalmology. Int J Mol Sci 2020; 21:ijms21114029. [PMID: 32512889 PMCID: PMC7312941 DOI: 10.3390/ijms21114029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023] Open
Abstract
For decades, the unique regenerative properties of the human amniotic membrane (hAM) have been successfully utilized in ophthalmology. As a directly applied biomaterial, the hAM should be available in a ready to use manner in clinical settings. However, an extended period of time is obligatory for performing quality and safety tests. Hence, the low temperature storage of the hAM is a virtually inevitable step in the chain from donor retrieval to patient application. At the same time, the impact of subzero temperatures carries an increased risk of irreversible alterations of the structure and composition of biological objects. In the present study, we performed a comprehensive analysis of the hAM as a medicinal product; this is intended for a novel strategy of application in ophthalmology requiring a GMP production protocol including double freezing–thawing cycles. We compared clinically relevant parameters, such as levels of growth factors and extracellular matrix proteins content, morphology, ultrastructure and mechanical properties, before and after one and two freezing cycles. It was found that epidermal growth factor (EGF), transforming growth factor beta 1 (TGF-β1), hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF), hyaluronic acid, and laminin could be detected in all studied conditions without significant differences. Additionally, histological and ultrastructure analysis, as well as transparency and mechanical tests, demonstrated that properties of the hAM required to support therapeutic efficacy in ophthalmology are not impaired by dual freezing.
Collapse
|
24
|
Hou M, Han J, Li G, Kwon MY, Jiang J, Emani S, Taglauer ES, Park JA, Choi EB, Vodnala M, Fong YW, Emani SM, Rosas IO, Perrella MA, Liu X. Multipotency of mouse trophoblast stem cells. Stem Cell Res Ther 2020; 11:55. [PMID: 32054514 PMCID: PMC7020558 DOI: 10.1186/s13287-020-1567-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/24/2019] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In a number of disease processes, the body is unable to repair injured tissue, promoting the need to develop strategies for tissue repair and regeneration, including the use of cellular therapeutics. Trophoblast stem cells (TSCs) are considered putative stem cells as they differentiate into other subtypes of trophoblast cells. To identify cells for future therapeutic strategies, we investigated whether TSCs have properties of stem/progenitor cells including self-renewal and the capacity to differentiate into parenchymal cells of fetal organs, in vitro and in vivo. METHODS TSCs were isolated using anti-CD117 micro-beads, from embryonic day 18.5 placentas. In vitro, CD117+ TSCs were cultured, at a limiting dilution in growth medium for the development of multicellular clones and in specialized medium for differentiation into lung epithelial cells, cardiomyocytes, and retinal photoreceptor cells. CD117+ TSCs were also injected in utero into lung, heart, and the sub-retinal space of embryonic day 13.5 fetuses, and the organs were harvested for histological assessment after a natural delivery. RESULTS We first identified CD117+ cells within the labyrinth zone and chorionic basal plate of murine placentas in late pregnancy, embryonic day 18.5. CD117+ TSCs formed multicellular clones that remained positive for CD117 in vitro, consistent with self-renewal properties. The clonal cells demonstrated multipotency, capable of differentiating into lung epithelial cells (endoderm), cardiomyocytes (mesoderm), and retinal photoreceptor cells (ectoderm). Finally, injection of CD117+ TSCs in utero into lungs, hearts, and the sub-retinal spaces of fetuses resulted in their engraftment on day 1 after birth, and the CD117+ TSCs differentiated into lung alveolar epithelial cells, heart cardiomyocytes, and retina photoreceptor cells, corresponding with the organs in which they were injected. CONCLUSIONS Our findings demonstrate that CD117+ TSCs have the properties of stem cells including clonogenicity, self-renewal, and multipotency. In utero administration of CD117+ TSCs engraft and differentiate into resident cells of the lung, heart, and retina during mouse development.
Collapse
Affiliation(s)
- Minmin Hou
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Junwen Han
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Gu Li
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Min-Young Kwon
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Jiani Jiang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Sirisha Emani
- Department of Cardiovascular Surgery, Children's Hospital, Boston, MA, USA
| | | | - Jin-Ah Park
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - Eun-Bee Choi
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Munender Vodnala
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Yick W Fong
- Department of Medicine, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Sitaram M Emani
- Department of Cardiovascular Surgery, Children's Hospital, Boston, MA, USA
| | - Ivan O Rosas
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Mark A Perrella
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaoli Liu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
25
|
Chen L, Merkhan MM, Forsyth NR, Wu P. Chorionic and amniotic membrane-derived stem cells have distinct, and gestational diabetes mellitus independent, proliferative, differentiation, and immunomodulatory capacities. Stem Cell Res 2019; 40:101537. [PMID: 31422237 DOI: 10.1016/j.scr.2019.101537] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/25/2019] [Accepted: 08/12/2019] [Indexed: 12/19/2022] Open
Abstract
Placental membrane-derived mesenchymal stem cells (MSCs), with the advantages of being non-invasive and having fewer ethical issues, are a promising source for cell therapy. Gestational diabetes (GDM) alters the uterine environment and may affect the therapeutic potential of MSCs derived from placenta. Therefore, we evaluated the biological properties of amniotic (AMSCs) and chorionic membrane MSCs (CMSCs) from human GDM placenta in order to explore their therapeutic potential. In comparison of GDM-/Healthy- CMSCs and AMSCs, the immunophenotypes and typical stellate morphology of MSC were similar in CMSCs irrespective of disease state while the MSC morphology in GDM-AMSCs was less evident. GDM- and Healthy- CMSCs displayed an enhanced proliferation rate and tri-lineage differentiation capacity compared with AMSCs. Notably, GDM-CMSCs had a significantly increased adipogenic ability than Healthy-CMSCs accompanied by increased transcriptional responsiveness of PPARγ and ADIPOQ induction. The secretome effect of Healthy- and GDM- CMSCs/AMSCs by using conditioned media and coculture experiments, suggests that GDM- and Healthy- CMSCs provided an equivalent immunoregulatory effect on suppressing T-cells activation but a reduced effect of GDM-CMSCs on macrophage regulation. However, Healthy- and GDM- CMSCs displayed a superior immunomodulatory capacity in regulation of both T-cells and macrophages than AMSCs. In summary, we highlight the importance of the maternal GDM intrauterine environment during pregnancy and its impact on CMSCs/AMSCs proliferation ability, CMSCs adipogenic potential, and macrophage regulatory capacity.
Collapse
Affiliation(s)
- Liyun Chen
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK.
| | - Marwan M Merkhan
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK; College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Nicholas R Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK.
| | - Pensee Wu
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK; Academic Unit of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, UK; Keele Cardiovascular Research Group, Institute for Applied Clinical Sciences and Centre for Prognosis Research, Institute of Primary Care and Health Sciences, Keele University, Stoke-on-Trent, UK.
| |
Collapse
|
26
|
Umezawa A, Hasegawa A, Inoue M, Tanuma-Takahashi A, Kajiwara K, Makino H, Chikazawa E, Okamoto A. Amnion-derived cells as a reliable resource for next-generation regenerative medicine. Placenta 2019; 84:50-56. [PMID: 31272680 DOI: 10.1016/j.placenta.2019.06.381] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022]
Abstract
The placenta is composed of the amnion, chorionic plate, villous and smooth chorion, decidua basalis, and umbilical cord. The amnion is a readily obtainable source of a large number of cells and cell types, including epithelium, mesenchyme, and endothelium, and is thus an allogeneic resource for regenerative medicine. Endothelial cells are obtained from large arteries and veins in the amniotic membrane as well as the umbilical cord. The amnion-derived cells exhibit transdifferentiation capabilities, including chondrogenesis and cardiomyogenesis, by introduction of transcription factors, in addition to their original and potential phenotypes. The amnion is also a source for production of induced pluripotent stem cells (AM-iPSCs). The AM-iPSCs exhibit stable phenotypes, such as multipotency and immortality, and a unique gene expression pattern. Through the use of amnion-derived cells, as well as other placenta-derived cells, preclinical proof of concept has been achieved in a mouse model of muscular dystrophy.
Collapse
Affiliation(s)
- Akihiro Umezawa
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan.
| | - Akihiro Hasegawa
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan; Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| | - Momoko Inoue
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan; Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| | - Akiko Tanuma-Takahashi
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan; Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| | - Kazuhiro Kajiwara
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan; Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| | - Hatsune Makino
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Emi Chikazawa
- Department of Reproductive Biology, National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, 105-8471, Japan
| |
Collapse
|
27
|
Isolation and expansion of high yield of pure mesenchymal stromal cells from fresh and cryopreserved placental tissues. Cryobiology 2019; 89:100-103. [PMID: 31128944 DOI: 10.1016/j.cryobiol.2019.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 01/15/2023]
Abstract
The injection of placental stromal cells isolated from fetal human tissues (f-hPSC) was reported to indirectly induce tissue regeneration in different animal models. A procedure of f-hPSC isolation from fragments of both selected fresh or cryopreserved bulk placental neonate tissues is proposed, based on their high migratory potential,. The fragments of the desired fetal placental tissues are adhered to a culture dish by traces of diluted fibrin and covered with culture medium. Spontaneous migration of pure f-hPSC from the tissue fragments to the cell culture dishes is followed by their rapid expansion by numerous passages. The isolated f-hPSC express typical mesenchymal surface antigens, including CD29, CD105, CD166 and CD146, with negative expression of white blood cell lineage and endothelial cells markers. Optimal yields of f-hPSC cultures can also be obtained from tissue samples cryopreserved in medium composed of 10% dimethyl sulfoxide (M2SO) and 50% fetal calf serum. Slightly better yields are obtained with media supplemented with 1% human albumin. Medium with 5% M2SO and/or 0.25 mg/ml PEG yielded inferior results. The f-hPSC from fresh or cryopreserved tissues express similar cell markers and growth kinetics. The proposed isolation protocol may also be applied for high yield isolation of stromal cells from fresh and cryopreserved tissue of other organs.
Collapse
|
28
|
Berebichez-Fridman R, Montero-Olvera PR. Sources and Clinical Applications of Mesenchymal Stem Cells: State-of-the-art review. Sultan Qaboos Univ Med J 2018; 18:e264-e277. [PMID: 30607265 DOI: 10.18295/squmj.2018.18.03.002] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/16/2018] [Accepted: 05/10/2018] [Indexed: 12/15/2022] Open
Abstract
First discovered by Friedenstein in 1976, mesenchymal stem cells (MSCs) are adult stem cells found throughout the body that share a fixed set of characteristics. Discovered initially in the bone marrow, this cell source is considered the gold standard for clinical research, although various other sources-including adipose tissue, dental pulp, mobilised peripheral blood and birth-derived tissues-have since been identified. Although similar, MSCs derived from different sources possess distinct characteristics, advantages and disadvantages, including their differentiation potential and proliferation capacity, which influence their applicability. Hence, they may be used for specific clinical applications in the fields of regenerative medicine and tissue engineering. This review article summarises current knowledge regarding the various sources, characteristics and therapeutic applications of MSCs.
Collapse
Affiliation(s)
- Roberto Berebichez-Fridman
- Department of Orthopaedic Surgery, American British Cowdray Medical Center, Mexico City, Mexico.,Tissue Engineering, Cell Therapy & Regenerative Medicine Unit, National Institute of Rehabilitation, Mexico City, Mexico
| | - Pablo R Montero-Olvera
- Tissue Engineering, Cell Therapy & Regenerative Medicine Unit, National Institute of Rehabilitation, Mexico City, Mexico
| |
Collapse
|
29
|
Alessio N, Pipino C, Mandatori D, Di Tomo P, Ferone A, Marchiso M, Melone MAB, Peluso G, Pandolfi A, Galderisi U. Mesenchymal stromal cells from amniotic fluid are less prone to senescence compared to those obtained from bone marrow: An in vitro study. J Cell Physiol 2018; 233:8996-9006. [PMID: 29904927 DOI: 10.1002/jcp.26845] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 05/09/2018] [Indexed: 12/27/2022]
Abstract
Mesenchymal stromal cells (MSCs) are considered to be an excellent source in regenerative medicine. They contain several cell subtypes, including multipotent stem cells. MSCs are of particular interest as they are currently being tested using cell and gene therapies for a number of human diseases. They represent a rare population in tissues; for this reason, they require, before being transplanted, an in vitro amplification. This process may induce replicative senescence, thus affecting differentiation and proliferative capacities. Increasing evidence suggests that MSCs from fetal tissues are significantly more plastic and grow faster than MSCs from bone marrow. Here, we compare amniotic fluid mesenchymal stromal cells (AF-MSCs) and bone marrow mesenchymal stromal cells (BM-MSCs) in terms of cell proliferation, surface markers, multidifferentiation potential, senescence, and DNA repair capacity. Our study shows that AF-MSCs are less prone to senescence with respect to BM-MSCs. Moreover, both cell models activate the same repair system after DNA damage, but AF-MSCs are able to return to the basal condition more efficiently with respect to BM-MSCs. Indeed, AF-MSCs are better able to cope with genotoxic stress that may occur either during in vitro cultivation or following transplantation in patients. Our findings suggest that AF-MSCs may represent a valid alternative to BM-MSCs in regenerative medicine, and, of great relevance, the investigation of the mechanisms involved in DNA repair capacity of both AF-MSCs and BM-MSCs may pave the way to their rational use in the medical field.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Caterina Pipino
- Department of Medical, Oral and Biotechnological Sciences, G. D'Annunzio University Chieti-Pescara, Chieti, Italy
| | - Domitilla Mandatori
- Department of Medicine and Aging Sciences, G. D'Annunzio University Chieti-Pescara, Chieti, Italy
| | - Pamela Di Tomo
- Department of Medicine and Aging Sciences, G. D'Annunzio University Chieti-Pescara, Chieti, Italy
| | - Angela Ferone
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marco Marchiso
- Department of Medicine and Aging Sciences, G. D'Annunzio University Chieti-Pescara, Chieti, Italy
| | - Mariarosa A B Melone
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, 2nd Division of Neurology, Center for Rare Diseases and InterUniversity Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| | | | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, G. D'Annunzio University Chieti-Pescara, Chieti, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania Luigi Vanvitelli, Naples, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Roberts EG, Piekarski BL, Huang K, Emani S, Wong JY, Emani SM. Evaluation of Placental Mesenchymal Stem Cell Sheets for Myocardial Repair and Regeneration. Tissue Eng Part A 2018; 25:867-877. [PMID: 30122114 DOI: 10.1089/ten.tea.2018.0035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
IMPACT STATEMENT This work explores placental tissue as a cell source for fabrication of tissue-engineered surgical patches for myocardial repair of congenital heart defects. This study demonstrates promising findings for the clinically driven evaluation of the cell source as defined by potential cardiac benefit, compatibility, cell source availability, and implant deliverability. It documents methods for the isolation of mesenchymal stem cells from human placental amnion and chorion tissues, characterization of these cells, and eventual cell sheet growth that can be leveraged going forward for patch fabrication. It establishes support to continue pursuing the placenta as a valuable cell source for myocardial repair.
Collapse
Affiliation(s)
- Erin G Roberts
- 1 Department of Materials Science and Engineering, Boston University, Boston, Massachusetts.,2 Department of Cardiovascular Surgery, Children's Hospital, Boston, Massachusetts
| | - Breanna L Piekarski
- 2 Department of Cardiovascular Surgery, Children's Hospital, Boston, Massachusetts
| | - Kevin Huang
- 3 Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Sirisha Emani
- 2 Department of Cardiovascular Surgery, Children's Hospital, Boston, Massachusetts
| | - Joyce Y Wong
- 1 Department of Materials Science and Engineering, Boston University, Boston, Massachusetts.,3 Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Sitaram M Emani
- 2 Department of Cardiovascular Surgery, Children's Hospital, Boston, Massachusetts
| |
Collapse
|
31
|
Pogozhykh O, Prokopyuk V, Prokopyuk O, Kuleshova L, Goltsev A, Figueiredo C, Pogozhykh D. Towards biobanking technologies for natural and bioengineered multicellular placental constructs. Biomaterials 2018; 185:39-50. [PMID: 30218835 DOI: 10.1016/j.biomaterials.2018.08.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 12/13/2022]
Abstract
Clinical application of a large variety of biomaterials is limited by the imperfections in storage technology. Perspective approaches utilizing low-temperature storage are especially challenging for multicellular structures, such as tissues, organs, and bioengineered constructs. Placenta, as a temporary organ, is a widely available unique biological material, being among the most promising sources of various cells and tissues for clinical and experimental use in regenerative medicine and tissue engineering. The aim of this study was to analyse the mechanisms of cryoinjuries in different placental tissues and bioengineered constructs as well as to support the viability after low temperature storage, which would contribute to development of efficient biobanking technologies. This study shows that specificity of cryodamage depends on the structure of the studied object, intercellular bonds, as well as interaction of its components with cryoprotective agents. Remarkably, it was possible to efficiently isolate cells after thawing from all of the studied tissues. While the outcome was lower in comparison to the native non-frozen samples, the phenotype and expression levels of pluripotency genes remained unaffected. Further progress in eliminating of recrystallization processes during thawing would significantly improve biobanking technologies for multicellular constructs and tissues.
Collapse
Affiliation(s)
- Olena Pogozhykh
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine
| | - Volodymyr Prokopyuk
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine
| | - Olga Prokopyuk
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine
| | - Larisa Kuleshova
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine
| | - Anatoliy Goltsev
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine
| | - Constança Figueiredo
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Denys Pogozhykh
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015 Kharkiv, Ukraine.
| |
Collapse
|
32
|
Moonshi SS, Zhang C, Peng H, Puttick S, Rose S, Fisk NM, Bhakoo K, Stringer BW, Qiao GG, Gurr PA, Whittaker AK. A unique 19F MRI agent for the tracking of non phagocytic cells in vivo. NANOSCALE 2018; 10:8226-8239. [PMID: 29682654 DOI: 10.1039/c8nr00703a] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
There is currently intense interest in new methods for understanding the fate of therapeutically-relevant cells, such as mesenchymal stem cells (MSCs). The absence of a confounding background signal and consequent unequivocal assignment makes 19F MRI one of the most attractive modalities for the tracking of injected cells in vivo. We describe here the synthesis of novel partly-fluorinated polymeric nanoparticles with small size and high fluorine content as MRI agents. The polymers, constructed from perfluoropolyether methacrylate (PFPEMA) and oligo(ethylene glycol) methacrylate (OEGMA) have favourable cell uptake profiles and excellent MRI performance. To facilitate cell studies the polymer was further conjugated with a fluorescent dye creating a dual-modal imaging agent. The efficacy of labelling of MSCs was assessed using 19F NMR, flow cytometry and confocal microscopy. The labelling efficiency of 2.6 ± 0.1 × 1012 19F atoms per cell, and viability of >90% demonstrates high uptake and good tolerance by the cells. This loading translates to a minimum 19F MRI detection sensitivity of ∼7.4 × 103 cells per voxel. Importantly, preliminary in vivo data demonstrate that labelled cells can be readily detected within a short acquisition scan period (12 minutes). Hence, these copolymers show outstanding potential for 19F MRI cellular tracking and for quantification of non-phagocytic and therapeutically-relevant cells in vivo.
Collapse
Affiliation(s)
- Shehzahdi S Moonshi
- Australian Institute for Bioengineering and Nanotechnology and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, QLD 4072, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kusuma GD, Yang MC, Brennecke SP, O'Connor AJ, Kalionis B, Heath DE. Transferable Matrixes Produced from Decellularized Extracellular Matrix Promote Proliferation and Osteogenic Differentiation of Mesenchymal Stem Cells and Facilitate Scale-Up. ACS Biomater Sci Eng 2018; 4:1760-1769. [PMID: 33445333 DOI: 10.1021/acsbiomaterials.7b00747] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Decellularized extracellular matrixes (dECM) derived from mesenchymal stem cell (MSC) cultures have recently emerged as cell culture substrates that improve the proliferation, differentiation, and maintenance of MSC phenotype during ex vivo expansion. These biomaterials have considerable potential in the fields of stem cell biology, tissue engineering, and regenerative medicine. Processing the dECMs into concentrated solutions of biomolecules that enable the useful properties of the native dECM to be transferred to a new surface via a simple adsorption step would greatly increase the usefulness and impact of this technology. The development of such solutions, hereafter referred to as transferable matrixes, is the focus of this article. In this work, we produced transferable matrixes from dECM derived from two human placental MSC cell lines (DMSC23 and CMSC29) using pepsin digestion (P-ECM), urea extraction (U-ECM), and mechanical homogenization in acetic acid (AA-ECM). Native dECMs improved primary DMSC proliferation as well as osteogenic and adipogenic differentiation, compared with traditional expansion procedures. Interestingly, tissue culture plastic coated with P-ECM was able to replicate the proliferative effects of native dECM, while U-ECM was able to replicate osteogenic differentiation. These data illustrate the feasibility of producing dECM-derived transferable matrixes that replicate key features of the native matrixes and show that different processing techniques produce transferable matrixes with varying bioactivities. Additionally, these transferable matrixes are able to coat 1.3-5.2 times the surface area covered by the native dECM, facilitating scale-up of this technology.
Collapse
Affiliation(s)
- Gina D Kusuma
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, 20 Flemington Road, Parkville, Victoria 3052, Australia.,School of Chemical and Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Michael C Yang
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, 20 Flemington Road, Parkville, Victoria 3052, Australia.,School of Chemical and Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Shaun P Brennecke
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, 20 Flemington Road, Parkville, Victoria 3052, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Andrea J O'Connor
- School of Chemical and Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, 20 Flemington Road, Parkville, Victoria 3052, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Daniel E Heath
- School of Chemical and Biomedical Engineering, Particulate Fluids Processing Centre, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
34
|
Musatova IB, Volina VV, Chub OV, Prokopyuk VY, Prokopyuk OS. Effects of Implantation of Cryopreserved Placental Explants on the Behavioral Indices and Morphological Characteristics of the Cerebral Structures in Senescent Mice. NEUROPHYSIOLOGY+ 2018. [DOI: 10.1007/s11062-018-9696-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Ventura Ferreira MS, Bienert M, Müller K, Rath B, Goecke T, Opländer C, Braunschweig T, Mela P, Brümmendorf TH, Beier F, Neuss S. Comprehensive characterization of chorionic villi-derived mesenchymal stromal cells from human placenta. Stem Cell Res Ther 2018; 9:28. [PMID: 29402304 PMCID: PMC5800083 DOI: 10.1186/s13287-017-0757-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/29/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Studies in which mesenchymal stromal cells (MSC) from the placenta are compared with multiple MSC types from other sources are rare. The chorionic plate of the human placenta is mainly composed of fetal blood vessels embedded in fetal stroma tissue, lined by trophoblastic cells and organized into chorionic villi (CV) structures. METHODS We comprehensively characterized human MSC collected from postnatal human chorionic villi of placenta (CV-MSC) by analyzing their growth and proliferation potential, differentiation, immunophenotype, extracellular matrix production, telomere length, aging phenotype, and plasticity. RESULTS Immunophenotypic characterization of CV-MSC confirmed the typical MSC marker expression as defined by the International Society for Cellular Therapy. The surface marker profile was consistent with increased potential for proliferation, vascular localization, and early myogenic marker expression. CV-MSC retained multilineage differentiation potential and extracellular matrix remodeling properties. They have undergone reduced telomere loss and delayed onset of cellular senescence as they aged in vitro compared to three other MSC sources. We present evidence that increased human telomerase reverse transcriptase gene expression could not explain the exceptional telomere maintenance and senescence onset delay in cultured CV-MSC. Our in-vitro tumorigenesis detection assay suggests that CV-MSC are not prone to undergo malignant transformation during long-term in-vitro culture. Besides SOX2 expression, no other pluripotency features were observed in early and late passages of CV-MSC. CONCLUSIONS Our work brings forward two remarkable characteristics of CV-MSC, the first being their extended life span as a result of delayed replicative senescence and the second being a delayed aged phenotype characterized by improved telomere length maintenance. MSC from human placenta are very attractive candidates for stem cell-based therapy applications.
Collapse
Affiliation(s)
- Mónica S. Ventura Ferreira
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Michaela Bienert
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aHelmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University, Aachen, Germany
| | - Katrin Müller
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Björn Rath
- 0000 0001 0728 696Xgrid.1957.aDepartment of Orthopedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Tamme Goecke
- 0000 0001 0728 696Xgrid.1957.aDepartment for Gynecology, RWTH Aachen University, Aachen, Germany
| | - Christian Opländer
- 0000 0000 9024 6397grid.412581.bDepartment of Translational Wound Research, Centre for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Till Braunschweig
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Petra Mela
- 0000 0001 0728 696Xgrid.1957.aDepartment of Tissue Engineering and Textile Implants, Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Tim H. Brümmendorf
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Fabian Beier
- 0000 0001 0728 696Xgrid.1957.aDepartment of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University, Aachen, Germany
| | - Sabine Neuss
- 0000 0001 0728 696Xgrid.1957.aInstitute of Pathology, RWTH Aachen University, Aachen, Germany
- 0000 0001 0728 696Xgrid.1957.aHelmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
36
|
Placenta and Placental Derivatives in Regenerative Therapies: Experimental Studies, History, and Prospects. Stem Cells Int 2018. [PMID: 29535770 PMCID: PMC5822788 DOI: 10.1155/2018/4837930] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Placental structures, capable to persist in a genetically foreign organism, are a natural model of allogeneic engraftment carrying a number of distinctive properties. In this review, the main features of the placenta and its derivatives such as structure, cellular composition, immunological and endocrine aspects, and the ability to invasion and deportation are discussed. These features are considered from a perspective that determines the placental material as a unique source for regenerative cell therapies and a lesson for immunological tolerance. A historical overview of clinical applications of placental extracts, cells, and tissue components is described. Empirically accumulated data are summarized and compared with modern research. Furthermore, we define scopes and outlooks of application of placental cells and tissues in the rapidly progressing field of regenerative medicine.
Collapse
|
37
|
Pleiotropic effects of sphingosine-1-phosphate signaling to control human chorionic mesenchymal stem cell physiology. Cell Death Dis 2017; 8:e2930. [PMID: 28703804 PMCID: PMC5550859 DOI: 10.1038/cddis.2017.312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/24/2017] [Accepted: 06/05/2017] [Indexed: 12/26/2022]
Abstract
Chorionic stem cells represent a promising opportunity for regenerative medicine. A deeper understanding of the stimuli that regulate their physiology, could lead to innovative clinical approaches. We revealed the presence of multiple sphingosine-1-phosphate (S1P) receptor isoforms in chorion-derived mesenchymal stem cells (CMSCs). Their activation simultaneously propagated from the plasma membrane through Gi and other heterotrimeric G proteins and further diverged toward extracellular-signal-regulated kinase 1/2 (ERK1/2), p38 and protein kinase D 1. At a functional level, S1P signaling inhibited CMSC migration, while promoting proliferation. Instead, a reduction of cell density was obtained when S1P was combined to treatments that increased cAMP intracellular concentration. Such surprising reduction of cell viability was relatively specific as it was not observed with stromal stem cells from bone marrow. Neither it was observed by activating analogous G proteins with bradykinin nor by inducing cell death via a cAMP-independent pathway. S1P could thus reveal novel keys to improve CMSC differentiation programs acting on cAMP concentration. Furthermore, S1P receptor agonists/antagonists could become instrumental in favoring CMSC engraftment by controlling cell motility.
Collapse
|
38
|
Trohatou O, Roubelakis MG. Mesenchymal Stem/Stromal Cells in Regenerative Medicine: Past, Present, and Future. Cell Reprogram 2017; 19:217-224. [PMID: 28520465 DOI: 10.1089/cell.2016.0062] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The concept of Regenerative Medicine combined with Cell based Therapy and Tissue Engineering represents the fourth pillar of healthcare and provides a promising approach for the treatment of serious diseases. Recently, cell based therapies are focused on the use of mesenchymal stem/stromal cells (MSCs). Human MSCs, that represent a mesoderm derived population of progenitors, are easily expanded in culture. They are capable to differentiate into osteoblasts, chondrocytes, and adipocytes and exhibit the potential to repair or regenerate damaged tissues. The best characterized source of human MSCs to date is the bone marrow; recently, fetal sources, such as amniotic fluid, umbilical cord, amniotic membranes, or placenta, have also attracted increased attention. Thus, MSCs may represent a valuable tool for tissue repair and cell therapeutic applications. To this end, the main focus of this review is to summarize and evaluate the key characteristics, the sources, and the potential use of MSCs in therapeutic approaches and modalities.
Collapse
Affiliation(s)
- Ourania Trohatou
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II , Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Maria G Roubelakis
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II , Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| |
Collapse
|
39
|
Pogozhykh D, Pogozhykh O, Prokopyuk V, Kuleshova L, Goltsev A, Blasczyk R, Mueller T. Influence of temperature fluctuations during cryopreservation on vital parameters, differentiation potential, and transgene expression of placental multipotent stromal cells. Stem Cell Res Ther 2017; 8:66. [PMID: 28284229 PMCID: PMC5346212 DOI: 10.1186/s13287-017-0512-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/11/2017] [Accepted: 02/15/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Successful implementation of rapidly advancing regenerative medicine approaches has led to high demand for readily available cellular suspensions. In particular, multipotent stromal cells (MSCs) of placental origin have shown therapeutic efficiency in the treatment of numerous pathologies of varied etiology. Up to now, cryopreservation is the only effective way to preserve the viability and unique properties of such cells in the long term. However, practical biobanking is often associated with repeated temperature fluctuations or interruption of a cold chain due to various technical, transportation, and stocking events. While biochemical processes are expected to be suspended during cryopreservation, such temperature fluctuations may lead to accumulation of stress as well as to periodic release of water fractions in the samples, possibly leading to damage during long-term storage. METHODS In this study, we performed a comprehensive analysis of changes in cell survival, vital parameters, and differentiation potential, as well as transgene expression of placental MSCs after temperature fluctuations within the liquid nitrogen steam storage, mimicking long-term preservation in practical biobanking, transportation, and temporal storage. RESULTS It was shown that viability and metabolic parameters of placental MSCs did not significantly differ after temperature fluctuations in the range from -196 °C to -100 °C in less than 20 cycles in comparison to constant temperature storage. However, increasing the temperature range to -80 °C as well as increasing the number of cycles leads to significant lowering of these parameters after thawing. The number of apoptotic changes increases depending on the number of cycles of temperature fluctuations. Besides, adhesive properties of the cells after thawing are significantly compromised in the samples subjected to temperature fluctuations during storage. Differentiation potential of placental MSCs was not compromised after cryopreservation with constant end temperatures or with temperature fluctuations. However, regulation of various genes after cryopreservation procedures significantly varies. Interestingly, transgene expression was not compromised in any of the studied samples. CONCLUSIONS Alterations in structural and functional parameters of placental MSCs after long-term preservation should be considered in practical biobanking due to potential temperature fluctuations in samples. At the same time, differentiation potential and transgene expression are not compromised during studied storage conditions, while variation in gene regulation is observed.
Collapse
Affiliation(s)
- Denys Pogozhykh
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany. .,Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015, Kharkiv, Ukraine.
| | - Olena Pogozhykh
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015, Kharkiv, Ukraine
| | - Volodymyr Prokopyuk
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015, Kharkiv, Ukraine
| | - Larisa Kuleshova
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015, Kharkiv, Ukraine
| | - Anatoliy Goltsev
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Pereyaslavskaya Str. 23, 61015, Kharkiv, Ukraine
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Thomas Mueller
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Synlab Medical Care Center Weiden Ltd., Zur Kesselschmiede 4, 92637, Weiden in der Oberpfalz, Germany
| |
Collapse
|
40
|
Qin SQ, Kusuma GD, Al-Sowayan B, Pace RA, Isenmann S, Pertile MD, Gronthos S, Abumaree MH, Brennecke SP, Kalionis B. Establishment and characterization of fetal and maternal mesenchymal stem/stromal cell lines from the human term placenta. Placenta 2016; 39:134-46. [DOI: 10.1016/j.placenta.2016.01.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 01/19/2016] [Accepted: 01/21/2016] [Indexed: 12/31/2022]
|
41
|
Pogozhykh D, Prokopyuk V, Pogozhykh O, Mueller T, Prokopyuk O. Influence of Factors of Cryopreservation and Hypothermic Storage on Survival and Functional Parameters of Multipotent Stromal Cells of Placental Origin. PLoS One 2015; 10:e0139834. [PMID: 26431528 PMCID: PMC4592233 DOI: 10.1371/journal.pone.0139834] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/17/2015] [Indexed: 12/19/2022] Open
Abstract
Human placenta is a highly perspective source of multipotent stromal cells (MSCs) both for the purposes of patient specific auto-banking and allogeneic application in regenerative medicine. Implementation of new GMP standards into clinical practice enforces the search for relevant methods of cryopreservation and short-term hypothermic storage of placental MSCs. In this paper we analyze the effect of different temperature regimes and individual components of cryoprotective media on viability, metabolic and culture properties of placental MSCs. We demonstrate (I) the possibility of short-term hypothermic storage of these cells; (II) determine DMSO and propanediol as the most appropriate cryoprotective agents; (III) show the possibility of application of volume expanders (plasma substituting solutions based on dextran or polyvinylpyrrolidone); (IV) reveal the priority of ionic composition over the serum content in cryopreservation media; (V) determine a cooling rate of 1°C/min down to -40°C followed by immersion into liquid nitrogen as the optimal cryopreservation regime for this type of cells. This study demonstrates perspectives for creation of new defined cryopreservation methods towards GMP standards.
Collapse
Affiliation(s)
- Denys Pogozhykh
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
- * E-mail:
| | - Volodymyr Prokopyuk
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Olena Pogozhykh
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Mueller
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Olga Prokopyuk
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| |
Collapse
|
42
|
Cox B, Leavey K, Nosi U, Wong F, Kingdom J. Placental transcriptome in development and pathology: expression, function, and methods of analysis. Am J Obstet Gynecol 2015; 213:S138-51. [PMID: 26428493 DOI: 10.1016/j.ajog.2015.07.046] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/29/2015] [Accepted: 07/30/2015] [Indexed: 12/18/2022]
Abstract
The placenta is the essential organ of mammalian pregnancy and errors in its development and function are associated with a wide range of human pathologies of pregnancy. Genome sequencing has led to methods for investigation of the transcriptome (all expressed RNA species) using microarrays and next-generation sequencing, and implementation of these techniques has identified many novel species of RNA including: micro-RNA, long noncoding RNA, and circular RNA. These species can physically interact with both each other and regulatory proteins to modify gene expression and messenger RNA to protein translation. Transcriptome analysis is actively used to investigate placental development and dysfunction in pathologies ranging from preeclampsia and fetal growth restriction to preterm labor. Genome-wide gene expression analysis is also being applied to identify prognostic and diagnostic biomarkers of these disorders. In this comprehensive review we summarize transcriptome biology, methods of isolation and analysis, application to placental development and pathology, and use in diagnostic analysis in maternal blood. Key information for analysis methods is organized into quick reference tables where current analysis techniques and tools are cited and compared. We have created this review as a practical guide and starting reference for those interested in beginning an investigation into the transcriptome of the placenta.
Collapse
|
43
|
Gamie Z, MacFarlane RJ, Tomkinson A, Moniakis A, Tran GT, Gamie Y, Mantalaris A, Tsiridis E. Skeletal tissue engineering using mesenchymal or embryonic stem cells: clinical and experimental data. Expert Opin Biol Ther 2015; 14:1611-39. [PMID: 25303322 DOI: 10.1517/14712598.2014.945414] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) can be obtained from a wide variety of tissues for bone tissue engineering such as bone marrow, adipose, birth-associated, peripheral blood, periosteum, dental and muscle. MSCs from human fetal bone marrow and embryonic stem cells (ESCs) are also promising cell sources. AREAS COVERED In vitro, in vivo and clinical evidence was collected using MEDLINE® (1950 to January 2014), EMBASE (1980 to January 2014) and Google Scholar (1980 to January 2014) databases. EXPERT OPINION Enhanced results have been found when combining bone marrow-derived mesenchymal stem cells (BMMSCs) with recently developed scaffolds such as glass ceramics and starch-based polymeric scaffolds. Preclinical studies investigating adipose tissue-derived stem cells and umbilical cord tissue-derived stem cells suggest that they are likely to become promising alternatives. Stem cells derived from periosteum and dental tissues such as the periodontal ligament have an osteogenic potential similar to BMMSCs. Stem cells from human fetal bone marrow have demonstrated superior proliferation and osteogenic differentiation than perinatal and postnatal tissues. Despite ethical concerns and potential for teratoma formation, developments have also been made for the use of ESCs in terms of culture and ideal scaffold.
Collapse
Affiliation(s)
- Zakareya Gamie
- Aristotle University Medical School, 'PapaGeorgiou' Hospital, Academic Orthopaedic Unit , Thessaloniki , Greece
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pipino C, Pierdomenico L, Di Tomo P, Di Giuseppe F, Cianci E, D'Alimonte I, Morabito C, Centurione L, Antonucci I, Mariggiò MA, Di Pietro R, Ciccarelli R, Marchisio M, Romano M, Angelucci S, Pandolfi A. Molecular and phenotypic characterization of human amniotic fluid-derived cells: a morphological and proteomic approach. Stem Cells Dev 2015; 24:1415-1428. [PMID: 25608581 DOI: 10.1089/scd.2014.0453] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal Stem Cells derived from Amniotic Fluid (AFMSCs) are multipotent cells of great interest for regenerative medicine. Two predominant cell types, that is, Epithelial-like (E-like) and Fibroblast-like (F-like), have been previously detected in the amniotic fluid (AF). In this study, we examined the AF from 12 donors and observed the prevalence of the E-like phenotype in 5, whereas the F-like morphology was predominant in 7 samples. These phenotypes showed slight differences in membrane markers, with higher CD90 and lower Sox2 and SSEA-4 expression in F-like than in E-like cells; whereas CD326 was expressed only in the E-like phenotype. They did not show any significant differences in osteogenic, adipogenic or chondrogenic differentiation. Proteomic analysis revealed that samples with a predominant E-like phenotype (HC1) showed a different profile than those with a predominant F-like phenotype (HC2). Twenty-five and eighteen protein spots were differentially expressed in HC1 and HC2 classes, respectively. Of these, 17 from HC1 and 4 from HC2 were identified by mass spectrometry. Protein-interaction networks for both phenotypes showed strong interactions between specific AFMSC proteins and molecular chaperones, such as preproteasomes and mature proteasomes, both of which are important for cell cycle regulation and apoptosis. Collectively, our results provide evidence that, regardless of differences in protein profiling, the prevalence of E-like or F-like cells in AF does not affect the differentiation capacity of AFMSC preparations. This may be valuable information with a view to the therapeutic use of AFMSCs.
Collapse
Affiliation(s)
- Caterina Pipino
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Laura Pierdomenico
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
- 4Department of Medicine and Aging Science, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Pamela Di Tomo
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Fabrizio Di Giuseppe
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Eleonora Cianci
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Iolanda D'Alimonte
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Caterina Morabito
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
- 5Department of Neuroscience and Imaging, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Lucia Centurione
- 3StemTeCh Group, Chieti, Italy
- 4Department of Medicine and Aging Science, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Ivana Antonucci
- 3StemTeCh Group, Chieti, Italy
- 6Psychological Sciences Humanities and Territory, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Maria A Mariggiò
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
- 5Department of Neuroscience and Imaging, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Roberta Di Pietro
- 3StemTeCh Group, Chieti, Italy
- 4Department of Medicine and Aging Science, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Renata Ciccarelli
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Marco Marchisio
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
- 4Department of Medicine and Aging Science, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
| | - Mario Romano
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Stefania Angelucci
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| | - Assunta Pandolfi
- 1Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences "G. d'Annunzio" University Chieti-Pescara, Chieti, Italy
- 2Aging Research Center (Ce.S.I.), "Università G. d'Annunzio" Foundation, Chieti, Italy
- 3StemTeCh Group, Chieti, Italy
| |
Collapse
|
45
|
Prokopyuk V, Prokopyuk O, Musatova I, Shevchenko N, Roenko A, Terehova E, Volina V. Safety of placental, umbilical cord and fetal membrane explants after cryopreservation. ACTA ACUST UNITED AC 2015. [DOI: 10.22494/cot.v3i1.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There have been studied morphological safety and functional state of the explants of human placenta, umbilical cord and fetal membranes by vital staining techniques using the MTT and resazurin reduction tests, level of glucose in incubation medium, activity of lactate dehydrogenase and alkaline phosphatase before and after cryopreservation. It has been found that proposed program of cryopreservation allows keeping a high level of viability of the explants of placenta, umbilical cord and fetal membranes, the most informative methods of assessing the safety of these biological objects before and after cryopreservation are method of vital staining, determination of glucose content in incubation medium, MTT-test and resazurin reduction test.
Collapse
|
46
|
Pipino C, Pandolfi A. Osteogenic differentiation of amniotic fluid mesenchymal stromal cells and their bone regeneration potential. World J Stem Cells 2015; 7:681-690. [PMID: 26029340 PMCID: PMC4444609 DOI: 10.4252/wjsc.v7.i4.681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/16/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
In orthopedics, tissue engineering approach using stem cells is a valid line of treatment for patients with bone defects. In this context, mesenchymal stromal cells of various origins have been extensively studied and continue to be a matter of debate. Although mesenchymal stromal cells from bone marrow are already clinically applied, recent evidence suggests that one may use mesenchymal stromal cells from extra-embryonic tissues, such as amniotic fluid, as an innovative and advantageous resource for bone regeneration. The use of cells from amniotic fluid does not raise ethical problems and provides a sufficient number of cells without invasive procedures. Furthermore, they do not develop into teratomas when transplanted, a consequence observed with pluripotent stem cells. In addition, their multipotent differentiation ability, low immunogenicity, and anti-inflammatory properties make them ideal candidates for bone regenerative medicine. We here present an overview of the features of amniotic fluid mesenchymal stromal cells and their potential in the osteogenic differentiation process. We have examined the papers actually available on this regard, with particular interest in the strategies applied to improve in vitro osteogenesis. Importantly, a detailed understanding of the behavior of amniotic fluid mesenchymal stromal cells and their osteogenic ability is desirable considering a feasible application in bone regenerative medicine.
Collapse
|
47
|
Ye JS, Su XS, Stoltz JF, de Isla N, Zhang L. Signalling pathways involved in the process of mesenchymal stem cells differentiating into hepatocytes. Cell Prolif 2015; 48:157-65. [PMID: 25656979 PMCID: PMC6496737 DOI: 10.1111/cpr.12165] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/01/2014] [Indexed: 12/18/2022] Open
Abstract
End‐stage liver disease can be the termination of acute or chronic liver diseases, with manifestations of liver failure; transplantation is currently an effective treatment for these. However, transplantation is severely limited due to the serious lack of donors, expense, graft rejection and requirement of long‐term immunosuppression. Mesenchymal stem cells (MSCs) have attracted considerable attention as therapeutic tools as they can be obtained with relative ease and expanded in culture, along with features of self‐renewal and multidirectional differentiation. Many scientific groups have sought to use MSCs differentiating into functional hepatocytes to be used in cell transplantation with liver tissue engineering to repair diseased organs. In most of the literature, hepatocyte differentiation refers to use of various additional growth factors and cytokines, such as hepatocyte growth factor (HGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), oncostatin M (OSM) and more, and most are involved in signalling pathway regulation and cell–cell/cell–matrix interactions. Signalling pathways have been shown to play critical roles in embryonic development, tumourigenesis, tumour progression, apoptosis and cell‐fate determination. However, mechanisms of MSCs differentiating into hepatocytes, particularly signalling pathways involved, have not as yet been completely illustrated. In this review, we have focused on progress of signalling pathways associated with mesenchymal stem cells differentiating into hepatocytes along with the stepwise differentiation procedure.
Collapse
Affiliation(s)
- Jun-Song Ye
- BRC, First Hospital of Kun Ming, Kun Ming, 650011, China; Lorraine University and CNRS UMR 7365, Medical College, Vandoeuvre-lès-Nancy, 54500, France
| | | | | | | | | |
Collapse
|
48
|
Ramachandra DL, Shaw SSW, Shangaris P, Loukogeorgakis S, Guillot PV, Coppi PD, David AL. In utero therapy for congenital disorders using amniotic fluid stem cells. Front Pharmacol 2014; 5:270. [PMID: 25566071 PMCID: PMC4271591 DOI: 10.3389/fphar.2014.00270] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/18/2014] [Indexed: 12/15/2022] Open
Abstract
Congenital diseases are responsible for over a third of all pediatric hospital admissions. Advances in prenatal screening and molecular diagnosis have allowed the detection of many life-threatening genetic diseases early in gestation. In utero transplantation (IUT) with stem cells could cure affected fetuses but so far in humans, successful IUT using allogeneic hematopoietic stem cells (HSCs), has been limited to fetuses with severe immunologic defects and more recently IUT with allogeneic mesenchymal stem cell transplantation, has improved phenotype in osteogenesis imperfecta. The options of preemptive treatment of congenital diseases in utero by stem cell or gene therapy changes the perspective of congenital diseases since it may avoid the need for postnatal treatment and reduce future costs. Amniotic fluid stem (AFS) cells have been isolated and characterized in human, mice, rodents, rabbit, and sheep and are a potential source of cells for therapeutic applications in disorders for treatment prenatally or postnatally. Gene transfer to the cells with long-term transgenic protein expression is feasible. Recently, pre-clinical autologous transplantation of transduced cells has been achieved in fetal sheep using minimally invasive ultrasound guided injection techniques. Clinically relevant levels of transgenic protein were expressed in the blood of transplanted lambs for at least 6 months. The cells have also demonstrated the potential of repair in a range of pre-clinical disease models such as neurological disorders, tracheal repair, bladder injury, and diaphragmatic hernia repair in neonates or adults. These results have been encouraging, and bring personalized tissue engineering for prenatal treatment of genetic disorders closer to the clinic.
Collapse
Affiliation(s)
- Durrgah L. Ramachandra
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Steven S. W. Shaw
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Obstetrics and Gynaecology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Panicos Shangaris
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Stavros Loukogeorgakis
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Pascale V. Guillot
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
- Cellular Reprogramming and Perinatal Therapy, Institute for Women’s Health, University College London, London, UK
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine, Institute of Child Health, University College London, London, UK
| | - Anna L. David
- Prenatal Therapy, Institute for Women’s Health, University College London, London, UK
| |
Collapse
|
49
|
Patel J, Shafiee A, Wang W, Fisk NM, Khosrotehrani K. Novel isolation strategy to deliver pure fetal-origin and maternal-origin mesenchymal stem cell (MSC) populations from human term placenta. Placenta 2014; 35:969-71. [PMID: 25239220 DOI: 10.1016/j.placenta.2014.09.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 08/26/2014] [Accepted: 09/02/2014] [Indexed: 12/22/2022]
Abstract
The placenta is an abundant source of mesenchymal stem/stromal cells (MSC). Although presumed of translationally-advantageous fetal origin, the literature instead suggests a high incidence of either contaminating or pure maternal MSC. Despite definitional criteria that MSC are CD34-, increasing evidence suggests that fetal MSC may be CD34 positive in vivo. We flow sorted term placental digests based on CD34+ expression and exploited differential culture media to isolate separately pure fetal and maternal MSC populations. This method has considerable translational implications, in particular to clinical trials underway with "placental" MSC of uncertain or decidual origin.
Collapse
Affiliation(s)
- J Patel
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, 4029, Australia.
| | - A Shafiee
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, 4029, Australia
| | - W Wang
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, 4029, Australia
| | - N M Fisk
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, 4029, Australia; Centre for Advanced Prenatal Care, Women's & Newborn Services, Royal Brisbane & Women's Hospital, Herston, QLD, 4029, Australia
| | - K Khosrotehrani
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, 4029, Australia; The University of Queensland, UQ Diamantina Institute, Translational Research Institute, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
50
|
Gucciardo L, De Koninck P, Verfaillie C, Lories R, Deprest J. Perception and knowledge about stem cell and tissue engineering research: a survey amongst researchers and medical practitioners in perinatology. Stem Cell Rev Rep 2014; 10:447-54. [PMID: 24647632 DOI: 10.1007/s12015-014-9506-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Stem cell and tissue engineering (SC&TE) research remain controversial. Polemics are potential hurdles for raising public funds for research and clinical implementation. In view of future applications of SC&TE in perinatal conditions, we aimed to measure the background knowledge, perceptions or beliefs on SC&TE research among clinicians and academic researchers with perinatal applications on the department's research agenda. MATERIAL AND METHODS We polled three professional categories: general obstetrician gynecologists, perinatologists and basic or translational researchers in development and regeneration. The survey included questions on demographics, work environment, educational background, general knowledge, expectations, opinions and ethical reflections of the respondent about SC&TE. RESULTS The response rate was 39 %. Respondents were mainly female (54 %) and under 40 years (63 %). The general background knowledge about SC&TE is low. Respondents confirm that remaining controversies still arise from the confusion that stem cell research coincides with embryo manipulation. Clinicians assume that stem cell research has reached the level of clinical implementation, and accept the risks associated of purposely harvesting fetal amniotic cells. Researchers in contrast are more cautious about both implementation and risks. CONCLUSION Professionals in the field of perinatology may benefit of a better background knowledge and information on current SC & TE research. Though clinicians may be less aware of the current state of knowledge, they are open to clinical implementation, whereas dedicated researchers remain cautious. In view of the clinical introduction of SC & TE, purposed designed informative action should be taken and safety studies executed, hence avoid sustaining needless polemics.
Collapse
Affiliation(s)
- Léonardo Gucciardo
- Department of Development and Regeneration, Cluster Organ Systems, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|