1
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [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: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
2
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
3
|
Shang WY, Ren YF, Li B, Huang XM, Zhang ZL, Huang J. Efficacy of growth factor gene-modified stem cells for motor function after spinal cord injury in rodents: a systematic review and meta‑analysis. Neurosurg Rev 2024; 47:87. [PMID: 38369598 DOI: 10.1007/s10143-024-02314-2] [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: 09/14/2023] [Revised: 01/15/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
The efficacy of growth factor gene-modified stem cells in treating spinal cord injury (SCI) remains unclear. This study aims to evaluate the effectiveness of growth factor gene-modified stem cells in restoring motor function after SCI. Two reviewers searched four databases, including PubMed, Embase, Web of Science, and Scopus, to identify relevant records. Studies on rodents assessing the efficacy of transplanting growth factor gene-modified stem cells in restoring motor function after SCI were included. The results were reported using the standardized mean difference (SMD) with a 95% confidence interval (95% CI). Analyses showed that growth factor gene-modified stem cell transplantation improved motor function recovery in rodents with SCI compared to the untreated (SMD = 3.98, 95% CI 3.26-4.70, I2 = 86.8%, P < 0.0001) and stem cell (SMD = 2.53, 95% CI 1.93-3.13, I2 = 86.9%, P < 0.0001) groups. Using growth factor gene-modified neural stem/histone cells enhanced treatment efficacy. In addition, the effectiveness increased when viral vectors were employed for gene modification and high transplantation doses were administered during the subacute phase. Stem cells derived from the human umbilical cord exhibited an advantage in motor function recovery. However, the transplantation of growth factor gene-modified stem cells did not significantly improve motor function in male rodents (P = 0.136). Transplantation of growth factor gene-modified stem cells improved motor function in rodents after SCI, but claims of enhanced efficacy should be approached with caution. The safety of gene modification remains a significant concern, requiring additional efforts to enhance its clinical translatability.
Collapse
Affiliation(s)
- Wen-Ya Shang
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Ya-Feng Ren
- The First Affiliated Hospital of Henan University of CM, Zhengzhou, China.
| | - Bing Li
- The First Affiliated Hospital of Henan University of CM, Zhengzhou, China
| | | | - Zhi-Lan Zhang
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Jing Huang
- Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
4
|
Kijima K, Ono G, Kobayakawa K, Saiwai H, Hara M, Yoshizaki S, Yokota K, Saito T, Tamaru T, Iura H, Haruta Y, Kitade K, Utsunomiya T, Konno D, Edgerton VR, Liu CY, Sakai H, Maeda T, Kawaguchi K, Matsumoto Y, Okada S, Nakashima Y. Zinc deficiency impairs axonal regeneration and functional recovery after spinal cord injury by modulating macrophage polarization via NF-κB pathway. Front Immunol 2023; 14:1290100. [PMID: 38022538 PMCID: PMC10666775 DOI: 10.3389/fimmu.2023.1290100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating disease that results in permanent paralysis. Currently, there is no effective treatment for SCI, and it is important to identify factors that can provide therapeutic intervention during the course of the disease. Zinc, an essential trace element, has attracted attention as a regulator of inflammatory responses. In this study, we investigated the effect of zinc status on the SCI pathology and whether or not zinc could be a potential therapeutic target. Methods We created experimental mouse models with three different serum zinc concentration by changing the zinc content of the diet. After inducing contusion injury to the spinal cord of three mouse models, we assessed inflammation, apoptosis, demyelination, axonal regeneration, and the number of nuclear translocations of NF-κB in macrophages by using qPCR and immunostaining. In addition, macrophages in the injured spinal cord of these mouse models were isolated by flow cytometry, and their intracellular zinc concentration level and gene expression were examined. Functional recovery was assessed using the open field motor score, a foot print analysis, and a grid walk test. Statistical analysis was performed using Wilcoxon rank-sum test and ANOVA with the Tukey-Kramer test. Results In macrophages after SCI, zinc deficiency promoted nuclear translocation of NF-κB, polarization to pro-inflammatory like phenotype and expression of pro-inflammatory cytokines. The inflammatory response exacerbated by zinc deficiency led to worsening motor function by inducing more apoptosis of oligodendrocytes and demyelination and inhibiting axonal regeneration in the lesion site compared to the normal zinc condition. Furthermore, zinc supplementation after SCI attenuated these zinc-deficiency-induced series of responses and improved motor function. Conclusion We demonstrated that zinc affected axonal regeneration and motor functional recovery after SCI by negatively regulating NF-κB activity and the subsequent inflammatory response in macrophages. Our findings suggest that zinc supplementation after SCI may be a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Utsunomiya
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - V. Reggie Edgerton
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
- Institut Guttmann. Hospital de Neurorehabilitació, Institut Universitari adscrit a la Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Charles Y. Liu
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
| | - Hiroaki Sakai
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Fukushima Medical University, Fukushima, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Ono G, Kobayakawa K, Saiwai H, Tamaru T, Iura H, Haruta Y, Kitade K, Iida K, Kawaguchi K, Matsumoto Y, Tsuda M, Tamura T, Ozato K, Inoue K, Konno DJ, Maeda T, Okada S, Nakashima Y. Macrophages play a leading role in determining the direction of astrocytic migration in spinal cord injury via ADP-P2Y1R axis. Sci Rep 2023; 13:11177. [PMID: 37429920 DOI: 10.1038/s41598-023-38301-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
After spinal cord injury (SCI), inflammatory cells such as macrophages infiltrate the injured area, and astrocytes migrate, forming a glial scar around macrophages. The glial scar inhibits axonal regeneration, resulting in significant permanent disability. However, the mechanism through which glial scar-forming astrocytes migrate to the injury site has not been clarified. Here we show that migrating macrophages attract reactive astrocytes toward the center of the lesion after SCI. Chimeric mice with bone marrow lacking IRF8, which controls macrophage centripetal migration after SCI, showed widely scattered macrophages in the injured spinal cord with the formation of a huge glial scar around the macrophages. To determine whether astrocytes or macrophages play a leading role in determining the directions of migration, we generated chimeric mice with reactive astrocyte-specific Socs3-/- mice, which showed enhanced astrocyte migration, and bone marrow from IRF8-/- mice. In this mouse model, macrophages were widely scattered, and a huge glial scar was formed around the macrophages as in wild-type mice that were transplanted with IRF8-/- bone marrow. In addition, we revealed that macrophage-secreted ATP-derived ADP attracts astrocytes via the P2Y1 receptor. Our findings revealed a mechanism through which migrating macrophages attract astrocytes and affect the pathophysiology and outcome after SCI.
Collapse
Affiliation(s)
- Gentaro Ono
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Hirokazu Saiwai
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Tamaru
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hirotaka Iura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yohei Haruta
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuki Kitade
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keiichiro Iida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Kawaguchi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0395, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, Section on Molecular Genetics of Immunity, Division of Developmental Biology, NICHD, National Institutes of Health, Building 6A, Room 2A01, 6 Center Drive, Bethesda, MD, 20892, USA
| | - Kazuhide Inoue
- Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0395, Japan
- Greenpharma Research Center for System Drug Discovery, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Dai-Jiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, 577-8502, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, 550-4 Igisu, Iizuka, Fukuoka, 820-8508, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
6
|
Azevedo-Pereira RL, Manley NC, Dong C, Zhang Y, Lee AG, Zatulovskaia Y, Gupta V, Vu J, Han S, Berry JE, Bliss TM, Steinberg GK. Decoding the molecular crosstalk between grafted stem cells and the stroke-injured brain. Cell Rep 2023; 42:112353. [PMID: 37043353 PMCID: PMC10562513 DOI: 10.1016/j.celrep.2023.112353] [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: 08/12/2021] [Revised: 01/25/2023] [Accepted: 03/21/2023] [Indexed: 04/13/2023] Open
Abstract
Stem cell therapy shows promise for multiple disorders; however, the molecular crosstalk between grafted cells and host tissue is largely unknown. Here, we take a step toward addressing this question. Using translating ribosome affinity purification (TRAP) with sequencing tools, we simultaneously decode the transcriptomes of graft and host for human neural stem cells (hNSCs) transplanted into the stroke-injured rat brain. Employing pathway analysis tools, we investigate the interactions between the two transcriptomes to predict molecular pathways linking host and graft genes; as proof of concept, we predict host-secreted factors that signal to the graft and the downstream molecular cascades they trigger in the graft. We identify a potential host-graft crosstalk pathway where BMP6 from the stroke-injured brain induces graft secretion of noggin, a known brain repair factor. Decoding the molecular interplay between graft and host is a critical step toward deciphering the molecular mechanisms of stem cell action.
Collapse
Affiliation(s)
| | - Nathan C Manley
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Chen Dong
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yue Zhang
- Stanford Genetics Bioinformatics Service Center, Stanford University, Stanford, CA 94305, USA
| | - Alex G Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, CA 94143, USA
| | - Yulia Zatulovskaia
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Varun Gupta
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jennifer Vu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Summer Han
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jack E Berry
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Tonya M Bliss
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Xu D, Tang L, Kapranov P. Complexities of mammalian transcriptome revealed by targeted RNA enrichment techniques. Trends Genet 2023; 39:320-333. [PMID: 36681580 DOI: 10.1016/j.tig.2022.12.004] [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: 08/29/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/21/2023]
Abstract
Studies using highly sensitive targeted RNA enrichment methods have shown that a large portion of the human transcriptome remains to be discovered and that most of the genome is transcribed in a complex, interleaved fashion characterized by a complex web of transcripts emanating from protein coding and noncoding loci. These results resonate with those from single-cell transcriptome profiling endeavors that reveal the existence of multiple novel, cell type-specific transcripts and clearly demonstrate that our understanding of the complexities of the human transcriptome is far from being complete. Here, we review the current status of the targeted RNA enrichment techniques, their application to the discovery of novel cell type-specific transcripts, and their impact on our understanding of the human genome and transcriptome.
Collapse
Affiliation(s)
- Dongyang Xu
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Lu Tang
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Philipp Kapranov
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China.
| |
Collapse
|
8
|
Kawai M, Nagoshi N, Okano H, Nakamura M. A review of regenerative therapy for spinal cord injury using human iPS cells. NORTH AMERICAN SPINE SOCIETY JOURNAL 2023; 13:100184. [PMID: 36479183 PMCID: PMC9720571 DOI: 10.1016/j.xnsj.2022.100184] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Spinal cord injury (SCI) has been considered to cause sudden, irreversible loss of function in patients. However, developments in stem cell biology and regenerative medicine are changing this conventional notion. Here we reviewed the overview of regenerative medicine of SCI. As a consequence of the establishment of human induced pluripotent stem cells (hiPSCs), hiPSC-based therapies for SCI, such as neural stem/progenitor cell (NS/PC) transplantation, have emerged as promising therapeutic modalities. Using several animal models, hiPSC-NS/PC transplantation into subacute injured spinal cords has been repeatedly demonstrated to improve locomotor function. Some biological mechanisms underlying this improvement have been proposed. In particular, combined with advanced neuroscience techniques such as designer receptors exclusively activated by designer drugs (DREADDs), neuronal relay theory, in which the transplanted cell-derived neurons reconstruct disrupted neuronal circuits, was proven to be involved histologically, pharmaceutically, electrophysiologically, and via in vivo bioimaging. Based on these findings, hiPSC-NS/PC transplantation for subacute SCI was moved ahead to a clinical study on human patients. At the same time, the search for effective treatments for chronic SCI is proceeding gradually, combining hiPSC-NS/PC transplantation with other treatment modalities such as rehabilitation, pharmaceutical interventions, or optimal scaffolds. In addition to NS/PCs, oligodendrocyte precursor cells (OPCs) are also a promising cell source for transplantation, as demyelinated axons affected by SCI can be repaired by OPCs. Therapies with OPCs derived from hiPSCs are still in preclinical studies but have shown favorable outcomes in animal models. In the future, several therapeutic options may be available according to the pathological conditions and the time period of SCI. Moreover, the application of regenerative therapy for the spinal cord could be broadened to degenerative disorders, such as spinal canal stenosis. Summary sentence: A historical review of human induced pluripotent stem cell (hiPSC) based cell transplantation therapy for spinal cord injury (SCI), in particular about footsteps of hiPSC-derived neural stem/progenitor cell transplantation, recent clinical study, and its future perspective.
Collapse
Affiliation(s)
- Momotaro Kawai
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
9
|
Kim DH, Cho HJ, Park CY, Cho MS, Kim DW. Transplantation of PSA-NCAM-Positive Neural Precursors from Human Embryonic Stem Cells Promotes Functional Recovery in an Animal Model of Spinal Cord Injury. Tissue Eng Regen Med 2022; 19:1349-1358. [PMID: 36036887 PMCID: PMC9679075 DOI: 10.1007/s13770-022-00483-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: 06/14/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) results in permanent impairment of motor and sensory functions at and below the lesion site. There is no therapeutic option to the functional recovery of SCI involving diverse injury responses of different cell types in the lesion that limit endogenous nerve regeneration. In this regard, cell replacement therapy utilizing stem cells or their derivatives has become a highly promising approach to promote locomotor recovery. For this reason, the demand for a safe and efficient multipotent cell source that can differentiate into various neural cells is increasing. In this study, we evaluated the efficacy and safety of human polysialylated-neural cell adhesion molecule (PSA-NCAM)-positive neural precursor cells (hNPCsPSA-NCAM+) as a treatment for SCI. METHODS One hundred thousand hNPCsPSA-NCAM+ isolated from human embryonic stem cell-derived NPCs were transplanted into the lesion site by microinjection 7 days after contusive SCI at the thoracic level. We examined the histological characteristics of the graft and behavioral improvement in the SCI rats 10 weeks after transplantation. RESULTS Locomotor activity improvement was estimated by the Basso-Beattie-Bresnahan locomotor rating scale. Behavioral tests revealed that the transplantation of the hNPCsPSA-NCAM+ into the injured spinal cords of rats significantly improved locomotor function. Histological examination showed that hNPCsPSA-NCAM+ had differentiated into neural cells and successfully integrated into the host tissue with no evidence of tumor formation. We investigated cytokine expressions, which led to the early therapeutic effect of hNPCsPSA-NCAM+, and found that some undifferentiated NPCs still expressed midkine, a well-known neurotrophic factor involved in neural development and inflammatory responses, 10 weeks after transplantation. CONCLUSION Our results demonstrate that hNPCsPSA-NCAM+ serve as a safe and efficient cell source which has the potential to improve impaired motor function following SCI.
Collapse
Affiliation(s)
- Do-Hun Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- S.Biomedics Co., Ltd, 2nd Floor, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, South Korea
| | - Hyun-Ju Cho
- S.Biomedics Co., Ltd, 2nd Floor, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, South Korea
| | - Chul-Yong Park
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- S.Biomedics Co., Ltd, 2nd Floor, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, South Korea
| | - Myung Soo Cho
- S.Biomedics Co., Ltd, 2nd Floor, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, South Korea.
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 PLUS Program for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- S.Biomedics Co., Ltd, 2nd Floor, 28 Seongsui-ro 26-gil, Seongdong-gu, Seoul, 04797, South Korea.
| |
Collapse
|
10
|
Shang Z, Wang M, Zhang B, Wang X, Wanyan P. Subacute traumatic spinal cord injury: a systematic review and network meta-analysis of therapeutic strategies based on bone marrow mesenchymal stromal cells in animal models. Cytotherapy 2022; 24:1181-1189. [PMID: 36117057 DOI: 10.1016/j.jcyt.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/21/2022] [Accepted: 08/11/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS To explore the optimal transplantation strategy of bone marrow mesenchymal stem cells in subacute traumatic spinal cord injury in animal experiments in order to provide reference for future animal studies and clinical research. METHODS The PubMed, Embase and Web of Science databases were systematically searched (inception to January 4, 2022). Literature search, data extraction and bias assessment were performed by two independent reviewers. RESULTS A total of 50 articles were included for analysis. Results of both traditional meta-analysis and network meta-analysis showed that high-dose (≥1 × 106) transplantation was significantly better than low-dose (<1 × 106) transplantation and intralesional transplantation was significantly better than intravenous transplantation. CONCLUSIONS Given the limited quality of evidence from current animal studies, more high-quality head-to-head comparisons are needed in the future to delve into the optimal transplantation strategy for stem cells.
Collapse
Affiliation(s)
- Zhizhong Shang
- First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xin Wang
- First Clinical Medical College of Lanzhou University, Lanzhou, China; Chengren Institute of Traditional Chinese Medicine, Lanzhou, China; Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, China; The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Giraldo E, Bonilla P, Mellado M, Garcia-Manau P, Rodo C, Alastrue A, Lopez E, Moratonas EC, Pellise F, Đorđević S, Vicent MJ, Moreno Manzano V. Transplantation of Human-Fetal-Spinal-Cord-Derived NPCs Primed with a Polyglutamate-Conjugated Rho/Rock Inhibitor in Acute Spinal Cord Injury. Cells 2022; 11:cells11203304. [PMID: 36291170 PMCID: PMC9600863 DOI: 10.3390/cells11203304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 12/31/2022] Open
Abstract
Neural precursor cell (NPC) transplantation represents a promising therapy for treating spinal cord injuries (SCIs); however, despite successful results obtained in preclinical models, the clinical translation of this approach remains challenging due, in part, to the lack of consensus on an optimal cell source for human neuronal cells. Depending on the cell source, additional limitations to NPC-based therapies include high tumorigenic potential, alongside poor graft survival and engraftment into host spinal tissue. We previously demonstrated that NPCs derived from rat fetal spinal cords primed with a polyglutamate (PGA)-conjugated form of the Rho/Rock inhibitor fasudil (PGA-SS-FAS) displayed enhanced neuronal differentiation and graft survival when compared to non-primed NPCs. We now conducted a similar study of human-fetal-spinal-cord-derived NPCs (hfNPCs) from legal gestational interruptions at the late gestational stage, at 19-21.6 weeks. In vitro, expanded hfNPCs retained neural features, multipotency, and self-renewal, which supported the development of a cell banking strategy. Before transplantation, we established a simple procedure to prime hfNPCs by overnight incubation with PGA-SS-FAS (at 50 μM FAS equiv.), which improved neuronal differentiation and overcame neurite-like retraction after lysophosphatidic-acid-induced Rho/Rock activation. The transplantation of primed hfNPCs into immune-deficient mice (NU(NCr)-Foxn1nu) immediately after the eighth thoracic segment compression prompted enhanced migration of grafted cells from the dorsal to the ventral spinal cord, increased preservation of GABAergic inhibitory Lbx1-expressing and glutamatergic excitatory Tlx3-expressing somatosensory interneurons, and elevated the numbers of preserved, c-Fos-expressing, activated neurons surrounding the injury epicenter, all in a low percentage. Overall, the priming procedure using PGA-SS-FAS could represent an alternative methodology to improve the capabilities of the hfNPC lines for a translational approach for acute SCI treatment.
Collapse
Affiliation(s)
- Esther Giraldo
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
- Department of Biotechnology. Universitat Politècnica de València, E-46022 Valencia, Spain
- UPV-CIPF Joint Research Unit Disease Mechanisms and Nanomedicine, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Pablo Bonilla
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Mara Mellado
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Pablo Garcia-Manau
- Maternal-Foetal Medicine Unit, Vall d’Hebron Hospital Campus, E-08035 Barcelona, Spain
| | - Carlota Rodo
- Maternal-Foetal Medicine Unit, Vall d’Hebron Hospital Campus, E-08035 Barcelona, Spain
| | - Ana Alastrue
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | - Eric Lopez
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
| | | | - Ferran Pellise
- Spine Surgery Unit, Hospital Universitari Vall d’Hebron, E-08035 Barcelona, Spain
| | - Snežana Đorđević
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, E-46012, Valencia, Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory, Centro de Investigación Príncipe Felipe, E-46012, Valencia, Spain
| | - Victoria Moreno Manzano
- Neuronal and Tissue Regeneration Laboratory, Centro de Investigación Príncipe Felipe, E-46012 Valencia, Spain
- Correspondence:
| |
Collapse
|
12
|
Lu Y, Zhang W, Tian Z, Liang Q, Liu C, Wu Y, Zhang L, Rong L. The optimal transplantation strategy of umbilical cord mesenchymal stem cells in spinal cord injury: a systematic review and network meta-analysis based on animal studies. Stem Cell Res Ther 2022; 13:441. [PMID: 36056386 PMCID: PMC9438219 DOI: 10.1186/s13287-022-03103-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/31/2022] [Indexed: 12/09/2022] Open
Abstract
Objective Umbilical cord mesenchymal stem cells (UCMSCs) have great potential in the treatment of spinal cord injury. However, the specific therapeutic effect and optimal transplantation strategy are still unclear. Therefore, exploring the optimal treatment strategy of UCMSCs in animal studies by systematic review can provide reference for the development of animal studies and clinical research in the future. Methods Databases of PubMed, Ovid-Embase, Web of Science, CNKI, WanFang, VIP, and CBM were searched for the literature in February 11, 2022. Two independent reviewers performed the literature search, identification, screening, quality assessment, and data extraction. Results and Discussion A total of 40 animal studies were included for combined analysis. In different subgroups, the results of traditional meta-analysis and network meta-analysis were consistent, that is, the therapeutic effect of high-dose (≥ 1 × 106) transplantation of UCMSCs was significantly better than that of low dose (< 1 × 106), the therapeutic effect of local transplantation of UCMSCs was significantly better than that of intravenous transplantation, and the therapeutic effect of subacute transplantation of UCMSCs was significantly better than that of acute and chronic transplantation. However, in view of the inherent risk of bias and limited internal and external validity of the current animal studies, more high-quality, direct comparison studies are needed to further explore the optimal transplantation strategy for UCMSCs in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03103-8.
Collapse
Affiliation(s)
- Yubao Lu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Wei Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Qian Liang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Chenrui Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Yingjie Wu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China. .,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China. .,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.
| |
Collapse
|
13
|
Shang Z, Wang R, Li D, Chen J, Zhang B, Wang M, Wang X, Wanyan P. Spinal Cord Injury: A Systematic Review and Network Meta-Analysis of Therapeutic Strategies Based on 15 Types of Stem Cells in Animal Models. Front Pharmacol 2022; 13:819861. [PMID: 35359872 PMCID: PMC8964098 DOI: 10.3389/fphar.2022.819861] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Objective: The optimal therapeutic strategies of stem cells for spinal cord injury (SCI) are fully explored in animal studies to promote the translation of preclinical findings to clinical practice, also to provide guidance for future animal experiments and clinical studies. Methods: PubMed, Web of Science, Embase, CNKI, Wangfang, VIP, and CBM were searched from inception to September 2021. Screening of search results, data extraction, and references quality evaluation were undertaken independently by two reviewers. Results and Discussion: A total of 188 studies were included for data analysis. Results of traditional meta-analysis showed that all 15 diverse types of stem cells could significantly improve locomotor function of animals with SCI, and results of further network meta-analysis showed that adipose-derived mesenchymal stem cells had the greatest therapeutic potential for SCI. Moreover, a higher dose (≥1 × 106) of stem cell transplantation had better therapeutic effect, transplantation in the subacute phase (3–14 days, excluding 3 days) was the optimal timing, and intralesional transplantation was the optimal route. However, the evidence of current animal studies is of limited quality, and more high-quality research is needed to further explore the optimal therapeutic strategies of stem cells, while the design and implementation of experiments, as well as measurement and reporting of results for animal studies, need to be further improved and standardized to reduce the risk when the results of animal studies are translated to the clinic. Systematic Review Registration: [website], identifier [registration number].
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ruirui Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Dongliang Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jinlei Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Xin Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, China
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| |
Collapse
|
14
|
Yuan T, Shao Y, Zhou X, Liu Q, Zhu Z, Zhou B, Dong Y, Stephanopoulos N, Gui S, Yan H, Liu D. Highly Permeable DNA Supramolecular Hydrogel Promotes Neurogenesis and Functional Recovery after Completely Transected Spinal Cord Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102428. [PMID: 34296471 DOI: 10.1002/adma.202102428] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Regeneration after severe spinal cord injury cannot occur naturally in mammals. Transplanting stem cells to the injury site is a highly promising method, but it faces many challenges because it relies heavily on the microenvironment provided by both the lesion site and delivery material. Although mechanical properties, biocompatibility, and biodegradability of delivery materials have been extensively explored, their permeability has rarely been recognized. Here, a DNA hydrogel is designed with extremely high permeability to repair a 2 mm spinal cord gap in Sprague-Dawley rats. The rats recover basic hindlimb function with detectable motor-evoked potentials, and a renascent neural network is formed via the proliferation and differentiation of both implanted and endogenous stem cells. The signal at the lesion area is conveyed by, on average, 15 newly formed synapses. This hydrogel system offers great potential in clinical trials. Further, it should be easily adaptable to other tissue regeneration applications.
Collapse
Affiliation(s)
- Taoyang Yuan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100071, China
| | - Yu Shao
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xu Zhou
- Center for Molecular Design and Biomimetics, The Biodesign Institute, School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Qian Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100071, China
| | - Zhichao Zhu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Bini Zhou
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yuanchen Dong
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Nicholas Stephanopoulos
- Center for Molecular Design and Biomimetics, The Biodesign Institute, School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China
| | - Hao Yan
- Center for Molecular Design and Biomimetics, The Biodesign Institute, School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Dongsheng Liu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
15
|
Liu A, Kang S, Yu P, Shi L, Zhou L. Transplantation of human urine-derived neural progenitor cells after spinal cord injury in rats. Neurosci Lett 2020; 735:135201. [PMID: 32585253 DOI: 10.1016/j.neulet.2020.135201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 12/25/2022]
Abstract
Spinal cord injury (SCI) is a worldwide problem and transplantation of neural progenitor cells (NPCs) represents a promising treatment strategy. Urine derived induced pluripotent stem cells (UiPSCs) which enable the generation of patient-specific NPCs, provide an invaluable source of autologous cells for future therapeutic applications after SCI. However, the fate and potential contribution of transplanted human UiPSCs-derived NPCs (UiPSC-NPCs) into injured spinal cords remain largely unknown. In this study, using a rat contusive SCI model, we evaluated the survival, migration and differentiation of UiPSC-NPCs after transplantation at subacute phase. Transplanted cells survived and migrated from the site of grafting towards the lesion epicenter. More than 25 % cells survived over 4 weeks post transplantation, with a few of them differentiated into neurons and astrocytes. Cytokine and chemokine levels within the injured spinal cord tissues were measured using multiplex immunoassays to evaluate the immune response. Pro-inflammatory factors and chemokines were significantly decreased at 3 days after UiPSC-NPCs transplantation. At 7 days post transplantation, a lower level of pro-inflammatory factor IFN-γ and a higher level of pro-inflammatory IL-2 were found in UiPSC-NPCs group than in the control. Transplantation of UiPSC-NPCs showed little effect on microglia activation at the lesion epicenter. However, the number of microglia cells at 4 mm rostral to the injury site was significantly decreased. The size of lesion cavity was reduced after transplantation of UiPSC-NPCs. In conclusions, the UiPSC-NPCs transplanted at the subacute phase of SCI showed a beneficial effect on tissue repairing.
Collapse
Affiliation(s)
- Aimei Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Sai Kang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Lingling Shi
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China.
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510530, PR China; Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu 226019, PR China.
| |
Collapse
|
16
|
Zhao Z, Hu X, Wu Z, Chen Q, Shao Q. A Selective P2Y Purinergic Receptor Agonist 2-MesADP Enhances Locomotor Recovery after Acute Spinal Cord Injury. Eur Neurol 2020; 83:195-212. [PMID: 32474563 DOI: 10.1159/000507854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/10/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Spinal cord injury (SCI) causes most severe motor and sensory dysfunctions. In Chinese traditional medicine, the agonist of a purinergic receptor is believed to have a positive effect on SCIs, and 2-Methylthio-adenosine-5'-diphosphate (2-MesADP) is a selective agonist of the P2Y purinergic receptor. METHODS To investigate its therapeutic function and molecular mechanism in SCI, transcriptome analysis associated with weighted gene co-expression network analysis (WGCNA) was carried out at various time points after T9 crush injury. RESULTS 2-MesADP demonstrated recovery of limb motor function at the 6 weeks after injury, accompanied by neuronal regeneration and axon remyelination at 2 and 6 weeks. Furthermore, gene profiling revealed alternated gene expression with the treatment of 2-MesADP. These genes were assigned to a total of 38 modules, followed by gene ontology analysis; of these, 18 represented neuronal apoptosis and regeneration, immune response, synaptic transmission, cell cycle, and angiogenesis. In the neuronal apoptosis and regeneration module, Nefh, NeuroD6, and Dcx in the 2-MesADP group were noticed due to their interesting expression pattern. The gene expression patterns of Mag, Mog, and Cnp, which played key roles in myelination, were significantly changed with the treatment of 2-MesADP. Wnt signal pathway was the most important pathway in 2-MesADP treatment for acute SCI. CONCLUSION 2-MesADP enhanced locomotor recovery in mouse SCI by altering the expression of neuronal apoptosis and remyelination-related genes and Wnt signaling pathways.
Collapse
Affiliation(s)
- Ziru Zhao
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Xiao Hu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Zhourui Wu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Qi Chen
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qihui Shao
- Department of Chinese Traditional Medicine, Tongji Hospital Affiliated to Tongji University, Shanghai, China,
| |
Collapse
|
17
|
Qian K, Xu TY, Wang X, Ma T, Zhang KX, Yang K, Qian TD, Shi J, Li LX, Wang Z. Effects of neural stem cell transplantation on the motor function of rats with contusion spinal cord injuries: a meta-analysis. Neural Regen Res 2020; 15:748-758. [PMID: 31638100 PMCID: PMC6975148 DOI: 10.4103/1673-5374.266915] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective To judge the efficacies of neural stem cell (NSC) transplantation on functional recovery following contusion spinal cord injuries (SCIs). Data sources Studies in which NSCs were transplanted into a clinically relevant, standardized rat model of contusion SCI were identified by searching the PubMed, Embase and Cochrane databases, and the extracted data were analyzed by Stata 14.0. Data selection Inclusion criteria were that NSCs were used in in vivo animal studies to treat contusion SCIs and that behavioral assessment of locomotor functional recovery was performed using the Basso, Beattie, and Bresnahan lo-comotor rating scale. Exclusion criteria included a follow-up of less than 4 weeks and the lack of control groups. Outcome measures The restoration of motor function was assessed by the Basso, Beattie, and Bresnahan locomotor rating scale. Results We identified 1756 non-duplicated papers by searching the aforementioned electronic databases, and 30 full-text articles met the inclusion criteria. A total of 37 studies reported in the 30 articles were included in the meta-analysis. The meta-analysis results showed that transplanted NSCs could improve the motor function recovery of rats following contusion SCIs, to a moderate extent (pooled standardized mean difference (SMD) = 0.73; 95% confidence interval (CI): 0.47-1.00; P < 0.001). NSCs obtained from different donor species (rat: SMD = 0.74; 95% CI: 0.36-1.13; human: SMD = 0.78; 95% CI: 0.31-1.25), at different donor ages (fetal: SMD = 0.67; 95% CI: 0.43-0.92; adult: SMD = 0.86; 95% CI: 0.50-1.22) and from different origins (brain-derived: SMD = 0.59; 95% CI: 0.27-0.91; spinal cord-derived: SMD = 0.51; 95% CI: 0.22-0.79) had similar efficacies on improved functional recovery; however, adult induced pluripotent stem cell-derived NSCs showed no significant efficacies. Furthermore, the use of higher doses of transplanted NSCs or the administration of immunosuppressive agents did not promote better locomotor function recovery (SMD = 0.45; 95% CI: 0.21-0.70). However, shorter periods between the contusion induction and the NSC transplantation showed slightly higher efficacies (acute: SMD = 1.22; 95% CI: 0.81-1.63; subacute: SMD = 0.75; 95% CI: 0.42-1.09). For chronic injuries, NSC implantation did not significantly improve functional recovery (SMD = 0.25; 95% CI: -0.16 to 0.65). Conclusion NSC transplantation alone appears to be a positive yet limited method for the treatment of contusion SCIs.
Collapse
Affiliation(s)
- Kai Qian
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tuo-Ye Xu
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xi Wang
- Department of Intensive Care Unit, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Ma
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Neurosurgery, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Kai-Xin Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province; Department of Neurosurgery, Huangshan City People's Hospital, Huangshan, Anhui Province, China
| | - Kun Yang
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University; Department of Neurosurgery, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Teng-Da Qian
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing; Department of Neurosurgery, Jintan Hospital Affiliated to Jiangsu University, Jintan, Jiangsu Province, China
| | - Jing Shi
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li-Xin Li
- Department of Neurosurgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zheng Wang
- Department of Gerontology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
18
|
High-Dose Neural Stem/Progenitor Cell Transplantation Increases Engraftment and Neuronal Distribution and Promotes Functional Recovery in Rats after Acutely Severe Spinal Cord Injury. Stem Cells Int 2019; 2019:9807978. [PMID: 31565061 PMCID: PMC6745168 DOI: 10.1155/2019/9807978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/04/2018] [Indexed: 01/12/2023] Open
Abstract
Severe spinal cord injury (SCI) leads to permanent, complete paraplegia and places considerable mental and economic burdens on patients, compared with mild to moderate SCI. However, the dose-related effects of the neural stem/precursor cell (NSPC) transplantation on the injury microenvironment, NSPC survival, axonal growth, neuronal distribution, the composition of neurons, oligodendrocytes, and astrocytes in the lesion area and functional recovery have not yet been quantitatively evaluated in the context of severe SCI. In our study, we acutely transplanted 2.5 × 104 or 1.5 × 105 NSPCs/μl into the site of transection SCI. We found that high-dose NSPC transplantation exerted immunomodulatory and neuroprotective effects in the acute phase of severe SCI. In addition, one week later, a remarkable positive relationship was observed between the transplantation dose and the number of surviving NSPCs in severe SCI. At 8 weeks postgrafting, subjects that received the higher cell dose exhibited abundant nerve regeneration, extensive neuronal distribution, increased proportions of neurons and oligodendrocytes, and nascent functional neural network formation in the lesion area. Notably, a significant functional recovery was also observed. Our data suggest that it is important to consider potential dose-related effects on donor cell survival, neuronal distribution, and locomotor recovery in the development of preclinical NSPC transplantation therapy for severe SCI.
Collapse
|
19
|
Cell therapy for spinal cord injury using induced pluripotent stem cells. Regen Ther 2019; 11:75-80. [PMID: 31245451 PMCID: PMC6581851 DOI: 10.1016/j.reth.2019.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/25/2019] [Accepted: 05/28/2019] [Indexed: 01/25/2023] Open
Abstract
For the past few decades, spinal cord injury (SCI) has been believed to be an incurable traumatic condition, but with recent developments in stem cell biology, the field of regenerative medicine has gained hopeful momentum in the development of a treatment for this challenging pathology. Among the treatment candidates, transplantation of neural precursor cells has gained remarkable attention as a reasonable therapeutic intervention to replace the damaged central nervous system cells and promote functional recovery. Here, we highlight transplantation therapy techniques using induced pluripotent stem cells to treat SCI and review the recent research giving consideration to future clinical applications.
Collapse
Key Words
- ASIA, American Spinal Injury Association
- C-ABC, chondroitinase ABC
- CSPGs, chondroitin sulfate proteoglycans
- CST, corticospinal tract
- CiRA, the Center for iPS Cell Research and Application
- Clinical application
- ESCs, embryonic stem cells
- GCV, ganciclovir
- GSI, γ-secretase inhibitor
- HLA, human leukocyte antigen
- HMGB1, high mobility group box-1
- HSVtk, herpes simplex virus type I thymidine kinase
- Induced pluripotent stem cells
- MLR, mixed lymphocyte reaction
- NHPs, nonhuman primates
- NPCs, neural precursor cells
- Neural precursor cells
- OPCs, oligodendrocyte progenitor cells
- PBMCs, peripheral blood mononuclear cells
- SCI, spinal cord injury
- SLA, swine leukocyte antigen
- Spinal cord injury
- drNPCs, directly reprogrammed neural precursor cells
- iPSCs, induced pluripotent stem cells
Collapse
|
20
|
Kobayakawa K, Ohkawa Y, Yoshizaki S, Tamaru T, Saito T, Kijima K, Yokota K, Hara M, Kubota K, Matsumoto Y, Harimaya K, Ozato K, Masuda T, Tsuda M, Tamura T, Inoue K, Edgerton VR, Iwamoto Y, Nakashima Y, Okada S. Macrophage centripetal migration drives spontaneous healing process after spinal cord injury. SCIENCE ADVANCES 2019; 5:eaav5086. [PMID: 31106270 PMCID: PMC6520026 DOI: 10.1126/sciadv.aav5086] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 04/11/2019] [Indexed: 05/31/2023]
Abstract
Traumatic spinal cord injury (SCI) brings numerous inflammatory cells, including macrophages, from the circulating blood to lesions, but pathophysiological impact resulting from spatiotemporal dynamics of macrophages is unknown. Here, we show that macrophages centripetally migrate toward the lesion epicenter after infiltrating into the wide range of spinal cord, depending on the gradient of chemoattractant C5a. However, macrophages lacking interferon regulatory factor 8 (IRF8) cannot migrate toward the epicenter and remain widely scattered in the injured cord with profound axonal loss and little remyelination, resulting in a poor functional outcome after SCI. Time-lapse imaging and P2X/YRs blockade revealed that macrophage migration via IRF8 was caused by purinergic receptors involved in the C5a-directed migration. Conversely, pharmacological promotion of IRF8 activation facilitated macrophage centripetal movement, thereby improving the SCI recovery. Our findings reveal the importance of macrophage centripetal migration via IRF8, providing a novel therapeutic target for central nervous system injury.
Collapse
Affiliation(s)
- Kazu Kobayakawa
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kensuke Kubota
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsumi Harimaya
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, NICHD, National Institutes of Health, Bethesda, MD, USA
| | - Takahiro Masuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - V. Reggie Edgerton
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
21
|
Sankavaram SR, Hakim R, Covacu R, Frostell A, Neumann S, Svensson M, Brundin L. Adult Neural Progenitor Cells Transplanted into Spinal Cord Injury Differentiate into Oligodendrocytes, Enhance Myelination, and Contribute to Recovery. Stem Cell Reports 2019; 12:950-966. [PMID: 31031190 PMCID: PMC6524946 DOI: 10.1016/j.stemcr.2019.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Long-term survival and integration of neural progenitor cells (NPCs) transplanted following spinal cord injury (SCI) have been observed. However, questions concerning the differentiation choice, the mechanism of action, and the contribution of NPCs to functional recovery remains unanswered. Therefore, we investigated the differentiation of NPCs, global transcriptomal changes in transplanted NPCs, the effect of NPCs on neuroinflammation, and the causality between NPC transplantation and functional recovery. We found that NPCs transplanted following SCI differentiate mainly into oligodendrocytes and enhance myelination, upregulate genes related to synaptic signaling and mitochondrial activity, and downregulate genes related to cytokine production and immune system response. NPCs suppress the expression of pro-inflammatory cytokines/chemokines; moreover, NPC ablation confirm that NPCs were responsible for enhanced recovery in hindlimb locomotor function. Understanding the reaction of transplanted NPCs is important for exploiting their full potential. Existence of causality implies that NPCs are useful in the treatment of SCI. NPCs differentiate mainly into oligodendrocytes and enhance myelination NPCs suppress expression of pro-inflammatory cytokines/chemokines Causality exists between transplantation of NPCs and functional recovery NPCs upregulate genes related to synaptic signaling, oligodendrocytes/myelination
Collapse
Affiliation(s)
- Sreenivasa Raghavan Sankavaram
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Departments of Neurology and Neurosurgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Ramil Hakim
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ruxandra Covacu
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Departments of Neurology and Neurosurgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Arvid Frostell
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Susanne Neumann
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Departments of Neurology and Neurosurgery, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; Center of Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden; Departments of Neurology and Neurosurgery, Karolinska University Hospital, 17176 Stockholm, Sweden.
| |
Collapse
|
22
|
The Fate of Transplanted Periodontal Ligament Stem Cells in Surgically Created Periodontal Defects in Rats. Int J Mol Sci 2019; 20:ijms20010192. [PMID: 30621073 PMCID: PMC6337301 DOI: 10.3390/ijms20010192] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/25/2018] [Accepted: 12/28/2018] [Indexed: 12/19/2022] Open
Abstract
Periodontal disease is chronic inflammation that leads to the destruction of tooth-supporting periodontal tissues. We devised a novel method (“cell transfer technology”) to transfer cells onto a scaffold surface and reported the potential of the technique for regenerative medicine. The aim of this study is to examine the efficacy of this technique in periodontal regeneration and the fate of transplanted cells. Human periodontal ligament stem cells (PDLSCs) were transferred to decellularized amniotic membrane and transplanted into periodontal defects in rats. Regeneration of tissues was examined by microcomputed tomography and histological observation. The fate of transplanted PDLSCs was traced using PKH26 and human Alu sequence detection by PCR. Imaging showed more bone in PDLSC-transplanted defects than those in control (amnion only). Histological examination confirmed the enhanced periodontal tissue formation in PDLSC defects. New formation of cementum, periodontal ligament, and bone were prominently observed in PDLSC defects. PKH26-labeled PDLSCs were found at limited areas in regenerated periodontal tissues. Human Alu sequence detection revealed that the level of Alu sequence was not increased, but rather decreased. This study describes a novel stem cell transplantation strategy for periodontal disease using the cell transfer technology and offers new insight for cell-based periodontal regeneration.
Collapse
|
23
|
Ryu CM, Yu HY, Lee HY, Shin JH, Lee S, Ju H, Paulson B, Lee S, Kim S, Lim J, Heo J, Hong KS, Chung HM, Kim JK, Shin DM, Choo MS. Longitudinal intravital imaging of transplanted mesenchymal stem cells elucidates their functional integration and therapeutic potency in an animal model of interstitial cystitis/bladder pain syndrome. Am J Cancer Res 2018; 8:5610-5624. [PMID: 30555567 PMCID: PMC6276303 DOI: 10.7150/thno.27559] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Rationale: Mesenchymal stem cell (MSC) therapy may be a novel approach to improve interstitial cystitis/bladder pain syndrome (IC/BPS), an intractable disease characterized by severe pelvic pain and urinary frequency. Unfortunately, the properties of transplanted stem cells have not been directly analyzed in vivo, which hampers elucidation of the therapeutic mechanisms of these cells and optimization of transplantation protocols. Here, we monitored the behaviors of multipotent stem cells (M-MSCs) derived from human embryonic stem cells (hESCs) in real time using a novel combination of in vivo confocal endoscopic and microscopic imaging and demonstrated their improved therapeutic potency in a chronic IC/BPS animal model. Methods: Ten-week-old female Sprague-Dawley rats were instilled with 10 mg of protamine sulfate followed by 750 μg of lipopolysaccharide weekly for 5 weeks. The sham group was instilled with phosphate-buffered saline (PBS). Thereafter, the indicated dose (0.1, 0.25, 0.5, and 1×106 cells) of M-MSCs or PBS was injected once into the outer layer of the bladder. The distribution, perivascular integration, and therapeutic effects of M-MSCs were monitored by in vivo endoscopic and confocal microscopic imaging, awake cystometry, and histological and gene expression analyses. Results: A novel combination of longitudinal intravital confocal fluorescence imaging and microcystoscopy in living animals, together with immunofluorescence analysis of bladder tissues, demonstrated that transplanted M-MSCs engrafted following differentiation into multiple cell types and gradually integrated into a perivascular-like structure until 30 days after transplantation. The beneficial effects of transplanted M-MSCs on bladder voiding function and the pathological characteristics of the bladder were efficient and long-lasting due to the stable engraftment of these cells. Conclusion: This longitudinal bioimaging study of transplanted hESC-derived M-MSCs in living animals reveals their long-term functional integration, which underlies the improved therapeutic effects of these cells on IC/BPS.
Collapse
|
24
|
Yu H, Wang X, Kang F, Chen Z, Meng Y, Dai M. Neuroprotective effects of midazolam on focal cerebral ischemia in rats through anti‑apoptotic mechanisms. Int J Mol Med 2018; 43:443-451. [PMID: 30431057 DOI: 10.3892/ijmm.2018.3973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 02/02/2018] [Indexed: 11/05/2022] Open
Abstract
Stroke is a cerebrovascular circulatory disorder and its high mortality rate represents a prominent threat to human health. Subsequent apoptosis and cytotoxicity are the main causes underlying the poor prognosis. Midazolam (MDZ) is a benzodiazepine drug that is clinically used during surgical procedures and for the treatment of insomnia, with a potential ability to treat stroke. The protective effect of MDZ was investigated on glutamate‑induced cortical neuronal injuries in vitro and transient middle cerebral artery occlusion (tMCAO) rat models in vivo. Western blot analysis and semi quantitative RT‑PCR were used to evaluate the potential underlying mechanisms. In vitro studies revealed that MDZ regulated apoptosis‑associated gene expression and inhibited lactate dehydrogenase (LDH) release, protecting against neuronal damage. In vivo studies revealed that MDZ reduced LDH‑induced neuronal damage by reducing LDH release from the peripheral blood, and brain tissue staining revealed that MDZ protected neurons during tMCAO. MDZ protected neurons under an ischemic environment by inhibiting LDH release and regulating apoptosis‑associated gene expression to reduce cytotoxicity and apoptosis. These results provide a reliable basis for further studies on the effect of MDZ, to improve the prognosis of cerebral infarction.
Collapse
Affiliation(s)
- Hang Yu
- Intensive Care Unit, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Xiaozhi Wang
- Intensive Care Unit, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fuxin Kang
- Intensive Care Unit, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Zhile Chen
- Intensive Care Unit, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Yunxia Meng
- Intensive Care Unit, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Mingming Dai
- Department of Internal Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
25
|
Bose RJC, Mattrey RF. Accomplishments and challenges in stem cell imaging in vivo. Drug Discov Today 2018; 24:492-504. [PMID: 30342245 DOI: 10.1016/j.drudis.2018.10.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 09/24/2018] [Accepted: 10/13/2018] [Indexed: 02/08/2023]
Abstract
Stem cell therapies have demonstrated promising preclinical results, but very few applications have reached the clinic owing to safety and efficacy concerns. Translation would benefit greatly if stem cell survival, distribution and function could be assessed in vivo post-transplantation, particularly in patients. Advances in molecular imaging have led to extraordinary progress, with several strategies being deployed to understand the fate of stem cells in vivo using magnetic resonance, scintigraphy, PET, ultrasound and optical imaging. Here, we review the recent advances, challenges and future perspectives and opportunities in stem cell tracking and functional assessment, as well as the advantages and challenges of each imaging approach.
Collapse
Affiliation(s)
- Rajendran J C Bose
- Department of Radiology and Advanced Imaging Research Center, 5323 Harry Hines Blvd, UT Southwestern Medical Center, Dallas, TX 75390-8514, USA; Current affiliation: Molecular Imaging Program at Stanford (MIPS) and the Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, Stanford, CA 94305-5427, USA
| | - Robert F Mattrey
- Department of Radiology and Advanced Imaging Research Center, 5323 Harry Hines Blvd, UT Southwestern Medical Center, Dallas, TX 75390-8514, USA.
| |
Collapse
|
26
|
Ramadan WS, Abdel-Hamid GA, Al-Karim S, Zakar NAMB, Elassouli MZ. Neuroectodermal stem cells: A remyelinating potential in acute compressed spinal cord injury in rat model. J Biosci 2018. [DOI: 10.1007/s12038-018-9812-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Matsui A, Uchida S, Hayashi A, Kataoka K, Itaka K. Prolonged engraftment of transplanted hepatocytes in the liver by transient pro-survival factor supplementation using ex vivo mRNA transfection. J Control Release 2018; 285:1-11. [PMID: 29966689 DOI: 10.1016/j.jconrel.2018.06.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 12/17/2022]
Abstract
Cell transplantation therapy needs engraftment efficiency improvement of transplanted cells to the host tissues. Ex vivo transfection of a pro-survival gene to transplanted cells is a possible solution; however prolonged expression and/or genomic integration of the gene can be cancer promoting. To supply pro-survival protein only when it is needed, we used mRNA transfection, which exhibits transient protein expression profiles without the risk of genomic integration. Ex vivo transfection of mRNA encoding Bcl-2, a pro-survival factor, led to enhanced hepatocyte engraftment in both of normal and diseased mouse liver, effectively supporting liver function in a model of chronic hepatitis. The transplanted hepatocytes maintained their viability and function in the liver for at least one month, though Bcl-2 expression from mRNA was sustained for just a few days. Mechanism analyses suggest that Bcl-2 inhibits Kupffer cell-mediated hepatocyte clearance, which occurs within 2 days after transplantation. Within 2 days, hepatocytes migrated to the liver parenchyma, presumably a suitable place for the hepatocytes to survive without Bcl-2 expression. Thus, the duration of Bcl-2 expression from mRNA was sufficient to achieve prolonged engraftment. Ex vivo mRNA transfection allows supply of pro-survival factors to transplanted cells with minimal safety concerns accompanying prolonged expression, providing an effective platform to improve engraftment efficiency in cell transplantation therapy.
Collapse
Affiliation(s)
- Akitsugu Matsui
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan; Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan
| | - Satoshi Uchida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo 113-8656, Japan.
| | - Akimasa Hayashi
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Keiji Itaka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa 210-0821, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Chiyoda, Tokyo 101-0062, Japan.
| |
Collapse
|
28
|
André EM, Daviaud N, Sindji L, Cayon J, Perrot R, Montero-Menei CN. A novel ex vivo Huntington's disease model for studying GABAergic neurons and cell grafts by laser microdissection. PLoS One 2018; 13:e0193409. [PMID: 29505597 PMCID: PMC5837106 DOI: 10.1371/journal.pone.0193409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/09/2018] [Indexed: 02/05/2023] Open
Abstract
Organotypic brain slice cultures have been recently used to study neurodegenerative disorders such as Parkinson’s disease and Huntington’s disease (HD). They preserve brain three-dimensional architecture, synaptic connectivity and brain cells microenvironment. Here, we developed an innovative model of Huntington’s disease from coronal rat brain slices, that include all the areas involved in the pathology. HD-like neurodegeneration was obtained in only one week, in a single step, during organotypic slice preparation, without the use of neurotoxins. HD-like histopathology was analysed and after one week, a reduction of 40% of medium spiny neurons was observed. To analyse new therapeutic approaches in this innovative HD model, we developed a novel protocol of laser microdissection to isolate and analyse by RT-qPCR, grafted cells as well as surrounding tissue of fresh organotypic slices. We determined that laser microdissection could be performed on a 400μm organotypic slice after alcohol dehydration protocol, allowing the analysis of mRNA expression in the rat tissue as well as in grafted cells. In conclusion, we developed a new approach for modeling Huntington's disease ex vivo, and provided a useful innovative method for screening new potential therapies for neurodegenerative diseases especially when associated with laser microdissection.
Collapse
Affiliation(s)
- E. M. André
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - N. Daviaud
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - L. Sindji
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
| | - J. Cayon
- PACEM, Angers University, Angers, France
| | - R. Perrot
- SCIAM, Angers University, Angers, France
| | - C. N. Montero-Menei
- CRCINA, INSERM, Université de Nantes, Université d’Angers, Angers, France
- * E-mail:
| |
Collapse
|
29
|
Nagoshi N, Okano H. iPSC-derived neural precursor cells: potential for cell transplantation therapy in spinal cord injury. Cell Mol Life Sci 2018; 75:989-1000. [PMID: 28993834 PMCID: PMC11105708 DOI: 10.1007/s00018-017-2676-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 09/03/2017] [Accepted: 10/02/2017] [Indexed: 12/12/2022]
Abstract
A number of studies have demonstrated that transplantation of neural precursor cells (NPCs) promotes functional recovery after spinal cord injury (SCI). However, the NPCs had been mostly harvested from embryonic stem cells or fetal tissue, raising the ethical concern. Yamanaka and his colleagues established induced pluripotent stem cells (iPSCs) which could be generated from somatic cells, and this innovative development has made rapid progression in the field of SCI regeneration. We and other groups succeeded in producing NPCs from iPSCs, and demonstrated beneficial effects after transplantation for animal models of SCI. In particular, efficacy of human iPSC-NPCs in non-human primate SCI models fostered momentum of clinical application for SCI patients. At the same time, however, artificial induction methods in iPSC technology created alternative issues including genetic and epigenetic abnormalities, and tumorigenicity after transplantation. To overcome these problems, it is critically important to select origins of somatic cells, use integration-free system during transfection of reprogramming factors, and thoroughly investigate the characteristics of iPSC-NPCs with respect to quality management. Moreover, since most of the previous studies have focused on subacute phase of SCI, establishment of effective NPC transplantation should be evaluated for chronic phase hereafter. Our group is currently preparing clinical-grade human iPSC-NPCs, and will move forward toward clinical study for subacute SCI patients soon in the near future.
Collapse
Affiliation(s)
- Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjukuku, Tokyo, 160-8582, Japan.
| |
Collapse
|
30
|
Improved efficacy and in vivo cellular properties of human embryonic stem cell derivative in a preclinical model of bladder pain syndrome. Sci Rep 2017; 7:8872. [PMID: 28827631 PMCID: PMC5567131 DOI: 10.1038/s41598-017-09330-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is an intractable disease characterized by severe pelvic pain and urinary frequency. Mesenchymal stem cell (MSC) therapy is a promising approach to treat incurable IC/BPS. Here, we show greater therapeutic efficacy of human embryonic stem cell (hESC)-derived multipotent stem cells (M-MSCs) than adult bone-marrow (BM)-derived counterparts for treating IC/BPS and also monitor long-term safety and in vivo properties of transplanted M-MSCs in living animals. Controlled hESC differentiation and isolation procedures resulted in pure M-MSCs displaying typical MSC behavior. In a hydrochloric-acid instillation-induced IC/BPS animal model, a single local injection of M-MSCs ameliorated bladder symptoms of IC/BPS with superior efficacy compared to BM-derived MSCs in ameliorating bladder voiding function and histological injuries including urothelium denudation, mast-cell infiltration, tissue fibrosis, apoptosis, and visceral hypersensitivity. Little adverse outcomes such as abnormal growth, tumorigenesis, or immune-mediated transplant rejection were observed over 12-months post-injection. Intravital confocal fluorescence imaging tracked the persistence of the transplanted cells over 6-months in living animals. The infused M-MSCs differentiated into multiple cell types and gradually integrated into vascular-like structures. The present study provides the first evidence for improved therapeutic efficacy, long-term safety, and in vivo distribution and cellular properties of hESC derivatives in preclinical models of IC/BPS.
Collapse
|
31
|
Hara M, Kobayakawa K, Ohkawa Y, Kumamaru H, Yokota K, Saito T, Kijima K, Yoshizaki S, Harimaya K, Nakashima Y, Okada S. Interaction of reactive astrocytes with type I collagen induces astrocytic scar formation through the integrin-N-cadherin pathway after spinal cord injury. Nat Med 2017. [PMID: 28628111 DOI: 10.1038/nm.4354] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central nervous system (CNS) injury transforms naive astrocytes into reactive astrocytes, which eventually become scar-forming astrocytes that can impair axonal regeneration and functional recovery. This sequential phenotypic change, known as reactive astrogliosis, has long been considered unidirectional and irreversible. However, we report here that reactive astrocytes isolated from injured spinal cord reverted in retrograde to naive astrocytes when transplanted into a naive spinal cord, whereas they formed astrocytic scars when transplanted into injured spinal cord, indicating the environment-dependent plasticity of reactive astrogliosis. We also found that type I collagen was highly expressed in the spinal cord during the scar-forming phase and induced astrocytic scar formation via the integrin-N-cadherin pathway. In a mouse model of spinal cord injury, pharmacological blockade of reactive astrocyte-type I collagen interaction prevented astrocytic scar formation, thereby leading to improved axonal regrowth and better functional outcomes. Our findings reveal environmental cues regulating astrocytic fate decisions, thereby providing a potential therapeutic target for CNS injury.
Collapse
Affiliation(s)
- Masamitsu Hara
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiromi Kumamaru
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsumi Harimaya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
32
|
Increasing Human Neural Stem Cell Transplantation Dose Alters Oligodendroglial and Neuronal Differentiation after Spinal Cord Injury. Stem Cell Reports 2017; 8:1534-1548. [PMID: 28479305 PMCID: PMC5469937 DOI: 10.1016/j.stemcr.2017.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 01/13/2023] Open
Abstract
Multipotent human central nervous system-derived neural stem cells transplanted at doses ranging from 10,000 (low) to 500,000 (very high) cells differentiated predominantly into the oligodendroglial lineage. However, while the number of engrafted cells increased linearly in relationship to increasing dose, the proportion of oligodendrocytic cells declined. Increasing dose resulted in a plateau of engraftment, enhanced neuronal differentiation, and increased distal migration caudal to the transplantation sites. Dose had no effect on terminal sensory recovery or open-field locomotor scores. However, total human cell number and decreased oligodendroglial proportion were correlated with hindlimb girdle coupling errors. Conversely, greater oligodendroglial proportion was correlated with increased Ab step pattern, decreased swing speed, and increased paw intensity, consistent with improved recovery. These data suggest that transplant dose, and/or target niche parameters can regulate donor cell engraftment, differentiation/maturation, and lineage-specific migration profiles. SCI niche may have a limited capacity for donor cell engraftment Dose alters the donor cell lineage-specific fate and migration profile Increasing hindlimb girdle couplings errors may be due to increased total cell numbers Greater proportion of oligodendroglial cells improves locomotor recovery
Collapse
|
33
|
Nagoshi N, Okano H. Applications of induced pluripotent stem cell technologies in spinal cord injury. J Neurochem 2017; 141:848-860. [PMID: 28199003 DOI: 10.1111/jnc.13986] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022]
Abstract
Numerous basic research studies have suggested the potential efficacy of neural precursor cell (NPC) transplantation in spinal cord injury (SCI). However, in most such studies, the origin of the cells used was mainly fetal tissue or embryonic stem cells, both of which carry potential ethical concerns with respect to clinical use. The development of induced pluripotent stem cells (iPSCs) opened a new path toward regenerative medicine for SCI. iPSCs can be generated from somatic cells by induction of transcription factors, and induced to differentiate into NPCs with characteristics of cells of the central nervous system. The beneficial effect of iPSC-derived NPC transplantation has been reported from our group and others working in rodent and non-human primate models. These promising results facilitate the application of iPSCs for clinical applications in SCI patients. However, iPSCs also have issues, such as genetic/epigenetic abnormalities and tumorigenesis because of the artificial induction method, that must be addressed prior to clinical use. The selection of somatic cells, generation of integration-free iPSCs, and characterization of differentiated NPCs with thorough quality management are all needed to address these potential risks. To enhance the efficacy of the transplanted iPSC-NPCs, especially at chronic phase of SCI, administration of a chondroitinase or semaphorin3A inhibitor represents a potentially important means of promoting axonal regeneration through the lesion site. The combined use of rehabilitation with such cell therapy approaches is also important, as repetitive training enhances neurite outgrowth of transplanted cells and strengthens neural circuits at central pattern generators. Our group has already evaluated clinical grade iPSC-derived NPCs, and we look forward to initiating clinical testing as the next step toward determining whether this approach is safe and effective for clinical use. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Yokota K, Kobayakawa K, Saito T, Hara M, Kijima K, Ohkawa Y, Harada A, Okazaki K, Ishihara K, Yoshida S, Kudo A, Iwamoto Y, Okada S. Periostin Promotes Scar Formation through the Interaction between Pericytes and Infiltrating Monocytes/Macrophages after Spinal Cord Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:639-653. [PMID: 28082119 DOI: 10.1016/j.ajpath.2016.11.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/11/2016] [Accepted: 11/22/2016] [Indexed: 01/13/2023]
Abstract
Scar formation is a prominent pathological feature of traumatic central nervous system (CNS) injury, which has long been implicated as a major impediment to the CNS regeneration. However, the factors affecting such scar formation remain to be elucidated. We herein demonstrate that the extracellular matrix protein periostin (POSTN) is a key player in scar formation after traumatic spinal cord injury (SCI). Using high-throughput RNA sequencing data sets, we found that the genes involved in the extracellular region, such as POSTN, were significantly expressed in the injured spinal cord. The expression of POSTN peaked at 7 days after SCI, predominantly in the scar-forming pericytes. Notably, we found that genetic deletion of POSTN in mice reduced scar formation at the lesion site by suppressing the proliferation of the pericytes. Conversely, we found that recombinant POSTN promoted the migration capacity of the monocytes/macrophages and increased the expression of tumor necrosis factor-α from the monocytes/macrophages in vitro, which facilitated the proliferation of pericytes. Furthermore, we revealed that the pharmacological blockade of POSTN suppressed scar formation and improved the long-term functional outcome after SCI. Our findings suggest a potential mechanism whereby POSTN regulates the scar formation after SCI and provide significant evidence that POSTN is a promising therapeutic target for CNS injury.
Collapse
Affiliation(s)
- Kazuya Yokota
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Department of Transcriptomics, Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihito Harada
- Department of Transcriptomics, Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Ishihara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigeo Yoshida
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Kudo
- Department of Biological Information, Tokyo Institute of Technology, Yokohama, Japan
| | - Yukihide Iwamoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
35
|
Abstract
Reactive astrogliosis occurs after central nervous system (CNS) injuries whereby resident astrocytes form rapid responses along a graded continuum. Following CNS lesions, naïve astrocytes are converted into reactive astrocytes and eventually into scar-forming astrocytes that block axon regeneration and neural repair. It has been known for decades that scarring development and its related extracellular matrix molecules interfere with regeneration of injured axons after CNS injury, but the cellular and molecular mechanisms for controlling astrocytic scar formation and maintenance are not well known. Recent use of various genetic tools has made tremendous progress in better understanding genesis of reactive astrogliosis. Especially, the latest experiments demonstrate environment-dependent plasticity of reactive astrogliosis because reactive astrocytes isolated from injured spinal cord form scarring astrocytes when transplanted into injured spinal cord, but revert in retrograde to naive astrocytes when transplanted into naive spinal cord. The interactions between upregulated type I collagen and its receptor integrin β1 and the N-cadherin-mediated cell adhesion appear to play major roles for local astrogliosis around the lesion. This review centers on the environment-dependent plasticity of reactive astrogliosis after spinal cord injury and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Fatima M Nathan
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
36
|
Nori S, Nakamura M, Okano H. Plasticity and regeneration in the injured spinal cord after cell transplantation therapy. PROGRESS IN BRAIN RESEARCH 2017; 231:33-56. [DOI: 10.1016/bs.pbr.2016.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Uchida S, Hayakawa K, Ogata T, Tanaka S, Kataoka K, Itaka K. Treatment of spinal cord injury by an advanced cell transplantation technology using brain-derived neurotrophic factor-transfected mesenchymal stem cell spheroids. Biomaterials 2016; 109:1-11. [DOI: 10.1016/j.biomaterials.2016.09.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/06/2016] [Accepted: 09/09/2016] [Indexed: 01/01/2023]
|
38
|
Yamashita T, Liu W, Matsumura Y, Miyagi R, Zhai Y, Kusaki M, Hishikawa N, Ohta Y, Kim SM, Kwak TH, Han DW, Abe K. Novel Therapeutic Transplantation of Induced Neural Stem Cells for Stroke. Cell Transplant 2016; 26:461-467. [PMID: 27653466 DOI: 10.3727/096368916x692988] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Somatic cells can be directly converted into induced neural stem cells (iNSCs) by defined transcription factors. However, the therapeutic effect of undifferentiated iNSCs on ischemic stroke has not been demonstrated. In this study, we used a mouse model of transient middle cerebral artery occlusion (tMCAO). iNSCs (5 × 105) were injected directly into the ipsilateral striatum and cortex 24 h after tMCAO. Histological analysis was performed at 7 days, 28 days, and 8 months after tMCAO. We found that iNSC transplantation successfully improved the survival rate of stroke model mice with significant functional recovery from the stroke. The fate of engrafted iNSCs was that the majority of iNSCs had differentiated into astroglial cells but not into neural cells in both the sham-operated brain and the poststroke brain without forming a tumor up to 8 months after tMCAO. Our data suggest that the directly converted iNSCs can be regarded as a candidate of safe cell resource for transplantation therapy in patients suffering from ischemic stroke.
Collapse
|
39
|
Teng M, Love MI, Davis CA, Djebali S, Dobin A, Graveley BR, Li S, Mason CE, Olson S, Pervouchine D, Sloan CA, Wei X, Zhan L, Irizarry RA. A benchmark for RNA-seq quantification pipelines. Genome Biol 2016; 17:74. [PMID: 27107712 PMCID: PMC4842274 DOI: 10.1186/s13059-016-0940-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 04/08/2016] [Indexed: 02/07/2023] Open
Abstract
Obtaining RNA-seq measurements involves a complex data analytical process with a large number of competing algorithms as options. There is much debate about which of these methods provides the best approach. Unfortunately, it is currently difficult to evaluate their performance due in part to a lack of sensitive assessment metrics. We present a series of statistical summaries and plots to evaluate the performance in terms of specificity and sensitivity, available as a R/Bioconductor package (http://bioconductor.org/packages/rnaseqcomp). Using two independent datasets, we assessed seven competing pipelines. Performance was generally poor, with two methods clearly underperforming and RSEM slightly outperforming the rest.
Collapse
Affiliation(s)
- Mingxiang Teng
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.,Department of Biostatistics, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Michael I Love
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.,Department of Biostatistics, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Carrie A Davis
- Functional Genomics Group, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY, 11724, USA
| | - Sarah Djebali
- Bioinformatics and Genomics Programme, Centre for Genomic Regulation (CRG) and UPF, Doctor Aiguader, 88, Barcelona, 08003, Spain
| | - Alexander Dobin
- Functional Genomics Group, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY, 11724, USA
| | - Brenton R Graveley
- Department of Genetics and Genome Sciences, Institute for System Genomics, UConn Health Center, Farmington, CT, 06030, USA
| | - Sheng Li
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Sara Olson
- Department of Genetics and Genome Sciences, Institute for System Genomics, UConn Health Center, Farmington, CT, 06030, USA
| | - Dmitri Pervouchine
- Bioinformatics and Genomics Programme, Centre for Genomic Regulation (CRG) and UPF, Doctor Aiguader, 88, Barcelona, 08003, Spain
| | - Cricket A Sloan
- Department of Genetics, Stanford University, 300 Pasteur Drive, MC-5477, Stanford, CA, 94305, USA
| | - Xintao Wei
- Department of Genetics and Genome Sciences, Institute for System Genomics, UConn Health Center, Farmington, CT, 06030, USA
| | - Lijun Zhan
- Department of Genetics and Genome Sciences, Institute for System Genomics, UConn Health Center, Farmington, CT, 06030, USA
| | - Rafael A Irizarry
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Department of Biostatistics, Harvard TH Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
40
|
Shoemaker LD, Kornblum HI. Neural Stem Cells (NSCs) and Proteomics. Mol Cell Proteomics 2015; 15:344-54. [PMID: 26494823 PMCID: PMC4739658 DOI: 10.1074/mcp.o115.052704] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Indexed: 01/09/2023] Open
Abstract
Neural stem cells (NSCs) can self-renew and give rise to the major cell types of the CNS. Studies of NSCs include the investigation of primary, CNS-derived cells as well as animal and human embryonic stem cell (ESC)-derived and induced pluripotent stem cell (iPSC)-derived sources. NSCs provide a means with which to study normal neural development, neurodegeneration, and neurological disease and are clinically relevant sources for cellular repair to the damaged and diseased CNS. Proteomics studies of NSCs have the potential to delineate molecules and pathways critical for NSC biology and the means by which NSCs can participate in neural repair. In this review, we provide a background to NSC biology, including the means to obtain them and the caveats to these processes. We then focus on advances in the proteomic interrogation of NSCs. This includes the analysis of posttranslational modifications (PTMs); approaches to analyzing different proteomic compartments, such the secretome; as well as approaches to analyzing temporal differences in the proteome to elucidate mechanisms of differentiation. We also discuss some of the methods that will undoubtedly be useful in the investigation of NSCs but which have not yet been applied to the field. While many proteomics studies of NSCs have largely catalogued the proteome or posttranslational modifications of specific cellular states, without delving into specific functions, some have led to understandings of functional processes or identified markers that could not have been identified via other means. Many challenges remain in the field, including the precise identification and standardization of NSCs used for proteomic analyses, as well as how to translate fundamental proteomics studies to functional biology. The next level of investigation will require interdisciplinary approaches, combining the skills of those interested in the biochemistry of proteomics with those interested in modulating NSC function.
Collapse
Affiliation(s)
- Lorelei D Shoemaker
- From the ‡Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University, 300 Pasteur Drive, Stanford, CA 94305
| | - Harley I Kornblum
- §NPI-Semel Institute for Neuroscience & Human Behavior, Departments of Psychiatry and Biobehavioral Sciences, and of Molecular and Medical Pharmacology, The Molecular Biology Institute, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los, Angeles, CA 90095
| |
Collapse
|
41
|
Kubota K, Kobayakaya K, Okada S, Shiba K, Iwamoto Y. Hyperglycemia During Acute Spinal Cord Injury Is a Detrimental Factor That Impairs Functional Improvement in Acute C3-C4 Cervical Cord Injury Patients Without Any Bony Damages. Intensive Care Med Exp 2015. [PMCID: PMC4797914 DOI: 10.1186/2197-425x-3-s1-a33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
42
|
Engrafted Neural Stem/Progenitor Cells Promote Functional Recovery through Synapse Reorganization with Spared Host Neurons after Spinal Cord Injury. Stem Cell Reports 2015; 5:264-77. [PMID: 26190527 PMCID: PMC4618657 DOI: 10.1016/j.stemcr.2015.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 12/26/2022] Open
Abstract
Neural stem/progenitor cell (NSPC) transplantation is a promising therapeutic strategy for spinal cord injury (SCI). However, the efficacy of NSPC transplantation on severe SCI is poorly understood. We herein show that NSPC transplantation promotes functional recovery after mild and moderate SCI, but not after severe SCI. In severe SCI mice, there were few remaining host neurons within the range of NSPC engraftment; thus, we examined whether the co-distribution of transplant and host is a contributory factor for functional improvement. A cellular selective analysis using laser microdissection revealed that drug-induced host neuronal ablation considerably decreased the synaptogenic potential of the engrafted NSPCs. Furthermore, following host neuronal ablation, neuronal retrograde tracing showed less propriospinal relay connections bridging the lesion after NSPC transplantation. Our findings suggest that the interactive synaptic reorganization between engrafted NSPCs and spared host neurons is crucial for functional recovery, providing significant insight for establishing therapeutic strategies for severe SCI.
Collapse
|
43
|
Iwai H, Shimada H, Nishimura S, Kobayashi Y, Itakura G, Hori K, Hikishima K, Ebise H, Negishi N, Shibata S, Habu S, Toyama Y, Nakamura M, Okano H. Allogeneic Neural Stem/Progenitor Cells Derived From Embryonic Stem Cells Promote Functional Recovery After Transplantation Into Injured Spinal Cord of Nonhuman Primates. Stem Cells Transl Med 2015; 4:708-19. [PMID: 26019226 DOI: 10.5966/sctm.2014-0215] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/25/2015] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED : Previous studies have demonstrated that neural stem/progenitor cells (NS/PCs) promote functional recovery in rodent animal models of spinal cord injury (SCI). Because distinct differences exist in the neuroanatomy and immunological responses between rodents and primates, it is critical to determine the effectiveness and safety of allografted embryonic stem cell (ESC)-derived NS/PCs (ESC-NS/PCs) in a nonhuman primate SCI model. In the present study, common marmoset ESC-NS/PCs were grafted into the lesion epicenter 14 days after contusive SCI in adult marmosets (transplantation group). In the control group, phosphate-buffered saline was injected instead of cells. In the presence of a low-dose of tacrolimus, several grafted cells survived without tumorigenicity and differentiated into neurons, astrocytes, or oligodendrocytes. Significant differences were found in the transverse areas of luxol fast blue-positive myelin sheaths, neurofilament-positive axons, corticospinal tract fibers, and platelet endothelial cell adhesion molecule-1-positive vessels at the lesion epicenter between the transplantation and control groups. Immunoelectron microscopic examination demonstrated that the grafted ESC-NS/PC-derived oligodendrocytes contributed to the remyelination of demyelinated axons. In addition, some grafted neurons formed synaptic connections with host cells, and some transplanted neurons were myelinated by host cells. Eventually, motor functional recovery significantly improved in the transplantation group compared with the control group. In addition, a mixed lymphocyte reaction assay indicated that ESC-NS/PCs modulated the allogeneic immune rejection. Taken together, our results indicate that allogeneic transplantation of ESC-NS/PCs from a nonhuman primate promoted functional recovery after SCI without tumorigenicity. SIGNIFICANCE This study demonstrates that allogeneic embryonic stem cell (ESC)-derived neural stem/progenitor cells (NS/PCs) promoted functional recovery after transplantation into the injured spinal cord in nonhuman primates. ESC-NS/PCs were chosen because ESC-NS/PCs are one of the controls for induced pluripotent stem cell-derived NS/PCs and because ESC derivatives are possible candidates for clinical use. This translational research using an allograft model of a nonhuman primate is critical for clinical application of grafting NS/PCs derived from various allogeneic pluripotent stem cells, especially induced pluripotent stem cells, into injured spinal cord at the subacute phase.
Collapse
Affiliation(s)
- Hiroki Iwai
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Hiroko Shimada
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Soraya Nishimura
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Yoshiomi Kobayashi
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Go Itakura
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Keiko Hori
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Keigo Hikishima
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Hayao Ebise
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Naoko Negishi
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Shinsuke Shibata
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Sonoko Habu
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Yoshiaki Toyama
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Masaya Nakamura
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| | - Hideyuki Okano
- Departments of Orthopaedic Surgery and Physiology, Keio University School of Medicine, Tokyo, Japan; Central Institute for Experimental Animals, Kawasaki, Japan; Genomic Science Laboratories, Dainippon Sumitomo Pharma Co., Ltd., Osaka, Japan; Department of Immunology, Juntendo University, Tokyo, Japan
| |
Collapse
|
44
|
Papastefanaki F, Matsas R. From demyelination to remyelination: the road toward therapies for spinal cord injury. Glia 2015; 63:1101-25. [PMID: 25731941 DOI: 10.1002/glia.22809] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/14/2022]
Abstract
Myelin integrity is crucial for central nervous system (CNS) physiology while its preservation and regeneration after spinal cord injury (SCI) is key to functional restoration. Disturbance of nodal organization acutely after SCI exposes the axon and triggers conduction block in the absence of overt demyelination. Oligodendrocyte (OL) loss and myelin degradation follow as a consequence of secondary damage. Here, we provide an overview of the major biological events and underlying mechanisms leading to OL death and demyelination and discuss strategies to restrain these processes. Another aspect which is critical for SCI repair is the enhancement of endogenously occurring spontaneous remyelination. Recent findings have unveiled the complex roles of innate and adaptive immune responses in remyelination and the immunoregulatory potential of the glial scar. Moreover, the intimate crosstalk between neuronal activity, oligodendrogenesis and myelination emphasizes the contribution of rehabilitation to functional recovery. With a view toward clinical applications, several therapeutic strategies have been devised to target SCI pathology, including genetic manipulation, administration of small therapeutic molecules, immunomodulation, manipulation of the glial scar and cell transplantation. The implementation of new tools such as cellular reprogramming for conversion of one somatic cell type to another or the use of nanotechnology and tissue engineering products provides additional opportunities for SCI repair. Given the complexity of the spinal cord tissue after injury, it is becoming apparent that combinatorial strategies are needed to rescue OLs and myelin at early stages after SCI and support remyelination, paving the way toward clinical translation.
Collapse
Affiliation(s)
- Florentia Papastefanaki
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, 11521, Greece
| | | |
Collapse
|
45
|
Nori S, Okada Y, Nishimura S, Sasaki T, Itakura G, Kobayashi Y, Renault-Mihara F, Shimizu A, Koya I, Yoshida R, Kudoh J, Koike M, Uchiyama Y, Ikeda E, Toyama Y, Nakamura M, Okano H. Long-term safety issues of iPSC-based cell therapy in a spinal cord injury model: oncogenic transformation with epithelial-mesenchymal transition. Stem Cell Reports 2015; 4:360-73. [PMID: 25684226 PMCID: PMC4375796 DOI: 10.1016/j.stemcr.2015.01.006] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Previously, we described the safety and therapeutic potential of neurospheres (NSs) derived from a human induced pluripotent stem cell (iPSC) clone, 201B7, in a spinal cord injury (SCI) mouse model. However, several safety issues concerning iPSC-based cell therapy remain unresolved. Here, we investigated another iPSC clone, 253G1, that we established by transducing OCT4, SOX2, and KLF4 into adult human dermal fibroblasts collected from the same donor who provided the 201B7 clone. The grafted 253G1-NSs survived, differentiated into three neural lineages, and promoted functional recovery accompanied by stimulated synapse formation 47 days after transplantation. However, long-term observation (for up to 103 days) revealed deteriorated motor function accompanied by tumor formation. The tumors consisted of Nestin+ undifferentiated neural cells and exhibited activation of the OCT4 transgene. Transcriptome analysis revealed that a heightened mesenchymal transition may have contributed to the progression of tumors derived from grafted cells. Grafted iPSC (253G1)-derived neurospheres formed tumors after long-term observation Activation of the OCT4 transgene is potentially related to tumor formation Tumor progression may have been caused by mesenchymal transition of grafted cells Integration-free iPSCs should be chosen to avoid transgene-induced tumorigenesis
Collapse
Affiliation(s)
- Satoshi Nori
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yohei Okada
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Kanrinmaru Project, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Soraya Nishimura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Takashi Sasaki
- Center for Integrated Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Go Itakura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Yoshiomi Kobayashi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Francois Renault-Mihara
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Atsushi Shimizu
- Department of Molecular Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Ikuko Koya
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Rei Yoshida
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Jun Kudoh
- Laboratory of Gene Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Yasuo Uchiyama
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyoku, Tokyo 113-8421, Japan
| | - Eiji Ikeda
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Department of Pathology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan.
| |
Collapse
|
46
|
Ou Y, Lv CJ, Yu W, Mao ZW, Wan LS, Xu ZK. Fabrication of perforated isoporous membranes via a transfer-free strategy: enabling high-resolution separation of cells. ACS APPLIED MATERIALS & INTERFACES 2014; 6:22400-22407. [PMID: 25421306 DOI: 10.1021/am506419z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Thin perforated membranes with ordered pores are ideal barriers for high-resolution and high-efficiency selective transport and separation of biological species. However, for self-assembled thin membranes with a thickness less than several micrometers, an additional step of transferring the membranes onto porous supports is generally required. In this article, we present a facile transfer-free strategy for fabrication of robust perforated composite membranes via the breath figure process, and for the first time, demonstrate the application of the membranes in high-resolution cell separation of yeasts and lactobacilli without external pressure, achieving almost 100% rejection of yeasts and more than 70% recovery of lactobacilli with excellent viability. The avoidance of the transfer step simplifies the fabrication procedure of composite membranes and greatly improves the membrane homogeneity. Moreover, the introduction of an elastic triblock copolymer increases the interfacial strength between the membrane and the support, and allows the preservation of composite membranes in a dry state. Such perforated ordered membranes can also be applied in other size-based separation systems, enabling new opportunities in bioseparation and biosensors.
Collapse
Affiliation(s)
- Yang Ou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, and ‡State Key Laboratory of Chemical Engineering, Department of Chemical and Biochemical Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Zupanc GKH, Sîrbulescu RF. Cell replacement therapy: lessons from teleost fish. Exp Neurol 2014; 263:272-6. [PMID: 25448008 DOI: 10.1016/j.expneurol.2014.10.006] [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: 05/13/2014] [Revised: 09/06/2014] [Accepted: 10/11/2014] [Indexed: 12/01/2022]
Abstract
Many disorders of the CNS are characterized by a massive loss of neurons. A promising therapeutic strategy to cure such conditions is based on the activation of endogenous stem cells. Implementation of this strategy will benefit from a better understanding of stem cell dynamics and the local CNS microenvironment in regeneration-competent vertebrate model systems. Using a spinal cord injury paradigm in zebrafish larvae, Briona and Dorsky (2014) have provided evidence for the existence of two distinct neural stem cell populations. One population has the characteristics of radial glia and expresses the homeobox transcription factor Dbx. The other lacks Dbx but expresses Olig2. These results are placed in the context of other studies that also support the notion of heterogeneity of adult stem cells in the CNS. The implication that differences among stem cell populations, in combination with specific factors from the local cellular microenvironment, might have a decisive impact on the fate choices of the new cells, is discussed. Reviewed evidence suggests that rather few modifications in the signaling pathways involved in the control of stem cell behavior have led, in the course of evolution, to the pronounced differences between mammals and regeneration-competent organisms. As a consequence, rather minor pharmacological manipulations may be sufficient to reactivate the hidden neurogenic potential of the mammalian CNS, and thus make it available for therapeutic applications.
Collapse
Affiliation(s)
- Günther K H Zupanc
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA 02115, USA.
| | - Ruxandra F Sîrbulescu
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
48
|
Hong JY, Lee SH, Lee SC, Kim JW, Kim KP, Kim SM, Tapia N, Lim KT, Kim J, Ahn HS, Ko K, Shin CY, Lee HT, Schöler HR, Hyun JK, Han DW. Therapeutic potential of induced neural stem cells for spinal cord injury. J Biol Chem 2014; 289:32512-25. [PMID: 25294882 DOI: 10.1074/jbc.m114.588871] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The spinal cord does not spontaneously regenerate, and treatment that ensures functional recovery after spinal cord injury (SCI) is still not available. Recently, fibroblasts have been directly converted into induced neural stem cells (iNSCs) by the forced expression defined transcription factors. Although directly converted iNSCs have been considered to be a cell source for clinical applications, their therapeutic potential has not yet been investigated. Here we show that iNSCs directly converted from mouse fibroblasts enhance the functional recovery of SCI animals. Engrafted iNSCs could differentiate into all neuronal lineages, including different subtypes of mature neurons. Furthermore, iNSC-derived neurons could form synapses with host neurons, thus enhancing the locomotor function recovery. A time course analysis of iNSC-treated SCI animals revealed that engrafted iNSCs effectively reduced the inflammatory response and apoptosis in the injured area. iNSC transplantation also promoted the active regeneration of the endogenous recipient environment in the absence of tumor formation. Therefore, our data suggest that directly converted iNSCs hold therapeutic potential for treatment of SCI and may thus represent a promising cell source for transplantation therapy in patients with SCI.
Collapse
Affiliation(s)
- Jin Young Hong
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | | | | | - Jong-Wan Kim
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | - Kee-Pyo Kim
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | - Natalia Tapia
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | | | - Hong-Sun Ahn
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea
| | - Kinarm Ko
- the Departments of Stem Cell Biology and
| | - Chan Young Shin
- Pharmacology, School of Medicine, and the Konkuk University Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, Gwangjin-gu, Seoul 143701, Republic of Korea
| | | | - Hans R Schöler
- the Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany, the University of Münster, Medical Faculty, 48149 Münster, Germany, and
| | - Jung Keun Hyun
- From the Department of Nanobiomedical Science and BK21PLUS NBM Global Research Center, Dankook University Graduate School, Cheonan 330714, Republic of Korea, the Institute of Tissue Regeneration Engineering, Dankook University, Cheonan 330714, Republic of Korea, the Department of Rehabilitation Medicine, College of Medicine, Dankook University, Cheonan 330714, Republic of Korea
| | - Dong Wook Han
- the Departments of Stem Cell Biology and the Konkuk University Open-Innovation Center, Institute of Biomedical Science & Technology, Konkuk University, Gwangjin-gu, Seoul 143701, Republic of Korea,
| |
Collapse
|
49
|
Kobayakawa K, Kumamaru H, Saiwai H, Kubota K, Ohkawa Y, Kishimoto J, Yokota K, Ideta R, Shiba K, Tozaki-Saitoh H, Inoue K, Iwamoto Y, Okada S. Acute hyperglycemia impairs functional improvement after spinal cord injury in mice and humans. Sci Transl Med 2014; 6:256ra137. [DOI: 10.1126/scitranslmed.3009430] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
50
|
Romanyuk N, Amemori T, Turnovcova K, Prochazka P, Onteniente B, Sykova E, Jendelova P. Beneficial Effect of Human Induced Pluripotent Stem Cell-Derived Neural Precursors in Spinal Cord Injury Repair. Cell Transplant 2014; 24:1781-97. [PMID: 25259685 DOI: 10.3727/096368914x684042] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Despite advances in our understanding and research of induced pluripotent stem cells (iPSCs), their use in clinical practice is still limited due to lack of preclinical experiments. Neural precursors (NPs) derived from a clone of human iPSCs (IMR90) were used to treat a rat spinal cord lesion 1 week after induction. Functional recovery was evaluated using the BBB, beam walking, rotarod, and plantar tests. Lesion morphology, endogenous axonal sprouting, graft survival, and iPSC-NP differentiation were analyzed immunohistochemically. Quantitative polymerase chain reaction (qPCR) was used to evaluate the effect of transplanted iPSC-NPs on endogenous regenerative processes and also to monitor their behavior after transplantation. Human iPSC-NPs robustly survived in the lesion, migrated, and partially filled the lesion cavity during the entire period of observation. Transplanted animals displayed significant motor improvement already from the second week after the transplantation of iPSC-NPs. qPCR revealed the increased expression of human neurotrophins 8 weeks after transplantation. Simultaneously, the white and gray matter were spared in the host tissue. The grafted cells were immunohistochemically positive for doublecortin, MAP2, βIII-tubulin, GFAP, and CNPase 8 weeks after transplantation. Human iPSC-NPs further matured, and 17 weeks after transplantation differentiated toward interneurons, dopaminergic neurons, serotoninergic neurons, and ChAT-positive motoneurons. Human iPSC-NPs possess neurotrophic properties that are associated with significant early functional improvement and the sparing of spinal cord tissue. Their ability to differentiate into tissue-specific neurons leads to the long-term restoration of the lesioned tissue, making the cells a promising candidate for future cell-based therapy of SCI.
Collapse
Affiliation(s)
- Nataliya Romanyuk
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|