1
|
Qu W, Wu X, Wu W, Wang Y, Sun Y, Deng L, Walker M, Chen C, Dai H, Han Q, Ding Y, Xia Y, Smith G, Li R, Liu NK, Xu XM. Chondroitinase ABC combined with Schwann cell transplantation enhances restoration of neural connection and functional recovery following acute and chronic spinal cord injury. Neural Regen Res 2025; 20:1467-1482. [PMID: 39075913 PMCID: PMC11624882 DOI: 10.4103/nrr.nrr-d-23-01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/20/2023] [Accepted: 05/16/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00029/figure1/v/2024-07-28T173839Z/r/image-tiff Schwann cell transplantation is considered one of the most promising cell-based therapy to repair injured spinal cord due to its unique growth-promoting and myelin-forming properties. A the Food and Drug Administration-approved Phase I clinical trial has been conducted to evaluate the safety of transplanted human autologous Schwann cells to treat patients with spinal cord injury. A major challenge for Schwann cell transplantation is that grafted Schwann cells are confined within the lesion cavity, and they do not migrate into the host environment due to the inhibitory barrier formed by injury-induced glial scar, thus limiting axonal reentry into the host spinal cord. Here we introduce a combinatorial strategy by suppressing the inhibitory extracellular environment with injection of lentivirus-mediated transfection of chondroitinase ABC gene at the rostral and caudal borders of the lesion site and simultaneously leveraging the repair capacity of transplanted Schwann cells in adult rats following a mid-thoracic contusive spinal cord injury. We report that when the glial scar was degraded by chondroitinase ABC at the rostral and caudal lesion borders, Schwann cells migrated for considerable distances in both rostral and caudal directions. Such Schwann cell migration led to enhanced axonal regrowth, including the serotonergic and dopaminergic axons originating from supraspinal regions, and promoted recovery of locomotor and urinary bladder functions. Importantly, the Schwann cell survival and axonal regrowth persisted up to 6 months after the injury, even when treatment was delayed for 3 months to mimic chronic spinal cord injury. These findings collectively show promising evidence for a combinatorial strategy with chondroitinase ABC and Schwann cells in promoting remodeling and recovery of function following spinal cord injury.
Collapse
Affiliation(s)
- Wenrui Qu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yan Sun
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heqiao Dai
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Ding
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yongzhi Xia
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Rui Li
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
2
|
Hajimirzaei P, Tabatabaei FSA, Nasibi-Sis H, Razavian RS, Nasirinezhad F. Schwann cell transplantation for remyelination, regeneration, tissue sparing, and functional recovery in spinal cord injury: A systematic review and meta-analysis of animal studies. Exp Neurol 2025; 384:115062. [PMID: 39579959 DOI: 10.1016/j.expneurol.2024.115062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
INTRODUCTION Spinal cord injury (SCI) is a significant global health challenge that results in profound physical and neurological impairments. Despite progress in medical care, the treatment options for SCI are still restricted and often focus on symptom management rather than promoting neural repair and functional recovery. This study focused on clarifying the impact of Schwann cell (SC) transplantation on the molecular, cellular, and functional basis of recovery in animal models of SCI. MATERIAL AND METHODS Relevant studies were identified by conducting searches across multiple databases, which included PubMed, Web of Science, Scopus, and ProQuest. The data were analyzed via comprehensive meta-analysis software. We assessed the risk of bias via the SYRCLE method. RESULTS The analysis included 59 studies, 48 of which provided quantitative data. The results revealed significant improvements in various outcome variables, including protein zero structures (SMD = 1.66, 95 %CI: 0.96-2.36; p < 0.001; I2 = 49.8 %), peripherally myelinated axons (SMD = 1.81, 95 %CI: 0.99-2.63; p < 0.001; I2 = 39.3 %), biotinylated dextran amine-labeled CST only rostral (SMD = 1.31, 95 % CI: 0.50-2.12, p < 0.01, I2 = 49.7 %), fast blue-labeled reticular formation (SMD = 0.96, 95 %CI: 0.43-1.49, p < 0.001, I2 = 0.0 %), 5-hydroxytryptamine caudally (SMD = 0.83, 95 %CI: 0.36-1.29, p < 0.001, I2 = 17.2 %) and epicenter (SMD = 0.85, 95 %CI: 0.17-1.53, p < 0.05, I2 = 62.7 %), tyrosine hydroxylase caudally (SMD = 1.86, 95 %CI: 1.14-2.59, p < 0.001, I2 = 0.0 %) and epicenter (SMD = 1.82, 95 %CI: 1.18-2.47, p < 0.001, I2 = 0.0 %), cavity volume (SMD = -2.07, 95 %CI: -2.90 - -1.24, p < 0.001, I2 = 67.2 %), and Basso, Beattie, and Bresnahan (SMD = 1.26, 95 %CI: 0.93-1.58; p < 0.001; I2 = 79.4 %). CONCLUSIONS This study demonstrates the promising potential of SC transplantation as a therapeutic approach for SCI, clarifying its impact on various biological processes critical for recovery.
Collapse
Affiliation(s)
- Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Nasibi-Sis
- Department of Medical Library and Information Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Farinaz Nasirinezhad
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, Iran University of Medical sciences, Tehran, Iran; Center of Experimental and Comparative Study, Iran University of Medical sciences, Tehran, Iran.
| |
Collapse
|
3
|
Guest JD, Santamaria AJ, Solano JP, de Rivero Vaccari JP, Dietrich WD, Pearse DD, Khan A, Levi AD. Challenges in advancing Schwann cell transplantation for spinal cord injury repair. Cytotherapy 2025; 27:36-50. [PMID: 39387736 DOI: 10.1016/j.jcyt.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AIMS In this article we aimed to provide an expert synthesis of the current status of Schwann cell (SC)therapeutics and potential steps to increase their clinical utility. METHODS We provide an expert synthesis based on preclinical, clinical and manufacturing experience. RESULTS Schwann cells (SCs) are essential for peripheral nerve regeneration and are of interest in supporting axonal repair after spinal cord injury (SCI). SCs can be isolated and cultivated in tissue culture from adult nerve biopsies or generated from precursors and neural progenitors using specific differentiation protocols leading to expanded quantities. In culture, they undergo dedifferentiation to a state similar to "repair" SCs. The known repertoire of SC functions is increasing beyond axon maintenance, myelination, and axonal regeneration to include immunologic regulation and the release of potentially therapeutic extracellular vesicles. Recently, autologous human SC cultures purified under cGMP conditions have been tested in both nerve repair and subacute and chronic SCI clinical trials. Although the effects of SCs to support nerve regeneration are indisputable, their efficacy for clinical SCI has been limited according to the outcomes examined. CONCLUSIONS This review discusses the current limitations of transplanted SCs within the damaged spinal cord environment. Limitations include limited post-transplant cell survival, the inability of SCs to migrate within astrocytic parenchyma, and restricted axonal regeneration out of SC-rich graft regions. We describe steps to amplify the survival and integration of transplanted SCs and to expand the repertoire of uses of SCs, including SC-derived extracellular vesicles. The relative merits of transplanting autologous versus allogeneic SCs and the role that endogenous SCs play in spinal cord repair are described. Finally, we briefly describe the issues requiring solutions to scale up SC manufacturing for commercial use.
Collapse
Affiliation(s)
- James D Guest
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Andrea J Santamaria
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P Solano
- Pediatric Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan P de Rivero Vaccari
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - William D Dietrich
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Aisha Khan
- The Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D Levi
- The Miami Project to Cure Paralysis and Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
4
|
Tai Z, Liu J, Wang B, Chen S, Liu C, Chen X. The Effect of Aligned and Random Electrospun Fibers Derived from Porcine Decellularized ECM on Mesenchymal Stem Cell-Based Treatments for Spinal Cord Injury. Bioengineering (Basel) 2024; 11:772. [PMID: 39199730 PMCID: PMC11351159 DOI: 10.3390/bioengineering11080772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
The impact of traumatic spinal cord injury (SCI) can be extremely devastating, as it often results in the disruption of neural tissues, impeding the regenerative capacity of the central nervous system. However, recent research has demonstrated that mesenchymal stem cells (MSCs) possess the capacity for multi-differentiation and have a proven track record of safety in clinical applications, thus rendering them effective in facilitating the repair of spinal cord injuries. It is urgent to develop an aligned scaffold that can effectively load MSCs for promoting cell aligned proliferation and differentiation. In this study, we prepared an aligned nanofiber scaffold using the porcine decellularized spinal cord matrix (DSC) to induce MSCs differentiation for spinal cord injury. The decellularization method removed 87% of the immune components while retaining crucial proteins in DSC. The electrospinning technique was employed to fabricate an aligned nanofiber scaffold possessing biocompatibility and a diameter of 720 nm. In in vitro and in vivo experiments, the aligned nanofiber scaffold induces the aligned growth of MSCs and promotes their differentiation into neurons, leading to tissue regeneration and nerve repair after spinal cord injury. The approach exhibits promising potential for the future development of nerve regeneration scaffolds for spinal cord injury treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; (Z.T.); (C.L.)
| |
Collapse
|
5
|
Khaled MM, Ibrahium AM, Abdelgalil AI, El-Saied MA, El-Bably SH. Regenerative Strategies in Treatment of Peripheral Nerve Injuries in Different Animal Models. Tissue Eng Regen Med 2023; 20:839-877. [PMID: 37572269 PMCID: PMC10519924 DOI: 10.1007/s13770-023-00559-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Peripheral nerve damage mainly resulted from traumatic or infectious causes; the main signs of a damaged nerve are the loss of sensory and/or motor functions. The injured nerve has limited regenerative capacity and is recovered by the body itself, the recovery process depends on the severity of damage to the nerve, nowadays the use of stem cells is one of the new and advanced methods for treatment of these problems. METHOD Following our review, data are collected from different databases "Google scholar, Springer, Elsevier, Egyptian Knowledge Bank, and PubMed" using different keywords such as Peripheral nerve damage, Radial Nerve, Sciatic Nerve, Animals, Nerve regeneration, and Stem cell to investigate the different methods taken in consideration for regeneration of PNI. RESULT This review contains tables illustrating all forms and types of regenerative medicine used in treatment of peripheral nerve injuries (PNI) including different types of stem cells " adipose-derived stem cells, bone marrow stem cells, Human umbilical cord stem cells, embryonic stem cells" and their effect on re-constitution and functional recovery of the damaged nerve which evaluated by physical, histological, Immuno-histochemical, biochemical evaluation, and the review illuminated the best regenerative strategies help in rapid peripheral nerve regeneration in different animal models included horse, dog, cat, sheep, monkey, pig, mice and rat. CONCLUSION Old surgical attempts such as neurorrhaphy, autogenic nerve transplantation, and Schwann cell implantation have a limited power of recovery in cases of large nerve defects. Stem cell therapy including mesenchymal stromal cells has a high potential differentiation capacity to renew and form a new nerve and also restore its function.
Collapse
Affiliation(s)
- Mona M Khaled
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt.
| | - Asmaa M Ibrahium
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Ahmed I Abdelgalil
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Mohamed A El-Saied
- Department of Pathology, Faculty of Veterinary of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Samah H El-Bably
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| |
Collapse
|
6
|
Cunha NSC, Malvea A, Sadat S, Ibrahim GM, Fehlings MG. Pediatric Spinal Cord Injury: A Review. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1456. [PMID: 37761417 PMCID: PMC10530251 DOI: 10.3390/children10091456] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023]
Abstract
A spinal cord injury (SCI) can be a devastating condition in children, with profound implications for their overall health and quality of life. In this review, we aim to provide a concise overview of the key aspects associated with SCIs in the pediatric population. Firstly, we discuss the etiology and epidemiology of SCIs in children, highlighting the diverse range of causes. We explore the unique anatomical and physiological characteristics of the developing spinal cord that contribute to the specific challenges faced by pediatric patients. Next, we delve into the clinical presentation and diagnostic methods, emphasizing the importance of prompt and accurate diagnosis to facilitate appropriate interventions. Furthermore, we approach the multidisciplinary management of pediatric SCIs, encompassing acute medical care, surgical interventions, and ongoing supportive therapies. Finally, we explore emerging research as well as innovative therapies in the field, and we emphasize the need for continued advancements in understanding and treating SCIs in children to improve their functional independence and overall quality of life.
Collapse
Affiliation(s)
| | - Anahita Malvea
- Division of Neurosurgery, Krembil Neuroscience Centre, University Health Network, Toronto, ON M5T 2S8, Canada;
| | - Sarah Sadat
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada;
| | - George M. Ibrahim
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON M5G 1E8, Canada;
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Michael G. Fehlings
- Division of Neurosurgery, Krembil Neuroscience Centre, University Health Network, Toronto, ON M5T 2S8, Canada;
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
7
|
Wang Y, Sheng H, Cong M, Wang W, He Q, Li H, Li S, Zhang J, Chen Y, Guo S, Fang L, Pluchino S, Biskup E, Artemyev M, Chen F, Li Y, Chen J, Feng S, Wo Y. Spatio-temporally deciphering peripheral nerve regeneration in vivo after extracellular vesicle therapy under NIR-II fluorescence imaging. NANOSCALE 2023; 15:7991-8005. [PMID: 37067249 DOI: 10.1039/d3nr00795b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Extracellular vesicles (EVs) show potential as a therapeutic tool for peripheral nerve injury (PNI), promoting neurological regeneration. However, there are limited data on the in vivo spatio-temporal trafficking and biodistribution of EVs. In this study, we introduce a new non-invasive near-infrared fluorescence imaging strategy based on glucose-conjugated quantum dot (QDs-Glu) labeling to target and track EVs in a sciatic nerve injury rat model in real-time. Our results demonstrate that the injected EVs migrated from the uninjured site to the injured site of the nerve, with an increase in fluorescence signals detected from 4 to 7 days post-injection, indicating the release of contents from the EVs with therapeutic effects. Immunofluorescence and behavioral tests revealed that the EV therapy promoted nerve regeneration and functional recovery at 28 days post-injection. We also found a relationship between functional recovery and the NIR-II fluorescence intensity change pattern, providing novel evidence for the therapeutic effects of EV therapy using real-time NIR-II imaging at the live animal level. This approach initiates a new path for monitoring EVs in treating PNI under in vivo NIR-II imaging, enhancing our understanding of the efficacy of EV therapy on peripheral nerve regeneration and its mechanisms.
Collapse
Affiliation(s)
- Yueming Wang
- Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| | - Huaixuan Sheng
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS 226001, China
| | - Wenjin Wang
- Department of Plastic and Reconstructive Surgery. Shanghai ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, JS 226001, China
| | - Huizhu Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Shunyao Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jian Zhang
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yuzhou Chen
- Department of Othopedic Surgery, Xin Hua Hospital affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Shuaicheng Guo
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Lu Fang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Stefano Pluchino
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Ewelina Biskup
- Department of Basic and Clinical Science, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Mikhail Artemyev
- Research Institute for Physical Chemical Problems of the Belarusian State University, Leningradskaya srt., 14, Minsk, 220006, Belarus
| | - Fuchun Chen
- Key Laboratory of Infrared System Detection and Imaging Technology, Shanghai Institute of Technical Physics, Chinese Academy of Sciences, Shanghai 200083, China
| | - Yunxia Li
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jun Chen
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Sijia Feng
- Sports Medicine Institute of Fudan University, Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Yan Wo
- Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China.
| |
Collapse
|
8
|
Kitagawa T, Nagoshi N, Okano H, Nakamura M. A Narrative Review of Advances in Neural Precursor Cell Transplantation Therapies for Spinal Cord Injury. Neurospine 2022; 19:935-945. [PMID: 36597632 PMCID: PMC9816589 DOI: 10.14245/ns.2244628.314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/11/2022] [Indexed: 12/27/2022] Open
Abstract
A spinal cord injury (SCI) is a destructive event that causes a permanent deficit in neurological function because of poor regenerative potential. Transplantation therapies have attracted attention for restoration of the injured spinal cord, and transplantation of neural precursor cells (NPCs) has been studied worldwide. Several groups have demonstrated functional recovery via this therapeutic intervention due to the multiple beneficial effects of NPC transplantation, such as reconstruction of neuronal circuits, remyelination of axons, and neuroprotection by trophic factors. Our group developed a method to induce NPCs from human induced pluripotent stem cells (hiPSCs) and established a transplantation strategy for SCI. Functional improvement in SCI animals treated with hiPSC-NPCs was observed, and the safety of transplanting these cells was evaluated from multiple perspectives. With selection of a safe cell line and pretreatment of the cells to encourage maturation and differentiation, hiPSC-NPC transplantation therapy is now in the clinical phase of testing for subacute SCI. In addition, a research challenge will be to expand the efficacy of transplantation therapy for chronic SCI. More comprehensive strategies involving combination treatments are required to treat this problematic situation.
Collapse
Affiliation(s)
- Takahiro Kitagawa
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan,Corresponding Author Narihito Nagoshi Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Heterozygous Dcc Mutant Mice Have a Subtle Locomotor Phenotype. eNeuro 2022; 9:ENEURO.0216-18.2021. [PMID: 35115383 PMCID: PMC8906791 DOI: 10.1523/eneuro.0216-18.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
Axon guidance receptors such as deleted in colorectal cancer (DCC) contribute to the normal formation of neural circuits, and their mutations can be associated with neural defects. In humans, heterozygous mutations in DCC have been linked to congenital mirror movements, which are involuntary movements on one side of the body that mirror voluntary movements of the opposite side. In mice, obvious hopping phenotypes have been reported for bi-allelic Dcc mutations, while heterozygous mutants have not been closely examined. We hypothesized that a detailed characterization of Dcc heterozygous mice may reveal impaired corticospinal and spinal functions. Anterograde tracing of the Dcc+/− motor cortex revealed a normally projecting corticospinal tract, intracortical microstimulation (ICMS) evoked normal contralateral motor responses, and behavioral tests showed normal skilled forelimb coordination. Gait analyses also showed a normal locomotor pattern and rhythm in adult Dcc+/− mice during treadmill locomotion, except for a decreased occurrence of out-of-phase walk and an increased duty cycle of the stance phase at slow walking speed. Neonatal isolated Dcc+/− spinal cords had normal left-right and flexor-extensor coupling, along with normal locomotor pattern and rhythm, except for an increase in the flexor-related motoneuronal output. Although Dcc+/− mice do not exhibit any obvious bilateral impairments like those in humans, they exhibit subtle motor deficits during neonatal and adult locomotion.
Collapse
|
10
|
Fu H, Hu D, Chen J, Wang Q, Zhang Y, Qi C, Yu T. Repair of the Injured Spinal Cord by Schwann Cell Transplantation. Front Neurosci 2022; 16:800513. [PMID: 35250447 PMCID: PMC8891437 DOI: 10.3389/fnins.2022.800513] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/27/2022] [Indexed: 01/12/2023] Open
Abstract
Spinal cord injury (SCI) can result in sensorimotor impairments or disability. Studies of the cellular response to SCI have increased our understanding of nerve regenerative failure following spinal cord trauma. Biological, engineering and rehabilitation strategies for repairing the injured spinal cord have shown impressive results in SCI models of both rodents and non-human primates. Cell transplantation, in particular, is becoming a highly promising approach due to the cells’ capacity to provide multiple benefits at the molecular, cellular, and circuit levels. While various cell types have been investigated, we focus on the use of Schwann cells (SCs) to promote SCI repair in this review. Transplantation of SCs promotes functional recovery in animal models and is safe for use in humans with subacute SCI. The rationales for the therapeutic use of SCs for SCI include enhancement of axon regeneration, remyelination of newborn or sparing axons, regulation of the inflammatory response, and maintenance of the survival of damaged tissue. However, little is known about the molecular mechanisms by which transplanted SCs exert a reparative effect on SCI. Moreover, SC-based therapeutic strategies face considerable challenges in preclinical studies. These issues must be clarified to make SC transplantation a feasible clinical option. In this review, we summarize the recent advances in SC transplantation for SCI, and highlight proposed mechanisms and challenges of SC-mediated therapy. The sparse information available on SC clinical application in patients with SCI is also discussed.
Collapse
Affiliation(s)
- Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Die Hu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao Eye Hospital, Shandong Eye Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Qingdao, China
| | - Jinli Chen
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qizun Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingze Zhang
- Key Laboratory of Biomechanics of Hebei Province, Department of Trauma Emergency Center, The Third Hospital of Hebei Medical University, Orthopaedics Research Institution of Hebei Province, Shijiazhuang, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- *Correspondence: Chao Qi,
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Tengbo Yu,
| |
Collapse
|
11
|
Zawadzka M, Kwaśniewska A, Miazga K, Sławińska U. Perspectives in the Cell-Based Therapies of Various Aspects of the Spinal Cord Injury-Associated Pathologies: Lessons from the Animal Models. Cells 2021; 10:cells10112995. [PMID: 34831217 PMCID: PMC8616284 DOI: 10.3390/cells10112995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic injury of the spinal cord (SCI) is a devastating neurological condition often leading to severe dysfunctions, therefore an improvement in clinical treatment for SCI patients is urgently needed. The potential benefits of transplantation of various cell types into the injured spinal cord have been intensively investigated in preclinical SCI models and clinical trials. Despite the many challenges that are still ahead, cell transplantation alone or in combination with other factors, such as artificial matrices, seems to be the most promising perspective. Here, we reviewed recent advances in cell-based experimental strategies supporting or restoring the function of the injured spinal cord with a particular focus on the regenerative mechanisms that could define their clinical translation.
Collapse
|
12
|
Moulson AJ, Squair JW, Franklin RJM, Tetzlaff W, Assinck P. Diversity of Reactive Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Front Cell Neurosci 2021; 15:703810. [PMID: 34381334 PMCID: PMC8349991 DOI: 10.3389/fncel.2021.703810] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are essential for the development and homeostatic maintenance of the central nervous system (CNS). They are also critical players in the CNS injury response during which they undergo a process referred to as "reactive astrogliosis." Diversity in astrocyte morphology and gene expression, as revealed by transcriptional analysis, is well-recognized and has been reported in several CNS pathologies, including ischemic stroke, CNS demyelination, and traumatic injury. This diversity appears unique to the specific pathology, with significant variance across temporal, topographical, age, and sex-specific variables. Despite this, there is limited functional data corroborating this diversity. Furthermore, as reactive astrocytes display significant environmental-dependent plasticity and fate-mapping data on astrocyte subsets in the adult CNS is limited, it remains unclear whether this diversity represents heterogeneity or plasticity. As astrocytes are important for neuronal survival and CNS function post-injury, establishing to what extent this diversity reflects distinct established heterogeneous astrocyte subpopulations vs. environmentally dependent plasticity within established astrocyte subsets will be critical for guiding therapeutic development. To that end, we review the current state of knowledge on astrocyte diversity in the context of three representative CNS pathologies: ischemic stroke, demyelination, and traumatic injury, with the goal of identifying key limitations in our current knowledge and suggesting future areas of research needed to address them. We suggest that the majority of identified astrocyte diversity in CNS pathologies to date represents plasticity in response to dynamically changing post-injury environments as opposed to heterogeneity, an important consideration for the understanding of disease pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Aaron J. Moulson
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| | - Jordan W. Squair
- Department of Clinical Neuroscience, Faculty of Life Sciences, Center for Neuroprosthetics and Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), NeuroRestore, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin J. M. Franklin
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Peggy Assinck
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Ma C, Zhang W, Wang W, Shen J, Cai K, Liu M, Cao M. SKP-SCs transplantation alleviates 6-OHDA-induced dopaminergic neuronal injury by modulating autophagy. Cell Death Dis 2021; 12:674. [PMID: 34226513 PMCID: PMC8257782 DOI: 10.1038/s41419-021-03967-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease is a common neurodegenerative disease. Cell transplantation is a promising therapeutic option for improving the survival and function of dopaminergic neurons, but the mechanisms underlying the interaction between the transplanted cells and the recipient neurons remain to be studied. In this study, we investigated the effects of skin precursor cell-derived Schwann cells (SKP-SCs) directly cocultured with 6-OHDA-injured dopaminergic neurons in vitro and of SKP-SCs transplanted into the brains of 6-OHDA-induced PD mice in vivo. In vitro and in vivo studies revealed that SKP-SCs could reduce the damage to dopaminergic neurons by enhancing self-autophagy and modulating neuronal autophagy. Thus, the present study provides the first evidence that cell transplantation mitigates 6-OHDA-induced damage to dopaminergic neurons by enhancing self-autophagy, suggesting that earlier transplantation of Schwann cells might help alleviate the loss of dopaminergic neurons.
Collapse
Affiliation(s)
- Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wen Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wengcong Wang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Kefu Cai
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
14
|
Monje PV, Deng L, Xu XM. Human Schwann Cell Transplantation for Spinal Cord Injury: Prospects and Challenges in Translational Medicine. Front Cell Neurosci 2021; 15:690894. [PMID: 34220455 PMCID: PMC8249939 DOI: 10.3389/fncel.2021.690894] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023] Open
Abstract
The benefits of transplanting cultured Schwann cells (SCs) for the treatment of spinal cord injury (SCI) have been systematically investigated in experimental animals since the early 1990s. Importantly, human SC (hSC) transplantation for SCI has advanced to clinical testing and safety has been established via clinical trials conducted in the USA and abroad. However, multiple barriers must be overcome to enable accessible and effective treatments for SCI patients. This review presents available information on hSC transplantation for SCI with the intention to uncover gaps in our knowledge and discuss areas for future development. To this end, we introduce the historical progression of the work that supports existing and prospective clinical initiatives and explain the reasons for the choice of hSCs while also addressing their limitations as cell therapy products. A search of the relevant literature revealed that rat SCs have served as a preclinical model of reference since the onset of investigations, and that hSC transplants are relatively understudied, possibly due to the sophisticated resources and expertise needed for the traditional processing of hSC cultures from human nerves. In turn, we reason that additional experimentation and a reexamination of the available data are needed to understand the therapeutic value of hSC transplants taking into consideration that the manufacturing of the hSCs themselves may require further development for extended uses in basic research and clinical settings.
Collapse
Affiliation(s)
- Paula V. Monje
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
15
|
Lindsay SL, Barnett SC. Therapeutic Potential of Niche-Specific Mesenchymal Stromal Cells for Spinal Cord Injury Repair. Cells 2021; 10:cells10040901. [PMID: 33919910 PMCID: PMC8070966 DOI: 10.3390/cells10040901] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
The use of mesenchymal stem/stromal cells (MSCs) for transplant-mediated repair represents an important and promising therapeutic strategy after spinal cord injury (SCI). The appeal of MSCs has been fuelled by their ease of isolation, immunosuppressive properties, and low immunogenicity, alongside the large variety of available tissue sources. However, despite reported similarities in vitro, MSCs sourced from distinct tissues may not have comparable biological properties in vivo. There is accumulating evidence that stemness, plasticity, immunogenicity, and adaptability of stem cells is largely controlled by tissue niche. The extrinsic impact of cellular niche for MSC repair potential is therefore important, not least because of its impact on ex vivo expansion for therapeutic purposes. It is likely certain niche-targeted MSCs are more suited for SCI transplant-mediated repair due to their intrinsic capabilities, such as inherent neurogenic properties. In addition, the various MSC anatomical locations means that differences in harvest and culture procedures can make cross-comparison of pre-clinical data difficult. Since a clinical grade MSC product is inextricably linked with its manufacture, it is imperative that cells can be made relatively easily using appropriate materials. We discuss these issues and highlight the importance of identifying the appropriate niche-specific MSC type for SCI repair.
Collapse
|
16
|
Liang J, Cui R, Wang J, Shen J, Chen Y, Cao M, Ke K. Intracarotid Transplantation of Skin-Derived Precursor Schwann Cells Promotes Functional Recovery After Acute Ischemic Stroke in Rats. Front Neurol 2021; 12:613547. [PMID: 33633668 PMCID: PMC7902026 DOI: 10.3389/fneur.2021.613547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/13/2021] [Indexed: 12/17/2022] Open
Abstract
Purpose: Skin-derived Precursor Schwann cells (SKP-SCs) have been reported to provide neuroprotection for the injured and dysmyelinated nervous system. However, little is known about SKP-SCs on acute ischemic stroke (AIS). We aimed to explore the efficacy and the potential mechanism of action of SKP-SCs on AIS in a rat ischemic stroke model. Methods: Adult male Sprague–Dawley rats were subjected to a middle cerebral artery occlusion (MCAO) for 1.5 h on Day 0 and subsequently received an intracarotid injection of 2 × 106 green fluorescent protein (GFP) -labeled SKP-SCs or phosphate buffered saline (PBS) during reperfusion. Neurological function was assessed by behavioral tests on Days 1, 4, 7, 14, and 28. In a satellite cohort, rat brains were harvested and infarct volume was measured with 2,3,5-triphenyltetrazolium chloride (TTC) staining on Days 1 and 7, and migration and survival of SKP-SCs in the brain were traced by monitoring green fluorescence at 6 and12 h on Day 0, and on Days 1, 4, 7, 14, and 28. Histopathology and immunofluorescence staining were used to analyze the morphology, survival and apoptosis of neurons. Additionally, in an in vitro SKP-SC co-culture model using fetal rat primary cortical neurons underwent oxygen glucose deprivation/reoxygenation (OGD/R), Western blot was used to detect the expression of apoptosis indicators including activated caspase-3, Bax, and Bcl-2. TUNEL staining was used to count apoptotic cells. Results: Intracarotid transplantation of SKP-SCs effectively migrated to the periinfarct area and survived for at least 4 weeks. Transplanted SKP-SCs inhibited neuronal apoptosis, reduced infarct volume, and improved neurological recovery in the MCAO rats. Moreover, in vitro data showed that SKP-SCs treatment inhibited OGD/R-induced neuronal apoptosis and promoted survival of the cultured primary cortical neurons. Conclusions: Intracarotid transplantation of SKP-SCs promoted functional recovery in the rat AIS model and possesses the potential to be further developed as a novel therapy to treat ischemic stroke in humans.
Collapse
Affiliation(s)
- Jingjing Liang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ronghui Cui
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China.,Nantong University, Nantong, China
| | - Jinglei Wang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiabing Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Maosheng Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Kaifu Ke
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
17
|
Choi SJ, Park SY, Shin YH, Heo SH, Kim KH, Lee HI, Kim JK. Mesenchymal Stem Cells Derived from Wharton's Jelly Can Differentiate into Schwann Cell-Like Cells and Promote Peripheral Nerve Regeneration in Acellular Nerve Grafts. Tissue Eng Regen Med 2021; 18:467-478. [PMID: 33515168 DOI: 10.1007/s13770-020-00329-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Schwann cells (SCs) secrete neurotrophic factors and provide structural support and guidance during axonal regeneration. However, nearby nerves may be damaged to obtain primary SCs, and there is a lack of nervous tissue donors. We investigated the potential of Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) in differentiating into Schwann cell-like cells (WJ-SCLCs) as an alternative to SCs. We also examined whether implantation of WJ-SCLCs-laden acellular nerve grafts (ANGs) are effective in inducing functional recovery and nerve regeneration in an animal model of peripheral nerve injury. METHODS The differentiation of WJ-MSCs into WJ-SCLCs was determined by analyzing SC-specific markers. The secretion of neurotrophic factors was assessed by the Neuro Discovery antibody array. Neurite outgrowth and myelination of axons were found in a co-culture system involving motor neuron cell lines. The effects of ANGs on repairing sciatic nerves were evaluated using video gait angle test, isometric tetanic force analysis, and toluidine blue staining. RESULTS Compared with undifferentiated WJ-MSCs, WJ-SCLCs showed higher expression levels of SC-specific markers such as S100β, GFAP, KROX20, and NGFR. WJ-SCLCs also showed higher secreted amounts of brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and granulocyte-colony stimulating factor than did WJ-MSCs. WJ-SCLCs effectively promoted the outgrowth and myelination of neurites in motor neuron cells, and WJ-SCLCs laden ANGs significantly facilitated peripheral nerve regeneration in an animal model of sciatic nerve injury. CONCLUSION WJ-MSCs were readily differentiated into WJ-SCLCs, which effectively promoted the regeneration of peripheral nerves. Transplantation of WJ-SCLCs with ANGs might be useful for assisting peripheral nerve regeneration.
Collapse
Affiliation(s)
- Soon Jin Choi
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Suk Young Park
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Young Ho Shin
- Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Seung-Ho Heo
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Kang-Hyun Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Hyo In Lee
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Jae Kwang Kim
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea. .,Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
18
|
Balakrishnan A, Belfiore L, Chu TH, Fleming T, Midha R, Biernaskie J, Schuurmans C. Insights Into the Role and Potential of Schwann Cells for Peripheral Nerve Repair From Studies of Development and Injury. Front Mol Neurosci 2021; 13:608442. [PMID: 33568974 PMCID: PMC7868393 DOI: 10.3389/fnmol.2020.608442] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries arising from trauma or disease can lead to sensory and motor deficits and neuropathic pain. Despite the purported ability of the peripheral nerve to self-repair, lifelong disability is common. New molecular and cellular insights have begun to reveal why the peripheral nerve has limited repair capacity. The peripheral nerve is primarily comprised of axons and Schwann cells, the supporting glial cells that produce myelin to facilitate the rapid conduction of electrical impulses. Schwann cells are required for successful nerve regeneration; they partially “de-differentiate” in response to injury, re-initiating the expression of developmental genes that support nerve repair. However, Schwann cell dysfunction, which occurs in chronic nerve injury, disease, and aging, limits their capacity to support endogenous repair, worsening patient outcomes. Cell replacement-based therapeutic approaches using exogenous Schwann cells could be curative, but not all Schwann cells have a “repair” phenotype, defined as the ability to promote axonal growth, maintain a proliferative phenotype, and remyelinate axons. Two cell replacement strategies are being championed for peripheral nerve repair: prospective isolation of “repair” Schwann cells for autologous cell transplants, which is hampered by supply challenges, and directed differentiation of pluripotent stem cells or lineage conversion of accessible somatic cells to induced Schwann cells, with the potential of “unlimited” supply. All approaches require a solid understanding of the molecular mechanisms guiding Schwann cell development and the repair phenotype, which we review herein. Together these studies provide essential context for current efforts to design glial cell-based therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren Belfiore
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak-Ho Chu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Taylor Fleming
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Bai YR, Lai BQ, Han WT, Sun JH, Li G, Ding Y, Zeng X, Ma YH, Zeng YS. Decellularized optic nerve functional scaffold transplant facilitates directional axon regeneration and remyelination in the injured white matter of the rat spinal cord. Neural Regen Res 2021; 16:2276-2283. [PMID: 33818513 PMCID: PMC8354131 DOI: 10.4103/1673-5374.310696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Axon regeneration and remyelination of the damaged region is the most common repair strategy for spinal cord injury. However, achieving good outcome remains difficult. Our previous study showed that porcine decellularized optic nerve better mimics the extracellular matrix of the embryonic porcine optic nerve and promotes the directional growth of dorsal root ganglion neurites. However, it has not been reported whether this material promotes axonal regeneration in vivo. In the present study, a porcine decellularized optic nerve was seeded with neurotrophin-3-overexpressing Schwann cells. This functional scaffold promoted the directional growth and remyelination of regenerating axons. In vitro, the porcine decellularized optic nerve contained many straight, longitudinal channels with a uniform distribution, and microscopic pores were present in the channel wall. The spatial micro topological structure and extracellular matrix were conducive to the adhesion, survival and migration of neural stem cells. The scaffold promoted the directional growth of dorsal root ganglion neurites, and showed strong potential for myelin regeneration. Furthermore, we transplanted the porcine decellularized optic nerve containing neurotrophin-3-overexpressing Schwann cells in a rat model of T10 spinal cord defect in vivo. Four weeks later, the regenerating axons grew straight, the myelin sheath in the injured/transplanted area recovered its structure, and simultaneously, the number of inflammatory cells and the expression of chondroitin sulfate proteoglycans were reduced. Together, these findings suggest that porcine decellularized optic nerve loaded with Schwann cells overexpressing neurotrophin-3 promotes the directional growth of regenerating spinal cord axons as well as myelin regeneration. All procedures involving animals were conducted in accordance with the ethical standards of the Institutional Animal Care and Use Committee of Sun Yat-sen University (approval No. SYSU-IACUC-2019-B034) on February 28, 2019.
Collapse
Affiliation(s)
- Yu-Rong Bai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wei-Tao Han
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
20
|
Wu X, Wang L, Cong M, Shen M, He Q, Ding F, Shi H. Extracellular vesicles from skin precursor-derived Schwann cells promote axonal outgrowth and regeneration of motoneurons via Akt/mTOR/p70S6K pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1640. [PMID: 33490152 PMCID: PMC7812244 DOI: 10.21037/atm-20-5965] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Skin precursor-derived Schwann cells (SKP-SCs) have been shown to benefit the recovery of spinal cord injury (SCI) and peripheral nerve injury (PNI) with motor dysfunction. However, the effect of extracellular vesicles (EVs) from SKP-SCs responsible for neuroregeneration remains unknown. Methods Based on the obtainment and identification of rat SKP-SCs and their derived EVs, the primary rat injury model of motoneurons resulting from axotomy in vitro or nerve crush in vivo, as well as the secondary rat ischemic hypoxic injury model of motoneuron exposure to oxygen-glucose-deprivation (OGD) in vitro, were treated with EVs from skin precursor-derived Schwann cells (SKP-SC-EVs), respectively. Then, the axonal outgrowth and regrowth was observed and compared, and cell viability as well as the protein kinase B/mammalian target of rapamycin/p70 S6 kinase (Akt/mTOR/p70S6K) signaling pathway was detected, moreover, rapamycin (an mTOR inhibitor) was used to further reveal the underlying molecular mechanism. Results The internalization of SKP-SC-EVs by neuronal cells was identified in vitro and in vivo. Besides the pro-axonal outgrowth effect of SKP-SC-EVs, prospectively, the treatment of OGD-injured motoneurons with SKP-SC-EVs potentiated the restoration of neuronal viability and axonal regrowth. Furthermore, the axotomizing injury could be improved with SKP-SC-EVs treatment in vitro and in vivo. Finally, it was shown that the application of SKP-SC-EVs could activate the Akt/mTOR/p70S6K signaling pathway that can be abolished by rapamycin. Conclusions In summary, the addition of SKP-SC-EVs could regulate the cell growth and death signaling pathway mediated by Akt/mTOR/p70S6K, owing to the transmission of cargos in EVs to damaged motoneurons, which leads to axonal regrowth and neuronal resurrection. Thus, SKP-SC-EVs treatment could be a novel promising strategy for improving the axonal outgrowth and regeneration of motoneurons.
Collapse
Affiliation(s)
- Xia Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Liting Wang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, China
| | - Meng Cong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Pathophysiology, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
21
|
Ahuja CS, Mothe A, Khazaei M, Badhiwala JH, Gilbert EA, van der Kooy D, Morshead CM, Tator C, Fehlings MG. The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury. Stem Cells Transl Med 2020; 9:1509-1530. [PMID: 32691994 PMCID: PMC7695641 DOI: 10.1002/sctm.19-0135] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injuries (SCIs) are associated with tremendous physical, social, and financial costs for millions of individuals and families worldwide. Rapid delivery of specialized medical and surgical care has reduced mortality; however, long-term functional recovery remains limited. Cell-based therapies represent an exciting neuroprotective and neuroregenerative strategy for SCI. This article summarizes the most promising preclinical and clinical cell approaches to date including transplantation of mesenchymal stem cells, neural stem cells, oligodendrocyte progenitor cells, Schwann cells, and olfactory ensheathing cells, as well as strategies to activate endogenous multipotent cell pools. Throughout, we emphasize the fundamental biology of cell-based therapies, critical features in the pathophysiology of spinal cord injury, and the strengths and limitations of each approach. We also highlight salient completed and ongoing clinical trials worldwide and the bidirectional translation of their findings. We then provide an overview of key adjunct strategies such as trophic factor support to optimize graft survival and differentiation, engineered biomaterials to provide a support scaffold, electrical fields to stimulate migration, and novel approaches to degrade the glial scar. We also discuss important considerations when initiating a clinical trial for a cell therapy such as the logistics of clinical-grade cell line scale-up, cell storage and transportation, and the delivery of cells into humans. We conclude with an outlook on the future of cell-based treatments for SCI and opportunities for interdisciplinary collaboration in the field.
Collapse
Affiliation(s)
- Christopher S. Ahuja
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Andrea Mothe
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Mohamad Khazaei
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Jetan H. Badhiwala
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Emily A. Gilbert
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Derek van der Kooy
- Department of Molecular GeneticsUniversity of TorontoTorontoOntarioCanada
| | - Cindi M. Morshead
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Charles Tator
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| |
Collapse
|
22
|
Hopf A, Schaefer DJ, Kalbermatten DF, Guzman R, Madduri S. Schwann Cell-Like Cells: Origin and Usability for Repair and Regeneration of the Peripheral and Central Nervous System. Cells 2020; 9:E1990. [PMID: 32872454 PMCID: PMC7565191 DOI: 10.3390/cells9091990] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
Functional recovery after neurotmesis, a complete transection of the nerve fiber, is often poor and requires a surgical procedure. Especially for longer gaps (>3 mm), end-to-end suturing of the proximal to the distal part is not possible, thus requiring nerve graft implantation. Artificial nerve grafts, i.e., hollow fibers, hydrogels, chitosan, collagen conduits, and decellularized scaffolds hold promise provided that these structures are populated with Schwann cells (SC) that are widely accepted to promote peripheral and spinal cord regeneration. However, these cells must be collected from the healthy peripheral nerves, resulting in significant time delay for treatment and undesired morbidities for the donors. Therefore, there is a clear need to explore the viable source of cells with a regenerative potential similar to SC. For this, we analyzed the literature for the generation of Schwann cell-like cells (SCLC) from stem cells of different origins (i.e., mesenchymal stem cells, pluripotent stem cells, and genetically programmed somatic cells) and compared their biological performance to promote axonal regeneration. Thus, the present review accounts for current developments in the field of SCLC differentiation, their applications in peripheral and central nervous system injury, and provides insights for future strategies.
Collapse
Affiliation(s)
- Alois Hopf
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
| | - Dirk J. Schaefer
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Daniel F. Kalbermatten
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| |
Collapse
|
23
|
Assinck P, Sparling JS, Dworski S, Duncan GJ, Wu DL, Liu J, Kwon BK, Biernaskie J, Miller FD, Tetzlaff W. Transplantation of Skin Precursor-Derived Schwann Cells Yields Better Locomotor Outcomes and Reduces Bladder Pathology in Rats with Chronic Spinal Cord Injury. Stem Cell Reports 2020; 15:140-155. [PMID: 32559459 PMCID: PMC7363874 DOI: 10.1016/j.stemcr.2020.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022] Open
Abstract
Cell transplantation for spinal cord injury (SCI) has largely been studied in sub-acute settings within 1–2 weeks of injury. In contrast, here we transplanted skin-derived precursors differentiated into Schwann cells (SKP-SCs) into the contused rat spinal cord 8 weeks post-injury (wpi). Twenty-one weeks later (29 wpi), SKP-SCs were found to have survived transplantation, integrated with host tissue, and mitigated the formation of a dense glial scar. Furthermore, transplanted SKP-SCs filled much of the lesion sites and greatly enhanced the presence of endogenous SCs, which myelinated thousands of sprouting/spared host axons in and around the injury site. In addition, SKP-SC transplantation improved locomotor outcomes and decreased pathological thickening of bladder wall. To date, functional improvements have very rarely been observed with cell transplantation beyond the sub-acute stage of injury. Hence, these findings indicate that skin-derived SCs are a promising candidate cell type for the treatment of chronic SCI. SKP-SCs injected 8 weeks after SCI survive long-term and integrate with host tissue SKP-SC transplants boosted the presence of endogenous SCs in the chronic SCI site Treated spinal cords showed enhanced growth and SC myelination of axons Treated rats displayed better locomotor outcomes with reduced bladder pathologies
Collapse
Affiliation(s)
- Peggy Assinck
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
| | - Joseph S Sparling
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shaalee Dworski
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Greg J Duncan
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Di L Wu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
| | - Jie Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
| | - Brian K Kwon
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Department of Orthopaedics, University of British Columbia, Vancouver, BC, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Freda D Miller
- Neuroscience and Mental Health Program, Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Department of Zoology, University of British Columbia, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
24
|
Antonios JP, Farah GJ, Cleary DR, Martin JR, Ciacci JD, Pham MH. Immunosuppressive mechanisms for stem cell transplant survival in spinal cord injury. Neurosurg Focus 2020; 46:E9. [PMID: 30835678 DOI: 10.3171/2018.12.focus18589] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022]
Abstract
Spinal cord injury (SCI) has been associated with a dismal prognosis-recovery is not expected, and the most standard interventions have been temporizing measures that do little to mitigate the extent of damage. While advances in surgical and medical techniques have certainly improved this outlook, limitations in functional recovery continue to impede clinically significant improvements. These limitations are dependent on evolving immunological mechanisms that shape the cellular environment at the site of SCI. In this review, we examine these mechanisms, identify relevant cellular components, and discuss emerging treatments in stem cell grafts and adjuvant immunosuppressants that target these pathways. As the field advances, we expect that stem cell grafts and these adjuvant treatments will significantly shift therapeutic approaches to acute SCI with the potential for more promising outcomes.
Collapse
Affiliation(s)
- Joseph P Antonios
- 1David Geffen School of Medicine, University of California, Los Angeles, Los Angeles; and
| | - Ghassan J Farah
- 2Department of Neurosurgery, University of California San Diego School of Medicine, San Diego, California
| | - Daniel R Cleary
- 2Department of Neurosurgery, University of California San Diego School of Medicine, San Diego, California
| | - Joel R Martin
- 2Department of Neurosurgery, University of California San Diego School of Medicine, San Diego, California
| | - Joseph D Ciacci
- 2Department of Neurosurgery, University of California San Diego School of Medicine, San Diego, California
| | - Martin H Pham
- 2Department of Neurosurgery, University of California San Diego School of Medicine, San Diego, California
| |
Collapse
|
25
|
Inflammation Alters the Secretome and Immunomodulatory Properties of Human Skin-Derived Precursor Cells. Cells 2020; 9:cells9040914. [PMID: 32276503 PMCID: PMC7226778 DOI: 10.3390/cells9040914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/25/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022] Open
Abstract
Human skin-derived precursors (SKP) represent a group of somatic stem/precursor cells that reside in dermal skin throughout life that harbor clinical potential. SKP have a high self-renewal capacity, the ability to differentiate into multiple cell types and low immunogenicity, rendering them key candidates for allogeneic cell-based, off-the-shelf therapy. However, potential clinical application of allogeneic SKP requires that these cells retain their therapeutic properties under all circumstances and, in particular, in the presence of an inflammation state. Therefore, in this study, we investigated the impact of pro-inflammatory stimulation on the secretome and immunosuppressive properties of SKP. We demonstrated that pro-inflammatory stimulation of SKP significantly changes their expression and the secretion profile of chemo/cytokines and growth factors. Most importantly, we observed that pro-inflammatory stimulated SKP were still able to suppress the graft-versus-host response when cotransplanted with human PBMC in severe-combined immune deficient (SCID) mice, albeit to a much lesser extent than unstimulated SKP. Altogether, this study demonstrates that an inflammatory microenvironment has a significant impact on the immunological properties of SKP. These alterations need to be taken into account when developing allogeneic SKP-based therapies.
Collapse
|
26
|
Developmental Potential and Plasticity of Olfactory Epithelium Stem Cells Revealed by Heterotopic Grafting in the Adult Brain. Stem Cell Reports 2020; 14:692-702. [PMID: 32243847 PMCID: PMC7160358 DOI: 10.1016/j.stemcr.2020.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
The neural stem cells (NSCs) residing in the olfactory epithelium (OE) regenerate damaged olfactory sensory neurons throughout adulthood. The accessibility and availability of these NSCs in living individuals, including humans, makes them a promising candidate for harvesting their potential for cell replacement therapies. However, this requires an in-depth understanding of their developmental potential after grafting. Here, we investigated the developmental potential and plasticity of mouse OE-derived NSCs after grafting into the adult subventricular zone (SVZ) neurogenic niche. Our results showed that OE-derived NSCs integrate and proliferate just like endogenous SVZ stem cells, migrate with similar dynamics as endogenous neuroblasts toward the olfactory bulb, and mature and acquire similar electrophysiological properties as endogenous adult-born bulbar interneurons. These results reveal the developmental potential and plasticity of OE-derived NSCs in vivo and show that they can respond to heterotopic neurogenic cues to adapt their phenotype and become functional neurons in ectopic brain regions. OE-derived NSCs integrate in the SVZ after heterotopic transplantation OE-derived NSCs respond to SVZ niche factors and change their developmental program The development of OE-derived and SVZ NSCs are indistinguishable OE-derived NSCs grafted into the SVZ become functional bulbar interneurons
Collapse
|
27
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
28
|
West CR, Poormasjedi-Meibod MS, Manouchehri N, Williams AM, Erskine EL, Webster M, Fisk S, Morrison C, Short K, So K, Cheung A, Streijger F, Kwon BK. A porcine model for studying the cardiovascular consequences of high-thoracic spinal cord injury. J Physiol 2020; 598:929-942. [PMID: 31876952 DOI: 10.1113/jp278451] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/24/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS We have developed a novel porcine model of high-thoracic midline contusion spinal cord injury (SCI) at the T2 spinal level. We describe this model and the ensuing cardiovascular and neurohormonal responses, and demonstrate the model is efficacious for studying clinically relevant cardiovascular dysfunction post-SCI. We demonstrate that the high-thoracic SCI model, but not a low-thoracic SCI model, induces persistent hypotension along with a gradual reduction in plasma noradrenaline and increases in plasma aldosterone and angiotensin II. We additionally conducted a proof-of-concept long-term (12 weeks) survival study in animals with T2 contusion SCI demonstrating the potential utility of this model for not only acute experimentation but also long-term drug studies prior to translation to the clinic. ABSTRACT Cardiovascular disease is a leading cause of morbidity and mortality in the spinal cord injury (SCI) population, especially in those with high-thoracic or cervical SCI. With this in mind, we aimed to develop a large animal (porcine) model of high-thoracic (T2 level) contusion SCI and compare the haemodynamic and neurohormonal responses of this injury against a low-thoracic (T10 level) model. Ten Yorkshire pigs were randomly subjected to 20 cm weight drop contusion SCI at either the T2 or the T10 spinal level. Systolic blood pressure (SBP), mean arterial pressure (MAP) and heart rate (HR) were continuously monitored until 4 h post-SCI. Plasma noradrenaline (NA), aldosterone and angiotensin II (ANGII) were measured pre-SCI and at 30, 60, 120 and 240 min post-SCI. Additionally, two Yucatan pigs were subjected to T2-SCI and survived up to 12 weeks post-injury to demonstrate the efficacy of this model for long-term survival studies. Immediately after T2-SCI, SBP, MAP and HR increased (P < 0.0001). Between decompression (5 min post-SCI) and 30 min post-decompression in T2-SCI, SBP and MAP were lower than pre-SCI (P < 0.038). At 3 and 4 h after T2-SCI, SBP remained lower than pre-SCI (P = 0.048). After T10-SCI, haemodynamic indices remained largely unaffected. Plasma NA was lower in T2- vs. T10-SCI post-SCI, whilst aldosterone and ANGII were higher. Both chronically injured pigs demonstrated a vast reduction in SBP at 12 weeks post-SCI. Our model of T2-SCI causes a rapid and sustained alteration in neurohormonal control and cardiovascular function, which does not occur in the T10 model.
Collapse
Affiliation(s)
- Christopher R West
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Malihe-Sadat Poormasjedi-Meibod
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Neda Manouchehri
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Alexandra M Williams
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Erin L Erskine
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Megan Webster
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Shera Fisk
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Charlotte Morrison
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Katelyn Short
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Kitty So
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Amanda Cheung
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Femke Streijger
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Brian K Kwon
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Vancouver Spine Surgery Institute, Department of Orthopedics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
29
|
Garcia-Diaz B, Baron-Van Evercooren A. Schwann cells: Rescuers of central demyelination. Glia 2020; 68:1945-1956. [PMID: 32027054 DOI: 10.1002/glia.23788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/31/2022]
Abstract
The presence of peripheral myelinating cells in the central nervous system (CNS) has gained the neurobiologist attention over the years. Despite the confirmed presence of Schwann cells in the CNS in pathological conditions, and the long list of their beneficial effects on central remyelination, the cues that impede or allow Schwann cells to successfully conquer and remyelinate central axons remain partially undiscovered. A better knowledge of these factors stands out as crucial to foresee a rational therapeutic approach for the use of Schwann cells in CNS repair. Here, we review the diverse origins of Schwann cells into the CNS, both peripheral and central, as well as the CNS components that inhibit Schwann survival and migration into the central parenchyma. Namely, we analyze the astrocyte- and the myelin-derived components that restrict Schwann cells into the CNS. Finally, we highlight the unveiled mode of invasion of these peripheral cells through the central environment, using blood vessels as scaffolds to pave their ways toward demyelinated lesions. In short, this review presents the so far uncovered knowledge of this complex CNS-peripheral nervous system (PNS) relationship.
Collapse
Affiliation(s)
- Beatriz Garcia-Diaz
- Unidad de Gestión Clínica de Neurociencias, IBIMA, Hospital Regional Universitario de Málaga, Universidad de Málaga, Málaga, Spain.,Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie Paris 06, Paris, France
| |
Collapse
|
30
|
Li Y, Xiong L, Tang J, Zhu G, Dai R, Li L. Mouse skin-derived precursors alleviates ultraviolet B irradiation damage via early activation of TGF-β/Smad pathway by thrombospondin1. Cell Cycle 2020; 19:492-503. [PMID: 31965893 DOI: 10.1080/15384101.2020.1717042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Our previous research implied mouse skin-derived precursors (mSKPs) possessed the capacity of anti-ultraviolet B (UVB) irradiation damage, and the mechanisms might be associated with transforming growth factor-β (TGF-β) signaling pathway activation. In this study, we investigated and compared the response to UVB irradiation between mSKPs and dermal mesenchymal stem cells (dMSCs), and explored the underlying mechanisms. Irradiation damage such as decreased cell viability, cell senescence, and cell death was observed in both mSKPs and dMSCs at 24 h after UVB exposure. In mSKPs, change in cell morphology, viability, cell senescence and death at the following time points implied the recovery of UVB irradiation damage. Additionally, thrombospondin1 (TSP1) and TGF-β1 increased significantly in mSKPs' supernatant after UVB irradiation. The gene expression of TSP1, TGF-β1, metalloproteinase 1 (MMP1), and Collagen I elevated shortly after the UVB exposure. The protein expression of TSP1, TGF-β1, MMP1, Collagen I, smad2/3, and p-smad2/3 at multiple time points after the UVB exposure was consistent with the gene expression results. In dMSCs, no obvious recovery was noticed. Together, these results revealed that in mSKPs, one of the mechanisms to attenuate the UVB irradiation damage might be the early activation of TGF-β/Smad pathway by TSP1. Given that mSKPs could differentiate into fibroblast-like SKP-derived fibroblasts (SFBs) in vivo or with the presence of serum, mSKPs might serve as a therapeutic potential for fibroblasts supplement and UVB irradiation damage treatment.Abbreviations: SKPs: skin-derived precursors; mSKPs: mouse SKPs; UVB: ultraviolet B; TGF-β/Smad: transforming growth factor-β/Smad; TSP1: thrombospondin 1; MMP 13: metalloproteinases 13; TβRII: TGF-β receptor II; SFBs: SKP-derived fibroblasts; KEGG: Kyoto encyclopedia of genes and genomes; DEGs: differentially expressed genes; dMSCs: dermal mesenchymal stem cells; LM: light microscope; CCK-8: cell counting kit 8; ELISA: Enzyme-linked immuno sorbent assay; qRT-PCR: quantitative real-time polymerase chain reaction; TSPs: thrombospondins; ECM: extracellular matrix; R-smads: receptor-regulated smads.
Collapse
Affiliation(s)
- Yiming Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, P.R.C
| | - Lidan Xiong
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, P.R.C
| | - Jie Tang
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, P.R.C
| | - Guonian Zhu
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, P.R.C
| | - Ru Dai
- Department of Dermatology, Ningbo First Hospital, Zhejiang University, Ningbo, P.R.C
| | - Li Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, P.R.C
| |
Collapse
|
31
|
Mehrotra P, Tseropoulos G, Bronner ME, Andreadis ST. Adult tissue-derived neural crest-like stem cells: Sources, regulatory networks, and translational potential. Stem Cells Transl Med 2019; 9:328-341. [PMID: 31738018 PMCID: PMC7031649 DOI: 10.1002/sctm.19-0173] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
Neural crest (NC) cells are a multipotent stem cell population that give rise to a diverse array of cell types in the body, including peripheral neurons, Schwann cells (SC), craniofacial cartilage and bone, smooth muscle cells, and melanocytes. NC formation and differentiation into specific lineages takes place in response to a set of highly regulated signaling and transcriptional events within the neural plate border. Premigratory NC cells initially are contained within the dorsal neural tube from which they subsequently emigrate, migrating to often distant sites in the periphery. Following their migration and differentiation, some NC‐like cells persist in adult tissues in a nascent multipotent state, making them potential candidates for autologous cell therapy. This review discusses the gene regulatory network responsible for NC development and maintenance of multipotency. We summarize the genes and signaling pathways that have been implicated in the differentiation of a postmigratory NC into mature myelinating SC. We elaborate on the signals and transcription factors involved in the acquisition of immature SC fate, axonal sorting of unmyelinated neuronal axons, and finally the path toward mature myelinating SC, which envelope axons within myelin sheaths, facilitating electrical signal propagation. The gene regulatory events guiding development of SC in vivo provides insights into means for differentiating NC‐like cells from adult human tissues into functional SC, which have the potential to provide autologous cell sources for the treatment of demyelinating and neurodegenerative disorders.
Collapse
Affiliation(s)
- Pihu Mehrotra
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York.,Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| |
Collapse
|
32
|
Zhou P, Guan J, Xu P, Zhao J, Zhang C, Zhang B, Mao Y, Cui W. Cell Therapeutic Strategies for Spinal Cord Injury. Adv Wound Care (New Rochelle) 2019; 8:585-605. [PMID: 31637103 PMCID: PMC6798812 DOI: 10.1089/wound.2019.1046] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022] Open
Abstract
Significance: Spinal cord injury (SCI) is a neurological disorder that resulted from destroyed long axis of spinal cord, affecting thousands of people every year. With the occurrence of SCI, the lesions can form cystic cavities and produce glial scar, myelin inhibitor, and inflammation that negatively impact repair of spinal cord. Therefore, SCI remains a difficult problem to overcome with present therapeutics. This review of cell therapeutics in SCI provides a systematic review of combinatory therapeutics of SCI and helps the realization of regeneration of spinal cord in the future. Recent Advances: With major breakthroughs in neurobiology in recent years, present therapeutic strategies for SCI mainly aim at nerve regeneration or neuroprotection. For nerve regeneration, the application approaches are tissue engineering and cell transplantation, while drug therapeutics is applied for neuroprotection. Cell therapeutics is a new approach that treats SCI by cell transplantation. Cell therapeutics possesses advantages of neuroprotection, immune regulation, axonal regeneration, neuron relay formation, and remyelination. Critical Issues: Neurons cannot regenerate at the site of injury. Therefore, it is essential to find a repair strategy for remyelination, axon regeneration, and functional recovery. Cell therapeutics is emerging as the most promising approach for treating SCI. Future Directions: The future application of SCI therapy in clinical practice may require a combination of multiple strategies. A comprehensive treatment of injury of spinal cord is the focus of the present research. With the combination of different cell therapy strategies, future experiments will achieve more dramatic success in spinal cord repair.
Collapse
Affiliation(s)
- Pinghui Zhou
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Jingjing Guan
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Panpan Xu
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Jingwen Zhao
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Changchun Zhang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Yingji Mao
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
- School of Life Science, Bengbu Medical College, Bengbu, P.R. China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
33
|
Seira O, Liu J, Assinck P, Ramer M, Tetzlaff W. KIF2A characterization after spinal cord injury. Cell Mol Life Sci 2019; 76:4355-4368. [PMID: 31041455 PMCID: PMC11105463 DOI: 10.1007/s00018-019-03116-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 04/05/2019] [Accepted: 04/24/2019] [Indexed: 01/23/2023]
Abstract
Axons in the central nervous system (CNS) typically fail to regenerate after injury. This failure is multi-factorial and caused in part by disruption of the axonal cytoskeleton. The cytoskeleton, in particular microtubules (MT), plays a critical role in axonal transport and axon growth during development. In this regard, members of the kinesin superfamily of proteins (KIFs) regulate the extension of primary axons toward their targets and control the growth of collateral branches. KIF2A negatively regulates axon growth through MT depolymerization. Using three different injury models to induce SCI in adult rats, we examined the temporal and cellular expression of KIF2A in the injured spinal cord. We observed a progressive increase of KIF2A expression with maximal levels at 10 days to 8 weeks post-injury as determined by Western blot analysis. KIF2A immunoreactivity was present in axons, spinal neurons and mature oligodendrocytes adjacent to the injury site. Results from the present study suggest that KIF2A at the injured axonal tips may contribute to neurite outgrowth inhibition after injury, and that its increased expression in inhibitory spinal neurons adjacent to the injury site might contribute to an intrinsic wiring-control mechanism associated with neuropathic pain. Further studies will determine whether KIF2A may be a potential target for the development of regeneration-promoting or pain-preventing therapies.
Collapse
Affiliation(s)
- Oscar Seira
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada.
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada.
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
| | - Peggy Assinck
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Graduate Program in Neuroscience, University of British Columbia (UBC), Vancouver, Canada
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Matt Ramer
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia (UBC), 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada
- Department of Zoology, University of British Columbia (UBC), Vancouver, Canada
- Department of Surgery, University of British Columbia (UBC), Vancouver, Canada
| |
Collapse
|
34
|
Wu S, Ni S, Jiang X, Kuss MA, Wang HJ, Duan B. Guiding Mesenchymal Stem Cells into Myelinating Schwann Cell-Like Phenotypes by Using Electrospun Core-Sheath Nanoyarns. ACS Biomater Sci Eng 2019; 5:5284-5294. [PMID: 33455233 DOI: 10.1021/acsbiomaterials.9b00748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nerve guidance conduit (NGC)-infilling substrates have been reported to facilitate the regeneration of injured peripheral nerves (PNs), especially for large nerve gaps. In this study, longitudinally oriented electrospun core-sheath nanoyarns (csNYs), consisting of a polylactic acid microfiber core and an electrospun nanofiber sheath, were fabricated for potential PN tissue engineering applications. Our novel csNY displayed a well-aligned nanofibrous surface topography, resembling the ultrastructure of axons and fascicles of a native PN system, and it also provided a mechanically stable structure. The biological results showed that the csNY significantly enhanced the attachment, growth, and proliferation of human adipose derived mesenchymal stem cells (hADMSC) and also promoted the migration, proliferation, and phenotype maintenance of rabbit Schwann cells (rSCs). Our csNY notably increased the differentiation capability of hADMSC into SC-like cells (hADMSC-SC), in comparison with a 2D tissue culture polystyrene plate. More importantly, when combined with the appropriate induction medium, our csNY promoted hADMSC-SC to express high levels of myelination-associated markers. Overall, this study demonstrates that our csNYs have great potential to serve as not only ideal in vitro culture models for understanding SC-axon interaction and SC myelination but also as promising NGC-infilling substrates for PN regeneration applications.
Collapse
Affiliation(s)
- Shaohua Wu
- College of Textiles & Clothing; Collaborative Innovation Center of Marine Biomass Fibers, Qingdao University, Qingdao 266071, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
| | | | | | | | - Bin Duan
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
35
|
Peña JS, Robles D, Zhang S, Vazquez M. A Milled Microdevice to Advance Glia-Mediated Therapies in the Adult Nervous System. MICROMACHINES 2019; 10:mi10080513. [PMID: 31370352 PMCID: PMC6723365 DOI: 10.3390/mi10080513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/19/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022]
Abstract
Neurodegenerative disorders affect millions of adults worldwide. Neuroglia have become recent therapeutic targets due to their reparative abilities in the recycling of exogenous neurotoxins and production of endogenous growth factors for proper functioning of the adult nervous system (NS). Since neuroglia respond effectively to stimuli within in vivo environments on the micron scale, adult glial physiology has remarkable synergy with microscale systems. While clinical studies have begun to explore the reparative action of Müller glia (MG) of the visual system and Schwann Cells (ShC) of the peripheral NS after neural injury, few platforms enable the study of intrinsic neuroglia responses to changes in the local microenvironment. This project developed a low-cost, benchtop-friendly microfluidic system called the glia line system, or gLL, to advance the cellular study needed for emerging glial-based therapies. The gLL was fabricated using elastomeric kits coupled with a metal mold milled via conventional computer numerical controlled (CNC) machines. Experiments used the gLL to measure the viability, adhesion, proliferation, and migration of MG and ShC within scales similar to their respective in vivo microenvironments. Results illustrate differences in neuroglia adhesion patterns and chemotactic behavior significant to advances in regenerative medicine using implants and biomaterials, as well as cell transplantation techniques. Data showed highest survival and proliferation of MG and ShC upon laminin and illustrated a four-fold and two-fold increase of MG migration to dosage-dependent signaling from vascular endothelial growth factor (VEGF) and epidermal growth factor (EGF), respectively, as well as a 20-fold increase of ShC migration toward exogenous brain-derived neurotrophic factor (BDNF), compared to media control. The ability to quantify these biological parameters within the gLL offers an effective and reliable alternative to photolithography study neuroglia in a local environment ranging from the tens to hundreds of microns, using a low-cost and easily fabricated system.
Collapse
Affiliation(s)
- Juan S Peña
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Denise Robles
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Stephanie Zhang
- Department of Biomedical Engineering, State University of New York at Binghamton, Binghamton, NY 13902, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA.
| |
Collapse
|
36
|
Badhiwala JH, Ahuja CS, Fehlings MG. Time is spine: a review of translational advances in spinal cord injury. J Neurosurg Spine 2019; 30:1-18. [PMID: 30611186 DOI: 10.3171/2018.9.spine18682] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 09/28/2018] [Indexed: 11/06/2022]
Abstract
Acute traumatic spinal cord injury (SCI) is a devastating event with far-reaching physical, emotional, and economic consequences for patients, families, and society at large. Timely delivery of specialized care has reduced mortality; however, long-term neurological recovery continues to be limited. In recent years, a number of exciting neuroprotective and regenerative strategies have emerged and have come under active investigation in clinical trials, and several more are coming down the translational pipeline. Among ongoing trials are RISCIS (riluzole), INSPIRE (Neuro-Spinal Scaffold), MASC (minocycline), and SPRING (VX-210). Microstructural MRI techniques have improved our ability to image the injured spinal cord at high resolution. This innovation, combined with serum and cerebrospinal fluid (CSF) analysis, holds the promise of providing a quantitative biomarker readout of spinal cord neural tissue injury, which may improve prognostication and facilitate stratification of patients for enrollment into clinical trials. Given evidence of the effectiveness of early surgical decompression and growing recognition of the concept that "time is spine," infrastructural changes at a systems level are being implemented in many regions around the world to provide a streamlined process for transfer of patients with acute SCI to a specialized unit. With the continued aging of the population, central cord syndrome is soon expected to become the most common form of acute traumatic SCI; characterization of the pathophysiology, natural history, and optimal treatment of these injuries is hence a key public health priority. Collaborative international efforts have led to the development of clinical practice guidelines for traumatic SCI based on robust evaluation of current evidence. The current article provides an in-depth review of progress in SCI, covering the above areas.
Collapse
Affiliation(s)
- Jetan H Badhiwala
- 1Division of Neurosurgery, Department of Surgery, and
- 2Institute of Medical Science, University of Toronto; and
| | - Christopher S Ahuja
- 1Division of Neurosurgery, Department of Surgery, and
- 2Institute of Medical Science, University of Toronto; and
- 3Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael G Fehlings
- 1Division of Neurosurgery, Department of Surgery, and
- 2Institute of Medical Science, University of Toronto; and
- 3Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Squair JW, Ruiz I, Phillips AA, Zheng MM, Sarafis ZK, Sachdeva R, Gopaul R, Liu J, Tetzlaff W, West CR, Krassioukov AV. Minocycline Reduces the Severity of Autonomic Dysreflexia after Experimental Spinal Cord Injury. J Neurotrauma 2018; 35:2861-2871. [DOI: 10.1089/neu.2018.5703] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jordan W. Squair
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- MD/PhD Training Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian Ruiz
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aaron A. Phillips
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mei M.Z. Zheng
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zoe K. Sarafis
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rahul Sachdeva
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rayshad Gopaul
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jie Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Zoology, Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher R. West
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrei V. Krassioukov
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medicine, Division of Physical Medicine and Rehabilitation, University of British Columbia, Vancouver, British Columbia, Canada
- GF Strong Rehabilitation Centre, Vancouver Health Authority, Vancouver, British Columbia, Canada
| |
Collapse
|
38
|
Farzamfar S, Ehterami A, Salehi M, Vaeez A, Atashi A, Sahrapeyma H. Unrestricted Somatic Stem Cells Loaded in Nanofibrous Conduit as Potential Candidate for Sciatic Nerve Regeneration. J Mol Neurosci 2018; 67:48-61. [DOI: 10.1007/s12031-018-1209-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/08/2018] [Indexed: 12/25/2022]
|
39
|
Galhom RA, Hussein Abd El Raouf HH, Mohammed Ali MH. Role of bone marrow derived mesenchymal stromal cells and Schwann-like cells transplantation on spinal cord injury in adult male albino rats. Biomed Pharmacother 2018; 108:1365-1375. [PMID: 30372839 DOI: 10.1016/j.biopha.2018.09.131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Spinal cord injury is a considerable health impact accompanied with physical, psychological and economic burden. Bone marrow derived mesenchymal stromal cells (BM-MSCs) transplantation was found to produce neuronal regenerative effects. Schwann-like cells differentiated from BM-MSCs have myelin-forming ability. AIM OF THE WORK To compare the ability of BM-MSCs versus Schwann like cells to promote recovery of spinal cord injury. MATERIAL AND METHODS Adult male albino rats were used throughout the study. BM-MSCs were harvested from femora of rats. Sciatic nerves were extracted and used in the preparation of the induction culture medium for differentiation of BM-MSCs into Schwann-like cells. Rats were divided into control, spinal cord injured (SCI), spinal cord injured plus BM-MSCs transplantation (BM-MSC) and spinal cord injured plus Schwann-like cells transplantation (Sn) groups. BBB scale assessment was performed before and after SCI in all rats. Rats were euthanized at the end of the 7th week and spinal cords were dissected and processed for light and transmission electron microscopic examinations. RESULTS Spinal cord sections of SCI group revealed cavitation, necrosis and demyelination. BM-MSC and Sn groups showed both functional and structural improvement compared to SCI group with better BBB score and histopathological features in the BM-MSC group and more expression of S100 in the Sn group. CONCLUSION Transplantation of BM-MSCs and Schwann-like cells improved the structural and functional alterations of spinal cord injury with better improvement in BM-MSC group.
Collapse
Affiliation(s)
- Rania A Galhom
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | | | - Mona H Mohammed Ali
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
40
|
Zhu C, Huang J, Xue C, Wang Y, Wang S, Bao S, Chen R, Li Y, Gu Y. Skin derived precursor Schwann cell-generated acellular matrix modified chitosan/silk scaffolds for bridging rat sciatic nerve gap. Neurosci Res 2018; 135:21-31. [DOI: 10.1016/j.neures.2017.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/24/2017] [Accepted: 12/25/2017] [Indexed: 12/12/2022]
|
41
|
Li Y, Li X, Xiong L, Tang J, Li L. Comparison of phenotypes and transcriptomes of mouse skin-derived precursors and dermal mesenchymal stem cells. Differentiation 2018; 102:30-39. [PMID: 30056221 DOI: 10.1016/j.diff.2018.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/17/2018] [Indexed: 02/06/2023]
Abstract
Both skin-derived precursors (SKPs) and dermal mesenchymal stem cells (dMSCs) are promising candidates for cellular therapy and regenerative medicine. To date the comparison of phenotypes and transcriptomes of mouse SKPs (mSKPs) and dMSCs has never been reported. Here we characterized and compared the biological properties and transcriptomes of mSKP and dMSCs from the same mouse dermis sample. Firstly, we analyzed mSKPs and dMSCs by use of immunocytochemistry, cell cycle analysis, and CD antigen expression. Then we conducted the osteogenic, adipogenic, and chondrogenic induced differentiation for both cell types. Lastly, we compared their genomic profiles by RNA-sequencing (RNA-Seq), and verified the results of RNA-Seq by quantitative real time reverse transcription PCR (qRT-PCR). The results suggested that mSKPs and dMSCs shared similarities in certain positive stem cells markers expression, but demonstrated difference in Nanog and Oct4 expression. mSKPs and dMSCs demonstrated similar cell cycle distribution and CD antigen expression. Both types of cells could be induced differentiated into osteocytes, adipocytes, and chondrocytes. However, RNA-Seq and qRT-PCR results indicated that mSKPs and dMSCs had distinct transcriptome profiles. The majority of enriched differentially expressed genes (DEGs) from mSKPs was immune-related, while the majority of enriched DEGs from dMSCs was differentiation/development/disease-related. Transcriptome profiles suggested that mSKPs and dMSCs might have potential usage in the relevant morbidity management. These results may indicate a molecular basis for novel stem cell-based therapeutic strategies.
Collapse
Affiliation(s)
- Yiming Li
- Department of Dermatology and Venerology, Huaxi Hospital, Chengdu, Sichuan Province, PR China; Department of dermatology and venerology, the First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Ave, Xindu district, Chengdu, Sichuan Province, PR China
| | - Xiaohua Li
- Department of Dermatology and Venerology, Huaxi Hospital, Chengdu, Sichuan Province, PR China
| | - Lidan Xiong
- Department of Dermatology and Venerology, Huaxi Hospital, Chengdu, Sichuan Province, PR China
| | - Jie Tang
- Department of Dermatology and Venerology, Huaxi Hospital, Chengdu, Sichuan Province, PR China
| | - Li Li
- Department of Dermatology and Venerology, Huaxi Hospital, Chengdu, Sichuan Province, PR China.
| |
Collapse
|
42
|
Him A, Onger ME, Delibas B. Periferik Sinir Rejenerasyonu ve Kök Hücre Tedavileri. ACTA ACUST UNITED AC 2018. [DOI: 10.31832/smj.404819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
43
|
May Z, Fuehrmann T, Shoichet MS, Tetzlaff W, Biernaskie J, Fouad K. Reply to Comment on 'Adult skin-derived precursor Schwann cell grafts form growths in the injured spinal cord of Fischer rats'. ACTA ACUST UNITED AC 2018. [PMID: 29532786 DOI: 10.1088/1748-605x/aab629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zacnicte May
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Saulite L, Vavers E, Zvejniece L, Dambrova M, Riekstina U. The Differentiation of Skin Mesenchymal Stem Cells Towards a Schwann Cell Phenotype: Impact of Sigma-1 Receptor Activation. Mol Neurobiol 2018; 55:2840-2850. [PMID: 28455697 DOI: 10.1007/s12035-017-0511-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
Abstract
Neural crest stem cells (NCSCs) are the source of mature Schwann cells in the peripheral nervous system (PNS). The NCSC population resides in the bulge of hair follicles and in the dermis. Recently, it was shown that 2-3% of the human dermis mesenchymal stem cell (MSC) population expresses the NCSC marker CD271, thus enabling the use of skin MSCs for studying Schwann cell differentiation in vitro. The aims of this study were to establish a protocol for human skin MSC differentiation towards Schwann cell-like cells (SC-lcs) and to analyse the expression of sigma-1 receptor (S1R) in SC-lcs. The impact of S1R ligands, namely the selective agonist PRE-084, the positive allosteric modulator E1R and the selective antagonist NE-100, on Schwann cell differentiation was assessed. The expression of the neuron-specific genes Tubulin-βIII and Integrin-6α, the Schwann cell-specific gene S100b, MBP and the NCSC-specific genes p75NTR, Sox10, Notch1, Integrin-4α, Ap2α and Pax6 was analysed in MSCs and SC-lcs by real-time RT-PCR. BDNF secretion was evaluated by ELISA. The effect of S1R ligands on SC-lc differentiation was measured using BDNF ELISA and MBP flow cytometry. After MSC differentiation, NCSC markers p75NTR and Integrin-4α were downregulated 3.5-fold and 2-fold, respectively. To the contrary, MBP and S100b were significantly upregulated in SC-lcs. S1R ligands showed a tendency to increase the secretion of BDNF by the SC-lc population. PRE-084 and E1R increased MBP expression in the SC-lc population, whereas 3 μM NE-100 inhibited MBP expression in SC-lcs. In conclusion, our data demonstrate that S1R plays an important role in skin MSC differentiation towards myelinating Schwann cells.
Collapse
Affiliation(s)
- L Saulite
- Faculty of Medicine, University of Latvia, Raina blvd. 19, Riga, LV-1586, Latvia
| | - E Vavers
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles Street 21, Riga, Latvia
- Faculty of Pharmacy, Riga Stradins University, Dzirciema Street 16, Riga, Latvia
| | - L Zvejniece
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles Street 21, Riga, Latvia
| | - M Dambrova
- Latvian Institute of Organic Synthesis, Laboratory of Pharmaceutical Pharmacology, Aizkraukles Street 21, Riga, Latvia
- Faculty of Pharmacy, Riga Stradins University, Dzirciema Street 16, Riga, Latvia
| | - U Riekstina
- Faculty of Medicine, University of Latvia, Raina blvd. 19, Riga, LV-1586, Latvia.
| |
Collapse
|
45
|
Cornelison RC, Gonzalez-Rothi EJ, Porvasnik SL, Wellman SM, Park JH, Fuller DD, Schmidt CE. Injectable hydrogels of optimized acellular nerve for injection in the injured spinal cord. Biomed Mater 2018; 13:034110. [PMID: 29380749 PMCID: PMC5911159 DOI: 10.1088/1748-605x/aaab82] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Spinal cord injury (SCI) affects a quarter million individuals in the United States, and there is currently no clinical treatment. Both fresh and acellular peripheral nerve grafts can induce spinal axon regeneration and support functional recovery in experimental injury models. Nonetheless, a scaffold that can be injected into a spinal contusion would be far less invasive to apply. We aimed to develop the first injectable acellular nerve graft for promoting repair after contusion SCI. APPROACH We report a method to enzymatically solubilize optimized acellular (OA) nerve-a decellularized peripheral nerve graft developed in our laboratory and currently used clinically-to obtain an injectable solution that undergoes thermal gelation under physiological conditions. We quantified multiple physical and compositional properties of this novel material as well as tested its efficacy at acute and chronic time points following cervical contusion SCI. MAIN RESULTS This injectable optimized acellular (iOA) nerve graft retains native chemical cues such as collagens and glycosaminoglycans. By varying hydrogel concentration, the rheological properties and compressive modulus of iOA were similar to that previous reported for rat central nervous tissue. iOA solution was compatible with rat Schwann cells in culture, and hydrogel injection into a rat cervical contusion model significantly reduced the ratio of M1:M2 macrophages after one week, favoring regenerative phenotypes (p < 0.05). Furthermore, while iOA treatment did not affect locomotor or respiratory recovery over an eight week period, the percentage of axonal coverage increased at the distal tissue interface (p < 0.05), suggesting enhanced axonal extension within this region. SIGNIFICANCE Our data indicate that this novel injectable form of acellular nerve grafts is amenable for use after contusion SCI and may bolster a simultaneous therapy by acutely modulating the inflammatory milieu and supporting axonal growth.
Collapse
Affiliation(s)
- R. Chase Cornelison
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | | | - Stacy L. Porvasnik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | - Steven M. Wellman
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | - James H. Park
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | - David D. Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL 32611
| | - Christine E. Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| |
Collapse
|
46
|
May Z, Kumar R, Fuehrmann T, Tam R, Vulic K, Forero J, Lucas Osma A, Fenrich K, Assinck P, Lee MJ, Moulson A, Shoichet MS, Tetzlaff W, Biernaskie J, Fouad K. Adult skin-derived precursor Schwann cell grafts form growths in the injured spinal cord of Fischer rats. ACTA ACUST UNITED AC 2018; 13:034101. [PMID: 29068322 DOI: 10.1088/1748-605x/aa95f8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, GFP+ skin-derived precursor Schwann cells (SKP-SCs) from adult rats were grafted into the injured spinal cord of immunosuppressed rats. Our goal was to improve grafted cell survival in the injured spinal cord, which is typically low. Cells were grafted in hyaluronan-methylcellulose hydrogel (HAMC) or hyaluronan-methylcellulose modified with laminin- and fibronectin-derived peptide sequences (eHAMC). The criteria for selection of hyaluronan was for its shear-thinning properties, making the hydrogel easy to inject, methylcellulose for its inverse thermal gelation, helping to keep grafted cells in situ, and fibronectin and laminin to improve cell attachment and, thus, prevent cell death due to dissociation from substrate molecules (i.e., anoikis). Post-mortem examination revealed large masses of GFP+ SKP-SCs in the spinal cords of rats that received cells in HAMC (5 out of n = 8) and eHAMC (6 out of n = 8). Cell transplantation in eHAMC caused significantly greater spinal lesions compared to lesion and eHAMC only control groups. A parallel study showed similar masses in the contused spinal cord of rats after transplantation of adult GFP+ SKP-SCs without a hydrogel or immunosuppression. These findings suggest that adult GFP+ SKP-SCs, cultured/transplanted under the conditions described here, have a capacity for uncontrolled proliferation. Growth-formation in pre-clinical research has also been documented after transplantation of: human induced pluripotent stem cell-derived neural stem cells (Itakura et al 2015 PLoS One 10 e0116413), embryonic stem cells and embryonic stem cell-derived neurons (Brederlau et al 2006 Stem Cells 24 1433-40; Dressel et al 2008 PLoS One 3 e2622), bone marrow derived mesenchymal stem cells (Jeong et al 2011 Circ. Res. 108 1340-47) and rat nerve-derived SCs following in vitro expansion for >11 passages (Funk et al 2007 Eur. J. Cell Biol. 86 207-19; Langford et al 1988 J. Neurocytology 17 521-9; Morrissey et al 1991 J. Neurosci. 11 2433-42). It is of upmost importance to define the precise culture/transplantation parameters for maintenance of normal cell function and safe and effective use of cell therapy.
Collapse
Affiliation(s)
- Zacnicte May
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Thiry L, Lemieux M, Bretzner F. Age- and speed-dependent modulation of gaits in DSCAM 2J mutant mice. J Neurophysiol 2017; 119:723-737. [PMID: 29093169 DOI: 10.1152/jn.00471.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gaits depend on the interplay between distributed spinal neural networks, termed central pattern generators, generating rhythmic and coordinated movements, primary afferents, and descending supraspinal inputs. Recent studies demonstrated that the mouse displays a rich repertoire of gaits. Changes in gaits occur in mutant mice lacking particular neurons or molecular signaling pathways implicated in the normal establishment of these neural networks. Given the role of the Down syndrome cell adherence molecule (DSCAM) to the formation and maintenance of spinal interneuronal circuits and sensorimotor integration, we have investigated its functional contribution to gaits over a wide range of locomotor speeds using freely walking mice. We show in this study that the DSCAM2J mutation, while not precluding any gait, impairs the age- and speed-dependent modulation of gaits. It impairs the ability of mice to maintain their locomotion at high treadmill speeds. DSCAM2J mutation induces the dominance of lateral walk over trot and the emergence of aberrant gaits for mice, such as pace and diagonal walk. Gaits were also more labile in DSCAM2J mutant mice, i.e., less stable, less attractive, and less predictable than in their wild-type littermates. Our results suggest that the DSCAM mutation affects the behavioral repertoire of gaits in an age- and speed-dependent manner. NEW & NOTEWORTHY Gaits evolve throughout development, up to adulthood, and according to the genetic background. Using mutant mice lacking DSCAM (a cell adherence molecule associated with Down syndrome), we show that the DSCAM2J mutation alters the repertoire of gaits according to the mouse's age and speed, and prevents fast gaits. Such an incapacity suggests a reorganization of spinal, propriospinal, and supraspinal neuronal circuits underlying locomotor control in DSCAM2J mutant mice.
Collapse
Affiliation(s)
- Louise Thiry
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences, Quebec City, Quebec , Canada
| | - Maxime Lemieux
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences, Quebec City, Quebec , Canada
| | - Frédéric Bretzner
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences, Quebec City, Quebec , Canada.,Faculty of Medicine, Department of Psychiatry and Neurosciences, Université Laval , Quebec City, Quebec , Canada
| |
Collapse
|
48
|
Cyclosporine-immunosuppression does not affect survival of transplanted skin-derived precursor Schwann cells in the injured rat spinal cord. Neurosci Lett 2017; 658:67-72. [PMID: 28843345 DOI: 10.1016/j.neulet.2017.08.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/01/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022]
Abstract
A major goal of Schwann cell (SC) transplantation for spinal cord injury (SCI) is to fill the injury site to create a bridge for regenerating axons. However, transplantation of peripheral nerve SCs requires an invasive biopsy, which may result in nerve damage and donor site morbidity. SCs derived from multipotent stem cells found in skin dermis (SKP-SCs) are a promising alternative. Regardless of source, loss of grafted SCs post-grafting is an issue in studies of regeneration, with survival rates ranging from ∼1 to 20% after ≥6 weeks in rodent models of SCI. Immune rejection has been implicated in these low survival rates. Therefore, our aim was to explore the role of the immune response on grafted SKP-SC survival in Fischer rats with a spinal hemisection injury. We compared SKP-SC survival 6 weeks post-transplantation in: (I) cyclosporine-immunosuppressed rats (n=8), (II) immunocompetent rats (n=9), and (III) rats of a different sub-strain than the SKP-SC donor rats (n=7). SKP-SC survival was similar in all groups, suggesting immune rejection was not a main factor in SKP-SC loss observed in this study. SKP-SCs were consistently found on laminin expressed at the injury site, indicating detachment-mediated apoptosis (i.e., anoikis) might play a major role in grafted cell loss.
Collapse
|
49
|
Myelinogenic Plasticity of Oligodendrocyte Precursor Cells following Spinal Cord Contusion Injury. J Neurosci 2017; 37:8635-8654. [PMID: 28760862 DOI: 10.1523/jneurosci.2409-16.2017] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 06/22/2017] [Accepted: 07/22/2017] [Indexed: 12/17/2022] Open
Abstract
Spontaneous remyelination occurs after spinal cord injury (SCI), but the extent of myelin repair and identity of the cells responsible remain incompletely understood and contentious. We assessed the cellular origin of new myelin by fate mapping platelet-derived growth factor receptor α (PDGFRα), Olig2+, and P0+ cells following contusion SCI in mice. Oligodendrocyte precursor cells (OPCs; PDGFRα+) produced oligodendrocytes responsible for de novo ensheathment of ∼30% of myelinated spinal axons at injury epicenter 3 months after SCI, demonstrating that these resident cells are a major contributor to oligodendrocyte regeneration. OPCs also produced the majority of myelinating Schwann cells in the injured spinal cord; invasion of peripheral myelinating (P0+) Schwann cells made only a limited contribution. These findings reveal that PDGFRα+ cells perform diverse roles in CNS repair, as multipotential progenitors that generate both classes of myelinating cells. This endogenous repair might be exploited as a therapeutic target for CNS trauma and disease.SIGNIFICANCE STATEMENT Spinal cord injury (SCI) leads to profound functional deficits, though substantial numbers of axons often survive. One possible explanation for these deficits is loss of myelin, creating conduction block at the site of injury. SCI leads to oligodendrocyte death and demyelination, and clinical trials have tested glial transplants to promote myelin repair. However, the degree and duration of myelin loss, and the extent and mechanisms of endogenous repair, have been contentious issues. Here, we use genetic fate mapping to demonstrate that spontaneous myelin repair by endogenous oligodendrocyte precursors is much more robust than previously recognized. These findings are relevant to many types of CNS pathology, raising the possibility that CNS precursors could be manipulated to repair myelin in lieu of glial transplantation.
Collapse
|
50
|
Comparative Analysis of the Cell Fates of Induced Schwann Cells from Subcutaneous Fat Tissue and Naïve Schwann Cells in the Sciatic Nerve Injury Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1252851. [PMID: 28713821 PMCID: PMC5496110 DOI: 10.1155/2017/1252851] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/14/2017] [Indexed: 01/11/2023]
Abstract
PURPOSE The fate and function of the induced Schwann cells (iSCs) like cells from adipose tissue have not been critically evaluated in vivo after transplantation. The objective of this study is to compare the fate of iSCs with naïve SCs (nSCs) after transplantation into the lesion sites of sciatic nerve, respectively. METHODS Adipose-derived stem cells from eGFP-expressing transgenic rat's subcutaneous fat were induced to iSCs in vitro. iSCs were injected to the sciatic nerve lesion area after crush injury and the cells fate was comparatively analyzed with that of nSCs from the same rat. RESULTS At 12 weeks after transplantation, nSCs were detected only in the restricted area of cell transplantation site but iSCs were widely distributed all over the sciatic nerve. Based on double fluorescence observations, both iSCs and naïve ones were colocalized with P0-expressing myelin sheath, outbound by laminin-expressing basal membrane, and terminated at contactin-associated protein-expressing doublets. However, some of iSCs were also differentiated to the fibrocyte/fibroblast-like cells. In the histological analysis of repaired sciatic nerves, axon density was higher in iSC-received group than in the nSCs group and normal sciatic nerve. CONCLUSION iSCs induced from subcutaneous fat tissues have higher engraftment and migration capacity than nSCs.
Collapse
|