1
|
Sakamoto T, Fuku A, Horie T, Kitajima H, Nakamura Y, Tanida I, Sunami H, Hirata H, Tachi Y, Iida Y, Yamada S, Yamamoto N, Shimizu Y, Ishigaki Y, Ichiseki T, Kaneuji A, Osawa S, Kawahara N. A novel cell source for therapy of knee osteoarthritis using atelocollagen microsphere-adhered adipose-derived stem cells: Impact of synovial fluid exposure on cell activity. Regen Ther 2024; 27:408-418. [PMID: 38694445 PMCID: PMC11061654 DOI: 10.1016/j.reth.2024.04.010] [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: 02/01/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Administration of adipose-derived stem cells (ADSCs) into the joint cavity has been shown to alleviate the symptoms of knee osteoarthritis (OA) by releasing exosomes and anti-inflammatory cytokines. However, the therapeutic effect of these cells is limited by their rapid disappearance after administration. Thus, it is necessary to prolong cell survival in the joint cavity. This study aimed to investigate the potential application of ADSCs adhered to atelocollagen microspheres (AMSs) for cell therapy of knee OA. Methods ADSCs were cultured for 2, 4, and 7 days in AMS suspension or adherent culture dishes. The supernatants were analyzed for IL-10 and exosome secretion via enzyme-linked immunosorbent assay and Nanosight. The effect of AMS was compared with that of adherent-cultured ADSCs (2D-cultured ADSCs) using transcriptome analysis. Moreover, the solubility of AMS and viability of ADSCs were evaluated using synovial fluid (SF) from patients with knee OA. Results Compared with 2D-cultured ADSCs, AMS-cultured ADSCs exhibited a significant increase in secretion of exosomes and IL-10, and the expression of several genes involved in extracellular matrix and immune regulation were altered. Furthermore, when AMS-cultured ADSCs were cultured in SF from knee OA patients to mimic the intra-articular environment, the SF dissolved the AMSs and released viable ADSCs. In addition, AMS-cultured ADSCs showed significantly higher long-term cell viability than 2D-cultured ADSCs. Conclusion Increased survival of AMS-adhered ADSCs was observed in the intra-articular environment, and AMSs were found to gradually dissipate. These results suggest that AMS-adhered ADSCs are promising source for cell therapy of knee OA.
Collapse
Affiliation(s)
- Takuya Sakamoto
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Atsushi Fuku
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Tetsuhiro Horie
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Hironori Kitajima
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Ikuhiro Tanida
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Hakusan, 924-0838, Ishikawa, Japan
| | - Hiroshi Sunami
- Faculty of Medicine, Advanced Medical Research Center, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroaki Hirata
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yoshiyuki Tachi
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Yasuo Iida
- Department of Mathematics, Division of General Education, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Sohsuke Yamada
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pathology, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Naoki Yamamoto
- Support Office for Bioresource Research, Center for Translational Research, Translational Research Headquarters, Fujita Health University, Toyoake, 470-1192, Aichi, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nakagami, 903-0215, Okinawa, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Toru Ichiseki
- Medical Research Institute, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Ayumi Kaneuji
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Satoshi Osawa
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Hakusan, 924-0838, Ishikawa, Japan
| | - Norio Kawahara
- Department of Orthopedic Surgery, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| |
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
|
He H, Yuan Y, Wu Y, Lu J, Yang X, Lu K, Liu A, Cao Z, Sun M, Yu M, Wang H. Exoskeleton Partial-Coated Stem Cells for Infarcted Myocardium Restoring. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2307169. [PMID: 37962473 DOI: 10.1002/adma.202307169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/30/2023] [Indexed: 11/15/2023]
Abstract
The integration of abiotic materials with live cells has emerged as an exciting strategy for the control of cellular functions. Exoskeletons consisting ofmetal-organic frameworks are generated to produce partial-coated bone marrow stem cells (BMSCs) to overcome low cell survival leading to disappointing effects for cell-based cardiac therapy. Partially coated exoskeletons can promote the survival of suspended BMSCs by integrating the support of exoskeletons and unimpaired cellular properties. In addition, partial exoskeletons exhibit protective effects against detrimental environmental conditions, including reactive oxygen species, pH changes, and osmotic pressure. The partial-coated cells exhibit increased intercellular adhesion forces to aggregate and adhere, promoting cell survival and preventing cell escape during cell therapy. The exoskeletons interact with cell surface receptors integrin α5β1, leading to augmented biological functions with profitable gene expression alteration, such as Vegfa, Cxcl12, and Adm. The partial-coated BMSCs display enhanced cell retention in infarcted myocardium through non-invasive intravenous injections. The repair of myocardial infarction has been achieved with improved cardiac function, myocardial angiogenesis, proliferation, and inhibition of cell apoptosis. This discovery advances the elucidation of potential molecular and cellular mechanisms for cell-exoskeleton interactions and benefits the rational design and manufacture of next-generation nanobiohybrids.
Collapse
Affiliation(s)
- Huihui He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yuan Yuan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, 310058, China
| | - Yunhong Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jingyi Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Xiaofu Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Kejie Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - An Liu
- Department of Orthopaedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310009, China
| | - Zelin Cao
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Miao Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
4
|
Zhuang K, Romagnuolo R, Sadikov Valdman T, Vollett KDW, Szulc DA, Cheng HYM, Laflamme MA, Cheng HLM. Bright ferritin for long-term MR imaging of human embryonic stem cells. Stem Cell Res Ther 2023; 14:330. [PMID: 37964388 PMCID: PMC10647036 DOI: 10.1186/s13287-023-03565-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND A non-invasive imaging technology that can monitor cell viability, retention, distribution, and interaction with host tissue after transplantation is needed for optimizing and translating stem cell-based therapies. Current cell imaging approaches are limited in sensitivity or specificity, or both, for in vivo cell tracking. The objective of this study was to apply a novel ferritin-based magnetic resonance imaging (MRI) platform to longitudinal tracking of human embryonic stem cells (hESCs) in vivo. METHODS Human embryonic stem cells (hESCs) were genetically modified to stably overexpress ferritin using the CRISPR-Cas9 system. Cellular toxicity associated with ferritin overexpression and manganese (Mn) supplementation were assessed based on cell viability, proliferation, and metabolic activity. Ferritin-overexpressing hESCs were characterized based on stem cell pluripotency and cardiac-lineage differentiation capability. Cells were supplemented with Mn and imaged in vitro as cell pellets on a preclinical 3 T MR scanner. T1-weighted images and T1 relaxation times were analyzed to assess contrast. For in vivo study, three million cells were injected into the leg muscle of non-obese diabetic severe combined immunodeficiency (NOD SCID) mice. Mn was administrated subcutaneously. T1-weighted sequences and T1 mapping were used to image the animals for longitudinal in vivo cell tracking. Cell survival, proliferation, and teratoma formation were non-invasively monitored by MRI. Histological analysis was used to validate MRI results. RESULTS Ferritin-overexpressing hESCs labeled with 0.1 mM MnCl2 provided significant T1-induced bright contrast on in vitro MRI, with no adverse effect on cell viability, proliferation, pluripotency, and differentiation into cardiomyocytes. Transplanted hESCs displayed significant bright contrast on MRI 24 h after Mn administration, with contrast persisting for 5 days. Bright contrast was recalled at 4-6 weeks with early teratoma outgrowth. CONCLUSIONS The bright-ferritin platform provides the first demonstration of longitudinal cell tracking with signal recall, opening a window on the massive cell death that hESCs undergo in the weeks following transplantation before the surviving cell fraction proliferates to form teratomas.
Collapse
Affiliation(s)
- Keyu Zhuang
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Room 1433, Toronto, ON, M5G 1M1, Canada
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | | | - Kyle D W Vollett
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Room 1433, Toronto, ON, M5G 1M1, Canada
| | - Daniel A Szulc
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Room 1433, Toronto, ON, M5G 1M1, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory of Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Room 1433, Toronto, ON, M5G 1M1, Canada.
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Cheng HLM. A primer on in vivo cell tracking using MRI. Front Med (Lausanne) 2023; 10:1193459. [PMID: 37324153 PMCID: PMC10264782 DOI: 10.3389/fmed.2023.1193459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Cell tracking by in vivo magnetic resonance imaging (MRI) offers a collection of multiple advantages over other imaging modalities, including high spatial resolution, unlimited depth penetration, 3D visualization, lack of ionizing radiation, and the potential for long-term cell monitoring. Three decades of innovation in both contrast agent chemistry and imaging physics have built an expansive array of probes and methods to track cells non-invasively across a diverse range of applications. In this review, we describe both established and emerging MRI cell tracking approaches and the variety of mechanisms available for contrast generation. Emphasis is given to the advantages, practical limitations, and persistent challenges of each approach, incorporating quantitative comparisons where possible. Toward the end of this review, we take a deeper dive into three key application areas - tracking cancer metastasis, immunotherapy for cancer, and stem cell regeneration - and discuss the cell tracking techniques most suitable to each.
Collapse
Affiliation(s)
- Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Translational Biology & Engineering Program, Toronto, ON, Canada
| |
Collapse
|
6
|
Study on Long-Term Tracing of Fibroblasts on Three-Dimensional Tissue Engineering Scaffolds Based on Graphene Quantum Dots. Int J Mol Sci 2022; 23:ijms231911040. [PMID: 36232342 PMCID: PMC9570154 DOI: 10.3390/ijms231911040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
In order to find a convenient and stable way to trace human skin fibroblasts (HSFs) in three-dimensional tissue engineering scaffolds for a long time, in this experiment, Graphene Oxide Quantum Dots (GOQDs), Amino Graphene Quantum Dots (AGQDs) and Carboxyl Graphene Quantum Dots (CGQDs) were used as the material source for labeling HSFs. Exploring the possibility of using it as a long-term tracer of HSFs in three-dimensional tissue engineering scaffolds, the contents of the experiment are as follows: the HSFs were cultured in a cell-culture medium composed of three kinds of Graphene Quantum Dots for 24 h, respectively; (1) using Cell Counting Kit 8 (CCK8), Transwell migration chamber and Phalloidin-iFlior 488 to detect the effect of Graphene Quantum Dots on the biocompatibility of HSFs; (2) using a living cell workstation to detect the fluorescence labeling results of three kinds of Graphene Quantum Dots on HSFs, and testing the fluorescence attenuation of HSFs for 7 days; (3) the HSFs labeled with Graphene Quantum Dots were inoculated on the three-dimensional chitosan demethylcellulose sodium scaffold, and the living cell workstation was used to detect the spatial distribution of the HSFs on the three-dimensional scaffold through the fluorescence properties of the HSFs.. Experimental results: (1) the results of CCK8, Transwell migration, and FITC-Phalloidin cytoskeleton test showed that the three kinds of Graphene Quantum Dots had no effect on the biological properties of HSFs (p < 0.05); (2) the results of the fluorescence labeling experiment showed that only AGQDs could make HSFs fluorescent, and cells showed orange−red fluorescence; (3) the results of long-range tracing of HSFs which were labeled by with AGQDs showed that the fluorescence life of the HSFs were as long as 7 days; (4) The spatial distribution of HSFs can be detected on the three-dimensional scaffold based on their fluorescence properties, and the detection time can be up to 7 days.
Collapse
|
7
|
Elhussieny A, Nogami K, Sakai-Takemura F, Maruyama Y, Takemura N, Soliman WT, Takeda S, Miyagoe-Suzuki Y. Mesenchymal stem cells derived from human induced pluripotent stem cells improve the engraftment of myogenic cells by secreting urokinase-type plasminogen activator receptor (uPAR). Stem Cell Res Ther 2021; 12:532. [PMID: 34627382 PMCID: PMC8501581 DOI: 10.1186/s13287-021-02594-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe X-linked recessive disease caused by mutations in the dystrophin gene. Transplantation of myogenic stem cells holds great promise for treating muscular dystrophies. However, poor engraftment of myogenic stem cells limits the therapeutic effects of cell therapy. Mesenchymal stem cells (MSCs) have been reported to secrete soluble factors necessary for skeletal muscle growth and regeneration. Methods We induced MSC-like cells (iMSCs) from induced pluripotent stem cells (iPSCs) and examined the effects of iMSCs on the proliferation and differentiation of human myogenic cells and on the engraftment of human myogenic cells in the tibialis anterior (TA) muscle of NSG-mdx4Cv mice, an immunodeficient dystrophin-deficient DMD model. We also examined the cytokines secreted by iMSCs and tested their effects on the engraftment of human myogenic cells. Results iMSCs promoted the proliferation and differentiation of human myogenic cells to the same extent as bone marrow-derived (BM)-MSCs in coculture experiments. In cell transplantation experiments, iMSCs significantly improved the engraftment of human myogenic cells injected into the TA muscle of NSG-mdx4Cv mice. Cytokine array analysis revealed that iMSCs produced insulin-like growth factor-binding protein 2 (IGFBP2), urokinase-type plasminogen activator receptor (uPAR), and brain-derived neurotrophic factor (BDNF) at higher levels than did BM-MSCs. We further found that uPAR stimulates the migration of human myogenic cells in vitro and promotes their engraftment into the TA muscles of immunodeficient NOD/Scid mice. Conclusions Our results indicate that iMSCs are a new tool to improve the engraftment of myogenic progenitors in dystrophic muscle. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02594-1.
Collapse
Affiliation(s)
- Ahmed Elhussieny
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Neurology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Ken'ichiro Nogami
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Fusako Sakai-Takemura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Yusuke Maruyama
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Natsumi Takemura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Wael Talaat Soliman
- Department of Neurology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Yuko Miyagoe-Suzuki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
8
|
Careccia G, Saclier M, Tirone M, Ruggieri E, Principi E, Raffaghello L, Torchio S, Recchia D, Canepari M, Gorzanelli A, Ferrara M, Castellani P, Rubartelli A, Rovere-Querini P, Casalgrandi M, Preti A, Lorenzetti I, Bruno C, Bottinelli R, Brunelli S, Previtali SC, Bianchi ME, Messina G, Vénéreau E. Rebalancing expression of HMGB1 redox isoforms to counteract muscular dystrophy. Sci Transl Med 2021; 13:13/596/eaay8416. [PMID: 34078746 DOI: 10.1126/scitranslmed.aay8416] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/03/2021] [Accepted: 05/07/2021] [Indexed: 12/21/2022]
Abstract
Muscular dystrophies (MDs) are a group of genetic diseases characterized by progressive muscle wasting associated to oxidative stress and persistent inflammation. It is essential to deepen our knowledge on the mechanism connecting these two processes because current treatments for MDs have limited efficacy and/or are associated with side effects. Here, we identified the alarmin high-mobility group box 1 (HMGB1) as a functional link between oxidative stress and inflammation in MDs. The oxidation of HMGB1 cysteines switches its extracellular activities from the orchestration of tissue regeneration to the exacerbation of inflammation. Extracellular HMGB1 is present at high amount and undergoes oxidation in patients with MDs and in mouse models of Duchenne muscular dystrophy (DMD) and limb-girdle muscular dystrophy 3 (LGMDR3) compared to controls. Genetic ablation of HMGB1 in muscles of DMD mice leads to an amelioration of the dystrophic phenotype as evidenced by the reduced inflammation and muscle degeneration, indicating that HMGB1 oxidation is a detrimental process in MDs. Pharmacological treatment with an engineered nonoxidizable variant of HMGB1, called 3S, improves functional performance, muscle regeneration, and satellite cell engraftment in dystrophic mice while reducing inflammation and fibrosis. Overall, our data demonstrate that the balance between HMGB1 redox isoforms dictates whether skeletal muscle is in an inflamed or regenerating state, and that the nonoxidizable form of HMGB1 is a possible therapeutic approach to counteract the progression of the dystrophic phenotype. Rebalancing the HMGB1 redox isoforms may also be a therapeutic strategy for other disorders characterized by chronic oxidative stress and inflammation.
Collapse
Affiliation(s)
- Giorgia Careccia
- Division of Genetics and Cell Biology, Tissue Regeneration and Homeostasis Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Marielle Saclier
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Mario Tirone
- Division of Genetics and Cell Biology, Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elena Ruggieri
- Division of Genetics and Cell Biology, Tissue Regeneration and Homeostasis Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Elisa Principi
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Lizzia Raffaghello
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Silvia Torchio
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Deborah Recchia
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Monica Canepari
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Andrea Gorzanelli
- Division of Genetics and Cell Biology, Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Michele Ferrara
- Division of Genetics and Cell Biology, Tissue Regeneration and Homeostasis Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Patrizia Castellani
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Anna Rubartelli
- Unità di Biologia Cellulare, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Division of Immunology, Transplantation and Infectious Immunity, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | | | | | - Isabella Lorenzetti
- Division of Neuroscience and Inspe, Neuromuscular Repair Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.,ICS-Maugeri (IRCCS), Scientific Institute of Pavia, 27100 Pavia, Italy
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Stefano Carlo Previtali
- Division of Neuroscience and Inspe, Neuromuscular Repair Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco Emilio Bianchi
- Vita-Salute San Raffaele University, 20132 Milan, Italy.,Division of Genetics and Cell Biology, Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Emilie Vénéreau
- Division of Genetics and Cell Biology, Tissue Regeneration and Homeostasis Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
9
|
Selective Proliferation of Highly Functional Adipose-Derived Stem Cells in Microgravity Culture with Stirred Microspheres. Cells 2021; 10:cells10030560. [PMID: 33806638 PMCID: PMC7998608 DOI: 10.3390/cells10030560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Therapeutic effects of adult stem-cell transplantations are limited by poor cell-retention in target organs, and a reduced potential for optimal cell differentiation compared to embryonic stem cells. However, contemporary studies have indicated heterogeneity within adult stem-cell pools, and a novel culturing technique may address these limitations by selecting those for cell proliferation which are highly functional. Here, we report the preservation of stemness in human adipose-derived stem cells (hASCs) by using microgravity conditions combined with microspheres in a stirred suspension. The cells were bound to microspheres (100-300 μm) and cultured using a wave-stirring shaker. One-week cultures using polystyrene and collagen microspheres increased the proportions of SSEA-3(+) hASCs 4.4- and 4.3-fold (2.7- and 2.9-fold increases in their numbers), respectively, compared to normal culture conditions. These cultured hASCs expressed higher levels of pluripotent markers (OCT4, SOX2, NANOG, MYC, and KLF), and had improved abilities for proliferation, colony formation, network formation, and multiple-mesenchymal differentiation. We believe that this novel culturing method may further enhance regenerative therapies using hASCs.
Collapse
|
10
|
Contreras-Muñoz P, Torrella JR, Venegas V, Serres X, Vidal L, Vila I, Lahtinen I, Viscor G, Martínez-Ibáñez V, Peiró JL, Järvinen TAH, Rodas G, Marotta M. Muscle Precursor Cells Enhance Functional Muscle Recovery and Show Synergistic Effects With Postinjury Treadmill Exercise in a Muscle Injury Model in Rats. Am J Sports Med 2021; 49:1073-1085. [PMID: 33719605 DOI: 10.1177/0363546521989235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Skeletal muscle injuries represent a major concern in sports medicine. Cell therapy has emerged as a promising therapeutic strategy for muscle injuries, although the preclinical data are still inconclusive and the potential clinical use of cell therapy has not yet been established. PURPOSE To evaluate the effects of muscle precursor cells (MPCs) on muscle healing in a small animal model. STUDY DESIGN Controlled laboratory study. METHODS A total of 27 rats were used in the study. MPCs were isolated from rat (n = 3) medial gastrocnemius muscles and expanded in primary culture. Skeletal muscle injury was induced in 24 rats, and the animals were assigned to 3 groups. At 36 hours after injury, animals received treatment based on a single ultrasound-guided MPC (105 cells) injection (Cells group) or MPC injection in combination with 2 weeks of daily exercise training (Cells+Exercise group). Animals receiving intramuscular vehicle injection were used as controls (Vehicle group). Muscle force was determined 2 weeks after muscle injury, and muscles were collected for histological and immunofluorescence evaluation. RESULTS Red fluorescence-labeled MPCs were successfully transplanted in the site of the injury by ultrasound-guided injection and were localized in the injured area after 2 weeks. Transplanted MPCs participated in the formation of regenerating muscle fibers as corroborated by the co-localization of red fluorescence with developmental myosin heavy chain (dMHC)-positive myofibers by immunofluorescence analysis. A strong beneficial effect on muscle force recovery was detected in the Cells and Cells+Exercise groups (102.6% ± 4.0% and 101.5% ± 8.5% of maximum tetanus force of the injured vs healthy contralateral muscle, respectively) compared with the Vehicle group (78.2% ± 5.1%). Both Cells and Cells+Exercise treatments stimulated the growth of newly formed regenerating muscles fibers, as determined by the increase in myofiber cross-sectional area (612.3 ± 21.4 µm2 and 686.0 ± 11.6 µm2, respectively) compared with the Vehicle group (247.5 ± 10.7 µm2), which was accompanied by a significant reduction of intramuscular fibrosis in Cells and Cells+Exercise treated animals (24.2% ± 1.3% and 26.0% ± 1.9% of collagen type I deposition, respectively) with respect to control animals (40.9% ± 4.1% in the Vehicle group). MPC treatment induced a robust acceleration of the muscle healing process as demonstrated by the decreased number of dMHC-positive regenerating myofibers (enhanced replacement of developmental myosin isoform by mature myosin isoforms) (4.3% ± 2.6% and 4.1% ± 1.5% in the Cells and Cells+Exercise groups, respectively) compared with the Vehicle group (14.8% ± 13.9%). CONCLUSION Single intramuscular administration of MPCs improved histological outcome and force recovery of the injured skeletal muscle in a rat injury model that imitates sports-related muscle injuries. Cell therapy showed a synergistic effect when combined with an early active rehabilitation protocol in rats, which suggests that a combination of treatments can generate novel therapeutic strategies for the treatment of human skeletal muscle injuries. CLINICAL RELEVANCE Our study demonstrates the strong beneficial effect of MPC transplant and the synergistic effect when the cell therapy is combined with an early active rehabilitation protocol for muscle recovery in rats; this finding opens new avenues for the development of effective therapeutic strategies for muscle healing and clinical trials in athletes undergoing MPC transplant and rehabilitation protocols.
Collapse
Affiliation(s)
- Paola Contreras-Muñoz
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Joan Ramón Torrella
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Vanessa Venegas
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Xavier Serres
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Laura Vidal
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ingrid Vila
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ilmari Lahtinen
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ginés Viscor
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Vicente Martínez-Ibáñez
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - José Luis Peiró
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Tero A H Järvinen
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Gil Rodas
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Mario Marotta
- Investigation performed at Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
11
|
Laurent A, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh AS, Raffoul W, Applegate LA. Holistic Approach of Swiss Fetal Progenitor Cell Banking: Optimizing Safe and Sustainable Substrates for Regenerative Medicine and Biotechnology. Front Bioeng Biotechnol 2020; 8:557758. [PMID: 33195124 PMCID: PMC7644790 DOI: 10.3389/fbioe.2020.557758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
Safety, quality, and regulatory-driven iterative optimization of therapeutic cell source selection has constituted the core developmental bedrock for primary fetal progenitor cell (FPC) therapy in Switzerland throughout three decades. Customized Fetal Transplantation Programs were pragmatically devised as straightforward workflows for tissue procurement, traceability maximization, safety, consistency, and robustness of cultured progeny cellular materials. Whole-cell bioprocessing standardization has provided plethoric insights into the adequate conjugation of modern biotechnological advances with current restraining legislative, ethical, and regulatory frameworks. Pioneer translational advances in cutaneous and musculoskeletal regenerative medicine continuously demonstrate the therapeutic potential of FPCs. Extensive technical and clinical hindsight was gathered by managing pediatric burns and geriatric ulcers in Switzerland. Concomitant industrial transposition of dermal FPC banking, following good manufacturing practices, demonstrated the extensive potential of their therapeutic value. Furthermore, in extenso, exponential revalorization of Swiss FPC technology may be achieved via the renewal of integrative model frameworks. Consideration of both longitudinal and transversal aspects of simultaneous fetal tissue differential processing allows for a better understanding of the quasi-infinite expansion potential within multi-tiered primary FPC banking. Multiple fetal tissues (e.g., skin, cartilage, tendon, muscle, bone, lung) may be simultaneously harvested and processed for adherent cell cultures, establishing a unique model for sustainable therapeutic cellular material supply chains. Here, we integrated fundamental, preclinical, clinical, and industrial developments embodying the scientific advances supported by Swiss FPC banking and we focused on advances made to date for FPCs that may be derived from a single organ donation. A renewed model of single organ donation bioprocessing is proposed, achieving sustained standards and potential production of billions of affordable and efficient therapeutic doses. Thereby, the aim is to validate the core therapeutic value proposition, to increase awareness and use of standardized protocols for translational regenerative medicine, potentially impacting millions of patients suffering from cutaneous and musculoskeletal diseases. Alternative applications of FPC banking include biopharmaceutical therapeutic product manufacturing, thereby indirectly and synergistically enhancing the power of modern therapeutic armamentariums. It is hypothesized that a single qualifying fetal organ donation is sufficient to sustain decades of scientific, medical, and industrial developments, as technological optimization and standardization enable high efficiency.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Tec-Pharma SA, Bercher, Switzerland
- LAM Biotechnologies SA, Épalinges, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
| | - Anthony S. de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Épalinges, Switzerland
- Oxford Suzhou Center for Advanced Research, Science and Technology Co., Ltd., Oxford University, Suzhou, China
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Abstract
PURPOSE To evaluate, if clinically translatable ferumoxytol nanoparticles can be used for in vivo detection and quantification of stem cell transplants with magnetic particle imaging (MPI). PROCEDURES Mesenchymal stem cells (MSCs) were labeled with ferumoxytol or ferucarbotran and underwent MPI, magnetic resonance imaging (MRI), Prussian blue staining, and inductively coupled plasma (ICP) spectrometry. Unlabeled, ferumoxytol, and ferucarbotran-labeled MSCs were implanted in calvarial defects of eight mice and underwent MPI, MRI, and histopathology. The iron concentration calculated according to the MPI signal intensity and T2 relaxation times of the three different groups were compared using an analysis of variance (ANOVA) with Bonferroni correction, and a p < 0.05. RESULTS Compared to unlabeled controls, ferumoxytol- and ferucarbotran-labeled MSC showed significantly increased iron content, MPI signal and MRI signal. The ferumoxytol MPI signal was approximately 4× weaker compared to ferucarbotran at equimolar concentrations (p = 0.0003) and approximately 1.5× weaker for labeled cells when using optimized labeling protocols (p = 0.002). In vivo, the MPI signal of ferumoxytol-labeled MSC decreased significantly between day 1 and day 14 (p = 0.0124). This was confirmed by histopathology where we observed a decrease in Prussian blue stain of MSCs at the transplant site. The MRI signal of the same transplants did not change significantly during this observation period (p = 0.93). CONCLUSION Ferumoxytol nanoparticles can be used for in vivo detection of stem cell transplants with MPI and provide quantitative information not attainable with MRI.
Collapse
|
13
|
Abstract
Muscle stem cells, or satellite cells, are required for skeletal muscle maintenance, growth, and repair. Following satellite cell activation, several factors drive asymmetric cell division to generate a stem cell and a proliferative progenitor that forms new muscle. The balance between symmetric self-renewal and asymmetric division significantly impacts the efficiency of regeneration. In this Review, we discuss the relationship of satellite cell heterogeneity and the establishment of polarity to asymmetric division, as well as how these processes are impacted in homeostasis, aging, and disease. We also highlight therapeutic opportunities for targeting satellite cell polarity and self-renewal to stimulate muscle regeneration.
Collapse
|
14
|
O’Connell CD, Onofrillo C, Duchi S, Li X, Zhang Y, Tian P, Lu L, Trengove A, Quigley A, Gambhir S, Khansari A, Mladenovska T, O’Connor A, Di Bella C, Choong PF, Wallace GG. Evaluation of sterilisation methods for bio-ink components: gelatin, gelatin methacryloyl, hyaluronic acid and hyaluronic acid methacryloyl. Biofabrication 2019; 11:035003. [DOI: 10.1088/1758-5090/ab0b7c] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Silva Garcia JM, Panitch A, Calve S. Functionalization of hyaluronic acid hydrogels with ECM-derived peptides to control myoblast behavior. Acta Biomater 2019; 84:169-179. [PMID: 30508655 DOI: 10.1016/j.actbio.2018.11.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/31/2018] [Accepted: 11/19/2018] [Indexed: 01/07/2023]
Abstract
Volumetric muscle loss (VML) occurs when skeletal muscle injury is too large for the body to fully self-repair. Typically, fibrotic tissue fills the void, which reduces muscle functionality and limb movement. Although a wide variety of natural and synthetic scaffolds have been studied with the purpose of providing the appropriate structural support, to date no scaffold has significantly restored muscle functionality after VML. Satellite cells, adult stem cells within the muscle capable of restoring smaller injuries, are sensitive to the stiffness and composition of the surrounding environment. Scaffolds that only address structural support are not sufficient to restore functionality and instead need to be designed to both promote satellite cell activation and prevent excessive fibroblast recruitment. The objective of this study was to design a scaffold that mimicked the regenerative environment and determine how the biomechanical properties differentially influence myogenic precursor and connective tissue cells. One of the main extracellular matrix (ECM) molecules upregulated during regeneration is hyaluronic acid (HA). Therefore, thiol-modified HA and poly(ethylene glycol) diacrylate hydrogels were generated and functionalized with peptides based on ECM known to influence regeneration, including fibronectin, laminin and tenascin-C. Scaffolds with different stiffness were created by varying HA content. The influence of HA stiffness and peptide functionalization on myogenic precursor and connective tissue cell proliferation, migration and gene expression was quantified. Our results indicated that HA hydrogels functionalized with the laminin peptide, IKVAV, show potential due to the enhanced promotion of myogenic cell behaviors including migration, proliferation and an increase in relevant transcription factors. STATEMENT OF SIGNIFICANCE: The goal of this study was to identify hyaluronic acid (HA) hydrogels with peptide and stiffness combinations that will direct muscle-derived cells towards regenerating phenotypes. While the interaction of skeletal muscle with RGD-functionalized HA hydrogels has been investigated, none of the other peptides described in this study had been used in the context of HA-based scaffolds and skeletal muscle-derived cells. Notably, the response of cells to variations in mechanics was dependent on ECM coating and lineage. The 3% HA functionalized with the laminin peptide, IKVAV, showed the most promise for future in vivo studies, as these hydrogels best promoted myoblast cell proliferation, attachment and spreading, enhanced migration over connective tissue cells and upregulated transcription factors associated with activated satellite cells.
Collapse
|
16
|
Shi L, Ye L, Liu P, Liu D, Ye G, Chen J, Dong Z. Ulinastatin inhibits apoptosis induced by serum deprivation in mesenchymal stem cells. Mol Med Rep 2019; 19:2397-2406. [PMID: 30664153 DOI: 10.3892/mmr.2019.9847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 10/10/2018] [Indexed: 11/06/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have exhibited great potential in the therapy of cardiovascular disease. However, the application of MSCs is hampered by apoptosis, which reduces the number of cells in the host cardiac microenvironment. Ulinastatin (UTI), a broad‑spectrum protease inhibitor that can be purified from human urine, has attracted attention for its protective effects through its immunomodulatory and anti‑inflammatory properties. The present study aimed to evaluate the effects of UTI on serum deprivation‑induced apoptosis of MSCs and investigate its molecular mechanisms. Cell viability was determined by the MTT assay. Apoptosis was assessed by flow cytometric analysis with Annexin V/propidium iodide staining. The protein levels of cleaved caspase‑3, B‑cell lymphoma‑2 (Bcl‑2) family proteins, total‑Akt and phospho‑Akt were evaluated by western blot. The results of the present study demonstrated that UTI exhibited a protective effect in serum deprived MSCs, as indicated by increased cell viability, and a reduction in the rate of apoptosis and caspase‑3 activation. In addition, treatment with UTI significantly decreased the expression levels of Bcl‑2, Bcl‑extra large and Bcl‑associated X protein. Furthermore, activation of the Akt signaling pathway was involved in the UTI‑induced anti‑apoptotic effects. The present findings indicated that UTI is able to promote the survival of MSCs under serum deprivation conditions. The present study may be helpful in improving the therapeutic efficacy of MSC transplantation used to cure chronic ischemic heart disease.
Collapse
Affiliation(s)
- Linhui Shi
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Longqiang Ye
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Panpan Liu
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Danqin Liu
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Gongjie Ye
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Jiahong Chen
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| | - Zhouzhou Dong
- Critical Care Unit, Ningbo Medical Center Lihuili Eastern Hospital, Taipei Medical University Ningbo Medical Center, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
17
|
|
18
|
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: 0.1] [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
|
19
|
Hejbøl EK, Sellathurai J, Nair PD, Schrøder HD. Injectable scaffold materials differ in their cell instructive effects on primary human myoblasts. J Tissue Eng 2017; 8:2041731417717677. [PMID: 28717506 PMCID: PMC5502935 DOI: 10.1177/2041731417717677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022] Open
Abstract
Scaffolds are materials used for delivery of cells for regeneration of tissues. They support three-dimensional organization and improve cell survival. For the repair of small skeletal muscles, injections of small volumes of cells are attractive, and injectable scaffolds for delivery of cells offer a minimally invasive technique. In this study, we examined in vitro the cell instructive effects of three types of injectable scaffolds, fibrin, alginate, and poly(lactic-co-glycolic acid)-based microparticles on primary human myoblasts. The myoblast morphology and progression in the myogenic program differed, depending on the type of scaffold material. In alginate gel, the cells obtained a round morphology, they ceased to proliferate, and entered quiescence. In the fibrin gels, differentiation was promoted, and myotubes were observed within a few days in culture, while poly(lactic-co-glycolic acid)-based microparticles supported prolonged proliferation. Myoblasts released from the alginate and fibrin gels were studied, and cells released from these scaffolds had retained the ability to proliferate and differentiate. Thus, the study shows that human myogenic cells combined with injectable scaffold materials are guided into different states depending on the choice of scaffold. This opens for in vivo experiments, including testing of the significance of the cell state on regeneration potential of primary human myoblasts.
Collapse
Affiliation(s)
- Eva Kildall Hejbøl
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark
| | - Jeeva Sellathurai
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark
| | - Prabha Damodaran Nair
- Division of Tissue Engineering and Regeneration Technologies, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Henrik Daa Schrøder
- Institute of Clinical Research, SDU Muscle Research Cluster, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
20
|
Bosio E, Lee-Pullen TF, Fragall CT, Beilharz MW, Bennett AL, Grounds MD, Hodgetts SI, Sammels LM. A Comparison between Real-Time Quantitative PCR and DNA Hybridization for Quantitation of Male DNA following Myoblast Transplantation. Cell Transplant 2017; 13:817-21. [PMID: 15690984 DOI: 10.3727/000000004783983369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The transplantation of muscle precursor cells (myoblasts) is a potential therapy for Duchenne muscular dystrophy. A commonly used method to detect cell survival is quantitation of the Y chromosome following transplantation of male donor cells into female hosts. This article presents a direct comparison between real-time quantitative PCR (Q-PCR) and the DNA hybridization (slot-blot) technique for quantitation of Y chromosome DNA. Q-PCR has a significantly greater linear quantitation range and is up to 40-fold more sensitive at low concentrations of male DNA, detecting as little as 1 ng of male DNA in each female tibialis anterior (TA) muscle. At high male DNA concentrations, accurate quantitation by Q-PCR is 2.5 times higher than the maximum possible with slot-blot. In conclusion, Q-PCR has a higher dynamic range and is more efficient than slot-blot analysis for the detection of donor cell engraftment in a transsexual transplantation model.
Collapse
Affiliation(s)
- Erika Bosio
- Discipline of Microbiology, School of Biomedical and Chemical Sciences, The University of Western Australia, Perth, WA, Australia
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Gimeno ML, Fuertes F, Barcala Tabarrozzi AE, Attorressi AI, Cucchiani R, Corrales L, Oliveira TC, Sogayar MC, Labriola L, Dewey RA, Perone MJ. Pluripotent Nontumorigenic Adipose Tissue-Derived Muse Cells have Immunomodulatory Capacity Mediated by Transforming Growth Factor-β1. Stem Cells Transl Med 2016; 6:161-173. [PMID: 28170177 PMCID: PMC5442729 DOI: 10.5966/sctm.2016-0014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stromal cell‐based interventions have shown promising results in a broad range of diseases. However, their use has faced limited effectiveness owing to the low survival rates and susceptibility to environmental stress on transplantation. We describe the cellular and molecular characteristics of multilineage‐differentiating stress‐enduring (Muse) cells derived from adipose tissue (AT), a subpopulation of pluripotent stem cells isolated from human lipoaspirates. Muse‐AT cells were efficiently obtained using a simple, fast, and affordable procedure, avoiding cell sorting and genetic manipulation methods. Muse‐AT cells isolated under severe cellular stress, expressed pluripotency stem cell markers and spontaneously differentiated into the three germ lineages. Muse‐AT cells grown as spheroids have a limited proliferation rate, a diameter of ∼15 µm, and ultrastructural organization similar to that of embryonic stem cells. Muse‐AT cells evidenced high stage‐specific embryonic antigen‐3 (SSEA‐3) expression (∼60% of cells) after 7–10 days growing in suspension and did not form teratomas when injected into immunodeficient mice. SSEA‐3+‐Muse‐AT cells expressed CD105, CD29, CD73, human leukocyte antigen (HLA) class I, CD44, and CD90 and low levels of HLA class II, CD45, and CD34. Using lipopolysaccharide‐stimulated macrophages and antigen‐challenged T‐cell assays, we have shown that Muse‐AT cells have anti‐inflammatory activities downregulating the secretion of proinflammatory cytokines, such as interferon‐γ and tumor necrosis factor‐α. Muse‐AT cells spontaneously gained transforming growth factor‐β1 expression that, in a phosphorylated SMAD2‐dependent manner, might prove pivotal in their observed immunoregulatory activity through decreased expression of T‐box transcription factor in T cells. Collectively, the present study has demonstrated the feasibility and efficiency of obtaining Muse‐AT cells that can potentially be harnessed as immunoregulators to treat immune‐related disorders. Stem Cells Translational Medicine2017;6:161–173
Collapse
Affiliation(s)
- María L. Gimeno
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Florencia Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Andres E. Barcala Tabarrozzi
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Alejandra I. Attorressi
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | | | - Luis Corrales
- Servicio de Cirugía Plástica, Hospital Austral, Derqui, Argentina
| | - Talita C. Oliveira
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, Brasil
| | - Mari C. Sogayar
- Cell and Molecular Therapy Center (Núcleo de Terapia Celular e Molecular/NETCEM), School of Medicine, University of São Paulo, São Paulo, Brasil
| | - Leticia Labriola
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, Brasil
| | - Ricardo A. Dewey
- Laboratorio de Terapia Génica y Células Madre, Instituto de Investigaciones Biotecnológicas–Instituto Tecnológico de Chascomús (IIB‐INTECH), National Scientific and Technical Research Council, National University of General San Martin, Chascomús, Argentina
| | - Marcelo J. Perone
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| |
Collapse
|
22
|
Wang Z, Fang B, Tan Z, Zhang D, Ma H. Hypoxic preconditioning increases the protective effect of bone marrow mesenchymal stem cells on spinal cord ischemia/reperfusion injury. Mol Med Rep 2016; 13:1953-60. [PMID: 26783161 PMCID: PMC4768971 DOI: 10.3892/mmr.2016.4753] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 07/23/2015] [Indexed: 11/05/2022] Open
Abstract
Transplantation of bone marrow mesenchymal stem cells (BMSCs) protect against spinal cord ischemia/reperfusion injury (SCIRI). However, a large number of transplanted BMSCs often undergo apoptosis, which severely affects the treatment outcome. Previous studies have demonstrated that hypoxic preconditioning effectively increases the survival rate of BMSCs following transplantation, and increases their protective effect on injured tissues. However, there have been few reports regarding roles of hypoxic preconditioning in SCIRI. The present study isolated rat BMSCs and separately transplanted hypoxia‑ and non‑hypoxia‑preconditioned BMSCs into the spinal cord tissues of rats with SCIRI. The role of hypoxic preconditioning in the promotion of the protective effect of BMSCs on SCIRI was investigated using neurological function scores, Evans blue staining, hematoxylin and eosin staining and terminal deoxynucleotidyl transferase dUTP nick end labeling. In addition, reverse transcription‑quantitative polymerase chain reaction and western blotting were used to detect the expression levels of hypoxia‑inducible factor 1α (HIF‑1α), and to investigate its possible underlying mechanism of action. The results indicated that hypoxic preconditioning effectively increased the protective effects of BMSCs on neurological function, blood spinal cord barrier and tissue damage following SCIRI, and inhibited apoptosis. Furthermore, hypoxic preconditioned BMSCs upregulated the expression of HIF‑1α in spinal cord tissues. Therefore, hypoxic preconditioning effectively increased the protective effect of BMSCs on SCIRI and may be associated with upregulation of the expression of HIF‑1α. Hypoxic preconditioning may serve as an effective means of increasing the protective effect of BMSCs on SCIRI.
Collapse
Affiliation(s)
- Zhilin Wang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Bo Fang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhibin Tan
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Dong Zhang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hong Ma
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
23
|
Kim MH, Lee YJ, Kang JH. Stem Cell Monitoring with a Direct or Indirect Labeling Method. Nucl Med Mol Imaging 2015; 50:275-283. [PMID: 27994682 DOI: 10.1007/s13139-015-0380-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/05/2015] [Accepted: 10/07/2015] [Indexed: 11/25/2022] Open
Abstract
The molecular imaging techniques allow monitoring of the transplanted cells in the same individuals over time, from early localization to the survival, migration, and differentiation. Generally, there are two methods of stem cell labeling: direct and indirect labeling methods. The direct labeling method introduces a labeling agent into the cell, which is stably incorporated or attached to the cells prior to transplantation. Direct labeling of cells with radionuclides is a simple method with relatively fewer adverse events related to genetic responses. However, it can only allow short-term distribution of transplanted cells because of the decreasing imaging signal with radiodecay, according to the physical half-lives, or the signal becomes more diffuse with cell division and dispersion. The indirect labeling method is based on the expression of a reporter gene transduced into the cell before transplantation, which is then visualized upon the injection of an appropriate probe or substrate. In this review, various imaging strategies to monitor the survival and behavior change of transplanted stem cells are covered. Taking these new approaches together, the direct and indirect labeling methods may provide new insights on the roles of in vivo stem cell monitoring, from bench to bedside.
Collapse
Affiliation(s)
- Min Hwan Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-gil, Gongneung-Dong, Nowon-Gu, Seoul, 139-706 Republic of Korea
| | - Yong Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-gil, Gongneung-Dong, Nowon-Gu, Seoul, 139-706 Republic of Korea
| | - Joo Hyun Kang
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences (KIRAMS), 75 Nowon-gil, Gongneung-Dong, Nowon-Gu, Seoul, 139-706 Republic of Korea
| |
Collapse
|
24
|
Mohamed SS, Ahmed LA, Attia WA, Khattab MM. Nicorandil enhances the efficacy of mesenchymal stem cell therapy in isoproterenol-induced heart failure in rats. Biochem Pharmacol 2015; 98:403-11. [PMID: 26453143 DOI: 10.1016/j.bcp.2015.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/02/2015] [Indexed: 12/14/2022]
Abstract
Stem cell transplantation has emerged as a promising technique for regenerative medicine in cardiovascular therapeutics. However, the results have been less than optimal. The aim of the present study was to investigate whether nicorandil could offer an additional benefit over bone marrow-derived mesenchymal stem cell therapy in isoproterenol-induced myocardial damage and its progression to heart failure in rats. Isoproterenol was injected subcutaneously for 2 consecutive days at doses of 85 and 170 mg/kg/day, respectively. Nicorandil (3 mg/kg/day) was then given orally with or without a single intravenous bone marrow-derived mesenchymal stem cell administration. Electrocardiography and echocardiography were recorded 2 weeks after the beginning of treatment. Rats were then sacrificed and the ventricle was isolated for estimation of tumor necrosis factor-alpha, vascular endothelial growth factor and transforming growth factor-beta. Moreover, protein expressions of caspase-3, connexin-43 as well as endothelial and inducible nitric oxide synthases were evaluated. Finally, histological studies of myocardial fibrosis and blood vessel density were performed and cryosections were done for estimation cell homing. Combined nicorandil/bone marrow-derived mesenchymal stem cell therapy provided an additional improvement compared to cell therapy alone toward reducing isoproterenol-induced cardiac hypertrophy, fibrosis and inflammation. Notably, combined therapy induced significant increase in angiogenesis and cell homing and prevented isoproterenol-induced changes in contractility and apoptotic markers. In conclusion, combined nicorandil/bone marrow-derived mesenchymal stem cell therapy was superior to cell therapy alone toward preventing isoproterenol-induced heart failure in rats through creation of a supportive environment for mesenchymal stem cells.
Collapse
Affiliation(s)
- Sarah S Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Lamiaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Wael A Attia
- Pediatric Department, Pediatric Cardiology Unit, Abou EL-Reesh Children Hospital, Cairo, Egypt.
| | - Mahmoud M Khattab
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
25
|
Zeng K, Deng BP, Jiang HQ, Wang M, Hua P, Zhang HW, Deng YB, Yang YQ. Prostaglandin E₁ protects bone marrow-derived mesenchymal stem cells against serum deprivation-induced apoptosis. Mol Med Rep 2015; 12:5723-9. [PMID: 26252504 PMCID: PMC4581785 DOI: 10.3892/mmr.2015.4176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 12/09/2014] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have become a recent focus of experimental and clinical research regarding myocardial regeneration. However, the therapeutic potential of these cells is limited by poor survival. Prostaglandin E1 (PGE1) is known to have anti-inflammatory and anti-apoptotic effects on the myocardium. The aim of the present study was to determine whether PGE1 could protect MSCs against serum deprivation (SD)-induced apoptosis. An SD model was used to induce apoptosis in MSCs in vitro. Apoptotic morphological changes were detected by Hoechst 33258 fluorescent nuclear staining; and Annexin V-fluorescein isothiocyanate/propidium iodide (PI) double staining and flow cytometry was used to quantify the rate of apoptosis. Western blot analysis was used to detect the expression levels of the apoptosis-associated proteins Bcl-2, Bax and caspase-3. The results of the present study demonstrated that SD induced apoptosis of MSCs, and that treatment with PGE1 attenuated the morphological changes characteristic of apoptosis. Annexin V/PI staining showed that the rate of apoptosis gradually increased with the duration of ischemia. Furthermore, treatment with PGE1 significantly reduced SD-induced apoptosis, decreased the protein expression levels of Bax and caspase-3, and increased the expression levels of Bcl-2. These data suggest that PGE1 is able to influence the survival of MSCs under certain conditions. These results may aid in improving the therapeutic efficacy of MSC transplantation used to treat chronic ischemic heart disease.
Collapse
Affiliation(s)
- Kuan Zeng
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Bao Ping Deng
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hui-Qi Jiang
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Meng Wang
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ping Hua
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hong-Wu Zhang
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yu-Bin Deng
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yan-Qi Yang
- Department of Cardiac Surgery, Sun Yat‑sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
26
|
Chen CH, Sereti KI, Wu BM, Ardehali R. Translational aspects of cardiac cell therapy. J Cell Mol Med 2015; 19:1757-72. [PMID: 26119413 PMCID: PMC4549027 DOI: 10.1111/jcmm.12632] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/13/2015] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has been intensely studied for over a decade as a potential treatment for ischaemic heart disease. While initial trials using skeletal myoblasts, bone marrow cells and peripheral blood stem cells showed promise in improving cardiac function, benefits were found to be short-lived likely related to limited survival and engraftment of the delivered cells. The discovery of putative cardiac ‘progenitor’ cells as well as the creation of induced pluripotent stem cells has led to the delivery of cells potentially capable of electromechanical integration into existing tissue. An alternative strategy involving either direct reprogramming of endogenous cardiac fibroblasts or stimulation of resident cardiomyocytes to regenerate new myocytes can potentially overcome the limitations of exogenous cell delivery. Complimentary approaches utilizing combination cell therapy and bioengineering techniques may be necessary to provide the proper milieu for clinically significant regeneration. Clinical trials employing bone marrow cells, mesenchymal stem cells and cardiac progenitor cells have demonstrated safety of catheter based cell delivery, with suggestion of limited improvement in ventricular function and reduction in infarct size. Ongoing trials are investigating potential benefits to outcome such as morbidity and mortality. These and future trials will clarify the optimal cell types and delivery conditions for therapeutic effect.
Collapse
Affiliation(s)
- Cheng-Han Chen
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Department of Bioengineering, UCLA, Los Angeles, CA, USA
| | - Konstantina-Ioanna Sereti
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Benjamin M Wu
- Department of Bioengineering, UCLA, Los Angeles, CA, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.,Eli and Edythe Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA
| |
Collapse
|
27
|
Pretreatment of Adipose Derived Stem Cells with Curcumin Facilitates Myocardial Recovery via Antiapoptosis and Angiogenesis. Stem Cells Int 2015; 2015:638153. [PMID: 26074974 PMCID: PMC4436501 DOI: 10.1155/2015/638153] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 02/04/2023] Open
Abstract
The poor survival rate of transplanted stem cells in ischemic myocardium has limited their therapeutic efficacy. Curcumin has potent antioxidant property. This study investigates whether prior curcumin treatment protects stem cells from oxidative stress injury and improves myocardial recovery following cells transplantation. Autologous Sprague-Dawley rat adipose derived mesenchymal stem cells (ADSCs) were pretreated with or without curcumin. The hydrogen peroxide/serum deprivation (H2O2/SD) medium was used to mimic the ischemic condition in vitro. Cytoprotective effects of curcumin on ADSCs were evaluated. Curcumin pretreatment significantly increased cell viability and VEGF secretion, and decreased cell injury and apoptosis via regulation of PTEN/Akt/p53 and HO-1 signal proteins expression. The therapeutic potential of ADSCs implantation was investigated in myocardial ischemia-reperfusion injury (IRI) model. Transplantation of curcumin pretreated ADSCs not only resulted in better heart function, higher cells retention, and smaller infarct size, but also decreased myocardial apoptosis, promoted neovascularization, and increased VEGF level in ischemic myocardium. Together, priming of ADSCs with curcumin improved tolerance to oxidative stress injury and resulted in enhancement of their therapeutic potential of ADSCs for myocardial repair. Curcumin pretreatment is a promising adjuvant strategy for stem cells transplantation in myocardial restoration.
Collapse
|
28
|
Kumar S, Ashraf M. Tadalafil, a Phosphodiesterase Inhibitor Protects Stem Cells over Longer Period Against Hypoxia/Reoxygenation Injury Through STAT3/PKG-I Signaling. Stem Cells Dev 2015; 24:1332-41. [PMID: 25602782 DOI: 10.1089/scd.2014.0288] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pharmacological preconditioning (PC) with tadalafil, a PDE5A inhibitor, enhances protein kinase G-1 (PKG-I) activity, resulting in stem cell survival. Protection by PC had two different phases, early (2 h) and late (24 h). However, the mechanism of protection during these phases remained grossly unknown. Mesenchymal stem cells (MSCs) from adult male Fischer-344 rats were cultured and pretreated with tadalafil (100 μM) for an hour and subjected to 2 h of hypoxia (1% O2), followed by reoxygenation (HR: in vitro model mimicking ischemia/reperfusion). We observed (i) increased MSC survival with reduced cell cytotoxicity as revealed by low lactate dehydrogenase release and trypan blue staining, respectively, in tadalafil-treated cells upon HR; (ii) decrease in TUNEL positivity as well as caspase activity; (iii) an increase in pAkt/Akt, iNOS, eNOS, and pGSK3β/GSK3β during the early protection phase of PC, and this protection seemed to be a spontaneous adaptive response of MSCs against HR and was independent of tadalafil, whereas an increase in Bcl2/Bax was tadalafil dependent; and (iv) during the late phase, we observed phosphorylation of STAT3 at serine727, leading to its entry inside the nucleus and binding onto the promoter of PKG-I by three-fold (P<0.05). In conclusion, an increase in Bcl2/Bax during the early phase and transcriptional upregulation of PKG-I by STAT3 during the late phase were responsible for stem cell protection by tadalafil against ischemic injury.
Collapse
Affiliation(s)
- Sanjay Kumar
- 1Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Muhammad Ashraf
- 2Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Rapamycin represses myotube hypertrophy and preserves viability of C2C12 cells during myogenesis in vitro. Transplantation 2014; 98:139-47. [PMID: 24926828 DOI: 10.1097/tp.0000000000000175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rapamycin (RAPA) has been successfully used for myoblast allotransplantation in X chromosome-linked muscular dystrophy mice. However, the mechanism of skeletal myogenesis, particularly in starved condition by RAPA, remains elusive. For this reason, we investigated the effect of RAPA on C2C12 myogenesis in serum-starved condition. METHODS Serum-free treated C2C12 cells were mimicked as skeletal myogenesis in nutrition shortage microenvironment. A methylthiazoletetrazolium (MTT) assay was used to investigate different RAPA concentrations on serum-free treated C2C12 cells and the following assays were used to detect the characteristic of C2C12 myogenesis by RAPA in vitro. RESULTS We found that 150 ng/mL of RAPA did not significantly suppress the viability of C2C12 differentiated cells by MTT assay. The RAPA concentration could protect myoblast serum-starved cells effectively from apoptosis through flow cytometry and retain myogenic regulatory factors through quantitative polymerase chain reaction analysis. However, RAPA significantly suppressed cell migration in wound healing assay (P<0.05). Morphological analyses indicated that RAPA also significantly suppressed myotube hypertrophy in serum-starved C2C12 cells. Western blot analysis revealed that the ratio of phosphate extracellular signal-regulated kinase/extracellular signal-regulated kinase and the protein level of p-Akt decreased in the proliferation medium and in the differentiation medium, respectively. CONCLUSION These findings suggest that myoblast cells are sensitive to RAPA under a serum-starved microenvironment. As an immunosuppressive agent, RAPA shall be used as a considering dosage and as a safe strategy for future myoblast allotransplantation.
Collapse
|
30
|
Dynamics of acute local inflammatory response after autologous transplantation of muscle-derived cells into the skeletal muscle. Mediators Inflamm 2014; 2014:482352. [PMID: 25242868 PMCID: PMC4163307 DOI: 10.1155/2014/482352] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/10/2014] [Accepted: 07/24/2014] [Indexed: 11/24/2022] Open
Abstract
The vast majority of myoblasts transplanted into the skeletal muscle die within the first week after injection. Inflammatory response to the intramuscular cell transfer was studied in allogeneic but not in autologous model. The aim of this study was to evaluate immune reaction to autotransplantation of myogenic cells and to assess its dynamics within the first week after injection. Muscle-derived cells or medium alone was injected into the intact skeletal muscles in autologous model. Tissue samples were collected 1, 3, and 7 days after the procedure. Our analysis revealed the peak increase of the gene expression of all evaluated cytokines (Il-1α, Il-1β, Il-6, Tgf-β, and Tnf-α) at day 1. The mRNA level of analyzed cytokines normalized in subsequent time points. The increase of Il-β
gene expression was further confirmed at the protein level. Analysis of the tissue sections revealed rapid infiltration of injected cell clusters with neutrophils and macrophages. The inflammatory infiltration was almost completely resolved at day 7. The survived cells were able to participate in the muscle regeneration process. Presented results demonstrate that autotransplanted muscle-derived cells induce classical early immune reaction in the site of injection which may contribute to cellular graft elimination.
Collapse
|
31
|
Stem cell transplantation for muscular dystrophy: the challenge of immune response. BIOMED RESEARCH INTERNATIONAL 2014; 2014:964010. [PMID: 25054157 PMCID: PMC4098613 DOI: 10.1155/2014/964010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/05/2014] [Indexed: 01/03/2023]
Abstract
Treating muscle disorders poses several challenges to the rapidly evolving field of regenerative medicine. Considerable progress has been made in isolating, characterizing, and expanding myogenic stem cells and, although we are now envisaging strategies to generate very large numbers of transplantable cells (e.g., by differentiating induced pluripotent stem cells), limitations directly linked to the interaction between transplanted cells and the host will continue to hamper a successful outcome. Among these limitations, host inflammatory and immune responses challenge the critical phases after cell delivery, including engraftment, migration, and differentiation. Therefore, it is key to study the mechanisms and dynamics that impair the efficacy of cell transplants in order to develop strategies that can ultimately improve the outcome of allogeneic and autologous stem cell therapies, in particular for severe disease such as muscular dystrophies. In this review we provide an overview of the main players and issues involved in this process and discuss potential approaches that might be beneficial for future regenerative therapies of skeletal muscle.
Collapse
|
32
|
Simerman AA, Dumesic DA, Chazenbalk GD. Pluripotent muse cells derived from human adipose tissue: a new perspective on regenerative medicine and cell therapy. Clin Transl Med 2014; 3:12. [PMID: 24940477 PMCID: PMC4041046 DOI: 10.1186/2001-1326-3-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/14/2014] [Indexed: 12/18/2022] Open
Abstract
In 2010, Multilineage Differentiating Stress Enduring (Muse) cells were introduced to the scientific community, offering potential resolution to the issue of teratoma formation that plagues both embryonic stem (ES) and induced pluripotent (iPS) stem cells. Isolated from human bone marrow, dermal fibroblasts, adipose tissue and commercially available adipose stem cells (ASCs) under severe cellular stress conditions, Muse cells self-renew in a controlled manner and do not form teratomas when injected into immune-deficient mice. Furthermore, Muse cells express classic pluripotency markers and differentiate into cells from the three embryonic germ layers both spontaneously and under media-specific induction. When transplanted in vivo, Muse cells contribute to tissue generation and repair. This review delves into the aspects of Muse cells that set them apart from ES, iPS, and various reported adult pluripotent stem cell lines, with specific emphasis on Muse cells derived from adipose tissue (Muse-AT), and their potential to revolutionize the field of regenerative medicine and stem cell therapy.
Collapse
Affiliation(s)
- Ariel A Simerman
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at 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 at the University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740, USA
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740, USA
| |
Collapse
|
33
|
Simerman AA, Perone MJ, Gimeno ML, Dumesic DA, Chazenbalk GD. A mystery unraveled: nontumorigenic pluripotent stem cells in human adult tissues. Expert Opin Biol Ther 2014; 14:917-29. [PMID: 24745973 DOI: 10.1517/14712598.2014.900538] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Embryonic stem cells and induced pluripotent stem cells have emerged as the gold standard of pluripotent stem cells and the class of stem cell with the highest potential for contribution to regenerative and therapeutic application; however, their translational use is often impeded by teratoma formation, commonly associated with pluripotency. We discuss a population of nontumorigenic pluripotent stem cells, termed Multilineage Differentiating Stress Enduring (Muse) cells, which offer an innovative and exciting avenue of exploration for the potential treatment of various human diseases. AREAS COVERED This review discusses the origin of Muse cells, describes in detail their various unique characteristics, and considers future avenues of their application and investigation with respect to what is currently known of adult pluripotent stem cells in scientific literature. We begin by defining cell potency, then discuss both mesenchymal and various reported populations of pluripotent stem cells, and finally delve into Muse cells and the characteristics that set them apart from their contemporaries. EXPERT OPINION Muse cells derived from adipose tissue (Muse-AT) are efficiently, routinely and painlessly isolated from human lipoaspirate material, exhibit tripoblastic differentiation both spontaneously and under media-specific induction, and do not form teratomas. We describe qualities specific to Muse-AT cells and their potential impact on the field of regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Ariel A Simerman
- The University of California, David Geffen School of Medicine, Department of Obstetrics and Gynecology , 10833 Le Conte Ave, Box 951740, Los Angeles, CA 90095-1740 , USA +1 310 206 3670 ;
| | | | | | | | | |
Collapse
|
34
|
Peng Y, Huang S, Wu Y, Cheng B, Nie X, Liu H, Ma K, Zhou J, Gao D, Feng C, Yang S, Fu X. Platelet rich plasma clot releasate preconditioning induced PI3K/AKT/NFκB signaling enhances survival and regenerative function of rat bone marrow mesenchymal stem cells in hostile microenvironments. Stem Cells Dev 2013; 22:3236-51. [PMID: 23885779 PMCID: PMC3868358 DOI: 10.1089/scd.2013.0064] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 07/25/2013] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been optimal targets in the development of cell based therapies, but their limited availability and high death rate after transplantation remains a concern in clinical applications. This study describes novel effects of platelet rich clot releasate (PRCR) on rat bone marrow-derived MSCs (BM-MSCs), with the former driving a gene program, which can reduce apoptosis and promote the regenerative function of the latter in hostile microenvironments through enhancement of paracrine/autocrine factors. By using reverse transcription-polymerase chain reaction, immunofluorescence and western blot analyses, we showed that PRCR preconditioning could alleviate the apoptosis of BM-MSCs under stress conditions induced by hydrogen peroxide (H2O2) and serum deprivation by enhancing expression of vascular endothelial growth factor and platelet-derived growth factor (PDGF) via stimulation of the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT/NF-κB signaling pathways. Furthermore, the effects of PRCR preconditioned GFP-BM-MSCs subcutaneously transplanted into rats 6 h after wound surgery were examined by histological and other tests from days 0-22 after transplantation. Engraftment of the PRCR preconditioned BM-MSCs not only significantly attenuated apoptosis and wound size but also improved epithelization and blood vessel regeneration of skin via regulation of the wound microenvironment. Thus, preconditioning with PRCR, which reprograms BM-MSCs to tolerate hostile microenvironments and enhance regenerative function by increasing levels of paracrine factors through PDGFR-α/PI3K/AKT/NF-κB signaling pathways would be a safe method for boosting the effectiveness of transplantation therapy in the clinic.
Collapse
Affiliation(s)
- Yan Peng
- The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area, PLA, Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Command, Guangzhou, People's Republic of China
- Southern Medical University, Guangzhou, People's Republic of China
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Sha Huang
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing, People's Republic of China
| | - Yan Wu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical College, Mudanjiang, People's Republic of China
| | - Biao Cheng
- The Key Laboratory of Trauma Treatment & Tissue Repair of Tropical Area, PLA, Department of Plastic Surgery, Guangzhou General Hospital of Guangzhou Command, Guangzhou, People's Republic of China
| | - Xiaohu Nie
- Southern Medical University, Guangzhou, People's Republic of China
| | - Hongwei Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Kui Ma
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Jiping Zhou
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Dongyun Gao
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Changjiang Feng
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Siming Yang
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
| | - Xiaobing Fu
- Burns Institute, Trauma Center of Postgraduate Medical College, The First Affiliated Hospital, General Hospital of PLA, Beijing, People's Republic of China
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing, People's Republic of China
| |
Collapse
|
35
|
Ye L, Zhang P, Duval S, Su L, Xiong Q, Zhang J. Thymosin β4 increases the potency of transplanted mesenchymal stem cells for myocardial repair. Circulation 2013; 128:S32-41. [PMID: 24030419 DOI: 10.1161/circulationaha.112.000025] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Thymosin β4 (Tβ4) has been shown to enhance the survival of cultured cardiomyocytes. Here, we investigated whether the cytoprotective effects of Tβ4 can increase the effectiveness of transplanted swine mesenchymal stem cells (sMSCs) for cardiac repair in a rat model of myocardial infarction (MI). METHODS AND RESULTS Under hypoxic conditions, cellular damage (lactate dehydrogenase leakage), apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labelingc cells), and caspase-8 activity were significantly lower, whereas B-cell lymphoma-extra large protein expression was significantly higher, in sMSCs cultured with Tβ4 (1 μg/mL) than in sMSCs cultured without Tβ4, and Tβ4 also increased sMSC proliferation. For in vivo experiments, animals were treated with basal medium (MI: n=6), a fibrin patch (Patch: n=6), a patch containing sMSCs (sMSC: n=9), or a patch containing sMSCs and Tβ4 (sMSC/Tβ4: n=11); Tβ4 was encapsulated in gelatin microspheres to extend Tβ4 delivery. Four weeks after treatment, echocardiographic assessments of left-ventricular ejection fraction and fractional shortening were significantly better (P<0.05) in sMSC/Tβ4 animals (left-ventricular ejection fraction=51.7 ± 1.1%; fractional shortening=26.7 ± 0.7%) than in animals from MI (39 ± 3%; 19.5 ± 1.7%) and Patch (43 ± 1.4%; 21.6 ± 0.9%) groups. Histological assessment of infarct wall thickness was significantly higher (P<0.05) in sMSC/Tβ4 animals (50%, [45%, 80%]) than in animals from MI (25%, [20%, 25%]) group. Measurements in sMSC (left-ventricular ejection fraction=45 ± 2.6%; fractional shortening=22.9 ± 1.6%; TH = 43% [25%, 45%]), Patch, and MI animals were similar. Tβ4 administration also significantly increased vascular growth, the retention/survival of the transplanted sMSCs, and the recruitment of endogenous c-Kit(+) progenitor cells to the infarcted region. CONCLUSIONS Extended-release Tβ4 administration improves the retention, survival, and regenerative potency of transplanted sMSCs after myocardial injury.
Collapse
Affiliation(s)
- Lei Ye
- Division of Cardiology, Department of Medicine (L.Y., P.Z., S.D., L.S., Q.X., J.Z.), Stem Cell Institute (L.Y., J.Z.), and Department of Biomedical Engineering (J.Z.), University of Minnesota, Minneapolis, MN
| | | | | | | | | | | |
Collapse
|
36
|
Don CW, Murry CE. Improving survival and efficacy of pluripotent stem cell-derived cardiac grafts. J Cell Mol Med 2013; 17:1355-62. [PMID: 24118766 PMCID: PMC4049630 DOI: 10.1111/jcmm.12147] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/28/2013] [Indexed: 01/02/2023] Open
Abstract
Human embryonic stem cells (hESCs) can be differentiated into structurally and electrically functional myocardial tissue and have the potential to regenerate large regions of infarcted myocardium. One of the key challenges that needs to be addressed towards full-scale clinical application of hESCs is enhancing survival of the transplanted cells within ischaemic or scarred, avascular host tissue. Shortly after transplantation, most hESCs are lost as a result of multiple mechanical, cellular and host factors, and a large proportion of the remaining cells undergo apoptosis or necrosis shortly thereafter, as a result of loss of adhesion-related signals, ischaemia, inflammation or immunological rejection. Blocking the apoptotic signalling pathways of the cells, using pro-survival cocktails, conditioning hESCs prior to transplant, promoting angiogenesis, immunosuppressing the host and using of bioengineered matrices are among the emerging techniques that have been shown to optimize cell survival. This review presents an overview of the current strategies for optimizing cell and host tissue to improve the survival and efficacy of cardiac cells derived from pluripotent stem cells.
Collapse
Affiliation(s)
- Creighton W Don
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
37
|
Heneidi S, Simerman AA, Keller E, Singh P, Li X, Dumesic DA, Chazenbalk G. Awakened by cellular stress: isolation and characterization of a novel population of pluripotent stem cells derived from human adipose tissue. PLoS One 2013; 8:e64752. [PMID: 23755141 PMCID: PMC3673968 DOI: 10.1371/journal.pone.0064752] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/17/2013] [Indexed: 12/15/2022] Open
Abstract
Advances in stem cell therapy face major clinical limitations, particularly challenged by low rates of post-transplant cell survival. Hostile host factors of the engraftment microenvironment such as hypoxia, nutrition deprivation, pro-inflammatory cytokines, and reactive oxygen species can each contribute to unwanted differentiation or apoptosis. In this report, we describe the isolation and characterization of a new population of adipose tissue (AT) derived pluripotent stem cells, termed Multilineage Differentiating Stress-Enduring (Muse) Cells, which are isolated using severe cellular stress conditions, including long-term exposure to the proteolytic enzyme collagenase, serum deprivation, low temperatures and hypoxia. Under these conditions, a highly purified population of Muse-AT cells is isolated without the utilization of cell sorting methods. Muse-AT cells grow in suspension as cell spheres reminiscent of embryonic stem cell clusters. Muse-AT cells are positive for the pluripotency markers SSEA3, TR-1-60, Oct3/4, Nanog and Sox2, and can spontaneously differentiate into mesenchymal, endodermal and ectodermal cell lineages with an efficiency of 23%, 20% and 22%, respectively. When using specific differentiation media, differentiation efficiency is greatly enhanced in Muse-AT cells (82% for mesenchymal, 75% for endodermal and 78% for ectodermal). When compared to adipose stem cells (ASCs), microarray data indicate a substantial up-regulation of Sox2, Oct3/4, and Rex1. Muse-ATs also exhibit gene expression patterns associated with the down-regulation of genes involved in cell death and survival, embryonic development, DNA replication and repair, cell cycle and potential factors related to oncogenecity. Gene expression analysis indicates that Muse-ATs and ASCs are mesenchymal in origin; however, Muse-ATs also express numerous lymphocytic and hematopoietic genes, such as CCR1 and CXCL2, encoding chemokine receptors and ligands involved in stem cell homing. Being highly resistant to severe cellular stress, Muse-AT cells have the potential to make a critical impact on the field of regenerative medicine and cell-based therapy.
Collapse
Affiliation(s)
- Saleh Heneidi
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Ariel A. Simerman
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
| | - Erica Keller
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
| | - Prapti Singh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
| | - Xinmin Li
- Clinical Microarray Core, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
| | - Daniel A. Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
| | - Gregorio Chazenbalk
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Gharaibeh B, Chun-Lansinger Y, Hagen T, Ingham SJM, Wright V, Fu F, Huard J. Biological approaches to improve skeletal muscle healing after injury and disease. ACTA ACUST UNITED AC 2012; 96:82-94. [PMID: 22457179 DOI: 10.1002/bdrc.21005] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Skeletal muscle injury and repair are complex processes, including well-coordinated steps of degeneration, inflammation, regeneration, and fibrosis. We have reviewed the recent literature including studies by our group that describe how to modulate the processes of skeletal muscle repair and regeneration. Antiinflammatory drugs that target cyclooxygenase-2 were found to hamper the skeletal muscle repair process. Muscle regeneration phase can be aided by growth factors, including insulin-like growth factor-1 and nerve growth factor, but these factors are typically short-lived, and thus more effective methods of delivery are needed. Skeletal muscle damage caused by traumatic injury or genetic diseases can benefit from cell therapy; however, the majority of transplanted muscle cells (myoblasts) are unable to survive the immune response and hypoxic conditions. Our group has isolated neonatal skeletal muscle derived stem cells (MDSCs) that appear to repair muscle tissue in a more effective manner than myoblasts, most likely due to their better resistance to oxidative stress. Enhancing antioxidant levels of MDSCs led to improved regenerative potential. It is becoming increasingly clear that stem cells tissue repair by direct differentiation and paracrine effects leading to neovascularization of injured site and chemoattraction of host cells. The factors invoked in paracrine action are still under investigation. Our group has found that angiotensin II receptor blocker (losartan) significantly reduces fibrotic tissue formation and improves repair of murine injured muscle. Based on these data, we have conducted a case study on two hamstring injury patients and found that losartan treatment was well tolerated and possibly improved recovery time. We believe this medication holds great promise to optimize muscle repair in humans.
Collapse
Affiliation(s)
- Burhan Gharaibeh
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Grabowska I, Brzoska E, Gawrysiak A, Streminska W, Moraczewski J, Polanski Z, Hoser G, Kawiak J, Machaj EK, Pojda Z, Ciemerych MA. Restricted Myogenic Potential of Mesenchymal Stromal Cells Isolated from Umbilical Cord. Cell Transplant 2012; 21:1711-26. [DOI: 10.3727/096368912x640493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Nonhematopoietic cord blood cells and mesenchymal cells of umbilical cord Wharton's jelly have been shown to be able to differentiate into various cell types. Thus, as they are readily available and do not raise any ethical issues, these cells are considered to be a potential source of material that can be used in regenerative medicine. In our previous study, we tested the potential of whole mononucleated fraction of human umbilical cord blood cells and showed that they are able to participate in the regeneration of injured mouse skeletal muscle. In the current study, we focused at the umbilical cord mesenchymal stromal cells isolated from Wharton's jelly. We documented that limited fraction of these cells express markers of pluripotent and myogenic cells. Moreover, they are able to undergo myogenic differentiation in vitro, as proved by coculture with C2C12 myoblasts. They also colonize injured skeletal muscle and, with low frequency, participate in the formation of new muscle fibers. Pretreatment of Wharton's jelly mesenchymal stromal cells with SDF-1 has no impact on their incorporation into regenerating muscle fibers but significantly increased muscle mass. As a result, transplantation of mesenchymal stromal cells enhances the skeletal muscle regeneration.
Collapse
Affiliation(s)
- Iwona Grabowska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Agnieszka Gawrysiak
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Wladyslawa Streminska
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Jerzy Moraczewski
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Zbigniew Polanski
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Grazyna Hoser
- Department of Clinical Cytology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Jerzy Kawiak
- Department of Clinical Cytology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Eugeniusz K. Machaj
- Department of Cellular Engineering, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Zygmunt Pojda
- Department of Cellular Engineering, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Maria A. Ciemerych
- Department of Cytology, Institute of Zoology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| |
Collapse
|
40
|
Dierickx P, Doevendans PA, Geijsen N, van Laake LW. Embryonic template-based generation and purification of pluripotent stem cell-derived cardiomyocytes for heart repair. J Cardiovasc Transl Res 2012; 5:566-80. [PMID: 22806916 DOI: 10.1007/s12265-012-9391-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/02/2012] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease remains a leading cause of death in Western countries. Many types of cardiovascular diseases are due to a loss of functional cardiomyocytes, which can result in irreversible cardiac failure. Since the adult human heart has limited regenerative potential, cardiac transplantation is still the only effective therapy to address this cardiomyocyte loss. However, drawbacks, such as immune rejection and insufficient donor availability, are limiting this last-resort solution. Recent developments in the stem cell biology field have improved the potential of cardiac regeneration. Improvements in reprogramming strategies of differentiated adult cells into induced pluripotent stem cells, together with increased efficiency of directed differentiation of pluripotent stem cells toward cardiac myocytes, have brought cell-based heart muscle regeneration a few steps closer to the clinic. In this review, we outline the status of research on cardiac regeneration with a focus on directed differentiation of pluripotent stem cells toward the cardiac lineage.
Collapse
Affiliation(s)
- Pieterjan Dierickx
- Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
41
|
Hoke NN, Salloum FN, Kass DA, Das A, Kukreja RC. Preconditioning by phosphodiesterase-5 inhibition improves therapeutic efficacy of adipose-derived stem cells following myocardial infarction in mice. Stem Cells 2012; 30:326-35. [PMID: 22102597 DOI: 10.1002/stem.789] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The rationale of this article is enhancing the therapeutic potential of stem cells in ischemic microenvironments by novel preconditioning strategies is critical for improving cellular therapy. We tested the hypothesis that inhibition of phosphodiesterase-5 (PDE-5) with sildenafil (Viagra) or knockdown with a silencing vector in adipose-derived stem cells (ASCs) would improve their survival and enhance cardiac function following myocardial implantation in vivo. ASCs were treated with sildenafil or PDE-5 silencing vector short hairpin RNA (shRNA(PDE-5)) and subjected to simulated ischemia/reoxygenation in vitro. Both sildenafil and shRNA(PDE-5) significantly improved viability, decreased necrosis, apoptosis, and enhanced the release of growth factors, vascular endothelial growth factor (VEGF), basic fibroblast growth factor (b-FGF), and insulin-like growth factor. Inhibition of protein kinase G reversed these effects. To show the beneficial effect of preconditioned ASCs in vivo, adult male CD-1 mice underwent myocardial infarction. Preconditioned ASCs (4 × 10(5)) were directly injected intramyocardially. Preconditioned ASC-treated hearts showed consistently superior cardiac function when compared with nonpreconditioned ASCs after 4 weeks of treatment. This was associated with significantly reduced fibrosis, increased vascular density, and decreased resident myocyte apoptosis when compared with mice receiving nonpreconditioned ASCs. VEGF, b-FGF, and Angiopoietin-1 were also significantly elevated 4 weeks after cell therapy with preconditioned ASCs. We conclude that preconditioning by inhibition of PDE-5 can be a powerful novel approach to improve stem cell therapy following myocardial infarction.
Collapse
Affiliation(s)
- Nicholas N Hoke
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | |
Collapse
|
42
|
Idris NM, Ashraf M, Ahmed RPH, Shujia J, Haider KH. Activation of IL-11/STAT3 pathway in preconditioned human skeletal myoblasts blocks apoptotic cascade under oxidant stress. Regen Med 2012; 7:47-57. [PMID: 22168497 DOI: 10.2217/rme.11.109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To determine whether our novel approach of diazoxide-induced stem cell preconditioning might be extrapolated to human skeletal myoblasts to support their survival under lethal oxidant stress. METHODS & RESULTS Using an in vitro model of H(2)O(2) treatment of human skeletal myoblasts, we report the ability of diazoxide-preconditioned human skeletal myoblasts to express cytokines and growth factors, which act in an autocrine and paracrine fashion to promote their own survival. Preconditioning of skeletal myoblasts was cytoprotective and significantly reduced their apoptotic index (p < 0.05). IL-11 gene and protein expression was significantly increased in preconditioned skeletal myoblasts. Transfection of skeletal myoblasts with IL-11-specific siRNA incurred their death under oxidant stress. The cytoprotective effect of diazoxide preconditioning was blocked by Erk1/2 inhibitor PD98059 (20-100 µM), which abrogated STAT-3 phosphorylation, thus confirming a possible involvement of Erk1/2/STAT3 signaling downstream of IL-11 in cell survival. We also investigated the time course of subcellular changes and signaling pathway of skeletal myoblasts apoptosis under oxidant stress before and after preconditioning. Apoptosis was induced in skeletal myoblasts with 100-500 µM H(2)O(2) for time points ranging from 1 to 24 h. Release of lactate dehydrogenase, disruption of the mitochondrial membrane potential and cytochrome-c translocation into cytoplasm were the earliest signs of apoptosis. Total Akt protein remained unchanged whereas marked reduction in pAkt was observed in the native skeletal myoblasts. Terminal dUTP nick end-labeling and annexin-V positivity were significantly increased after 4 h. Ultra-structure studies showed condensed chromatin, shriveled nuclei and swollen mitochondria. CONCLUSION These data suggest that skeletal myoblasts undergo apoptosis under oxidant stress in a time-dependent manner and preconditioning of skeletal myoblasts significantly prevented their apoptosis via IL-11/STAT3 signaling.
Collapse
Affiliation(s)
- Niagara Muhammad Idris
- Department of Pathology, 231 Albert Sabin Way, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | | | | | | |
Collapse
|
43
|
Bajek A, Drewa T, Joachimiak R, Marszałek A, Gagat M, Grzanka A. Stem cells for urinary tract regeneration. Cent European J Urol 2012; 65:7-10. [PMID: 24578913 PMCID: PMC3921771 DOI: 10.5173/ceju.2012.01.art2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 07/16/2011] [Accepted: 09/15/2011] [Indexed: 01/14/2023] Open
Abstract
Regeneration of the urinary bladder is a complicated task, due to organ dimensions and diseases (cancer, interstitial cystitis) when autologous bladder cells cannot be used. Cancer is the most frequent indication for bladder removal (cystectomy). Stem cells can be used with the guarantee of the sufficient cell number for the in vitro construction of the urinary bladder wall. Tissue engineering techniques hold great promise for regeneration of dysfunctional urinary sphincter. Denervation following surgical procedures or injuries results in weakness of the urethral sphincter and stress urinary incontinence. Injectable therapies and the potential of stem cells for sphincter restoration was presented in this review. The aim of this review was to present possibilities of urinary bladder regeneration with the use of stem cells and tissue engineering techniques.
Collapse
Affiliation(s)
- Anna Bajek
- Department of Tissue Engineering, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Drewa
- Department of Tissue Engineering, Nicolaus Copernicus University, Bydgoszcz, Poland ; Department of Urology, Institute of Oncology, Bydgoszcz, Poland
| | - Romana Joachimiak
- Department of Tissue Engineering, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Andrzej Marszałek
- Department of Clinical Pathomorphology, Nicolaus Copernicus University, Bydgoszcz, Poland ; Department of Clinical Pathomorphology, University of Medical Sciences, Poznań, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
44
|
Mohsin S, Siddiqi S, Collins B, Sussman MA. Empowering adult stem cells for myocardial regeneration. Circ Res 2012; 109:1415-28. [PMID: 22158649 DOI: 10.1161/circresaha.111.243071] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Treatment strategies for heart failure remain a high priority for ongoing research due to the profound unmet need in clinical disease coupled with lack of significant translational progress. The underlying issue is the same whether the cause is acute damage, chronic stress from disease, or aging: progressive loss of functional cardiomyocytes and diminished hemodynamic output. To stave off cardiomyocyte losses, a number of strategic approaches have been embraced in recent years involving both molecular and cellular approaches to augment myocardial structure and performance. Resultant excitement surrounding regenerative medicine in the heart has been tempered by realizations that reparative processes in the heart are insufficient to restore damaged myocardium to normal functional capacity and that cellular cardiomyoplasty is hampered by poor survival, proliferation, engraftment, and differentiation of the donated population. To overcome these limitations, a combination of molecular and cellular approaches must be adopted involving use of genetic engineering to enhance resistance to cell death and increase regenerative capacity. This review highlights biological properties of approached to potentiate stem cell-mediated regeneration to promote enhanced myocardial regeneration, persistence of donated cells, and long-lasting tissue repair. Optimizing cell delivery and harnessing the power of survival signaling cascades for ex vivo genetic modification of stem cells before reintroduction into the patient will be critical to enhance the efficacy of cellular cardiomyoplasty. Once this goal is achieved, then cell-based therapy has great promise for treatment of heart failure to combat the loss of cardiac structure and function associated with acute damage, chronic disease, or aging.
Collapse
|
45
|
Bartoszuk-Bruzzone U, Burdzińska A, Orzechowski A, Kłos Z. Protective effect of sodium ascorbate on efficacy of intramuscular transplantation of autologous muscle-derived cells. Muscle Nerve 2012; 45:32-8. [PMID: 22190303 DOI: 10.1002/mus.22248] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION The possible reason for elimination of myogenic cells after transplantation is inflammation at the injection site associated with oxidative stress. The aim of this study was to determine whether preconditioning of muscle-derived cells with an antioxidant, sodium ascorbate, can influence the fate of transplanted cells. METHODS Autologous transplantation of muscle-derived cells was performed in rabbits. Isolated cells were identified, lipofected with β-galactosidase, preincubated or not with sodium ascorbate, and injected intramuscularly. RESULTS Two weeks after autologous transplantation in the edge of a previous muscle defect, donor cells formed multinucleated young myotubes. Pretreatment of cells with sodium ascorbate before injection resulted in a significant increase of donor cells at the injection site 2 weeks after transfer. CONCLUSIONS These results show that: (1) preincubation with antioxidant can increase the efficacy of myogenic cell transplantation; and (2) oxidative stress may play a role in elimination of cells after autologous transplantation.
Collapse
|
46
|
Shao SX, Zhang L, Chen HX, Liu Y, Zhang JP, Chen W, Xue GY. Diazoxide pretreatment enhances L6 skeletal myoblast survival and inhibits apoptosis induced by hydrogen peroxide. Anat Rec (Hoboken) 2012; 295:632-40. [PMID: 22262406 DOI: 10.1002/ar.22410] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 11/17/2011] [Indexed: 11/09/2022]
Abstract
Skeletal myoblast (SKM) transplantation is a promising approach to regenerate tissue and improve the function of the injured heart. However, the number of survival cells transplanted to host myocardium is quite poor due to high rate of apoptosis; diazoxide (DZ) is a highly selective mito-KATP channel opener that may reduce cell apoptosis by relieving reactive oxygen species (ROS) damage. The aim of this study is to explore the protective effects of DZ on L6 SKM damage induced by hydrogen peroxide (H(2)O(2) ) in vitro. Different dose and time of H(2)O(2) and DZ treatment were performed and only 24 hr of 1.00 mmol/L H(2) O(2) treatment and 200 μmol/L DZ pretreatment for 30 min were used for further experiment. L6 SKMs were cultured and divided into control group (no treatment), H(2)O(2) group (24 hr of 1.00 mmol/L H(2) O(2) treatment) and DZ + H(2)O(2) group (pretreated with 200 μmol/L DZ for 30 min before 24 hr of 1.00 mmol/L H(2) O(2) treatment). Compared with control group, H(2)O(2) treatment caused cell damage, increased lactate dehydrogenase release, cell apoptosis, and bax gene expression, while reduced cell proliferation and decreased bcl-2 expression. DZ pretreatment may protect cells from damage induced by H(2)O(2) and reduce cell apoptosis by increasing bcl-2 and decreasing bax expression. DZ pretreatment may also promote cell proliferation measured by both PCNA expression and flow cytometry method. These results suggest that DZ may protect L6 SKMs from damage induced by H(2)O(2) by maintaining integrity of cell membrane, reducing apoptosis and increasing proliferation in vitro.
Collapse
Affiliation(s)
- Su-Xia Shao
- Department of Histology and Embryology, Hebei Medical University, Shijiazhuang City, Hebei Province, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
47
|
Haider KH, Ashraf M. Preconditioning approach in stem cell therapy for the treatment of infarcted heart. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:323-56. [PMID: 22917238 DOI: 10.1016/b978-0-12-398459-3.00015-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nearly two decades of research in regenerative medicine have been focused on the development of stem cells as a therapeutic option for treatment of the ischemic heart. Given the ability of stem cells to regenerate the damaged tissue, stem-cell-based therapy is an ideal approach for cardiovascular disorders. Preclinical studies in experimental animal models and clinical trials to determine the safety and efficacy of stem cell therapy have produced encouraging results that promise angiomyogenic repair of the ischemically damaged heart. Despite these promising results, stem cell therapy is still confronted with issues ranging from uncertainty about the as-yet-undetermined "ideal" donor cell type to the nonoptimized cell delivery strategies to harness optimal clinical benefits. Moreover, these lacunae have significantly hampered the progress of the heart cell therapy approach from bench to bedside for routine clinical applications. Massive death of donor cells in the infarcted myocardium during acute phase postengraftment is one of the areas of prime concern, which immensely lowers the efficacy of the procedure. An overview of the published data relevant to stem cell therapy is provided here and the various strategies that have been adopted to develop and optimize the protocols to enhance donor stem cell survival posttransplantation are discussed, with special focus on the preconditioning approach.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | |
Collapse
|
48
|
White JD, Grounds MD. Harnessing the therapeutic potential of myogenic stem cells. Cytotechnology 2011; 41:153-64. [PMID: 19002952 DOI: 10.1023/a:1024830924103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The potential clinical use of stem cells for cell transplantation therapies to replace defective genes in myopathies is an area of intense investigation. Precursor cells derived from non-muscle tissue with myogenic potential have been identified in many tissues, including bone marrow and dermis, although the status of these putative stem cells requires clarification. The incorporation of circulating bone-marrow derived stem cells into regenerating adult skeletal muscle has been demonstrated in mice but the contribution of donor cells is so minimal that it would appear clinically irrelevant at this stage. The possibility of a true stem cell subpopulation within skeletal muscle that replenishes the satellite cells (conventional muscle precursors on the surface of myofibres) is also very attractive as a superior source of myoblasts for muscle construction. A full understanding of the intrinsic factors (i.e. gene expression within the stem cell) and extrinsic factors (i.e. signals from the external environment) which control the commitment of stem cells to the myogenic lineage, and the conditions which favour stem cell expansion in vivo is required before stem cells can be seriously considered for clinical cell therapy.
Collapse
Affiliation(s)
- Jason D White
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Perth, Western Australia, Australia (Author for correspondence; E-mail,
| | | |
Collapse
|
49
|
Mingliang R, Bo Z, Zhengguo W. Stem cells for cardiac repair: status, mechanisms, and new strategies. Stem Cells Int 2011; 2011:310928. [PMID: 21776280 PMCID: PMC3137967 DOI: 10.4061/2011/310928] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/03/2011] [Accepted: 04/10/2011] [Indexed: 01/09/2023] Open
Abstract
Faced with the end stage of heart disease, the current treatments only slow worsening of heart failure. Stem cells have the potential of self-renewal and differentiation. It is expected to replace and repair damaged myocardium. But many clinical trials have shown that the stem cell therapy of heart failure is modest or not effective. The possible causes for the limited effects of stem cell in curing heart failure are the stem cells which have been transplanted into the ischemic heart muscle may suffer low survival rate, affected by inflammatory molecules, proapoptotic factor, and lack of nutrients and oxygen, and then the stem cells which home and have been completely transplanted to the site of myocardial infarction become very small. Therefore, through preconditioning of stem cells and appropriate choice of genes for mesenchymal stem cell modification to improve the survival rate of stem cells, ability in homing and promoting angiogenesis may become the newly effective strategies for the application of stem cells therapy in heart failure.
Collapse
Affiliation(s)
- Ren Mingliang
- Department 4, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | | | | |
Collapse
|
50
|
Overexpression of cellular repressor of E1A-stimulated genes inhibits TNF-α-induced apoptosis via NF-κB in mesenchymal stem cells. Biochem Biophys Res Commun 2011; 406:601-7. [DOI: 10.1016/j.bbrc.2011.02.100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/19/2011] [Indexed: 11/23/2022]
|