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 DOI: 10.4103/nrr.nrr-d-23-01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [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
|
Zhu J, Ouyang X, Liu Y, Qian Y, Chen Y, Xu B. LncRNA GAS5 modulates Schwann cell function and enhances facial nerve injury repair via the miR-138-5p/CXCL12 axis. J Mol Histol 2024; 55:741-752. [PMID: 39068616 DOI: 10.1007/s10735-024-10227-z] [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: 02/05/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Facial nerve is an integral part of peripheral nerve. Schwann cells are important microglia involved in the repair and regulation of facial nerve injury. LncRNA growth arrest‑specific transcript 5 (GAS5) is involved in the behavioral regulation of Schwann cell and the regeneration of peripheral nervous system. However, there is little research about the effect of GAS5 on the repair of facial nerve injury (FNI) by regulating Schwann cells. This study aimed to investigate the role of GAS5 in Schwann cell function and FNI repair, focusing on the miR-138-5p/CXCL12 axis. Hematoxylin and eosin staining, Luxol fast blue staining, transmission electron microscope, and immunofluorescence (IF) experiments were used to verify the effect of GAS5 on FNI rats. Reverse transcription real-time polymerase chain reaction was performed to detect GAS5, miR-138-5p, and C-X-C motif chemokine ligand 12 (CXCL12) mRNA expression. IF staining was used to detect the inflorescence of S100 calcium binding protein B (S100β), SRY-box transcription factor 10 (SOX10), and tubulin beta 3 class III (β-Tubulin III). Glial fibrillary acidic protein (GFAP), nerve growth factor receptor (NGFR), S100β, brain derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and CXCL12 proteins were detected using western blot. The 5-bromo-2'-deoxyuridine staining, Transwell, and flow cytometry assays were conducted to detect Schwann cell function. Dual-luciferase, RNA immunoprecipitation, and RNA pulldown assay were used to identify the interaction among GAS5, miR-138-5p, and CXCL12. Results found that GAS5 was downregulated in facial nerve tissues of FNI rats. Overexpressed GAS5 decreased facial grading, inhibited demyelination, and promoted proliferation, migration, and suppressed apoptosis of Schwann cells. Mechanistically, GAS5 was a sponge of miR-138-5p and positively regulated CXCL12 expression. GAS5 inhibition repressed CXCL12 expression and decreased cell proliferation and migration, increased apoptosis rate of Schwann cells by sponging miR-138-5p. In conclusion, overexpression of GAS5 accelerates facial nerve repair in FNI rats by regulating miR-138-5p/CXCL12 axis.
Collapse
Affiliation(s)
- Jin Zhu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, 1088 HaiYuan middle road, Kunming, 650106, Yunnan, China
| | - Xin Ouyang
- Stomatology Centre, The First People's Hospital of Yunnan Province, Kunming, 650032, Yunnan, China
| | - Yu Liu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, 1088 HaiYuan middle road, Kunming, 650106, Yunnan, China
| | - Yemei Qian
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, 1088 HaiYuan middle road, Kunming, 650106, Yunnan, China
| | - Yuancan Chen
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, 1088 HaiYuan middle road, Kunming, 650106, Yunnan, China
| | - Biao Xu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital of Kunming Medical University, 1088 HaiYuan middle road, Kunming, 650106, Yunnan, China.
| |
Collapse
|
3
|
Du X, Zhang S, Khabbaz A, Cohen KL, Zhang Y, Chakraborty S, Smith GM, Wang H, Yadav AP, Liu N, Deng L. Regeneration of Propriospinal Axons in Rat Transected Spinal Cord Injury through a Growth-Promoting Pathway Constructed by Schwann Cells Overexpressing GDNF. Cells 2024; 13:1160. [PMID: 38995011 PMCID: PMC11240522 DOI: 10.3390/cells13131160] [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: 04/13/2024] [Revised: 06/01/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Unsuccessful axonal regeneration in transected spinal cord injury (SCI) is mainly attributed to shortage of growth factors, inhibitory glial scar, and low intrinsic regenerating capacity of severely injured neurons. Previously, we constructed an axonal growth permissive pathway in a thoracic hemisected injury by transplantation of Schwann cells overexpressing glial-cell-derived neurotrophic factor (SCs-GDNF) into the lesion gap as well as the caudal cord and proved that this novel permissive bridge promoted the regeneration of descending propriospinal tract (dPST) axons across and beyond the lesion. In the current study, we subjected rats to complete thoracic (T11) spinal cord transections and examined whether these combinatorial treatments can support dPST axons' regeneration beyond the transected injury. The results indicated that GDNF significantly improved graft-host interface by promoting integration between SCs and astrocytes, especially the migration of reactive astrocyte into SCs-GDNF territory. The glial response in the caudal graft area has been significantly attenuated. The astrocytes inside the grafted area were morphologically characterized by elongated and slim process and bipolar orientation accompanied by dramatically reduced expression of glial fibrillary acidic protein. Tremendous dPST axons have been found to regenerate across the lesion and back to the caudal spinal cord which were otherwise difficult to see in control groups. The caudal synaptic connections were formed, and regenerated axons were remyelinated. The hindlimb locomotor function has been improved.
Collapse
Affiliation(s)
- Xiaolong Du
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210005, China
| | - Shengqi Zhang
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing 210009, China;
| | - Aytak Khabbaz
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kristen Lynn Cohen
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yihong Zhang
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Samhita Chakraborty
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George M. Smith
- Shriners Hospitals Pediatric Research Center, School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing 210009, China;
| | - Amol P. Yadav
- Department of Biomedical Engineering, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Naikui Liu
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lingxiao Deng
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.D.); (S.Z.); (A.K.); (K.L.C.); (Y.Z.); (S.C.)
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Goodman and Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
4
|
Agarwal G, Shumard S, McCrary MW, Osborne O, Santiago JM, Ausec B, Schmidt CE. Decellularized porcine peripheral nerve based injectable hydrogels as a Schwann cell carrier for injured spinal cord regeneration. J Neural Eng 2024; 21:046002. [PMID: 38885674 DOI: 10.1088/1741-2552/ad5939] [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: 01/29/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
Objective.To develop a clinically relevant injectable hydrogel derived from decellularized porcine peripheral nerves and with mechanical properties comparable to native central nervous system (CNS) tissue to be used as a delivery vehicle for Schwann cell transplantation to treat spinal cord injury (SCI).Approach.Porcine peripheral nerves (sciatic and peroneal) were decellularized by chemical decellularization using a sodium deoxycholate and DNase (SDD) method previously developed by our group. The decellularized nerves were delipidated using dichloromethane and ethanol solvent and then digested using pepsin enzyme to form injectable hydrogel formulations. Genipin was used as a crosslinker to enhance mechanical properties. The injectability, mechanical properties, and gelation kinetics of the hydrogels were further analyzed using rheology. Schwann cells encapsulated within the injectable hydrogel formulations were passed through a 25-gauge needle and cell viability was assessed using live/dead staining. The ability of the hydrogel to maintain Schwann cell viability against an inflammatory milieu was assessedin vitrousing inflamed astrocytes co-cultured with Schwann cells.Mainresults. The SDD method effectively removes cells and retains extracellular matrix in decellularized tissues. Using rheological studies, we found that delipidation of decellularized porcine peripheral nerves using dichloromethane and ethanol solvent improves gelation kinetics and mechanical strength of hydrogels. The delipidated and decellularized hydrogels crosslinked using genipin mimicked the mechanical strength of CNS tissue. The hydrogels were found to have shear thinning properties desirable for injectable formulations and they also maintained higher Schwann cell viability during injection compared to saline controls. Usingin vitroco-culture experiments, we found that the genipin-crosslinked hydrogels also protected Schwann cells from astrocyte-mediated inflammation.Significance. Injectable hydrogels developed using delipidated and decellularized porcine peripheral nerves are a potential clinically relevant solution to deliver Schwann cells, and possibly other therapeutic cells, at the SCI site by maintaining higher cellular viability and increasing therapeutic efficacy for SCI treatment.
Collapse
Affiliation(s)
- Gopal Agarwal
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Samantha Shumard
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Olivia Osborne
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Jorge Mojica Santiago
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Breanna Ausec
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32610, United States of America
| |
Collapse
|
5
|
Yang Y, Ma B, Chen J, Liu D, Ma J, Li B, Hao J, Zhou X. Epigenetic regulation and factors that influence the effect of iPSCs-derived neural stem/progenitor cells (NS/PCs) in the treatment of spinal cord injury. Clin Epigenetics 2024; 16:30. [PMID: 38383473 PMCID: PMC10880347 DOI: 10.1186/s13148-024-01639-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disorder that causes neurological impairment and disability. Neural stem/progenitor cells (NS/PCs) derived from induced pluripotent stem cells (iPSCs) represent a promising cell therapy strategy for spinal cord regeneration and repair. However, iPSC-derived NS/PCs face many challenges and issues in SCI therapy; one of the most significant challenges is epigenetic regulation and that factors that influence this mechanism. Epigenetics refers to the regulation of gene expression and function by DNA methylation, histone modification, and chromatin structure without changing the DNA sequence. Previous research has shown that epigenetics plays a crucial role in the generation, differentiation, and transplantation of iPSCs, and can influence the quality, safety, and outcome of transplanted cells. In this study, we review the effects of epigenetic regulation and various influencing factors on the role of iPSC-derived NS/PCs in SCI therapy at multiple levels, including epigenetic reprogramming, regulation, and the adaptation of iPSCs during generation, differentiation, and transplantation, as well as the impact of other therapeutic tools (e.g., drugs, electrical stimulation, and scaffolds) on the epigenetic status of transplanted cells. We summarize our main findings and insights in this field and identify future challenges and directions that need to be addressed and explored.
Collapse
Affiliation(s)
- Yubiao Yang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Boyuan Ma
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Jinyu Chen
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Derong Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jun Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jian Hao
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| | - Xianhu Zhou
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
6
|
Yari D, Saberi A, Salmasi Z, Ghoreishi SA, Etemad L, Movaffagh J, Ganjeifar B. Recent Advances in the Treatment of Spinal Cord Injury. THE ARCHIVES OF BONE AND JOINT SURGERY 2024; 12:380-399. [PMID: 38919744 PMCID: PMC11195032 DOI: 10.22038/abjs.2023.73944.3424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/07/2023] [Indexed: 06/27/2024]
Abstract
Spinal cord injury (SCI) is a complex, multifaceted, progressive, and yet incurable complication that can cause irreversible damage to the individual, family, and society. In recent years strategies for the management and rehabilitation of SCI besides axonal regeneration, remyelination, and neuronal plasticity of the injured spinal cord have significantly improved. Although most of the current research and therapeutic advances have been made in animal models, so far, no specific and complete treatment has been reported for SCI in humans. The failure to treat this complication has been due to the inherent neurological complexity and the structural, cellular, molecular, and biochemical characteristics of spinal cord injury. In this review, in addition to elucidating the causes of spinal cord injury from a molecular and pathophysiological perspective, the complexity and drawbacks of neural regeneration that lead to the failure in SCI treatment are described. Also, recent advances and cutting-edge strategies in most areas of SCI treatment are presented.
Collapse
Affiliation(s)
- Davood Yari
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Ghoreishi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Orthopedic Surgery, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jebrail Movaffagh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Ganjeifar
- Department of Neurosurgery, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Zhang F, Gao H, Jiang X, Yang F, Zhang J, Song S, Shen J. Biomedical Application of Decellularized Scaffolds. ACS APPLIED BIO MATERIALS 2023; 6:5145-5168. [PMID: 38032114 DOI: 10.1021/acsabm.3c00778] [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] [Indexed: 12/01/2023]
Abstract
Tissue loss and end-stage organ failure are serious health problems across the world. Natural and synthetic polymer scaffold material based artificial organs play an important role in the field of tissue engineering and organ regeneration, but they are not from the body and may cause side effects such as rejection. In recent years, the biomimetic decellularized scaffold based materials have drawn great attention in the tissue engineering field for their good biocompatibility, easy modification, and excellent organism adaptability. Therefore, in this review, we comprehensively summarize the application of decellularized scaffolds in tissue engineering and biomedicine in recent years. The preparation methods, modification strategies, construction of artificial tissues, and application in biomedical applications are discussed. We hope that this review will provide a useful reference for research on decellularized scaffolds and promote their application tissue engineering.
Collapse
Affiliation(s)
- Fang Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Huimin Gao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xuefeng Jiang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fang Yang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jun Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Saijie Song
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
- Jiangsu Engineering Research Center of Interfacial Chemistry, Nanjing University, Nanjing 210023, China
| |
Collapse
|
8
|
Ghosh M, Pearse DD. Schwann Cell-Derived Exosomal Vesicles: A Promising Therapy for the Injured Spinal Cord. Int J Mol Sci 2023; 24:17317. [PMID: 38139147 PMCID: PMC10743801 DOI: 10.3390/ijms242417317] [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: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles released by cells into their extracellular milieu. Within these nanovesicles reside a multitude of bioactive molecules, which orchestrate essential biological processes, including cell differentiation, proliferation, and survival, in the recipient cells. These bioactive properties of exosomes render them a promising choice for therapeutic use in the realm of tissue regeneration and repair. Exosomes possess notable positive attributes, including a high bioavailability, inherent safety, and stability, as well as the capacity to be functionalized so that drugs or biological agents can be encapsulated within them or to have their surface modified with ligands and receptors to imbue them with selective cell or tissue targeting. Remarkably, their small size and capacity for receptor-mediated transcytosis enable exosomes to cross the blood-brain barrier (BBB) and access the central nervous system (CNS). Unlike cell-based therapies, exosomes present fewer ethical constraints in their collection and direct use as a therapeutic approach in the human body. These advantageous qualities underscore the vast potential of exosomes as a treatment option for neurological injuries and diseases, setting them apart from other cell-based biological agents. Considering the therapeutic potential of exosomes, the current review seeks to specifically examine an area of investigation that encompasses the development of Schwann cell (SC)-derived exosomal vesicles (SCEVs) as an approach to spinal cord injury (SCI) protection and repair. SCs, the myelinating glia of the peripheral nervous system, have a long history of demonstrated benefit in repair of the injured spinal cord and peripheral nerves when transplanted, including their recent advancement to clinical investigations for feasibility and safety in humans. This review delves into the potential of utilizing SCEVs as a therapy for SCI, explores promising engineering strategies to customize SCEVs for specific actions, and examines how SCEVs may offer unique clinical advantages over SC transplantation for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
9
|
Aparicio GI, Monje PV. Human Schwann Cells in vitro I. Nerve Tissue Processing, Pre-degeneration, Isolation, and Culturing of Primary Cells. Bio Protoc 2023; 13:e4748. [PMID: 38023787 PMCID: PMC10665635 DOI: 10.21769/bioprotoc.4748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 12/01/2023] Open
Abstract
This paper presents versatile protocols to prepare primary human Schwann cell (hSC) cultures from mature peripheral nervous system tissues, including fascicles from long spinal nerves, nerve roots, and ganglia. This protocol starts with a description of nerve tissue procurement, handling, and dissection to obtain tissue sections suitable for hSC isolation and culturing. A description follows on how to disintegrate the nerve tissue by delayed enzymatic dissociation, plate the initial cell suspensions on a two-dimensional substrate, and culture the primary hSCs. Each section contains detailed procedures, technical notes, and background information to aid investigators in understanding and managing all steps. Some general recommendations are made to optimize the recovery, growth, and purity of the hSC cultures irrespective of the tissue source. These recommendations include: (1) pre-culturing epineurium- and perineurium-free nerve fascicles under conditions of adherence or suspension depending on the size of the explants to facilitate the release of proliferative, in vitro-activated hSCs; (2) plating the initial cell suspensions as individual droplets on a laminin-coated substrate to expedite cell adhesion and thereby increase the recovery of viable cells; and (3) culturing the fascicles (pre-degeneration step) and the cells derived therefrom in mitogen- and serum-supplemented medium to accelerate hSC dedifferentiation and promote mitogenesis before and after tissue dissociation, respectively. The hSC cultures obtained as suggested in this protocol are suitable for assorted basic and translational research applications. With the appropriate adaptations, donor-relevant hSC cultures can be prepared using fresh or postmortem tissue biospecimens of a wide range of types and sizes.
Collapse
Affiliation(s)
- Gabriela I. Aparicio
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Paula V. Monje
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
10
|
Monje PV. Human Schwann Cells in vitro III. Analytical Methods and a Practical Approach for Quality Control. Bio Protoc 2023; 13:e4840. [PMID: 38034849 PMCID: PMC10682955 DOI: 10.21769/bioprotoc.4840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 12/02/2023] Open
Abstract
This paper introduces simple analytical methods and bioassays to promptly assess the identity and function of in vitro cultured human Schwann cells (hSCs). A systematic approach is proposed to unequivocally discriminate hSCs from other glial cells, non-glial cells, and non-human SCs (authentication), identify hSCs at different stages of differentiation, and determine whether individual hSCs are proliferative or senescent. Examples of how to use distinct cell-based approaches for quality control and routine troubleshooting are provided to confirm the constitution (identity, purity, and heterogeneity) and potency (bioactivity) of hSC cultures from multiple sources. The bioassays are valuable for rapidly gauging the responses of hSCs to mitogenic and differentiating factors and ascertaining the cells' basic properties before performing co-culture or cell grafting studies. The assays are image based and use adherent hSCs established in monoculture to simplify the experimental setup and interpretation of results. Finally, all sections contain thorough background information, notes, and references to facilitate decision making, data interpretation, and ad hoc method development for diverse applications.
Collapse
Affiliation(s)
- Paula V. Monje
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
11
|
Monje PV. Human Schwann Cells in vitro II. Passaging, Purification, Banking, and Labeling of Established Cultures. Bio Protoc 2023; 13:e4882. [PMID: 38023793 PMCID: PMC10665714 DOI: 10.21769/bioprotoc.4882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
This manuscript describes step-by-step procedures to establish and manage fresh and cryopreserved cultures of nerve-derived human Schwann cells (hSCs) at the desired scale. Adaptable protocols are provided to propagate hSC cultures through serial passaging and perform routine manipulations such as enzymatic dissociation, purification, cryogenic preservation, live-cell labeling, and gene delivery. Expanded hSCs cultures are metabolically active, proliferative, and phenotypically stable for at least three consecutive passages. Cell yields are expected to be variable as determined by the rate of growth of individual batches and the rounds of subculture. The purity, however, can be maintained high at >95% hSC regardless of passage. The cells obtained in this manner are suitable for various applications, including small drug screens, in vitro modeling of neurodevelopmental processes, and cell transplantation. One caveat of this protocol is that continued expansion of same-batch hSC populations is eventually restricted due to senescence-linked growth arrest.
Collapse
Affiliation(s)
- Paula V. Monje
- Department of Neurosurgery, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
12
|
Garcia E, Buzoianu-Anguiano V, Silva-Garcia R, Esparza-Salazar F, Arriero-Cabañero A, Escandon A, Doncel-Pérez E, Ibarra A. Use of Cells, Supplements, and Peptides as Therapeutic Strategies for Modulating Inflammation after Spinal Cord Injury: An Update. Int J Mol Sci 2023; 24:13946. [PMID: 37762251 PMCID: PMC10531377 DOI: 10.3390/ijms241813946] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury is a traumatic lesion that causes a catastrophic condition in patients, resulting in neuronal deficit and loss of motor and sensory function. That loss is caused by secondary injury events following mechanical damage, which results in cell death. One of the most important events is inflammation, which activates molecules like proinflammatory cytokines (IL-1β, IFN-γ, and TNF-α) that provoke a toxic environment, inhibiting axonal growth and exacerbating CNS damage. As there is no effective treatment, one of the developed therapies is neuroprotection of the tissue to preserve healthy tissue. Among the strategies that have been developed are the use of cell therapy, the use of peptides, and molecules or supplements that have been shown to favor an anti-inflammatory environment that helps to preserve tissue and cells at the site of injury, thus favoring axonal growth and improved locomotor function. In this review, we will explain some of these strategies used in different animal models of spinal cord injury, their activity as modulators of the immune system, and the benefits they have shown.
Collapse
Affiliation(s)
- Elisa Garcia
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (E.G.); (F.E.-S.); (A.E.)
| | - Vinnitsa Buzoianu-Anguiano
- Grupo Regeneración Neural, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (V.B.-A.); (A.A.-C.)
| | - Raúl Silva-Garcia
- Unidad de Investigación Médica en Inmunología Hospital de Pediatría, CMN-SXXI, IMSS, Mexico City 06720, Mexico;
| | - Felipe Esparza-Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (E.G.); (F.E.-S.); (A.E.)
| | - Alejandro Arriero-Cabañero
- Grupo Regeneración Neural, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (V.B.-A.); (A.A.-C.)
| | - Adela Escandon
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (E.G.); (F.E.-S.); (A.E.)
| | - Ernesto Doncel-Pérez
- Grupo Regeneración Neural, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (V.B.-A.); (A.A.-C.)
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico; (E.G.); (F.E.-S.); (A.E.)
| |
Collapse
|
13
|
Zeng CW, Tsai HJ. The Promising Role of a Zebrafish Model Employed in Neural Regeneration Following a Spinal Cord Injury. Int J Mol Sci 2023; 24:13938. [PMID: 37762240 PMCID: PMC10530783 DOI: 10.3390/ijms241813938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating event that results in a wide range of physical impairments and disabilities. Despite the advances in our understanding of the biological response to injured tissue, no effective treatments are available for SCIs at present. Some studies have addressed this issue by exploring the potential of cell transplantation therapy. However, because of the abnormal microenvironment in injured tissue, the survival rate of transplanted cells is often low, thus limiting the efficacy of such treatments. Many studies have attempted to overcome these obstacles using a variety of cell types and animal models. Recent studies have shown the utility of zebrafish as a model of neural regeneration following SCIs, including the proliferation and migration of various cell types and the involvement of various progenitor cells. In this review, we discuss some of the current challenges in SCI research, including the accurate identification of cell types involved in neural regeneration, the adverse microenvironment created by SCIs, attenuated immune responses that inhibit nerve regeneration, and glial scar formation that prevents axonal regeneration. More in-depth studies are needed to fully understand the neural regeneration mechanisms, proteins, and signaling pathways involved in the complex interactions between the SCI microenvironment and transplanted cells in non-mammals, particularly in the zebrafish model, which could, in turn, lead to new therapeutic approaches to treat SCIs in humans and other mammals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
14
|
Liu Y, Yu H, Yu P, Peng P, Li C, Xiang Z, Ban D. Gelatin methacryloyl hydrogel scaffold loaded with activated Schwann cells attenuates apoptosis and promotes functional recovery following spinal cord injury. Exp Ther Med 2023; 25:144. [PMID: 36911380 PMCID: PMC9995797 DOI: 10.3892/etm.2023.11843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/21/2022] [Indexed: 02/17/2023] Open
Abstract
Spinal cord injury (SCI) is a refractory disease of the central nervous system with a high disability and incidence rate. In recent years, bioactive material combined with cell transplantation has been considered an effective method for the treatment of SCI. The present study encapsulated activated Schwann cells (ASCs) in a 3D gelatin methacryloyl (GelMA) hydrogel in order to investigate its therapeutic effects on SCI. ASCs were isolated from previously ligated rat sciatic nerves. Scanning electron microscopy and live/dead staining were used to evaluate the biocompatibility of hydrogels with the ASCs. The scaffold was transplanted into the spinal cord of rats in the hemisection model. Behavioral tests and hematoxylin and eosin staining were employed to assess the locomotion recovery and lesion areas before and after treatment. Cell apoptosis was evaluated using TUNEL staining and immunochemistry, and apoptosis-related protein expression was detected using western blot analysis. The ASCs exhibited a favorable survival and proliferative ability in the 3D GelMA hydrogel. The scaffold transplantation significantly reduced the cavities and improved functional recovery. Moreover, the GelMA/ASCs implants significantly inhibited cell apoptosis following SCI and this effect may be mediated via the p38 MAPK pathway. Overall, these findings indicated that ASCs combined with the 3D GelMA hydrogel may be a promising therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Hao Yu
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Peng Yu
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Peng Peng
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Chao Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Zhenyang Xiang
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| | - Dexiang Ban
- Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Heping, Tianjin 300052, P.R. China
| |
Collapse
|
15
|
Aparicio GI, León A, Gutiérrez Fuster R, Ravenscraft B, Monje PV, Scorticati C. Endogenous Glycoprotein GPM6a Is Involved in Neurite Outgrowth in Rat Dorsal Root Ganglion Neurons. Biomolecules 2023; 13:biom13040594. [PMID: 37189342 DOI: 10.3390/biom13040594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
The peripheral nervous system (PNS) has a unique ability for self-repair. Dorsal root ganglion (DRG) neurons regulate the expression of different molecules, such as neurotrophins and their receptors, to promote axon regeneration after injury. However, the molecular players driving axonal regrowth need to be better defined. The membrane glycoprotein GPM6a has been described to contribute to neuronal development and structural plasticity in central-nervous-system neurons. Recent evidence indicates that GPM6a interacts with molecules from the PNS, although its role in DRG neurons remains unknown. Here, we characterized the expression of GPM6a in embryonic and adult DRGs by combining analysis of public RNA-seq datasets with immunochemical approaches utilizing cultures of rat DRG explants and dissociated neuronal cells. M6a was detected on the cell surfaces of DRG neurons throughout development. Moreover, GPM6a was required for DRG neurite elongation in vitro. In summary, we provide evidence on GPM6a being present in DRG neurons for the first time. Data from our functional experiments support the idea that GPM6a could contribute to axon regeneration in the PNS.
Collapse
|
16
|
A Multi-Stage Bioprocess for the Expansion of Rodent Skin-Derived Schwann Cells in Computer-Controlled Bioreactors. Int J Mol Sci 2023; 24:ijms24065152. [PMID: 36982227 PMCID: PMC10049355 DOI: 10.3390/ijms24065152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
Regenerative therapies for the treatment of peripheral nerve and spinal cord injuries can require hundreds of millions of autologous cells. Current treatments involve the harvest of Schwann cells (SCs) from nerves; however, this is an invasive procedure. Therefore, a promising alternative is using skin-derived Schwann cells (Sk-SCs), in which between 3–5 million cells can be harvested from a standard skin biopsy. However, traditional static planar culture is still inefficient at expanding cells to clinically relevant numbers. As a result, bioreactors can be used to develop reproducible bioprocesses for the large-scale expansion of therapeutic cells. Here, we present a proof-of-concept SC manufacturing bioprocess using rat Sk-SCs. With this integrated process, we were able to simulate a feasible bioprocess, taking into consideration the harvest and shipment of cells to a production facility, the generation of the final cell product, and the cryopreservation and shipment of cells back to the clinic and patient. This process started with 3 million cells and inoculated and expanded them to over 200 million cells in 6 days. Following the harvest and post-harvest cryopreservation and thaw, we were able to maintain 150 million viable cells that exhibited a characteristic Schwann cell phenotype throughout each step of the process. This process led to a 50-fold expansion, producing a clinically relevant number of cells in a 500 mL bioreactor in just 1 week, which is a dramatic improvement over current methods of expansion.
Collapse
|
17
|
Khaing ZZ, Chen JY, Safarians G, Ezubeik S, Pedroncelli N, Duquette RD, Prasse T, Seidlits SK. Clinical Trials Targeting Secondary Damage after Traumatic Spinal Cord Injury. Int J Mol Sci 2023; 24:3824. [PMID: 36835233 PMCID: PMC9960771 DOI: 10.3390/ijms24043824] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Spinal cord injury (SCI) often causes loss of sensory and motor function resulting in a significant reduction in quality of life for patients. Currently, no therapies are available that can repair spinal cord tissue. After the primary SCI, an acute inflammatory response induces further tissue damage in a process known as secondary injury. Targeting secondary injury to prevent additional tissue damage during the acute and subacute phases of SCI represents a promising strategy to improve patient outcomes. Here, we review clinical trials of neuroprotective therapeutics expected to mitigate secondary injury, focusing primarily on those in the last decade. The strategies discussed are broadly categorized as acute-phase procedural/surgical interventions, systemically delivered pharmacological agents, and cell-based therapies. In addition, we summarize the potential for combinatorial therapies and considerations.
Collapse
Affiliation(s)
- Zin Z. Khaing
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Jessica Y. Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Gevick Safarians
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sohib Ezubeik
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Nicolas Pedroncelli
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rebecca D. Duquette
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Tobias Prasse
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
- Department of Orthopedics and Trauma Surgery, University of Cologne, 50931 Cologne, Germany
| | - Stephanie K. Seidlits
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
18
|
Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032528. [PMID: 36768846 PMCID: PMC9917245 DOI: 10.3390/ijms24032528] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
Spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically, with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in preclinical research and clinical trials. In the near future, several more are expected to come down the translational pipeline. Among ongoing and completed trials are those reporting the use of biomaterial scaffolds. The advancements in biomaterial technology, combined with stem cell therapy or other regenerative therapy, can now accelerate the progress of promising novel therapeutic strategies from bench to bedside. Various types of approaches to regeneration therapy for SCI have been combined with the use of supportive biomaterial scaffolds as a drug and cell delivery system to facilitate favorable cell-material interactions and the supportive effect of neuroprotection. In this review, we summarize some of the most recent insights of preclinical and clinical studies using biomaterial scaffolds in regenerative therapy for SCI and summarized the biomaterial strategies for treatment with simplified results data. One hundred and sixty-eight articles were selected in the present review, in which we focused on biomaterial scaffolds. We conducted our search of articles using PubMed and Medline, a medical database. We used a combination of "Spinal cord injury" and ["Biomaterial", or "Scaffold"] as search terms and searched articles published up until 30 April 2022. Successful future therapies will require these biomaterial scaffolds and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, the loss of a structural framework, and biocompatibility. This database could serve as a benchmark to progress in future clinical trials for SCI using biomaterial scaffolds.
Collapse
|
19
|
Zheng B, Tuszynski MH. Regulation of axonal regeneration after mammalian spinal cord injury. Nat Rev Mol Cell Biol 2023; 24:396-413. [PMID: 36604586 DOI: 10.1038/s41580-022-00562-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 01/06/2023]
Abstract
One hundred years ago, Ramón y Cajal, considered by many as the founder of modern neuroscience, stated that neurons of the adult central nervous system (CNS) are incapable of regenerating. Yet, recent years have seen a tremendous expansion of knowledge in the molecular control of axon regeneration after CNS injury. We now understand that regeneration in the adult CNS is limited by (1) a failure to form cellular or molecular substrates for axon attachment and elongation through the lesion site; (2) environmental factors, including inhibitors of axon growth associated with myelin and the extracellular matrix; (3) astrocyte responses, which can both limit and support axon growth; and (4) intraneuronal mechanisms controlling the establishment of an active cellular growth programme. We discuss these topics together with newly emerging hypotheses, including the surprising finding from transcriptomic analyses of the corticospinal system in mice that neurons revert to an embryonic state after spinal cord injury, which can be sustained to promote regeneration with neural stem cell transplantation. These gains in knowledge are steadily advancing efforts to develop effective treatment strategies for spinal cord injury in humans.
Collapse
Affiliation(s)
- Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| | - Mark H Tuszynski
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| |
Collapse
|
20
|
Wu Y, Tang Z, Zhang J, Wang Y, Liu S. Restoration of spinal cord injury: From endogenous repairing process to cellular therapy. Front Cell Neurosci 2022; 16:1077441. [PMID: 36523818 PMCID: PMC9744968 DOI: 10.3389/fncel.2022.1077441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 09/26/2023] Open
Abstract
Spinal cord injury (SCI) disrupts neurological pathways and impacts sensory, motor, and autonomic nerve function. There is no effective treatment for SCI currently. Numerous endogenous cells, including astrocytes, macrophages/microglia, and oligodendrocyte, are involved in the histological healing process following SCI. By interfering with cells during the SCI repair process, some advancements in the therapy of SCI have been realized. Nevertheless, the endogenous cell types engaged in SCI repair and the current difficulties these cells confront in the therapy of SCI are poorly defined, and the mechanisms underlying them are little understood. In order to better understand SCI and create new therapeutic strategies and enhance the clinical translation of SCI repair, we have comprehensively listed the endogenous cells involved in SCI repair and summarized the six most common mechanisms involved in SCI repair, including limiting the inflammatory response, protecting the spared spinal cord, enhancing myelination, facilitating neovascularization, producing neurotrophic factors, and differentiating into neural/colloidal cell lines.
Collapse
Affiliation(s)
| | | | | | | | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
22
|
Chu TH, Baral K, Labit E, Rosin N, Sinha S, Umansky D, Alzahrani S, Rancourt D, Biernaskie J, Midha R. Comparison of human skin- and nerve-derived Schwann cells reveals many similarities and subtle genomic and functional differences. Glia 2022; 70:2131-2156. [PMID: 35796321 DOI: 10.1002/glia.24242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 11/08/2022]
Abstract
Skin is an easily accessible tissue and a rich source of Schwann cells (SCs). Toward potential clinical application of autologous SC therapies, we aim to improve the reliability and specificity of our protocol to obtain SCs from small skin samples. As well, to explore potential functional distinctions between skin-derived SCs (Sk-SCs) and nerve-derived SCs (N-SCs), we used single-cell RNA-sequencing and a series of in vitro and in vivo assays. Our results showed that Sk-SCs expressed typical SC markers. Single-cell sequencing of Sk- and N-SCs revealed an overwhelming overlap in gene expression with the exception of HLA genes which were preferentially up-regulated in Sk-SCs. In vitro, both cell types exhibited similar levels of proliferation, migration, uptake of myelin debris and readily associated with neurites when co-cultured with human iPSC-induced motor neurons. Both exhibited ensheathment of multiple neurites and early phase of myelination, especially in N-SCs. Interestingly, dorsal root ganglion (DRG) neurite outgrowth assay showed substantially more complexed neurite outgrowth in DRGs exposed to Sk-SC conditioned media compared to those from N-SCs. Multiplex ELISA array revealed shared growth factor profiles, but Sk-SCs expressed a higher level of VEGF. Transplantation of Sk- and N-SCs into injured peripheral nerve in nude rats and NOD-SCID mice showed close association of both SCs to regenerating axons. Myelination of rodent axons was observed infrequently by N-SCs, but absent in Sk-SC xenografts. Overall, our results showed that Sk-SCs share near-identical properties to N-SCs but with subtle differences that could potentially enhance their therapeutic utility.
Collapse
Affiliation(s)
- Tak-Ho Chu
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kabita Baral
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elodie Labit
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel Umansky
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Saud Alzahrani
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Derrick Rancourt
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Xu X, Liang Z, Lin Y, Rao J, Lin F, Yang Z, Wang R, Chen C. Comparing the Efficacy and Safety of Cell Transplantation for Spinal Cord Injury: A Systematic Review and Bayesian Network Meta-Analysis. Front Cell Neurosci 2022; 16:860131. [PMID: 35444516 PMCID: PMC9013778 DOI: 10.3389/fncel.2022.860131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo compare the safety and effectiveness of transplanted cells from different sources for spinal cord injury (SCI).DesignA systematic review and Bayesian network meta-analysis.Data SourcesMedline, Embase, and the Cochrane Central Register of Controlled Trials.Study SelectionWe included randomized controlled trials, case–control studies, and case series related to cell transplantation for SCI patients, that included at least 1 of the following outcome measures: American Spinal Cord Injury Association (ASIA) Impairment Scale (AIS grade), ASIA motor score, ASIA sensory score, the Functional Independence Measure score (FIM), International Association of Neurorestoratology Spinal Cord Injury Functional Rating Scale (IANR-SCIFRS), or adverse events. Follow-up data were analyzed at 6 and 12 months.ResultsForty-four eligible trials, involving 1,266 patients, investigated 6 treatments: olfactory ensheathing cells (OECs), neural stem cells/ neural progenitor cells (NSCs), mesenchymal stem cells (MSCs), Schwann cells, macrophages, and combinations of cells (MSCs plus Schwann cells). Macrophages improved the AIS grade at 12 months (mean 0.42, 95% credible interval: 0–0.91, low certainty) and FIM score at 12 months (42.83, 36.33–49.18, very low certainty). MSCs improved the AIS grade at 6 months (0.42, 0.15–0.73, moderate certainty), the motor score at 6 months (4.43, 0.91–7.78, moderate certainty), light touch at 6 (10.01, 5.81–13.88, moderate certainty) and 12 months (11.48, 6.31–16.64, moderate certainty), pinprick score at 6 (14.54, 9.76–19.46, moderate certainty) and 12 months (12.48, 7.09–18.12, moderate certainty), and the IANR-SCIFRS at 6 (3.96, 0.62–6.97, moderate certainty) and 12 months (5.54, 2.45–8.42, moderate certainty). OECs improved the FIM score at 6 months (9.35, 1.71–17.00, moderate certainty). No intervention improved the motor score significantly at 12 months. The certainty of other interventions was low or very low. Overall, the number of adverse events associated with transplanted cells was low.ConclusionsPatients with SCI who receive transplantation of macrophages, MSCs, NSCs, or OECs may have improved disease prognosis. MSCs are the primary recommendations. Further exploration of the mechanism of cell transplantation in the treatment of SCI, transplantation time window, transplantation methods, and monitoring of the number of transplanted cells and cell survival is needed.Systematic Review Registrationhttps://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier: CRD 42021282043.
Collapse
|
24
|
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: 4.5] [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
|