1
|
Zhang W, Wang K, Suo M, Liu X, Wang J, Liu X, Huang H, Chen S, Wang H, Chen X, Li Z. E7 peptide for in situ tissue engineering applications in bone repair. JOURNAL OF THE AMERICAN CERAMIC SOCIETY 2025. [DOI: 10.1111/jace.20524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/05/2025] [Indexed: 05/16/2025]
Abstract
AbstractIn regenerative medicine, leveraging bioactive molecules to enhance endogenous repair mechanisms represents a critical advancement. The E7 peptide, a novel short peptide, has emerged as a key candidate for bone defect repair due to its unique ability to interact with stem cells directly. Unlike traditional tissue‐engineered bone constructs that rely on exogenous cell delivery or scaffold‐based strategies, E7 enables in situ regeneration by actively recruiting and anchoring endogenous stem cells to the defect site. Studies demonstrate that E7‐functionalized materials significantly enhance stem cell proliferation and osteogenic differentiation while concurrently stimulating local angiogenesis through VEGF upregulation. These dual effects—stem cell homing and vascularization—address two major bottlenecks in bone repair: insufficient cell supply and poor nutrient diffusion in avascular regions. Despite these advantages, optimizing E7's spatiotemporal presentation and elucidating its signaling mechanisms remain critical. Further investigations into E7's receptor specificity, dose dependency, and long‐term safety will accelerate its clinical translation. It is of great guiding significance to clarify what role. E7 peptide plays in various bone repair materials and which pathways are activated for future research of bone defect repair.
Collapse
Affiliation(s)
- Wentao Zhang
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
- Department of Spine Surgery The Second Affiliated Hospital of Chongqing Medical University Chongqing People's Republic of China
| | - Kaizhong Wang
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Moran Suo
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Xiangyan Liu
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Jinzuo Wang
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Xin Liu
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Huagui Huang
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Shuang Chen
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Hui Wang
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| | - Xin Chen
- Faculty of Medicine Department of Orthopedics & Traumatology Musculoskeletal Research Laboratory The Chinese University of Hong Kong Hong Kong People's Republic of China
| | - Zhonghai Li
- Department of Orthopaedics First Affiliated Hospital of Dalian Medical University Dalian People's Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Diseases Shenyang Liaoning Province People's Republic of China
| |
Collapse
|
2
|
Zhong J, Li W, Li H, Zhang J, Hou Z, Wang X, Zhou E, Lu K, Zhuang W, Sang H. A self-forming bone membrane generated by periosteum-derived stem cell spheroids enhances the repair of bone defects. Acta Biomater 2025; 193:185-201. [PMID: 39742905 DOI: 10.1016/j.actbio.2024.12.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
The periosteum, a highly specialized thin tissue, is instrumental in contributing to as much as 70 % of early bone formation. Recognizing the periosteum's vital physiological roles, the fabrication of a biomimetic periosteum has risen as an auspicious strategy for addressing extensive bone defects. In the study, we obtained such biomimetic periosteum by utilizing periosteum-derived stem cells (PDSCs) spheroids. These spheroids are induced to spontaneously generate a bioactive membrane on a delicate 3D-printed polycaprolactone (PCL) substrate. This process yields a biomimetic periosteum rich in the resources needed for bone repair. The in vitro evaluations demonstrated that this membrane can act as a repository for growth factors and stem cells. The release kinetics confirmed a sustained delivery of BMP-2 and VEGF, which promoted enhanced osteogenesis and angiogenesis in vitro, respectively. The in vivo results further highlighted robust bone regeneration from critical cranial defects upon the application of this biomimetic periosteum. The biomimetic periosteum, easily harvested and potent in bioactivity, presents substantial clinical potential, particularly for the treatment of critical-sized bone defects. STATEMENT OF SIGNIFICANCE: PDSC theoretically demonstrates substantial potential in membrane construction, a value we've harnessed in this pioneering application. By employing cell spheroids, we've successfully integrated a substantial number of cells into the membrane framework. PDSC spheroids exhibit the remarkable ability to self-assemble into functional membranes, endowing them with robust biological capabilities that enhance their performance in biological systems. The in vitro evaluations demonstrated that this membrane can act as a repository for growth factors and stem cells. The in vivo bone repair facilitated by this membrane is notably effective, characterized by superior bone quality and accelerated formation rates. This process mirrors the natural intramembrane ossification, offering a promising approach to bone integration and regeneration.
Collapse
Affiliation(s)
- Jintao Zhong
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Wenhua Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Hetong Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Jin Zhang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| | - Zuoxu Hou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Xiao Wang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Enhui Zhou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Ke Lu
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Weida Zhuang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China.
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen 518000, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510000, PR China.
| |
Collapse
|
3
|
Thai NLB, Fittante E, Ma Z, Monroe MB. Rapid Fabrication of Polyvinyl Alcohol Hydrogel Foams With Encapsulated Mesenchymal Stem Cells for Chronic Wound Treatment. J Biomed Mater Res A 2025; 113:e37868. [PMID: 39794931 DOI: 10.1002/jbm.a.37868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025]
Abstract
Chronic wounds present a major healthcare challenge around the world, and significant hurdles remain in their effective treatment due to limitations in accessible treatment options. Mesenchymal stem cells (MSCs) with multifunctional differentiation and modulatory properties have been delivered to chronic wounds to enhance closure but have limited engraftment when delivered without a scaffold. In this study, hybrid porous hydrogel foams composed of modified polyvinyl alcohol and gelatin were developed that are suitable for rapid and facile MSC encapsulation, fully degradable, and supportive of wound healing. Rapid fabrication and encapsulation within porous foams was achieved using a cytocompatible gas blowing process. The hybrid hydrogels have tunable degradation rates based on chemistry, with complete mass loss achieved within 2-6 weeks, which is compatible with chronic wound closure rates. High encapsulated A375 epithelial cell and MSC viability with maintained cell functionality over 2 weeks reveals the potential of these hydrogels to serve as cell delivery systems for chronic wound treatment. An ex vivo porcine skin wound model demonstrated enhanced healing after application of cell-laden hydrogel foams. Overall, hybrid hydrogel foams with encapsulated therapeutic cells have the capacity for robust wound healing and are a promising platform for chronic wound dressings.
Collapse
Affiliation(s)
- Nghia Le Ba Thai
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Emily Fittante
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Zhen Ma
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| | - Mary Beth Monroe
- Biomedical and Chemical Engineering and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York, USA
| |
Collapse
|
4
|
Li Z, Wang Y, Wang H, Wang H, Shang Y, Wang S, Han Q, Li J, Zhao RC, Jiang Q, Ding B. Self-Assembled DNA Composite-Engineered Mesenchymal Stem Cells for Improved Skin-Wound Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310241. [PMID: 38441385 DOI: 10.1002/smll.202310241] [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: 11/09/2023] [Revised: 02/15/2024] [Indexed: 08/02/2024]
Abstract
The direct use of mesenchymal stem cells (MSCs) as therapeutics for skin injuries is a promising approach, yet it still faces several obstacles, including limited adhesion, retention, and engraftment of stem cells in the wound area, as well as impaired regenerative and healing functions. Here, DNA-based self-assembled composites are reported that can aid the adhesion of MSCs in skin wounds, enhance MSC viability, and accelerate wound closure and re-epithelialization. Rolling-circle amplification (RCA)-derived DNA flowers, equipped with multiple copies of cyclic Arg-Gly-Asp (cRGD) peptides and anti-von Willebrand factor (vWF) aptamers, act as robust scavengers of reactive oxygen species (ROS) and enable synergistic recognition and adhesion to stem cells and damaged vascular endothelial cells. These DNA structure-aided stem cells are retained at localized wound sites, maintain repair function, and promote angiogenesis and growth factor secretion. In both normal and diabetes-prone db/db mice models with excisional skin injuries, facile topical administration of DNA flower-MSCs elicits rapid blood vessel formation and enhances the sealing of the wound edges in a single dose. DNA composite-engineered stem cells warrant further exploration as a new strategy for the treatment of skin and tissue damage.
Collapse
Affiliation(s)
- Zhuoting Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Yiming Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Hong Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Haiyan Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Yingxu Shang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Shihua Wang
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, 100005, China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
5
|
Quezada M, Ponce C, Berríos‐Cárcamo P, Santapau D, Gallardo J, De Gregorio C, Quintanilla ME, Morales P, Ezquer M, Herrera‐Marschitz M, Israel Y, Andrés‐Herrera P, Hipólito L, Ezquer F. Amelioration of morphine withdrawal syndrome by systemic and intranasal administration of mesenchymal stem cell-derived secretome in preclinical models of morphine dependence. CNS Neurosci Ther 2024; 30:e14517. [PMID: 37927136 PMCID: PMC11017443 DOI: 10.1111/cns.14517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/21/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Morphine is an opiate commonly used in the treatment of moderate to severe pain. However, prolonged administration can lead to physical dependence and strong withdrawal symptoms upon cessation of morphine use. These symptoms can include anxiety, irritability, increased heart rate, and muscle cramps, which strongly promote morphine use relapse. The morphine-induced increases in neuroinflammation, brain oxidative stress, and alteration of glutamate levels in the hippocampus and nucleus accumbens have been associated with morphine dependence and a higher severity of withdrawal symptoms. Due to its rich content in potent anti-inflammatory and antioxidant factors, secretome derived from human mesenchymal stem cells (hMSCs) is proposed as a preclinical therapeutic tool for the treatment of this complex neurological condition associated with neuroinflammation and brain oxidative stress. METHODS Two animal models of morphine dependence were used to evaluate the therapeutic efficacy of hMSC-derived secretome in reducing morphine withdrawal signs. In the first model, rats were implanted subcutaneously with mini-pumps which released morphine at a concentration of 10 mg/kg/day for seven days. Three days after pump implantation, animals were treated with a simultaneous intravenous and intranasal administration of hMSC-derived secretome or vehicle, and withdrawal signs were precipitated on day seven by i.p. naloxone administration. In this model, brain alterations associated with withdrawal were also analyzed before withdrawal precipitation. In the second animal model, rats voluntarily consuming morphine for three weeks were intravenously and intranasally treated with hMSC-derived secretome or vehicle, and withdrawal signs were induced by morphine deprivation. RESULTS In both animal models secretome administration induced a significant reduction of withdrawal signs, as shown by a reduction in a combined withdrawal score. Secretome administration also promoted a reduction in morphine-induced neuroinflammation in the hippocampus and nucleus accumbens, while no changes were observed in extracellular glutamate levels in the nucleus accumbens. CONCLUSION Data presented from two animal models of morphine dependence suggest that administration of secretome derived from hMSCs reduces the development of opioid withdrawal signs, which correlates with a reduction in neuroinflammation in the hippocampus and nucleus accumbens.
Collapse
Affiliation(s)
- Mauricio Quezada
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - Carolina Ponce
- Department of Neuroscience, Faculty of MedicineUniversidad de ChileSantiagoChile
| | - Pablo Berríos‐Cárcamo
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - Daniela Santapau
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - Javiera Gallardo
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - Cristian De Gregorio
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of MedicineUniversidad de ChileSantiagoChile
| | - Paola Morales
- Department of Neuroscience, Faculty of MedicineUniversidad de ChileSantiagoChile
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of MedicineUniversidad de ChileSantiagoChile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
| | - Mario Herrera‐Marschitz
- Department of Neuroscience, Faculty of MedicineUniversidad de ChileSantiagoChile
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of MedicineUniversidad de ChileSantiagoChile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Science, Faculty of MedicineUniversidad de ChileSantiagoChile
| | - Paula Andrés‐Herrera
- Department of Pharmacy and Pharmaceutical Technology and ParasitologyUniversity of ValenciaValenciaSpain
- University Institute of Biotechnology and Biomedicine (BIOTECMED)University of ValenciaValenciaSpain
| | - Lucia Hipólito
- Department of Pharmacy and Pharmaceutical Technology and ParasitologyUniversity of ValenciaValenciaSpain
- University Institute of Biotechnology and Biomedicine (BIOTECMED)University of ValenciaValenciaSpain
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of MedicineClínica Alemana‐Universidad del DesarrolloSantiagoChile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use DisordersSantiagoChile
| |
Collapse
|
6
|
Venkatakrishnan J, Yuan Y, Zhang J, Yu Y, Hu YC, Kao WWY. Self-complementary AAV vector therapy for treating corneal cloudiness of mucopolysaccharidosis type VII (MPS VII). Ocul Surf 2024; 32:39-47. [PMID: 38218582 DOI: 10.1016/j.jtos.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/26/2023] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
PURPOSE To design a novel efficacious scAAV-Gusb viral vector for treating Mucopolysaccharidosis Type VII (MPS VII) caused by a mutation in the β-Glu gene (Gusb allele). METHODS β-Glu expression of single-stranded AAV-Gusb (ssAAV-Gusb) and self-complementary AAV (scAAV-Gusb) vectors are tested with cultured murine Gusb fibroblasts. The scAAV-Gusb vector was chosen in further studies to prolong the life span and treat corneal pathology of Gusb mice via intrahepatic injection of neonates and intrastromal injection in adults, respectively. Corneal pathology was studied using HRT2 in vivo confocal microscope and histochemistry in mice corneas. RESULTS Both ssAAV-Gusb and scAAV-Gusb vectors expressed murine β-Glu in cultured Gusb fibroblasts. The scAAV-Gusb vector had higher transduction efficiency than the ssAAV-Gusb vector. To prolong the life span of Gusb mice, neonates (3 days old) were administered with scAAV-Gusb virus via intrahepatic injection. The treatment improves the survival rate of Gusb mice, prolonging the median survival rate from 22.5 weeks (untreated) to 50 weeks (treated). Thereafter, we determined the efficacy of the scAAV-Gusb virus in ameliorating corneal cloudiness observed in aged Gusb mice. Both corneal cloudiness and stroma thickness decreased, and there was the presence of β-Glu enzyme activity in the Gusb corneas receiving scAAV-Gusb virus associated with morphology change of amoeboid stromal cells in untreated to characteristic dendritic keratocytes morphology after 4-12 weeks of scAAV-Gusb virus injection. CONCLUSION Intrahepatic injection of scAAV-Gusb is efficacious in prolonging the life span of Gusb mice, and intrastromal injection can ameliorate corneal phenotypes. Both strategies can be adapted for treating other MPS.
Collapse
Affiliation(s)
- Jhuwala Venkatakrishnan
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA; Department of Biomedical Engineering, University of Cincinnati, OH, USA
| | - Yong Yuan
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Jianhua Zhang
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA
| | - Yang Yu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, OH, USA
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, OH, USA
| | - Winston W-Y Kao
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
7
|
Zheng J, Lu Y, Lin Y, Si S, Guo B, Zhao X, Cui L. Epitranscriptomic modifications in mesenchymal stem cell differentiation: advances, mechanistic insights, and beyond. Cell Death Differ 2024; 31:9-27. [PMID: 37985811 PMCID: PMC10782030 DOI: 10.1038/s41418-023-01238-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/24/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
RNA modifications, known as the "epitranscriptome", represent a key layer of regulation that influences a wide array of biological processes in mesenchymal stem cells (MSCs). These modifications, catalyzed by specific enzymes, often termed "writers", "readers", and "erasers", can dynamically alter the MSCs' transcriptomic landscape, thereby modulating cell differentiation, proliferation, and responses to environmental cues. These enzymes include members of the classes METTL, IGF2BP, WTAP, YTHD, FTO, NAT, and others. Many of these RNA-modifying agents are active during MSC lineage differentiation. This review provides a comprehensive overview of the current understanding of different RNA modifications in MSCs, their roles in regulating stem cell behavior, and their implications in MSC-based therapies. It delves into how RNA modifications impact MSC biology, the functional significance of individual modifications, and the complex interplay among these modifications. We further discuss how these intricate regulatory mechanisms contribute to the functional diversity of MSCs, and how they might be harnessed for therapeutic applications. The review also highlights current challenges and potential future directions in the study of RNA modifications in MSCs, emphasizing the need for innovative tools to precisely map these modifications and decipher their context-specific effects. Collectively, this work paves the way for a deeper understanding of the role of the epitranscriptome in MSC biology, potentially advancing therapeutic strategies in regenerative medicine and MSC-based therapies.
Collapse
Affiliation(s)
- Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Shanshan Si
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, 90095, CA, USA.
| |
Collapse
|
8
|
Sun W, Lv J, Guo S, Lv M. Cellular microenvironment: a key for tuning mesenchymal stem cell senescence. Front Cell Dev Biol 2023; 11:1323678. [PMID: 38111850 PMCID: PMC10725964 DOI: 10.3389/fcell.2023.1323678] [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: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess the ability to self-renew and differentiate into multiple cell types, making them highly suitable for use as seed cells in tissue engineering. These can be derived from various sources and have been found to play crucial roles in several physiological processes, such as tissue repair, immune regulation, and intercellular communication. However, the limited capacity for cell proliferation and the secretion of senescence-associated secreted phenotypes (SASPs) pose challenges for the clinical application of MSCs. In this review, we provide a comprehensive summary of the senescence characteristics of MSCs and examine the different features of cellular microenvironments studied thus far. Additionally, we discuss the mechanisms by which cellular microenvironments regulate the senescence process of MSCs, offering insights into preserving their functionality and enhancing their effectiveness.
Collapse
Affiliation(s)
| | | | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Chouaib B, Haack-Sørensen M, Chaubron F, Cuisinier F, Collart-Dutilleul PY. Towards the Standardization of Mesenchymal Stem Cell Secretome-Derived Product Manufacturing for Tissue Regeneration. Int J Mol Sci 2023; 24:12594. [PMID: 37628774 PMCID: PMC10454619 DOI: 10.3390/ijms241612594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/29/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cell secretome or conditioned medium (MSC-CM) is a combination of biomolecules and growth factors in cell culture growth medium, secreted by mesenchymal stem cells (MSCs), and the starting point of several derived products. MSC-CM and its derivatives could be applied after injuries and could mediate most of the beneficial regenerative effects of MSCs without the possible side effects of using MSCs themselves. However, before the clinical application of these promising biopharmaceuticals, several issues such as manufacturing protocols and quality control must be addressed. This review aims to underline the influence of the procedure for conditioned medium production on the quality of the secretome and its derivatives and highlights the questions considering cell sources and donors, cell expansion, cell passage number and confluency, conditioning period, cell culture medium, microenvironment cues, and secretome-derived product purification. A high degree of variability in MSC secretomes is revealed based on these parameters, confirming the need to standardize and optimize protocols. Understanding how bioprocessing and manufacturing conditions interact to determine the quantity, quality, and profile of MSC-CM is essential to the development of good manufacturing practice (GMP)-compliant procedures suitable for replacing mesenchymal stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Batoul Chouaib
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, 92262 Fontenay-aux-Roses, France
| | - Mandana Haack-Sørensen
- Cardiology Stem Cell Centre 9302, Rigshospitalet University of Copenhagen, Henrik Harpestrengsvej 4C, 2100 Copenhagen, Denmark
| | - Franck Chaubron
- Institut Clinident BioPharma, Biopôle Clermont-Limagne, 63360 Saint Beauzire, France;
| | - Frederic Cuisinier
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Faculty of Dentistry, University of Montpellier, 34000 Montpellier, France
- Service Odontologie, CHU Montpellier, 34000 Montpellier, France
| | - Pierre-Yves Collart-Dutilleul
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Faculty of Dentistry, University of Montpellier, 34000 Montpellier, France
- Service Odontologie, CHU Montpellier, 34000 Montpellier, France
| |
Collapse
|
10
|
Liang J, Liu P, Yang X, Liu L, Zhang Y, Wang Q, Zhao H. Biomaterial-based scaffolds in promotion of cartilage regeneration: Recent advances and emerging applications. J Orthop Translat 2023; 41:54-62. [PMID: 37691640 PMCID: PMC10485599 DOI: 10.1016/j.jot.2023.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 08/05/2023] [Indexed: 09/12/2023] Open
Abstract
Osteoarthritis (OA) poses a significant burden for countless individuals, inflicting relentless pain and impairing their quality of life. Although traditional treatments for OA focus on pain management and surgical interventions, they often fall short of addressing the underlying cause of the disease. Fortunately, emerging biomaterial-based scaffolds offer hope for OA therapy, providing immense promise for cartilage regeneration in OA. These innovative scaffolds are ingeniously designed to provide support and mimic the intricate structure of the natural extracellular matrix, thus stimulating the regeneration of damaged cartilage. In this comprehensive review, we summarize and discuss current landscape of biomaterial-based scaffolds for cartilage regeneration in OA. Furthermore, we delve into the diverse range of biomaterials employed in their construction and explore the cutting-edge techniques utilized in their fabrication. By examining both preclinical and clinical studies, we aim to illuminate the remarkable versatility and untapped potential of biomaterial-based scaffolds in the context of OA. Thetranslational potential of this article By thoroughly examining the current state of research and clinical studies, this review provides valuable insights that bridge the gap between scientific knowledge and practical application. This knowledge is crucial for clinicians and researchers who strive to develop innovative treatments that go beyond symptom management and directly target the underlying cause of OA. Through the comprehensive analysis and multidisciplinary approach, the review paves the way for the translation of scientific knowledge into practical applications, ultimately improving the lives of individuals suffering from OA and shaping the future of orthopedic medicine.
Collapse
Affiliation(s)
| | | | - Xinquan Yang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Liu
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiong Wang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
11
|
Borkowska P, Morys J, Zielinska A, Kowalski J. Effects of the Co-Overexpression of the BCL and BDNF Genes on the Gamma-Aminobutyric Acid-Ergic Differentiation of Wharton's-Jelly-Derived Mesenchymal Stem Cells. Biomedicines 2023; 11:1751. [PMID: 37371846 DOI: 10.3390/biomedicines11061751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
One of the problems with using MSCs (mesenchymal stem cells) to treat different neurodegenerative diseases of the central nervous system is their low ability to spontaneously differentiate into functional neurons. The aim of this study was to investigate how the co-overexpression of the BCL and BDNF genes affects the ability of genetically modified MSCs to differentiate into GABA-ergic neurons. A co-overexpression of two genes was performed, one of which, BCL, was supposed to increase the resistance of the cells to the toxic agents in the brain environment. The second one, BDNF, was supposed to direct the cells onto the neuronal differentiation pathway. As a result, the co-overexpression of both BCL2 + BDNF and BCLXL + BDNF caused an increase in the MAP2 gene expression level (a marker of the neuronal pathway) and the SYP gene that is associated with synaptogenesis. In both cases, approximately 18% of the genetically modified and then differentiated cells exhibited the presence of the GAD protein, which is characteristic of GABA-ergic neurons. Despite the presence of GAD, after both modifications, only the BCL2 and BDNF co-overexpression correlated with the ability of the modified cells to release gamma-aminobutyric acid (GABA) after depolarization. Our study identified a novel model of genetically engineered MSCs that can be used as a tool to deliver the antiapoptotic proteins (BCL) and neurotrophic factor (BDNF) directly into the brain microenvironment. Additionally, in the investigated model, the genetically modified MSCs could easily differentiate into functional GABA-ergic neurons and, moreover, due to the secreted BCL and BDNF, promote endogenous neuronal growth and encourage synaptic connections between neurons.
Collapse
Affiliation(s)
- Paulina Borkowska
- Department of Medical Genetics, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Julia Morys
- Department of Medical Genetics, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Aleksandra Zielinska
- Department of Medical Genetics, Medical University of Silesia, 41-200 Sosnowiec, Poland
| | - Jan Kowalski
- Department of Medical Genetics, Medical University of Silesia, 41-200 Sosnowiec, Poland
| |
Collapse
|
12
|
Valsecchi C, Croce S, Lenta E, Acquafredda G, Comoli P, Avanzini MA. TITLE: New therapeutic approaches in pediatric diseases: Mesenchymal stromal cell and mesenchymal stromal cell-derived extracellular vesicles as new drugs. Pharmacol Res 2023; 192:106796. [PMID: 37207738 DOI: 10.1016/j.phrs.2023.106796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Mesenchymal Stromal Cell (MSC) clinical applications have been widely reported and their therapeutic potential has been documented in several diseases. MSCs can be isolated from several human tissues and easily expanded in vitro, they are able to differentiate in a variety of cell lineages, and they are known to interact with most immunological cells, showing immunosuppressive and tissue repair properties. Their therapeutic efficacy is closely associated with the release of bioactive molecules, namely Extracellular Vesicles (EVs), effective as their parental cells. EVs isolated from MSCs act by fusing with target cell membrane and releasing their content, showing a great potential for the treatment of injured tissues and organs, and for the modulation of the host immune system. EV-based therapies provide, as major advantages, the possibility to cross the epithelium and blood barrier and their activity is not influenced by the surrounding environment. In the present review, we deal with pre-clinical reports and clinical trials to provide data in support of MSC and EV clinical efficacy with particular focus on neonatal and pediatric diseases. Considering pre-clinical and clinical data so far available, it is likely that cell-based and cell-free therapies could become an important therapeutic approach for the treatment of several pediatric diseases.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Stefania Croce
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Elisa Lenta
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Gloria Acquafredda
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Patrizia Comoli
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| |
Collapse
|
13
|
Brown SV, Dewitt S, Clayton A, Waddington RJ. Identifying the Efficacy of Extracellular Vesicles in Osteogenic Differentiation: An EV-Lution in Regenerative Medicine. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.849724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have long been the focus for regenerative medicine and the restoration of damaged or aging cells throughout the body. However, the efficacy of MSCs in cell-based therapy still remains unpredictable and carries with it enumerable risks. It is estimated that only 3-10% of MSCs survive transplantation, and there remains undefined and highly variable heterogeneous biological potency within these administered cell populations. The mode of action points to secreted factors produced by MSCs rather than the reliance on engraftment. Hence harnessing such secreted elements as a replacement for live-cell therapies is attractive. Extracellular vesicles (EVs) are heterogenous lipid bounded structures, secreted by cells. They comprise a complex repertoire of molecules including RNA, proteins and other factors that facilitate cell-to-cell communication. Described as protected signaling centers, EVs can modify the cellular activity of recipient cells and are emerging as a credible alternative to cell-based therapies. EV therapeutics demonstrate beneficial roles for wound healing by preventing apoptosis, moderating immune responses, and stimulating angiogenesis, in addition to promoting cell proliferation and differentiation required for tissue matrix synthesis. Significantly, EVs maintain their signaling function following transplantation, circumventing the issues related to cell-based therapies. However, EV research is still in its infancy in terms of their utility as medicinal agents, with many questions still surrounding mechanistic understanding, optimal sourcing, and isolation of EVs for regenerative medicine. This review will consider the efficacy of using cell-derived EVs compared to traditional cell-based therapies for bone repair and regeneration. We discuss the factors to consider in developing productive lines of inquiry and establishment of standardized protocols so that EVs can be harnessed from optimal secretome production, to deliver reproducible and effective therapies.
Collapse
|
14
|
Shang Z, Wang R, Li D, Chen J, Zhang B, Wang M, Wang X, Wanyan P. Spinal Cord Injury: A Systematic Review and Network Meta-Analysis of Therapeutic Strategies Based on 15 Types of Stem Cells in Animal Models. Front Pharmacol 2022; 13:819861. [PMID: 35359872 PMCID: PMC8964098 DOI: 10.3389/fphar.2022.819861] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Objective: The optimal therapeutic strategies of stem cells for spinal cord injury (SCI) are fully explored in animal studies to promote the translation of preclinical findings to clinical practice, also to provide guidance for future animal experiments and clinical studies. Methods: PubMed, Web of Science, Embase, CNKI, Wangfang, VIP, and CBM were searched from inception to September 2021. Screening of search results, data extraction, and references quality evaluation were undertaken independently by two reviewers. Results and Discussion: A total of 188 studies were included for data analysis. Results of traditional meta-analysis showed that all 15 diverse types of stem cells could significantly improve locomotor function of animals with SCI, and results of further network meta-analysis showed that adipose-derived mesenchymal stem cells had the greatest therapeutic potential for SCI. Moreover, a higher dose (≥1 × 106) of stem cell transplantation had better therapeutic effect, transplantation in the subacute phase (3–14 days, excluding 3 days) was the optimal timing, and intralesional transplantation was the optimal route. However, the evidence of current animal studies is of limited quality, and more high-quality research is needed to further explore the optimal therapeutic strategies of stem cells, while the design and implementation of experiments, as well as measurement and reporting of results for animal studies, need to be further improved and standardized to reduce the risk when the results of animal studies are translated to the clinic. Systematic Review Registration: [website], identifier [registration number].
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ruirui Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Dongliang Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jinlei Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Xin Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, China
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| |
Collapse
|
15
|
Deng P, Halmai JANM, Beitnere U, Cameron D, Martinez ML, Lee CC, Waldo JJ, Thongphanh K, Adhikari A, Copping N, Petkova SP, Lee RD, Lock S, Palomares M, O’Geen H, Carter J, Gonzalez CE, Buchanan FKB, Anderson JD, Fierro FA, Nolta JA, Tarantal AF, Silverman JL, Segal DJ, Fink KD. An in vivo Cell-Based Delivery Platform for Zinc Finger Artificial Transcription Factors in Pre-clinical Animal Models. Front Mol Neurosci 2022; 14:789913. [PMID: 35153670 PMCID: PMC8829036 DOI: 10.3389/fnmol.2021.789913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/01/2021] [Indexed: 11/28/2022] Open
Abstract
Zinc finger (ZF), transcription activator-like effectors (TALE), and CRISPR/Cas9 therapies to regulate gene expression are becoming viable strategies to treat genetic disorders, although effective in vivo delivery systems for these proteins remain a major translational hurdle. We describe the use of a mesenchymal stem/stromal cell (MSC)-based delivery system for the secretion of a ZF protein (ZF-MSC) in transgenic mouse models and young rhesus monkeys. Secreted ZF protein from mouse ZF-MSC was detectable within the hippocampus 1 week following intracranial or cisterna magna (CM) injection. Secreted ZF activated the imprinted paternal Ube3a in a transgenic reporter mouse and ameliorated motor deficits in a Ube3a deletion Angelman Syndrome (AS) mouse. Intrathecally administered autologous rhesus MSCs were well-tolerated for 3 weeks following administration and secreted ZF protein was detectable within the cerebrospinal fluid (CSF), midbrain, and spinal cord. This approach is less invasive when compared to direct intracranial injection which requires a surgical procedure.
Collapse
Affiliation(s)
- Peter Deng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Julian A. N. M. Halmai
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ulrika Beitnere
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - David Cameron
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Michele L. Martinez
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Charles C. Lee
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Jennifer J. Waldo
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Krista Thongphanh
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Anna Adhikari
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Nycole Copping
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Stela P. Petkova
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ruth D. Lee
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Samantha Lock
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Miranda Palomares
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Henriette O’Geen
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Jasmine Carter
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Casiana E. Gonzalez
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Fiona K. B. Buchanan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Johnathan D. Anderson
- Department of Otolaryngology, University of California, Davis, Davis, CA, United States
| | - Fernando A. Fierro
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Jan A. Nolta
- Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States
| | - Alice F. Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, Gene Therapy Center, and California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Jill L. Silverman
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States
| | - David J. Segal
- Department of Biochemistry and Molecular Medicine, Genome Center, University of California, Davis, Davis, CA, United States
| | - Kyle D. Fink
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States,Stem Cell Program and Gene Therapy Center, University of California, Davis, Sacramento, CA, United States,Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California Davis School of Medicine, Sacramento, CA, United States,*Correspondence: Kyle D. Fink,
| |
Collapse
|
16
|
Jeong G, Im G, Lee T, Kim S, Jeon HR, Lee D, Baik S, Pang C, Kim T, Kim D, Jang YC, Bhang SH. Development of a stem cell spheroid‐laden patch with high retention at skin wound site. Bioeng Transl Med 2021; 7:e10279. [PMID: 35600658 PMCID: PMC9115685 DOI: 10.1002/btm2.10279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/19/2021] [Accepted: 12/09/2021] [Indexed: 11/08/2022] Open
Abstract
Mesenchymal stem cells such as human adipose tissue‐derived stem cells (hADSCs) have been used as a representative therapeutic agent for tissue regeneration because of their high proliferation and paracrine factor‐secreting abilities. However, certain points regarding conventional ADSC delivery systems, such as low cell density, secreted cytokine levels, and cell viability, still need to be addressed for treating severe wounds. In this study, we developed a three‐dimensional (3D) cavity‐structured stem cell‐laden system for overdense delivery of cells into severe wound sites. Our system includes a hydrophobic surface and cavities that can enhance the efficiency of cell delivery to the wound site. In particular, the cavities in the system facilitate hADSC spheroid formation, increasing therapeutic growth factor expression compared with 2D cultured cells. Our hADSC spheroid‐loaded patch exhibited remarkably improved cell localization at the wound site and dramatic therapeutic efficacy compared to the conventional cell injection method. Taken together, the hADSC spheroid delivery system focused on cell delivery, and stem cell homing effect at the wound site showed a significantly enhanced wound healing effect. By overcoming the limitations of conventional cell delivery methods, our overdense cell delivery system can contribute to biomedical and clinical applications.
Collapse
Affiliation(s)
- Gun‐Jae Jeong
- School of Biological Sciences Georgia Institute of Technology Atlanta Georgia USA
| | - Gwang‐Bum Im
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| | - Tae‐Jin Lee
- Department of Bio‐Health Convergence Kangwon National University Chuncheon Gwangwon South Korea
- Department of Medical Biotechnology, School of Biomedical Science Kangwon National University Chuncheon South Korea
| | - Sung‐Won Kim
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| | - Hye Ran Jeon
- Division of Vascular Surgery, Samsung Medical Center School of Medicine, Sungkyunkwan University Seoul South Korea
| | - Dong‐Hyun Lee
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| | - Sangyul Baik
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| | - Changhyun Pang
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| | - Tae‐Hyung Kim
- School of Integrative Engineering Chung‐Ang University Seoul South Korea
| | - Dong‐Ik Kim
- Division of Vascular Surgery, Samsung Medical Center School of Medicine, Sungkyunkwan University Seoul South Korea
| | - Young Charles Jang
- School of Biological Sciences Georgia Institute of Technology Atlanta Georgia USA
| | - Suk Ho Bhang
- School of Chemical Engineering Sungkyunkwan University Suwon South Korea
| |
Collapse
|
17
|
Raghav PK, Mann Z, Ahlawat S, Mohanty S. Mesenchymal stem cell-based nanoparticles and scaffolds in regenerative medicine. Eur J Pharmacol 2021; 918:174657. [PMID: 34871557 DOI: 10.1016/j.ejphar.2021.174657] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/05/2021] [Accepted: 11/24/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells owing to their regenerative potential and multilineage potency. MSCs have wide-scale applications either in their native cellular form or in conjugation with specific biomaterials as nanocomposites. Majorly, these natural or synthetic biomaterials are being used in the form of metallic and non-metallic nanoparticles (NPs) to encapsulate MSCs within hydrogels like alginate or chitosan or drug cargo loading into MSCs. In contrast, nanofibers of polymer scaffolds such as polycaprolactone (PCL), poly-lactic-co-glycolic acid (PLGA), poly-L-lactic acid (PLLA), silk fibroin, collagen, chitosan, alginate, hyaluronic acid (HA), and cellulose are used to support or grow MSCs directly on it. These MSCs based nanotherapies have application in multiple domains of biomedicine including wound healing, bone and cartilage engineering, cardiac disorders, and neurological disorders. This study focused on current approaches of MSCs-based therapies and has been divided into two major sections. The first section elaborates on MSC-based nano-therapies and their plausible applications including exosome engineering and NPs encapsulation. The following section focuses on the various MSC-based scaffold approaches in tissue engineering. Conclusively, this review mainly focused on MSC-based nanocomposite's current approaches and compared their advantages and limitations for building effective regenerative medicines.
Collapse
Affiliation(s)
- Pawan Kumar Raghav
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Zoya Mann
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Swati Ahlawat
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Sujata Mohanty
- Stem Cell Facility, DBT Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
18
|
Assunção Silva RC, Pinto L, Salgado AJ. Cell transplantation and secretome based approaches in spinal cord injury regenerative medicine. Med Res Rev 2021; 42:850-896. [PMID: 34783046 DOI: 10.1002/med.21865] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/12/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
The axonal growth-restrictive character of traumatic spinal cord injury (SCI) makes finding a therapeutic strategy a very demanding task, due to the postinjury events impeditive to spontaneous axonal outgrowth and regeneration. Considering SCI pathophysiology complexity, it has been suggested that an effective therapy should tackle all the SCI-related aspects and provide sensory and motor improvement to SCI patients. Thus, the current aim of any therapeutic approach for SCI relies in providing neuroprotection and support neuroregeneration. Acknowledging the current SCI treatment paradigm, cell transplantation is one of the most explored approaches for SCI with mesenchymal stem cells (MSCs) being in the forefront of many of these. Studies showing the beneficial effects of MSC transplantation after SCI have been proposing a paracrine action of these cells on the injured tissues, through the secretion of protective and trophic factors, rather than attributing it to the action of cells itself. This manuscript provides detailed information on the most recent data regarding the neuroregenerative effect of the secretome of MSCs as a cell-free based therapy for SCI. The main challenge of any strategy proposed for SCI treatment relies in obtaining robust preclinical evidence from in vitro and in vivo models, before moving to the clinics, so we have specifically focused on the available vertebrate and mammal models of SCI currently used in research and how can SCI field benefit from them.
Collapse
Affiliation(s)
- Rita C Assunção Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
19
|
Zhao Y, Yang X, Li S, Zhang B, Li S, Wang X, Wang Y, Jia C, Chang Y, Wei W. sTNFRII-Fc modification protects human UC-MSCs against apoptosis/autophagy induced by TNF-α and enhances their efficacy in alleviating inflammatory arthritis. Stem Cell Res Ther 2021; 12:535. [PMID: 34627365 PMCID: PMC8502322 DOI: 10.1186/s13287-021-02602-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tumor necrosis factor (TNF)-α inhibitors represented by Etanercept (a fusion protein containing soluble TNF receptor II (sTNFRII) and the Fc segment of human IgG1) play a pivotal role in Rheumatoid arthritis (RA) treatment. However, long-term use increases the risk of infection and tumors for their systemic inhibition of TNF-α, which disrupts the regular physiological function of this molecular. Mesenchymal stem cells (MSCs)-based delivery system provides new options for RA treatment with their "homing" and immune-regulation capacities, whereas inflammatory environment (especially TNF-α) is not conducive to MSCs' therapeutic effects by inducing apoptosis/autophagy. Here, we constructed a strain of sTNFRII-Fc-expressing MSCs (sTNFRII-MSC), aiming to offset the deficiency of those two interventions. METHODS Constructed sTNFRII-Fc lentiviral vector was used to infect human umbilical cord-derived MSCs, and sTNFRII-MSC stable cell line was generated by monoclonal cultivation. In vitro and vivo characteristics of sTNFRII-MSC were assessed by coculture assay and an acute inflammatory model in NOD/SCID mice. The sTNFRII-MSC were transplanted into CIA model, pathological and immunological indicators were detected to evaluate the therapeutic effects of sTNFRII-MSC. The distribution of sTNFRII-MSC was determined by immunofluorescence assay. Apoptosis and autophagy were analyzed by flow cytometry, western blot and immunofluorescence. RESULTS sTNFRII-Fc secreted by sTNFRII-MSC present biological activity both in vitro and vivo. sTNFRII-MSC transplantation effectively alleviates mice collagen-induced arthritis (CIA) via migrating to affected area, protecting articular cartilage destruction, modulating immune balance and sTNFRII-MSC showed prolonged internal retention via resisting apoptosis/autophagy induced by TNF-α. CONCLUSION sTNFRII-Fc modification protects MSCs against apoptosis/autophagy induced by TNF-α, in addition to releasing sTNFRII-Fc neutralizing TNF-α to block relevant immune-inflammation cascade, and thus exert better therapeutic effects in alleviating inflammatory arthritis.
Collapse
Affiliation(s)
- Yingjie Zhao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.,Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Xuezhi Yang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Siyu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Bingjie Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Susu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Xinwei Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Yueye Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Chengyan Jia
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Yan Chang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
20
|
Rafiei Alavi SN, Madani Neishaboori A, Hossein H, Sarveazad A, Yousefifard M. Efficacy of adipose tissue-derived stem cells in locomotion recovery after spinal cord injury: a systematic review and meta-analysis on animal studies. Syst Rev 2021; 10:213. [PMID: 34330329 PMCID: PMC8325264 DOI: 10.1186/s13643-021-01771-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Considerable disparities exist on the use of adipose tissue-derived stem cells (ADSCs) for treatment of spinal cord injury (SCI). Hence, the current systematic review aimed to investigate the efficacy of ADSCs in locomotion recovery following SCI in animal models. METHODS A search was conducted in electronic databases of MEDLINE, Embase, Scopus, and Web of Science until the end of July 2019. Reference and citation tracking and searching Google and Google Scholar search engines were performed to achieve more studies. Animal studies conducted on rats having SCI which were treated with ADSCs were included in the study. Exclusion criteria were lacking a non-treated control group, not evaluating locomotion, non-rat studies, not reporting the number of transplanted cells, not reporting isolation and preparation methods of stem cells, review articles, combination therapy, use of genetically modified ADSCs, use of induced pluripotent ADSCs, and human trials. Risk of bias was assessed using Hasannejad et al.'s proposed method for quality control of SCI-animal studies. Data were analyzed in STATA 14.0 software, and based on a random effect model, pooled standardized mean difference with a 95% confidence interval was presented. RESULTS Of 588 non-duplicated papers, data from 18 articles were included. Overall risk of bias was high risk in 8 studies, some concern in 9 studies and low risk in 1 study. Current evidence demonstrated that ADSCs transplantation could improve locomotion following SCI (standardized mean difference = 1.71; 95%CI 1.29-2.13; p < 0.0001). A considerable heterogeneity was observed between the studies (I2 = 72.0%; p < 0.0001). Subgroup analysis and meta-regression revealed that most of the factors like injury model, the severity of SCI, treatment phase, injury location, and number of transplanted cells did not have a significant effect on the efficacy of ADSCs in improving locomotion following SCI (pfor odds ratios > 0.05). CONCLUSION We conclude that any number of ADSCs by any prescription routes can improve locomotion recovery in an SCI animal model, at any phase of SCI, with any severity. Given the remarkable bias about blinding, clinical translation of the present results is tough, because in addition to the complexity of the nervous system and the involvement of far more complex motor circuits in the human, blinding compliance and motor outcome assessment tests in animal studies and clinical trials are significantly different.
Collapse
Affiliation(s)
| | - Arian Madani Neishaboori
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Hasti Hossein
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Niayesh St, Satarkhan Av, P.O Box: 14665-354, 1449614535, Tehran, Iran. .,Nursing Care Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran.
| |
Collapse
|
21
|
Hua T(T, Bejoy J, Song L, Wang Z, Zeng Z, Zhou Y, Li Y, Sang QXA. Cerebellar Differentiation from Human Stem Cells Through Retinoid, Wnt, and Sonic Hedgehog Pathways. Tissue Eng Part A 2021; 27:881-893. [PMID: 32873223 PMCID: PMC8336229 DOI: 10.1089/ten.tea.2020.0135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023] Open
Abstract
Differentiating cerebellar organoids can be challenging due to complex cell organization and structure in the cerebellum. Different approaches were investigated to recapitulate differentiation process of the cerebellum from human-induced pluripotent stem cells (hiPSCs) without high efficiency. This study was carried out to test the hypothesis that the combination of different signaling factors including retinoic acid (RA), Wnt activator, and sonic hedgehog (SHH) activator promotes the cerebellar differentiation of hiPSCs. Wnt, RA, and SHH pathways were activated by CHIR99021 (CHIR), RA, and purmorphamine (PMR), respectively. Different combinations of the morphogens (RA/CHIR, RA/PMR, CHIR/PMR, and RA/CHIR/PMR) were utilized, and the spheroids (day 35) were characterized for the markers of three cerebellum layers (the molecular layer, the Purkinje cell layer, and the granule cell layer). Of all the combinations tested, RA/CHIR/PMR promoted both the Purkinje cell layer and the granule cell layer differentiation. The cells also exhibited electrophysiological characteristics using whole-cell patch clamp recording, especially demonstrating Purkinje cell electrophysiology. This study should advance the understanding of different signaling pathways during cerebellar development to engineer cerebellum organoids for drug screening and disease modeling. Impact statement This study investigated the synergistic effects of retinoic acid, Wnt activator, and sonic hedgehog activator on cerebellar patterning of human-induced pluripotent stem cell (hiPSC) spheroids and organoids. The results indicate that the combination promotes the differentiation of the Purkinje cell layer and the granule cell layer. The cells also exhibit electrophysiological characteristics using whole-cell patch clamp recording, especially demonstrating Purkinje cell electrophysiology. The findings are significant for understanding the biochemical signaling of three-dimensional microenvironment on neural patterning of hiPSCs for applications in organoid engineering, disease modeling, and drug screening.
Collapse
Affiliation(s)
- Thien (Timothy) Hua
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Julie Bejoy
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Zhe Wang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
| | - Ziwei Zeng
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
- Department of Colorectal Surgery, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
22
|
Galipeau J, Krampera M, Leblanc K, Nolta JA, Phinney DG, Shi Y, Tarte K, Viswanathan S, Martin I. Mesenchymal stromal cell variables influencing clinical potency: the impact of viability, fitness, route of administration and host predisposition. Cytotherapy 2021; 23:368-372. [PMID: 33714704 PMCID: PMC11708105 DOI: 10.1016/j.jcyt.2020.11.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/14/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
The International Society for Cell & Gene Therapy mesenchymal stromal cell (MSC) committee has been an interested observer of community interests in all matters related to MSC identity, mechanism of action, potency assessment and etymology, and it has regularly contributed to this conversation through a series of MSC pre-conferences and committee publications dealing with these matters. Arising from these reflections, the authors propose that an overlooked and potentially disruptive perspective is the impact of in vivo persistence on potency that is not predicted by surrogate cellular potency assays performed in vitro and how this translates to in vivo outcomes. Systemic delivery or extravascular implantation at sites removed from the affected organ system seems to be adequate in affecting clinical outcomes in many pre-clinical murine models of acute tissue injury and inflammatory pathology, including the recent European Medicines Agency-approved use of MSCs in Crohn-related fistular disease. The authors further propose that MSC viability and metabolic fitness likely dominate as a potency quality attribute, especially in recipients poised for salutary benefits as defined by emerging predictive biomarkers of response.
Collapse
Affiliation(s)
- Jacques Galipeau
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA.
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Katarina Leblanc
- Department of Laboratory Medicine, Center for Allogeneic Stem Cell Transplantation, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jan A Nolta
- Stem Cell Program, University of California Davis, Sacramento, California, USA
| | - Donald G Phinney
- Department of Molecular Therapeutics, Scripps Research Institute, Jupiter, Florida, USA
| | - Yufang Shi
- Institute for Translational Medicine, Soochow University, Suzhou, China
| | - Karin Tarte
- Établissement Français du Sang Bretagne, Institute for Health and Medical Research, University of Rennes, Rennes, France
| | - Sowmya Viswanathan
- Department of Medicine and Institute of Biomedical Engineering, Krembil Research Institute, University Health Network, University of Toronto, Toronto, Canada
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
23
|
Li L, Liu X, Gaihre B, Li Y, Lu L. Mesenchymal stem cell spheroids incorporated with collagen and black phosphorus promote osteogenesis of biodegradable hydrogels. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 121:111812. [PMID: 33579456 DOI: 10.1016/j.msec.2020.111812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/28/2020] [Accepted: 12/14/2020] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cell (MSC)-spheroids have sparked significant interest in bone tissue engineering due to their resemblance to natural bone tissue, especially in terms of cell-cell and cell-extracellular matrix interactions. Many biomaterials or biomolecules have been incorporated into MSC-spheroids to enhance their osteogenic abilities. In this respect, we assessed the osteogenic responses of MSC spheroids leveraged through the unique combination of collagen and black phosphorus (BP). The MSC spheroids were successfully constructed with 6 μg/mL collagen and/or a concentration gradient (0 μg/mL, 4 μg/mL, 8 μg/mL, and 16 μg/mL) of BP and were evaluated for MSC viability and their osteogenic differentiation over a time period of 14 days. Improved MSC viability and osteogenic ability were observed for the spheroids with collagen and BP at the concentration of 4 μg/mL and 8 μg/mL. Next, blank spheroids (Control) or the optimized MSC spheroids with 6 μg/mL collagen and 4 μg/mL BP (Col+BP4) were further encapsulated into two types of hydrogel scaffolds: porous oligo[poly(ethylene glycol) fumarate] (OPF) hydrogel and hydroxyapatite-collagen I scaffold (HE-COL). The osteogenic abilities of these four groups were evaluated after 14 and 21 days of osteogenic induction. The MSC spheroids incorporated with collagen and BP implanted into OPF porous hydrogel (Col+BP/OPF) elicited a higher expression of Runx2, osteopontin, and alkaline phosphatase than blank spheroids implanted into OPF porous hydrogel (Control/OPF). Enhanced osteogenesis was also observed in the Col+BP/HE-COL group as compared to Control/HE-COL. Taken together, the results from this study showed the perspectives of collagen and BP incorporated MSC spheroids for the development of injectable cellular therapies for bone regeneration.
Collapse
Affiliation(s)
- Linli Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Yong Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
24
|
Analysis of Same Selected Immunomodulatory Properties of Chorionic Mesenchymal Stem Cells. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10249040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) represent a population of adherent cells that can be isolated from multiple adult tissues. MSCs have immunomodulatory capacity and the ability to differentiate into many cell lines. Research study examines the immunomodulatory properties of MSCs isolated from chorion (CMSCs). Following the stimulation process, it was found that MSCs are capable of immunomodulatory action via the release of bioactive molecules as well as through direct contact with the immune cells. Immunomodulatory potential of the CMSCs was analyzed by modifying proliferative capacity of mitogen-activated lymphocytes. CMSCs and lymphocytes were tested in cell-to-cell contact. Lymphocytes were stained with carboxyfluorescein diacetate succinimidyl ester. Inhibition of the proliferation of activated lymphocytes was observed. Following the co-cultivation, the expression of markers involved in the immune response modulation was assessed. Afterwards, an increase in CMSCs expression of IL-10 was detected. Following the co-cultivation with activated lymphocyte, adhesion molecules CD54 and CD44 in the CMSCs increased. An increase of CD54 expression was observed. The properties of CMSCs, adherence and differentiation ability, were confirmed. The phenotype of CMSCs CD105+, CD90+, CD73+, CD44+, CD29+, CD45−, CD34−, CD54+ was characterized. It was demonstrated that chorion-derived MSCs have important immunomodulatory effects.
Collapse
|
25
|
Oxidative Stress and Neuroinflammation as a Pivot in Drug Abuse. A Focus on the Therapeutic Potential of Antioxidant and Anti-Inflammatory Agents and Biomolecules. Antioxidants (Basel) 2020; 9:antiox9090830. [PMID: 32899889 PMCID: PMC7555323 DOI: 10.3390/antiox9090830] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
Drug abuse is a major global health and economic problem. However, there are no pharmacological treatments to effectively reduce the compulsive use of most drugs of abuse. Despite exerting different mechanisms of action, all drugs of abuse promote the activation of the brain reward system, with lasting neurobiological consequences that potentiate subsequent consumption. Recent evidence shows that the brain displays marked oxidative stress and neuroinflammation following chronic drug consumption. Brain oxidative stress and neuroinflammation disrupt glutamate homeostasis by impairing synaptic and extra-synaptic glutamate transport, reducing GLT-1, and system Xc− activities respectively, which increases glutamatergic neurotransmission. This effect consolidates the relapse-promoting effect of drug-related cues, thus sustaining drug craving and subsequent drug consumption. Recently, promising results as experimental treatments to reduce drug consumption and relapse have been shown by (i) antioxidant and anti-inflammatory synthetic molecules whose effects reach the brain; (ii) natural biomolecules secreted by mesenchymal stem cells that excel in antioxidant and anti-inflammatory properties, delivered via non-invasive intranasal administration to animal models of drug abuse and (iii) potent anti-inflammatory microRNAs and anti-miRNAs which target the microglia and reduce neuroinflammation and drug craving. In this review, we address the neurobiological consequences of brain oxidative stress and neuroinflammation that follow the chronic consumption of most drugs of abuse, and the current and potential therapeutic effects of antioxidants and anti-inflammatory agents and biomolecules to reduce these drug-induced alterations and to prevent relapse.
Collapse
|
26
|
Genetically Modified Mesenchymal Stem Cells: The Next Generation of Stem Cell-Based Therapy for TBI. Int J Mol Sci 2020; 21:ijms21114051. [PMID: 32516998 PMCID: PMC7312789 DOI: 10.3390/ijms21114051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are emerging as an attractive approach for restorative medicine in central nervous system (CNS) diseases and injuries, such as traumatic brain injury (TBI), due to their relatively easy derivation and therapeutic effect following transplantation. However, the long-term survival of the grafted cells and therapeutic efficacy need improvement. Here, we review the recent application of MSCs in TBI treatment in preclinical models. We discuss the genetic modification approaches designed to enhance the therapeutic potency of MSCs for TBI treatment by improving their survival after transplantation, enhancing their homing abilities and overexpressing neuroprotective and neuroregenerative factors. We highlight the latest preclinical studies that have used genetically modified MSCs for TBI treatment. The recent developments in MSCs’ biology and potential TBI therapeutic targets may sufficiently improve the genetic modification strategies for MSCs, potentially bringing effective MSC-based therapies for TBI treatment in humans.
Collapse
|
27
|
Dong Y, Sun X, Zhang Z, Liu Y, Zhang L, Zhang X, Huang Y, Zhao Y, Qi C, Midgley AC, Wang S, Yang Q. Regional and sustained dual-release of growth factors from biomimetic tri-layered scaffolds for the repair of large-scale osteochondral defects. APPLIED MATERIALS TODAY 2020; 19:100548. [DOI: 10.1016/j.apmt.2019.100548] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
|
28
|
Sun J, Lyu J, Xing F, Chen R, Duan X, Xiang Z. A biphasic, demineralized, and Decellularized allograft bone‐hydrogel scaffold with a cell‐based
BMP
‐7 delivery system for osteochondral defect regeneration. J Biomed Mater Res A 2020; 108:1909-1921. [PMID: 32323455 DOI: 10.1002/jbm.a.36954] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/18/2020] [Accepted: 03/28/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Jiachen Sun
- Department of Orthopedics, West China HospitalSichuan University Chengdu P. R. China
| | - Jingtong Lyu
- Department of Orthopedics, Southwest HospitalThird Military Medical University Chongqing P. R. China
| | - Fei Xing
- Department of Orthopedics, West China HospitalSichuan University Chengdu P. R. China
| | - Ran Chen
- Department of Orthopedics, West China HospitalSichuan University Chengdu P. R. China
| | - Xin Duan
- Department of Orthopedics, West China HospitalSichuan University Chengdu P. R. China
| | - Zhou Xiang
- Department of Orthopedics, West China HospitalSichuan University Chengdu P. R. China
- Division of Stem Cell and Tissue Engineering, State Key Laboratory of BiotherapyWest China Hospital, Sichuan University Chengdu P. R. China
| |
Collapse
|
29
|
mRNA as a Tool for Gene Transfection in 3D Cell Culture for Future Regenerative Therapy. MICROMACHINES 2020; 11:mi11040426. [PMID: 32325734 PMCID: PMC7231348 DOI: 10.3390/mi11040426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 01/31/2023]
Abstract
A combination of three-dimensional (3D) cell culturing and non-viral gene transfection is promising in improving outcomes of cell transplantation therapy. Herein, gene transfection profiles in 3D cell culture were compared between plasmid DNA (pDNA) and messenger RNA (mRNA) introduction, using mesenchymal stem cell (MSC) 3D spheroids. Green fluorescence protein (GFP) mRNA induced GFP protein expression in 77% of the cells in the spheroids, whereas only 34% of the cells became GFP positive following pDNA introduction. In mechanistic analyses, most of the cells in MSC spheroids were non-dividing, and pDNA failed to induce GFP expression in most of the non-dividing cells. In contrast, both dividing and non-dividing cells became GFP-positive after mRNA introduction, which led to a high overall percentage of GFP-positive cells in the spheroids. Consequently, mRNA encoding an osteogenic factor, runt-related transcription factor 2 (Runx2), allowed in vitro osteogenic differentiation of MSCs in spheroids more efficiently compared to Runx2 pDNA. Conclusively, mRNA exhibits high potential in gene transfection in 3D cell culture, in which the cell division rate is lower than that in monolayer culture, and the combination of mRNA introduction and 3D cell culture is a promising approach to improve outcomes of cell transplantation in future regenerative therapy.
Collapse
|
30
|
Zhao Y, Ding X, Dong Y, Sun X, Wang L, Ma X, Zhu M, Xu B, Yang Q. Role of the Calcified Cartilage Layer of an Integrated Trilayered Silk Fibroin Scaffold Used to Regenerate Osteochondral Defects in Rabbit Knees. ACS Biomater Sci Eng 2020; 6:1208-1216. [PMID: 33464868 DOI: 10.1021/acsbiomaterials.9b01661] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The repair of osteochondral defects remains challenging, given the complexity of native osteochondral tissue and the limited self-repair capacity of cartilage. Osteochondral tissue engineering is a promising strategy. Here, we fabricated a biomimetic osteochondral scaffold using silk fibroin and hydroxyapatite, including a calcified cartilage layer (CCL). We studied the role played by the CCL in terms of cell viability in vivo. We established osteochondral defects in rabbit knees to investigate the effects of CCL-containing scaffolds with or without adipose tissue-derived stem cells (ADSCs). We evaluated osteochondral tissue regeneration by calculating gross observational scores, via histological and immunohistochemical assessments, by performing quantitative biochemical and biomechanical analyses of new osteochondral tissue, and via microcomputed tomography of new bone at 4, 8, and 12 weeks after surgery. In terms of surface roughness and integrity, the CCL + ADSCs group was better than the CCL and the non-CCL + ADSCs groups at all time points tested; the glycosaminoglycan and collagen type II levels of the CCL + ADSCs group were highest, reflecting the important role played by the CCL in cartilage tissue repair. Subchondral bone smoothness was better in the CCL + ADSCs group than in the non-CCL + ADSCs and CCL groups. The CCL promoted smooth subchondral bone regeneration but did not obviously affect bone strength or quality. In conclusion, a biomimetic osteochondral scaffold with a CCL, combined with autologous ADSCs, satisfactorily regenerated a rabbit osteochondral defect. The CCL enhances cartilage and subchondral bone regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Stomatological Hospital of Tianjin Medical University, 12 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Xiaoming Ding
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China.,Department of Orthopedics, Rizhao Traditional Chinese Medicine Hospital, 35 Haiwang Road, Donggang District, Rizhao, Shandong 276800, People's Republic of China
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Lianyong Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Xinlong Ma
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Nankai District, Tianjin 300071, People's Republic of China
| | - Baoshan Xu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 406 Jiefang Nan Road, Hexi District, Tianjin 300211, People's Republic of China
| |
Collapse
|
31
|
Yu-Taeger L, Stricker-Shaver J, Arnold K, Bambynek-Dziuk P, Novati A, Singer E, Lourhmati A, Fabian C, Magg J, Riess O, Schwab M, Stolzing A, Danielyan L, Nguyen HHP. Intranasal Administration of Mesenchymal Stem Cells Ameliorates the Abnormal Dopamine Transmission System and Inflammatory Reaction in the R6/2 Mouse Model of Huntington Disease. Cells 2019; 8:E595. [PMID: 31208073 PMCID: PMC6628278 DOI: 10.3390/cells8060595] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022] Open
Abstract
Intrastriatal administration of mesenchymal stem cells (MSCs) has shown beneficial effects in rodent models of Huntington disease (HD). However, the invasive nature of surgical procedure and its potential to trigger the host immune response may limit its clinical use. Hence, we sought to evaluate the non-invasive intranasal administration (INA) of MSC delivery as an effective alternative route in HD. GFP-expressing MSCs derived from bone marrow were intranasally administered to 4-week-old R6/2 HD transgenic mice. MSCs were detected in the olfactory bulb, midbrain and striatum five days post-delivery. Compared to phosphate-buffered saline (PBS)-treated littermates, MSC-treated R6/2 mice showed an increased survival rate and attenuated circadian activity disruption assessed by locomotor activity. MSCs increased the protein expression of DARPP-32 and tyrosine hydroxylase (TH) and downregulated gene expression of inflammatory modulators in the brain 7.5 weeks after INA. While vehicle treated R6/2 mice displayed decreased Iba1 expression and altered microglial morphology in comparison to the wild type littermates, MSCs restored both, Iba1 level and the thickness of microglial processes in the striatum of R6/2 mice. Our results demonstrate significantly ameliorated phenotypes of R6/2 mice after MSCs administration via INA, suggesting this method as an effective delivering route of cells to the brain for HD therapy.
Collapse
Affiliation(s)
- Libo Yu-Taeger
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Janice Stricker-Shaver
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Katrin Arnold
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Patrycja Bambynek-Dziuk
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Arianna Novati
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Elisabeth Singer
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Ali Lourhmati
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
| | - Claire Fabian
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), D-04103 Leipzig, Germany.
| | - Janine Magg
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
| | - Matthias Schwab
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, D-70376 Stuttgart, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Alexandra Stolzing
- Interdisciplinary Centre for Bioinformatics (IZBI), University of Leipzig, D-04107 Leipzig, Germany.
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| | - Lusine Danielyan
- Department of Clinical Pharmacology, University Hospital of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Laboratory of Neuroscience, Yerevan State Medical University, 0025 Yerevan, Armenia.
| | - Hoa Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, D-72076 Tuebingen, Germany.
- Centre for Rare Diseases (ZSE), University of Tuebingen, D-72076 Tuebingen, Germany.
- Departments of Biochemistry and Clinical Pharmacology, Yerevan State Medical University, 0025 Yerevan, Armenia.
- Department of Human Genetics, Ruhr University of Bochum, D-44801 Bochum, Germany.
- Departments of Medical Chemistry and Biochemistry, Yerevan State Medical University, 0025 Yerevan, Armenia.
| |
Collapse
|
32
|
Yuan O, Lin C, Wagner J, Archard JA, Deng P, Halmai J, Bauer G, Fink KD, Fury B, Perotti NH, Walker JE, Pollock K, Apperson M, Butters J, Belafsky P, Farwell DG, Kuhn M, Nolta J, Anderson JD. Exosomes Derived from Human Primed Mesenchymal Stem Cells Induce Mitosis and Potentiate Growth Factor Secretion. Stem Cells Dev 2019; 28:398-409. [PMID: 30638129 PMCID: PMC6441283 DOI: 10.1089/scd.2018.0200] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) facilitate functional recovery in numerous animal models of inflammatory and ischemic tissue-related diseases with a growing body of research suggesting that exosomes mediate many of these therapeutic effects. It remains unclear, however, which types of proteins are packaged into exosomes compared with the cells from which they are derived. In this study, using comprehensive proteomic analysis, we demonstrated that human primed MSCs secrete exosomes (pMEX) that are packaged with markedly higher fractions of specific protein subclasses compared with their cells of origin, indicating regulation of their contents. Notably, we found that pMEX are also packaged with substantially elevated levels of extracellular-associated proteins. Fibronectin was the most abundant protein detected, and data established that fibronectin mediates the mitogenic properties of pMEX. In addition, treatment of SHSY5Y cells with pMEX induced the secretion of growth factors known to possess mitogenic and neurotrophic properties. Taken together, our comprehensive analysis indicates that pMEX are packaged with specific protein subtypes, which may provide a molecular basis for their distinct functional properties.
Collapse
Affiliation(s)
- Oliver Yuan
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - Clayton Lin
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - Joseph Wagner
- Drug Discovery Consortium, University of California, San Francisco, San Francisco, California
| | - Joehleen A. Archard
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - Peter Deng
- Department of Neurology, University of California, Davis, Davis, California
| | - Julian Halmai
- Department of Neurology, University of California, Davis, Davis, California
| | - Gerhard Bauer
- Good Manufacturing Practice Facility, University of California, Davis, Davis, California
| | - Kyle D. Fink
- Department of Neurology, University of California, Davis, Davis, California
| | - Brian Fury
- Good Manufacturing Practice Facility, University of California, Davis, Davis, California
| | - Nicholas H. Perotti
- Good Manufacturing Practice Facility, University of California, Davis, Davis, California
| | - Jon E. Walker
- Stem Cell Program, University of California, Davis, Davis, California
| | - Kari Pollock
- Stem Cell Program, University of California, Davis, Davis, California
| | - Michelle Apperson
- Department of Neurology, University of California, Davis, Davis, California
| | - Janelle Butters
- Department of Neurology, University of California, Davis, Davis, California
| | - Peter Belafsky
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - D. Gregory Farwell
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - Maggie Kuhn
- Department of Otolaryngology, University of California, Davis, Davis, California
| | - Jan Nolta
- Stem Cell Program, University of California, Davis, Davis, California
| | | |
Collapse
|
33
|
Preclinical Evaluation of Long-Term Neuroprotective Effects of BDNF-Engineered Mesenchymal Stromal Cells as Intravitreal Therapy for Chronic Retinal Degeneration in Rd6 Mutant Mice. Int J Mol Sci 2019; 20:ijms20030777. [PMID: 30759764 PMCID: PMC6387230 DOI: 10.3390/ijms20030777] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/03/2019] [Accepted: 02/10/2019] [Indexed: 12/13/2022] Open
Abstract
This study aimed to investigate whether the transplantation of genetically engineered bone marrow-derived mesenchymal stromal cells (MSCs) to overexpress brain-derived neurotrophic factor (BDNF) could rescue the chronic degenerative process of slow retinal degeneration in the rd6 (retinal degeneration 6) mouse model and sought to identify the potential underlying mechanisms. Rd6 mice were subjected to the intravitreal injection of lentivirally modified MSC-BDNF or unmodified MSC or saline. In vivo morphology, electrophysiological retinal function (ERG), and the expression of apoptosis-related genes, as well as BDNF and its receptor (TrkB), were assessed in retinas collected at 28 days and three months after transplantation. We observed that cells survived for at least three months after transplantation. MSC-BDNF preferentially integrated into the outer retinal layers and considerably rescued damaged retinal cells, as evaluated by ERG and immunofluorescence staining. Additionally, compared with controls, the therapy with MSC-BDNF was associated with the induction of molecular changes related to anti-apoptotic signaling. In conclusion, BDNF overexpression observed in retinas after MSC-BDNF treatment could enhance the neuroprotective properties of transplanted autologous MSCs alone in the chronically degenerated retina. This research provides evidence for the long-term efficacy of genetically-modified MSC and may represent a strategy for treating various forms of degenerative retinopathies in the future.
Collapse
|
34
|
Marques CR, Marote A, Mendes-Pinheiro B, Teixeira FG, Salgado AJ. Cell secretome based approaches in Parkinson’s disease regenerative medicine. Expert Opin Biol Ther 2018; 18:1235-1245. [DOI: 10.1080/14712598.2018.1546840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Cláudia R. Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bárbara Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fábio G. Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
35
|
El Baz H, Demerdash Z, Kamel M, Atta S, Salah F, Hassan S, Hammam O, Khalil H, Bayoumi A. Potentials of Differentiated Human Cord Blood-Derived Unrestricted Somatic Stem Cells in Treatment of Liver Cirrhosis. EXP CLIN TRANSPLANT 2018; 17:251-258. [PMID: 30346265 DOI: 10.6002/ect.2017.0249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Liver transplantation is the well-known treatment for chronic liver diseases; however, postoperative complications and lack of donors continue to be limitations with this treatment. Investigating new modalities for treatment of chronic liver illness is a must. In the present study, we aimed to clarify the effects of an in vitro hepatocyte-differentiated human unrestricted somatic stem cell transplant as a new cell-based therapy in an experimental model of chronic liver failure. MATERIALS AND METHODS Human umbilical cord blood-derived unrestricted somatic stem cells were isolated, cultured, propagated, and characterized. Cells were directed to differentiate into hepatocyte-like cells. An animal model of carbon tetrachloride cirrhotic liver failure was prepared, and the human in vitro differentiated unrestricted somatic stem cells were transplanted into the experimental model. Animals that did not receive transplant served as the pathologic control group. Animals were euthanized 12 weeks after transplant, and liver functions and histopathology were assessed. RESULTS Compared with the pathologic control group, the transplant group showed improvements in levels of alanine aminotransferase, aspartate aminotransferase, albumin, and bilirubin. Histopathologic examination of the transplant group also showed improvements in hydropic degeneration and fibrosis. CONCLUSIONS The use of unrestricted somatic stem cells, isolated and propagated from cord blood and then differentiated into hepatocyte-like cells, improved both fibrosis and normal function of cirrhotic livers. These cells could be considered as a line of cell-based therapy in cases of chronic liver disease.
Collapse
Affiliation(s)
- Hanan El Baz
- From the Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
He XT, Wang J, Li X, Yin Y, Sun HH, Chen FM. The Critical Role of Cell Homing in Cytotherapeutics and Regenerative Medicine. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiao-Tao He
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Jia Wang
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Hai-Hua Sun
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- National Clinical Research Center for Oral Diseases; Department of Periodontology; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
- Shaanxi Engineering Research Center for Dental Materials, and Advanced Manufacture; Biomaterials Unit; School of Stomatology; Fourth Military Medical University; 710032 Xi'an P. R. China
| |
Collapse
|
37
|
The use of hydrogels for cell-based treatment of chronic kidney disease. Clin Sci (Lond) 2018; 132:1977-1994. [PMID: 30220651 DOI: 10.1042/cs20180434] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/01/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is a major and growing public health concern with increasing incidence and prevalence worldwide. The therapeutic potential of stem cell therapy, including mesenchymal stem cells (MSCs) and endothelial progenitor cells (EPCs) holds great promise for treatment of CKD. However, there are significant bottlenecks in the clinical translation due to the reduced number of transplanted cells and the duration of their presence at the site of tissue damage. Bioengineered hydrogels may provide a route of cell delivery to enhance treatment efficacy and optimise the targeting effectiveness while minimising any loss of cell function. In this review, we highlight the advances in stem cell therapy targeting kidney disease and discuss the emerging role of hydrogel delivery systems to fully realise the potential of adult stem cells as a regenerative therapy for CKD in humans. MSCs and EPCs mediate kidney repair through distinct paracrine effects. As a delivery system, hydrogels can prolong these paracrine effects by improving retention at the site of injury and protecting the transplanted cells from the harsh inflammatory microenvironment. We also discuss the features of a hydrogel, which may be tuned to optimise the therapeutic potential of encapsulated stem cells, including cell-adhesive epitopes, material stiffness, nanotopography, modes of gelation and degradation and the inclusion of bioactive molecules. This review concludes with a discussion of the challenges to be met for the widespread clinical use of hydrogel delivery system of stem cell therapy for CKD.
Collapse
|
38
|
Yanagihara K, Uchida S, Ohba S, Kataoka K, Itaka K. Treatment of Bone Defects by Transplantation of Genetically Modified Mesenchymal Stem Cell Spheroids. Mol Ther Methods Clin Dev 2018; 9:358-366. [PMID: 30038939 PMCID: PMC6054700 DOI: 10.1016/j.omtm.2018.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 04/11/2018] [Indexed: 11/18/2022]
Abstract
Cell transplantation is promising for regenerative medicine. A combination of a three-dimensional spheroid culture system with gene transfection was developed to enhance the therapeutic effects of mesenchymal stem cell (MSC) transplantation. The spheroid cell culture system is based on micropatterned substrates composed of a regular array of 100-μm-diameter cell-adhesion areas coated with a temperature-responsive polymer, poly (N-isopropylacrylamide-co-methacrylic acid), which allows for spheroid detachment by simply cooling the plates. In this study, MSC spheroids were transfected with plasmid DNA encoding runt-related transcription factor 2 (Runx2) and tested for their ability to enhance bone regeneration. In vitro analyses revealed that osteogenic differentiation of the MSCs was enhanced by forming spheroids and was further promoted by Runx2 expression. The enhanced osteogenic differentiation was maintained under pathological conditions, such as hypoxia and inflammation. Transplanting Runx2-transfected MSC spheroids into bone defects on rat femurs induced significantly faster bone regeneration compared with nontransfected MSC spheroids or genetically modified MSCs from conventional monolayer culture. MSC migration into the bone defect area was enhanced by upregulation of cell-migration-related genes. In conclusion, genetically modified MSC spheroids are effective for enhancing bone regeneration, providing a promising option for cell transplantation therapy in the fields of regenerative medicine.
Collapse
Affiliation(s)
- Kayoko Yanagihara
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Satoshi Uchida
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8656, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Shinsuke Ohba
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Keiji Itaka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
39
|
Music E, Futrega K, Doran MR. Sheep as a model for evaluating mesenchymal stem/stromal cell (MSC)-based chondral defect repair. Osteoarthritis Cartilage 2018; 26:730-740. [PMID: 29580978 DOI: 10.1016/j.joca.2018.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 02/05/2018] [Accepted: 03/19/2018] [Indexed: 02/02/2023]
Abstract
Osteoarthritis results from the degradation of articular cartilage and is one of the leading global causes of pain and immobility. Cartilage has a limited capacity for self-repair. While repair can be enhanced through surgical intervention, current methods often generate inferior fibrocartilage and repair is transient. The development of tissue engineering strategies to improve repair outcomes is an active area of research. While small animal models such as rodents and rabbits are often used in early pre-clinical work, larger animals that better recapitulate the anatomy and loading of the human joint are required for late-stage preclinical evaluation. Because of their physiological similarities to humans, and low cost relative to other large animals, sheep are routinely used in orthopedic research, including cartilage repair studies. In recent years, there has been considerable research investment into the development of cartilage repair strategies that utilize mesenchymal stem/stromal cells (MSC). In contrast to autologous chondrocytes derived from biopsies of articular cartilage, MSC offer some benefits including greater expansion capacity and elimination of the risk of morbidity at the cartilage biopsy site. The disadvantages of MSC are related to the challenges of inducing and maintaining a stable chondrocyte-like cell population capable of generating hyaline cartilage. Ovine MSC (oMSC) biology and their utility in sheep cartilage repair models have not been reviewed. Herein, we review the biological properties of MSC derived from sheep tissues, and the use of these cells to study articular cartilage repair in this large animal model.
Collapse
Affiliation(s)
- E Music
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia.
| | - K Futrega
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia.
| | - M R Doran
- Queensland University of Technology, Institute of Health and Biomedical Innovation, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD, Australia; Mater Research Institute - University of Queensland, Brisbane, QLD, Australia; Australian National Centre for the Public Awareness of Science, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
40
|
Zhang YJ, Chen X, Li G, Chan KM, Heng BC, Yin Z, Ouyang HW. Concise Review: Stem Cell Fate Guided By Bioactive Molecules for Tendon Regeneration. Stem Cells Transl Med 2018; 7:404-414. [PMID: 29573225 PMCID: PMC5905226 DOI: 10.1002/sctm.17-0206] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
Tendon disorders, which are commonly presented in the clinical setting, disrupt the patients' normal work and life routines, and they damage the careers of athletes. However, there is still no effective treatment for tendon disorders. In the field of tissue engineering, the potential of the therapeutic application of exogenous stem cells to treat tendon pathology has been demonstrated to be promising. With the development of stem cell biology and chemical biology, strategies that use inductive tenogenic factors to program stem cell fate in situ are the most easily and readily translatable to clinical applications. In this review, we focus on bioactive molecules that can potentially induce tenogenesis in adult stem cells, and we summarize the various differentiation factors found in comparative studies. Moreover, we discuss the molecular regulatory mechanisms of tenogenesis, and we examine the various challenges in developing standardized protocols for achieving efficient and reproducible tenogenesis. Finally, we discuss and predict future directions for tendon regeneration. Stem Cells Translational Medicine 2018;7:404-414.
Collapse
Affiliation(s)
- Yan-Jie Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| | - Gang Li
- China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Kai-Ming Chan
- Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Boon Chin Heng
- Faculty of Dentistry, Department of Endodontology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Zi Yin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China.,Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China.,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | - Hong-Wei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,China Orthopaedic Regenerative Medicine (CORMed), Hangzhou, People's Republic of China
| |
Collapse
|
41
|
Teixeira FG, Serra SC, Salgado AJ. Tips on How to Collect and Administer the Mesenchymal Stem Cell Secretome for Central Nervous System Applications. Methods Mol Biol 2017; 1416:457-65. [PMID: 27236689 DOI: 10.1007/978-1-4939-3584-0_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Human mesenchymal stem cells (hMSCs) have been proposed as possible therapeutic agents for central nervous system (CNS) disorders. Recently, it has been suggested that their effects are mostly mediated through their secretome, which contains a number of neuroregulatory molecules capable of increasing cell proliferation, differentiation, and survival in different physiological conditions. Here, we present an overview of the hMSC secretome as a possible candidate in the creation of new cell-free therapies, demonstrating the process of its collection and route of administration, focusing our attention on their effects in CNS regenerative medicine.
Collapse
Affiliation(s)
- F G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S C Serra
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus Gualtar, Braga, 4710-057, Portugal.
- ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
42
|
Kuljanin M, Bell GI, Sherman SE, Lajoie GA, Hess DA. Proteomic characterisation reveals active Wnt-signalling by human multipotent stromal cells as a key regulator of beta cell survival and proliferation. Diabetologia 2017; 60:1987-1998. [PMID: 28710530 DOI: 10.1007/s00125-017-4355-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/23/2017] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Novel strategies to stimulate the expansion of beta cell mass in situ are warranted for diabetes therapy. The aim of this study was to elucidate the secretome of human bone marrow (BM)-derived multipotent stromal cells (MSCs) with documented islet regenerative paracrine function. We hypothesised that regenerative MSCs will secrete a unique combination of protein factors that augment islet regeneration. METHODS Human BM-derived MSCs were examined for glucose-lowering capacity after transplantation into streptozotocin-treated NOD/severe combined immunodeficiency (SCID) mice and segregated into samples with regenerative (MSCR) vs nonregenerative (MSCNR) capacity. Secreted proteins associated with islet regenerative function were identified using stable isotope labelling with amino acids in cell culture (SILAC)-based quantitative proteomics. To functionally validate the importance of active Wnt signalling, we stimulated the Wnt-signalling pathway in MSCNR samples during ex vivo expansion using glycogen synthase kinase 3 (GSK3) inhibition (CHIR99201), and the conditioned culture media (CM) generated was tested for the capacity to support cultured human islet cell survival and proliferation in vitro. RESULTS MSCR showed increased secretion of proteins associated with cell growth, matrix remodelling, immunosuppressive and proangiogenic properties. In contrast, MSCNR uniquely secreted proteins known to promote inflammation and negatively regulate angiogenesis. Most notably, MSCR maintained Wnt signalling via Wnt5A/B (~2.5-fold increase) autocrine activity during ex vivo culture, while MSCNR repressed Wnt signalling via Dickkopf-related protein (DKK)1 (~2.5-fold increase) and DKK3 secretion. Inhibition of GSK3 activity in MSCNR samples increased the accumulation of nuclear β-catenin and generated CM that augmented beta cell survival (13% increases) and proliferation when exposed to cultured human islets. CONCLUSIONS/INTERPRETATION Maintenance of active Wnt signalling within human MSCs promotes the secretion of matricellular and proangiogenic proteins that formulate a niche for islet regeneration.
Collapse
Affiliation(s)
- Miljan Kuljanin
- Don Rix Protein Identification Facility, Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, N5A 6C1, Canada
| | - Gillian I Bell
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Stephen E Sherman
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Molecular Medicine Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, 100 Perth Drive, London, ON, N6A 5K8, Canada
| | - Gilles A Lajoie
- Don Rix Protein Identification Facility, Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, N5A 6C1, Canada.
| | - David A Hess
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
- Molecular Medicine Laboratories, Krembil Centre for Stem Cell Biology, Robarts Research Institute, 100 Perth Drive, London, ON, N6A 5K8, Canada.
| |
Collapse
|
43
|
Fischer G, Wang F, Xiang H, Bai X, Yu H, Hogan QH. Inhibition of neuropathic hyperalgesia by intrathecal bone marrow stromal cells is associated with alteration of multiple soluble factors in cerebrospinal fluid. Exp Brain Res 2017; 235:2627-2638. [PMID: 28573310 PMCID: PMC6688185 DOI: 10.1007/s00221-017-5000-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/24/2017] [Indexed: 01/09/2023]
Abstract
Injury-induced neuropathic pain remains a serious clinical problem. Recent studies indicate that bone marrow stromal cells (BMSCs) effectively attenuate chronic neuropathic pain in animal models. Here, we examined the therapeutic effect of intrathecal administration of BMSCs isolated from young (1-month-old) rats on pain hypersensitivity induced by tibial nerve injury. Cerebrospinal fluid (CSF) was collected and analyzed to examine the effect of BMSC administration on the expression of 67 soluble factors in CSF. A sustained remission in injury-induced mechanical hyperalgesia was observed in BMSC-treated rats but not in control animals. Engrafted BMSCs were observed in spinal cords and dorsal root ganglia at 5 weeks after cell injection. Injury significantly decreased the levels of six soluble factors in CSF: intercellular adhesion molecule 1 (ICAM-1), interleukin-1β (IL-1β), IL-10, hepatocyte growth factor (HGF), Nope protein, and neurogenic locus notch homolog protein 1 (Notch-1). Intrathecal BMSCs significantly attenuated the injury-induced reduction of ICAM-1, IL-1β, HGF, IL-10, and Nope. This study adds to evidence supporting the use of intrathecal BMSCs in pain control and shows that this effect is accompanied by the reversal of injury-induced reduction of multiple CSF soluble factors. Our findings suggest that these soluble factors may be potential targets for treating chronic pain.
Collapse
Affiliation(s)
- Gregory Fischer
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, People's Republic of China
| | - Hongfei Xiang
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Xiaowen Bai
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| |
Collapse
|
44
|
Mechanisms of stem cell based cardiac repair-gap junctional signaling promotes the cardiac lineage specification of mesenchymal stem cells. Sci Rep 2017; 7:9755. [PMID: 28852100 PMCID: PMC5574972 DOI: 10.1038/s41598-017-10122-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023] Open
Abstract
Different subtypes of bone marrow-derived stem cells are characterized by varying functionality and activity after transplantation into the infarcted heart. Improvement of stem cell therapeutics requires deep knowledge about the mechanisms that mediate the benefits of stem cell treatment. Here, we demonstrated that co-transplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) led to enhanced synergistic effects on cardiac remodeling. While HSCs were associated with blood vessel formation, MSCs were found to possess transdifferentiation capacity. This cardiomyogenic plasticity of MSCs was strongly promoted by a gap junction-dependent crosstalk between myocytes and stem cells. The inhibition of cell-cell coupling significantly reduced the expression of the cardiac specific transcription factors NKX2.5 and GATA4. Interestingly, we observed that small non-coding RNAs are exchanged between MSCs and cardiomyocytes in a GJ-dependent manner that might contribute to the transdifferentiation process of MSCs within a cardiac environment. Our results suggest that the predominant mechanism of HSCs contribution to cardiac regeneration is based on their ability to regulate angiogenesis. In contrast, transplanted MSCs have the capability for intercellular communication with surrounding cardiomyocytes, which triggers the intrinsic program of cardiogenic lineage specification of MSCs by providing cardiomyocyte-derived cues.
Collapse
|
45
|
A New Chapter for Mesenchymal Stem Cells: Decellularized Extracellular Matrices. Stem Cell Rev Rep 2017; 13:587-597. [DOI: 10.1007/s12015-017-9757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Gonzalez-King H, García NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia Inducible Factor-1α Potentiates Jagged 1-Mediated Angiogenesis by Mesenchymal Stem Cell-Derived Exosomes. Stem Cells 2017; 35:1747-1759. [PMID: 28376567 DOI: 10.1002/stem.2618] [Citation(s) in RCA: 284] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/05/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022]
Abstract
Insufficient vessel growth associated with ischemia remains an unresolved issue in vascular medicine. Mesenchymal stem cells (MSCs) have been shown to promote angiogenesis via a mechanism that is potentiated by hypoxia. Overexpression of hypoxia inducible factor (HIF)-1α in MSCs improves their therapeutic potential by inducing angiogenesis in transplanted tissues. Here, we studied the contribution of exosomes released by HIF-1α-overexpressing donor MSCs (HIF-MSC) to angiogenesis by endothelial cells. Exosome secretion was enhanced in HIF-MSC. Omics analysis of miRNAs and proteins incorporated into exosomes pointed to the Notch pathway as a candidate mediator of exosome communication. Interestingly, we found that Jagged1 was the sole Notch ligand packaged into MSC exosomes and was more abundant in HIF-MSC than in MSC controls. The addition of Jagged1-containing exosomes from MSC and HIF-MSC cultures to endothelial cells triggered transcriptional changes in Notch target genes and induced angiogenesis in an in vitro model of capillary-like tube formation, and both processes were stimulated by HIF-1α. Finally, subcutaneous injection of Jagged 1-containing exosomes from MSC and HIF-MSC cultures in the Matrigel plug assay induced angiogenesis in vivo, which was more robust when they were derived from HIF-MSC cultures. All Jagged1-mediated effects could be blocked by prior incubation of exosomes with an anti-Jagged 1 antibody. All together, the results indicate that exosomes derived from MSCs stably overexpressing HIF-1α have an increased angiogenic capacity in part via an increase in the packaging of Jagged1, which could have potential applications for the treatment of ischemia-related disease. Stem Cells 2017;35:1747-1759.
Collapse
Affiliation(s)
- Hernán Gonzalez-King
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Nahuel A García
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María Ciria
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - José Anastasio Montero
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
47
|
Moisseiev E, Anderson JD, Oltjen S, Goswami M, Zawadzki RJ, Nolta JA, Park SS. Protective Effect of Intravitreal Administration of Exosomes Derived from Mesenchymal Stem Cells on Retinal Ischemia. Curr Eye Res 2017. [PMID: 28636406 DOI: 10.1080/02713683.2017.1319491] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Exosomes derived from human mesenchymal stem cells (hMSCs) cultured under hypoxic conditions contain proteins and growth factors that promote angiogenesis. This study investigated the effect of intravitreal administration of these exosomes on retinal ischemia using a murine model. METHODS Oxygen-induced retinopathy (OIR) was induced by exposing one-week-old male C57BL/6J mice to 5 days of 75% hyperoxic conditioning, and returning to room air. After hyperoxic conditioning, the right eye of each mouse was injected intravitreally with 1 µl saline or exosomes derived from hMSCs and compared to control mice of the same age raised in room air without OIR injected intravitreally with saline. Two weeks post-injection, fluorescein angiography (FA) and phase-variance optical coherence tomography angiography (pvOCTA) were used to assess retinal perfusion. Retinal thickness was determined by OCT. The extent of retinal neovascularization was quantitated histologically by counting vascular nuclei on the retinal surface. RESULTS Among eyes with OIR, intravitreal exosome treatment partially preserved retinal vascular flow in vivo and reduced associated retinal thinning; retinal thickness on OCT was 111.1 ± 7.4µm with saline versus 132.1 ± 11.6µm with exosome, p < 0.001. Retinal neovascularization among OIR eyes was reduced with exosome treatment when compared to saline-treated eyes (7.75 ± 3.68 versus 2.68 ± 1.35 neovascular nuclei per section, p < 0.0001). No immunogenicity or ocular/systemic adverse effect was associated with intravitreal exosome treatment. CONCLUSIONS Intravitreal administration of exosomes derived from hMSCs was well tolerated without immunosuppression and decreased the severity of retinal ischemia in this murine model. This appealing novel non-cellular therapeutic approach warrants further exploration.
Collapse
Affiliation(s)
- Elad Moisseiev
- a Department of Ophthalmology and Vision Science , University of California Davis Eye Center , Sacramento , CA , USA.,b Sackler School of Medicine , Tel Aviv University , Tel Aviv , Israel
| | - Johnathon D Anderson
- c Stem Cell Program , Institute for Regenerative Cures, UC Davis Medical Center , Sacramento , CA , USA
| | - Sharon Oltjen
- d Vitreoretinal Research Laboratory, Department of Ophthalmology and Vision Science , University of California Davis Eye Center , Sacramento , CA , USA
| | - Mayank Goswami
- e UC Davis RISE Eye-Pod Laboratory, Department of Cell Biology and Human Anatomy , University of California Davis , Davis , CA , USA
| | - Robert J Zawadzki
- a Department of Ophthalmology and Vision Science , University of California Davis Eye Center , Sacramento , CA , USA.,e UC Davis RISE Eye-Pod Laboratory, Department of Cell Biology and Human Anatomy , University of California Davis , Davis , CA , USA
| | - Jan A Nolta
- c Stem Cell Program , Institute for Regenerative Cures, UC Davis Medical Center , Sacramento , CA , USA
| | - Susanna S Park
- a Department of Ophthalmology and Vision Science , University of California Davis Eye Center , Sacramento , CA , USA
| |
Collapse
|
48
|
|
49
|
Uchida S, Hayakawa K, Ogata T, Tanaka S, Kataoka K, Itaka K. Treatment of spinal cord injury by an advanced cell transplantation technology using brain-derived neurotrophic factor-transfected mesenchymal stem cell spheroids. Biomaterials 2016; 109:1-11. [DOI: 10.1016/j.biomaterials.2016.09.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/06/2016] [Accepted: 09/09/2016] [Indexed: 01/01/2023]
|
50
|
Anderson JD, Pham MT, Contreras Z, Hoon M, Fink KD, Johansson HJ, Rossignol J, Dunbar GL, Showalter M, Fiehn O, Bramlett CS, Bardini RL, Bauer G, Fury B, Hendrix KJ, Chedin F, EL-Andaloussi S, Hwang B, Mulligan MS, Lehtiö J, Nolta JA. Mesenchymal stem cell-based therapy for ischemic stroke. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0053-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|