1
|
Zhou Q, Fang Q, Zhang C, Liu W, Sun Y. BDNF-GABA signaling in astrocytes: enhancing neural repair after SCI through MSC therapies. Spinal Cord 2025:10.1038/s41393-025-01077-x. [PMID: 40229538 DOI: 10.1038/s41393-025-01077-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
STUDY DESIGN An integrated bioinformatics data study. OBJECTIVE This study, through bioinformatics analysis, aims to map the landscape of astrocytes, explore key signaling pathways, and uncover molecular mechanisms that support SCI recovery facilitated by MSCs and iPSCs. SETTING Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University. METHODS We performed a bioinformatics analysis of single-cell transcriptomes (scRNA-seq), spatial transcriptomics, and bulk RNA-seq data sourced from Gene Expression Omnibus (GEO) datasets. The data processing involved R packages like "Seurat," "DESeq2," and "WGCNA." For pathway enrichment, we used Gene Set Enrichment Analysis (GSEA) and the Enrichr web server. RESULTS Single-cell and spatial transcriptomic analysis revealed notable changes in the astrocyte landscape after SCI, highlighting a significant disruption in astrocyte populations within the injured region. Findings suggest that BDNF regulation of GABA neurotransmission and GABA receptor signaling in astrocytes plays a key role in promoting neuronal regeneration. Additionally, hUC-MSCs were found to enhance neural repair by activating BDNF-regulated GABA signaling of astrocytes. A promising alternative involves iPS-derived MSCs, which have shown potential to boost neural regeneration through BDNF, GABA, and GABA receptor signaling pathways of astrocytes. CONCLUSIONS In summary, SCI disrupts astrocyte populations, impacting their ability to support neural repair. BDNF-regulated GABA signaling in astrocytes is essential for neuron regeneration. Both hUC-MSCs and iPS-derived MSCs show promise in enhancing neural recovery by activating these pathways, offering potential new therapeutic options for SCI.
Collapse
Affiliation(s)
- Qingsheng Zhou
- Department of Spine Surgery, Yantaishan Hospital, Binzhou Medical University, 10087 Science and Technology Avenue, Yantai, Shandong, 264003, P. R. China
| | - Qiongxuan Fang
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Chunming Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, P. R. China
| | - Wei Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, P. R. China
| | - Yifeng Sun
- Orthopaedic and Sports Medicine Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing,102218, P. R. China.
| |
Collapse
|
2
|
Abbas A, Huang X, Ullah A, Luo L, Xi W, Qiao Y, Zeng K. Enhanced spinal cord repair using bioengineered induced pluripotent stem cell-derived exosomes loaded with miRNA. Mol Med 2024; 30:168. [PMID: 39354344 PMCID: PMC11446086 DOI: 10.1186/s10020-024-00940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND A spinal cord injury (SCI) can result in severe impairment and fatality as well as significant motor and sensory abnormalities. Exosomes produced from IPSCs have demonstrated therapeutic promise for accelerating spinal cord injury recovery, according to a recent study. OBJECTIVE This study aims to develop engineered IPSCs-derived exosomes (iPSCs-Exo) capable of targeting and supporting neurons, and to assess their therapeutic potential in accelerating recovery from spinal cord injury (SCI). METHODS iPSCs-Exo were characterized using Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and western blot. To enhance neuronal targeting, iPSCs-Exo were bioengineered, and their uptake by neurons was visualized using PKH26 labeling and fluorescence microscopy. In vitro, the anti-inflammatory effects of miRNA-loaded engineered iPSCs-Exo were evaluated by exposing neurons to LPS and IFN-γ. In vivo, biodistribution of engineered iPSC-Exo was monitored using a vivo imaging system. The therapeutic efficacy of miRNA-loaded engineered iPSC-Exo in a SCI mouse model was assessed by Basso Mouse Scale (BMS) scores, H&E, and Luxol Fast Blue (LFB) staining. RESULTS The results showed that engineered iPSC-Exo loaded with miRNA promoted the spinal cord injure recovery. Thorough safety assessments using H&E staining on major organs revealed no evidence of systemic toxicity, with normal organ histology and biochemistry profiles following engineered iPSC-Exo administration. CONCLUSION These results suggest that modified iPSC-derived exosomes loaded with miRNA have great potential as a cutting-edge therapeutic approach to improve spinal cord injury recovery. The observed negligible systemic toxicity further underscores their potential safety and efficacy in clinical applications.
Collapse
Affiliation(s)
- Azar Abbas
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, 518055, P.R. China
| | - Xiaosheng Huang
- Department of Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Affiliated Hospital of Jinan University, No. 18, Zetian Road, Futian District, Shenzhen, Guangdong Province, 518040, P.R. China
| | - Aftab Ullah
- School of Medicine, Huaqiao University, No. 269, Chenghua North Road, Fengze District, Quanzhou, Fujian, 362021, P.R. China
| | - Lishi Luo
- Department of Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Affiliated Hospital of Jinan University, No. 18, Zetian Road, Futian District, Shenzhen, Guangdong Province, 518040, P.R. China
| | - Wenqun Xi
- Department of Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Affiliated Hospital of Jinan University, No. 18, Zetian Road, Futian District, Shenzhen, Guangdong Province, 518040, P.R. China
| | - Yuanjiao Qiao
- Department of Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Affiliated Hospital of Jinan University, No. 18, Zetian Road, Futian District, Shenzhen, Guangdong Province, 518040, P.R. China
| | - Kun Zeng
- Department of Shenzhen Eye Hospital, Shenzhen Key Laboratory of Ophthalmology, Affiliated Hospital of Jinan University, No. 18, Zetian Road, Futian District, Shenzhen, Guangdong Province, 518040, P.R. China.
| |
Collapse
|
3
|
Kim JW, Kim J, Lee SM, Rim YA, Sung YC, Nam Y, Kim HJ, Kim H, Jung SI, Lim J, Ju JH. Combination of induced pluripotent stem cell-derived motor neuron progenitor cells with irradiated brain-derived neurotrophic factor over-expressing engineered mesenchymal stem cells enhanced restoration of axonal regeneration in a chronic spinal cord injury rat model. Stem Cell Res Ther 2024; 15:173. [PMID: 38886817 PMCID: PMC11184802 DOI: 10.1186/s13287-024-03770-9] [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: 11/28/2023] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a disease that causes permanent impairment of motor, sensory, and autonomic nervous system functions. Stem cell transplantation for neuron regeneration is a promising strategic treatment for SCI. However, selecting stem cell sources and cell transplantation based on experimental evidence is required. Therefore, this study aimed to investigate the efficacy of combination cell transplantation using the brain-derived neurotrophic factor (BDNF) over-expressing engineered mesenchymal stem cell (BDNF-eMSC) and induced pluripotent stem cell-derived motor neuron progenitor cell (iMNP) in a chronic SCI rat model. METHOD A contusive chronic SCI was induced in Sprague-Dawley rats. At 6 weeks post-injury, BDNF-eMSC and iMNP were transplanted into the lesion site via the intralesional route. At 12 weeks post-injury, differentiation and growth factors were evaluated through immunofluorescence staining and western blot analysis. Motor neuron differentiation and neurite outgrowth were evaluated by co-culturing BDNF-eMSC and iMNP in vitro in 2-dimensional and 3-dimensional. RESULTS Combination cell transplantation in the chronic SCI model improved behavioral recovery more than single-cell transplantation. Additionally, combination cell transplantation enhanced mature motor neuron differentiation and axonal regeneration at the injured spinal cord. Both BDNF-eMSC and iMNP played a critical role in neurite outgrowth and motor neuron maturation via BDNF expression. CONCLUSIONS Our results suggest that the combined transplantation of BDNF- eMSC and iMNP in chronic SCI results in a significant clinical recovery. The transplanted iMNP cells predominantly differentiated into mature motor neurons. Additionally, BDNF-eMSC exerts a paracrine effect on neuron regeneration through BDNF expression in the injured spinal cord.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Juryun Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Yoojun Nam
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Hyewon Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | - Se In Jung
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jooyoung Lim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- YiPSCELL, Inc., Seoul, Republic of Korea.
| |
Collapse
|
4
|
Kim JW, Kim J, Mo H, Han H, Rim YA, Ju JH. Stepwise combined cell transplantation using mesenchymal stem cells and induced pluripotent stem cell-derived motor neuron progenitor cells in spinal cord injury. Stem Cell Res Ther 2024; 15:114. [PMID: 38650015 PMCID: PMC11036722 DOI: 10.1186/s13287-024-03714-3] [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: 05/10/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is an intractable neurological disease in which functions cannot be permanently restored due to nerve damage. Stem cell therapy is a promising strategy for neuroregeneration after SCI. However, experimental evidence of its therapeutic effect in SCI is lacking. This study aimed to investigate the efficacy of transplanted cells using stepwise combined cell therapy with human mesenchymal stem cells (hMSC) and induced pluripotent stem cell (iPSC)-derived motor neuron progenitor cells (iMNP) in a rat model of SCI. METHODS A contusive SCI model was developed in Sprague-Dawley rats using multicenter animal spinal cord injury study (MASCIS) impactor. Three protocols were designed and conducted as follows: (Subtopic 1) chronic SCI + iMNP, (Subtopic 2) acute SCI + multiple hMSC injections, and (Main topic) chronic SCI + stepwise combined cell therapy using multiple preemptive hMSC and iMNP. Neurite outgrowth was induced by coculturing hMSC and iPSC-derived motor neuron (iMN) on both two-dimensional (2D) and three-dimensional (3D) spheroid platforms during mature iMN differentiation in vitro. RESULTS Stepwise combined cell therapy promoted mature motor neuron differentiation and axonal regeneration at the lesional site. In addition, stepwise combined cell therapy improved behavioral recovery and was more effective than single cell therapy alone. In vitro results showed that hMSC and iMN act synergistically and play a critical role in the induction of neurite outgrowth during iMN differentiation and maturation. CONCLUSIONS Our findings show that stepwise combined cell therapy can induce alterations in the microenvironment for effective cell therapy in SCI. The in vitro results suggest that co-culturing hMSC and iMN can synergistically promote induction of MN neurite outgrowth.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | | | - Hyunkyung Mo
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Heeju Han
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- YiPSCELL, Inc, Seoul, South Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| |
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
|
Li J, Jing Y, Bai F, Wu Y, Wang L, Yan Y, Jia Y, Yu Y, Jia B, Ali F. Induced pluripotent stem cells as natural biofactories for exosomes carrying miR-199b-5p in the treatment of spinal cord injury. Front Pharmacol 2023; 13:1078761. [PMID: 36703756 PMCID: PMC9871459 DOI: 10.3389/fphar.2022.1078761] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Induced pluripotent stem cells-derived exosomes (iPSCs-Exo) can effectively treat spinal cord injury (SCI) in mice. But the role of iPSCs-Exo in SCI mice and its molecular mechanisms remain unclear. This research intended to study the effects and molecular mechanism of iPSCs-Exo in SCI mice models. Methods: The feature of iPSCs-Exo was determined by transmission electron microscope (TEM), nanoparticle tracking analysis (NTA), and western blot. The effects of iPSCs-Exo in the SCI mice model were evaluated by Basso Mouse Scale (BMS) scores and H&E staining. The roles of iPSCs-Exo and miR-199b-5p in LPS-treated BMDM were verified by immunofluorescence, RT-qPCR, and Cytokine assays. The target genes of miR-199b-5p were identified, and the function of miR-199b-5p and its target genes on LPS-treated BMDM was explored by recuse experiment. Results: iPSCs-Exo improved motor function in SCI mice model in vivo, shifted the polarization from M1 macrophage to M2 phenotype, and regulated related inflammatory factors expression to accelerate the SCI recovery in LPS-treated BMDM in vitro. Meanwhile, miR-199b-5p was a functional player of iPSCs-Exo, which could target hepatocyte growth factor (Hgf). Moreover, miR-199b-5p overexpression polarized M1 macrophage into M2 phenotype and promoted neural regeneration in SCI. The rescue experiments confirmed that miR-199b-5p induced macrophage polarization and SCI recovery by regulating Hgf and Phosphoinositide 3-kinase (PI3K) signaling pathways. Conclusion: The miR-199b-5p-bearing iPSCs-Exo might become an effective method to treat SCI.
Collapse
Affiliation(s)
- Jun Li
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,School of Rehabilitation Medicine, Capital Medical University, Beijing, China
| | - Yingli Jing
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China,China Rehabilitation Science Institute, Beijing, China
| | - Fan Bai
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China,China Rehabilitation Science Institute, Beijing, China
| | - Ying Wu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Limiao Wang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China,China Rehabilitation Science Institute, Beijing, China
| | - Yitong Yan
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China,China Rehabilitation Science Institute, Beijing, China
| | - Yunxiao Jia
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yan Yu
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China,China Rehabilitation Science Institute, Beijing, China,*Correspondence: Yan Yu, ; Benzhi Jia,
| | - Benzhi Jia
- Department of Spinal cord injury rehabilitation, Shanxi Kangfu Hospital, Xi’an, Shanxi, China,*Correspondence: Yan Yu, ; Benzhi Jia,
| | - Fawad Ali
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, Pakistan
| |
Collapse
|
7
|
Lohrasbi F, Ghasemi-Kasman M, Soghli N, Ghazvini S, Vaziri Z, Abdi S, Darban YM. The Journey of iPSC-derived OPCs in Demyelinating Disorders: From In vitro Generation to In vivo Transplantation. Curr Neuropharmacol 2023; 21:1980-1991. [PMID: 36825702 PMCID: PMC10514531 DOI: 10.2174/1570159x21666230220150010] [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: 08/27/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 02/22/2023] Open
Abstract
Loss of myelination is common among neurological diseases. It causes significant disability, even death, if it is not treated instantly. Different mechanisms involve the pathophysiology of demyelinating diseases, such as genetic background, infectious, and autoimmune inflammation. Recently, regenerative medicine and stem cell therapy have shown to be promising for the treatment of demyelinating disorders. Stem cells, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult stem cells (ASCs), can differentiate into oligodendrocyte progenitor cells (OPCs), which may convert to oligodendrocytes (OLs) and recover myelination. IPSCs provide an endless source for OPCs generation. However, the restricted capacity of proliferation, differentiation, migration, and myelination of iPSC-derived OPCs is a notable gap for future studies. In this article, we have first reviewed stem cell therapy in demyelinating diseases. Secondly, methods of different protocols have been discussed among in vitro and in vivo studies on iPSC-derived OPCs to contrast OPCs' transplantation efficacy. Lastly, we have reviewed the results of iPSCs-derived OLs production in each demyelination model.
Collapse
Affiliation(s)
- Fatemeh Lohrasbi
- Student Research Committee, Babol University of Medical Science, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Science, Babol, Iran
- Department of Physiology, School of Medical Sciences, Babol University of Medical Science, Babol, Iran
| | - Negar Soghli
- Student Research Committee, Babol University of Medical Science, Babol, Iran
| | - Sobhan Ghazvini
- Student Research Committee, Babol University of Medical Science, Babol, Iran
| | - Zahra Vaziri
- Student Research Committee, Babol University of Medical Science, Babol, Iran
| | - Sadaf Abdi
- Student Research Committee, Babol University of Medical Science, Babol, Iran
| | | |
Collapse
|
8
|
Huang L, Sun X, Wang L, Pei G, Wang Y, Zhang Q, Liang Z, Wang D, Fu C, He C, Wei Q. Enhanced effect of combining bone marrow mesenchymal stem cells (BMMSCs) and pulsed electromagnetic fields (PEMF) to promote recovery after spinal cord injury in mice. MedComm (Beijing) 2022; 3:e160. [PMID: 35949547 PMCID: PMC9350428 DOI: 10.1002/mco2.160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 12/03/2022] Open
Abstract
Spinal cord injury (SCI) is a destructive traumatic disease of the central nervous system without satisfying therapy efficiency. Bone marrow mesenchymal stem cells (BMMSCs) therapy promotes the neurotrophic factors' secretion and axonal regeneration, thereby promoting recovery of SCI. Pulsed electromagnetic fields (PEMF) therapy has been proven to promote neural growth and regeneration. Both BMMSCs and PEMF have shown curative effects for SCI; PEMF can further promote stem cell differentiation. Thus, we explored the combined effects of BMMSCs and PEMF and the potential interaction between these two therapies in SCI. Compared with the SCI control, BMMSCs, and PEMF groups, the combinational therapy displayed the best therapeutic effect. Combinational therapy increased the expression levels of nutritional factors including brain-derived neurotrophic factor (BDNF), nerve growth factors (NGF) and vascular endothelial growth factor (VEGF), enhanced neuron preservation (NeuN and NF-200), and increased axonal growth (MBP and myelin sheath). Additionally, PEMF promoted the expression levels of BDNF and VEGF in BMMSCs via Wnt/β-catenin signaling pathway. In summary, the combined therapy of BMMSCs and PEMF displayed a more satisfactory effect than BMMSCs and PEMF therapy alone, indicating a promising application of combined therapy for the therapy of SCI.
Collapse
Affiliation(s)
- Liyi Huang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Xin Sun
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Gaiqing Pei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Yang Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Qing Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Zejun Liang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Dong Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
- Aging and Geriatric Mechanism Laboratory, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| |
Collapse
|
9
|
The Inflammatory Response after Moderate Contusion Spinal Cord Injury: A Time Study. BIOLOGY 2022; 11:biology11060939. [PMID: 35741460 PMCID: PMC9220050 DOI: 10.3390/biology11060939] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary The neuroinflammatory response is a rather complex event in spinal cord injury (SCI) and has the capacity to exacerbate cell damage but also to contribute to the repair of the injury. This complexity is thought to depend on a variety of inflammatory mediators, of which tumor necrosis factor (TNF) plays a key role. Evidence indicates that TNF can be both protective and detrimental in SCI. In the present study, we studied the temporal and cellular expression of TNF and its receptors after SCI in mice. We found TNF to be significantly increased in both the acute and the delayed phases after SCI, alongside a robust neuroinflammatory response. As we could verify some of our results in human postmortem tissue, our results imply that diminishing the detrimental immune signaling after SCI could also enhance recovery in humans. Abstract Spinal cord injury (SCI) initiates detrimental cellular and molecular events that lead to acute and delayed neuroinflammation. Understanding the role of the inflammatory response in SCI requires insight into the temporal and cellular synthesis of inflammatory mediators. We subjected C57BL/6J mice to SCI and investigated inflammatory reactions. We examined activation, recruitment, and polarization of microglia and infiltrating immune cells, focusing specifically on tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. In the acute phase, TNF expression increased in glial cells and neuron-like cells, followed by infiltrating immune cells. TNFR1 and TNFR2 levels increased in the delayed phase and were found preferentially on neurons and glial cells, respectively. The acute phase was dominated by the infiltration of granulocytes and macrophages. Microglial/macrophage expression of Arg1 increased from 1–7 days after SCI, followed by an increase in Itgam, Cx3cr1, and P2ry12, which remained elevated throughout the study. By 21 and 28 days after SCI, the lesion core was populated by galectin-3+, CD68+, and CD11b+ microglia/macrophages, surrounded by a glial scar consisting of GFAP+ astrocytes. Findings were verified in postmortem tissue from individuals with SCI. Our findings support the consensus that future neuroprotective immunotherapies should aim to selectively neutralize detrimental immune signaling while sustaining pro-regenerative processes.
Collapse
|
10
|
Assunção Silva RC, Pinto L, Salgado AJ. Cell transplantation and secretome based approaches in spinal cord injury regenerative medicine. Med Res Rev 2021; 42:850-896. [PMID: 34783046 DOI: 10.1002/med.21865] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/12/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
The axonal growth-restrictive character of traumatic spinal cord injury (SCI) makes finding a therapeutic strategy a very demanding task, due to the postinjury events impeditive to spontaneous axonal outgrowth and regeneration. Considering SCI pathophysiology complexity, it has been suggested that an effective therapy should tackle all the SCI-related aspects and provide sensory and motor improvement to SCI patients. Thus, the current aim of any therapeutic approach for SCI relies in providing neuroprotection and support neuroregeneration. Acknowledging the current SCI treatment paradigm, cell transplantation is one of the most explored approaches for SCI with mesenchymal stem cells (MSCs) being in the forefront of many of these. Studies showing the beneficial effects of MSC transplantation after SCI have been proposing a paracrine action of these cells on the injured tissues, through the secretion of protective and trophic factors, rather than attributing it to the action of cells itself. This manuscript provides detailed information on the most recent data regarding the neuroregenerative effect of the secretome of MSCs as a cell-free based therapy for SCI. The main challenge of any strategy proposed for SCI treatment relies in obtaining robust preclinical evidence from in vitro and in vivo models, before moving to the clinics, so we have specifically focused on the available vertebrate and mammal models of SCI currently used in research and how can SCI field benefit from them.
Collapse
Affiliation(s)
- Rita C Assunção Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
11
|
Suzuki H, Sakai T. Current Concepts of Stem Cell Therapy for Chronic Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22147435. [PMID: 34299053 PMCID: PMC8308009 DOI: 10.3390/ijms22147435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic 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 clinical trials. In addition, several more are coming down the translational pipeline. Among ongoing and completed trials are those reporting the use of mesenchymal stem cells, neural stem/progenitor cells, induced pluripotent stem cells, olfactory ensheathing cells, and Schwann cells. The advancements in stem cell technology, combined with the powerful neuroimaging modalities, can now accelerate the pathway of promising novel therapeutic strategies from bench to bedside. Various combinations of different molecular therapies have been combined with supportive scaffolds to facilitate favorable cell–material interactions. In this review, we summarized some of the most recent insights into the preclinical and clinical studies using stem cells and other supportive drugs to unlock the microenvironment in chronic SCI to treat patients with this condition. Successful future therapies will require these stem cells and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, loss of structural framework, and immunorejection.
Collapse
|
12
|
Abstract
Traumatic spinal cord injury (SCI) results in direct and indirect damage to neural tissues, which results in motor and sensory dysfunction, dystonia, and pathological reflex that ultimately lead to paraplegia or tetraplegia. A loss of cells, axon regeneration failure, and time-sensitive pathophysiology make tissue repair difficult. Despite various medical developments, there are currently no effective regenerative treatments. Stem cell therapy is a promising treatment for SCI due to its multiple targets and reactivity benefits. The present review focuses on SCI stem cell therapy, including bone marrow mesenchymal stem cells, umbilical mesenchymal stem cells, adipose-derived mesenchymal stem cells, neural stem cells, neural progenitor cells, embryonic stem cells, induced pluripotent stem cells, and extracellular vesicles. Each cell type targets certain features of SCI pathology and shows therapeutic effects via cell replacement, nutritional support, scaffolds, and immunomodulation mechanisms. However, many preclinical studies and a growing number of clinical trials found that single-cell treatments had only limited benefits for SCI. SCI damage is multifaceted, and there is a growing consensus that a combined treatment is needed.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, 34753West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
13
|
Stem Cells in Clinical Research and Therapy. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
14
|
Farzaneh M, Anbiyaiee A, Khoshnam SE. Human Pluripotent Stem Cells for Spinal Cord Injury. Curr Stem Cell Res Ther 2020; 15:135-143. [PMID: 31656156 DOI: 10.2174/1574362414666191018121658] [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: 06/08/2019] [Revised: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) as a serious public health issue and neurological insult is one of the most severe cause of long-term disability. To date, a variety of techniques have been widely developed to treat central nervous system injury. Currently, clinical treatments are limited to surgical decompression and pharmacotherapy. Because of their negative effects and inefficiency, novel therapeutic approaches are required in the management of SCI. Improvement and innovation of stem cell-based therapies have a huge potential for biological and future clinical applications. Human pluripotent stem cells (hPSCs) including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are defined by their abilities to divide asymmetrically, self-renew and ultimately differentiate into various cell lineages. There are considerable research efforts to use various types of stem cells, such as ESCs, neural stem cells (NSCs), and mesenchymal stem cells (MSCs) in the treatment of patients with SCI. Moreover, the use of patient-specific iPSCs holds great potential as an unlimited cell source for generating in vivo models of SCI. In this review, we focused on the potential of hPSCs in treating SCI.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Anbiyaiee
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Department of Physiology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
15
|
Zavvarian MM, Toossi A, Khazaei M, Hong J, Fehlings M. Novel innovations in cell and gene therapies for spinal cord injury. F1000Res 2020; 9. [PMID: 32399196 PMCID: PMC7194487 DOI: 10.12688/f1000research.21989.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) leads to chronic and multifaceted disability, which severely impacts the physical and mental health as well as the socio-economic status of affected individuals. Permanent disabilities following SCI result from the failure of injured neurons to regenerate and rebuild functional connections with their original targets. Inhibitory factors present in the SCI microenvironment and the poor intrinsic regenerative capacity of adult spinal cord neurons are obstacles for regeneration and functional recovery. Considerable progress has been made in recent years in developing cell and molecular approaches to enable the regeneration of damaged spinal cord tissue. In this review, we highlight several potent cell-based approaches and genetic manipulation strategies (gene therapy) that are being investigated to reconstruct damaged or lost spinal neural circuits and explore emerging novel combinatorial approaches for enhancing recovery from SCI.
Collapse
Affiliation(s)
- Mohammad-Masoud Zavvarian
- Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Amirali Toossi
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Mohamad Khazaei
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - James Hong
- Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Michael Fehlings
- Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
16
|
Csobonyeiova M, Polak S, Zamborsky R, Danisovic L. Recent Progress in the Regeneration of Spinal Cord Injuries by Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 20:3838. [PMID: 31390782 PMCID: PMC6695701 DOI: 10.3390/ijms20153838] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/27/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022] Open
Abstract
Regeneration of injuries occurring in the central nervous system, particularly spinal cord injuries (SCIs), is extremely difficult. The complex pathological events following a SCI often restrict regeneration of nervous tissue at the injury site and frequently lead to irreversible loss of motor and sensory function. Neural stem/progenitor cells (NSCs/NPCs) possess neuroregenerative and neuroprotective features, and transplantation of such cells into the site of damaged tissue is a promising stem cell-based therapy for SCI. However, NSC/NPCs have mostly been induced from embryonic stem cells or fetal tissue, leading to ethical concerns. The pioneering work of Yamanaka and colleagues gave rise to the technology to induce pluripotent stem cells (iPSCs) from somatic cells, overcoming these ethical issues. The advent of iPSCs technology has meant significant progress in the therapy of neurodegenerative disease and nerve tissue damage. A number of published studies have described the successful differentiation of NSCs/NPCs from iPSCs and their subsequent engraftment into SCI animal models, followed by functional recovery of injury. The aim of this present review is to summarize various iPSC- NPCs differentiation methods, SCI modelling, and the current status of possible iPSC- NPCs- based therapy of SCI.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Polak
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Radoslav Zamborsky
- Department of Orthopaedics, Faculty of Medicine, Comenius University and National Institute of Children's Diseases, Limbova 1, 833 40 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia.
- Regenmed Ltd., Medena 29, 811 01 Bratislava, Slovakia.
| |
Collapse
|
17
|
Beldick SR, Hong J, Altamentova S, Khazaei M, Hundal A, Zavvarian MM, Rumajogee P, Chio J, Fehlings MG. Severe-combined immunodeficient rats can be used to generate a model of perinatal hypoxic-ischemic brain injury to facilitate studies of engrafted human neural stem cells. PLoS One 2018; 13:e0208105. [PMID: 30485360 PMCID: PMC6261629 DOI: 10.1371/journal.pone.0208105] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/12/2018] [Indexed: 01/12/2023] Open
Abstract
Cerebral palsy (CP) encompasses a group of non-progressive brain disorders that are often acquired through perinatal hypoxic-ischemic (HI) brain injury. Injury leads to a cascade of cell death events, resulting in lifetime motor and cognitive deficits. There are currently no treatments that can repair the resulting brain damage and improve functional outcomes. To date, preclinical research using neural precursor cell (NPC) transplantation as a therapy for HI brain injury has shown promise. To translate this treatment to the clinic, it is essential that human-derived NPCs also be tested in animal models, however, a major limitation is the high risk of xenograft rejection. A solution is to transplant the cells into immune-deficient rodents, but there are currently no models of HI brain injury established in such a cohort of animals. Here, we demonstrate that a model of HI brain injury can be generated in immune-deficient Prkdc knockout (KO) rats. Long-term deficits in sensorimotor function were similar between KO and wildtype (WT) rats. Interestingly, some aspects of the injury were more severe in KO rats. Additionally, human induced pluripotent stem cell derived (hiPSC)-NPCs had higher survival at 10 weeks post-transplant in KO rats when compared to their WT counterparts. This work establishes a reliable model of neonatal HI brain injury in Prkdc KO rats that will allow for future transplantation, survival, and long-term evaluation of the safety and efficacy of hiPSC-NPCs for neonatal brain damage. This model will enable critical preclinical translational research using human NPCs.
Collapse
Affiliation(s)
- Stephanie R. Beldick
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - James Hong
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Svetlana Altamentova
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Anisha Hundal
- Life Sciences Program, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Mohammad-Masoud Zavvarian
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Prakasham Rumajogee
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Jonathon Chio
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Michael G. Fehlings
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Division of Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
- Division of Neurosurgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Abstract
INTRODUCTION Spinal cord injury (SCI) is a devastating condition, where regenerative failure and cell loss lead to paralysis. The heterogeneous and time-sensitive pathophysiology has made it difficult to target tissue repair. Despite many medical advances, there are no effective regenerative therapies. As stem cells offer multi-targeted and environmentally responsive benefits, cell therapy is a promising treatment approach. Areas covered: This review highlights the cell therapies being investigated for SCI, including Schwann cells, olfactory ensheathing cells, mensenchymal stem/stromal cells, neural precursors, oligodendrocyte progenitors, embryonic stem cells, and induced pluripotent stem cells. Through mechanisms of cell replacement, scaffolding, trophic support and immune modulation, each approach targets unique features of SCI pathology. However, as the injury is multifaceted, it is increasingly recognized that a combinatorial approach will be necessary to treat SCI. Expert opinion: Most preclinical studies, and an increasing number of clinical trials, are finding that single cell therapies have only modest benefits after SCI. These considerations, alongside issues of therapy cost-effectiveness, need to be addressed at the bench. In addition to exploring combinatorial strategies, researchers should consider cell reproducibility and storage parameters when designing animal experiments. Equally important, clinical trials must follow strict regulatory guidelines that will enable transparency of results.
Collapse
Affiliation(s)
- Anna Badner
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada.,b Institute of Medical Sciences , University of Toronto , Toronto , ON , Canada
| | - Ahad M Siddiqui
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada
| | - Michael G Fehlings
- a Department of Genetics and Development , Krembil Research Institute, University Health Network , Toronto , ON , Canada.,b Institute of Medical Sciences , University of Toronto , Toronto , ON , Canada.,c Canada Spinal Program , University Health Network, Toronto Western Hospital , Toronto , ON , Canada
| |
Collapse
|
19
|
Iyer NR, Wilems TS, Sakiyama-Elbert SE. Stem cells for spinal cord injury: Strategies to inform differentiation and transplantation. Biotechnol Bioeng 2017; 114:245-259. [PMID: 27531038 PMCID: PMC5642909 DOI: 10.1002/bit.26074] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 12/13/2022]
Abstract
The complex pathology of spinal cord injury (SCI), involving a cascade of secondary events and the formation of inhibitory barriers, hampers regeneration across the lesion site and often results in irreversible loss of motor function. The limited regenerative capacity of endogenous cells after SCI has led to a focus on the development of cell therapies that can confer both neuroprotective and neuroregenerative benefits. Stem cells have emerged as a candidate cell source because of their ability to self-renew and differentiate into a multitude of specialized cell types. While ethical and safety concerns impeded the use of stem cells in the past, advances in isolation and differentiation methods have largely mitigated these issues. A confluence of work in stem cell biology, genetics, and developmental neurobiology has informed the directed differentiation of specific spinal cell types. After transplantation, these stem cell-derived populations can replace lost cells, provide trophic support, remyelinate surviving axons, and form relay circuits that contribute to functional recovery. Further refinement of stem cell differentiation and transplantation methods, including combinatorial strategies that involve biomaterial scaffolds and drug delivery, is critical as stem cell-based treatments enter clinical trials. Biotechnol. Bioeng. 2017;114: 245-259. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nisha R Iyer
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Thomas S Wilems
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| |
Collapse
|
20
|
Lane MA, Lepore AC, Fischer I. Improving the therapeutic efficacy of neural progenitor cell transplantation following spinal cord injury. Expert Rev Neurother 2016; 17:433-440. [PMID: 27927055 DOI: 10.1080/14737175.2017.1270206] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION There have been a wide range of preclinical studies testing cellular therapies to repair the injured spinal cord, yet they remain a challenge to translate because of inconsistencies in efficacy, limited number of patients with acute/subacute SCI and the high costs of clinical trials. Area covered: This paper focusses on the therapeutic potential of neural precursor cells (NPCs) because they can provide the cellular components capable of promoting repair and enhancing functional improvement following spinal cord injury (SCI). The authors discuss the challenges of NPC transplantation with respect to different populations of NPCs of glial and neuronal lineages, the timing of treatment relative to acute and chronic injury, and the progress in ongoing clinical trials. Expert commentary: Preclinical research will continue to elucidate mechanisms of recovery associated with NPC transplants, including increasing the partnership with related fields such as spinal atrophies and multiple sclerosis. The clinical trials landscape will grow and include both acute and chronic SCI with increased partnership and strengthened communication between biotechnology, government and academia. There will also be growing effort to develop better biomarkers, imaging and outcome measures for detailed assessment of neurological function and measures of quality of life.
Collapse
Affiliation(s)
- Michael A Lane
- a Department of Neurobiology & Anatomy, Spinal Cord Research Center , Drexel University , Philadelphia , PA , USA
| | - Angelo C Lepore
- b Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience , Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia , PA , USA
| | - Itzhak Fischer
- a Department of Neurobiology & Anatomy, Spinal Cord Research Center , Drexel University , Philadelphia , PA , USA
| |
Collapse
|
21
|
Stem cell regenerative potential for plastic and reconstructive surgery. Cell Tissue Bank 2016; 17:735-744. [PMID: 27604466 DOI: 10.1007/s10561-016-9583-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022]
Abstract
Stem cells represent heterogeneous population of undifferentiated cells with unique characteristics of long term self renewal and plasticity. Moreover, they are capable of active migration to diseased tissues, secretion of different bioactive molecules, and they have immunosuppressive potential as well. They occur in all tissues through life and are involved in process of embryogenesis and regeneration. During last decades stem cells attracted significant attention in each field of medicine, including plastic and reconstructive surgery. The main goal of the present review article is to present and discuss the potential of stem cells and to provide information about their safe utilization in chronic wounds and fistulae healing, scar management, breast reconstruction, as well as in bone, tendon and peripheral nerve regeneration.
Collapse
|