1
|
Li H, Wei J, Li M, Li Y, Zhang T, Tian J, Liu X, Li K, Lin J. Biological characteristics of Muse cells derived from MenSCs and their application in acute liver injury and intracerebral hemorrhage diseases. Regen Ther 2024; 27:48-62. [PMID: 38496012 PMCID: PMC10940801 DOI: 10.1016/j.reth.2024.03.003] [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: 11/28/2023] [Revised: 02/16/2024] [Accepted: 03/03/2024] [Indexed: 03/19/2024] Open
Abstract
The increasing interest in multilineage differentiating stress-enduring (Muse) cells within the field of regenerative medicine is attributed to their exceptional homing capabilities, prolonged viability in adverse conditions, and enhanced three-germ-layer differentiate ability, surpassing their parent mesenchymal stem cells. Given their abundant sources, non-invasive collection procedure, and periodic availability, human menstrual blood-derived endometrium stem cells (MenSCs) have been extensively investigated as a potential resource for stem cell-based therapies. However, there is no established modality to isolate Muse cells from MenSCs and disparity in gene expression profiles between Muse cells and MenSCs remain unknown. In this study, Muse cells were isolated from MenSCs by long-time trypsin incubation method. Muse cells expressed pluripotency markers and could realize multilineage differentiation in vitro. Compared with MenSCs, Muse cells showed enhanced homing ability and superior therapeutic efficacy in animal models of acute liver injury (ALI) and intracerebral hemorrhage (ICH). Furthermore, the RNA-seq analysis offers insights into the mechanism underlying the disparity in trypsin resistance and migration ability between Muse and MenSCs cells. This research offers a significant foundation for further exploration of cell-based therapies using MenSCs-derived Muse cells in the context of various human diseases, highlighting their promising application in the field of regenerative medicine.
Collapse
Affiliation(s)
- Han Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jinghui Wei
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Mingzhi Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yaoqiang Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Tong Zhang
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jialu Tian
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Xuejia Liu
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Kangjia Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
2
|
Que H, Mai E, Hu Y, Li H, Zheng W, Jiang Y, Han F, Li X, Gong P, Gu J. Multilineage-differentiating stress-enduring cells: a powerful tool for tissue damage repair. Front Cell Dev Biol 2024; 12:1380785. [PMID: 38872932 PMCID: PMC11169632 DOI: 10.3389/fcell.2024.1380785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are a type of pluripotent cell with unique characteristics such as non-tumorigenic and pluripotent differentiation ability. After homing, Muse cells spontaneously differentiate into tissue component cells and supplement damaged/lost cells to participate in tissue repair. Importantly, Muse cells can survive in injured tissue for an extended period, stabilizing and promoting tissue repair. In addition, it has been confirmed that injection of exogenous Muse cells exerts anti-inflammatory, anti-apoptosis, anti-fibrosis, immunomodulatory, and paracrine protective effects in vivo. The discovery of Muse cells is an important breakthrough in the field of regenerative medicine. The article provides a comprehensive review of the characteristics, sources, and potential mechanisms of Muse cells for tissue repair and regeneration. This review serves as a foundation for the further utilization of Muse cells as a key clinical tool in regenerative medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
3
|
Andreone L, dos Santos A, Wailemann R, Terra L, Gomes V, Macedo da Silva J, Rosa-Fernandes L, Sogayar M, Palmisano G, Labriola L, Perone M. Cotransplantation of marginal mass allogeneic islets with 3D culture-derived adult human skin cells improves glycemia in diabetic mice. Braz J Med Biol Res 2023; 56:e12611. [PMID: 37792778 PMCID: PMC10515501 DOI: 10.1590/1414-431x2023e12611] [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: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 10/06/2023] Open
Abstract
Islet transplantation represents a therapeutic option for type 1 diabetes (T1D). Long-term viability of transplanted islets requires improvement. Mesenchymal stromal cells (MSCs) have been proposed as adjuvants for islet transplantation facilitating grafting and functionality. Stem cell aggregation provides physiological interactions between cells and enhances the in situ concentration of modulators of inflammation and immunity. We established a hanging-drop culture of adult human skin fibroblast-like cells as spheroids, and skin spheroid-derived cells (SphCs) were characterized. We assessed the potential of SphCs in improving islet functionality by cotransplantation with a marginal mass of allogeneic islets in an experimental diabetic mouse model and characterized the secretome of SphCs by mass spectrometry-based proteomics. SphCs were characterized as multipotent progenitors and their coculture with anti-CD3 stimulated mouse splenocytes decreased CD4+ T cell proliferation with skewed cytokine secretion through an increase in the Th2/Th1 ratio profile. SphCs-conditioned media attenuated apoptosis of islets induced by cytokine challenge in vitro and importantly, intratesticular SphCs administration did not show tumorigenicity in immune-deficient mice. Moreover, SphCs improved glycemic control when cotransplanted with a marginal mass of allogeneic islets in a diabetic mouse model without pharmacological immunosuppression. SphCs' protein secretome differed from its paired skin fibroblast-like counterpart in containing 70% of up- and downregulated proteins and biological processes that overall positively influenced islets such as cytoprotection, cellular stress, metabolism, and survival. In summary, SphCs improved the performance of transplanted allogeneic islets in an experimental T1D model, without pharmacological immunosuppression. Future research is warranted to identify SphCs-secreted factors responsible for islets' endurance.
Collapse
Affiliation(s)
- L. Andreone
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - A.F. dos Santos
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - R.A.M. Wailemann
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L.F. Terra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V.M. Gomes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - J. Macedo da Silva
- Departamento de Parasitologia, Instituto de Biosciências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L. Rosa-Fernandes
- Departamento de Parasitologia, Instituto de Biosciências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M.C. Sogayar
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
- Centro de Terapia Celular e Molecular, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - G. Palmisano
- Departamento de Parasitologia, Instituto de Biosciências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L. Labriola
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M.J. Perone
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| |
Collapse
|
4
|
Dushime H, Moreno SG, Linard C, Adrait A, Couté Y, Peltzer J, Messiaen S, Torres C, Bensemmane L, Lewandowski D, Romeo PH, Petit V, Gault N. Fetal Muse-based therapy prevents lethal radio-induced gastrointestinal syndrome by intestinal regeneration. Stem Cell Res Ther 2023; 14:201. [PMID: 37568164 PMCID: PMC10416451 DOI: 10.1186/s13287-023-03425-1] [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: 10/21/2022] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Human multilineage-differentiating stress enduring (Muse) cells are nontumorigenic endogenous pluripotent-like stem cells that can be easily obtained from various adult or fetal tissues. Regenerative effects of Muse cells have been shown in some disease models. Muse cells specifically home in damaged tissues where they exert pleiotropic effects. Exposition of the small intestine to high doses of irradiation (IR) delivered after radiotherapy or nuclear accident results in a lethal gastrointestinal syndrome (GIS) characterized by acute loss of intestinal stem cells, impaired epithelial regeneration and subsequent loss of the mucosal barrier resulting in sepsis and death. To date, there is no effective medical treatment for GIS. Here, we investigate whether Muse cells can prevent lethal GIS and study how they act on intestinal stem cell microenvironment to promote intestinal regeneration. METHODS Human Muse cells from Wharton's jelly matrix of umbilical cord (WJ-Muse) were sorted by flow cytometry using the SSEA-3 marker, characterized and compared to bone-marrow derived Muse cells (BM-Muse). Under gas anesthesia, GIS mice were treated or not through an intravenous retro-orbital injection of 50,000 WJ-Muse, freshly isolated or cryopreserved, shortly after an 18 Gy-abdominal IR. No immunosuppressant was delivered to the mice. Mice were euthanized either 24 h post-IR to assess early small intestine tissue response, or 7 days post-IR to assess any regenerative response. Mouse survival, histological stainings, apoptosis and cell proliferation were studied and measurement of cytokines, recruitment of immune cells and barrier functional assay were performed. RESULTS Injection of WJ-Muse shortly after abdominal IR highly improved mouse survival as a result of a rapid regeneration of intestinal epithelium with the rescue of the impaired epithelial barrier. In small intestine of Muse-treated mice, an early enhanced secretion of IL-6 and MCP-1 cytokines was observed associated with (1) recruitment of monocytes/M2-like macrophages and (2) proliferation of Paneth cells through activation of the IL-6/Stat3 pathway. CONCLUSION Our findings indicate that a single injection of a small quantity of WJ-Muse may be a new and easy therapeutic strategy for treating lethal GIS.
Collapse
Affiliation(s)
- Honorine Dushime
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Stéphanie G Moreno
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Christine Linard
- Laboratory of Medical Radiobiology, Institute of Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Annie Adrait
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, FR2048, CEA, 38000, Grenoble, France
| | - Yohann Couté
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, FR2048, CEA, 38000, Grenoble, France
| | - Juliette Peltzer
- Institut de Recherche Biomédicale des Armées (IRBA), 92141, Clamart, France
- UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Sébastien Messiaen
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Claire Torres
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Lydia Bensemmane
- Laboratory of Medical Radiobiology, Institute of Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Daniel Lewandowski
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Paul-Henri Romeo
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Vanessa Petit
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France.
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France.
| | - Nathalie Gault
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France.
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France.
| |
Collapse
|
5
|
Velasco MG, Satué K, Chicharro D, Martins E, Torres-Torrillas M, Peláez P, Miguel-Pastor L, Del Romero A, Damiá E, Cuervo B, Carrillo JM, Cugat R, Sopena JJ, Rubio M. Multilineage-Differentiating Stress-Enduring Cells (Muse Cells): The Future of Human and Veterinary Regenerative Medicine. Biomedicines 2023; 11:biomedicines11020636. [PMID: 36831171 PMCID: PMC9953712 DOI: 10.3390/biomedicines11020636] [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: 01/31/2023] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
In recent years, several studies have been conducted on Muse cells mainly due to their pluripotency, high tolerance to stress, self-renewal capacity, ability to repair DNA damage and not being tumoral. Additionally, since these stem cells can be isolated from different tissues in the adult organism, obtaining them is not considered an ethical problem, providing an advantage over embryonic stem cells. Regarding their therapeutic potential, few studies have reported clinical applications in the treatment of different diseases, such as aortic aneurysm and chondral injuries in the mouse or acute myocardial infarction in the swine, rabbit, sheep and in humans. This review aims to describe the characterization of Muse cells, show their biological characteristics, explain the differences between Muse cells and mesenchymal stem cells, and present their contribution to the treatment of some diseases.
Collapse
Affiliation(s)
- María Gemma Velasco
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Katy Satué
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Emma Martins
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Pau Peláez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Laura Miguel-Pastor
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Ayla Del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Elena Damiá
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - Belén Cuervo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
| | - José María Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Ramón Cugat
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| | - Joaquín Jesús Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
- Correspondence:
| | - Mónica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, C/Tirant lo Blanc, 7, Alfara del Patriarca, 46115 Valencia, Spain
- Garcia Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, 08006 Barcelona, Spain
| |
Collapse
|
6
|
Castillo MG, Peralta TM, Locatelli P, Velazquez C, Herrero Y, Crottogini AJ, Olea FD, Cuniberti LA. Promoting early neovascularization by allotransplanted adipose-derived Muse cells in an ovine model of acute myocardial infarction. PLoS One 2023; 18:e0277442. [PMID: 36662847 PMCID: PMC9858827 DOI: 10.1371/journal.pone.0277442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/26/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent preclinical studies have demonstrated that bone marrow (BM)-derived Muse cells have a homing mechanism to reach damaged cardiac tissue while also being able to reduce myocardial infarct size and improve cardiac function; however, the potential of BM-Muse cells to foster new blood-vessel formation has not been fully assessed. Up to date, adipose tissue (AT)-derived Muse cells remain to be studied in acute myocardial infarction (AMI). The aim of the present study was to analyze in vitro and in vivo the neovascularization capacity of AT-Muse cells while exploring their biodistribution and differentiation potential in a translational ovine model of AMI. METHODS AND RESULTS AT-Muse cells were successfully isolated from ovine adipose tissue. In adult sheep, one or more diagonal branches of the left anterior descending coronary artery were permanently ligated for thirty minutes. Sheep were randomized in two groups and treated with intramyocardial injections: Vehicle (PBS, n = 4) and AT-Muse (2x107 AT-Muse cells labeled with PKH26 Red Fluorescent Dye, n = 4). Molecular characterization showed higher expression of angiogenic genes (VEGF, PGF and ANG) and increased number of tube formation in AT-Muse cells group compared to Adipose-derived mesenchymal stromal cells (ASCs) group. At 7 days post-IAM, the AT-Muse group showed significantly more arterioles and capillaries than the Vehicle group. Co-localization of PKH26+ cells with desmin, sarcomeric actin and troponin T implied the differentiation of Muse cells to a cardiac fate; moreover, PKH26+ cells also co-localized with a lectin marker, suggesting a possible differentiation to a vascular lineage. CONCLUSION Intramyocardially administered AT-Muse cells displayed a significant neovascularization activity and survival capacity in an ovine model of AMI.
Collapse
Affiliation(s)
- Martha G. Castillo
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Tomás M. Peralta
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Candela Velazquez
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Yamila Herrero
- Instituto de Biología y Medicina Experimental—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto J. Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda D. Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| | - Luis A. Cuniberti
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB)—Universidad Favaloro—CONICET, Ciudad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Yamashita T, Nakano Y, Sasaki R, Tadokoro K, Omote Y, Yunoki T, Kawahara Y, Matsumoto N, Taira Y, Matsuoka C, Morihara R, Abe K. Safety and Clinical Effects of a Muse Cell-Based Product in Patients With Amyotrophic Lateral Sclerosis: Results of a Phase 2 Clinical Trial. Cell Transplant 2023; 32:9636897231214370. [PMID: 38014622 PMCID: PMC10686030 DOI: 10.1177/09636897231214370] [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: 08/03/2023] [Revised: 10/23/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by progressive loss of motor neurons. Multilineage-differentiating stress-enduring (Muse) cells are unique endogenous stem cells that show therapeutic effects on motor function in ALS mouse models. We conducted a single-center open phase II clinical trial to evaluate the safety and clinical effects of repeated intravenous injections of an allogenic Muse cell-based product, CL2020, in patients with ALS. Five patients with ALS received CL2020 intravenously once a month for a total of six doses. The primary endpoints were safety and tolerability, and the secondary endpoint was the rate of change in the Revised Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS-R) score. In addition, serum tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), sphingosine-1-phosphate (S1P), cerebrospinal fluid chitotriosidase-1 (CHIT-1), and neurofilament light chain (NfL) levels were evaluated. The CL2020 treatment was highly tolerated without serious side effects. The ALSFRS-R score change trended upward at 12 months post-CL2020 treatment compared with that at 3 months pre-administration, but the difference was not statistically significant. Among five patients diagnosed with ALS, three exhibited a decrease in the rate of ALSFRS-R score change, one demonstrated an increase, and another showed no change. In addition, the patients' serum IL-6 and TNF-α levels and cerebrospinal fluid CHIT-1 and NfL levels increased for up to 6 months post-treatment; however, their serum S1P levels continuously decreased over 12 months. These findings indicate a favorable safety profile of CL2020 therapy. In the near future, a double-blind study of a larger number of ALS patients should be conducted to confirm the efficacy of ALS treatment with CL2020.
Collapse
Affiliation(s)
- Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshio Omote
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuko Kawahara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Namiko Matsumoto
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuki Taira
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chika Matsuoka
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
8
|
Chen X, Yin XY, Wang CC, Du P, Wang XS, Lu YC, Sun YW, Sun YH, Hu YM. Muse cells decrease the neuroinflammatory response by modulating the proportion of M1 and M2 microglia in vitro. Neural Regen Res 2023. [PMID: 35799545 PMCID: PMC9241390 DOI: 10.4103/1673-5374.343885] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation hinders repair of the central nervous system (CNS). Stem cell transplantation is a very promising approach for treatment of CNS injuries. However, it is difficult to select seed cells that can both facilitate nerve regeneration and improve the microenvironment in the CNS. In this study, we isolated multilineage-differentiating stress-enduring (Muse) cells from bone marrow mesenchymal stem cells. We explored the anti-inflammatory effect and mechanism of Muse cells in vitro by coculture of Muse cells with lipopolysaccharide-stimulated microglia. Our results showed that Muse cells effectively reduced the transcription and secretion of tumor necrosis factor α and interleukin-1β and increased the expression of transforming growth factor-β and interleukin-10 in microglia. In addition, Muse cells decreased the number of M1 microglia and increased the proportion of M2 microglia in an inflammatory environment more effectively than bone marrow mesenchymal stem cells. We also show that Muse cells inhibited the protein expression of toll-like receptor 4 (TLR4) and myeloid differentiation primary response protein (MyD88) and inhibited the expression of the phosphorylated forms of transcription factor p65, nuclear factor (NF)-κB inhibitor alpha, and p38 mitogen-activated protein kinase (MAPK) in microglia. Therefore, we suggest Muse cells cause antineuroinflammatory effects by inhibition of the TLR4/MyD88/NF-κB and p38 MAPK signaling pathways in microglia. Our results shed light on the function of Muse cells in relation to CNS diseases and provide insight into the selection of seed cells.
Collapse
|
9
|
Hori Y, Kitani T, Yanishi K, Suga T, Kogure M, Kusaba T, Kushida Y, Dezawa M, Matoba S. Intravenous administration of human Muse cells recovers blood flow in a mouse model of hindlimb ischemia. Front Cardiovasc Med 2022; 9:981088. [PMID: 36440014 PMCID: PMC9692087 DOI: 10.3389/fcvm.2022.981088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cell-based therapies hold great promise for the treatment of peripheral arterial disease (PAD), especially in patients presenting with severe limb ischemia, although the optimal strategy remains to be explored. In this study, we evaluated the therapeutic effect of intravenous administration of human Muse cells, a unique subpopulation of mesenchymal stem cells (MSC), using a mouse model of hindlimb ischemia (HLI) without an immunosuppressant. Compared with the phosphate buffered saline (PBS) or non-Muse MSC groups, the Muse group showed significantly higher laser doppler blood flow in the ischemic limb at days 7 and 14 after HLI. Increased microvascular density [percent area of CD31(+) cells] and reduced interstitial fibrosis in the ischemic limb muscle were also observed in the Muse group. mCherry-expressing Muse cells were found in the ischemic border zone and expressed CD31 but did not in the non-ischemic limb. Muse cells produced higher amounts of vascular endothelial growth factor (VEGF) than non-Muse cells under normoxic and hypoxic conditions in vitro. In the ischemic muscle, tissue VEGF concentration and angiogenesis-related genes such as Vegfa, Angpt1, Pdgfb, and Igf1 were significantly higher in the Muse group than in the other two groups. In addition, the proportion of M2 macrophages to total macrophages and the ratio of anti-inflammatory-related genes such as IL-10, Arg1, and CD206 per iNOS were significantly higher in the Muse group than in the other two groups. In summary, Muse cells exert pleiotropic effects in a mouse model of HLI, and therefore may provide a novel therapeutic approach for the treatment of PAD patients with severe limb ischemia.
Collapse
|
10
|
Kuroda Y, Oguma Y, Hall K, Dezawa M. Endogenous reparative pluripotent Muse cells with a unique immune privilege system: Hint at a new strategy for controlling acute and chronic inflammation. Front Pharmacol 2022; 13:1027961. [PMID: 36339573 PMCID: PMC9627303 DOI: 10.3389/fphar.2022.1027961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/21/2022] Open
Abstract
Multilineage-differentiating stress enduring (Muse) cells, non-tumorigenic endogenous pluripotent stem cells, reside in the bone marrow (BM), peripheral blood, and connective tissue as pluripotent surface marker SSEA-3(+) cells. They express other pluripotent markers, including Nanog, Oct3/4, and Sox2 at moderate levels, differentiate into triploblastic lineages, self-renew at a single cell level, and exhibit anti-inflammatory effects. Cultured mesenchymal stromal cells (MSCs) and fibroblasts contain several percent of SSEA-3(+)-Muse cells. Circulating Muse cells, either endogenous or administered exogenously, selectively accumulate at the damaged site by sensing sphingosine-1-phosphate (S1P), a key mediator of inflammation, produced by damaged cells and replace apoptotic and damaged cells by spontaneously differentiating into multiple cells types that comprise the tissue and repair the tissue. Thus, intravenous injection is the main route for Muse cell treatment, and surgical operation is not necessary. Furthermore, gene introduction or cytokine induction are not required for generating pluripotent or differentiated states prior to treatment. Notably, allogenic and xenogenic Muse cells escape host immune rejection after intravenous injection and survive in the tissue as functioning cells over 6 and ∼2 months, respectively, without immunosuppressant treatment. Since Muse cells survive in the host tissue for extended periods of time, therefore their anti-inflammatory, anti-fibrotic, and trophic effects are long-lasting. These unique characteristics have led to the administration of Muse cells via intravenous drip in clinical trials for stroke, acute myocardial infarction, epidermolysis bullosa, spinal cord injury, neonatal hypoxic ischemic encephalopathy, amyotrophic lateral sclerosis, and COVID-19 acute respiratory distress syndrome without HLA-matching or immunosuppressive treatment.
Collapse
Affiliation(s)
| | | | | | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
11
|
Di Stefano AB, Urrata V, Trapani M, Moschella F, Cordova A, Toia F. Systematic review on spheroids from adipose‐derived stem cells: Spontaneous or artefact state? J Cell Physiol 2022; 237:4397-4411. [PMID: 36209478 PMCID: PMC10091738 DOI: 10.1002/jcp.30892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022]
Abstract
Three-dimensional (3D) cell cultures represent the spontaneous state of stem cells with specific gene and protein molecular expression that are more alike the in vivo condition. In vitro two-dimensional (2D) cell adhesion cultures are still commonly employed for various cellular studies such as movement, proliferation and differentiation phenomena; this procedure is standardized and amply used in laboratories, however their representing the original tissue has recently been subject to questioning. Cell cultures in 2D require a support/substrate (flasks, multiwells, etc.) and use of fetal bovine serum as an adjuvant that stimulates adhesion that most likely leads to cellular aging. A 3D environment stimulates cells to grow in suspended aggregates that are defined as "spheroids." In particular, adipose stem cells (ASCs) are traditionally observed in adhesion conditions, but a recent and vast literature offers many strategies that obtain 3D cell spheroids. These cells seem to possess a greater ability in maintaining their stemness and differentiate towards all mesenchymal lineages, as demonstrated in in vitro and in vivo studies compared to adhesion cultures. To date, standardized procedures that form ASC spheroids have not yet been established. This systematic review carries out an in-depth analysis of the 76 articles produced over the past 10 years and discusses the similarities and differences in materials, techniques, and purposes to standardize the methods aimed at obtaining ASC spheroids as already described for 2D cultures.
Collapse
Affiliation(s)
- Anna Barbara Di Stefano
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
| | - Valentina Urrata
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
| | - Marco Trapani
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
| | - Francesco Moschella
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
| | - Adriana Cordova
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
- Department of Surgical, Oncological and Oral Sciences, Unit of Plastic and Reconstructive Surgery University of Palermo Palermo Italy
- Department of D.A.I. Chirurgico, Plastic and Reconstructive Unit Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone” Palermo Italy
| | - Francesca Toia
- BIOPLAST‐Laboratory of BIOlogy and Regenerative Medicine‐PLASTic Surgery, Plastic and Reconstructive Surgery Unit, Department of Surgical, Oncological and Oral Sciences University of Palermo Palermo Italy
- Department of Surgical, Oncological and Oral Sciences, Unit of Plastic and Reconstructive Surgery University of Palermo Palermo Italy
- Department of D.A.I. Chirurgico, Plastic and Reconstructive Unit Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone” Palermo Italy
| |
Collapse
|
12
|
Wakao S, Oguma Y, Kushida Y, Kuroda Y, Tatsumi K, Dezawa M. Phagocytosing differentiated cell-fragments is a novel mechanism for controlling somatic stem cell differentiation within a short time frame. Cell Mol Life Sci 2022; 79:542. [PMID: 36203068 PMCID: PMC9537123 DOI: 10.1007/s00018-022-04555-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/29/2022]
Abstract
Stem cells undergo cytokine-driven differentiation, but this process often takes longer than several weeks to complete. A novel mechanism for somatic stem cell differentiation via phagocytosing ‘model cells’ (apoptotic differentiated cells) was found to require only a short time frame. Pluripotent-like Muse cells, multipotent mesenchymal stem cells (MSCs), and neural stem cells (NSCs) phagocytosed apoptotic differentiated cells via different phagocytic receptor subsets than macrophages. The phagocytosed-differentiated cell-derived contents (e.g., transcription factors) were quickly released into the cytoplasm, translocated into the nucleus, and bound to promoter regions of the stem cell genomes. Within 24 ~ 36 h, the cells expressed lineage-specific markers corresponding to the phagocytosed-differentiated cells, both in vitro and in vivo. At 1 week, the gene expression profiles were similar to those of the authentic differentiated cells and expressed functional markers. Differentiation was limited to the inherent potential of each cell line: triploblastic-, adipogenic-/chondrogenic-, and neural-lineages in Muse cells, MSCs, and NSCs, respectively. Disruption of phagocytosis, either by phagocytic receptor inhibition via small interfering RNA or annexin V treatment, impeded differentiation in vitro and in vivo. Together, our findings uncovered a simple mechanism by which differentiation-directing factors are directly transferred to somatic stem cells by phagocytosing apoptotic differentiated cells to trigger their rapid differentiation into the target cell lineage.
Collapse
Affiliation(s)
- Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan.
| | - Yo Oguma
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Yasumasa Kuroda
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan
| | - Kazuki Tatsumi
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan.,Regenerative Medicine Division, Analytical Research Department, Technology Development Unit, Life Science Institute, Inc., Tokyo, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, 980-8575, Japan.
| |
Collapse
|
13
|
Kikuchi K, Katagiri H, Suzuki Y, Nitta H, Sasaki A. Mobilization of multilineage-differentiating stress-enduring cells into the peripheral blood in liver surgery. PLoS One 2022; 17:e0271698. [PMID: 35862404 PMCID: PMC9302816 DOI: 10.1371/journal.pone.0271698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/05/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose This study investigated whether liver damage severity relates to the mobilization of multilineage-differentiating stress-enduring (Muse) cells, which are endogenous reparative pluripotent stem cells, into the peripheral blood (PB) and whether the degree of mobilization relates to the recovery of liver volume following human liver surgery. Methods Forty-seven patients who underwent liver surgery were included in the present study. PB-Muse cells were counted before surgery, on postoperative days (PODs) 3 and on POD 7. Liver volume was measured using computed tomography before and after surgery. Results The PB-Muse cell count increased after surgery. The number of PB-Muse cells before surgery was higher, but without statistical significance in the group with neoplasms than in the healthy group that included liver donors (p = 0.065). Forty-seven patients who underwent liver surgery were divided into major hepatic resection (MHR; hepatectomy of three or more segments according to the Couinaud classification, n = 22) and minor hepatic resection (mhr; hepatectomy of two segments or less according to the Couinaud classification, n = 25) groups. PB-Muse cells increased at high rates among MHR patients (p = 0.033). Except for complication cases, PB-Muse cells increased at higher rates in the group with advanced liver volume recovery (p = 0.043). The predictive impact of the rate of increase in PB-Muse cells on the recovery of liver volume was demonstrated by multivariate analysis (OR 11.0, p = 0.014). Conclusions PB-Muse cell mobilization correlated with the volume of liver resection, suggesting that the PB-Muse cell number reflects the degree of liver injury. Given that the degree of PB-Muse cell mobilization was related to liver volume recovery, PB-Muse cells were suggested to contribute to liver regeneration, although this mechanism remains unclear.
Collapse
Affiliation(s)
- Koji Kikuchi
- Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
- * E-mail:
| | - Hirokatsu Katagiri
- Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| | - Yuji Suzuki
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| | - Hiroyuki Nitta
- Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| | - Akira Sasaki
- Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| |
Collapse
|
14
|
Nguyen LT, Tran NT, Than UTT, Nguyen MQ, Tran AM, Do PTX, Chu TT, Nguyen TD, Bui AV, Ngo TA, Hoang VT, Hoang NTM. Optimization of human umbilical cord blood-derived mesenchymal stem cell isolation and culture methods in serum- and xeno-free conditions. Stem Cell Res Ther 2022; 13:15. [PMID: 35012671 PMCID: PMC8751356 DOI: 10.1186/s13287-021-02694-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/24/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Although umbilical cord blood (UCB) is identified as a source of mesenchymal stem cells (MSCs) with various advantages, the success in cell isolation is volatile. Therefore, it is necessary to optimize methods of cord blood-derived MSC (UCB-MSC) isolation and culture. In this study, we evaluated the efficiency of UCB-MSC isolation and expansion using different commercially available serum- and xeno-free media and investigated the capacity of autologous serum and plasma as a supplement to support cell proliferation. Additionally, we defined the presence of multilineage-differentiating stress-enduring (Muse) cells in the UCB-MSC population. Functions of UCB-MSC in in vitro angiogenesis processes and anti-cancer were also verified. METHODS Mononuclear cells were isolated using density gradient separation and cultured in four commercial media kits, as well as four surface coating solutions. UCB-MSCs were characterized and tested on tube formation assay, and co-cultured with SK-MEL cells in a transwell system. RESULTS The results showed that only StemMACS™ MSC Expansion Media is more appropriate to isolate and culture UCB-MSCs. The cells exhibited a high cell proliferation rate, CFU forming capability, MSC surface marker expression, trilineage differentiate potential, and chromosome stability. In addition, the culture conditions with autologous serum coating and autologous plasma supplement enhanced cell growth and colony forming. This cell population contained Muse cells at rate of 0.3%. Moreover, UCB-MSCs could induce the tube formation of human umbilical vein endothelial cells and inhibit more than 50% of SK-MEL cell growth. CONCLUSIONS UCB-MSCs could be high-yield isolated and expanded under serum- and xeno-free conditions by using the StemMACS™ MSC Expansion Media kit. Autologous serum coating and plasma supplement enhanced cell proliferation. These UCB-MSCs had effected the tube formation process and an anti-cancer impact.
Collapse
Affiliation(s)
- Liem Thanh Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam.,College of Health Sciences, VinUniversity, Hanoi, Vietnam
| | - Nghia Trung Tran
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam.,VNU University of Science, Vietnam National University, Hanoi, Vietnam.,Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon, Korea
| | - Uyen Thi Trang Than
- Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Minh Quang Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam.,VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Anh Minh Tran
- VNU University of Science, Vietnam National University, Hanoi, Vietnam.,Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Phuong Thi Xuan Do
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam.,VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Thao Thi Chu
- Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Tu Dac Nguyen
- Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Anh Viet Bui
- Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Tien Anh Ngo
- Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam
| | - Van Thanh Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam
| | - Nhung Thi My Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Hanoi, Vietnam. .,VNU University of Science, Vietnam National University, Hanoi, Vietnam. .,Center of Applied sciences, Regenerative medicine, and Advance technologies (CARA), Hanoi, Vietnam.
| |
Collapse
|
15
|
Wang P, Wang S, Ji F, Zhang R. Muse Cells Have Higher Stress Tolerance than Adipose Stem Cells due to the Overexpression of the CCNA2 Gene. Stem Cells Dev 2021; 30:1056-1069. [PMID: 34486391 DOI: 10.1089/scd.2021.0088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
This study aimed to investigate the stress tolerance mechanism of multilineage-differentiating stress enduring (Muse) cells and elucidate the means to improve the stress tolerance of mesenchymal stem cells. Cell viability, apoptosis, and senescence-related protein expression were detected under H2O2 stress by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium reduction assay, flow cytometry in combination with Annexin V-FITC/PI staining, and western blotting analysis, respectively. A significant increase in the CCNA2 gene level within Muse cells relative to adipose stem cells (ASCs) was observed. In the H2O2 stress environment in vitro, the survival rate of Muse cells remarkably increased compared with the survival rate of the ASCs. In addition, a reduced level of apoptosis and senescence-related protein expression of Muse cells relative to ASCs was documented. The miR-29b-3p-induced negative regulation of CCNA2 gene expression was confirmed by in vitro luciferase assay. A significant upregulation of CCNA2 gene expression in ASCs, transfected with antagomir-29b-3p, improved the survival rate of ASCs under H2O2 stress but dramatically reduced the apoptosis and expression of the senescence-related gene; agomir-29b-3p could partially reverse these effects. In conclusion, high expression of the CCNA2 gene is associated with an increased stress tolerance of Muse cells. Regulating the expression of CCNA2 by miR-29b-3p can alter the stress tolerance of ASCs.
Collapse
Affiliation(s)
- Peng Wang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Intensive Care Unit, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shengyi Wang
- The Dermal and Venereal Department, Xuzhou Central Hospital, Xuzhou, China.,The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| | - Fuhai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruzhi Zhang
- The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| |
Collapse
|
16
|
Sato T, Wakao S, Kushida Y, Tatsumi K, Kitada M, Abe T, Niizuma K, Tominaga T, Kushimoto S, Dezawa M. A Novel Type of Stem Cells Double-Positive for SSEA-3 and CD45 in Human Peripheral Blood. Cell Transplant 2021; 29:963689720923574. [PMID: 32525407 PMCID: PMC7586270 DOI: 10.1177/0963689720923574] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Peripheral blood (PB) contains several types of stem/progenitor cells, including hematopoietic stem and endothelial progenitor cells. We identified a population positive for both the pluripotent surface marker SSEA-3 and leukocyte common antigen CD45 that comprises 0.04% ± 0.003% of the mononuclear cells in human PB. The average size of the SSEA-3(+)/CD45(+) cells was 10.1 ± 0.3 µm and ∼22% were positive for CD105, a mesenchymal marker; ∼85% were positive for CD19, a B cell marker; and ∼94% were positive for HLA-DR, a major histocompatibility complex class II molecule relevant to antigen presentation. These SSEA-3(+)/CD45(+) cells expressed the pluripotency markers Nanog, Oct3/4, and Sox2, as well as sphingosine-1-phosphate (S1P) receptor 2, and migrated toward S1P, although their adherence and proliferative activities in vitro were low. They expressed NeuN at 7 d, Pax7 and desmin at 7 d, and alpha-fetoprotein and cytokeratin-19 at 3 d when supplied to mouse damaged tissues of the brain, skeletal muscle and liver, respectively, suggesting the ability to spontaneously differentiate into triploblastic lineages compatible to the tissue microenvironment. Multilineage-differentiating stress enduring (Muse) cells, identified as SSEA-3(+) in tissues such as the bone marrow and organ connective tissues, express pluripotency markers, migrate to sites of damage via the S1P-S1P receptor 2 system, and differentiate spontaneously into tissue-compatible cells after homing to the damaged tissue where they participate in tissue repair. After the onset of acute myocardial infarction and stroke, patients are reported to have an increase in the number of SSEA-3(+) cells in the PB. The SSEA-3(+)/CD45(+) cells in the PB showed similarity to tissue-Muse cells, although with difference in surface marker expression and cellular properties. Thus, these findings suggest that human PB contains a subset of cells that are distinct from known stem/progenitor cells, and that CD45(+)-mononuclear cells in the PB comprise a novel subpopulation of cells that express pluripotency markers.
Collapse
Affiliation(s)
- Tetsuya Sato
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuki Tatsumi
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Regenerative Medicine Division, Life Science Institute, Inc., Tokyo, Japan
| | - Masaaki Kitada
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takatsugu Abe
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigeki Kushimoto
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
17
|
Guo Y, Xue Y, Wang P, Cui Z, Cao J, Liu S, Yu Q, Zeng Q, Zhu D, Xie M, Zhang J, Li Z, Liu H, Zhong J, Chen J. Muse cell spheroids have therapeutic effect on corneal scarring wound in mice and tree shrews. Sci Transl Med 2021; 12:12/562/eaaw1120. [PMID: 32967971 DOI: 10.1126/scitranslmed.aaw1120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/02/2019] [Accepted: 06/23/2020] [Indexed: 12/16/2022]
Abstract
Stem cell therapy holds promises for treating corneal scarring. Here, we use multilineage-differentiating stress-enduring (Muse) cells to study their differentiation and therapeutic potential for treating corneal injury. Muse cells were isolated from lipoaspirate, which presented biphenotype properties of both pluripotent stem cells and some mesenchymal stem cells. Muse cells expanded by about 100-fold from the initial seeding cell number to Muse spheroids with the maintenance of the Muse cell phenotype and high cell viability at 33 days by static spheroid culture. We revealed that Muse spheroids were activated by the dynamic rotary cell culture system (RCCS), as characterized by increased stemness, improved activity, and enhanced adherence. Gene and protein expression of the pluripotent markers OCT3/4, SOX2, and NANOG and of the proliferation marker KI67 in Muse spheroids cultured under RCCS were higher than those in the static group. These activated Muse spheroids enabled ready differentiation into corneal stromal cells (CSCs) expressing characteristic marker genes and proteins. Furthermore, implantation of Muse cells-differentiated CSCs (Muse-CSCs) laden assembled with two orthogonally stacked stretched compressed collagen (cell-SCC) in mouse and tree shrew wounded corneas prevented the formation of corneal scarring, increased corneal re-epithelialization and nerve regrowth, and reduced the severity of corneal inflammation and neovascularization. cell-SCC retained the capacity to suppress corneal scarring after long-distance cryopreserved transport. Thus, Muse cell therapy is a promising avenue for developing therapeutics for treating corneal scarring.
Collapse
Affiliation(s)
- Yonglong Guo
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China.,Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Yunxia Xue
- Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Peiyuan Wang
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zekai Cui
- Aier Eye Institute, 18th floor, the New century building, #198 Furong Middle Road, Changsha, Hunan 410015, China
| | - Jixing Cao
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Shiwei Liu
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Quan Yu
- Centric Laboratory, Medical College, Jinan University, Guangzhou, China
| | - Qiaolang Zeng
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Deliang Zhu
- Key Laboratory of Optoelectronic Information and Sensing Technologies, Guangdong Higher Educational Institutes, Jinan University, Guangzhou 510632, China
| | - Mengyuan Xie
- Key Laboratory of Optoelectronic Information and Sensing Technologies, Guangdong Higher Educational Institutes, Jinan University, Guangzhou 510632, China
| | - Jun Zhang
- Key Laboratory of Optoelectronic Information and Sensing Technologies, Guangdong Higher Educational Institutes, Jinan University, Guangzhou 510632, China
| | - Zhijie Li
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China.,Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China
| | - Hongwei Liu
- Department of Plastic Surgery, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingxiang Zhong
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Jiansu Chen
- Ophthalmology Department, First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China.,Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China.,Aier Eye Institute, 18th floor, the New century building, #198 Furong Middle Road, Changsha, Hunan 410015, China
| |
Collapse
|
18
|
Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol 2021; 477:117-132. [PMID: 34048734 DOI: 10.1016/j.ydbio.2021.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
During embryonic stages, vascular endothelial cells (ECs) originate from the mesoderm, at specific extraembryonic and embryonic regions, through a process called vasculogenesis. In the adult, EC renewal/replacement mostly depend on local resident ECs or endothelial progenitor cells (EPCs). Nevertheless, contribution from circulating ECs/EPCs was also reported. In addition, cells lacking from EC/EPC markers with in vitro extended plasticity were shown to originate endothelial-like cells (ELCs). Most of these cells consist of mesenchymal stromal progenitors, which would eventually get mobilized from the bone marrow after injury. Based on that, current knowledge on different mouse and human bone marrow stromal cell (BM-SC) subpopulations, able to contribute with mesenchymal stromal/stem cells (MSCs), is herein reviewed. Such analyses underline an unexpected heterogeneity among sinusoidal LepR+ stromal/CAR cells. For instance, in a recent report a subgroup of LepR+ stromal/CAR progenitors, which express GLAST and is traced in Wnt1Cre;R26RTom mice, was found to contribute with ELCs in vivo. These GLAST + Wnt1+ BM-SCs were shown to get mobilized to the peripheral blood and to contribute with liver regeneration. Other sources of ELCs, such as adipose, neural and dental pulp tissues, were also published. Finally, mechanisms likely involved in the enhanced cellular plasticity properties of bone marrow/adipose tissue stromal cells, able to originate ELCs, are assessed. In the future, strategies to analyze the in vivo expression profile of stromal cells, with MSC properties, in combination with screening of active genomic regions at the single cell-level, during early postnatal development and/or after injury, will likely help understanding properties of these ELC sources.
Collapse
Affiliation(s)
- Jorge B Aquino
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina.
| | - Romina Sierra
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| | - Laura A Montaldo
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| |
Collapse
|
19
|
Lwin SM, Snowden JA, Griffiths CEM. The promise and challenges of cell therapy for psoriasis. Br J Dermatol 2021; 185:887-898. [PMID: 34036569 DOI: 10.1111/bjd.20517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The management of moderate-to-severe psoriasis has been transformed by the introduction of biological therapies. These medicines, particularly those targeting interleukin (IL)-17 and IL-23p19, can offer clear or nearly clear skin for the majority of patients with psoriasis, with good long-term drug survival. However, as currently used, none of these therapies is curative and disconcertingly there is a small but increasing number of patients with severe psoriasis who have failed all currently available therapeutic modalities. A similar scenario has occurred in other immune-mediated inflammatory diseases (IMIDs) where treatment options are limited in severely affected patients. In these cases, cell therapy, including haematopoietic stem cell transplantation (HSCT) and mesenchymal stromal cells (MSC), has been utilized. This review discusses the various forms of cell therapy currently available, their utility in the management of IMIDs and emerging evidence for efficacy in severe psoriasis that is unresponsive to biological therapy. Balancing the risks and benefits of treatment vs. the underlying disease is key; cell therapy carries significant risks, costs, regulation and other complexities, which must be justified by outcomes. Although HSCT has anecdotally been reported to benefit severe psoriasis, sometimes with apparent cure, this has mainly been in the setting of other coincidental 'routine' indications. In psoriasis, cell therapies, such as MSC and regulatory T cells, with a lower risk of complications are likely to be more appropriate. Well-designed controlled trials coupled with mechanistic studies are warranted if advanced cell therapies are to be developed and delivered as a realistic option for severe psoriasis.
Collapse
Affiliation(s)
- S M Lwin
- St John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - J A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.,Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - C E M Griffiths
- Dermatology Centre, Salford Royal Hospital, NIHR Manchester Biomedical Research Centre Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
20
|
An open-label phase 1 clinical trial of the allogeneic side population adipose-derived mesenchymal stem cells in SMA type 1 patients. Neurol Sci 2021; 43:399-410. [PMID: 34032944 DOI: 10.1007/s10072-021-05291-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Spinal muscular atrophy (SMA), an autosomal recessive neurodegenerative disorder of alpha motor neurons of spinal cord associated with progressive muscle weakness and hypotonia, is the most common genetic cause of infant mortality. Although there is few promising treatment for SMA, but the field of translational research is active in it, and stem cell-based therapy clinical trials or case studies are ongoing. Combination of different therapeutic approaches for noncurative treatments may increase their effectiveness and compliance of patients. We present a phase 1 clinical trial in patients with SMA1 who received side population adipose-derived mesenchymal stem cells (SPADMSCs). METHODS The intervention group received three intrathecal administrations of escalating doses of SPADMSCs and followed until 24 months or the survival time. The safety analysis was assessed by controlling the side effects and efficacy evaluations performed by the Hammersmith Infant Neurological Examination (HINE), Ballard score, and electrodiagnostic (EDX) evaluation. These evaluations were performed before intervention and at the end of the follow-up. RESULTS The treatment was safe and well tolerated, without any adverse event related to the stem cell administration. One of the patients in the intervention group was alive after 24 months of study follow-up. He is a non-sitter 62-month-old boy with appropriate weight gain and need for noninvasive ventilation (NIV) for about 8 h per day. Clinical scores, need for supportive ventilation, and number of hospitalizations were not meaningful parameters in the response of patients in the intervention and control groups. All five patients in the intervention group showed significant improvement in the motor amplitude response of the tibial nerve (0.56mV; p: 0.029). CONCLUSION This study showed that SPADMSCs therapy is tolerable and safe with promising efficacy in SMA I. Probably same as other treatment strategies, early intervention will increase its efficacy and prepare time for more injections. We suggest EDX evaluation for the follow-up of treatment efficacy.
Collapse
|
21
|
Yamashita T, Kushida Y, Abe K, Dezawa M. Non-Tumorigenic Pluripotent Reparative Muse Cells Provide a New Therapeutic Approach for Neurologic Diseases. Cells 2021; 10:cells10040961. [PMID: 33924240 PMCID: PMC8074773 DOI: 10.3390/cells10040961] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023] Open
Abstract
Muse cells are non-tumorigenic endogenous reparative pluripotent cells with high therapeutic potential. They are identified as cells positive for the pluripotent surface marker SSEA-3 in the bone marrow, peripheral blood, and connective tissue. Muse cells also express other pluripotent stem cell markers, are able to differentiate into cells representative of all three germ layers, self-renew from a single cell, and are stress tolerant. They express receptors for sphingosine-1-phosphate (S1P), which is actively produced by damaged cells, allowing circulating cells to selectively home to damaged tissue. Muse cells spontaneously differentiate on-site into multiple tissue-constituent cells with few errors and replace damaged/apoptotic cells with functional cells, thereby contributing to tissue repair. Intravenous injection of exogenous Muse cells to increase the number of circulating Muse cells enhances their reparative activity. Muse cells also have a specific immunomodulatory system, represented by HLA-G expression, allowing them to be directly administered without HLA-matching or immunosuppressant treatment. Owing to these unique characteristics, clinical trials using intravenously administered donor-Muse cells have been conducted for myocardial infarction, stroke, epidermolysis bullosa, spinal cord injury, perinatal hypoxic ischemic encephalopathy, and amyotrophic lateral sclerosis. Muse cells have the potential to break through the limitations of current cell therapies for neurologic diseases, including amyotrophic lateral sclerosis. Muse cells provide a new therapeutic strategy that requires no HLA-matching or immunosuppressant treatment for administering donor-derived cells, no gene introduction or differentiation induction for cell preparation, and no surgery for delivering the cells to patients.
Collapse
Affiliation(s)
- Toru Yamashita
- Department of Neurology, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (T.Y.); (K.A.)
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, School of Medicine, Tohoku University, Sendai 980-8575, Japan;
| | - Koji Abe
- Department of Neurology, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (T.Y.); (K.A.)
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, School of Medicine, Tohoku University, Sendai 980-8575, Japan;
- Correspondence: ; Tel.: +81-22-717-8025; Fax: +81-22-717-8030
| |
Collapse
|
22
|
Zhou G, Wang Y, Gao S, Fu X, Cao Y, Peng Y, Zhuang J, Hu J, Shao A, Wang L. Potential Mechanisms and Perspectives in Ischemic Stroke Treatment Using Stem Cell Therapies. Front Cell Dev Biol 2021; 9:646927. [PMID: 33869200 PMCID: PMC8047216 DOI: 10.3389/fcell.2021.646927] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke (IS) remains one of the major causes of death and disability due to the limited ability of central nervous system cells to regenerate and differentiate. Although several advances have been made in stroke therapies in the last decades, there are only a few approaches available to improve IS outcome. In the acute phase of IS, mechanical thrombectomy and the administration of tissue plasminogen activator have been widely used, while aspirin or clopidogrel represents the main therapy used in the subacute or chronic phase. However, in most cases, stroke patients fail to achieve satisfactory functional recovery under the treatments mentioned above. Recently, cell therapy, especially stem cell therapy, has been considered as a novel and potential therapeutic strategy to improve stroke outcome through mechanisms, including cell differentiation, cell replacement, immunomodulation, neural circuit reconstruction, and protective factor release. Different stem cell types, such as mesenchymal stem cells, marrow mononuclear cells, and neural stem cells, have also been considered for stroke therapy. In recent years, many clinical and preclinical studies on cell therapy have been carried out, and numerous results have shown that cell therapy has bright prospects in the treatment of stroke. However, some cell therapy issues are not yet fully understood, such as its optimal parameters including cell type choice, cell doses, and injection routes; therefore, a closer relationship between basic and clinical research is needed. In this review, the role of cell therapy in stroke treatment and its mechanisms was summarized, as well as the function of different stem cell types in stroke treatment and the clinical trials using stem cell therapy to cure stroke, to reveal future insights on stroke-related cell therapy, and to guide further studies.
Collapse
Affiliation(s)
- Guoyang Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjie Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shiqi Gao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiongjie Fu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Cao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yucong Peng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junwen Hu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
Mitani K, Ito Y, Takene Y, Hatoya S, Sugiura K, Inaba T. Long-Term Trypsin Treatment Promotes Stem Cell Potency of Canine Adipose-Derived Mesenchymal Stem Cells. Stem Cells Dev 2021; 30:337-349. [PMID: 33528297 DOI: 10.1089/scd.2020.0175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) isolated from adipose tissue (adipose-derived stem cells [ADSCs]) are considered one of the most promising cell types for applications in regenerative medicine. However, the regenerative potency of ADSCs may vary because of heterogeneity. Long-term trypsin treatment (LTT) is known to significantly concentrate multilineage-differentiating stress-enduring (Muse) cells from human MSCs. In this study, we aimed to generate cells with high stem cell potency from canine ADSCs using LTT. After 16 h of treatment with trypsin, surviving ADSCs (LTT-tolerant cells) had significantly enhanced expression of stage-specific embryonic antigen (SSEA)-1, a mouse embryonic stem cell marker, and fucosyltransferase 9, one of several fucosyltransferases for SSEA-1 biosynthesis. However, LTT-tolerant cells did not enhance the expression of SSEA-3, a known human Muse cell marker. LTT-tolerant cells, however, showed significantly higher self-renewal capacity in the colony-forming unit fibroblast assay than ADSCs. In addition, the LTT-tolerant cells formed cell clusters similar to embryoid bodies and expressed undifferentiated markers. Moreover, these cells differentiated into cells of all three germ layers and showed significantly higher levels of α 2-6 sialic acid (Sia)-specific lectins, known as differentiation potential markers of human MSCs, than ADSCs. LTT-tolerant cells had a normal karyotype and had low telomerase activity, showing little carcinogenetic potency. LTT-tolerant cells also showed significantly increased activity of transmigration in the presence of chemoattractants and had increased expression of migration-related genes compared with ADSCs. In addition, LTT-tolerant cells had stronger suppressive activity against mitogen-stimulated lymphocyte proliferation than ADSCs. Overall, these results indicated that the LTT-tolerant cells in canine ADSCs have similar properties as human Muse cells (although one of the undifferentiated markers is different) and are expected to be a promising tool for regenerative therapy in dogs.
Collapse
Affiliation(s)
- Kosuke Mitani
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan.,Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Yuki Ito
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Yukio Takene
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Shingo Hatoya
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Kikuya Sugiura
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Toshio Inaba
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| |
Collapse
|
24
|
Cao J, Yang Z, Xiao R, Pan B. Regenerative potential of pluripotent nontumorgenetic stem cells: Multilineage differentiating stress enduring cells (Muse cells). Regen Ther 2020; 15:92-96. [PMID: 33426206 PMCID: PMC7770368 DOI: 10.1016/j.reth.2020.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/17/2020] [Accepted: 04/25/2020] [Indexed: 12/15/2022] Open
Abstract
Multilineage differentiating stress enduring cells (Muse cells), double positive for SSEA-3 and CD105, can be isolated by fluorescence-activated cell sorting (FACS) or sever cellular conditions from dermal fibroblasts, bone marrow stem cells (BMSCs), adipose tissue derived stem cells (ADSCs), fresh bone marrow and liposuction fat. When cultured in a single-cell suspension, Muse cells can grow into characteristic cell clusters. Muse cells maintain pluripotency as evidenced by pluripotent markers in vitro. Besides, Muse cells have no tumorigenesis up to 6 months in SCID mice. Muse cells differentiate into cells representative of all three germ layers both spontaneously and under specific induction. In comparison to mesenchymal stem cells (MSCs), Muse cells show higher homing and migration capabilities to damaged sites which is predominantly attributed to S1P–S1PR2 axis. The regenerative effects of Muse cells have been demonstrated by many models in vivo or in vitro, including stroke, intracerebral hemorrhage, myocardial infarction, aortic aneurysm, lung injuries, liver fibrosis, focal segmental glomerulosclerosis, osteochondral defects and skin ulcer. In general, migration, differentiation and paracrine play a pivotal role in the regeneration capability. Here we review the isolation, core properties, preclinical studies as well as the underling molecular and cellular details to highlight their regenerative potential.
Collapse
Affiliation(s)
- Jiankun Cao
- Plastic Surgery Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhigang Yang
- Plastic Surgery Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ran Xiao
- Plastic Surgery Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Pan
- th Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Sun D, Yang L, Cao H, Shen ZY, Song HL. Study of the protective effect on damaged intestinal epithelial cells of rat multilineage-differentiating stress-enduring (Muse) cells. Cell Biol Int 2019; 44:549-559. [PMID: 31642560 PMCID: PMC7003933 DOI: 10.1002/cbin.11255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 10/19/2019] [Indexed: 12/12/2022]
Abstract
In this study, we determined whether multilineage‐differentiating stress‐enduring (Muse) cells exist in rat bone marrow and elucidated their effects on protection against the injury of intestinal epithelial cells associated with inflammation. Rat Muse cells were separated from bone marrow mesenchymal stem cells (BMMSCs) by trypsin‐incubation stress. The group of cells maintained the characteristics of BMMSCs; however, there were high positive expression levels of stage‐specific embryonic antigen‐3 (SSEA‐3; 75.6 ± 2.8%) and stage‐specific embryonic antigen‐1 (SSEA‐1; 74.8 ± 3.1%), as well as specific antigens including Nanog, POU class 5 homeobox 1 (OCT 3/4), and SRY‐box 2 (SOX 2). After inducing differentiation, α‐fetoprotein (endodermal), α‐smooth muscle actin and neurofilament medium polypeptide (ectodermal) were positive in Muse cells. Injuries of intestinal epithelial crypt cell‐6 (IEC‐6) and colorectal adenocarcinoma 2 (Caco‐2) cells as models were induced by tumor necrosis factor‐α stimulation in vitro. Muse cells exhibited significant protective effects on the proliferation and intestinal barrier structure, the underlying mechanisms of which were related to reduced levels of interleukin‐6 (IL‐6) and interferon‐γ (IFN‐γ), and the restoration of transforming growth factor‐β (TGF‐β) and IL‐10 in the inflammation microenvironment. In summary, there were minimal levels of pluripotent stem cells in rat bone marrow, which exhibit similar properties to human Muse cells. Rat Muse cells could provide protection against damage to intestinal epithelial cells depending on their anti‐inflammatory and immune regulatory functionality. Their functional impact was more obvious than that of BMMSCs.
Collapse
Affiliation(s)
- Dong Sun
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, P.R. China
| | - Liu Yang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, P.R. China.,Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, P.R. China
| | - Huan Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, P.R. China.,NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, P.R. China
| | - Zhong-Yang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, P.R. China.,Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, 300192, P.R. China
| | - Hong-Li Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, 300192, P.R. China.,Tianjin Key Laboratory of Organ Transplantation, Tianjin, 300192, P.R. China
| |
Collapse
|
26
|
Leng Z, Sun D, Huang Z, Tadmori I, Chiang N, Kethidi N, Sabra A, Kushida Y, Fu YS, Dezawa M, He X, Young W. Quantitative Analysis of SSEA3+ Cells from Human Umbilical Cord after Magnetic Sorting. Cell Transplant 2019; 28:907-923. [PMID: 30997834 PMCID: PMC6719495 DOI: 10.1177/0963689719844260] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are a population of pluripotent stage-specific embryonic antigen 3 (SSEA3)+ mesenchymal stem cells first described by Mari Dezawa in 2010. Although some investigators have reported SSEA3+ mesenchymal cells in umbilical cord tissues, none have quantitatively compared SSEA3+ cells isolated from Wharton’s jelly (WJ) and the cord lining (CL) of human umbilical cords (HUCs). We separated WJ and the CL from HUCs, cultured mesenchymal stromal cells (MSCs) isolated from these two tissues with collagenase, and quantified the percentage of SSEA3+ cells over three passages. The first passage had 5.0% ± 4.3% and 5.3% ± 5.1% SSEA3+ cells from WJ and the CL, respectively, but the percentage of SSEA3+ cells decreased significantly (P < 0.05) between P0 and P2 in the CL group and between P0 and P1 in the WJ group. Magnetic-activated cell sorting (MACS) markedly enriched SSEA3+ cells to 91.4% ± 3.2%. Upon culture of the sorted population, we found that the SSEA3+ percentage ranged from 62.5% to 76.0% in P2–P5 and then declined to 42.0%–54.7% between P6 and P9. At P10, the cultures contained 37.4% SSEA3+ cells. After P10, we resorted the cells and achieved 89.4% SSEA3+ cells in culture. The procedure for MACS-based enrichment of SSEA3+ cells, followed by expansion in culture and a re-enrichment step, allows the isolation of many millions of SSEA3+ cells in relatively pure culture. When cultured, the sorted SSEA3+ cells differentiated into embryoid spheres and survived 4 weeks after transplant into a contused Sprague-Dawley rat spinal cord. The transplanted SSEA3+ cells migrated into the injury area from four injection points around the contusion site and did not produce any tumors. The umbilical cord is an excellent source of fetal Muse cells, and our method allows the practical and efficient isolation and expansion of relatively pure populations of SSEA3+ Muse cells that can be matched by human leukocyte antigen for transplantation in human trials.
Collapse
Affiliation(s)
- Zikuan Leng
- 1 Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Dongming Sun
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Zihao Huang
- 3 Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei
| | - Iman Tadmori
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Ning Chiang
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Nikhit Kethidi
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Ahmed Sabra
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| | - Yoshihiro Kushida
- 4 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu-Show Fu
- 3 Department of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei
| | - Mari Dezawa
- 4 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Xijing He
- 1 Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wise Young
- 2 W.M. Keck Center for Collaborative Neuroscience, Rutgers, the State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
27
|
Amin M, Kushida Y, Wakao S, Kitada M, Tatsumi K, Dezawa M. Cardiotrophic Growth Factor-Driven Induction of Human Muse Cells Into Cardiomyocyte-Like Phenotype. Cell Transplant 2019; 27:285-298. [PMID: 29637816 PMCID: PMC5898685 DOI: 10.1177/0963689717721514] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous nontumorigenic stem cells collectable as stage-specific embryonic antigen 3 (SSEA-3) + from various organs including the bone marrow and are pluripotent-like. The potential of human bone marrow-derived Muse cells to commit to cardiac lineage cells was evaluated. We found that (1) initial treatment of Muse cells with 5'-azacytidine in suspension culture successfully accelerated demethylation of cardiac marker Nkx2.5 promoter; (2) then transferring the cells onto adherent culture and treatment with early cardiac differentiation factors including wingless-int (Wnt)-3a, bone morphogenetic proteins (BMP)-2/4, and transforming growth factor (TGF) β1; and (3) further treatment with late cardiac differentiation cytokines including cardiotrophin-1 converted Muse cells into cardiomyocyte-like cells that expressed α-actinin and troponin-I with a striation-like pattern. MLC2a expression in the final step suggested differentiation of the cells into an atrial subtype. MLC2v, a marker for a mature ventricular subtype, was expressed when cells were treated with Dickkopf-related protein 1 (DKK-1) and Noggin, inhibitors of Wnt3a and BMP-4, respectively, between steps (2) and (3). None of the steps included exogenous gene transfection, making induced cells feasible for future clinical application.
Collapse
Affiliation(s)
- Mohamed Amin
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,2 Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia, Egypt
| | - Yoshihiro Kushida
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shohei Wakao
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masaaki Kitada
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuki Tatsumi
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan.,3 Life Science Institute Inc., Regenerative Medicine Division, Nagoya, Japan
| | - Mari Dezawa
- 1 Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
28
|
Labusca L, Mashayekhi K. Human adult pluripotency: Facts and questions. World J Stem Cells 2019; 11:1-12. [PMID: 30705711 PMCID: PMC6354101 DOI: 10.4252/wjsc.v11.i1.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/16/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
Cellular reprogramming and induced pluripotent stem cell (IPSC) technology demonstrated the plasticity of adult cell fate, opening a new era of cellular modelling and introducing a versatile therapeutic tool for regenerative medicine. While IPSCs are already involved in clinical trials for various regenerative purposes, critical questions concerning their medium- and long-term genetic and epigenetic stability still need to be answered. Pluripotent stem cells have been described in the last decades in various mammalian and human tissues (such as bone marrow, blood and adipose tissue). We briefly describe the characteristics of human-derived adult stem cells displaying in vitro and/or in vivo pluripotency while highlighting that the common denominators of their isolation or occurrence within tissue are represented by extreme cellular stress. Spontaneous cellular reprogramming as a survival mechanism favoured by senescence and cellular scarcity could represent an adaptative mechanism. Reprogrammed cells could initiate tissue regeneration or tumour formation dependent on the microenvironment characteristics. Systems biology approaches and lineage tracing within living tissues can be used to clarify the origin of adult pluripotent stem cells and their significance for regeneration and disease.
Collapse
Affiliation(s)
- Luminita Labusca
- National Institute of Research and Development for Advanced Technical Physics Iasi, Iasi 700349, Romania
| | - Kaveh Mashayekhi
- Systems Biomedical Informatics and Modeling, Frankfurt D-45367, Germany
| |
Collapse
|
29
|
Leng Z, Kethidi N, Chang AJ, Sun L, Zhai J, Yang Y, Xu J, He X. Muse cells and Neurorestoratology. JOURNAL OF NEURORESTORATOLOGY 2019. [DOI: 10.26599/jnr.2019.9040005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells were discovered in 2010 as a subpopulation of mesenchymal stroma cells (MSCs). Muse cells can self-renew and tolerate severe culturing conditions. These cells can differentiate into three lineage cells spontaneously or in induced medium but do not form teratoma in vitro or in vivo. Central nervous system (CNS) diseases, such as intracerebral hemorrhage (ICH), cerebral infarction, and spinal cord injury are normally disastrous. Despite numerous therapy strategies, CNS diseases are difficult to recover. As a novel kind of pluripotent stem cells, Muse cells have shown great regeneration capacity in many animal models, including acute myocardial infarction, hepatectomy, and acute cerebral ischemia (ACI). After injection into injury sites, Muse cells survived, migrated, and differentiated into functional neurons with synaptic junctions to local neurons and contributed to recovery of function. Furthermore, Muse cell differentiation did not need to be induced pre-transplantation and no tumors were observed post- transplantation. The Muse cell population is promising and may lead to a revolution in regenerative medicine. This review focuses on recent advances regarding the Muse cells therapies in Neurorestoratology and discusses future perspectives in this field.
Collapse
|
30
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
31
|
Wang Z, Sun D. Adipose-Derived Mesenchymal Stem Cells: A New Tool for the Treatment of Renal Fibrosis. Stem Cells Dev 2018; 27:1406-1411. [PMID: 30032706 DOI: 10.1089/scd.2017.0304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As chronic kidney disease progresses, kidney tissue inevitably undergoes cell loss, accumulation of extracellular matrix, and kidney tissue fibrosis, eventually leading to end-stage renal disease. With the continuous innovation of cell therapy technology, mesenchymal stem cells are used in numerous fields, including cardiovascular diseases, diabetes, and kidney tissue injury repair. Adipose-derived mesenchymal stem cells (AMSCs), a type of pluripotent stem cells, have the potential for self-renewal and proliferation with low immunogenicity and significant anti-inflammatory properties. AMSCs can promote impaired cell regeneration and remodeling in renal lesions, thus avoiding further worsening of renal disease and even blocking or reversing the process of renal fibrosis. In this review, we discuss the mechanisms involved in the treatment of renal fibrosis with AMSCs and summarize the potential hazards that may exist in cell therapy.
Collapse
Affiliation(s)
- Zhuojun Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China
| | - Dong Sun
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China .,2 Department of Internal Medicine and Diagnostics, Xuzhou Medical University , Xuzhou, China
| |
Collapse
|
32
|
Yabuki H, Wakao S, Kushida Y, Dezawa M, Okada Y. Human Multilineage-differentiating Stress-Enduring Cells Exert Pleiotropic Effects to Ameliorate Acute Lung Ischemia-Reperfusion Injury in a Rat Model. Cell Transplant 2018; 27:979-993. [PMID: 29707971 PMCID: PMC6050908 DOI: 10.1177/0963689718761657] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/29/2018] [Indexed: 12/26/2022] Open
Abstract
Posttransplantation lung ischemia-reperfusion (IR) injuries affect both patient survival and graft function. In this study, we evaluated the protective effects of infused human multilineage-differentiating stress-enduring (Muse) cells, a novel, easily harvested type of nontumorigenic endogenous reparative stem cell, against acute IR lung injury in a rat model. After a 2-h warm IR injury induction in a left rat lung, human Muse cells, human mesenchymal stem cells (MSCs), and vehicle were injected via the left pulmonary artery after reperfusion. Functionality, histological findings, and protein expression were subsequently assessed in the injured lung. In vitro, we also compared human Muse cells with human MSCs in terms of migration abilities and the secretory properties of protective substances. The arterial oxygen partial pressure to fractional inspired oxygen ratio, alveolar-arterial oxygen gradient, left lung compliance, and histological injury score on hematoxylin-eosin sections were significantly better in the Muse group relative to the MSC and vehicle groups. Compared to MSCs, human Muse cells homed more efficiently to the injured lung, where they suppressed the apoptosis and stimulated proliferation of host alveolar cells. Human Muse cells also migrated to serum from lung-injured model rats and produced beneficial substances (keratinocyte growth factor [KGF], hepatocyte growth factor, angiopoietin-1, and prostaglandin E2) in vitro. Western blot of lung tissue confirmed high expression of KGF and their target molecules (interleukin-6, protein kinase B, and B-cell lymphoma-2) in the Muse group. Thus, Muse cells efficiently ameliorated lung IR injury via pleiotropic effects in a rat model. These findings support further investigation on the use of human Muse cells for lung IR injury.
Collapse
Affiliation(s)
- Hiroshi Yabuki
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer,
Tohoku University, Sendai, Miyagi, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate
School of Medicine, Sendai, Miyagi, Japan
| | - Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate
School of Medicine, Sendai, Miyagi, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate
School of Medicine, Sendai, Miyagi, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer,
Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
33
|
Hosoyama K, Wakao S, Kushida Y, Ogura F, Maeda K, Adachi O, Kawamoto S, Dezawa M, Saiki Y. Intravenously injected human multilineage-differentiating stress-enduring cells selectively engraft into mouse aortic aneurysms and attenuate dilatation by differentiating into multiple cell types. J Thorac Cardiovasc Surg 2018; 155:2301-2313.e4. [DOI: 10.1016/j.jtcvs.2018.01.098] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/13/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
|
34
|
Della Corte A, Di Mauro M, Forte A. Musing on cell therapy for aortic aneurysms. J Thorac Cardiovasc Surg 2018; 155:2314-2315. [PMID: 29555089 DOI: 10.1016/j.jtcvs.2018.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/11/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Alessandro Della Corte
- Department of Cardiothoracic Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Michele Di Mauro
- Department of Cardiovascular Disease, University of L'Aquila, L'Aquila, Italy.
| | - Amalia Forte
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
35
|
Muse Cells Are Endogenous Reparative Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:43-68. [PMID: 30484223 DOI: 10.1007/978-4-431-56847-6_3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The dynamics and actions of Muse cells at a time of physical crisis are unique and highly remarkable compared with other stem cell types. When the living body is in a steady state, low levels of Muse cells are mobilized to the peripheral blood, possibly from the bone marrow, and supplied to the connective tissue of nearly every organ. Under conditions of serious tissue damage, such as acute myocardial infarction and stroke, Muse cells are highly mobilized to the peripheral blood, drastically increasing their numbers in the peripheral blood within 24 h after the onset of tissue injury. The alerting signal, sphingosine-1-phosphate, attracts Muse cells to the damaged site mainly via the sphingosine-1-phosphate receptor 2, enabling them to preferentially home to site of injury. After homing, Muse cells spontaneously differentiate into tissue-compatible cells and replenish new functional cells for tissue repair. Because Muse cells have pleiotropic effects, including paracrine, anti-inflammatory, anti-fibrotic, and anti-apoptotic effects, these cells synergistically deliver long-lasting functional and structural recovery. This chapter describes how Muse cells exert their reparative effects in vivo.
Collapse
|
36
|
Hosoyama K, Saiki Y. Muse Cells and Aortic Aneurysm. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:273-291. [PMID: 30484235 DOI: 10.1007/978-4-431-56847-6_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The aorta is a well-organized, multilayered structure comprising several cell types, namely, endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and fibroblasts, as well as an extracellular matrix (ECM), which includes elastic and collagen fibers. Aortic aneurysms (AAs) are defined as progressive enlargements of the aorta that carries an incremental risk of rupture as the diameter increases over time. The destruction of the aortic wall tissue is triggered by atherosclerosis, inflammation, and oxidative stress, leading to the activation of matrix metalloproteinases (MMPs), and inflammatory cytokines and chemokines, resulting in the loss of the structural back bone of VSMCs, ECM, and ECs. To date, cell-based therapy has been applied to animal models using several types of cells, such as VSMCs, ECs, and mesenchymal stem cells (MSCs). Although these cells indeed deliver beneficial outcomes for AAs, particularly by paracrine and immunomodulatory effects, the attenuation of aneurysmal dilation with a robust tissue repair is insufficient. Meanwhile, multilineage-differentiating stress-enduring (Muse) cells are known to be endogenous non-tumorigenic pluripotent-like stem cells that are included as several percent of MSCs. Since Muse cells are pluripotent-like, they have the ability to differentiate into cells representative of all three germ layers from a single cell and to self-renew. Moreover, Muse cells are able to home to the site of damage following simple intravenous injection and repair the tissue by replenishing new functional cells through spontaneous differentiation into tissue-compatible cells. Given these unique properties, Muse cells are expected to provide an efficient therapeutic efficacy for AA by simple intravenous injection. In this chapter, we summarize several studies on Muse cell therapy for AA including our recent data, in comparison with other kinds of cell therapies.
Collapse
Affiliation(s)
- Katsuhiro Hosoyama
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshikatsu Saiki
- Division of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
37
|
Muse Cells and Ischemia-Reperfusion Lung Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:293-303. [DOI: 10.1007/978-4-431-56847-6_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
38
|
Perone MJ, Gimeno ML, Fuertes F. Immunomodulatory Properties and Potential Therapeutic Benefits of Muse Cells Administration in Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:115-129. [PMID: 30484226 DOI: 10.1007/978-4-431-56847-6_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is well established the link between inflammation and the development of insulin resistance and pathogenesis of type 2 diabetes. Type 1 diabetes is an autoimmune disease characterized by the destruction of insulin-producing pancreatic β cells mediated by autoreactive T lymphocytes and pro-inflammatory agents. Therefore, developing new strategies to efficiently control dysregulated inflammation could have substantial benefits in the treatment of diabetes. Recently, a novel population of non-tumorigenic pluripotent stem cells, named multilineage-differentiating stress-enduring (Muse) cells, was discovered. Muse cells secrete significant amounts of TGF-β1, a key cytokine governing down-modulation of T lymphocytes and macrophages. In this chapter, we discuss the immunomodulatory properties of Muse cells as well as the molecular mechanism of TGF-β1 as mediator of Muse cell action. We also describe the role of certain cytokines/growth factors highly expressed in Muse cells as potential mediators of their effects. Finally, we provide evidence of the beneficial effects of adipose tissue-derived Muse cells in an experimental mice model of type 1 diabetes.
Collapse
Affiliation(s)
- Marcelo Javier Perone
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - María Laura Gimeno
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Florencia Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| |
Collapse
|
39
|
Yamauchi T, Yamasaki K, Tsuchiyama K, Koike S, Aiba S. The Potential of Muse Cells for Regenerative Medicine of Skin: Procedures to Reconstitute Skin with Muse Cell-Derived Keratinocytes, Fibroblasts, and Melanocytes. J Invest Dermatol 2017; 137:2639-2642. [DOI: 10.1016/j.jid.2017.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 11/30/2022]
|
40
|
Fisch SC, Gimeno ML, Phan JD, Simerman AA, Dumesic DA, Perone MJ, Chazenbalk GD. Pluripotent nontumorigenic multilineage differentiating stress enduring cells (Muse cells): a seven-year retrospective. Stem Cell Res Ther 2017; 8:227. [PMID: 29041955 PMCID: PMC5646122 DOI: 10.1186/s13287-017-0674-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multilineage differentiating stress enduring (Muse) cells, discovered in the spring of 2010 at Tohoku University in Sendai, Japan, were quickly recognized by scientists as a possible source of pluripotent cells naturally present within mesenchymal tissues. Muse cells normally exist in a quiescent state, singularly activated by severe cellular stress in vitro and in vivo. Muse cells have the capacity for self-renewal while maintaining pluripotent cell characteristics indicated by the expression of pluripotent stem cell markers. Muse cells differentiate into cells representative of all three germ cell layers both spontaneously and under media-specific induction. In contrast to embryonic stem and induced pluripotent stem cells, Muse cells exhibit low telomerase activity, a normal karyotype, and do not undergo tumorigenesis once implanted in SCID mice. Muse cells efficiently home into damaged tissues and differentiate into specific cells leading to tissue regeneration and functional recovery as described in different animal disease models (i.e., fulminant hepatitis, muscle degeneration, skin ulcers, liver cirrhosis, cerebral stroke, vitiligo, and focal segmental glomerulosclerosis). Circulating Muse cells have been detected in peripheral blood, with higher levels present in stroke patients during the acute phase. Furthermore, Muse cells have inherent immunomodulatory properties, which could contribute to tissue generation and functional repair in vivo. Genetic studies in Muse cells indicate a highly conserved cellular mechanism as seen in more primitive organisms (yeast, Saccharomyces cerevisiae, Caenorhabditis elegans, chlamydomonas, Torpedo californica, drosophila, etc.) in response to cellular stress and acute injury. This review details the molecular and cellular properties of Muse cells as well as their capacity for tissue repair and functional recovery, highlighting their potential for clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Samantha C. Fisch
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, The University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 USA
| | - María L. Gimeno
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)—CONICET—Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Julia D. Phan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, The University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 USA
| | - Ariel A. Simerman
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, The University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 USA
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, The University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 USA
| | - Marcelo J. Perone
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)—CONICET—Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD Buenos Aires, Argentina
| | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, The University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 USA
| |
Collapse
|
41
|
Uchida N, Kushida Y, Kitada M, Wakao S, Kumagai N, Kuroda Y, Kondo Y, Hirohara Y, Kure S, Chazenbalk G, Dezawa M. Beneficial Effects of Systemically Administered Human Muse Cells in Adriamycin Nephropathy. J Am Soc Nephrol 2017; 28:2946-2960. [PMID: 28674043 DOI: 10.1681/asn.2016070775] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 05/08/2017] [Indexed: 01/24/2023] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are nontumorigenic endogenous pluripotent-like stem cells that can be collected from various organs. Intravenously administered Muse cells have been shown to spontaneously migrate to damaged tissue and replenish lost cells, but the effect in FSGS is unknown. We systemically administered human bone marrow-derived Muse cells without concurrent administration of immunosuppressants to severe combined immune-deficient (SCID) and BALB/c mouse models with adriamycin-induced FSGS (FSGS-SCID and FSGS-BALB/c, respectively). In FSGS-SCID mice, human Muse cells preferentially integrated into the damaged glomeruli and spontaneously differentiated into cells expressing markers of podocytes (podocin; 31%), mesangial cells (megsin; 13%), and endothelial cells (CD31; 41%) without fusing to the host cells; attenuated glomerular sclerosis and interstitial fibrosis; and induced the recovery of creatinine clearance at 7 weeks. Human Muse cells induced similar effects in FSGS-BALB/c mice at 5 weeks, despite xenotransplant without concurrent immunosuppressant administration, and led to improvement in urine protein, creatinine clearance, and plasma creatinine levels more impressive than that in the FSGS-SCID mice at 5 weeks. However, functional recovery in FSGS-BALB/c mice was impaired at 7 weeks due to immunorejection, suggesting the importance of Muse cell survival as glomerular cells in the FSGS kidney for tissue repair and functional recovery. In conclusion, Muse cells are unique reparative stem cells that preferentially home to damaged glomeruli and spontaneously differentiate into glomerular cells after systemic administration. Introduction of genes to induce differentiation is not required before Muse cell administration; thus, Muse cells may be a feasible therapeutic strategy in FSGS.
Collapse
Affiliation(s)
- Nao Uchida
- Departments of Stem Cell Biology and Histology and.,Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | - Shohei Wakao
- Departments of Stem Cell Biology and Histology and
| | - Naonori Kumagai
- Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Yoshiaki Kondo
- Department of Healthcare Services Management, Nihon University School of Medicine, Tokyo, Japan
| | - Yukari Hirohara
- Departments of Stem Cell Biology and Histology and.,Regenerative Medicine Division, Life Science Institute, Inc., Tokyo, Japan; and
| | - Shigeo Kure
- Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Gregorio Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Mari Dezawa
- Departments of Stem Cell Biology and Histology and
| |
Collapse
|
42
|
Yamasaki T, Wakao S, Kawaji H, Koizumi S, Sameshima T, Dezawa M, Namba H. Genetically Engineered Multilineage-Differentiating Stress-Enduring Cells as Cellular Vehicles against Malignant Gliomas. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:45-56. [PMID: 28748212 PMCID: PMC5514691 DOI: 10.1016/j.omto.2017.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/10/2017] [Indexed: 01/14/2023]
Abstract
Malignant glioma, the most common malignant brain tumor in adults, is difficult to treat due to its aggressive invasive nature. Enzyme/prodrug suicide gene therapy based on the herpes simplex virus thymidine kinase (HSVtk)/ganciclovir (GCV) system is an efficient strategy for treating malignant gliomas. In the present study, we evaluated treatment with multilineage-differentiating stress-enduring (Muse) cells, which are endogenous non-tumorigenic pluripotent-like stem cells that are easily collectable from the bone marrow as SSEA-3+ cells, as carriers of the HSVtk gene. Human Muse cells showed potent migratory activity toward glioma cells both in vitro and in vivo. HSVtk gene-transduced Muse cells (Muse-tk cells) at a cell number of only 1/32 that of U87 human glioma cells completely eradicated U87 gliomas in nude mouse brains, showing a robust in vivo bystander effect. Pre-existing intracranial U87 gliomas in nude mouse brains injected intratumorally with Muse-tk cells followed by intraperitoneal GCV administration were significantly reduced in size within 2 weeks, and 4 of 10 treated mice survived over 200 days. These findings suggest that intratumoral Muse-tk cell injection followed by systemic GCV administration is safe and effective and that allogeneic Muse-tk cell-medicated suicide gene therapy for malignant glioma is clinically feasible.
Collapse
Affiliation(s)
- Tomohiro Yamasaki
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 1-1 Seiryocho, Aoba-ku, Sendai, 980-8574, Japan
| | - Hiroshi Kawaji
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Tetsuro Sameshima
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 1-1 Seiryocho, Aoba-ku, Sendai, 980-8574, Japan
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan
| |
Collapse
|
43
|
Simerman AA, Phan JD, Dumesic DA, Chazenbalk GD. Muse Cells: Nontumorigenic Pluripotent Stem Cells Present in Adult Tissues-A Paradigm Shift in Tissue Regeneration and Evolution. Stem Cells Int 2016; 2016:1463258. [PMID: 28070194 PMCID: PMC5192335 DOI: 10.1155/2016/1463258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/15/2016] [Accepted: 10/24/2016] [Indexed: 01/26/2023] Open
Abstract
Muse cells are a novel population of nontumorigenic pluripotent stem cells, highly resistant to cellular stress. These cells are present in every connective tissue and intrinsically express pluripotent stem markers such as Nanog, Oct3/4, Sox2, and TRA1-60. Muse cells are able to differentiate into cells from all three embryonic germ layers both spontaneously and under media-specific induction. Unlike ESCs and iPSCs, Muse cells exhibit low telomerase activity and asymmetric division and do not undergo tumorigenesis or teratoma formation when transplanted into a host organism. Muse cells have a high capacity for homing into damaged tissue and spontaneous differentiation into cells of compatible tissue, leading to tissue repair and functional restoration. The ability of Muse cells to restore tissue function may demonstrate the role of Muse cells in a highly conserved cellular mechanism related to cell survival and regeneration, in response to cellular stress and acute injury. From an evolutionary standpoint, genes pertaining to the regenerative capacity of an organism have been lost in higher mammals from more primitive species. Therefore, Muse cells may offer insight into the molecular and evolutionary bases of autonomous tissue regeneration and elucidate the molecular and cellular mechanisms that prevent mammals from regenerating limbs and organs, as planarians, newts, zebrafish, and salamanders do.
Collapse
Affiliation(s)
- Ariel A. Simerman
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at The University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Julia D. Phan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at The University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at The University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gregorio D. Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at The University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
44
|
Iseki M, Kushida Y, Wakao S, Akimoto T, Mizuma M, Motoi F, Asada R, Shimizu S, Unno M, Chazenbalk G, Dezawa M. Muse Cells, Nontumorigenic Pluripotent-Like Stem Cells, Have Liver Regeneration Capacity Through Specific Homing and Cell Replacement in a Mouse Model of Liver Fibrosis. Cell Transplant 2016; 26:821-840. [PMID: 27938474 DOI: 10.3727/096368916x693662] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Muse cells, a novel type of nontumorigenic pluripotent-like stem cells, reside in the bone marrow, skin, and adipose tissue and are collectable as cells positive for pluripotent surface marker SSEA-3. They are able to differentiate into cells representative of all three germ layers. The capacity of intravenously injected human bone marrow-derived Muse cells to repair an immunodeficient mouse model of liver fibrosis was evaluated in this study. The cells exhibited the ability to spontaneously differentiate into hepatoblast/hepatocyte lineage cells in vitro. They demonstrated a high migration capacity toward the serum and liver section of carbon tetrachloride-treated mice in vitro. In vivo, they specifically accumulated in the liver, but not in other organs except, to a lesser extent, in the lungs at 2 weeks after intravenous injection in the liver fibrosis model. After homing, Muse cells spontaneously differentiated in vivo into HepPar-1 (71.1 ± 15.2%), human albumin (54.3 ± 8.2%), and anti-trypsin (47.9 ± 4.6%)-positive cells without fusing with host hepatocytes, and expressed mature functional markers such as human CYP1A2 and human Glc-6-Pase at 8 weeks after injection. Recovery in serum, total bilirubin, and albumin and significant attenuation of fibrosis were recognized with statistical differences between the Muse cell-transplanted group and the control groups, which received the vehicle or the same number of a non-Muse cell population of MSCs (MSCs in which Muse cells were eliminated). Thus, unlike ESCs and iPSCs, Muse cells are unique in their efficient migration and integration into the damaged liver after intravenous injection, nontumorigenicity, and spontaneous differentiation into hepatocytes, rendering induction into hepatocytes prior to transplantation unnecessary. They may repair liver fibrosis by two simple steps: expansion after collection from the bone marrow and intravenous injection. A therapeutic strategy such as this is feasible and may provide significant advancements toward liver regeneration in patients with liver disease.
Collapse
|