1
|
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
|
2
|
Entessari M, Oliveira LP. Current evidence on mesenchymal stem cells for hip osteoarthritis: a narrative review. Regen Med 2023; 18:749-758. [PMID: 37496424 DOI: 10.2217/rme-2023-0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023] Open
Abstract
There are limited data on the use of mesenchymal stem cell injections for hip osteoarthritis. The goal of this study was to evaluate the literature by analyzing outcomes and comparing methodologies. Online search of PubMed, SportsDiscus and Case Reports Keywords was completed using the keywords 'stem cells' and 'hip' and 'osteoarthritis'. Six studies met the inclusion and exclusion criteria. Five out the six studies had statistically significant improvement in patient reported outcomes after mesenchymal stem cell injections. Only two studies provided information on radiological changes and findings were positive. None of the studies reported major complications. Small series of non-randomized controlled trials completed to date in the use of mesenchymal stem cells for the treatment of hip osteoarthritis reported the procedures to be safe and provide a positive clinical response. Randomized controlled trials must be performed to further confirm mesenchymal stem cells as a treatment option for hip osteoarthritis.
Collapse
Affiliation(s)
- Mina Entessari
- Florida International University, Herbert Wertheim College of Medicine, 11200 SW 8th Street, AHC2, Miami, FL 33199, USA
| | - Leonardo P Oliveira
- Levitetz Department of Orthopaedic Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd, Weston, FL 33331, USA
| |
Collapse
|
3
|
Li H, Wei J, Zhang Z, Li J, Ma Y, Zhang P, Lin J. Menstrual blood-derived endometrial stem cells alleviate neuroinflammation by modulating M1/M2 polarization in cell and rat Parkinson's disease models. Stem Cell Res Ther 2023; 14:85. [PMID: 37055866 PMCID: PMC10099022 DOI: 10.1186/s13287-023-03330-7] [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: 07/28/2022] [Accepted: 04/05/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Neuroinflammation is closely related to the development of Parkinson's disease (PD). Because of the extensive sources, non-invasive and periodical collection method, human menstrual blood-derived endometrial stem cells (MenSCs) have been explored as a promising tool for treatment of PD. This study aimed to investigate if MenSCs could inhibit neuroinflammation in PD rats by regulating M1/M2 polarization and to excavate the underlying mechanisms. METHODS MenSCs were co-cultured with 6-OHDA-exposed microglia cell lines. Then the morphology of microglia cells and the level of inflammatory factors were assessed by immunofluorescence and qRT-PCR. After MenSCs were transplanted into the brain of PD rats, animal motor function, the expression of tyrosine hydroxylase, and the level of inflammatory factors in the cerebrospinal fluid (CSF) and serum were detected to evaluate the therapeutic potential of MenSCs. Meanwhile, the expression of M1/M2 phenotype related genes was detected by qRT-PCR. One protein array kit containing 1000 kinds of factors was used to detect the protein components in the conditioned medium of MenSCs. Finally, bioinformatic analysis was performed to analyze the function of factors secreted by MenSCs and the signal pathways involved in. RESULTS MenSCs could suppress 6-OHDA-induced microglia cell activation and significantly decrease inflammation in vitro. After transplantation into the brain of PD rats, MenSCs improved animal motor function, which was indicated by the increased movement distance, ambulatory episodes, exercise time on the rotarod, and less contralateral rotation. Additionally, MenSCs reduced the loss of dopaminergic neurons and down-regulated the level of pro-inflammatory factors in the CSF and serum. Moreover, q-PCR and WB results showed the transplantation of MenSCs significantly down-regulated the expression of M1 phenotype cell markers and meanwhile up-regulated the expression of M2 phenotype cell markers in the brain of PD rats. 176 biological processes including inflammatory response, negative regulation of apoptotic process, and microglial cell activation were enriched by GO-BP analysis. 58 signal pathways including PI3K/Akt and MAPK were enriched by KEGG analysis. CONCLUSIONS In conclusion, our results provide preliminary evidence for the anti-inflammation capacity of MenSCs by regulating M1/M2 polarization. We firstly demonstrated the biological process of factors secreted by MenSCs and the signal pathways involved in using protein array and bioinformatic analysis.
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
| | - Zhigang Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Junyao 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
| | - Yaokai Ma
- 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
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 45003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
4
|
Wang S, Wang P, Zhang R. Adipose tissue-derived Muse cells promote autophagy and oxidative stress tolerance in human epidermal melanocytes. Cell Tissue Bank 2023; 24:253-264. [PMID: 35986799 DOI: 10.1007/s10561-022-10031-7] [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: 01/19/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
Abstract
To investigate the effect of human adipose tissue-derived multilineage-differentiating stress-enduring (Muse) cells on the oxidative stress injury of human epidermal melanocytes (HEMs) in vitro. HEMs were treated with H2O2 to establish an oxidative stress injury model and then were co-cultured with adipose tissue-derived Muse cells. Immunohistochemistry, flow cytometry and Western blotting were used to assess changes in autophagy flux, apoptosis, expression of melanin synthesis related proteins and proliferation of melanocytes. Our findings demonstrate that co-culture with Muse cells significantly increased the tolerance of HEMs to oxidative stress, enhanced autophagy flux and reduced apoptosis. The expression of proteins related to the formation of melanin increased as did cell proliferation. Treatment with the autophagy inhibitor, 3-methyladenine (3MA), partially counteracted the improvement of oxidative stress tolerance in melanocytes elicited by co-culture with Muse cells. Muse cells promote autophagy and oxidative stress tolerance of melanocytes.
Collapse
Affiliation(s)
- Shengyi Wang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Peng Wang
- Department of Intensive Care Unit, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruzhi Zhang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
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
|
Quintero Sierra LA, Biswas R, Conti A, Busato A, Ossanna R, Zingaretti N, Parodi PC, Conti G, Riccio M, Sbarbati A, De Francesco F. Highly Pluripotent Adipose-Derived Stem Cell-Enriched Nanofat: A Novel Translational System in Stem Cell Therapy. Cell Transplant 2023; 32:9636897231175968. [PMID: 37243545 DOI: 10.1177/09636897231175968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023] Open
Abstract
Fat graft is widely used in plastic and reconstructive surgery. The size of the injectable product, the unpredictable fat resorption rates, and subsequent adverse effects make it tricky to inject untreated fat into the dermal layer. Mechanical emulsification of fat tissue, which Tonnard introduced, solves these problems, and the product obtained was called nanofat. Nanofat is widely used in clinical and aesthetic settings to treat facial compartments, hypertrophic and atrophic scars, wrinkle attenuation, skin rejuvenation, and alopecia. Several studies demonstrate that the tissue regeneration effects of nanofat are attributable to its rich content of adipose-derived stem cells. This study aimed to characterize Hy-Tissue Nanofat product by investigating morphology, cellular yield, adipose-derived stem cell (ASC) proliferation rate and clonogenic capability, immunophenotyping, and differential potential. The percentage of SEEA3 and CD105 expression was also analyzed to establish the presence of multilineage-differentiating stress-enduring (MUSE) cell. Our results showed that the Hy-Tissue Nanofat kit could isolate 3.74 × 104 ± 1.31 × 104 proliferative nucleated cells for milliliter of the treated fat. Nanofat-derived ASC can grow in colonies and show high differentiation capacity into adipocytes, osteocytes, and chondrocytes. Moreover, immunophenotyping analysis revealed the expression of MUSE cell antigen, making this nanofat enriched of pluripotent stem cell, increasing its potential in regenerative medicine. The unique characteristics of MUSE cells give a simple, feasible strategy for treating a variety of diseases.
Collapse
Affiliation(s)
| | - Reetuparna Biswas
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
| | - Anita Conti
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
| | - Alice Busato
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
- Safety Assessment Department, Aptuit (Verona) S.r.l., an Evotec Company, Verona, Italy
| | - Riccardo Ossanna
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
| | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, Montelabbate, Italy
| | - Giamaica Conti
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
| | - Michele Riccio
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, Montelabbate, Italy
- Department of General and Specialties Surgery, SOD of Reconstructive Surgery and Hand Surgery, Azienda Ospedaliera Universitaria delle Marche, Ancona, Italy
| | - Andrea Sbarbati
- Human Anatomy and Histology Section, Department of Neuroscience, Biomedicine, and Movement, University of Verona, Verona, Italy
- Research and Training Center in Regenerative Surgery, Accademia del Lipofilling, Montelabbate, Italy
| | - Francesco De Francesco
- Department of General and Specialties Surgery, SOD of Reconstructive Surgery and Hand Surgery, Azienda Ospedaliera Universitaria delle Marche, Ancona, Italy
| |
Collapse
|
7
|
Leng Z, Li L, Zhou X, Dong G, Chen S, Shang G, Kou H, Yang B, Liu H. Novel Insights into the Stemness and Immune Privilege of Mesenchymal Stem Cells from Human Wharton Jelly by Single-Cell RNA Sequencing. Med Sci Monit 2022; 28:e934660. [PMID: 35153292 PMCID: PMC8855628 DOI: 10.12659/msm.934660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/24/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Fundamental and clinical interest in mesenchymal stem cells (MSCs) has risen dramatically over the past 3 decades. The immunomodulatory and differentiation abilities are the main mechanisms in vitro and in vivo. However, increasing evidence casts doubt on the stemness and immunogenicity of MSCs. MATERIAL AND METHODS We conducted a high-throughput 10x RNA sequencing and Smart-seq2 scRNA-seq analysis to reveal gene expression of Wharton jelly MSCs (WJ-MSCs) at a single-cell level. Multipotent differentiation, subpopulations, marker genes, human leucocyte antigen (HLA) gene expression, and cell cluster trajectory analysis were evaluated. RESULTS The WJ-MSCs had considerable heterogeneity between cells in terms of gene expression. They highly, partially, and hardly expressed genes related to mesodermal differentiation, endodermal differentiation, and ectodermal differentiation, respectively. Some cells seem to be bipotent or unipotent stem cells. Further, Monocle and cell cluster trajectory analysis demonstrated that 1 of the 3 divided clusters performed as stem cells, accounting for 12.6% of the population. The marker genes for a stem cell cluster were CRIM1, GLS, PLOD2, NEXN, ACTR2, FN1, MBNL1, LMOD1, COL3A1, NCL, SEC62, EPRS, COL5A2, COL8A1, and VCAN. In addition, the MSCs also highly, partially, and hardly expressed HLA-I antigen genes, HLA-II genes, and the HLA-G gene, respectively, indicating that MSCs probably have immunogenicity. A Kyoto Encyclopedia of Genes and Genomes pathway analysis of the 3 clusters demonstrated that they were mainly connected with viral infectious diseases, cancer, and endocrine and metabolic disorders. The most expressed transcription factors were zf-C2H2, HMG/HMGY, and Homeobox. CONCLUSIONS We found that only a subpopulation of WJ-MSCs are real stem cells and WJ-MSCs probably do not have immune privilege.
Collapse
Affiliation(s)
- Zikuan Leng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Longyu Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Xiang Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China
| | - Guangyao Dong
- Department of Obstetrics, Kaifeng Maternal and Child Health Hospital, Kaifeng, Henan, PR China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Hongwei Kou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Bo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| |
Collapse
|
8
|
Zocchi ML, Facchin F, Pagani A, Bonino C, Sbarbati A, Conti G, Vindigni V, Bassetto F. New perspectives in regenerative medicine and surgery: the bioactive composite therapies (BACTs). EUROPEAN JOURNAL OF PLASTIC SURGERY 2021; 45:1-25. [PMID: 34728900 PMCID: PMC8554210 DOI: 10.1007/s00238-021-01874-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/06/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine and surgery is a rapidly expanding branch of translational research in tissue engineering, cellular and molecular biology. To date, the methods to improve cell intake, survival, and isolation need to comply with a complex and still unclear regulatory frame, becoming everyday more restrictive and often limiting the effectiveness and outcome of the therapeutic choices. Thus, the authors developed a novel 360° regenerative strategy based on the synergic action of several new components called the bioactive composite therapies (BACTs) to improve grafted cells intake, and survival in total compliance with the legal and ethical limits of the current regulatory frame. The rationale at the origin of this new technology is based on the evidence that cells need supportive substrate to survive in vitro and this observation, applying the concept of translational medicine, is true also in vivo. Bioactive composite mixtures (BACMs) are tailor-made bioactive mixtures containing several bioactive components that support cells' survival and induce a regenerative response in vivo by stimulating the recipient site to act as an in situ real bioreactor. Many different tissues have been used in the past for the isolation of cells, molecules, and growth factors, but the adipose tissue and its stromal vascular fraction (SVF) remains the most valuable, abundant, safe, and reliable source of regenerative components and particularly of adipose-derived stems cells (ADSCs). The role of plastic surgeons as the historical experts in all the most advanced techniques for harvesting, manipulating, and grafting adipose tissue is fundamental in this constant process of expansion of regenerative procedures. In this article, we analyze the main causes of cell death and the strategies for preventing it, and we present all the technical steps for preparing the main components of BACMs and the different mixing modalities to obtain the most efficient regenerative action on different clinical and pathological conditions. The second section of this work is dedicated to the logical and sequential evolution from simple bioactive composite grafts (BACGs) that distinguished our initial approach to regenerative medicine, to BACTs where many other fundamental technical steps are analyzed and integrated for supporting and enhancing the most efficient regenerative activity. Level of Evidence: Not gradable.
Collapse
Affiliation(s)
- Michele L Zocchi
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy.,Remix Institute for Regenerative Surgery, Turin, Italy
| | - Federico Facchin
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| | - Andrea Pagani
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Claudia Bonino
- Department of Rheumatology and Immune Diseases, Humanitas Gradenigo Hospital, Turin, Italy
| | - Andrea Sbarbati
- Institute of Human Anatomy, University of Verona, Verona, Italy
| | - Giamaica Conti
- Institute of Human Anatomy, University of Verona, Verona, Italy
| | - Vincenzo Vindigni
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| | - Franco Bassetto
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| |
Collapse
|
9
|
Chen X, Yin XY, Zhao YY, Wang CC, Du P, Lu YC, Jin HB, Yang CC, Ying JL. Human Muse cells-derived neural precursor cells as the novel seed cells for the repair of spinal cord injury. Biochem Biophys Res Commun 2021; 568:103-109. [PMID: 34214874 DOI: 10.1016/j.bbrc.2021.06.070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022]
Abstract
At present, stem cell transplantation has a significant therapeutic effect on spinal cord injury (SCI), however, it is still challenging for the seed cells selection. In this study, in order to explore cells with wide neural repair potentials, we selected the pluripotent stem cells multilineage-differentiating stress-enduring (Muse) cells, inducing the in vitro differentiation of human Muse cells into neural precursor cells (Muse-NPCs) by applying neural induction medium. Here, we found induced Muse-NPCs expressed neural stem cell markers Nestin and NCAM, capable of differentiating into three types of neural cells (neuron, astrocyte and oligodendrocyte), and have certain biological functions. When Muse-NPCs were transplanted into rats suffering from T10 SCI, motor function was improved. These results provide an insight for application of Muse-NPCs in cell therapy or tissue engineering for the repair of SCI in future.
Collapse
Affiliation(s)
- Xue Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| | - Xin-Yao Yin
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ya-Yu Zhao
- Jiangsu Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chen-Chun Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Pan Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yi-Chi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Hong-Bo Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Cheng-Cheng Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Jia-Lu Ying
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
10
|
Ong WK, Chakraborty S, Sugii S. Adipose Tissue: Understanding the Heterogeneity of Stem Cells for Regenerative Medicine. Biomolecules 2021; 11:biom11070918. [PMID: 34206204 PMCID: PMC8301750 DOI: 10.3390/biom11070918] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been increasingly used as a versatile source of mesenchymal stem cells (MSCs) for diverse clinical investigations. However, their applications often become complicated due to heterogeneity arising from various factors. Cellular heterogeneity can occur due to: (i) nomenclature and criteria for definition; (ii) adipose tissue depots (e.g., subcutaneous fat, visceral fat) from which ASCs are isolated; (iii) donor and inter-subject variation (age, body mass index, gender, and disease state); (iv) species difference; and (v) study design (in vivo versus in vitro) and tools used (e.g., antibody isolation and culture conditions). There are also actual differences in resident cell types that exhibit ASC/MSC characteristics. Multilineage-differentiating stress-enduring (Muse) cells and dedifferentiated fat (DFAT) cells have been reported as an alternative or derivative source of ASCs for application in regenerative medicine. In this review, we discuss these factors that contribute to the heterogeneity of human ASCs in detail, and what should be taken into consideration for overcoming challenges associated with such heterogeneity in the clinical use of ASCs. Attempts to understand, define, and standardize cellular heterogeneity are important in supporting therapeutic strategies and regulatory considerations for the use of ASCs.
Collapse
Affiliation(s)
- Wee Kiat Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Selangor, Malaysia
- Correspondence: (W.K.O.); (S.S.)
| | - Smarajit Chakraborty
- Institute of Bioengineering and Bioimaging (IBB), A*STAR, 31 Biopolis Way, Singapore 138669, Singapore;
| | - Shigeki Sugii
- Institute of Bioengineering and Bioimaging (IBB), A*STAR, 31 Biopolis Way, Singapore 138669, Singapore;
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Correspondence: (W.K.O.); (S.S.)
| |
Collapse
|
11
|
Saeedi M, Nezhad MS, Mehranfar F, Golpour M, Esakandari MA, Rashmeie Z, Ghorbani M, Nasimi F, Hoseinian SN. Biological Aspects and Clinical Applications of Mesenchymal Stem Cells: Key Features You Need to be Aware of. Curr Pharm Biotechnol 2021; 22:200-215. [PMID: 32895040 DOI: 10.2174/1389201021666200907121530] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 11/22/2022]
Abstract
Mesenchymal Stem Cells (MSCs), a form of adult stem cells, are known to have a selfrenewing property and the potential to specialize into a multitude of cells and tissues such as adipocytes, cartilage cells, and fibroblasts. MSCs can migrate and home to the desired target zone where inflammation is present. The unique characteristics of MSCs in repairing, differentiation, regeneration, and the high capacity of immune modulation have attracted tremendous attention for exerting them in clinical purposes, as they contribute to the tissue regeneration process and anti-tumor activity. The MSCs-based treatment has demonstrated remarkable applicability towards various diseases such as heart and bone malignancies, and cancer cells. Importantly, genetically engineered MSCs, as a stateof- the-art therapeutic approach, could address some clinical hurdles by systemic secretion of cytokines and other agents with a short half-life and high toxicity. Therefore, understanding the biological aspects and the characteristics of MSCs is an imperative issue of concern. Herein, we provide an overview of the therapeutic application and the biological features of MSCs against different inflammatory diseases and cancer cells. We further shed light on MSCs' physiological interaction, such as migration, homing, and tissue repairing mechanisms in different healthy and inflamed tissues.
Collapse
Affiliation(s)
- Mohammad Saeedi
- Department of Laboratory Science, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Muhammad S Nezhad
- Stem Cells and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mehranfar
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahdieh Golpour
- School of Paramedical Sciences, Semnan University of Medical Sciences, Sorkheh, Semnan, Iran
| | - Mohammad A Esakandari
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Rashmeie
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Ghorbani
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nasimi
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed N Hoseinian
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
12
|
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
|
13
|
Generation of Retinal Pigmented Epithelium-Like Cells from Pigmented Spheres Differentiated from Bone Marrow Stromal Cell-Derived Neurospheres. Tissue Eng Regen Med 2019; 16:253-263. [PMID: 31205854 DOI: 10.1007/s13770-019-00183-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 01/31/2023] Open
Abstract
Background Retinal degeneration causes blindness, and cell replacement is a potential therapy. The purpose of this study is to formation of pigmented neurospheres in a simple medium, low-cost, high-performance manner over a short period of time while expressing markers of RPE cells and the activation of specific genes of the pigment cells. Also, these neurospheres have the ability to produce a monolayer of retinal pigment epithelium-like cells (RPELC) with the ability of photoreceptor outer segment phagocytosis. Methods BMSC were isolated from pigmented hooded male rats and were immunoreactive to BMSC markers, then converted into neurospheres, differentiated into pigmented spheres (PS), and characterized using Retinal pigment epithelium-specific 65 kDa protein (RPE65), Retinaldehyde-binding protein 1 (CRALBP) and orthodenticle homeobox 2 (OTX2) markers by immunocytochemistry, RT-PCR and RT-qPCR. The PS were harvested into RPELC. The functionality of RPELC was evaluated by phagocytosis of fluorescein-labeled photoreceptor outer segment. Results The BMSC immunophenotype was confirmed by immunostained for fibronectin, CD90, CD166 and CD44. These cells differentiated into osteogenic and lipogenic cells. The generated neurospheres were immunoreactive to nestin and stemness genes. The PS after 7-14 days were positive for RPE65 (92.76-100%), CRALBP (95.21-100%) and OTX2 (94.88-100%), and after 30 days RT-PCR, qPCR revealed increasing in gene expression. The PS formed a single layer of RPELC after cultivation and phagocyte photoreceptor outer segments. Conclusion Bone marrow stromal stem cells can differentiate into functional retinal pigmented epithelium cells in a simple, low-cost, high-performance manner over a short period of time. These cells due to expressing the RPELC genes and markers can be used in cell replacement therapy for degenerative diseases including age-related macular degeneration as well as retinitis pigmentosa.
Collapse
|
14
|
Hill ABT, Bressan FF, Murphy BD, Garcia JM. Applications of mesenchymal stem cell technology in bovine species. Stem Cell Res Ther 2019; 10:44. [PMID: 30678726 PMCID: PMC6345009 DOI: 10.1186/s13287-019-1145-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have received a great deal of attention over the past 20 years mainly because of the results that showed regeneration potential and plasticity that were much stronger than expected in prior decades. Recent findings in this field have contributed to progress in the establishment of cell differentiation methods, which have made stem cell therapy more clinically attractive. In addition, MSCs are easy to isolate and have anti-inflammatory and angiogenic capabilities. The use of stem cell therapy is currently supported by scientific literature in the treatment of several animal health conditions. MSC may be administered for autologous or allogenic therapy following either a fresh isolation or a thawing of a previously frozen culture. Despite the fact that MSCs have been widely used for the treatment of companion and sport animals, little is known about their clinical and biotechnological potential in the economically relevant livestock industry. This review focuses on describing the key characteristics of potential applications of MSC therapy in livestock production and explores the themes such as the concept, culture, and characterization of mesenchymal stem cells; bovine mesenchymal stem cell isolation; applications and perspectives on commercial interests and farm relevance of MSC in bovine species; and applications in translational research.
Collapse
Affiliation(s)
- Amanda Baracho Trindade Hill
- Department of Preventive Veterinary Medicine and Animal Reproduction, São Paulo State University, Via de Acesso Professor Paulo Donato Castelane - Vila Industrial, s/n, Jaboticabal, SP, 14884-900, Brazil. .,Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint Hyacinthe, QC, J2S 7C6, Canada.
| | - Fabiana Fernandes Bressan
- Campus Fernando Costa, University of São Paulo, Av. Duque de Caxias Norte, 225 - Zona Rural, Pirassununga, SP, 13635-900, Brazil
| | - Bruce D Murphy
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint Hyacinthe, QC, J2S 7C6, Canada
| | - Joaquim Mansano Garcia
- Department of Preventive Veterinary Medicine and Animal Reproduction, São Paulo State University, Via de Acesso Professor Paulo Donato Castelane - Vila Industrial, s/n, Jaboticabal, SP, 14884-900, Brazil
| |
Collapse
|
15
|
Wysoczynski M, Khan A, Bolli R. New Paradigms in Cell Therapy: Repeated Dosing, Intravenous Delivery, Immunomodulatory Actions, and New Cell Types. Circ Res 2018; 123:138-158. [PMID: 29976684 PMCID: PMC6050028 DOI: 10.1161/circresaha.118.313251] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Perhaps the most important advance in the field of cell therapy for heart disease has been the recognition that all stem/progenitor cells (both adult and embryonic) fail to engraft in the heart to a significant extent and thus work via paracrine mechanisms. This fundamental advance has led to 4 new paradigms that are discussed in this review and that may importantly shape, or even revolutionize, the future of the field: (1) repeated cell therapy, (2) intravenous cell therapy, (3) immunomodulatory actions of cell therapy, and (4) new cell types. Because virtually all of our current knowledge of cell therapy is predicated on the effects of a single cell dose, the idea that the full therapeutic effects of a cell product require repeated doses is disruptive and has far-reaching implications. For example, inadequate dosing (single-dose protocols) may be responsible, at least in part, for the borderline or disappointing results obtained to date in clinical trials; furthermore, future studies (both preclinical and clinical) may need to incorporate repeated cell administrations. Another disruptive idea, supported by emerging preclinical and clinical evidence, is that intravenously injected cells can produce beneficial effects on the heart, presumably via release of paracrine factors in extracardiac organs or endocrine factors into the systemic circulation. Intravenous administration would obviate the need for direct delivery of cells to the heart, making cell therapy simpler, cheaper, safer, more scalable, and more broadly available, even on an outpatient basis. Although the mechanism of action of cell therapy remains elusive, there is compelling in vitro evidence that transplanted cells modulate the function of various immune cell types via release of paracrine factors, such as extracellular vesicles, although in vivo evidence is still limited. Investigation of the new paradigms reviewed herein should be a top priority because it may profoundly transform cell therapy and finally make it a reality.
Collapse
Affiliation(s)
- Marcin Wysoczynski
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Abdur Khan
- From the Institute of Molecular Cardiology, University of Louisville, KY
| | - Roberto Bolli
- From the Institute of Molecular Cardiology, University of Louisville, KY.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Nakabayashi H, Kawahara M, Nagamune T. Cell-Surface Expression Levels Are Important for Fine-Tuning the Performance of Receptor Tyrosine Kinase-Based Signalobodies. Biotechnol J 2017; 12. [PMID: 28881109 DOI: 10.1002/biot.201700441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 08/18/2017] [Indexed: 11/07/2022]
Abstract
As receptor tyrosine kinases (RTKs) play important roles in cell-fate control of various cell types, engineered RTKs that could respond to inexpensive ligands might drastically reduce the cost of producing desired cells for various applications in regenerative medicine. We developed several engineered RTKs named "signalobodies" in which the ligand-recognition domain of RTKs is replaced by single-chain Fv for enabling recognition of a specific antigen. However, the remaining concern was the dysregulation of antigen-dependent on/off signaling of the signalobodies. This study aims at fine-tuning the performance of the signalobodies based on three RTKs (fibroblast growth factor receptor 1, insulin receptor, and c-fms). To this end, the cell-surface expression levels of the RTK-based signalobodies were altered by locating their genes either upstream or downstream of the internal ribosomal entry site, and by inserting 1 to 3 alanine residue(s) at the intracellular juxtamembrane region. As a result, while the signaling response was different among the three signalobodies, the antigen-dependent on/off regulation became tighter when the cell-surface expression levels of the signalobodies were lowered. Therefore, we successfully developed a method to diminish the leaky signaling of RTK-based signalobodies, which will be important for establishing the signalobody-based platform technology that can produce cells of interest for regenerative medicine.
Collapse
Affiliation(s)
- Hideto Nakabayashi
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Masahiro Kawahara
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Teruyuki Nagamune
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
19
|
Loesch A, Dashwood MR. Nerve-perivascular fat communication as a potential influence on the performance of blood vessels used as coronary artery bypass grafts. J Cell Commun Signal 2017; 12:181-191. [PMID: 28601937 PMCID: PMC5842173 DOI: 10.1007/s12079-017-0393-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/14/2022] Open
Abstract
Perivascular fat, the cushion of adipose tissue surrounding blood vessels, possesses dilator, anti-contractile and constrictor actions. The majority of these effects have been demonstrated in vitro and may depend on the vessel and/or the experimental method or species used. In general, the relaxant effect of perivascular adipose tissue is local and may be either endothelium-dependent or endothelium-independent. However, nerve stimulation studies show that, in general, perivascular adipose tissue (PVAT) has an anti-contractile vascular effect likely to involve an action of the autonomic vascular nerves. Apart from a direct effect of perivascular fat-derived factors on bypass conduits, an interaction with a number of neurotransmitters and other agents may play an important role in graft performance. Although the vascular effects of PVAT are now well-established there is a lack of information regarding the role and/or involvement of peripheral nerves including autonomic nerves. For example, are perivascular adipocytes innervated and does PVAT affect neuronal control of vessels used as grafts? To date there is a paucity of electrophysiological studies into nerve-perivascular fat control. This review provides an overview of the vascular actions of PVAT, focussing on its potential relevance on blood vessels used as bypass grafts. In particular, the anatomical relationship between the perivascular nerves and fat are considered and the role of the perivascular-nerve/fat axis in the performance of bypass grafts is also discussed.
Collapse
Affiliation(s)
- Andrzej Loesch
- Centre for Rheumatology and Connective Tissue Diseases, Division of Medicine, University College London Medical School, Royal Free Campus, Rowland Hill Street, NW3 2PF, London, UK.
| | - Michael R Dashwood
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London Medical School, Royal Free Campus, Rowland Hill Street, NW3 2PF, London, UK
| |
Collapse
|
20
|
Impact of early subcultures on stemness, migration and angiogenic potential of adipose tissue-derived stem cells and their resistance to in vitro ischemic condition. Cytotechnology 2017; 69:885-900. [PMID: 28536871 DOI: 10.1007/s10616-017-0104-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 04/28/2017] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) are capable of multipotential differentiation and express several angiogenic, anti-apoptotic and immunomodulatory markers. These features make adipose tissue as a promising source of stem cells for regenerative medicine. However, for efficient translational use, culture-induced changes in the gene expression profile and resistance of the ADSCs to ischemic environment should be taken into consideration. We compared the expression of some clinically important markers between the unpassaged and third-passaged ADSCs by RT-PCR, qPCR and flow cytometry. Our results demonstrated that the embryonic stem cell (ESC)-specific markers were expressed in the unpassaged ADSCs but were downregulated after three passages. The expression of stemness-related genes, TGFB and FGF2, was upregulated while FGF4 and LIF were downregulated after three passages. The expression of angiogenic genes in the third-passaged ADSCs was higher than the unpassaged cells. Epithelial-mesenchymal transition (EMT) markers were either expressed in the third-passaged ADSCs or significantly upregulated after three passages. In contrast, cell cycle inhibitors, CDKN1A and TP53, were downregulated with early subcultures. The unpassaged and third-passaged ADSCs showed nearly similar resistance to oxidative stress, hypoxia and serum deprivation. In conclusion, the primary cultures of human adipose tissue contain a subpopulation of cells expressing ESC-specific genes and proteins, but the expression of these pluripotency markers subsides rapidly in standard mesenchymal stem cell culture medium. The expression of angiogenic and EMT markers also varies with early subcultures. Altogether, early-passaged ADSCs may be better choices for transplantation therapy of injured tissues, especially after ischemic conditions.
Collapse
|
21
|
García-Domínguez X, Vicente JS, Vera-Donoso CD, Marco-Jimenez F. Current Bioengineering and Regenerative Strategies for the Generation of Kidney Grafts on Demand. Curr Urol Rep 2017; 18:2. [PMID: 28092070 DOI: 10.1007/s11934-017-0650-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Currently in the USA, one name is added to the organ transplant waiting list every 15 min. As this list grows rapidly, fewer than one-third of waiting patients can receive matched organs from donors. Unfortunately, many patients who require a transplant have to wait for long periods of time, and many of them die before receiving the desired organ. In the USA alone, over 100,000 patients are waiting for a kidney transplant. However, it is a problem that affects around 6% of the word population. Therefore, seeking alternative solutions to this problem is an urgent work. Here, we review the current promising regenerative technologies for kidney function replacement. Despite many approaches being applied in the different ways outlined in this work, obtaining an organ capable of performing complex functions such as osmoregulation, excretion or hormone synthesis is still a long-term goal. However, in the future, the efforts in these areas may eliminate the long waiting list for kidney transplants, providing a definitive solution for patients with end-stage renal disease.
Collapse
Affiliation(s)
- Ximo García-Domínguez
- Instituto de Ciencia y Tecnología Animal, Universidad Politécnica de Valencia, C/Camino de Vera s/n, 46022, Valencia, Spain
| | - Jose S Vicente
- Instituto de Ciencia y Tecnología Animal, Universidad Politécnica de Valencia, C/Camino de Vera s/n, 46022, Valencia, Spain
| | - Cesar D Vera-Donoso
- Servicio de Urología, Hospital Universitari i Politècnic La Fe, Avinguda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Francisco Marco-Jimenez
- Instituto de Ciencia y Tecnología Animal, Universidad Politécnica de Valencia, C/Camino de Vera s/n, 46022, Valencia, Spain.
| |
Collapse
|
22
|
Tian T, Zhang RZ, Yang YH, Liu Q, Li D, Pan XR. Muse Cells Derived from Dermal Tissues Can Differentiate into Melanocytes. Cell Reprogram 2017; 19:116-122. [PMID: 28170296 DOI: 10.1089/cell.2016.0032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The objective of the authors has been to obtain multilineage-differentiating stress-enduring cells (Muse cells) from primary cultures of dermal fibroblasts, identify their pluripotency, and detect their ability to differentiate into melanocytes. The distribution of SSEA-3-positive cells in human scalp skin was assessed by immunohistochemistry, and the distribution of Oct4, Sox2, Nanog, and SSEA-3-positive cells was determined by immunofluorescence staining. The expression levels of Sox2, Oct4, hKlf4, and Nanog mRNAs and proteins in Muse cells were determined by reverse transcription polymerase chain reaction (RT-PCR) analyses and Western blots, respectively. These Muse cells differentiated into melanocytes in differentiation medium. The SSEA-3-positive cells were scattered in the basement membrane zone and the dermis, with comparatively more in the sebaceous glands, vascular and sweat glands, as well as the outer root sheath of hair follicles, the dermal papillae, and the hair bulbs. Muse cells, which have the ability to self-renew, were obtained from scalp dermal fibroblasts by flow cytometry sorting with an anti-SSEA-3 antibody. The results of RT-PCR, Western blot, and immunofluorescence staining showed that the expression levels of Oct4, Nanog, Sox2, and Klf4 mRNAs and proteins in Muse cells were significantly different from their parental dermal fibroblasts. Muse cells differentiated into melanocytes when cultured in melanocyte differentiation medium, and the Muse cell-derived melanocytes expressed the melanocyte-specific marker HMB45. Muse cells could be obtained by flow cytometry from primary cultures of scalp dermal fibroblasts, which possessed the ability of pluripotency and self-renewal, and could differentiate into melanocytes in vitro.
Collapse
Affiliation(s)
- Ting Tian
- 1 The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University , Changzhou, China
| | - Ru-Zhi Zhang
- 1 The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University , Changzhou, China
| | - Yu-Hua Yang
- 1 The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University , Changzhou, China
| | - Qi Liu
- 1 The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University , Changzhou, China
| | - Di Li
- 1 The Dermal and Venereal Department, The Third Affiliated Hospital of Suzhou University , Changzhou, China
| | - Xiao-Ru Pan
- 2 The Dermal and Venereal Department, Bengbu Medical College , Bengbu, China
| |
Collapse
|
23
|
Liu Q, Zhang RZ, Li D, Cheng S, Yang YH, Tian T, Pan XR. Muse Cells, a New Type of Pluripotent Stem Cell Derived from Human Fibroblasts. Cell Reprogram 2016; 18:67-77. [PMID: 27055628 DOI: 10.1089/cell.2015.0085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A new type of mesenchymal stem cells (MSCs) that expresses stage-specific embryonic antigen 3 (SSEA-3) and the mesenchymal cell marker CD105 are known as multilineage-differentiating stress-enduring (Muse) cells. Studies have shown that stem cells in suspension cultures are more likely to generate embryoid body-like stem cell spheres and maintain an undifferentiated phenotype and pluripotency. We separated Muse cells derived from human dermal fibroblasts by long-term trypsin incubation (LTT) through suspension cultures in methylcellulose. The Muse cells obtained expressed several pluripotency markers, including Nanog, Oct4, Sox2, and SSEA-3, and could differentiate in vitro into cells of the three germ layers, such as hepatocytes (endodermal), neural cells (ectodermal) and adipocytes, and osteocytes (mesodermal cells). These cells showed a low level of DNA methylation and a high nucleo-cytoplasmic ratio. Our study provides an innovative and exciting platform for exploring the potential cell-based therapy of various human diseases using Muse cells as well as their great possibility for regenerative medicine.
Collapse
Affiliation(s)
- Qi Liu
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Ru-zhi Zhang
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Di Li
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Sai Cheng
- 2 Department of Dermatology, The First Affiliated Hospital of Bengbu Medical College , Anhui, 213003, China
| | - Yu-hua Yang
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Ting Tian
- 1 Department of Dermatology, The Third Affiliated Hospital of Suzhou University , Changzhou, 213003, China
| | - Xiao-ru Pan
- 2 Department of Dermatology, The First Affiliated Hospital of Bengbu Medical College , Anhui, 213003, China
| |
Collapse
|
24
|
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
|
25
|
Dezawa M. Muse Cells Provide the Pluripotency of Mesenchymal Stem Cells: Direct Contribution of Muse Cells to Tissue Regeneration. Cell Transplant 2016; 25:849-61. [PMID: 26884346 DOI: 10.3727/096368916x690881] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While mesenchymal stem cells (MSCs) are easily accessible from mesenchymal tissues, such as bone marrow and adipose tissue, they are heterogeneous, and their entire composition is not fully identified. MSCs are not only able to differentiate into osteocytes, chondrocytes, and adipocytes, which belong to the same mesodermal lineage, but they are also able to cross boundaries between mesodermal, ectodermal, and endodermal lineages, and differentiate into neuronal- and hepatocyte-like cells. However, the ratio of such differentiation is not very high, suggesting that only a subpopulation of the MSCs participates in this cross-lineage differentiation phenomenon. We have identified unique cells that we named multilineage-differentiating stress-enduring (Muse) cells that may explain the pluripotent-like properties of MSCs. Muse cells comprise a small percentage of MSCs, are able to generate cells representative of all three germ layers from a single cell, and are nontumorigenic and self-renewable. Importantly, cells other than Muse cells in MSCs do not have these pluripotent-like properties. Muse cells are particularly unique compared with other stem cells in that they efficiently migrate and integrate into damaged tissue when supplied into the bloodstream, and spontaneously differentiate into cells compatible with the homing tissue. Such a repairing action of Muse cells via intravenous injection is recognized in various tissues including the brain, liver, and skin. Therefore, unlike ESCs/iPSCs, Muse cells render induction into the target cell type prior to transplantation unnecessary. They can repair tissues in two simple steps: collection from mesenchymal tissues, such as the bone marrow, and intravenous injection. The impressive regenerative performance of these cells provides a simple, feasible strategy for treating a variety of diseases. This review details the unique characteristics of Muse cells and describes their future application for regenerative medicine.
Collapse
Affiliation(s)
- Mari Dezawa
- Department of Stem Cell Biology and Histology and Department of Anatomy and Anthropology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
26
|
Togliatto G, Dentelli P, Gili M, Gallo S, Deregibus C, Biglieri E, Iavello A, Santini E, Rossi C, Solini A, Camussi G, Brizzi MF. Obesity reduces the pro-angiogenic potential of adipose tissue stem cell-derived extracellular vesicles (EVs) by impairing miR-126 content: impact on clinical applications. Int J Obes (Lond) 2015; 40:102-11. [PMID: 26122028 PMCID: PMC4722244 DOI: 10.1038/ijo.2015.123] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 05/18/2015] [Accepted: 06/22/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES Soluble factors and cell-derived extracellular vesicles (EVs) are crucial tissue repair mediators in cell-based therapy. In the present study, we investigate the therapeutic impact of EVs released by adipose tissue-derived stem cells (ASCs) recovered from obese subjects' visceral and subcutaneous tissues. METHODS ASCs were recovered from 10 obese (oASCs) and 6 non-obese (nASCs) participants and characterized. In selected experiments, nASCs and oASCs were cultured with palmitic acid (PA) or high glucose (HG), respectively. EVs from obese (oEVs) and non-obese (nEVs) subjects' visceral and subcutaneous ASCs were collected after ultracentrifugation and analyzed for their cargo: microRNA-126 (miR-126), vascular endothelial growth factor (VEGF), and matrix metalloproteinase 2 (MMP-2), and for their biological effects on endothelial cells (ECs). Western blotting analysis and loss- and gain-of function experiments were performed. RESULTS oEVs show impaired angiogenic potential compared with nEVs. This effect depends on EV cargo: reduced content of VEGF, MMP-2 and, more importantly, miR-126. We demonstrate, using gain- and loss-of-function experiments, that this reduced miR-126 content leads to Spred1 upregulation and the inhibition of the extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway in ECs. We also show that PA treatment of nASCs translates into the release of EVs that recapitulate oEV cargo. Moreover, HG treatment of oASCs further reduces miR-126 EV content and EV-mediated in vitro angiogenesis. Finally, impaired pro-angiogenic potential is also detected in EVs released from obese subcutaneous adipose tissue-derived ASCs. CONCLUSIONS These results indicate that obesity impacts on EV pro-angiogenic potential and may raise concerns about the use of adipose tissue-derived EVs in cell-based therapy in the obese setting.
Collapse
Affiliation(s)
- G Togliatto
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - P Dentelli
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - M Gili
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - S Gallo
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - C Deregibus
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - E Biglieri
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - A Iavello
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - E Santini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Solini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - G Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - M F Brizzi
- Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
27
|
Current Bioengineering Methods for Whole Kidney Regeneration. Stem Cells Int 2015; 2015:724047. [PMID: 26089921 PMCID: PMC4452081 DOI: 10.1155/2015/724047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 02/07/2023] Open
Abstract
Kidney regeneration is likely to provide an inexhaustible source of tissues and organs for immunosuppression-free transplantation. It is currently garnering considerable attention and might replace kidney dialysis as the ultimate therapeutic strategy for renal failure. However, anatomical complications make kidney regeneration difficult. Here, we review recent advances in the field of kidney regeneration, including (i) the directed differentiation of induced pluripotent stem cells/embryonic stem cells into kidney cells; (ii) blastocyst decomplementation; (iii) use of a decellularized cadaveric scaffold; (iv) embryonic organ transplantation; and (v) use of a nephrogenic niche for growing xenoembryos for de novo kidney regeneration from stem cells. All these approaches represent potentially promising therapeutic strategies for the treatment of patients with chronic kidney disease. Although many obstacles to kidney regeneration remain, we hope that innovative strategies and reliable research will ultimately allow the restoration of renal function in patients with end-stage kidney disease.
Collapse
|
28
|
Díaz-Flores L, Gutiérrez R, Lizartza K, Goméz MG, García MDP, Sáez FJ, Díaz-Flores L, Madrid JF. Behavior of in situ human native adipose tissue CD34+ stromal/progenitor cells during different stages of repair. Tissue-resident CD34+ stromal cells as a source of myofibroblasts. Anat Rec (Hoboken) 2014; 298:917-30. [PMID: 25387858 DOI: 10.1002/ar.23086] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/12/2014] [Indexed: 12/22/2022]
Abstract
CD34+ adipose stromal cells are scattered in the adipose tissue and found in the CD34+ population of the stromal vascular fraction (SVF). This fraction includes adipose-derived stromal/stem/progenitor cells (ASCs), which have attracted considerable attention and show great promise for the future of regenerative medicine. Studies in this field have been undertaken mainly in vitro. In this work, however, we assessed the characteristics of human adipose tissue-resident CD34+ stromal cells in normal conditions and when activated in vivo during inflammatory/repair processes at different stages of evolution. In normal adipose tissue, these cells showed a characteristic location (peri/paravascular and between adipocytes), a fusiform or stellate morphology, long and moniliform processes, and scarce organelles. During inflammatory/repair stages, native CD34+ stromal cells increased in size, proliferated, developed numerous organelles of synthesis, lost CD34 expression, and differentiated into myofibroblasts (αSMA expression and typical ultrastructure). In double-stained sections, cells expressing both CD34 and αSMA were observed. CD34 expression correlated positively with a high proliferative capacity (Ki-67 expression). Conversely, CD34 expression was lost with successive mitoses and with increased numbers of macrophages in the granulation tissue. CD34+ stromal cell behavior varied depending on proximity to (with myofibroblast differentiation) or remoteness from (with activated plump cells conserving CD34 expression) injury. In conclusion, our observations point to human adipose tissue-resident CD34+ stromal cells as an important source of myofibroblasts during inflammatory/repair processes. Moreover, stromal cell activation may occur with or without αSMA expression (with or without myofibroblast transformation) and with loss or persistence of CD34 expression, respectively.
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Anatomy, Histology, Pathology and Radiology, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | | | | | | | | | | | | | | |
Collapse
|