1
|
Lee SW, Lim YJ, Kim HY, Kim W, Park WT, Ma MJ, Lee J, Seo MS, Kim YI, Park S, Choi SK, Lee GW. Immortalization of epidural fat-derived mesenchymal stem cells: In vitro characterization and adipocyte differentiation potential. World J Stem Cells 2025; 17:98777. [PMID: 39866894 PMCID: PMC11752455 DOI: 10.4252/wjsc.v17.i1.98777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising candidates for regenerative therapy due to their self-renewal capability, multilineage differentiation potential, and immunomodulatory effects. The molecular characteristics of MSCs are influenced by their location. Recently, epidural fat (EF) and EF-derived MSCs (EF-MSCs) have garnered attention due to their potential benefits to the spinal microenvironment and their high expression of neural SC markers. However, their clinical applications are limited due to cell senescence and limited accessibility of EF. Although many studies have attempted to establish an immortalized, stable SC line, the characteristics of immortalized EF-MSCs remain to be clarified. AIM To establish and analyze stable immortalized EF-MSCs. METHODS The phenotypes of EF-MSCs were analyzed using optical microscopy. Cell immortalization was performed using lentiviral vectors. The biomolecular characteristics of the cells were analyzed by immunoblotting, quantitative PCR, and proteomics. RESULTS The immortalized EF-MSCs demonstrated a significantly extended lifespan compared to the control group, with well-preserved adipogenic potential and SC surface marker expression. Introduction of human telomerase reverse transcriptase genes markedly increased the lifespan of EF-MSCs. Proteomics analysis revealed substantial increase in the expression of DNA replication pathway components in immortalized EF-MSCs. CONCLUSION Immortalized EF-MSCs exhibited significantly enhanced proliferative capacity, retained adipogenic potential, and upregulated the expression of DNA replication pathway components.
Collapse
Affiliation(s)
- Seoung-Woo Lee
- Core Protein Resources Center, Daegu-Gyeongbuk Institute of Science and Technology, Daegu 42988, South Korea
| | - Young-Ju Lim
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu 42415, South Korea
| | - Hee-Yeon Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, South Korea
| | - Wansoo Kim
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, South Korea
| | - Wook-Tae Park
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, Daegu 42415, South Korea
| | - Min-Jung Ma
- Laboratory of Veterinary Tissue Engineering, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, South Korea
| | - Junho Lee
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, South Korea
| | - Min-Soo Seo
- Department of Veterinary Tissue Engineering, Kyungpook National University, Daegu 41566, South Korea
| | | | - Sangbum Park
- Michigan State University, Institute for Quantitative Health Science & Engineering, East Lansing, MI 48824, United States
| | - Seong-Kyoon Choi
- Core Protein Resources Center, Daegu-Gyeongbuk Institute of Science and Technology, Daegu 42988, South Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Daegu 42415, South Korea.
| |
Collapse
|
2
|
Li Z, Xu Z, Zhu L, Qin T, Ma J, Feng Z, Yue H, Guan Q, Zhou B, Han G, Zhang G, Li C, Jia S, Qiu Q, Hao D, Wang Y, Wang W. High-quality sika deer omics data and integrative analysis reveal genic and cellular regulation of antler regeneration. Genome Res 2025; 35:188-201. [PMID: 39542648 PMCID: PMC11789637 DOI: 10.1101/gr.279448.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
The antler is the only organ that can fully regenerate annually in mammals. However, the regulatory pattern and mechanism of gene expression and cell differentiation during this process remain largely unknown. Here, we obtain comprehensive assembly and gene annotation of the sika deer (Cervus nippon) genome. We construct, together with large-scale chromatin accessibility and gene expression data, gene regulatory networks involved in antler regeneration, identifying four transcription factors, MYC, KLF4, NFE2L2, and JDP2, with high regulatory activity across the whole regeneration process. Comparative studies and luciferase reporter assay suggest the MYC expression driven by a cervid-specific regulatory element might be important for antler regenerative ability. We further develop a model called combinatorial TF Oriented Program (cTOP), which integrates single-cell data with bulk regulatory networks and find PRDM1, FOSL1, BACH1, and NFATC1 as potential pivotal factors in antler stem cell activation and osteogenic differentiation. Additionally, we uncover interactions within and between cell programs and pathways during the regeneration process. These findings provide insights into the gene and cell regulatory mechanisms of antler regeneration, particularly in stem cell activation and differentiation.
Collapse
Affiliation(s)
- Zihe Li
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ziyu Xu
- CEMS, NCMIS, HCMS, MADIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Zhu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an, Shaanxi 710054, China
| | - Tao Qin
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jinrui Ma
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhanying Feng
- CEMS, NCMIS, HCMS, MADIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- Department of Statistics, Department of Biomedical Data Science, Bio-X Program, Stanford University, Stanford, California 94305, USA
| | - Huishan Yue
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Qing Guan
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Botong Zhou
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ge Han
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, 130600 Changchun, China
| | - Shuaijun Jia
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an, Shaanxi 710054, China
| | - Qiang Qiu
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China;
| | - Dingjun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China;
- Shaanxi Key Laboratory of Spine Bionic Treatment, Xi'an, Shaanxi 710054, China
| | - Yong Wang
- CEMS, NCMIS, HCMS, MADIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China;
- School of Mathematics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Wen Wang
- New Cornerstone Science Laboratory, Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an 710072, China;
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| |
Collapse
|
3
|
Yamaguchi N, Horio E, Sonoda J, Yamagishi M, Miyakawa S, Murakami F, Hasegawa H, Katahira Y, Mizoguchi I, Fujii Y, Chikazu D, Yoshimoto T. Immortalization of Mesenchymal Stem Cells for Application in Regenerative Medicine and Their Potential Risks of Tumorigenesis. Int J Mol Sci 2024; 25:13562. [PMID: 39769322 PMCID: PMC11676347 DOI: 10.3390/ijms252413562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Regenerative medicine utilizes stem cells to repair damaged tissues by replacing them with their differentiated cells and activating the body's inherent regenerative abilities. Mesenchymal stem cells (MSCs) are adult stem cells that possess tissue repair and regenerative capabilities and immunomodulatory properties with a much lower risk of tumorigenicity, making them a focus of numerous clinical trials worldwide. MSCs primarily exert their therapeutic effects through paracrine effects via secreted factors, such as cytokines and exosomes. This has led to increasing interest in cell-free therapy, where only the conditioned medium (also called secretome) from MSC cultures is used for regenerative applications. However, MSCs face certain limitations, including cellular senescence, scarcity, donor heterogeneity, complexity, short survival post-implantation, and regulatory and ethics hurdles. To address these challenges, various types of immortalized MSCs (ImMSCs) capable of indefinite expansion have been developed. These cells offer significant promise and essential tools as a reliable source for both cell-based and cell-free therapies with the aim of translating them into practical medicine. However, the process of immortalization, often involving the transduction of immortalizing genes, poses potential risks of genetic instability and resultant malignant transformation. Cell-free therapy is particularly attractive, as it circumvents the risks of tumorigenicity and ethical concerns associated with live cell therapies. Rigorous safety tests, such as monitoring chromosomal abnormalities, are critical to ensure safety. Technologies like inducible or suicide genes may allow for the controlled proliferation of MSCs and induce apoptosis after their therapeutic task is completed. This review highlights recent advancements in the immortalization of MSCs and the associated risks of tumorigenesis.
Collapse
Affiliation(s)
- Natsuki Yamaguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Eri Horio
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Jukito Sonoda
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Miu Yamagishi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Satomi Miyakawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Fumihiro Murakami
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Yasuhiro Katahira
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| | - Yasuyuki Fujii
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Daichi Chikazu
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan
| |
Collapse
|
4
|
Guillot-Ferriols M, Costa CM, Correia DM, Rodríguez-Hernández JC, Tsimbouri PM, Lanceros-Méndez S, Dalby MJ, Gómez Ribelles JL, Gallego-Ferrer G. Piezoelectric Stimulation Induces Osteogenesis in Mesenchymal Stem Cells Cultured on Electroactive Two-Dimensional Substrates. ACS APPLIED POLYMER MATERIALS 2024; 6:13710-13722. [PMID: 39606252 PMCID: PMC11590054 DOI: 10.1021/acsapm.4c02485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Physical cues have been shown to be effective in inducing osteogenic differentiation of mesenchymal stem cells (MSCs). Here, we propose piezoelectric stimulation as a potential osteogenic cue mimicking the electroactive properties of bone's extracellular matrix. When combined with a magnetostrictive component, piezoelectric polymers can be used for MSC stimulation by applying an external magnetic field. The deformation of the magnetostrictive component produces a deformation in the polymer matrix, generating a change in the surface charge that induces an electric field that can be transmitted to the cells. Cell adhesion, cytoskeleton changes, and metabolomics are the first evidence of MSC osteoblastogenesis and can be used to study initial MSC response to this kind of stimulation. In the current study, poly(vinylidene) fluoride (PVDF) piezoelectric films with and without cobalt ferrite oxide (CFO) crystallized from the melt in the presence of the ionic liquid 1-butyl-3-methyl-imidazolium chloride ([Bmim][Cl]) were produced. [Bmim][Cl] allowed the production of the β-phase, the most electroactive phase, even without CFO. After ionic liquid removal, PVDF and PVDF-CFO films presented high percentages of the β-phase and similar crystalline content. Incorporating CFO nanoparticles was effective, allowing the electromechanical stimulation of MSCs by applying a magnetic field with a bioreactor. Before stimulation, the initial response of MSCs was characterized in static conditions, showing that the produced films were biocompatible and noncytotoxic, allowing MSC adhesion and proliferation in the short term. Stimulation experiments revealed that MSCs electromechanically stimulated for 3 days in PVDF-CFO supports showed longer focal adhesions and decreased vimentin cytoskeletal density, both signals of early osteogenic differentiation. Furthermore, they rearranged their energy metabolism toward an osteogenic phenotype after 7 days of culture under the same stimulation. The results prove that MSCs respond to electromechanical stimulation by osteogenic differentiation.
Collapse
Affiliation(s)
- Maria Guillot-Ferriols
- Center for
Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Valencia 46022, Spain
- Biomedical
Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia 46022, Spain
| | - Carlos M. Costa
- Physics Centre
of Minho and Porto Universities (CF-UM-UP) and Laboratory of Physics
for Materials and Emergent Technologies, LapMET, University of Minho, Braga 4710-057, Portugal
- Institute
of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, Braga 4710-057, Portugal
| | | | | | - Penelope M. Tsimbouri
- Center for
the Cellular Microenvironment, School of Molecular Biosciences, College
of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United
Kingdom
| | - Senentxu Lanceros-Méndez
- Physics Centre
of Minho and Porto Universities (CF-UM-UP) and Laboratory of Physics
for Materials and Emergent Technologies, LapMET, University of Minho, Braga 4710-057, Portugal
- BCMaterials,
Basque Center for Materials, Applications and Nanostructures, UPV/EHU, Science Park, Leioa 48940, Spain
- Basque Foundation
for Science, IKERBASQUE, Bilbao 48009, Spain
| | - Matthew J. Dalby
- Center for
the Cellular Microenvironment, School of Molecular Biosciences, College
of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United
Kingdom
| | - José Luis Gómez Ribelles
- Center for
Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Valencia 46022, Spain
- Biomedical
Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia 46022, Spain
| | - Gloria Gallego-Ferrer
- Center for
Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Valencia 46022, Spain
- Biomedical
Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia 46022, Spain
| |
Collapse
|
5
|
Hassanpour Khodaei S, Sabetkam S, Kalarestaghi H, Dizaji Asl K, Mazloumi Z, Bahramloo M, Norouzi N, Naderali E, Rafat A. Mesenchymal stem cells and mesenchymal stem cell-derived exosomes: attractive therapeutic approaches for female reproductive dysfunction. Mol Biol Rep 2024; 52:10. [PMID: 39576370 DOI: 10.1007/s11033-024-10106-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Infertility is a reproductive health problem in the male or female reproductive system. Traditional assisted reproductive technology (ART) has been unable to solve various cases of infertility for years. Clinical researchers have sought to treat infertility using new methods that are more effective and noninvasive than the old methods. Recently, Mesenchymal stem cells (MSCs) and MSCs-derived Exosomes (MSC-Exos) via paracrine activity play an important role in treating various causes of infertility and improving pregnancy outcomes. In this review, we focus on the roles of MSCs and MSC-Exos cell therapy in female infertility in the different types of female reproductive disorders.
Collapse
Affiliation(s)
- Sepideh Hassanpour Khodaei
- Department of Dentistry, Eastern Mediterranean University (EMU) Famagusta, North Cyprus Mersin 10, Famagusta, Turkey
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, University of Kyrenia, Kyrenia, Northern Cyprus
| | - Hossein Kalarestaghi
- Research Laboratory for Embryology and Stem Cell, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Khadijeh Dizaji Asl
- Department of Histopathology and Anatomy, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Zeinab Mazloumi
- Department of Medical Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadmahdi Bahramloo
- Department of Medical Sciences, Student Research Committee, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Nahid Norouzi
- Nursing Trauma Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Elahe Naderali
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Rivera Orsini MA, Ozmen EB, Miles A, Newby SD, Springer N, Millis D, Dhar M. Isolation and Characterization of Canine Adipose-Derived Mesenchymal Stromal Cells: Considerations in Translation from Laboratory to Clinic. Animals (Basel) 2024; 14:2974. [PMID: 39457904 PMCID: PMC11503832 DOI: 10.3390/ani14202974] [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: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
In allogeneic MSC implantation, the cells are isolated from a donor different from the recipient. When tested, allogeneic MSCs have several advantages over autologous ones: faster cell growth, sufficient cell concentration, and readily available cells for clinics. To ensure the safe and efficient use of allogeneic MSCs in clinics, the MSCs need to be first tested in vitro. With this study, we paved the way by addressing the in vitro aspects of canine adipose-derived MSCs, considering the limited studies on the clinical use of canine cells. We isolated cAD-MSCs from canine falciform ligament fat and evaluated their viability and proliferation using an MTS assay. Then, we characterized the MSC-specific antigens using immunophenotyping and immunofluorescence and demonstrated their potential for in vitro differentiation. Moreover, we established shipping and cryobanking procedures to lead the study to become an off-the-shelf therapy. During expansion, the cells demonstrated a linear increase in cell numbers, confirming their proliferation quantitatively. The cells showed viability before and after cryopreservation, demonstrating that cell viability can be preserved. From a clinical perspective, the established shipping conditions demonstrated that the cells retain their viability for up to 48 h. This study lays the groundwork for the potential use of allogeneic cAD-MSCs in clinical applications.
Collapse
Affiliation(s)
- Michael A. Rivera Orsini
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| | - Emine Berfu Ozmen
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
- Genome Science and Technology, University of Tennessee Knoxville, Knoxville, TN 37996, USA
| | - Alyssa Miles
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (A.M.); (N.S.)
| | - Steven D. Newby
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| | - Nora Springer
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (A.M.); (N.S.)
| | - Darryl Millis
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| | - Madhu Dhar
- Regenerative Medicine and Tissue Engineering, Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA; (M.A.R.O.); (E.B.O.); (S.D.N.)
| |
Collapse
|
7
|
Azevedo PL, Maradei S, de Sá Bigni R, Santos Ramires Aragao J, Abdelhay E, Binato R. SLPI overexpression in hMSCs could be implicated in the HSC gene expression profile in AML. Sci Rep 2024; 14:15550. [PMID: 38969699 PMCID: PMC11226598 DOI: 10.1038/s41598-024-66400-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a severe haematological neoplasm that originates from the transformation of haematopoietic stem cells (HSCs) into leukaemic stem cells (LSCs). The bone marrow (BM) microenvironment, particularly that of mesenchymal stromal cells (hMSCs), plays a crucial role in the maintenance of HSCs. In this context, we explored whether alterations in the secretome of hMSCs derived from AML patients (hMSC-AML) could impact HSC gene expression. Proteomic analysis revealed that the secretome of coculture assays with hMSC-AMLs and HSC from healthy donor is altered, with increased levels of secretory leukocyte protease inhibitor (SLPI), a protein associated with important processes for maintenance of the haematopoietic niche that has already been described to be altered in several tumours. Increased SLPI expression was also observed in the BM plasma of AML patients. Transcriptome analysis of HSCs cocultured with hMSC-AML in comparison with HSCs cocultured with hMSC-HD revealed altered expression of SLPI target genes associated with the cell cycle, proliferation, and apoptosis. Important changes were identified, such as increased expression levels of CCNA2, CCNE2, CCND2, CD133 and CDK1 and decreased levels of CDKN2A and IGFBP3, among others. Overall, these findings suggest that the altered secretome of coculture assays with hMSC-AMLs and HSC from healthy donor, particularly increased SLPI expression, can contribute to gene expression changes in HSCs, potentially influencing important molecular mechanisms related to AML development and progression.
Collapse
Affiliation(s)
- Pedro L Azevedo
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil.
| | - Simone Maradei
- Bone Marrow Transplantation Unit, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | - Ricardo de Sá Bigni
- Haematology Service, National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
| | | | - Eliana Abdelhay
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil
| | - Renata Binato
- Stem Cell Laboratory, Lab. de Células-Tronco (LCT) Centro, National Cancer Institute (INCA), Praça da Cruz Vermelha 23, 6° andar, Ala C, Rio de Janeiro, RJ, CEP: 20230-130, Brazil
| |
Collapse
|
8
|
Ghufran H, Azam M, Mehmood A, Umair M, Baig MT, Tasneem S, Butt H, Riazuddin S. Adipose Tissue and Umbilical Cord Tissue: Potential Sources of Mesenchymal Stem Cells for Liver Fibrosis Treatment. J Clin Exp Hepatol 2024; 14:101364. [PMID: 38449506 PMCID: PMC10912848 DOI: 10.1016/j.jceh.2024.101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/11/2024] [Indexed: 03/08/2024] Open
Abstract
Background/Aims Mesenchymal stem cells (MSCs) are potential alternatives for liver fibrosis treatment; however, their optimal sources remain uncertain. This study compares the ex-vivo expansion characteristics of MSCs obtained from adipose tissue (AT) and umbilical cord (UC) and assesses their therapeutic potential for liver fibrosis treatment. Methods Since MSCs from early to mid-passage numbers (P2-P6) are preferable for cellular therapy, we investigated the growth kinetics of AT-MSCs and UC-MSCs up to P6 and evaluated their therapeutic effects in a rat model of liver fibrosis induced by diethylnitrosamine. Results Results from the expansion studies demonstrated that both cell types exhibited bona fide characteristics of MSCs, including surface antigens, pluripotent gene expression, and differentiation potential. However, AT-MSCs demonstrated a shorter doubling time (58.2 ± 7.3 vs. 82.3 ± 4.3 h; P < 0.01) and a higher population doubling level (10.1 ± 0.7 vs. 8.2 ± 0.3; P < 0.01) compared to UC-MSCs, resulting in more cellular yield (230 ± 9.0 vs. 175 ± 13.2 million) in less time. Animal studies demonstrated that both MSC types significantly reduced liver fibrosis (P < 0.05 vs. the control group) while also improving liver function and downregulating fibrosis-associated gene expression. Conclusion AT-MSCs and UC-MSCs effectively reduce liver fibrosis. However, adipose cultures display an advantage by yielding a higher number of MSCs in a shorter duration, rendering them a viable choice for scenarios requiring immediate single-dose administration, often encountered in clinical settings.
Collapse
Affiliation(s)
- Hafiz Ghufran
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Maryam Azam
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Azra Mehmood
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Muhammad Umair
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Maria T. Baig
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Saba Tasneem
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Hira Butt
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Sheikh Riazuddin
- Jinnah Burn and Reconstructive Surgery Centre, Allama Iqbal Medical College, Lahore, Pakistan
| |
Collapse
|
9
|
Velikova T, Dekova T, Miteva DG. Controversies regarding transplantation of mesenchymal stem cells. World J Transplant 2024; 14:90554. [PMID: 38947963 PMCID: PMC11212595 DOI: 10.5500/wjt.v14.i2.90554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 06/13/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have tantalized regenerative medicine with their therapeutic potential, yet a cloud of controversies looms over their clinical transplantation. This comprehensive review navigates the intricate landscape of MSC controversies, drawing upon 15 years of clinical experience and research. We delve into the fundamental properties of MSCs, exploring their unique immunomodulatory capabilities and surface markers. The heart of our inquiry lies in the controversial applications of MSC transplantation, including the perennial debate between autologous and allogeneic sources, concerns about efficacy, and lingering safety apprehensions. Moreover, we unravel the enigmatic mechanisms surrounding MSC transplantation, such as homing, integration, and the delicate balance between differentiation and paracrine effects. We also assess the current status of clinical trials and the ever-evolving regulatory landscape. As we peer into the future, we examine emerging trends, envisioning personalized medicine and innovative delivery methods. Our review provides a balanced and informed perspective on the controversies, offering readers a clear understanding of the complexities, challenges, and potential solutions in MSC transplantation.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Department of Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Tereza Dekova
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
| | | |
Collapse
|
10
|
Farag M, Rezk R, Hutchinson H, Zankevich A, Lucke‐Wold B. Intervertebral disc degeneration and regenerative medicine. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/15/2024] [Indexed: 01/08/2025]
Abstract
AbstractIntervertebral disc (IVD) degeneration is a common phenomenon that affects patients with increasing prevalence with increasing age. Both conservative treatments, such as the use of pain medication or physical therapy, and surgical treatments, such as fusion or disc replacement therapies, are offered to patients. Both non‐invasive and invasive treatments have been shown to improve pain and quality of life for patients. This review explores the role of regenerative medicine techniques as a promising therapeutic intervention that can be used before or in combination with conservative therapy and surgery to enhance the treatment process in patients with IVD degeneration or disc pathology. Currently, there are four major modules of regenerative medicine: genetic therapy, platelet‐rich plasma therapy, stem cell transplantation and tissue engineering. Several research studies have shown promising outcomes of stem cell transplantation and tissue engineering when combined with either surgical or conservative treatment, resulting in improved pain outcomes. The additional benefit of regenerative medicine techniques, specifically stem cell transplantation, is the potential for treating the root pathology of degeneration. Regenerative medicine techniques also have the potential to either halt or reverse degeneration as opposed to current standards of care for managing symptoms. There is a plethora of current research highlighting the benefits of regenerative medicine techniques; however, there remains clinical concerns and ethical concerns regarding the use of regenerative therapy techniques such as stem cell transplantation in the context of IVD degeneration.
Collapse
Affiliation(s)
| | - Rogina Rezk
- University of Florida Gainesville Florida USA
| | | | | | | |
Collapse
|
11
|
Yoon JY, Vu HT, Lee JH, Shin JS, Kim HW, Lee HH, Kim JB, Lee JH. Evaluation of Human Platelet Lysate as an Alternative to Fetal Bovine Serum for Potential Clinical Applications of Stem Cells from Human Exfoliated Deciduous Teeth. Cells 2024; 13:847. [PMID: 38786069 PMCID: PMC11120611 DOI: 10.3390/cells13100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
In recent years, there has been a surge in demand for and research focus on cell therapy, driven by the tissue-regenerative and disease-treating potentials of stem cells. Among the candidates, dental pulp stem cells (DPSCs) or human exfoliated deciduous teeth (SHED) have garnered significant attention due to their easy accessibility (non-invasive), multi-lineage differentiation capability (especially neurogenesis), and low immunogenicity. Utilizing these stem cells for clinical purposes requires careful culture techniques such as excluding animal-derived supplements. Human platelet lysate (hPL) has emerged as a safer alternative to fetal bovine serum (FBS) for cell culture. In our study, we assessed the impact of hPL as a growth factor supplement for culture medium, also conducting a characterization of SHED cultured in hPL-supplemented medium (hPL-SHED). The results showed that hPL has effects in enhancing cell proliferation and migration and increasing cell survivability in oxidative stress conditions induced by H2O2. The morphology of hPL-SHED exhibited reduced size and elongation, with a differentiation capacity comparable to or even exceeding that of SHED cultured in a medium supplemented with fetal bovine serum (FBS-SHED). Moreover, no evidence of chromosome abnormalities or tumor formation was detected. In conclusion, hPL-SHED emerges as a promising candidate for cell therapy, exhibiting considerable potential for clinical investigation.
Collapse
Affiliation(s)
- Ji-Young Yoon
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
| | - Huong Thu Vu
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Pediatric Dentistry, Faculty of Odonto-Stomatology, University of Medincine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 17000, Vietnam
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Jun Hee Lee
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Ji-Sun Shin
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Hae-Won Kim
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Jong-Bin Kim
- Department of Pediatric Dentistry, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea;
| | - Jung-Hwan Lee
- Institute for Stem Cell & Matters, Cell & Matter Corporation, Cheonan 31116, Republic of Korea; (J.-Y.Y.); (J.H.L.); (H.-W.K.)
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea; (H.T.V.); (H.-H.L.)
- Department of Biomaterials Science, College of Dentistry, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| |
Collapse
|
12
|
Sarvari A, Niasari-Naslaji A, Shirazi A, Heidari B, Boroujeni SB, Moradi MH, Naderi MM, Behzadi B, Mehrazar MM, Dehghan MM. Effect of Intra-ovarian Injection of Mesenchymal Stem Cells or its Conditioned Media on Repeated OPU-IVEP Outcomes in Jersey Heifers and Its Relationship with Follicular Fluid Inflammatory Markers. Avicenna J Med Biotechnol 2024; 16:16-28. [PMID: 38605741 PMCID: PMC11005394 DOI: 10.18502/ajmb.v16i1.14167] [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: 07/27/2023] [Accepted: 08/26/2023] [Indexed: 04/13/2024] Open
Abstract
Background Repeated Ovum Pick Up (OPU) could have a detrimental effect on ovarian function, reducing In Vitro Embryo Production (IVEP). The present study examined the therapeutic effect of adipose-derived Mesenchymal Stem Cells (MSCs) or its Conditioned Medium (ConM) on ovarian trauma following repeated OPU. Resolvin E1 (RvE1) and Interleukin-12 (IL-12) were investigated as biomarkers. Methods Jersey heifers (n=8) experienced 11 OPU sessions including 5 pre-treatment and 6 treatment sessions. Heifers received intra-ovarian administration of MSCs or ConM (right ovary) and Dulbecco's Modified Phosphate Buffer Saline (DMPBS; left ovary) after OPU in sessions 5 and 8 and 2 weeks after session 11. The concentrations of RvE1 and IL-12 in follicular fluid was evaluated on sessions 1, 5, 6, 9, and 4 weeks after session 11. Following each OPU session, the IVEP parameters were recorded. Results Intra-ovarian administration of MSCs, ConM, and DMPBS did not affect IVEP parameters (p>0.05). The concentration of IL-12 in follicular fluid increased at the last session of pre-treatment (Session 5; p<0.05) and remained elevated throughout the treatment period. There was no correlation between IL-12 and IVEP parameters (p>0.05). However, RvE1 remained relatively high during the pre-treatment and decreased toward the end of treatment period (p<0.05). This in turn was associated with decline in some IVEP parameters (p<0.05). Conclusion Intra-ovarian administration of MSCs or ConM during repeated OPU did not enhance IVEP outcomes in Bos taurus heifers. The positive association between RvE1 and some of IVEP parameters could nominate RvE1 as a promising biomarker to predict IVEP parameters following repeated OPU.
Collapse
Affiliation(s)
- Ali Sarvari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir Niasari-Naslaji
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Banafsheh Heidari
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Sara Borjian Boroujeni
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Moradi
- Department of Animal Sciences, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
| | - Mohammad-Mahdi Naderi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Bahareh Behzadi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Mahdi Mehrazar
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
13
|
Lu B, Klomjit N, Zhu XY, Jordan KL, Grande JP, Lerman LO. A Pseudotumor in a Mouse Kidney Following Human MSCs Injection. Stem Cell Rev Rep 2023; 19:2551-2553. [PMID: 37310668 PMCID: PMC10592440 DOI: 10.1007/s12015-023-10572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 06/14/2023]
Affiliation(s)
- Bo Lu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Nattawat Klomjit
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kyra L Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Joseph P Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
14
|
Wu Y, Li B, Yu D, Zhou Z, Shen M, Jiang F. CBX7 Rejuvenates Late Passage Dental Pulp Stem Cells by Maintaining Stemness and Pro-angiogenic Ability. Tissue Eng Regen Med 2023; 20:473-488. [PMID: 36920677 PMCID: PMC10219923 DOI: 10.1007/s13770-023-00521-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/08/2023] [Accepted: 01/13/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Ever-growing tissue regeneration causes pressing need for large population of stem cells. However, extensive cell expansion eventually leads to impaired regenerative potentials. In this study, chromobox protein homolog 7 (CBX7) was overexpressed to rejuvenate late passage dental pulp stem cells (DPSCs-P9). METHODS The recruitment of copper ions (Cu2+)-activated hypoxia-inducible factor-1α (HIF-1α) to the CBX7 gene promoter was confirmed by chromatin immunoprecipitation assay. Functions subsequent to Cu2+-induced or recombinant overexpression of CBX7 on proliferation, multipotency, odontoblastic differentiation and angiogenesis were investigated in vitro, while murine subcutaneous transplantation model was used to further detect the effects of Cu2+-induced CBX7 overexpression in vivo. RESULTS Our data displayed that CBX7 overexpression maintain proliferation and multipotency of DPSCs-P9 almost as strong as those of DPSCs-P3. Both gene level of odontoblast-lineage markers and calcium precipitation were nearly the same between CBX7 overexpressed DPSCs-P9 and normal DPSCs-P3. Moreover, we also found upregulated expression of vascular endothelial growth factor in DPSCs-P9 with CBX7 overexpression, which increased the number of capillary-like structures and migrating co-cultured human umbilical vein endothelial cells as well. These findings indicate CBX7 as an effective factor to rejuvenate late passage stem cells insusceptible to cell expansion. Cu2+ has been proved to achieve CBX7 overexpression in DPSCs through the initiation of HIF-1α-CBX7 cascade. Under Cu2+ stimulation since P3, DPSCs-P9 exhibited ameliorated regenerative potential both in vitro and in vivo. CONCLUSION Long-term stimulation of Cu2+ to overexpress CBX7 could be a new strategy to manufacture large population of self-renewing stem cells.
Collapse
Affiliation(s)
- Yu Wu
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China
| | - Bing Li
- Department of Oral Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 1, Shanghai Road, Nanjing, 210029, China
| | - Dandan Yu
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 1, Shanghai Road, Nanjing, 210029, China
| | - Zhixuan Zhou
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China.
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 1, Shanghai Road, Nanjing, 210029, China.
| | - Ming Shen
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China.
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 1, Shanghai Road, Nanjing, 210029, China.
| | - Fei Jiang
- Jiangsu Key Laboratory of Oral Diseases, Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 140, Hanzhong Road, Nanjing, 210029, China.
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, No. 1, Shanghai Road, Nanjing, 210029, China.
| |
Collapse
|
15
|
Kim SG, George NP, Hwang JS, Park S, Kim MO, Lee SH, Lee G. Human Bone Marrow-Derived Mesenchymal Stem Cell Applications in Neurodegenerative Disease Treatment and Integrated Omics Analysis for Successful Stem Cell Therapy. Bioengineering (Basel) 2023; 10:bioengineering10050621. [PMID: 37237691 DOI: 10.3390/bioengineering10050621] [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: 04/27/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Neurodegenerative diseases (NDDs), which are chronic and progressive diseases, are a growing health concern. Among the therapeutic methods, stem-cell-based therapy is an attractive approach to NDD treatment owing to stem cells' characteristics such as their angiogenic ability, anti-inflammatory, paracrine, and anti-apoptotic effects, and homing ability to the damaged brain region. Human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) are attractive NDD therapeutic agents owing to their widespread availability, easy attainability and in vitro manipulation and the lack of ethical issues. Ex vivo hBM-MSC expansion before transplantation is essential because of the low cell numbers in bone marrow aspirates. However, hBM-MSC quality decreases over time after detachment from culture dishes, and the ability of hBM-MSCs to differentiate after detachment from culture dishes remains poorly understood. Conventional analysis of hBM-MSCs characteristics before transplantation into the brain has several limitations. However, omics analyses provide more comprehensive molecular profiling of multifactorial biological systems. Omics and machine learning approaches can handle big data and provide more detailed characterization of hBM-MSCs. Here, we provide a brief review on the application of hBM-MSCs in the treatment of NDDs and an overview of integrated omics analysis of the quality and differentiation ability of hBM-MSCs detached from culture dishes for successful stem cell therapy.
Collapse
Affiliation(s)
- Seok Gi Kim
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Nimisha Pradeep George
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Ji Su Hwang
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Seokho Park
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Biomedical Science, Graduate School of Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Soo Hwan Lee
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Biomedical Science, Graduate School of Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Suwon 16499, Republic of Korea
- Department of Physiology, Ajou University School of Medicine, 206 World Cup-ro, Suwon 16499, Republic of Korea
| |
Collapse
|
16
|
Jankovic MG, Stojkovic M, Bojic S, Jovicic N, Kovacevic MM, Ivosevic Z, Juskovic A, Kovacevic V, Ljujic B. Scaling up human mesenchymal stem cell manufacturing using bioreactors for clinical uses. Curr Res Transl Med 2023; 71:103393. [PMID: 37163885 DOI: 10.1016/j.retram.2023.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are multipotent cells and an attractive therapeutic agent in regenerative medicine and intensive clinical research. Despite the great potential, the limitation that needs to be overcome is the necessity of ex vivo expansion because of insufficient number of hMSCs presented within adult organs and the high doses required for a transplantation. As a result, numerous research studies aim to provide novel expansion methods in order to achieve appropriate numbers of cells with preserved therapeutic quality. Bioreactor-based cell expansion provide high-level production of hMSCs in accordance with good manufacturing practice (GMP) and quality standards. This review summarizes current knowledge about the hMSCs manufacturing platforms with a main focus to the application of bioreactors for large-scale production of GMP-grade hMSCs.
Collapse
Affiliation(s)
- Marina Gazdic Jankovic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia.
| | | | - Sanja Bojic
- Newcastle University, School of Computing, Newcastle upon Tyne, UK
| | - Nemanja Jovicic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Marina Miletic Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Zeljko Ivosevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| | - Aleksandar Juskovic
- Department of Orthopaedic Surgery, Clinical Centre of Montenegro, 81110 Podgorica, Montenegro
| | - Vojin Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Surgery, Serbia
| | - Biljana Ljujic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| |
Collapse
|
17
|
Li Y, Li D, You L, Deng T, Pang Q, Meng X, Zhu B. dCas9-Based PDGFR-β Activation ADSCs Accelerate Wound Healing in Diabetic Mice through Angiogenesis and ECM Remodeling. Int J Mol Sci 2023; 24:ijms24065949. [PMID: 36983022 PMCID: PMC10057415 DOI: 10.3390/ijms24065949] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The chronic wound represents a serious disease characterized by a failure to heal damaged skin and surrounding soft tissue. Mesenchymal stem cells (MSCs) derived from adipose tissue (ADSCs) are a promising therapeutic strategy, but their heterogeneity may result in varying or insufficient therapeutic capabilities. In this study, we discovered that all ADSCs populations expressed platelet-derived growth factor receptor β (PDGFR-β), while the expression level decreased dynamically with passages. Thus, using a CRISPRa-based system, we endogenously overexpressed PDGFR-β in ADSCs. Moreover, a series of in vivo and in vitro experiments were conducted to determine the functional changes in PDGFR-β activation ADSCs (AC-ADSCs) and to investigate the underlying mechanisms. With the activation of PDGFR-β, AC-ADSCs exhibited enhanced migration, survival, and paracrine capacity relative to control ADSCs (CON-ADSCs). In addition, the secretion components of AC-ADSCs contained more pro-angiogenic factors and extracellular matrix-associated molecules, which promoted the function of endothelial cells (ECs) in vitro. Additionally, in in vivo transplantation experiments, the AC-ADSCs transplantation group demonstrated improved wound healing rates, stronger collagen deposition, and angiogenesis. Consequently, our findings revealed that PDGFR-β overexpression enhanced the migration, survival, and paracrine capacity of ADSCs and improved therapeutic effects after transplantation to diabetic mice.
Collapse
Affiliation(s)
- Yumeng Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Deyong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
18
|
Volkova MV, Boyarintsev VV, Trofimenko AV, Kovaleva EV, Othman AA, Melerzanov AV, Filkov GI, Rybalkin SP, Durymanov MO. Local injection of bone-marrow derived mesenchymal stromal cells alters a molecular expression profile of a contact frostbite injury wound and improves healing in a rat model. Burns 2023; 49:432-443. [PMID: 35610075 DOI: 10.1016/j.burns.2022.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Frostbite is a traumatic injury of the tissues upon low temperature environment exposure, which is characterized by direct cell injury due to freezing-thawing followed by development of an acute inflammatory process. Severe frostbite can lead to necrosis of soft tissues and loss of a limb. Mesenchymal stromal cells (MSCs) have a unique ability to modulate pathogenic immune response by secretion of paracrine factors, which suppress inflammation and mediate more efficient tissue regeneration. It should be noted that potential of stem cell therapy for frostbite injury treatment has not been investigated so far. Here, we evaluated a healing capacity of bone-marrow derived MSCs for the treatment of contact frostbite injury wound in a rat model. METHODS Cold-contact injury in a Wistar rat model was induced by 1-minute tight application of the cooled probe (-196 ⁰C) to the skin surface of the left hip. Rat bone marrow MSCs were phenotypically characterized and used for local injections into non-damaged tissues surrounding the wound of animals from the experimental group. The second group of rats was treated in the same manner with 1 mL of isotonic sodium chloride solution. Analysis of cytokine and growth factor expression profile in сold-contact injury wounds was performed on days 5, 9, and 16 using immunoblotting and enzyme-linked immunosorbent assay. Animal recovery in MSC-treated and vehicle-treated groups was evaluated by several criteria including body weight recording, determination of eschar desquamation and re-epithelialization terms, assessment of wound closure kinetics, and histological scoring of the wounds on day 23. RESULTS It turned out that a single subcutaneous administration of MSCs around the wound site resulted in elevated expression of pro-survival and pro-angiogenic VEGF-A and PDGF and 3-5-fold decrease in pro-inflammatory IL-1β as compared with the frostbite wound treated with a vehicle. Moreover, treatment with MSCs caused accelerated wound re-epithelialization (p < 0.05) as well as a better histological score of the MSC-treated wounds. CONCLUSIONS Thus, our data suggested that the use of MSCs is a promising therapeutic strategy for the treatment of cold-induced injury wounds.
Collapse
Affiliation(s)
- Marina V Volkova
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Valery V Boyarintsev
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Alexander V Trofimenko
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Elena V Kovaleva
- Research Center of Toxicology and Hygienic Regulation of Biopreparations, NRC Institute of Immunology FMBA of Russia, Ul. Lenina 102A, Dashkovka, Serpukhov district, Moscow Region 142253, Russia
| | - Aya Al Othman
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Alexander V Melerzanov
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Gleb I Filkov
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Sergey P Rybalkin
- Research Center of Toxicology and Hygienic Regulation of Biopreparations, NRC Institute of Immunology FMBA of Russia, Ul. Lenina 102A, Dashkovka, Serpukhov district, Moscow Region 142253, Russia
| | - Mikhail O Durymanov
- Moscow Institute of Physics and Technology (National Research University), Institutsky per. 9, Dolgoprudny, Moscow Region 141701, Russia.
| |
Collapse
|
19
|
Mizuno M, Ozeki N, Sekiya I. Safety of using cultured cells with trisomy 7 in cell therapy for treating osteoarthritis. Regen Ther 2022; 21:81-86. [PMID: 35785042 PMCID: PMC9234008 DOI: 10.1016/j.reth.2022.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/25/2022] [Accepted: 06/01/2022] [Indexed: 12/27/2022] Open
Abstract
Cell therapy is a promising alternative treatment approach currently under study for osteoarthritis (OA), the most common chronic musculoskeletal disease. However, the mesenchymal stem cells (MSCs) used in cell therapy to treat OA are usually expanded in vitro to obtain sufficient numbers for transplantation, and their safety has not been fully assessed from multiple perspectives. Analysis of karyotypic abnormalities, in particular, is important to ensure the safety of cells; however, chromosomal mutations may also occur during the cell-expansion process. In addition, there have been many reports showing chromosome abnormalities, mainly trisomy 7, in the cartilage and synovium of patients with OA as well as in normal tissues. The suitability of cells with these karyotypic abnormalities as cells for cell therapy has not been evaluated. Recently, we assessed the safety of using cells with trisomy 7 from the osteoarthritic joint of a patient for transplantation, and we followed up with the patient for 5 years. This study showed analysis for copy number variant and whole-genome sequencing, compared with blood DNA from the same patient. We did not find any abnormalities in the genes regardless of trisomy 7. No side effects were observed for at least 5 years in the human clinical study. This suggests that the transplantation of cultured cells with trisomy 7 isolated from an osteoarthritic joint and transplanted into the osteoarthritic joints of the same person is not expected to cause serious adverse events. However, it is unclear what problems may arise in the case of allogeneic transplantation. Different types of risks will also exist depending on other transplantation routes, such as localization to the knee-joint only or circulation inflow and lung entrapment. In addition, since the cause of trisomy 7 occurrence remains unclear, it is necessary to clarify the mechanism of trisomy 7 in OA to perform cell therapy for OA patients in a safe manner. Trisomy 7 is frequently observed in the cartilage and synovium of patients with OA. MSCs with trisomy 7 did not form tumor after transplantation into mice. No side effects were observed 5 years after transplantation of MSCs with trisomy 7.
Collapse
Affiliation(s)
- Mitsuru Mizuno
- Corresponding author. Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University,1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan. Fax: +81 3 5803-0192.
| | | | | |
Collapse
|
20
|
Suyama T, Takemoto Y, Miyauchi H, Kato Y, Matsuzaki Y, Kato R. Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells. Inflamm Regen 2022; 42:30. [PMID: 36182958 PMCID: PMC9526913 DOI: 10.1186/s41232-022-00214-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/29/2022] [Indexed: 11/12/2022] Open
Abstract
Background Rapidly expanding clones (RECs) are one of the single-cell-derived mesenchymal stem cell clones sorted from human bone marrow mononuclear cells (BMMCs), which possess advantageous features. The RECs exhibit long-lasting proliferation potency that allows more than 10 repeated serial passages in vitro, considerably benefiting the manufacturing process of allogenic MSC-based therapeutic products. Although RECs aid the preparation of large-variation clone libraries for a greedy selection of better-quality clones, such a selection is only possible by establishing multiple-candidate cell banks for quality comparisons. Thus, there is a high demand for a novel method that can predict “low-risk and high-potency clones” early and in a feasible manner given the excessive cost and effort required to maintain such an establishment. Methods LNGFR and Thy-1 co-positive cells from BMMCs were single-cell-sorted into 96-well plates, and only fast-growing clones that reached confluency in 2 weeks were picked up and passaged as RECs. Fifteen RECs were prepared as passage 3 (P3) cryostock as the primary cell bank. From this cryostock, RECs were passaged until their proliferation limitation; their serial-passage limitation numbers were labeled as serial-passage potencies. At the P1 stage, phase-contrast microscopic images were obtained over 6–90 h to identify time-course changes of 24 morphological descriptors describing cell population information. Machine learning models were constructed using the morphological descriptors for predicting serial-passage potencies. The time window and field-of-view-number effects were evaluated to identify the most efficient image data usage condition for realizing high-performance serial-passage potency models. Results Serial-passage test results indicated variations of 7–13-repeated serial-passage potencies within RECs. Such potency values were predicted quantitatively with high performance (RMSE < 1.0) from P1 morphological profiles using a LASSO model. The earliest and minimum effort predictions require 6–30 h with 40 FOVs and 6–90 h with 15 FOVs, respectively. Conclusion We successfully developed a noninvasive morphology-based machine learning model to enhance the efficiency of establishing cell banks with single-cell-derived RECs for quantitatively predicting the future serial-passage potencies of clones. Conventional methods that can make noninvasive and quantitative predictions without wasting precious cells in the early stage are lacking; the proposed method will provide a more efficient and robust cell bank establishment process for allogenic therapeutic product manufacturing. Supplementary Information The online version contains supplementary material available at 10.1186/s41232-022-00214-w.
Collapse
Affiliation(s)
- Takashi Suyama
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.,PuREC Co. Ltd, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yuto Takemoto
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Hiromi Miyauchi
- PuREC Co. Ltd, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yuko Kato
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.,PuREC Co. Ltd, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yumi Matsuzaki
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan. .,PuREC Co. Ltd, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
| | - Ryuji Kato
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan. .,Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Tokai National Higher Education and Research System, Furocho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
21
|
Preclinical Evaluation of the Tumorigenic and Immunomodulatory Properties of Human Bone Marrow Mesenchymal Stromal Cell Populations with Clonal Trisomy 5. Stem Cells Int 2022; 2022:1613636. [PMID: 36035513 PMCID: PMC9417782 DOI: 10.1155/2022/1613636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/04/2022] [Indexed: 01/22/2023] Open
Abstract
Cytogenetic aberrations may emerge in human mesenchymal stromal cells (MSC) during ex vivo expansion for cell therapy. We have detected clonal trisomy 5 in two distinct autologous MSC products expanded from bone marrow which, based on the current quality control criteria, could not be released for clinical use. Although a safety concern, it is still unclear to what extent recurrent aneuploidies detected in MSC products may affect the threshold for neoplastic transformation or the medicinal properties of these cells. We have carried out an exploratory preclinical study to evaluate these MSC products with clonal trisomy 5, regarding their oncogenic and immunomodulatory potential. Cell population growth in vitro was reduced in MSC cultures with clonal trisomy 5 compared with the population growth of their euploid MSC counterparts, based on a lower cumulative population doubling level, reduced cell proliferation index, and increased senescence-associated beta-galactosidase activity. Subcutaneous injection of clinically relevant amount of MSC population, either with or without clonal trisomy 5, did not generate tumors in immunodeficient mice within a follow-up period of six months. Most importantly, MSC population with clonal trisomy 5 kept immunomodulatory properties upon interferon gamma (IFNγ) licensing, displaying overexpression of IDO, CXCL9, CXCL10, and CXCL11, in a similar fashion than that of IFNγ-licensed euploid MSC. Our findings suggest that bone marrow MSC products with clonal trisomy 5 may retain their therapeutic potential, based on poor tumor initiating capability and preserved immunomodulatory potency. This preclinical evidence may further support the definition of release criteria of autologous MSC products for cell therapy under critical clinical scenarios. This trial is registered with Clinical Study registration number: RBR-29x2pr.
Collapse
|
22
|
Ahn SY, Sung DK, Chang YS, Sung SI, Kim YE, Kim HJ, Lee SM, Park WS. BDNF-Overexpressing Engineered Mesenchymal Stem Cells Enhances Their Therapeutic Efficacy against Severe Neonatal Hypoxic Ischemic Brain Injury. Int J Mol Sci 2021; 22:ijms222111395. [PMID: 34768827 PMCID: PMC8583727 DOI: 10.3390/ijms222111395] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
We investigated whether irradiated brain-derived neurotropic factor (BDNF)-overexpressing engineered human mesenchymal stem cells (BDNF-eMSCs) improve paracrine efficiency and, thus, the beneficial potency of naïve MSCs against severe hypoxic ischemic (HI) brain injury in newborn rats. Irradiated BDNF-eMSCs hyper-secreted BDNF > 10 fold and were >5 fold more effective than naïve MSCs in attenuating the oxygen-glucose deprivation-induced increase in cytotoxicity, oxidative stress, and cell death in vitro. Only the irradiated BDNF-eMSCs, but not naïve MSCs, showed significant attenuating effects on severe neonatal HI-induced short-term brain injury scores, long-term progress of brain infarct, increased apoptotic cell death, astrogliosis and inflammatory responses, and impaired negative geotaxis and rotarod tests in vivo. Our data, showing better paracrine potency and the resultant better therapeutic efficacy of the irradiated BDNF-eMSCs, compared to naïve MSCs, suggest that MSCs transfected with the BDNF gene might represent a better, new therapeutic strategy against severe neonatal HI brain injury.
Collapse
Affiliation(s)
- So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.A.); (D.K.S.); (Y.S.C.); (S.I.S.)
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
| | - Dong Kyung Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.A.); (D.K.S.); (Y.S.C.); (S.I.S.)
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.A.); (D.K.S.); (Y.S.C.); (S.I.S.)
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.A.); (D.K.S.); (Y.S.C.); (S.I.S.)
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
| | - Young Eun Kim
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
| | - Hyo-Jin Kim
- SL BiGen, Inc., SL BIGEN Research Hall, 85, Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (H.-J.K.); (S.M.L.)
| | - Soon Min Lee
- SL BiGen, Inc., SL BIGEN Research Hall, 85, Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Korea; (H.-J.K.); (S.M.L.)
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (S.Y.A.); (D.K.S.); (Y.S.C.); (S.I.S.)
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea;
- Correspondence: ; Tel.: +82-2-3410-3523
| |
Collapse
|
23
|
Guan Y, Yang B, Xu W, Li D, Wang S, Ren Z, Zhang J, Zhang T, Liu XZ, Li J, Li C, Meng F, Han F, Wu T, Wang Y, Peng J. Cell-derived extracellular matrix materials for tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1007-1021. [PMID: 34641714 DOI: 10.1089/ten.teb.2021.0147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The involvement of cell-derived extracellular matrix (CDM) in assembling tissue engineering scaffolds has yielded significant results. CDM possesses excellent characteristics, such as ideal cellular microenvironment mimicry and good biocompatibility, which make it a popular research direction in the field of bionanomaterials. CDM has significant advantages as an expansion culture substrate for stem cells, including stabilization of phenotype, reversal of senescence, and guidance of specific differentiation. In addition, the applications of CDM-assembled tissue engineering scaffolds for disease simulation and tissue organ repair are comprehensively summarized; the focus is mainly on bone and cartilage repair, skin defect or wound healing, engineered blood vessels, peripheral nerves, and periodontal tissue repair. We consider CDM a highly promising bionic biomaterial for tissue engineering applications and propose a vision for its comprehensive development.
Collapse
Affiliation(s)
- Yanjun Guan
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Boyao Yang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Wenjing Xu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Dongdong Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Sidong Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, Beijing, China;
| | - Zhiqi Ren
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Jian Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tieyuan Zhang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics, Chinese PLA, General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Xiu-Zhi Liu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Junyang Li
- Nankai University School of Medicine, 481107, Tianjin, Tianjin, China.,Chinese PLA General Hospital, 104607, Beijing, Beijing, China;
| | - Chaochao Li
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Fanqi Meng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Peking University People's Hospital, 71185, Department of spine surgery, Beijing, China;
| | - Feng Han
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Tong Wu
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China;
| | - Yu Wang
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| | - Jiang Peng
- Chinese PLA General Hospital, 104607, Institute of Orthopedics; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, Beijing, China.,Nantong University, 66479, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China;
| |
Collapse
|
24
|
Lee DY, Lee SE, Kwon DH, Nithiyanandam S, Lee MH, Hwang JS, Basith S, Ahn JH, Shin TH, Lee G. Strategies to Improve the Quality and Freshness of Human Bone Marrow-Derived Mesenchymal Stem Cells for Neurological Diseases. Stem Cells Int 2021; 2021:8444599. [PMID: 34539792 PMCID: PMC8445711 DOI: 10.1155/2021/8444599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hBM-MSCs) have been studied for their application to manage various neurological diseases, owing to their anti-inflammatory, immunomodulatory, paracrine, and antiapoptotic ability, as well as their homing capacity to specific regions of brain injury. Among mesenchymal stem cells, such as BM-MSCs, adipose-derived MSCs, and umbilical cord MSCs, BM-MSCs have many merits as cell therapeutic agents based on their widespread availability and relatively easy attainability and in vitro handling. For stem cell-based therapy with BM-MSCs, it is essential to perform ex vivo expansion as low numbers of MSCs are obtained in bone marrow aspirates. Depending on timing, before hBM-MSC transplantation into patients, after detaching them from the culture dish, cell viability, deformability, cell size, and membrane fluidity are decreased, whereas reactive oxygen species generation, lipid peroxidation, and cytosolic vacuoles are increased. Thus, the quality and freshness of hBM-MSCs decrease over time after detachment from the culture dish. Especially, for neurological disease cell therapy, the deformability of BM-MSCs is particularly important in the brain for the development of microvessels. As studies on the traditional characteristics of hBM-MSCs before transplantation into the brain are very limited, omics and machine learning approaches are needed to evaluate cell conditions with indepth and comprehensive analyses. Here, we provide an overview of hBM-MSCs, the application of these cells to various neurological diseases, and improvements in their quality and freshness based on integrated omics after detachment from the culture dish for successful cell therapy.
Collapse
Affiliation(s)
- Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung Eun Lee
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Do Hyeon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | | | - Mi Ha Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Ji Su Hwang
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jung Hwan Ahn
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
25
|
Mizuno M, Endo K, Katano H, Amano N, Nomura M, Hasegawa Y, Ozeki N, Koga H, Takasu N, Ohara O, Morio T, Sekiya I. Transplantation of human autologous synovial mesenchymal stem cells with trisomy 7 into the knee joint and 5 years of follow-up. Stem Cells Transl Med 2021; 10:1530-1543. [PMID: 34342383 PMCID: PMC8550709 DOI: 10.1002/sctm.20-0491] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/20/2021] [Accepted: 06/16/2021] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can show trisomy 7; however, the safety of these cells has not been fully investigated. The purposes of this study were to determine the ratio of patients whose synovial MSCs were transplanted clinically, to intensively investigate MSCs with trisomy 7 from a safety perspective, and to follow up the patients for 5 years after transplantation. Synovial MSCs at passage 0 were transplanted into a knee for degenerative meniscus tears in 10 patients, and the patients were checked at 5 years. The synovial MSCs were evaluated at passages 0 to 15 by G‐bands and digital karyotyping, and trisomy 7 was found in 3 of 10 patients. In those three patients, 5% to 10% of the synovial MSCs showed trisomy 7. The mRNA expressions of representative oncogenes and genes on chromosome 7 did not differ between MSCs with and without trisomy 7. Whole‐genome sequencing and DNA methylation analysis showed similar results for MSCs with and without trisomy 7. Transplantation of human synovial MSCs with trisomy 7 into eight mouse knees did not result in tumor formation under the skin or in the knees after 8 weeks in any mouse, whereas transplanted HT1080 cells formed tumors. In vitro chondrogenic potentials were similar between MSCs with and without trisomy 7. Five‐year follow‐ups revealed no serious adverse events in all 10 human patients, including 3 who had received MSCs with trisomy 7. Overall, our findings indicated that synovial MSCs with trisomy 7 were comparable with MSCs without trisomy 7 from a safety perspective.
Collapse
Affiliation(s)
- Mitsuru Mizuno
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Kentaro Endo
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hisako Katano
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Naoki Amano
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | - Masaki Nomura
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | | | - Nobutake Ozeki
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports MedicineGraduate School, Tokyo Medical and Dental University (TMDU)TokyoJapan
| | - Naoko Takasu
- Department of Fundamental Cell TechnologyCenter for iPS Cell Research and Application, Kyoto UniversityKyotoJapan
| | - Osamu Ohara
- Department of Applied GenomicsKazusa DNA Research InstituteChibaJapan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental BiologyGraduate School, Tokyo Medical and Dental University (TMDU)TokyoJapan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative MedicineTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
26
|
Hassan SA, Elghait ATA, Abdelqader ZS, Meligy FY. Therapeutic efficiency of adipose-derived mesenchymal stem cells in healing of experimentally induced gastric ulcers in rats. Anat Cell Biol 2021; 54:361-374. [PMID: 34290152 PMCID: PMC8493023 DOI: 10.5115/acb.21.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/27/2022] Open
Abstract
Gastric (peptic) ulcer is a major gastrointestinal disorder with high morbidity and mortality. While several drugs have been used to treat gastric ulcers, such as proton pump inhibitor-based triple therapy for Helicobacter pylori eradication, but hey result in adverse side effects. Therefore, development of new alternative therapies is desirable. Many recent studies have shown that mesenchymal stem cells (MSCs) might have an enhancing effect on the ulcerated gastric mucosa. The aim of this study is to evaluate the efficacy of MSCs in the treatment of indomethacin-induced gastric ulcer, and to compare it with the normal ulcer autohealing. This work was performed on 36 adult male albino rats, divided into four groups: Group I (control group), Group II (ulcer group), Group III (autohealing group), and Group IV (stem cells-treated group). The histological changes of gastric mucosa were examined in sections stained with H&E using light microscope for expression of vascular endothelial growth factors (VEGF) and proliferating cell nuclear antigen (PCNA) in immunohistochemical stained sections using image analyzer. The results from MSCs-treated group revealed restoration of the normal architecture of the gastric mucosa with comparison to the autohealing group which showed excessive granulation tissue and heavy cellular infiltration with disorganized architecture of the fundic mucosa. Immunohistochemical examination showed strong expression of both VEGF and PCNA in the MSCs-treated group. So it was concluded that MSCs accelerate gastric ulcer healing when injected intraperitoneally, compared to autohealing process which showed delayed healing.
Collapse
Affiliation(s)
- Safaa A Hassan
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Taha Abou Elghait
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt.,Histology and Cell Biology Department, Sphinx University, Assiut, Egypt
| | - Zainab S Abdelqader
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma Y Meligy
- Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
27
|
Matheni C, Dsouza W. Xeno-Free Human Wharton's Jelly Mesenchymal Stromal Cells Maintain Their Characteristic Properties after Long-Term Cryopreservation. CELL JOURNAL 2021; 23:145-153. [PMID: 34096215 PMCID: PMC8181313 DOI: 10.22074/cellj.2021.7131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/22/2019] [Indexed: 11/30/2022]
Abstract
Objective The past decade has witnessed a rapid growth in harnessing the potential of adult stem cells for regenerative
medicine. An investigational new drug (IND) or a regenerative medicine advanced therapy (RMAT) product must fulfil
many requirements, such as stability studies, after cryopreservation. Such studies are important to ascertain the utility
of off-the-shelf allogeneic cells for clinical applications. The present work describes a complete characterisation of xeno-
free human Wharton’s Jelly mesenchymal stromal cells (hWJ-MSCs) before and up to 28 months post-cryopreservation.
Materials and Methods In this experimental study, culture methods that involved plasma derived human serum and
recombinant trypsin were used to develop clinical grade cells. Complete cell characterisation involved flow cytometry
studies for viability, positive and negative markers, colony forming unit (CFU) potential, population doubling time (PDT),
soft agar assay to evaluate in vitro tumourigenicity, karyotype analysis and differentiation studies which were performed
before and at 6, 12, 18 and 28 months post-cryopreservation.
Results Our data showed consistency in the flow cytometry, CFU assay, PDT, soft agar assay, karyotyping and
differentiation studies.
Conclusion Using our protocols for extended xeno-free culture and cryopreservation of hWJ-MSCs, we could establish
the shelf life of the cell-based product for up to 28 months.
Collapse
|
28
|
Towards Physiologic Culture Approaches to Improve Standard Cultivation of Mesenchymal Stem Cells. Cells 2021; 10:cells10040886. [PMID: 33924517 PMCID: PMC8069108 DOI: 10.3390/cells10040886] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest for their use in cell-based therapies due to their multipotent differentiation and immunomodulatory capacities. In consequence of limited numbers following their isolation from the donor tissue, MSCs require extensive expansion performed in traditional 2D cell culture setups to reach adequate amounts for therapeutic use. However, prolonged culture of MSCs in vitro has been shown to decrease their differentiation potential and alter their immunomodulatory properties. For that reason, preservation of these physiological characteristics of MSCs throughout their in vitro culture is essential for improving the efficiency of therapeutic and in vitro modeling applications. With this objective in mind, many studies already investigated certain parameters for enhancing current standard MSC culture protocols with regard to the effects of specific culture media components or culture conditions. Although there is a lot of diversity in the final therapeutic uses of the cells, the primary stage of standard isolation and expansion is imperative. Therefore, we want to review on approaches for optimizing standard MSC culture protocols during this essential primary step of in vitro expansion. The reviewed studies investigate and suggest improvements focused on culture media components (amino acids, ascorbic acid, glucose level, growth factors, lipids, platelet lysate, trace elements, serum, and xenogeneic components) as well as culture conditions and processes (hypoxia, cell seeding, and dissociation during passaging), in order to preserve the MSC phenotype and functionality during the primary phase of in vitro culture.
Collapse
|
29
|
Li JK, Yang C, Su Y, Luo JC, Luo MH, Huang DL, Tu GW, Luo Z. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Potential Therapeutic Strategy for Acute Kidney Injury. Front Immunol 2021; 12:684496. [PMID: 34149726 PMCID: PMC8209464 DOI: 10.3389/fimmu.2021.684496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/18/2021] [Indexed: 02/05/2023] Open
Abstract
Acute kidney injury (AKI) is a common and potential life-threatening disease in patients admitted to hospital, affecting 10%-15% of all hospitalizations and around 50% of patients in the intensive care unit. Severe, recurrent, and uncontrolled AKI may progress to chronic kidney disease or end-stage renal disease. AKI thus requires more efficient, specific therapies, rather than just supportive therapy. Mesenchymal stem cells (MSCs) are considered to be promising cells for cellular therapy because of their ease of harvesting, low immunogenicity, and ability to expand in vitro. Recent research indicated that the main therapeutic effects of MSCs were mediated by MSC-derived extracellular vesicles (MSC-EVs). Furthermore, compared with MSCs, MSC-EVs have lower immunogenicity, easier storage, no tumorigenesis, and the potential to be artificially modified. We reviewed the therapeutic mechanism of MSCs and MSC-EVs in AKI, and considered recent research on how to improve the efficacy of MSC-EVs in AKI. We also summarized and analyzed the potential and limitations of EVs for the treatment of AKI to provide ideas for future clinical trials and the clinical application of MSC-EVs in AKI.
Collapse
Affiliation(s)
- Jia-Kun Li
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Su
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing-Chao Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming-Hao Luo
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan-Lei Huang
- Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Zhe Luo, ; Guo-Wei Tu,
| | - Guo-Wei Tu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Zhe Luo, ; Guo-Wei Tu,
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Critical Care Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- *Correspondence: Zhe Luo, ; Guo-Wei Tu,
| |
Collapse
|
30
|
IL-17 Triggers Invasive and Migratory Properties in Human MSCs, while IFNy Favors their Immunosuppressive Capabilities: Implications for the "Licensing" Process. Stem Cell Rev Rep 2020; 16:1266-1279. [PMID: 33067729 PMCID: PMC7667142 DOI: 10.1007/s12015-020-10051-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stromal cells (MSCs) were first used as a source for cell therapy in 1995; however, despite their versatility and unambiguous demonstration of efficacy and safety in preclinical/phase I studies, the positive effect of MSCs in human phase III studies did not resemble the success obtained in mouse models of disease. This dissonance highlights the need to more thoroughly study the immunobiology of MSCs to make better use of these cells. Thus, we aimed to study the immunobiology of MSCs by using chip array analysis as a method for general screening to obtain a global picture in our model study and found IFNy and IL-17 signaling as the first two “top canonical pathways” involved in MSCs immunomodulation. The role of IFNy in triggering the immunosuppressive properties of MSCs is well recognized by many groups; however, the role of IL-17 in this process remains uncertain. Interestingly, in contrast to IFNy, which actively improved the MSCs-mediated immunosuppression, IL-17 did not improve directly the MSCs-mediated immunosuppression. Instead, IL-17 signaling induced the migration of MSCs and inflammatory cells, bringing these cell types together and increasing the likelihood of the lymphocytes sensing the immunosuppressive molecules produced by the MSCs. These effects also correlated with high levels of cytokine/chemokine production and metalloprotease activation by MSCs. Importantly, this treatment maintained the MSCs safety profile by not inducing the expression of molecules related to antigen presentation. In this way, our findings highlight the possibility of using IL-17, in combination with IFNy, to prime MSCs for cell therapy to improve their biological properties and thus their therapeutic efficacy. Finally, the use of preactivated MSCs may also minimize variations among MSCs to produce more uniform therapeutic products. In the not-so-distant future, we envisage a portfolio of MSCs activated by different cocktails specifically designed to target and treat specific diseases. Graphical abstract ![]()
Collapse
|
31
|
Kerstan A, Niebergall-Roth E, Esterlechner J, Schröder HM, Gasser M, Waaga-Gasser AM, Goebeler M, Rak K, Schrüfer P, Endres S, Hagenbusch P, Kraft K, Dieter K, Ballikaya S, Stemler N, Sadeghi S, Tappenbeck N, Murphy GF, Orgill DP, Frank NY, Ganss C, Scharffetter-Kochanek K, Frank MH, Kluth MA. Ex vivo-expanded highly pure ABCB5 + mesenchymal stromal cells as Good Manufacturing Practice-compliant autologous advanced therapy medicinal product for clinical use: process validation and first in-human data. Cytotherapy 2020; 23:165-175. [PMID: 33011075 PMCID: PMC8310651 DOI: 10.1016/j.jcyt.2020.08.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/05/2020] [Accepted: 08/29/2020] [Indexed: 12/13/2022]
Abstract
Background aim: Mesenchymal stromal cells (MSCs) hold promise for the treatment of tissue damage and injury. However, MSCs comprise multiple subpopulations with diverse properties, which could explain inconsistent therapeutic outcomes seen among therapeutic attempts. Recently, the adenosine triphosphate-binding cassette transporter ABCB5 has been shown to identify a novel dermal immunomodulatory MSC subpopulation. Methods: The authors have established a validated Good Manufacturing Practice (GMP)-compliant expansion and manufacturing process by which ABCB5+ MSCs can be isolated from skin tissue and processed to generate a highly functional homogeneous cell population manufactured as an advanced therapy medicinal product (ATMP). This product has been approved by the German competent regulatory authority to be tested in a clinical trial to treat therapy-resistant chronic venous ulcers. Results: As of now, 12 wounds in nine patients have been treated with 5 × 105 autologous ABCB5+ MSCs per cm2 wound area, eliciting a median wound size reduction of 63% (range, 32–100%) at 12 weeks and early relief of pain. Conclusions: The authors describe here their GMP- and European Pharmacopoeia-compliant production and quality control process, report on a pre-clinical dose selection study and present the first in-human results. Together, these data substantiate the idea that ABCB5+ MSCs manufactured as ATMPs could deliver a clinically relevant wound closure strategy for patients with chronic therapy-resistant wounds.
Collapse
Affiliation(s)
- Andreas Kerstan
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Martin Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Ana M Waaga-Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany; Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Goebeler
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Katrin Rak
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Philipp Schrüfer
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Sabrina Endres
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Petra Hagenbusch
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | - George F Murphy
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Natasha Y Frank
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA; Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Christoph Ganss
- TICEBA GmbH, Heidelberg, Germany; RHEACELL GmbH & Co. KG, Heidelberg, Germany
| | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA; School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Mark A Kluth
- TICEBA GmbH, Heidelberg, Germany; RHEACELL GmbH & Co. KG, Heidelberg, Germany.
| |
Collapse
|
32
|
Iwanaka T, Yamaza T, Sonoda S, Yoshimaru K, Matsuura T, Yamaza H, Ohga S, Oda Y, Taguchi T. A model study for the manufacture and validation of clinical-grade deciduous dental pulp stem cells for chronic liver fibrosis treatment. Stem Cell Res Ther 2020; 11:134. [PMID: 32213198 PMCID: PMC7093986 DOI: 10.1186/s13287-020-01630-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/30/2020] [Accepted: 03/02/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human deciduous pulp stem cells (hDPSCs) have remarkable stem cell potency associated with cell proliferation, mesenchymal multipotency, and immunosuppressive function and have shown beneficial effects in a variety of animal disease models. Recent studies demonstrated that hDPSCs exhibited in vivo anti-fibrotic and anti-inflammatory action and in vivo hepatogenic-associated liver regeneration, suggesting that hDPSCs may offer a promising source with great clinical demand for treating liver diseases. However, how to manufacture ex vivo large-scale clinical-grade hDPSCs with the appropriate quality, safety, and preclinical efficacy assurances remains unclear. METHODS We isolated hDPSCs from human deciduous dental pulp tissues formed by the colony-forming unit-fibroblast (CFU-F) method and expanded them under a xenogeneic-free and serum-free (XF/SF) condition; hDPSC products were subsequently stored by two-step banking including a master cell bank (MCB) and a working cell bank (WCB). The final products were directly thawed hDPSCs from the WCB. We tested the safety and quality check, stem cell properties, and preclinical potentials of final hDPSC products and hDPSC products in the MCB and WCB. RESULTS We optimized manufacturing procedures to isolate and expand hDPSC products under a XF/SF culture condition and established the MCB and the WCB. The final hDPSC products and hDPSC products in the MCB and WCB were validated the safety and quality including population doubling ability, chromosome stability, microorganism safety, and stem cell properties including morphology, cell surface marker expression, and multipotency. We also evaluated the in vivo immunogenicity and tumorigenicity and validated in vivo therapeutic efficacy for liver regeneration in a CCl4-induced chronic liver fibrosis mouse model in the final hDPSC products and hDPSC products in the WCB. CONCLUSION The manufacture and quality control results indicated that the present procedure could produce sufficient numbers of clinical-grade hDPSC products from a tiny deciduous dental pulp tissue to enhance clinical application of hDPSC products in chronic liver fibrosis.
Collapse
Affiliation(s)
- Tsuyoshi Iwanaka
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takayoshi Yamaza
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Soichiro Sonoda
- Department of Molecular Cell Biology and Oral Anatomy, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichiro Yoshimaru
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Toshiharu Matsuura
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruyoshi Yamaza
- Department of Pediatric Dentistry, Kyushu University Graduate School of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
33
|
Paracrine Mechanisms of Mesenchymal Stromal Cells in Angiogenesis. Stem Cells Int 2020; 2020:4356359. [PMID: 32215017 PMCID: PMC7085399 DOI: 10.1155/2020/4356359] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
The role of the mesenchymal stromal cell- (MSC-) derived secretome is becoming increasingly intriguing from a clinical perspective due to its ability to stimulate endogenous tissue repair processes as well as its effective regulation of the immune system, mimicking the therapeutic effects produced by the MSCs. The secretome is a composite product secreted by MSC in vitro (in conditioned medium) and in vivo (in the extracellular milieu), consisting of a protein soluble fraction (mostly growth factors and cytokines) and a vesicular component, extracellular vesicles (EVs), which transfer proteins, lipids, and genetic material. MSC-derived secretome differs based on the tissue from which the MSCs are isolated and under specific conditions (e.g., preconditioning or priming) suggesting that clinical applications should be tailored by choosing the tissue of origin and a priming regimen to specifically correct a given pathology. MSC-derived secretome mediates beneficial angiogenic effects in a variety of tissue injury-related diseases. This supports the current effort to develop cell-free therapeutic products that bring both clinical benefits (reduced immunogenicity, persistence in vivo, and no genotoxicity associated with long-term cell cultures) and manufacturing advantages (reduced costs, availability of large quantities of off-the-shelf products, and lower regulatory burden). In the present review, we aim to give a comprehensive picture of the numerous components of the secretome produced by MSCs derived from the most common tissue sources for clinical use (e.g., AT, BM, and CB). We focus on the factors involved in the complex regulation of angiogenic processes.
Collapse
|
34
|
Denninger JK, Chen X, Turkoglu AM, Sarchet P, Volk AR, Rieskamp JD, Yan P, Kirby ED. Defining the adult hippocampal neural stem cell secretome: In vivo versus in vitro transcriptomic differences and their correlation to secreted protein levels. Brain Res 2020; 1735:146717. [PMID: 32035887 DOI: 10.1016/j.brainres.2020.146717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Adult hippocampal neural stem and progenitor cells (NSPCs) secrete a variety of proteins that affect tissue function. Though several individual NSPC-derived proteins have been shown to impact key cellular processes, a broad characterization is lacking. Secretome profiling of low abundance stem cell populations is typically achieved via proteomic characterization of in vitro, isolated cells. Here, we identified hundreds of secreted proteins in conditioned media from in vitro adult mouse hippocampal NSPCs using an antibody array and mass spectrometry. Comparison of protein abundance between antibody array and mass spectrometry plus quantification of several key secreted proteins by ELISA revealed notable disconnect between methods in what proteins were identified as being high versus low abundance, suggesting that data from antibody arrays in particular should be approached with caution. We next assessed the NSPC secretome on a transcriptional level with single cell and bulk RNA sequencing (RNAseq) of cultured NSPCs. Comparison of RNAseq transcript levels of highly secreted proteins revealed that quantification of gene expression did not necessarily predict relative protein abundance. Interestingly, comparing our in vitro NSPC gene expression data with similar data from freshly isolated, in vivo hippocampal NSPCs revealed strong correlations in global gene expression between in vitro and in vivo NSPCs. Understanding the components and functions of the NSPC secretome is essential to understanding how these cells may modulate the hippocampal neurogenic niche. Cumulatively, our data emphasize the importance of using proteomics in conjunction with transcriptomics and highlights the need for better methods of unbiased secretome profiling.
Collapse
Affiliation(s)
- Jiyeon K Denninger
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States
| | - Xi Chen
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Altan M Turkoglu
- College of Arts and Sciences, The Ohio State University, United States
| | - Patricia Sarchet
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Abby R Volk
- College of Arts and Sciences, The Ohio State University, United States
| | - Joshua D Rieskamp
- Neuroscience Graduate Program, The Ohio State University, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, The Ohio State University, United States; Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, United States
| | - Elizabeth D Kirby
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States; Department of Neuroscience, The Ohio State University, United States; Chronic Brain Injury Initiative, The Ohio State University, United States.
| |
Collapse
|
35
|
Neri S. Genetic Stability of Mesenchymal Stromal Cells for Regenerative Medicine Applications: A Fundamental Biosafety Aspect. Int J Mol Sci 2019; 20:ijms20102406. [PMID: 31096604 PMCID: PMC6566307 DOI: 10.3390/ijms20102406] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) show widespread application for a variety of clinical conditions; therefore, their use necessitates continuous monitoring of their safety. The risk assessment of mesenchymal stem cell-based therapies cannot be separated from an accurate and deep knowledge of their biological properties and in vitro and in vivo behavior. One of the most relevant safety issues is represented by the genetic stability of MSCs, that can be altered during in vitro manipulation, frequently required before clinical application. MSC genetic stability has the potential to influence the transformation and the therapeutic effect of these cells. At present, karyotype evaluation represents the definitely prevailing assessment of MSC stability, but DNA alterations of smaller size should not be underestimated. This review will focus on current scientific knowledge about the genetic stability of mesenchymal stem cells. The techniques used and possible improvements together with regulatory aspects will also be discussed.
Collapse
Affiliation(s)
- Simona Neri
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
36
|
Kureel SK, Mogha P, Khadpekar A, Kumar V, Joshi R, Das S, Bellare J, Majumder A. Soft substrate maintains proliferative and adipogenic differentiation potential of human mesenchymal stem cells on long-term expansion by delaying senescence. Biol Open 2019; 8:bio039453. [PMID: 31023646 PMCID: PMC6503999 DOI: 10.1242/bio.039453] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs), during in vitro expansion, gradually lose their distinct spindle morphology, self-renewal ability, multi-lineage differentiation potential and enter replicative senescence. This loss of cellular function is a major roadblock for clinical applications which demand cells in large numbers. Here, we demonstrate a novel role of substrate stiffness in the maintenance of hMSCs over long-term expansion. When serially passaged for 45 days from passage 3 to passage 18 on polyacrylamide gel of Young's modulus E=5 kPa, hMSCs maintained their proliferation rate and showed nine times higher population doubling in comparison to their counterparts cultured on plastic Petri-plates. They did not express markers of senescence, maintained their morphology and other mechanical properties such as cell stiffness and cellular traction, and were significantly superior in adipogenic differentiation potential. These results were demonstrated in hMSCs from two different sources, umbilical cord and bone marrow. In summary, our result shows that a soft gel is a suitable substrate to maintain the stemness of mesenchymal stem cells. As preparation of polyacrylamide gel is a well-established, and well-standardized protocol, we propose that this novel system of cell expansion will be useful in therapeutic and research applications of hMSCs.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Pankaj Mogha
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Akshada Khadpekar
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Vardhman Kumar
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Rohit Joshi
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Siddhartha Das
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Jayesh Bellare
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, Indian Institute of Technology Bombay (IITB), Mumbai 400076, India
| |
Collapse
|
37
|
Therapeutic Potential of Autologous Adipose-Derived Stem Cells for the Treatment of Liver Disease. Int J Mol Sci 2018; 19:ijms19124064. [PMID: 30558283 PMCID: PMC6321531 DOI: 10.3390/ijms19124064] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/30/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022] Open
Abstract
Currently, the most effective therapy for liver diseases is liver transplantation, but its use is limited by organ donor shortage, economic reasons, and the requirement for lifelong immunosuppression. Mesenchymal stem cell (MSC) transplantation represents a promising alternative for treating liver pathologies in both human and veterinary medicine. Interestingly, these pathologies appear with a common clinical and pathological profile in the human and canine species; as a consequence, dogs may be a spontaneous model for clinical investigations in humans. The aim of this work was to characterize canine adipose-derived MSCs (cADSCs) and compare them to their human counterpart (hADSCs) in order to support the application of the canine model in cell-based therapy of liver diseases. Both cADSCs and hADSCs were successfully isolated from adipose tissue samples. The two cell populations shared a common fibroblast-like morphology, expression of stemness surface markers, and proliferation rate. When examining multilineage differentiation abilities, cADSCs showed lower adipogenic potential and higher osteogenic differentiation than human cells. Both cell populations retained high viability when kept in PBS at controlled temperature and up to 72 h, indicating the possibility of short-term storage and transportation. In addition, we evaluated the efficacy of autologous ADSCs transplantation in dogs with liver diseases. All animals exhibited significantly improved liver function, as evidenced by lower liver biomarkers levels measured after cells transplantation and evaluation of cytological specimens. These beneficial effects seem to be related to the immunomodulatory properties of stem cells. We therefore believe that such an approach could be a starting point for translating the results to the human clinical practice in future.
Collapse
|
38
|
Stacey G, Andrews P, Asante C, Barbaric I, Barry J, Bisset L, Braybrook J, Buckle R, Chandra A, Coffey P, Crouch S, Driver P, Evans A, Gardner J, Ginty P, Goldring C, Hay DC, Healy L, Hows A, Hutchinson C, Jesson H, Kalber T, Kimber S, Leathers R, Moyle S, Murray T, Neale M, Pan D, Park BK, Rebolledo RE, Rees I, Rivolta MN, Ritchie A, Roos EJ, Saeb-Parsy K, Schröder B, Sebastian S, Thomas A, Thomas RJ, Turner M, Vallier L, Vitillo L, Webster A, Williams D. Science-based assessment of source materials for cell-based medicines: report of a stakeholders workshop. Regen Med 2018; 13:935-944. [PMID: 30488776 DOI: 10.2217/rme-2018-0120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) have the potential to transform medicine. However, hurdles remain to ensure safety for such cellular products. Science-based understanding of the requirements for source materials is required as are appropriate materials. Leaders in hPSC biology, clinical translation, biomanufacturing and regulatory issues were brought together to define requirements for source materials for the production of hPSC-derived therapies and to identify other key issues for the safety of cell therapy products. While the focus of this meeting was on hPSC-derived cell therapies, many of the issues are generic to all cell-based medicines. The intent of this report is to summarize the key issues discussed and record the consensus reached on each of these by the expert delegates.
Collapse
Affiliation(s)
- Glyn Stacey
- International Stem Cell Banking Initiative, 2 High Street, Barley, Hertfordshire, SG8 8HZ, UK
| | - Peter Andrews
- Department of Biomedical Sciences, Centre for Stem Cell Biology, University of Sheffield, Sheffield, South Yourkshire, S10 2TN, UK
| | - Curtis Asante
- Centre for Stem Cells & Regenerative Medicine, King's College London, Guy's Campus, London, SE1 9RT, UK
| | - Ivana Barbaric
- Department of Biomedical Sciences, Centre for Stem Cell Biology, University of Sheffield, Sheffield, South Yourkshire, S10 2TN, UK
| | | | - Louise Bisset
- Medicines & Healthcare Products Regulatory Agency, London, E14 4PU, UK
| | - Julian Braybrook
- LGC Ltd, National Measurement Laboratory, Teddington, TW11 0LY, UK
| | | | | | - Peter Coffey
- University of California, Neuroscience Research Institue, Santa Barbara, CA, 93106, USA
- Institute of Opthalmology, University College London, London, WC1E 6BT, UK
| | - Sharon Crouch
- Financial & Business Services, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | - Amanda Evans
- University of Cambridge/NHS Blood & Transplant, Long Road, CB2 0PT, Cambridge, UK
| | - John Gardner
- School of Social Sciences, Monash University, Victoria, Australia
| | - Patrick Ginty
- Regulatory Affairs, Cell and Gene Therapy Catapult, London, SE1 9RT, UK
| | - Christopher Goldring
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - David C Hay
- University of Edinburgh, MRC Centre for Regenerative Medicine, Edinburgh, EH16 4UU, UK
| | - Lyn Healy
- Francis Crick Institute, London, NW1 1AT, UK
| | - Anna Hows
- Miltenyi Biotec, Bisley, GU24 9DR, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Helen Jesson
- Centre for Biological Engineering, Loughborough University, Loughborough, LE11 3TU, UK
| | - Tammy Kalber
- Centre for Advanced Biomedical Imaging, University College London, WC1E 6BT, UK
| | - Sue Kimber
- Division of Cell Matrix Biology and Regenerative Medicine; Director EPSRC/MRC in Regenerative Medicine, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, M13 9PT, UK
| | | | - Sarah Moyle
- Clinical Biomanufacturing Facility, Oxford University, Oxford, OX3 7JT, UK
| | - Trish Murray
- Department of Physiology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Michael Neale
- Institute of Opthalmology, University College London, London, WC1E 6BT, UK
| | - David Pan
- Medical Research Council, London, WC2B 4AN, UK
| | - B Kevin Park
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | | | - Ian Rees
- Medicines & Healthcare Products Regulatory Agency, London, E14 4PU, UK
| | - Marcelo N Rivolta
- Department of Biomedical Sciences, Centre for Stem Cell Biology, University of Sheffield, Sheffield, South Yourkshire, S10 2TN, UK
| | - Allan Ritchie
- Allan Ritchie Medical Device Consulting, Harrogate, HG1 1BX, UK
| | | | - Kourosh Saeb-Parsy
- Department of Surgery, University fo Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Bernd Schröder
- Miltenyi Biotec GmbH, 51429 Bergisch Gladbach, Nordrhein-Westfalen, Germany
| | - Sujith Sebastian
- School of Bioscience, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Angela Thomas
- College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Robert J Thomas
- Centre for Biological Engineering, Loughborough University, Loughborough, LE11 3TU, UK
| | - Marc Turner
- Scottish National Blood Transfusion Service, Edinburgh, EH14 4BE, UK
| | - Ludovic Vallier
- Wellcome-Medical Research Council Cambridge Stem Cell Institute and Department of Surgery, University of Cambridge, Cambridge UK
| | - Loriana Vitillo
- Institute of Opthalmology, University College London, London, WC1E 6BT, UK
| | - Andrew Webster
- SATSU, Department of Sociology, University of York, York, YO10 5DD, UK
| | - David Williams
- Centre for Biological Engineering, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
39
|
Bahsoun S, Coopman K, Forsyth NR, Akam EC. The Role of Dissolved Oxygen Levels on Human Mesenchymal Stem Cell Culture Success, Regulatory Compliance, and Therapeutic Potential. Stem Cells Dev 2018; 27:1303-1321. [DOI: 10.1089/scd.2017.0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
40
|
Peltzer J, Aletti M, Frescaline N, Busson E, Lataillade JJ, Martinaud C. Mesenchymal Stromal Cells Based Therapy in Systemic Sclerosis: Rational and Challenges. Front Immunol 2018; 9:2013. [PMID: 30271402 PMCID: PMC6146027 DOI: 10.3389/fimmu.2018.02013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Systemic Sclerosis (SSc) is a rare chronic disease, related to autoimmune connective tissue diseases such as Systemic Lupus Erythematosus and Sjögren's Syndrome. Although its clinical heterogeneity, main features of the disease are: extensive tissue fibrosis with increase matrix deposition in skin and internal organ, microvascular alterations and activation of the immune system with autoantibodies against various cellular antigens. In the diffuse cutaneous scleroderma subtype, the disease is rapidly progressive with a poor prognosis, leading to failure of almost any internal organ, especially lung which is the leading cause of death. Primary trigger is unknown but may involve an immune process against mesenchymal cells in a genetically receptive host. Pathophysiology reveals a pivotal role of fibrosis and inflammation alterations implicating different cell subtypes, cytokines and growth factors, autoantibodies and reactive oxygen species. Despite improvement, the overall survival of SSc patients is still lower than that of other inflammatory diseases. Recommended drugs are agents capable of modulating fibrotic and inflammatory pathways. Cellular therapy has recently emerged as a credible option. Besides autologous hematopoietic stem cell transplantation which demonstrated remarkable improvement, mesenchymal stromal cells (MSCs) represent promising therapeutic candidates. Indeed, these cells possess anti-inflammatory, antiproliferative, antifibrotic, and immunomodulary properties especially by secreting a large panel of bioactive molecules, addressing the most important key points of the SSc. In addition, these cells are very sensitive to their environment and are able to modulate their activity according to the pathophysiological context in which they are located. Autologous or allogeneic MSCs from various sources have been tested in many trials in different auto-immune diseases such as multiple sclerosis, Crohn's disease or systemic lupus erythematosus. They are characterized by a broad availability and no or low acute toxicity. However, few randomized prospective clinical trials were published and their production under ATMP regulatory procedures is complex and time-consuming. Many aspects have still to be addressed to ascertain their potential as well as the potential of their derived products in the management of SSc, probably in association with other therapies.
Collapse
Affiliation(s)
- Juliette Peltzer
- Unité de Thérapie tissulaire et traumatologie de guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Marc Aletti
- Service de Médecine Interne, Hôpital d'Instruction des Armées Percy, Clamart, France
| | - Nadira Frescaline
- UMR7648 Laboratoire de physique des plasmas, École Polytechnique, Palaiseau, France
| | - Elodie Busson
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Jean-Jacques Lataillade
- Unité de Thérapie tissulaire et traumatologie de guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Christophe Martinaud
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| |
Collapse
|
41
|
Zeng Y, Wang X, Wang J, Yi R, Long H, Zhou M, Luo Q, Zhai Z, Song Y, Qi S. The Tumorgenicity of Glioblastoma Cell Line U87MG Decreased During Serial In Vitro Passage. Cell Mol Neurobiol 2018; 38:1245-1252. [PMID: 29948550 DOI: 10.1007/s10571-018-0592-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/17/2018] [Indexed: 02/08/2023]
Abstract
Established cancer cell lines are routinely used to study cancer. Several factors such as serial passage may affect the reproducibility of experiments with cancer cell lines, but few researches focused on these changes. In the present study, different morphology and decreased tumorigenicity were observed in late passage U87MG cells. In vitro experiments further revealed that late passage U87MG cells possessed lower invasion properties than early passage, whereas no significant differences of proliferation and migration were found between early and late passage U87MG cells. In particular, we confirmed that late passage U87MG cells exhibited more epithelial phenotype with decreased PI3K/Akt pathway and TGF-β pathway expressions at protein level. In summary, our results focused on the changes of U87MG cells during serial in vitro passage, suggested that passage-induced changes may lead to notable changes of biological characteristics and several molecular transitions in cancer cell lines, indicating the necessity to shorten experiment-span and accomplish experiments with the same or similar passage cancer cell strains.
Collapse
Affiliation(s)
- Yu Zeng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xizhao Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Jizhou Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, People's Republic of China
| | - Renhui Yi
- Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Department of Neurosurgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Mingfeng Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, 533000, Guangxi, People's Republic of China
| | - Zhihao Zhai
- Department of Neurosurgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Futian, Shenzhen, 518000, People's Republic of China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
42
|
Wang L, Zhu CY, Ma DX, Gu ZY, Xu CC, Wang FY, Chen JG, Liu CJ, Guan LX, Gao R, Gao Z, Fang S, Zhuo DJ, Liu SF, Gao CJ. Efficacy and safety of mesenchymal stromal cells for the prophylaxis of chronic graft-versus-host disease after allogeneic hematopoietic stem cell transplantation: a meta-analysis of randomized controlled trials. Ann Hematol 2018; 97:1941-1950. [PMID: 29947972 DOI: 10.1007/s00277-018-3384-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
Abstract
A meta-analysis of randomized controlled trials (RCTs) was conducted to evaluate the efficacy and safety of mesenchymal stromal cells (MSCs) for the prophylaxis of chronic graft-versus-host disease (cGVHD) in patients with hematological malignancies undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). Six studies involving 365 patients were included. The pooled results showed that MSCs significantly reduced the incidence of cGVHD (risk ratio [RR] 0.63, 95% confidence interval [CI] 0.46 to 0.86, P = 0.004). Favorable prophylactic effects of MSCs on cGVHD were observed with umbilical cord-derived, high-dose, and late-infusion MSCs, while bone marrow-derived, low-dose, and coinfused MSCs did not confer beneficial prophylactic effects. In addition, MSC infusion did not increase the risk of primary disease relapse and infection (RR 1.02, 95% CI 0.70 to 1.50, P = 0.913; RR 0.89, 95% CI 0.44 to 1.81, P = 0.752; respectively). Moreover, there was an apparent trend toward increased overall survival (OS) in the MSC group compared with that in the control group (RR 1.13, 95% CI 0.98 to 1.29, P = 0.084). In conclusion, this meta-analysis demonstrated that MSC infusion is an effective and safe prophylactic strategy for cGVHD in patients with hematological malignancies undergoing allo-HSCT.
Collapse
Affiliation(s)
- Li Wang
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Cheng-Ying Zhu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - De-Xun Ma
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Zhen-Yang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Chang-Chun Xu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Fei-Yan Wang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Ji-Gang Chen
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Cheng-Jun Liu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Li-Xun Guan
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Rui Gao
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Zhe Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Shu Fang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Du-Jun Zhuo
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China
| | - Shu-Feng Liu
- Department of Hematology, Laoshan Branch of No. 401 Hospital of Chinese People's Liberation Army (PLA), 109 Laoshan Road, Qingdao, 266101, China.
| | - Chun-Ji Gao
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
43
|
Villatoro AJ, Claros S, Fernández V, Alcoholado C, Fariñas F, Moreno A, Becerra J, Andrades JA. Safety and efficacy of the mesenchymal stem cell in feline eosinophilic keratitis treatment. BMC Vet Res 2018; 14:116. [PMID: 29587744 PMCID: PMC5870249 DOI: 10.1186/s12917-018-1413-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/06/2018] [Indexed: 01/07/2023] Open
Abstract
Background Feline eosinophilic keratitis (FEK) is a chronic keratopathy caused by a suspected immune mediated response to an unknown antigenic stimulus. The purpose of this study was to investigate the safety and therapeutic effects of allogeneic feline adipose-derived mesenchymal stromal cells (fAd-MSCs) implanted subconjunctival around the ocular surface lesion in five cats with FEK refractory to current available treatments. Results FEK was diagnosed by clinical appearance and evidence of eosinophil and/or mast cells in corneal cytology. Each animal was treated with two applications of 2 × 106 million of fAd-MSCs 2 months apart. Ocular surface integrity was assessed before treatment and at 1, 3, 6 and 11 months after treatment. Clinical signs showed a significant change during the follow-up with resolution of the corneal and conjunctiva lesions and there were no signs of regression or worsening. Conclusions Implanted cells were well-tolerated and effective reducing clinical signs of FEK with a sustained effect during the study period. None of the animals showed systemic or local complications during the study. To our knowledge, this is the first time in literature that local implantation of allogeneic fAd-MSCs has been found as an effective therapeutic alternative to treat cats with FEK.
Collapse
Affiliation(s)
- Antonio J Villatoro
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Silvia Claros
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Viviana Fernández
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - Fernando Fariñas
- ImmuneStem, Instituto de Inmunología Clínica y Terapia Celular, 29018, Málaga, Spain.,Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain
| | - Antonio Moreno
- Hospital veterinario Alhaurín el Grande. Alhaurín el Grande, 29120, Málaga, Spain
| | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.,Laboratory of Bioengineering and Tissue Regeneration, Andalusian Center for Nanomedicine and Biotechnology-BIONAND, 29590, Málaga, Spain
| | - José A Andrades
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedicine Research Institute of Malaga (IBIMA), Campus Universitario de Teatinos, 29071, Málaga, Spain. .,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain.
| |
Collapse
|
44
|
Dabrowska S, Del Fattore A, Karnas E, Frontczak-Baniewicz M, Kozlowska H, Muraca M, Janowski M, Lukomska B. Imaging of extracellular vesicles derived from human bone marrow mesenchymal stem cells using fluorescent and magnetic labels. Int J Nanomedicine 2018; 13:1653-1664. [PMID: 29593411 PMCID: PMC5865569 DOI: 10.2147/ijn.s159404] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stem cells have been shown therapeutic in various neurological disorders. Recent studies support the notion that the predominant mechanism by which MSCs act is through the release of extracellular vesicles (EVs). EVs seem to have similar therapeutic activity as their cellular counterparts and may represent an interesting alternative standalone therapy for various diseases. The aim of the study was to optimize the method of EV imaging to better understand therapeutic effects mediated by EVs. Methods The fluorescent lipophilic stain PKH26 and superparamagnetic iron oxide nanoparticles conjugated with rhodamine (Molday ION Rhodamine B™) were used for the labeling of vesicles in human bone marrow MSCs (hBM-MSCs). The entire cycle from intracellular vesicles to EVs followed by their uptake by hBM-MSCs has been studied. The identity of vesicles has been proven by antibodies against: anti-CD9, -CD63, and -CD81 (tetraspanins). NanoSight particle tracking analysis (NTA), high-resolution flow cytometric analysis, transmission electron microscopy (TEM), ELYRA PS.1 super-resolution microscopy, and magnetic resonance imaging (MRI) were used for the characterization of vesicles. Results The PKH26 and Molday ION were exclusively localized in intracellular vesicles positively stained for EV markers: CD9, CD63, and CD81. The isolated EVs represent heterogeneous population of various sizes as confirmed by NTA. The TEM and MRI were capable to show successful labeling of EVs using ION. Co-culture of EVs with hBM-MSCs revealed their uptake by cells in vitro, as visualized by the co-localization of PKH26 or Molday ION with tetraspanins inside hBM-MSCs. Conclusion PKH26 and Molday ION seem to be biocompatible with EVs, and the labeling did not interfere with the capability of EVs to re-enter hBM-MSCs during co-culture in vitro. Magnetic properties of IONs provide an additional advantage for the imaging of EV using TEM and MRI.
Collapse
Affiliation(s)
- Sylwia Dabrowska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Andrea Del Fattore
- Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Elzbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Krakow, Poland
| | | | - Hanna Kozlowska
- Laboratory of Advanced Microscopy Techniques, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Russel H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
45
|
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Research Institut, Badalona, Spain
- Center of Regenerative Medicine in Barcelona, Barcelona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institut, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Research Institut, Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Cardiology Service, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| |
Collapse
|
46
|
Clonal chromosomal and genomic instability during human multipotent mesenchymal stromal cells long-term culture. PLoS One 2018; 13:e0192445. [PMID: 29432491 PMCID: PMC5809118 DOI: 10.1371/journal.pone.0192445] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
Background aims Spontaneous mutagenesis often leads to appearance of genetic changes in cells. Although human multipotent mesenchymal stromal cells (hMSC) are considered as genetically stable, there is a risk of genomic and structural chromosome instability and, therefore, side effects of cell therapy associated with long-term effects. In this study, the karyotype, genetic variability and clone formation analyses have been carried out in the long-term culture MSC from human gingival mucosa. Methods The immunophenotype of MSC has been examined using flow cytofluorometry and short tandem repeat (STR) analysis has been carried out for authentication. The karyotype has been examined using GTG staining and mFISH, while the assessment of the aneuploidy 8 frequency has been performed using centromere specific chromosome FISH probes in interphase cells. Results The immunophenotype and STR loci combination did not change during the process of cultivation. From passage 23 the proliferative activity of cultured MSCs was significantly reduced. From passage 12 of cultivation, clones of cells with stable chromosome aberrations have been identified and the biggest of these (12%) are tetrasomy of chromosome 8. The random genetic and structural chromosomal aberrations and the spontaneous level of chromosomal aberrations in the hMSC long-term cultures were also described. Conclusions The spectrum of spontaneous chromosomal aberrations in MSC long-term cultivation has been described. Clonal chromosomal aberrations have been identified. A clone of cells with tetrasomy 8 has been detected in passage 12 and has reached the maximum size by passage 18 before and decreased along with the reduction of proliferative activity of cell line by passage 26. At later passages, the MSC line exhibited a set of cells with structural variants of the karyotype with a preponderance of normal diploid cells. The results of our study strongly suggest a need for rigorous genetic analyses of the clone formation in cultured MSCs before use in medicine.
Collapse
|
47
|
Lee JY, McMurtry SA, Stevens T. Single cell cloning generates lung endothelial colonies with conserved growth, angiogenic, and bioenergetic characteristics. Pulm Circ 2017; 7:777-792. [PMID: 28841087 PMCID: PMC5703126 DOI: 10.1177/2045893217731295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 08/03/2017] [Indexed: 01/13/2023] Open
Abstract
Pulmonary artery, capillary, and vein endothelial cells possess distinctive structures and functions, which represent a form of vascular segment specific macroheterogeneity. However, within each of these segmental populations, individual cell functional variability represents a poorly characterized microheterogeneity. Here, we hypothesized that single cell clonogenic assays would reveal microheterogeneity among the parent cell population and enable isolation of highly representative cells with committed parental characteristics. To test this hypothesis, pulmonary microvascular endothelial cells (PMVECs) and pulmonary arterial endothelial cells (PAECs) were isolated from different Sprague Dawley rats. Serum stimulated proliferation of endothelial populations and single cell clonogenic potential were evaluated. In vitro Matrigel assays were utilized to analyze angiogenic potential and the Seahorse assay was used to evaluate bioenergetic profiles. PMVEC populations grew faster and had a higher proliferative potential than PAEC populations. Fewer PMVECs were needed to form networks on Matrigel when compared with PAECs. PMVECs primarily utilized aerobic glycolysis, while PAECs relied more heavily on oxidative phosphorylation, to support bioenergetic demands. Repeated single cell cloning and expansion of PAEC colonies generated homogeneous first-generation clones that were highly reflective of the parental population in terms of growth, angiogenic potential, and bioenergetic profiles. Repeated single cell cloning of the first-generation clones generated second-generation clones with increased proliferative potential while maintaining other parental characteristics. Second-generation clones were highly homogeneous populations. Thus, single cell cloning reveals microheterogeneity among the parent cell population and enables isolation of highly representative cells with parental characteristics.
Collapse
Affiliation(s)
- Ji Young Lee
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
- Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care Medicine, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Sarah A. McMurtry
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
- Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
48
|
Cryopreserved or Fresh Mesenchymal Stromal Cells: Only a Matter of Taste or Key to Unleash the Full Clinical Potential of MSC Therapy? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:77-98. [PMID: 27837556 DOI: 10.1007/978-3-319-45457-3_7] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) harbor great therapeutic potential for numerous diseases. From early clinical trials, success and failure analysis, bench-to-bedside and back-to-bench approaches, there has been a great gain in knowledge, still leaving a number of questions to be answered regarding optimal manufacturing and quality of MSCs for clinical application. For treatment of many acute indications, cryobanking may remain a prerequisite, but great uncertainty exists considering the therapeutic value of freshly thawed (thawed) and continuously cultured (fresh) MSCs. The field has seen an explosion of new literature lately, outlining the relevance of the topic. MSCs appear to have compromised immunomodulatory activity directly after thawing for clinical application. This may provide a possible explanation for failure of early clinical trials. It is not clear if and how quickly MSCs recover their full therapeutic activity, and if the "cryo stun effect" is relevant for clinical success. Here, we will share our latest insights into the relevance of these observations for clinical practice that will be discussed in the context of the published literature. We argue that the differences of fresh and thawed MSCs are limited but significant. A key issue in evaluating potency differences is the time point of analysis after thawing. To date, prospective double-blinded randomized clinical studies to evaluate potency of both products are lacking, although recent progress was made with preclinical assessment. We suggest refocusing therapeutic MSC development on potency and safety assays with close resemblance of the clinical reality.
Collapse
|
49
|
|
50
|
Shakouri-Motlagh A, O'Connor AJ, Brennecke SP, Kalionis B, Heath DE. Native and solubilized decellularized extracellular matrix: A critical assessment of their potential for improving the expansion of mesenchymal stem cells. Acta Biomater 2017; 55:1-12. [PMID: 28412553 DOI: 10.1016/j.actbio.2017.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/03/2017] [Accepted: 04/11/2017] [Indexed: 02/08/2023]
Abstract
Capturing the promise of mesenchymal stem cell (MSC)-based treatments is currently limited by inefficient production of cells needed for clinical therapies. During conventional ex vivo expansion, a large portion of MSCs lose the properties that make them attractive for use in cell therapies. Decellularized extracellular matrix (dECM) has recently emerged as a promising substrate for the improved expansion of MSCs. MSCs cultured on these surfaces exhibit improved proliferation capacity, maintenance of phenotype, and increased differentiation potential. Additionally, these dECMs can be solubilized and used to coat new cell culture surfaces, imparting key biological properties of the native matrices to other surfaces such as tissue engineering scaffolds. Although this technology is still developing, there is potential for an impact in the fields of MSC biology, biomaterials, tissue engineering, and therapeutics. In this article, we review the role of dECM in MSC expansion by first detailing the decellularization methods that have been used to produce the dECM substrates; discussing the shortcomings of current decellularization methods; describing the improved MSC characteristics obtained when the cells are cultured on these surfaces; and considering the effect of the passage number, age of donor, and dECM preparation method on the quality of the dECM. Finally we describe the critical roadblocks that must be addressed before this technology can fulfil its potential, including elucidating the mechanism by which the dECMs improve the expansion of primary MSCs and the identification of a readily available source of dECM. STATEMENT OF SIGNIFICANCE Current mesenchymal stem cell (MSC) culture methods result in premature cellular senescence or loss of differentiation potential. This creates a major bottleneck in their clinical application, as prolonged expansion is necessary to achieve clinically relevant numbers of cells. Recently, decellularized extracellular matrix (dECM) produced by primary MSC has emerged as an attractive substrate for the improved expansion of MSC; cells cultured on these surfaces retain their desired stem cell characteristics for prolonged times during culture. This review article describes the inception and development of this dECM-based technology, points out existing challenges that must be addressed, and suggests future directions of research. To our knowledge, this is the first review written on the use of dECM for improved mesenchymal stem cell expansion.
Collapse
|