1
|
Poulen G, Perrin FE. Advances in spinal cord injury: insights from non-human primates. Neural Regen Res 2024; 19:2354-2364. [PMID: 38526271 PMCID: PMC11090432 DOI: 10.4103/nrr.nrr-d-23-01505] [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: 09/06/2023] [Revised: 11/13/2023] [Accepted: 12/22/2023] [Indexed: 03/26/2024] Open
Abstract
Spinal cord injury results in significant sensorimotor deficits, currently, there is no curative treatment for the symptoms induced by spinal cord injury. Basic and pre-clinical research on spinal cord injury relies on the development and characterization of appropriate animal models. These models should replicate the symptoms observed in human, allowing for the exploration of functional deficits and investigation into various aspects of physiopathology of spinal cord injury. Non-human primates, due to their close phylogenetic association with humans, share more neuroanatomical, genetic, and physiological similarities with humans than rodents. Therefore, the responses to spinal cord injury in nonhuman primates most likely resemble the responses to traumatism in humans. In this review, we will discuss nonhuman primate models of spinal cord injury, focusing on in vivo assessments, including behavioral tests, magnetic resonance imaging, and electrical activity recordings, as well as ex vivo histological analyses. Additionally, we will present therapeutic strategies developed in non-human primates and discuss the unique specificities of non-human primate models of spinal cord injury.
Collapse
Affiliation(s)
- Gaetan Poulen
- University of Montpellier, INSERM, EPHE, Montpellier, France
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Florence E. Perrin
- University of Montpellier, INSERM, EPHE, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
2
|
Marinova D, Ivanov M, Yamashima T, Tonchev A. Quantity, distribution and phenotype of newly generated cells in the intact spinal cord of adult macaque monkeys. Heliyon 2024; 10:e28856. [PMID: 38596108 PMCID: PMC11002253 DOI: 10.1016/j.heliyon.2024.e28856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
The existence of proliferating cells in the intact spinal cord, their distribution and phenotype, are well studied in rodents. A limited number of studies also address the proliferation after spinal cord injury, in non-human primates. However, a detailed description of the quantity, distribution and phenotype of proliferating cells at different anatomical levels of the intact adult non-human primate spinal cord is lacking at present. In the present study, we analyzed normal spinal cord tissues from adult macaque monkeys (Macaca fuscata), infused with Bromo-2'-deoxyuridine (BrdU), and euthanized at 2h, 2 weeks, 5 weeks and 10 weeks after BrdU. We found a significantly higher density of BrdU + cells in the gray matter of cervical segments as compared to thoracic or lumbar segments, and a significantly higher density of proliferating cells in the posterior as compared to the anterior horn of the gray matter. BrdU + cells exhibited phenotype of microglia or endothelial cells (∼50%) or astroglial and oligodendroglial cells (∼40%), including glial progenitor phenotypes marked by the transcription factors Sox9 and Sox10. BrdU + cells also co-expressed other transcription factors known for their involvement in embryonic development, including Emx2, Sox1, Sox2, Ngn1, Olig1, Olig2, Olig3. In the central canal, BrdU + cells were located along the dorso-ventral axis and co-labeled for the markers Vimentin and Nestin. These results reveal the extent of cellular plasticity in the spinal cord of non-human primates under normal conditions.
Collapse
Affiliation(s)
- D. Marinova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Marin Drinov str. 55, Medical University, Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University, Varna, Bulgaria
| | - M.N. Ivanov
- Department of Anatomy and Cell Biology, Faculty of Medicine, Marin Drinov str. 55, Medical University, Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University, Varna, Bulgaria
| | - T. Yamashima
- Departnent of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Takara-machi 13-1, Kanazawa, Japan
| | - A.B. Tonchev
- Department of Anatomy and Cell Biology, Faculty of Medicine, Marin Drinov str. 55, Medical University, Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University, Varna, Bulgaria
| |
Collapse
|
3
|
Liu Y, Luo Z, Xie Y, Sun Y, Yuan F, Jiang L, Lu H, Hu J. Extracellular vesicles from UTX-knockout endothelial cells boost neural stem cell differentiation in spinal cord injury. Cell Commun Signal 2024; 22:155. [PMID: 38424563 PMCID: PMC10903014 DOI: 10.1186/s12964-023-01434-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Vascular endothelial cells are pivotal in the pathophysiological progression following spinal cord injury (SCI). The UTX (Ubiquitously Transcribed Tetratripeptide Repeat on Chromosome X) serves as a significant regulator of endothelial cell phenotype. The manipulation of endogenous neural stem cells (NSCs) offers a compelling strategy for the amelioration of SCI. METHODS Two mouse models were used to investigate SCI: NSCs lineage-traced mice and mice with conditional UTX knockout (UTX KO) in endothelial cells. To study the effects of UTX KO on neural differentiation, we harvested extracellular vesicles (EVs) from both UTX KO spinal cord microvascular endothelial cells (SCMECs) and negative control SCMECs. These EVs were then employed to modulate the differentiation trajectory of endogenous NSCs in the SCI model. RESULTS In our NSCs lineage-traced mice model of SCI, a marked decrease in neurogenesis was observed post-injury. Notably, NSCs in UTX KO SCMECs mice showed enhanced neuronal differentiation compared to controls. RNA sequencing and western blot analyses revealed an upregulation of L1 cell adhesion molecule (L1CAM), a gene associated with neurogenesis, in UTX KO SCMECs and their secreted EVs. This aligns with the observed promotion of neurogenesis in UTX KO conditions. In vivo administration of L1CAM-rich EVs from UTX KO SCMECs (KO EVs) to the mice significantly enhanced neural differentiation. Similarly, in vitro exposure of NSCs to KO EVs resulted in increased activation of the Akt signaling pathway, further promoting neural differentiation. Conversely, inhibiting Akt phosphorylation or knocking down L1CAM negated the beneficial effects of KO EVs on NSC neuronal differentiation. CONCLUSIONS In conclusion, our findings substantiate that EVs derived from UTX KO SCMECs can act as facilitators of neural differentiation following SCI. This study not only elucidates a novel mechanism but also opens new horizons for therapeutic interventions in the treatment of SCI. Video Abstract.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixiang Luo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Xie
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Sun
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Feifei Yuan
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyuan Jiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
4
|
Pukos N, Marion CM, Arnold WD, Noble BT, Popovich PG, McTigue DM. Chronic demyelination and myelin repair after spinal cord injury in mice: A potential link for glutamatergic axon activity. Glia 2023; 71:2096-2116. [PMID: 37208933 PMCID: PMC10330449 DOI: 10.1002/glia.24382] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/08/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023]
Abstract
Our prior work examining endogenous repair after spinal cord injury (SCI) in mice revealed that large numbers of new oligodendrocytes (OLs) are generated in the injured spinal cord, with peak oligodendrogenesis between 4 and 7 weeks post-injury (wpi). We also detected new myelin formation over 2 months post-injury (mpi). Our current work significantly extends these results, including quantification of new myelin through 6 mpi and concomitant examination of indices of demyelination. We also examined electrophysiological changes during peak oligogenesis and a potential mechanism driving OL progenitor cell (OPC) contact with axons. Results reveal peak in remyelination occurs during the 3rd mpi, and that myelin generation continues for at least 6 mpi. Further, motor evoked potentials significantly increased during peak remyelination, suggesting enhanced axon potential conduction. Interestingly, two indices of demyelination, nodal protein spreading and Nav1.2 upregulation, were also present chronically after SCI. Nav1.2 was expressed through 10 wpi and nodal protein disorganization was detectable throughout 6 mpi suggesting chronic demyelination, which was confirmed with EM. Thus, demyelination may continue chronically, which could trigger the long-term remyelination response. To examine a potential mechanism that may initiate post-injury myelination, we show that OPC processes contact glutamatergic axons in the injured spinal cord in an activity-dependent manner. Notably, these OPC/axon contacts were increased 2-fold when axons were activated chemogenetically, revealing a potential therapeutic target to enhance post-SCI myelin repair. Collectively, results show the surprisingly dynamic nature of the injured spinal cord over time and that the tissue may be amenable to treatments targeting chronic demyelination.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
| | - Christina M Marion
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - W David Arnold
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
- Division of Neuromuscular Disorders, Department of Neurology, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
- Department of Physical Medicine and Rehabilitation, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Benjamin T Noble
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
| | - Phillip G Popovich
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| | - Dana M McTigue
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio, USA
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
5
|
Hu X, Xu W, Ren Y, Wang Z, He X, Huang R, Ma B, Zhao J, Zhu R, Cheng L. Spinal cord injury: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:245. [PMID: 37357239 DOI: 10.1038/s41392-023-01477-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/27/2023] Open
Abstract
Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate. The challenges of SCI repair include its complex pathological mechanisms and the difficulties of neural regeneration in the central nervous system. In the past few decades, researchers have attempted to completely elucidate the pathological mechanism of SCI and identify effective strategies to promote axon regeneration and neural circuit remodeling, but the results have not been ideal. Recently, new pathological mechanisms of SCI, especially the interactions between immune and neural cell responses, have been revealed by single-cell sequencing and spatial transcriptome analysis. With the development of bioactive materials and stem cells, more attention has been focused on forming intermediate neural networks to promote neural regeneration and neural circuit reconstruction than on promoting axonal regeneration in the corticospinal tract. Furthermore, technologies to control physical parameters such as electricity, magnetism and ultrasound have been constantly innovated and applied in neural cell fate regulation. Among these advanced novel strategies and technologies, stem cell therapy, biomaterial transplantation, and electromagnetic stimulation have entered into the stage of clinical trials, and some of them have already been applied in clinical treatment. In this review, we outline the overall epidemiology and pathophysiology of SCI, expound on the latest research progress related to neural regeneration and circuit reconstruction in detail, and propose future directions for SCI repair and clinical applications.
Collapse
Affiliation(s)
- Xiao Hu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Wei Xu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Yilong Ren
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Zhaojie Wang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Xiaolie He
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Runzhi Huang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Bei Ma
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Jingwei Zhao
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Rongrong Zhu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| |
Collapse
|
6
|
Gupta S, Dutta S, Hui SP. Regenerative Potential of Injured Spinal Cord in the Light of Epigenetic Regulation and Modulation. Cells 2023; 12:1694. [PMID: 37443728 PMCID: PMC10341208 DOI: 10.3390/cells12131694] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
A spinal cord injury is a form of physical harm imposed on the spinal cord that causes disability and, in many cases, leads to permanent mammalian paralysis, which causes a disastrous global issue. Because of its non-regenerative aspect, restoring the spinal cord's role remains one of the most daunting tasks. By comparison, the remarkable regenerative ability of some regeneration-competent species, such as some Urodeles (Axolotl), Xenopus, and some teleost fishes, enables maximum functional recovery, even after complete spinal cord transection. During the last two decades of intensive research, significant progress has been made in understanding both regenerative cells' origins and the molecular signaling mechanisms underlying the regeneration and reconstruction of damaged spinal cords in regenerating organisms and mammals, respectively. Epigenetic control has gradually moved into the center stage of this research field, which has been helped by comprehensive work demonstrating that DNA methylation, histone modifications, and microRNAs are important for the regeneration of the spinal cord. In this review, we concentrate primarily on providing a comparison of the epigenetic mechanisms in spinal cord injuries between non-regenerating and regenerating species. In addition, we further discuss the epigenetic mediators that underlie the development of a regeneration-permissive environment following injury in regeneration-competent animals and how such mediators may be implicated in optimizing treatment outcomes for spinal cord injurie in higher-order mammals. Finally, we briefly discuss the role of extracellular vesicles (EVs) in the context of spinal cord injury and their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Samudra Gupta
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
| | - Subhra Prakash Hui
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India;
| |
Collapse
|
7
|
Yu H, Yang S, Li H, Wu R, Lai B, Zheng Q. Activating Endogenous Neurogenesis for Spinal Cord Injury Repair: Recent Advances and Future Prospects. Neurospine 2023; 20:164-180. [PMID: 37016865 PMCID: PMC10080446 DOI: 10.14245/ns.2245184.296] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/29/2022] [Indexed: 04/03/2023] Open
Abstract
After spinal cord injury (SCI), endogenous neural stem cells are activated and migrate to the injury site where they differentiate into astrocytes, but they rarely differentiate into neurons. It is difficult for brain-derived information to be transmitted through the injury site after SCI because of the lack of neurons that can relay neural information through the injury site, and the functional recovery of adult mammals is difficult to achieve. The development of bioactive materials, tissue engineering, stem cell therapy, and physiotherapy has provided new strategies for the treatment of SCI and shown broad application prospects, such as promoting endogenous neurogenesis after SCI. In this review, we focus on novel approaches including tissue engineering, stem cell technology, and physiotherapy to promote endogenous neurogenesis and their therapeutic effects on SCI. Moreover, we explore the mechanisms and challenges of endogenous neurogenesis for the repair of SCI.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shangbin Yang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haotao Li
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Rongjie Wu
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Biqin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Co-corresponding Author Biqin Lai Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Southern Medical University, Guangzhou, China
- Corresponding Author Qiujian Zheng Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Lazic A, Balint V, Stanisavljevic Ninkovic D, Peric M, Stevanovic M. Reactive and Senescent Astroglial Phenotypes as Hallmarks of Brain Pathologies. Int J Mol Sci 2022; 23:ijms23094995. [PMID: 35563385 PMCID: PMC9100382 DOI: 10.3390/ijms23094995] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes, as the most abundant glial cells in the central nervous system, are tightly integrated into neural networks and participate in numerous aspects of brain physiology and pathology. They are the main homeostatic cells in the central nervous system, and the loss of astrocyte physiological functions and/or gain of pro-inflammatory functions, due to their reactivation or cellular senescence, can have profound impacts on the surrounding microenvironment with pathological outcomes. Although the importance of astrocytes is generally recognized, and both senescence and reactive astrogliosis have been extensively reviewed independently, there are only a few comparative overviews of these complex processes. In this review, we summarize the latest data regarding astrocyte reactivation and senescence, and outline similarities and differences between these phenotypes from morphological, functional, and molecular points of view. A special focus has been given to neurodegenerative diseases, where these phenotypic alternations of astrocytes are significantly implicated. We also summarize current perspectives regarding new advances in model systems based on astrocytes as well as data pointing to these glial cells as potential therapeutic targets.
Collapse
Affiliation(s)
- Andrijana Lazic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Correspondence:
| | - Vanda Balint
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Danijela Stanisavljevic Ninkovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Mina Peric
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
| | - Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (V.B.); (D.S.N.); (M.P.); (M.S.)
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11001 Belgrade, Serbia
| |
Collapse
|
9
|
Havelikova K, Smejkalova B, Jendelova P. Neurogenesis as a Tool for Spinal Cord Injury. Int J Mol Sci 2022; 23:ijms23073728. [PMID: 35409088 PMCID: PMC8998995 DOI: 10.3390/ijms23073728] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Spinal cord injury is a devastating medical condition with no effective treatment. One approach to SCI treatment may be provided by stem cells (SCs). Studies have mainly focused on the transplantation of exogenous SCs, but the induction of endogenous SCs has also been considered as an alternative. While the differentiation potential of neural stem cells in the brain neurogenic regions has been known for decades, there are ongoing debates regarding the multipotent differentiation potential of the ependymal cells of the central canal in the spinal cord (SCECs). Following spinal cord insult, SCECs start to proliferate and differentiate mostly into astrocytes and partly into oligodendrocytes, but not into neurons. However, there are several approaches concerning how to increase neurogenesis in the injured spinal cord, which are discussed in this review. The potential treatment approaches include drug administration, the reduction of neuroinflammation, neuromodulation with physical factors and in vivo reprogramming.
Collapse
Affiliation(s)
- Katerina Havelikova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; (K.H.); (B.S.)
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Barbora Smejkalova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; (K.H.); (B.S.)
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Pavla Jendelova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; (K.H.); (B.S.)
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
- Correspondence: ; Tel.: +420-24-106-2828
| |
Collapse
|
10
|
Assunção Silva RC, Pinto L, Salgado AJ. Cell transplantation and secretome based approaches in spinal cord injury regenerative medicine. Med Res Rev 2021; 42:850-896. [PMID: 34783046 DOI: 10.1002/med.21865] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 07/12/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
The axonal growth-restrictive character of traumatic spinal cord injury (SCI) makes finding a therapeutic strategy a very demanding task, due to the postinjury events impeditive to spontaneous axonal outgrowth and regeneration. Considering SCI pathophysiology complexity, it has been suggested that an effective therapy should tackle all the SCI-related aspects and provide sensory and motor improvement to SCI patients. Thus, the current aim of any therapeutic approach for SCI relies in providing neuroprotection and support neuroregeneration. Acknowledging the current SCI treatment paradigm, cell transplantation is one of the most explored approaches for SCI with mesenchymal stem cells (MSCs) being in the forefront of many of these. Studies showing the beneficial effects of MSC transplantation after SCI have been proposing a paracrine action of these cells on the injured tissues, through the secretion of protective and trophic factors, rather than attributing it to the action of cells itself. This manuscript provides detailed information on the most recent data regarding the neuroregenerative effect of the secretome of MSCs as a cell-free based therapy for SCI. The main challenge of any strategy proposed for SCI treatment relies in obtaining robust preclinical evidence from in vitro and in vivo models, before moving to the clinics, so we have specifically focused on the available vertebrate and mammal models of SCI currently used in research and how can SCI field benefit from them.
Collapse
Affiliation(s)
- Rita C Assunção Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal.,BnML, Behavioral and Molecular Lab, Braga, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal.,ICVS/3B's e PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
11
|
Kwiecien JM, Dąbrowski W, Yaron JR, Zhang L, Delaney KH, Lucas AR. The Role of Astrogliosis in Formation of the Syrinx in Spinal Cord Injury. Curr Neuropharmacol 2021; 19:294-303. [PMID: 32691715 PMCID: PMC8033977 DOI: 10.2174/1570159x18666200720225222] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/28/2022] Open
Abstract
A massive localized trauma to the spinal cord results in complex pathologic events driven by necrosis and vascular damage which in turn leads to hemorrhage and edema. Severe, destructive and very protracted inflammatory response is characterized by infiltration by phagocytic macrophages of a site of injury which is converted into a cavity of injury (COI) surrounded by astroglial reaction mounted by the spinal cord. The tissue response to the spinal cord injury (SCI) has been poorly understood but the final outcome appears to be a mature syrinx filled with the cerebrospinal fluid with related neural tissue loss and permanent neurologic deficits. This paper reviews known pathologic mechanisms involved in the formation of the COI after SCI and discusses the integrative role of reactive astrogliosis in mechanisms involved in the removal of edema after the injury. A large proportion of edema fluid originating from the trauma and then from vasogenic edema related to persistent severe inflammation, may be moved into the COI in an active process involving astrogliosis and specifically over-expressed aquaporins.
Collapse
Affiliation(s)
- Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Wojciech Dąbrowski
- Department of Anaesthesiology and Intensive Therapy, Medical University of Lublin, ul. Jaczewskiego 8, Lublin 20-090 Poland
| | - Jordan R Yaron
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ, U.S.A
| | - Liqiang Zhang
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ, U.S.A
| | - Kathleen H. Delaney
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ, U.S.A
| |
Collapse
|
12
|
Bal E, Hanalioglu S, Kopru CZ, Köse S, Basak AT, Pehlivan SB, Cetinkaya DU, Purali N, Korkusuz P, Bozkurt G. Effect of mesenchymal stem cells therapy in experimental kaolin induced syringomyelia model. J Neurosurg Sci 2020; 66:40-48. [PMID: 33056946 DOI: 10.23736/s0390-5616.20.05026-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Syringomyelia is a pathological cavitation of the spinal cord. In this study, we examined whether a syrinx cavity would limit itself with axonal regeneration and stem cell activity in the cavity, and we evaluated subjects on a functional basis. METHODS Groups were designated as kaolin, trauma, kaolin-trauma, and saline groups. Also divided out of the syringomyelia treated groups were those given human mesenchymal stem cells (hMSCs). All groups were evaluated with immunohistochemistry, electron microscopy, confocal microscopy and functionally. RESULTS The kaolin-trauma group had a significant correction of BBB score with hMSCs therapy. The syrinx cavity measurements showed significant improvement in groups treated with hMSCs. The tissue surrounding the syrinx cavity, however, appeared to be better organized in groups treated with hMSCs. The process of repair and regeneration of damaged axons in the lesion were more improved in groups treated with hMSCs. Using confocal microscopy, fluorescence of hMSCs was observed in the central canal, in the ependymal tissue, and around the lesion. CONCLUSIONS It was concluded that axonal repair accelerated in groups receiving stem cells, and thus, stem cells may be effective in recovery of neural tissue and myelin damage in syringomyelia.
Collapse
Affiliation(s)
- Ercan Bal
- Department of Neurosurgery, Ankara Yıldırım Beyazıt University School of Medicine, Ankara, Turkey -
| | - Sahin Hanalioglu
- Department of Neurosurgery, Hacettepe University School of Hospital, Ankara, Turkey
| | - Cagla Z Kopru
- Department of Histology and Embryology, Yüksek İhtisas University School of Medicine, Ankara, Turkey
| | - Sevil Köse
- Department of Medical Biology, Atilim University Faculty of Medicine, Ankara, Turkey
| | - Ahmet T Basak
- Department of Neurosurgery, Medipol Mega Hospital University of School, İstanbul, Turkey
| | - Sibel B Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Duygu U Cetinkaya
- Center for Stem Cell Research and Development (PEDI-STEM), Hacettepe University School of Medicine, Ankara Turkey
| | - Nuhan Purali
- Department of Biophysics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Gökhan Bozkurt
- Department of Neurosurgery, Memorial Hospital, İstanbul, Turkey
| |
Collapse
|
13
|
Luzzi S, Crovace AM, Del Maestro M, Giotta Lucifero A, Elbabaa SK, Cinque B, Palumbo P, Lombardi F, Cimini A, Cifone MG, Crovace A, Galzio R. The cell-based approach in neurosurgery: ongoing trends and future perspectives. Heliyon 2019; 5:e02818. [PMID: 31844735 PMCID: PMC6889232 DOI: 10.1016/j.heliyon.2019.e02818] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/11/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Examination of the current trends and future perspectives of the cell-based therapies in neurosurgery. METHODS A PubMed/MEDLINE-based systematic review has been performed combining the main Medical Subject Headings (MeSH) regarding the cell- and tissue-based therapies with the "Brain", "Spinal Cord", "Spine" and "Skull" MeSH terms. Only articles in English published in the last 10 years and pertinent to neurosurgery have been selected. RESULTS A total of 1,173 relevant articles have been chosen. Somatic cells and gene-modification technologies have undergone the greatest development. Immunotherapies and gene therapies have been tested for the cure of glioblastoma, stem cells mainly for brain and spinal cord traumatic injuries. Stem cells have also found a rationale in the treatment of the cranial and spinal bony defects, and of the intervertebral disc degeneration, as well.Most of the completed or ongoing trials concerning the cell-based therapies in neurosurgery are on phase 2. Future perspectives involve the need to overcome issues related to immunogenicity, oncogenicity and routes for administration. Refinement and improvement of vector design and delivery are required within the gene therapies. CONCLUSION The last decade has been characterised by a progressive evolution of neurosurgery from a purely mechanical phase to a new biological one. This trend has followed the rapid and parallel development of translational medicine and nanotechnologies.The introduction of new technologies, the optimisation of the already existing ones, and the reduction of costs are among the main challenges of the foreseeable future.
Collapse
Affiliation(s)
- Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| | - Alberto Maria Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Mattia Del Maestro
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
- PhD School in Experimental Medicine, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Samer K. Elbabaa
- Pediatric Neurosurgery, Pediatric Neuroscience Center of Excellence, Arnold Palmer Hospital for Children, 1222 S. Orange Avenue, 2nd Floor, MP 154, Orlando, FL, 32806, USA
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Annamaria Cimini
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Renato Galzio
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| |
Collapse
|
14
|
Pukos N, Goodus MT, Sahinkaya FR, McTigue DM. Myelin status and oligodendrocyte lineage cells over time after spinal cord injury: What do we know and what still needs to be unwrapped? Glia 2019; 67:2178-2202. [PMID: 31444938 DOI: 10.1002/glia.23702] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) affects over 17,000 individuals in the United States per year, resulting in sudden motor, sensory and autonomic impairments below the level of injury. These deficits may be due at least in part to the loss of oligodendrocytes and demyelination of spared axons as it leads to slowed or blocked conduction through the lesion site. It has long been accepted that progenitor cells form new oligodendrocytes after SCI, resulting in the acute formation of new myelin on demyelinated axons. However, the chronicity of demyelination and the functional significance of remyelination remain contentious. Here we review work examining demyelination and remyelination after SCI as well as the current understanding of oligodendrocyte lineage cell responses to spinal trauma, including the surprisingly long-lasting response of NG2+ oligodendrocyte progenitor cells (OPCs) to proliferate and differentiate into new myelinating oligodendrocytes for months after SCI. OPCs are highly sensitive to microenvironmental changes, and therefore respond to the ever-changing post-SCI milieu, including influx of blood, monocytes and neutrophils; activation of microglia and macrophages; changes in cytokines, chemokines and growth factors such as ciliary neurotrophic factor and fibroblast growth factor-2; glutamate excitotoxicity; and axon degeneration and sprouting. We discuss how these changes relate to spontaneous oligodendrogenesis and remyelination, the evidence for and against demyelination being an important clinical problem and if remyelination contributes to motor recovery.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio.,Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio
| | - Matthew T Goodus
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| | - Fatma R Sahinkaya
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio
| | - Dana M McTigue
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| |
Collapse
|
15
|
Liu S, Chen Z. Employing Endogenous NSCs to Promote Recovery of Spinal Cord Injury. Stem Cells Int 2019; 2019:1958631. [PMID: 31191666 PMCID: PMC6525819 DOI: 10.1155/2019/1958631] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/07/2019] [Indexed: 12/15/2022] Open
Abstract
Endogenous neural stem cells (NSCs) exist in the central canal of mammalian spinal cords. Under normal conditions, these NSCs remain quiescent and express FoxJ1. After spinal cord injury (SCI), the endogenous NSCs of a heterogeneous nature are activated and proliferate and migrate towards the lesion site and mainly differentiate into astrocytes to repair the injured tissue. In vitro, spinal cord NSCs are multipotent and can differentiate into neurons, astrocytes, and oligodendrocytes. The altered microenvironments after SCI play key roles on the fate determination of activated NSCs, especially on the neuronal specification potential. Studies show that the activated spinal cord NSCs can generate interneurons when transplanted into the adult hippocampus. In addition, the spinal cord NSCs exhibit low immunogenicity in a transplantation context, thus implicating a promising therapeutic potential on SCI recovery. Here, we summarize the characteristics of spinal cord NSCs, especially their properties after injury. With a better understanding of endogenous NSCs under normal and SCI conditions, we may be able to employ endogenous NSCs for SCI repair in the future.
Collapse
Affiliation(s)
- Sumei Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
16
|
Zupanc GK. Stem‐Cell‐Driven Growth and Regrowth of the Adult Spinal Cord in Teleost Fish. Dev Neurobiol 2019; 79:406-423. [DOI: 10.1002/dneu.22672] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Günther K.H. Zupanc
- Laboratory of Neurobiology, Department of Biology Northeastern University Boston Massachusetts
| |
Collapse
|
17
|
Oligodendrogliogenesis and Axon Remyelination after Traumatic Spinal Cord Injuries in Animal Studies: A Systematic Review. Neuroscience 2019; 402:37-50. [PMID: 30685542 DOI: 10.1016/j.neuroscience.2019.01.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
Extensive oligodendrocyte death after acute traumatic spinal cord injuries (TSCI) leads to axon demyelination and subsequently may leave axons vulnerable to degeneration. Despite the present evidence showing spontaneous remyelination after TSCI the cellular origin of new myelin and the time course of the axon ensheathment/remyelination remained controversial issue. In this systematic review the trend of oligodendrocyte death after injury as well as the extent and the cellular origin of oligodendrogliogenesis were comprehensively evaluated. The study design was based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA)-guided systematic review. PubMed and EMBASE were searched with no temporal or linguistic restrictions. Also, hand-search was performed in the bibliographies of relevant articles. Non-interventional animal studies discussing different types of myelinating cells including oligodendrocytes, Schwann cells and oligodendrocyte progenitor cells (OPCs) were evaluated. The extent of oligodendrocyte death, oligodendrocyte differentiation and remyelination were the pathophysiological outcome measures. We found 12,359 studies, 34 of which met the inclusion criteria. The cumulative evidence shows extensive oligodendrocytes cell death during the first week post-injury (pi). OPCs and peripheral invading Schwann cells are the dominant cells contributing in myelin formation. The maximum OPC proliferation was observed at around 2 weeks pi and oligodendrogliogenesis continues at later stages until the number of oligodendrocytes return to normal tissue by one month pi. Taken together, the evidence in animals reveals the potential role for endogenous myelinating cells in the axon ensheathment/remyelination after TSCI and this can be the target of pharmacotherapy to induce oligodendrocyte differentiation and myelin formation post-injury.
Collapse
|
18
|
Yang Q, Zhou J. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2018; 67:1017-1035. [DOI: 10.1002/glia.23571] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Qiao‐qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
| | - Jia‐wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Shanghai 200031 China
| |
Collapse
|
19
|
Guest JD, Moore SW, Aimetti AA, Kutikov AB, Santamaria AJ, Hofstetter CP, Ropper AE, Theodore N, Ulich TR, Layer RT. Internal decompression of the acutely contused spinal cord: Differential effects of irrigation only versus biodegradable scaffold implantation. Biomaterials 2018; 185:284-300. [DOI: 10.1016/j.biomaterials.2018.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/04/2018] [Accepted: 09/16/2018] [Indexed: 12/13/2022]
|
20
|
Becker CG, Becker T, Hugnot JP. The spinal ependymal zone as a source of endogenous repair cells across vertebrates. Prog Neurobiol 2018; 170:67-80. [DOI: 10.1016/j.pneurobio.2018.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
|
21
|
Nagoshi N, Khazaei M, Ahlfors JE, Ahuja CS, Nori S, Wang J, Shibata S, Fehlings MG. Human Spinal Oligodendrogenic Neural Progenitor Cells Promote Functional Recovery After Spinal Cord Injury by Axonal Remyelination and Tissue Sparing. Stem Cells Transl Med 2018; 7:806-818. [PMID: 30085415 PMCID: PMC6216444 DOI: 10.1002/sctm.17-0269] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/01/2018] [Accepted: 06/05/2018] [Indexed: 12/22/2022] Open
Abstract
Cell transplantation therapy utilizing neural precursor cells (NPCs) is a conceptually attractive strategy for traumatic spinal cord injury (SCI) to replace lost cells, remyelinate denuded host axons and promote tissue sparing. However, the number of mature oligodendrocytes that differentiate from typical NPCs remains limited. Herein, we describe a novel approach to bias the differentiation of directly reprogrammed human NPCs (drNPCs) toward a more oligodendrogenic fate (oNPCs) while preserving their tripotency. The oNPCs derived from different lines of human NPCs showed similar characteristics in vitro. To assess the in vivo efficacy of this approach, we used oNPCs derived from drNPCs and transplanted them into a SCI model in immunodeficient Rowett Nude (RNU) rats. The transplanted cells showed significant migration along the rostrocaudal axis and proportionally greater differentiation into oligodendrocytes. These cells promoted perilesional tissue sparing and axonal remyelination, which resulted in recovery of motor function. Moreover, after transplantation of the oNPCs into intact spinal cords of immunodeficient NOD/SCID mice, we detected no evidence of tumor formation even after 5 months of observation. Thus, biasing drNPC differentiation along an oligodendroglial lineage represents a promising approach to promote tissue sparing, axonal remyelination, and neural repair after traumatic SCI. Stem Cells Translational Medicine 2018;7:806-818.
Collapse
Affiliation(s)
- Narihito Nagoshi
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | - Christopher S Ahuja
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery and Spine Program, University of Toronto, Toronto, Ontario, Canada
| | - Satoshi Nori
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Jian Wang
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery and Spine Program, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Merten N, Fischer J, Simon K, Zhang L, Schröder R, Peters L, Letombe AG, Hennen S, Schrage R, Bödefeld T, Vermeiren C, Gillard M, Mohr K, Lu QR, Brüstle O, Gomeza J, Kostenis E. Repurposing HAMI3379 to Block GPR17 and Promote Rodent and Human Oligodendrocyte Differentiation. Cell Chem Biol 2018; 25:775-786.e5. [PMID: 29706593 DOI: 10.1016/j.chembiol.2018.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/11/2018] [Accepted: 03/23/2018] [Indexed: 12/22/2022]
Abstract
Identification of additional uses for existing drugs is a hot topic in drug discovery and a viable alternative to de novo drug development. HAMI3379 is known as an antagonist of the cysteinyl-leukotriene CysLT2 receptor, and was initially developed to treat cardiovascular and inflammatory disorders. In our study we identified HAMI3379 as an antagonist of the orphan G protein-coupled receptor GPR17. HAMI3379 inhibits signaling of recombinant human, rat, and mouse GPR17 across various cellular backgrounds, and of endogenous GPR17 in primary rodent oligodendrocytes. GPR17 blockade by HAMI3379 enhanced maturation of primary rat and mouse oligodendrocytes, but was without effect in oligodendrocytes from GPR17 knockout mice. In human oligodendrocytes prepared from inducible pluripotent stem cells, GPR17 is expressed and its activation impaired oligodendrocyte differentiation. HAMI3379, conversely, efficiently favored human oligodendrocyte differentiation. We propose that HAMI3379 holds promise for pharmacological exploitation of orphan GPR17 to enhance regenerative strategies for the promotion of remyelination in patients.
Collapse
Affiliation(s)
- Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Julia Fischer
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, 53105 Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Liguo Zhang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Ralf Schröder
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Lucas Peters
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | | | - Stephanie Hennen
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Ramona Schrage
- UCB Biopharma, CNS Research, 1420 Braine-l'Alleud, Belgium
| | - Theresa Bödefeld
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, 53115 Bonn, Germany
| | | | - Michel Gillard
- UCB Biopharma, CNS Research, 1420 Braine-l'Alleud, Belgium
| | - Klaus Mohr
- Pharmacology and Toxicology Section, Institute of Pharmacy, University of Bonn, 53115 Bonn, Germany
| | - Qing Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE&BRAIN Center, University of Bonn, 53105 Bonn, Germany
| | - Jesus Gomeza
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
23
|
The p53 Pathway Controls SOX2-Mediated Reprogramming in the Adult Mouse Spinal Cord. Cell Rep 2017; 17:891-903. [PMID: 27732862 DOI: 10.1016/j.celrep.2016.09.038] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/06/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
Although the adult mammalian spinal cord lacks intrinsic neurogenic capacity, glial cells can be reprogrammed in vivo to generate neurons after spinal cord injury (SCI). How this reprogramming process is molecularly regulated, however, is not clear. Through a series of in vivo screens, we show here that the p53-dependent pathway constitutes a critical checkpoint for SOX2-mediated reprogramming of resident glial cells in the adult mouse spinal cord. While it has no effect on the reprogramming efficiency, the p53 pathway promotes cell-cycle exit of SOX2-induced adult neuroblasts (iANBs). As such, silencing of either p53 or p21 markedly boosts the overall production of iANBs. A neurotrophic milieu supported by BDNF and NOG can robustly enhance maturation of these iANBs into diverse but predominantly glutamatergic neurons. Together, these findings have uncovered critical molecular and cellular checkpoints that may be manipulated to boost neuron regeneration after SCI.
Collapse
|
24
|
Epigenetic regulation of neural stem cell differentiation towards spinal cord regeneration. Cell Tissue Res 2017; 371:189-199. [PMID: 28695279 DOI: 10.1007/s00441-017-2656-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/31/2017] [Indexed: 12/20/2022]
Abstract
Severe spinal cord injury (SCI) leads to almost complete neural cell loss at the injured site, causing the irreversible disruption of neuronal circuits. The transplantation of neural stem or precursor cells (NS/PCs) has been regarded as potentially effective for SCI treatment because NS/PCs can compensate for the injured sites by differentiating into neurons and glial cells (astrocytes and oligodendrocytes). An understanding of the molecular mechanisms that regulate the proliferation, fate specification and maturation of NS/PCs and their progeny would facilitate the establishment of better therapeutic strategies for regeneration after SCI. In recent years, several studies of SCI animal models have demonstrated that the modulation of specific epigenetic marks by histone modifiers and non-coding RNAs directs the setting of favorable cellular environments that promote the neuronal differentiation of NS/PCs and/or the elongation of the axons of the surviving neurons at the injured sites. In this review, we provide an overview of recent progress in the epigenetic regulation/manipulation of neural cells for the treatment of SCI.
Collapse
|
25
|
Nardone R, Florea C, Höller Y, Brigo F, Versace V, Lochner P, Golaszewski S, Trinka E. Rodent, large animal and non-human primate models of spinal cord injury. ZOOLOGY 2017; 123:101-114. [PMID: 28720322 DOI: 10.1016/j.zool.2017.06.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 01/05/2023]
Abstract
In this narrative review we aimed to assess the usefulness of the different animal models in identifying injury mechanisms and developing therapies for humans suffering from spinal cord injury (SCI). Results obtained from rodent studies are useful but, due to the anatomical, molecular and functional differences, confirmation of these findings in large animals or non-human primates may lead to basic discoveries that cannot be made in rodent models and that are more useful for developing treatment strategies in humans. SCI in dogs can be considered as intermediate between rodent models and human clinical trials, but the primate models could help to develop appropriate methods that might be more relevant to humans. Ideally, an animal model should meet the requirements of availability and repeatability as well as reproduce the anatomical features and the clinical pathological changing process of SCI. An animal model that completely simulates SCI in humans does not exist. The different experimental models of SCI have advantages and disadvantages for investigating the different aspects of lesion development, recovery mechanisms and potential therapeutic interventions. The potential advantages of non-human primate models include genetic similarities, similar caliber/length of the spinal cord as well as biological and physiological responses to injury which are more similar to humans. Among the potential disadvantages, high operating costs, infrastructural requirements and ethical concerns should be considered. The translation from experimental repair strategies to clinical applications needs to be investigated in future carefully designed studies.
Collapse
Affiliation(s)
- Raffaele Nardone
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria; Department of Neurology, Franz Tappeiner Hospital, Via Rossini 5, I-39012, Merano, Italy; Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria.
| | - Cristina Florea
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Yvonne Höller
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Francesco Brigo
- Department of Neurology, Franz Tappeiner Hospital, Via Rossini 5, I-39012, Merano, Italy; Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, Piazzale L.A. Scuro, I-37134 Verona, Italy
| | - Viviana Versace
- Department of Neurorehabilitation, Hospital of Vipiteno, Via Santa Margherita 24, I-39049, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, Kirrberger-Str. 100, D-66421 Homburg, Germany
| | - Stefan Golaszewski
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Klinik, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Ignaz-Harrer-Str. 79, A-5020, Salzburg, Austria
| |
Collapse
|
26
|
Zhao Y, Xiao Z, Chen B, Dai J. The neuronal differentiation microenvironment is essential for spinal cord injury repair. Organogenesis 2017; 13:63-70. [PMID: 28598297 DOI: 10.1080/15476278.2017.1329789] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Spinal cord injury (SCI) often leads to substantial disability due to loss of motor function and sensation below the lesion. Neural stem cells (NSCs) are a promising strategy for SCI repair. However, NSCs rarely differentiate into neurons; they mostly differentiate into astrocytes because of the adverse microenvironment present after SCI. We have shown that myelin-associated inhibitors (MAIs) inhibited neuronal differentiation of NSCs. Given that MAIs activate epidermal growth factor receptor (EGFR) signaling, we used a collagen scaffold-tethered anti-EGFR antibody to attenuate the inhibitory effects of MAIs and create a neuronal differentiation microenvironment for SCI repair. The collagen scaffold modified with anti-EGFR antibody prevented the inhibition of NSC neuronal differentiation by myelin. After transplantation into completely transected SCI animals, the scaffold-linked antibodies induced production of nascent neurons from endogenous and transplanted NSCs, which rebuilt the neuronal relay by forming connections with each other or host neurons to transmit electrophysiological signals and promote functional recovery. Thus, a scaffold-based strategy for rebuilding the neuronal differentiation microenvironment could be useful for SCI repair.
Collapse
Affiliation(s)
- Yannan Zhao
- a State Key Laboratory of Molecular Developmental Biology , Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Zhifeng Xiao
- a State Key Laboratory of Molecular Developmental Biology , Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Bing Chen
- a State Key Laboratory of Molecular Developmental Biology , Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Jianwu Dai
- a State Key Laboratory of Molecular Developmental Biology , Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| |
Collapse
|
27
|
Führmann T, Anandakumaran PN, Shoichet MS. Combinatorial Therapies After Spinal Cord Injury: How Can Biomaterials Help? Adv Healthc Mater 2017; 6. [PMID: 28247563 DOI: 10.1002/adhm.201601130] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/05/2016] [Indexed: 12/31/2022]
Abstract
Traumatic spinal cord injury (SCI) results in an immediate loss of motor and sensory function below the injury site and is associated with a poor prognosis. The inhibitory environment that develops in response to the injury is mainly due to local expression of inhibitory factors, scarring and the formation of cystic cavitations, all of which limit the regenerative capacity of endogenous or transplanted cells. Strategies that demonstrate promising results induce a change in the microenvironment at- and around the lesion site to promote endogenous cell repair, including axonal regeneration or the integration of transplanted cells. To date, many of these strategies target only a single aspect of SCI; however, the multifaceted nature of SCI suggests that combinatorial strategies will likely be more effective. Biomaterials are a key component of combinatorial strategies, as they have the potential to deliver drugs locally over a prolonged period of time and aid in cell survival, integration and differentiation. Here we summarize the advantages and limitations of widely used strategies to promote recovery after injury and highlight recent research where biomaterials aided combinatorial strategies to overcome some of the barriers of spinal cord regeneration.
Collapse
Affiliation(s)
- Tobias Führmann
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Department of Chemical Engineering and Applied Chemistry; 200 College Street Toronto ON M5S 3E5 Canada
| | - Priya N. Anandakumaran
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Institute of Biomaterials and Biomedical Engineering; 164 College Street Toronto ON M5S 3G9 Canada
| | - Molly S. Shoichet
- The Donnelly Centre for Cellular and Biomolecular Research; 160 College Street, Room 514 Toronto ON M5S 3E1 Canada
- Department of Chemical Engineering and Applied Chemistry; 200 College Street Toronto ON M5S 3E5 Canada
- Institute of Biomaterials and Biomedical Engineering; 164 College Street Toronto ON M5S 3G9 Canada
- Department of Chemistry; University of Toronto; 80 St George St Toronto ON M5S 3H6 Canada
| |
Collapse
|
28
|
Iyer NR, Wilems TS, Sakiyama-Elbert SE. Stem cells for spinal cord injury: Strategies to inform differentiation and transplantation. Biotechnol Bioeng 2017; 114:245-259. [PMID: 27531038 PMCID: PMC5642909 DOI: 10.1002/bit.26074] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 12/13/2022]
Abstract
The complex pathology of spinal cord injury (SCI), involving a cascade of secondary events and the formation of inhibitory barriers, hampers regeneration across the lesion site and often results in irreversible loss of motor function. The limited regenerative capacity of endogenous cells after SCI has led to a focus on the development of cell therapies that can confer both neuroprotective and neuroregenerative benefits. Stem cells have emerged as a candidate cell source because of their ability to self-renew and differentiate into a multitude of specialized cell types. While ethical and safety concerns impeded the use of stem cells in the past, advances in isolation and differentiation methods have largely mitigated these issues. A confluence of work in stem cell biology, genetics, and developmental neurobiology has informed the directed differentiation of specific spinal cell types. After transplantation, these stem cell-derived populations can replace lost cells, provide trophic support, remyelinate surviving axons, and form relay circuits that contribute to functional recovery. Further refinement of stem cell differentiation and transplantation methods, including combinatorial strategies that involve biomaterial scaffolds and drug delivery, is critical as stem cell-based treatments enter clinical trials. Biotechnol. Bioeng. 2017;114: 245-259. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nisha R Iyer
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Thomas S Wilems
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| |
Collapse
|
29
|
Abstract
Demyelination of central nervous system axons, associated with traumatic injury and demyelinating diseases such as multiple sclerosis, causes impaired neural transmission and ultimately axon degeneration. Consequently, extensive research has focused on signaling systems that promote myelinating activity of oligodendrocytes or promote production of new oligodendrocytes from oligodendrocyte progenitor cells. Many receptor systems, notably including growth factor receptors and G protein-coupled receptors, control myelination. A number of recent clinical trials target these receptor signaling pathways.
Collapse
Affiliation(s)
- Mark Bothwell
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195;
| |
Collapse
|
30
|
Castejón OJ. Ultrastructural Pathology of Oligodendroglial Cells in Traumatic and Hydrocephalic Human Brain Edema: A Review. Ultrastruct Pathol 2016; 39:359-68. [PMID: 26548433 DOI: 10.3109/01913123.2012.750408] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Oligodendroglial cell changes in human traumatic brain injuries and hydrocephalus have been reviewed and compared with experimental brain edema. Resting unreactive oligodendrocytes, reactive oligodendrocytes, anoxic-ischemic oligodendrocytes, hyperthrophic phagocytic oligodendrocytes, and apoptotic oligodendrocytes are found. Anoxic-ischemic oligodendrocytes exhibit enlargement of endoplasmic reticulum, Golgi complex, and enlargement and disassembly of nuclear envelope. They appear in contact with degenerated myelinated axons. Hypertrophic phagocytic oligodendrocytes engulf degenerated myelinated axons exerting myelinolytic effects. A continuum oncotic and apoptotic cell death type leading to necrosis is observed. The vasogenic and cytotoxic components of brain edema are discussed in relation to oligodendroglial cell changes and reactivity.
Collapse
Affiliation(s)
- Orlando J Castejón
- a Biological Research Institute "Drs. Orlando Castejón and Haydée Viloria de Castejón," School of Medicine, Universidad del Zulia , Maracaibo , Venezuela
| |
Collapse
|
31
|
Neuroprotective and Neurorestorative Processes after Spinal Cord Injury: The Case of the Bulbospinal Respiratory Neurons. Neural Plast 2016; 2016:7692602. [PMID: 27563469 PMCID: PMC4987469 DOI: 10.1155/2016/7692602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/29/2016] [Indexed: 11/18/2022] Open
Abstract
High cervical spinal cord injuries interrupt the bulbospinal respiratory pathways projecting to the cervical phrenic motoneurons resulting in important respiratory defects. In the case of a lateralized injury that maintains the respiratory drive on the opposite side, a partial recovery of the ipsilateral respiratory function occurs spontaneously over time, as observed in animal models. The rodent respiratory system is therefore a relevant model to investigate the neuroplastic and neuroprotective mechanisms that will trigger such phrenic motoneurons reactivation by supraspinal pathways. Since part of this recovery is dependent on the damaged side of the spinal cord, the present review highlights our current understanding of the anatomical neuroplasticity processes that are developed by the surviving damaged bulbospinal neurons, notably axonal sprouting and rerouting. Such anatomical neuroplasticity relies also on coordinated molecular mechanisms at the level of the axotomized bulbospinal neurons that will promote both neuroprotection and axon growth.
Collapse
|
32
|
Götz M, Nakafuku M, Petrik D. Neurogenesis in the Developing and Adult Brain-Similarities and Key Differences. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a018853. [PMID: 27235475 DOI: 10.1101/cshperspect.a018853] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adult neurogenesis in the mammalian brain is often viewed as a continuation of neurogenesis at earlier, developmental stages. Here, we will critically review the extent to which this is the case highlighting similarities as well as key differences. Although many transcriptional regulators are shared in neurogenesis at embryonic and adult stages, recent findings on the molecular mechanisms by which these neuronal fate determinants control fate acquisition and maintenance have revealed profound differences between development and adulthood. Importantly, adult neurogenesis occurs in a gliogenic environment, hence requiring adult-specific additional and unique mechanisms of neuronal fate specification and maintenance. Thus, a better understanding of the molecular logic for continuous adult neurogenesis provides important clues to develop strategies to manipulate endogenous stem cells for the purpose of repair.
Collapse
Affiliation(s)
- Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany Synergy, Munich Cluster for Systems Neurology, 81377 Munich, Germany
| | - Masato Nakafuku
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45140 Departments of Pediatrics and Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Petrik
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany
| |
Collapse
|
33
|
Neuroplasticity and Repair in Rodent Neurotoxic Models of Spinal Motoneuron Disease. Neural Plast 2016; 2016:2769735. [PMID: 26862439 PMCID: PMC4735933 DOI: 10.1155/2016/2769735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 07/12/2015] [Accepted: 08/19/2015] [Indexed: 12/14/2022] Open
Abstract
Retrogradely transported toxins are widely used to set up protocols for selective lesioning of the nervous system. These methods could be collectively named "molecular neurosurgery" because they are able to destroy specific types of neurons by using targeted neurotoxins. Lectins such as ricin, volkensin, or modeccin and neuropeptide- or antibody-conjugated saporin represent the most effective toxins used for neuronal lesioning. Some of these specific neurotoxins could be used to induce selective depletion of spinal motoneurons. In this review, we extensively describe two rodent models of motoneuron degeneration induced by volkensin or cholera toxin-B saporin. In particular, we focus on the possible experimental use of these models to mimic neurodegenerative diseases, to dissect the molecular mechanisms of neuroplastic changes underlying the spontaneous functional recovery after motoneuron death, and finally to test different strategies of neural repair. The potential clinical applications of these approaches are also discussed.
Collapse
|
34
|
Xu L, Ryu J, Hiel H, Menon A, Aggarwal A, Rha E, Mahairaki V, Cummings BJ, Koliatsos VE. Transplantation of human oligodendrocyte progenitor cells in an animal model of diffuse traumatic axonal injury: survival and differentiation. Stem Cell Res Ther 2015; 6:93. [PMID: 25971252 PMCID: PMC4453242 DOI: 10.1186/s13287-015-0087-0] [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: 12/19/2014] [Revised: 03/13/2015] [Accepted: 05/01/2015] [Indexed: 12/14/2022] Open
Abstract
Introduction Diffuse axonal injury is an extremely common type of traumatic brain injury encountered in motor vehicle crashes, sports injuries, and in combat. Although many cases of diffuse axonal injury result in chronic disability, there are no current treatments for this condition. Its basic lesion, traumatic axonal injury, has been aggressively modeled in primate and rodent animal models. The inexorable axonal and perikaryal degeneration and dysmyelination often encountered in traumatic axonal injury calls for regenerative therapies, including therapies based on stem cells and precursors. Here we explore the proof of concept that treatments based on transplants of human oligodendrocyte progenitor cells can replace or remodel myelin and, eventually, contribute to axonal regeneration in traumatic axonal injury. Methods We derived human oligodendrocyte progenitor cells from the human embryonic stem cell line H9, purified and characterized them. We then transplanted these human oligodendrocyte progenitor cells into the deep sensorimotor cortex next to the corpus callosum of nude rats subjected to traumatic axonal injury based on the impact acceleration model of Marmarou. We explored the time course and spatial distribution of differentiation and structural integration of these cells in rat forebrain. Results At the time of transplantation, over 90 % of human oligodendrocyte progenitor cells expressed A2B5, PDGFR, NG2, O4, Olig2 and Sox10, a profile consistent with their progenitor or early oligodendrocyte status. After transplantation, these cells survived well and migrated massively via the corpus callosum in both injured and uninjured brains. Human oligodendrocyte progenitor cells displayed a striking preference for white matter tracts and were contained almost exclusively in the corpus callosum and external capsule, the striatopallidal striae, and cortical layer 6. Over 3 months, human oligodendrocyte progenitor cells progressively matured into myelin basic protein(+) and adenomatous polyposis coli protein(+) oligodendrocytes. The injured environment in the corpus callosum of impact acceleration subjects tended to favor maturation of human oligodendrocyte progenitor cells. Electron microscopy revealed that mature transplant-derived oligodendrocytes ensheathed host axons with spiral wraps intimately associated with myelin sheaths. Conclusions Our findings suggest that, instead of differentiating locally, human oligodendrocyte progenitor cells migrate massively along white matter tracts and differentiate extensively into ensheathing oligodendrocytes. These features make them appealing candidates for cellular therapies of diffuse axonal injury aiming at myelin remodeling and axonal protection or regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0087-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leyan Xu
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Jiwon Ryu
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Hakim Hiel
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Adarsh Menon
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Ayushi Aggarwal
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Elizabeth Rha
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Vasiliki Mahairaki
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Brian J Cummings
- Departments of Physical and Medical Rehabilitation, Neurological Surgery, and Anatomy and Neurobiology, Sue and Bill Gross Stem Cell Research Center, Institute for Memory Impairments and Neurological Disorders, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Vassilis E Koliatsos
- Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
35
|
Novel Mechanisms of Spinal Cord Plasticity in a Mouse Model of Motoneuron Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:654637. [PMID: 26064939 PMCID: PMC4433663 DOI: 10.1155/2015/654637] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
A hopeful spinal cord repairing strategy involves the activation of neural precursor cells. Unfortunately, their ability to generate neurons after injury appears limited. Another process promoting functional recovery is synaptic plasticity. We have previously studied some mechanisms of spinal plasticity involving BDNF, Shh, Notch-1, Numb, and Noggin, by using a mouse model of motoneuron depletion induced by cholera toxin-B saporin. TDP-43 is a nuclear RNA/DNA binding protein involved in amyotrophic lateral sclerosis. Interestingly, TDP-43 could be localized at the synapse and affect synaptic strength. Here, we would like to deepen the investigation of this model of spinal plasticity. After lesion, we observed a glial reaction and an activity-dependent modification of Shh, Noggin, and Numb proteins. By using multivariate regression models, we found that Shh and Noggin could affect motor performance and that these proteins could be associated with both TDP-43 and Numb. Our data suggest that TDP-43 is likely an important regulator of synaptic plasticity, probably in collaboration with other proteins involved in both neurogenesis and synaptic plasticity. Moreover, given the rapidly increasing knowledge about spinal cord plasticity, we believe that further efforts to achieve spinal cord repair by stimulating the intrinsic potential of spinal cord will produce interesting results.
Collapse
|
36
|
Garcia-Ovejero D, Arevalo-Martin A, Paniagua-Torija B, Florensa-Vila J, Ferrer I, Grassner L, Molina-Holgado E. The ependymal region of the adult human spinal cord differs from other species and shows ependymoma-like features. Brain 2015; 138:1583-97. [PMID: 25882650 DOI: 10.1093/brain/awv089] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/30/2015] [Indexed: 12/20/2022] Open
Abstract
Several laboratories have described the existence of undifferentiated precursor cells that may act like stem cells in the ependyma of the rodent spinal cord. However, there are reports showing that this region is occluded and disassembled in humans after the second decade of life, although this has been largely ignored or interpreted as a post-mortem artefact. To gain insight into the patency, actual structure, and molecular properties of the adult human spinal cord ependymal region, we followed three approaches: (i) with MRI, we estimated the central canal patency in 59 control subjects, 99 patients with traumatic spinal cord injury, and 26 patients with non-traumatic spinal cord injuries. We observed that the central canal is absent from the vast majority of individuals beyond the age of 18 years, gender-independently, throughout the entire length of the spinal cord, both in healthy controls and after injury; (ii) with histology and immunohistochemistry, we describe morphological properties of the non-lesioned ependymal region, which showed the presence of perivascular pseudorosettes, a common feature of ependymoma; and (iii) with laser capture microdissection, followed by TaqMan® low density arrays, we studied the gene expression profile of the ependymal region and found that it is mainly enriched in genes compatible with a low grade or quiescent ependymoma (53 genes); this region is enriched only in 14 genes related to neurogenic niches. In summary, we demonstrate here that the central canal is mainly absent in the adult human spinal cord and is replaced by a structure morphologically and molecularly different from that described for rodents and other primates. The presented data suggest that the ependymal region is more likely to be reminiscent of a low-grade ependymoma. Therefore, a direct translation to adult human patients of an eventual therapeutic potential of this region based on animal models should be approached with caution.
Collapse
Affiliation(s)
- Daniel Garcia-Ovejero
- 1 Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos (SESCAM), Toledo, Spain
| | - Angel Arevalo-Martin
- 1 Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos (SESCAM), Toledo, Spain
| | - Beatriz Paniagua-Torija
- 1 Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos (SESCAM), Toledo, Spain
| | - José Florensa-Vila
- 2 Radiology Unit, Hospital Nacional de Paraplejicos (SESCAM), Toledo, Spain
| | - Isidro Ferrer
- 3 Institut de Neuropatologia, Servei d'Anatomia Patolo`gica, IDIBELL-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Lukas Grassner
- 4 Center for Spinal Cord Injuries, Trauma Center Murnau, Germany 5 Institute of Molecular Regenerative Medicine, SCI-TReCS (Spinal Cord Injury and Tissue Regeneration Center Salzburg), Paracelsus Medical University, Salzburg, Austria
| | - Eduardo Molina-Holgado
- 1 Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos (SESCAM), Toledo, Spain
| |
Collapse
|
37
|
Liu J, Yang X, Jiang L, Wang C, Yang M. Neural plasticity after spinal cord injury. Neural Regen Res 2015; 7:386-91. [PMID: 25774179 PMCID: PMC4350123 DOI: 10.3969/j.issn.1673-5374.2012.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 01/10/2012] [Indexed: 11/18/2022] Open
Abstract
Plasticity changes of uninjured nerves can result in a novel neural circuit after spinal cord injury, which can restore sensory and motor functions to different degrees. Although processes of neural plasticity have been studied, the mechanism and treatment to effectively improve neural plasticity changes remain controversial. The present study reviewed studies regarding plasticity of the central nervous system and methods for promoting plasticity to improve repair of injured central nerves. The results showed that synaptic reorganization, axonal sprouting, and neurogenesis are critical factors for neural circuit reconstruction. Directed functional exercise, neurotrophic factor and transplantation of nerve-derived and non-nerve-derived tissues and cells can effectively ameliorate functional disturbances caused by spinal cord injury and improve quality of life for patients.
Collapse
Affiliation(s)
- Jian Liu
- Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Xiaoyu Yang
- Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Lianying Jiang
- Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chunxin Wang
- Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Maoguang Yang
- Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
38
|
Wu H, Hu M, Yuan D, Wu H, Wang Y, Wang J, Li T, Qian C, Yu H. Electroacupuncture promotes the proliferation of endogenous neural stem cells and oligodendrocytes in the injured spinal cord of adult rats. Neural Regen Res 2015; 7:1138-44. [PMID: 25722706 PMCID: PMC4340030 DOI: 10.3969/j.issn.1673-5374.2012.15.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 04/23/2012] [Indexed: 11/24/2022] Open
Abstract
A contusive model of spinal cord injury at spinal segment T8-9 was established in rats. Huantiao (GB30) and Huatuojiaji (Ex-B05) were punctured with needles, and endogenous neural stem cells were labeled with 5-bromo-2’-deoxyuridine (BrdU) and NG2. Double immunofluorescence staining showed that electroacupuncture markedly increased the numbers of BrdU+/NG2+ cells at spinal cord tissue 15 mm away from the injury center in the rostral and caudal directions. The results suggest that electroacupuncture promotes the proliferation of endogenous neural stem cells and oligodendrocytes in rats with spinal cord injury.
Collapse
Affiliation(s)
- Haiying Wu
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Min Hu
- Kunming University, Kunming 650118, Yunnan Province, China
| | - Dekai Yuan
- Kunming University, Kunming 650118, Yunnan Province, China
| | - Haiying Wu
- Department of Otolaryngology, Second Affiliated Hospital, Kunming Medical College, Kunming 650101, Yunnan Province, China
| | - Yunhui Wang
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Jing Wang
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Tao Li
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Chuanyun Qian
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| | - Hualin Yu
- Department of Minimally Invasive Neurosurgery, First Affiliated Hospital, Kunming Medical College, Kunming 650032, Yunnan Province, China
| |
Collapse
|
39
|
Corticospinal sprouting differs according to spinal injury location and cortical origin in macaque monkeys. J Neurosci 2015; 34:12267-79. [PMID: 25209269 DOI: 10.1523/jneurosci.1593-14.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The primate corticospinal tract (CST), the major descending pathway mediating voluntary hand movements, comprises nine or more functional subdivisions. The role of subcomponents other than that from primary motor cortex, however, is not well understood. We have previously shown that following a cervical dorsal rhizotomy (Darian-Smith et al., 2013), CST projections originating from primary somatosensory (S1) and motor (M1) cortex responded quite differently to injury. Terminal projections from the S1 (areas 3b/1/2) shrank to <60% of the contralateral side, while M1 CST projections remained robust or expanded (>110%). Here, we asked what happens when a central lesion is added to the equation, to better simulate clinical injury. Monkeys (n = 6) received either a unilateral (1) dorsal root lesion (DRL), (2) or a combined DRL/dorsal column lesion (DRL/DCL), or (3) a DRL/DCL where the DCL was made 4 months following the initial DRL. Electrophysiological recordings were made in S1 4 months postlesion in the first two groups, and 6 weeks after the DCL in the third lesion group, to identify the reorganized region of D1-D3 (thumb, index finger, and middle finger) representation. Anterograde tracers were then injected bilaterally to assess spinal terminal labeling. Remarkably, in all DRL/DCL animals, terminal projections from the S1 and M1 extended bilaterally and caudally well beyond terminal territories in normal animals or following a DRL. These data were highly significant. Extensive sprouting from the S1 CST has not been reported previously, and these data raise important questions about S1 CST involvement in recovery following spinal injury.
Collapse
|
40
|
Juhasova J, Juhas S, Hruska-Plochan M, Dolezalova D, Holubova M, Strnadel J, Marsala S, Motlik J, Marsala M. Time course of spinal doublecortin expression in developing rat and porcine spinal cord: implication in in vivo neural precursor grafting studies. Cell Mol Neurobiol 2015; 35:57-70. [PMID: 25487013 DOI: 10.1007/s10571-014-0145-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022]
Abstract
Expression of doublecortin (DCX), a 43-53 kDa microtubule binding protein, is frequently used as (i) an early neuronal marker to identify the stage of neuronal maturation of in vivo grafted neuronal precursors (NSCs), and (ii) a neuronal fate marker transiently expressed by immature neurons during development. Reliable identification of the origin of DCX-immunoreactive cells (i.e., host vs. graft) requires detailed spatial and temporal mapping of endogenous DCX expression at graft-targeted brain or spinal cord regions. Accordingly, in the present study, we analyzed (i) the time course of DCX expression in pre- and postnatal rat and porcine spinal cord, and (ii) the DCX expression in spinally grafted porcine-induced pluripotent stem cells (iPS)-derived NSCs and human embryonic stem cell (ES)-derived NSCs. In addition, complementary temporospatial GFAP expression study in porcine spinal cord was also performed. In 21-day-old rat fetuses, an intense DCX immunoreactivity distributed between the dorsal horn (DH) and ventral horn was seen and was still present in the DH neurons on postnatal day 20. In animals older than 8 weeks, no DCX immunoreactivity was seen at any spinal cord laminae. In contrast to rat, in porcine spinal cord (gestational period 113-114 days), DCX was only expressed during the pre-natal period (up to 100 days) but was no longer present in newborn piglets or in adult animals. Immunohistochemical analysis was confirmed with a comparable expression profile by western blot analysis. Contrary, the expression of porcine GFAP started within 70-80 days of the pre-natal period. Spinally grafted porcine iPS-NSCs and human ES-NSCs showed clear DCX expression at 3-4 weeks postgrafting. These data indicate that in spinal grafting studies which employ postnatal or adult porcine models, the expression of DCX can be used as a reliable marker of grafted neurons. In contrast, if grafted neurons are to be analyzed during the first 4 postnatal weeks in the rat spinal cord, additional markers or grafted cell-specific labeling techniques need to be employed to reliably identify grafted early postmitotic neurons and to differentiate the DCX expression from the neurons of the host.
Collapse
Affiliation(s)
- J Juhasova
- Laboratory of Cell Regeneration and Plasticity, Institute of Animal Physiology and Genetics, AS CR, v.v.i., Rumburska 89, 27721, Libechov, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhang G, Vidal Pizarro I, Swain GP, Kang SH, Selzer ME. Neurogenesis in the lamprey central nervous system following spinal cord transection. J Comp Neurol 2014; 522:1316-32. [PMID: 24151158 DOI: 10.1002/cne.23485] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/23/2022]
Abstract
After spinal cord transection, lampreys recover functionally and axons regenerate. It is not known whether this is accompanied by neurogenesis. Previous studies suggested a baseline level of nonneuronal cell proliferation in the spinal cord and rhombencephalon (where most supraspinal projecting neurons are located). To determine whether cell proliferation increases after injury and whether this includes neurogenesis, larval lampreys were spinally transected and injected with 5-bromo-2&prime-deoxyuridine (BrdU) at 0-3 weeks posttransection. Labeled cells were counted in the lesion site, within 0.5 mm rostral and caudal to the lesion, and in the rhombencephalon. One group of animals was processed in the winter and a second group was processed in the summer. The number of labeled cells was greater in winter than in summer. The lesion site had the most BrdU labeling at all times, correlating with an increase in the number of cells. In the adjacent spinal cord, the percentage of BrdU labeling was higher in the ependymal than in nonependymal regions. This was also true in the rhombencephalon but only in summer. In winter, BrdU labeling was seen primarily in the subventricular and peripheral zones. Some BrdU-labeled cells were also double labeled by antibodies to glial-specific (antikeratin) as well as neuron-specific (anti-Hu) antigens, indicating that both gliogenesis and neurogenesis occurred after spinal cord transection. However, the new neurons were restricted to the ependymal zone, were never labeled by antineurofilament antibodies, and never migrated away from the ependyma even at 5 weeks after BrdU injection. They would appear to be cerebrospinal fluid-contacting neurons.
Collapse
Affiliation(s)
- Guixin Zhang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Penhnsylvania, 19140; Department of Neurology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, 19104
| | | | | | | | | |
Collapse
|
42
|
Paniagua-Torija B, Arevalo-Martin A, Molina-Holgado E, Molina-Holgado F, Garcia-Ovejero D. Spinal cord injury induces a long-lasting upregulation of interleukin-1β in astrocytes around the central canal. Neuroscience 2014; 284:283-289. [PMID: 25453765 DOI: 10.1016/j.neuroscience.2014.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/23/2014] [Accepted: 10/08/2014] [Indexed: 01/25/2023]
Abstract
Under inflammatory conditions, interleukin-1β (IL-1β) modulates neural stem cells at neurogenic niches. Here we show that spinal cord injury in rats increases IL-1β expression in astrocytes located around the spinal cord ependyma, a region that also holds a neurogenic potential. IL-1β increases from day 1 after lesion, reaches maximal levels between days 3 and 7, and declines from 14 days to low levels after 28 days. At the time of maximal expression, periependymal upregulation of IL-1β extends beyond 5 mm from the epicenter of the lesion both rostral and caudally. Since IL-1β controls proliferation and cell fate of neural stem/precursor cells, its modulation in periependymal astrocytes might create an appropriate environment for cell replacement after injury.
Collapse
Affiliation(s)
- B Paniagua-Torija
- Laboratory of Neuroinflammation, Unidad de Neurologia Experimental, Hospital Nacional de Paraplejicos (SESCAM), 45071 Toledo, Spain.
| | - A Arevalo-Martin
- Laboratory of Neuroinflammation, Unidad de Neurologia Experimental, Hospital Nacional de Paraplejicos (SESCAM), 45071 Toledo, Spain.
| | - E Molina-Holgado
- Laboratory of Neuroinflammation, Unidad de Neurologia Experimental, Hospital Nacional de Paraplejicos (SESCAM), 45071 Toledo, Spain.
| | - F Molina-Holgado
- Neural Stem Cell Laboratory, Department of Life Sciences, Health Sciences Research Centre, University of Roehampton, London SW15 4JD, UK.
| | - D Garcia-Ovejero
- Laboratory of Neuroinflammation, Unidad de Neurologia Experimental, Hospital Nacional de Paraplejicos (SESCAM), 45071 Toledo, Spain.
| |
Collapse
|
43
|
Neural stem cells in the adult spinal cord. Exp Neurol 2014; 260:44-9. [DOI: 10.1016/j.expneurol.2013.01.026] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 01/18/2013] [Accepted: 01/23/2013] [Indexed: 11/20/2022]
|
44
|
Franz S, Ciatipis M, Pfeifer K, Kierdorf B, Sandner B, Bogdahn U, Blesch A, Winner B, Weidner N. Thoracic rat spinal cord contusion injury induces remote spinal gliogenesis but not neurogenesis or gliogenesis in the brain. PLoS One 2014; 9:e102896. [PMID: 25050623 PMCID: PMC4106835 DOI: 10.1371/journal.pone.0102896] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/24/2014] [Indexed: 12/16/2022] Open
Abstract
After spinal cord injury, transected axons fail to regenerate, yet significant, spontaneous functional improvement can be observed over time. Distinct central nervous system regions retain the capacity to generate new neurons and glia from an endogenous pool of progenitor cells and to compensate neural cell loss following certain lesions. The aim of the present study was to investigate whether endogenous cell replacement (neurogenesis or gliogenesis) in the brain (subventricular zone, SVZ; corpus callosum, CC; hippocampus, HC; and motor cortex, MC) or cervical spinal cord might represent a structural correlate for spontaneous locomotor recovery after a thoracic spinal cord injury. Adult Fischer 344 rats received severe contusion injuries (200 kDyn) of the mid-thoracic spinal cord using an Infinite Horizon Impactor. Uninjured rats served as controls. From 4 to 14 days post-injury, both groups received injections of bromodeoxyuridine (BrdU) to label dividing cells. Over the course of six weeks post-injury, spontaneous recovery of locomotor function occurred. Survival of newly generated cells was unaltered in the SVZ, HC, CC, and the MC. Neurogenesis, as determined by identification and quantification of doublecortin immunoreactive neuroblasts or BrdU/neuronal nuclear antigen double positive newly generated neurons, was not present in non-neurogenic regions (MC, CC, and cervical spinal cord) and unaltered in neurogenic regions (dentate gyrus and SVZ) of the brain. The lack of neuronal replacement in the brain and spinal cord after spinal cord injury precludes any relevance for spontaneous recovery of locomotor function. Gliogenesis was increased in the cervical spinal cord remote from the injury site, however, is unlikely to contribute to functional improvement.
Collapse
Affiliation(s)
- Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Mareva Ciatipis
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Kathrin Pfeifer
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Birthe Kierdorf
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Beatrice Sandner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Winner
- IZKF Junior Group III and BMBF Research Group Neuroscience, Interdisciplinary Center for Clinical Research, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University-Erlangen-Nürnberg, Erlangen, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
45
|
Thomas AM, Seidlits SK, Goodman AG, Kukushliev TV, Hassani DM, Cummings BJ, Anderson AJ, Shea LD. Sonic hedgehog and neurotrophin-3 increase oligodendrocyte numbers and myelination after spinal cord injury. Integr Biol (Camb) 2014; 6:694-705. [PMID: 24873988 DOI: 10.1039/c4ib00009a] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) results in loss of sensory and motor function below the level of injury and has limited available therapies. Multiple channel bridges have been investigated as a means to create a permissive environment for regeneration, with channels supporting axonal growth through the injury. Bridges support robust axon growth and myelination. Here, we investigated the cell types that myelinate axons in the bridges and whether over-expression of trophic factors can enhance myelination. Lentivirus encoding for neurotrophin-3 (NT3), sonic hedgehog (SHH) and the combination of these factors was delivered from bridges implanted into a lateral hemisection defect at T9/T10 in mice, and the response of endogenous progenitor cells within the spinal cord was investigated. Relative to control, the localized, sustained expression of these factors significantly increased growth of regenerating axons into the bridge and enhanced axon myelination 8 weeks after injury. SHH decreased the number of Sox2(+) cells and increased the number of Olig2(+) cells, whereas NT3 alone or in combination with SHH enhanced the numbers of GFAP(+) and Olig2(+) cells relative to control. For delivery of lentivirus encoding for either factor, we identified cells at various stages of differentiation along the oligodendrocyte lineage (e.g., O4(+), GalC(+)). Expression of NT3 enhanced myelination primarily by infiltrating Schwann cells, whereas SHH over-expression substantially increased myelination by oligodendrocytes. These studies further establish biomaterial-mediated gene delivery as a promising tool to direct activation and differentiation of endogenous progenitor cells for applications in regenerative medicine.
Collapse
Affiliation(s)
- Aline M Thomas
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Stephanie K Seidlits
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.,Institute for BioNanotechnology in Medicine (IBNAM), Northwestern University, Chicago, IL, USA
| | - Ashley G Goodman
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Todor V Kukushliev
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Donna M Hassani
- Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Brian J Cummings
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA.,Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.,Sue and Bill Gross Stem Cell Center, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders (MIND), Irvine, CA, USA
| | - Aileen J Anderson
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA.,Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.,Sue and Bill Gross Stem Cell Center, Irvine, CA, USA.,Institute for Memory Impairments and Neurological Disorders (MIND), Irvine, CA, USA
| | - Lonnie D Shea
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA.,Institute for BioNanotechnology in Medicine (IBNAM), Northwestern University, Chicago, IL, USA.,Center for Reproductive Science (CRS), Northwestern University, Evanston, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.,Chemistry of Life Processes Institute (CLP), Northwestern University, Evanston, IL, USA
| |
Collapse
|
46
|
Dengler EC, Alberti LA, Bowman BN, Kerwin AA, Wilkerson JL, Moezzi DR, Limanovich E, Wallace JA, Milligan ED. Improvement of spinal non-viral IL-10 gene delivery by D-mannose as a transgene adjuvant to control chronic neuropathic pain. J Neuroinflammation 2014; 11:92. [PMID: 24884664 PMCID: PMC4046049 DOI: 10.1186/1742-2094-11-92] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Peri-spinal subarachnoid (intrathecal; i.t.) injection of non-viral naked plasmid DNA encoding the anti-inflammatory cytokine, IL-10 (pDNA-IL-10) suppresses chronic neuropathic pain in animal models. However, two sequential i.t. pDNA injections are required within a discrete 5 to 72-hour period for prolonged efficacy. Previous reports identified phagocytic immune cells present in the peri-spinal milieu surrounding the i.t injection site that may play a role in transgene uptake resulting in subsequent IL-10 transgene expression. METHODS In the present study, we aimed to examine whether factors known to induce pro-phagocytic anti-inflammatory properties of immune cells improve i.t. IL-10 transgene uptake using reduced naked pDNA-IL-10 doses previously determined ineffective. Both the synthetic glucocorticoid, dexamethasone, and the hexose sugar, D-mannose, were factors examined that could optimize i.t. pDNA-IL-10 uptake leading to enduring suppression of neuropathic pain as assessed by light touch sensitivity of the rat hindpaw (allodynia). RESULTS Compared to dexamethasone, i.t. mannose pretreatment significantly and dose-dependently prolonged pDNA-IL-10 pain suppressive effects, reduced spinal IL-1β and enhanced spinal and dorsal root ganglia IL-10 immunoreactivity. Macrophages exposed to D-mannose revealed reduced proinflammatory TNF-α, IL-1β, and nitric oxide, and increased IL-10 protein release, while IL-4 revealed no improvement in transgene uptake. Separately, D-mannose dramatically increased pDNA-derived IL-10 protein release in culture supernatants. Lastly, a single i.t. co-injection of mannose with a 25-fold lower pDNA-IL-10 dose produced prolonged pain suppression in neuropathic rats. CONCLUSIONS Peri-spinal treatment with D-mannose may optimize naked pDNA-IL-10 transgene uptake for suppression of allodynia, and is a novel approach to tune spinal immune cells toward pro-phagocytic phenotype for improved non-viral gene therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Erin D Milligan
- Department of Neurosciences, UNM School of Medicine, University of New Mexico Health Sciences Center, 1 University of New Mexico, Albuquerque, NM 87131-0001, USA.
| |
Collapse
|
47
|
Alfaro-Cervello C, Cebrian-Silla A, Soriano-Navarro M, Garcia-Tarraga P, Matías-Guiu J, Gomez-Pinedo U, Molina Aguilar P, Alvarez-Buylla A, Luquin MR, Garcia-Verdugo JM. The Adult Macaque Spinal Cord Central Canal Zone Contains Proliferative Cells And Closely Resembles The Human. J Comp Neurol 2014; 522:1800-17. [DOI: 10.1002/cne.23501] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 10/04/2013] [Accepted: 10/30/2013] [Indexed: 01/28/2023]
Affiliation(s)
- Clara Alfaro-Cervello
- Laboratorio de Neurobiología Comparada; Instituto Cavanilles, Universidad de Valencia; CIBERNED Valencia 46980 Spain
| | - Arantxa Cebrian-Silla
- Laboratorio de Neurobiología Comparada; Instituto Cavanilles, Universidad de Valencia; CIBERNED Valencia 46980 Spain
| | - Mario Soriano-Navarro
- Laboratorio de Neurobiología Comparada; Instituto Cavanilles, Universidad de Valencia; CIBERNED Valencia 46980 Spain
| | - Patricia Garcia-Tarraga
- Laboratorio de Neurobiología Comparada; Instituto Cavanilles, Universidad de Valencia; CIBERNED Valencia 46980 Spain
| | - Jorge Matías-Guiu
- Instituto de Neurociencias; IdISSC, Hospital Clínico San Carlos Madrid 28040 Spain
| | - Ulises Gomez-Pinedo
- Instituto de Neurociencias; IdISSC, Hospital Clínico San Carlos Madrid 28040 Spain
| | - Pilar Molina Aguilar
- Servicio de Patología; Instituto de Medicina Legal de Valencia; Valencia 46013 Spain
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery and Institute for Regeneration Medicine; University of California at San Francisco; San Francisco California 94143, USA
| | - Maria-Rosario Luquin
- Laboratorio de Terapia Regenerativa, Departmento de Neurología, División de Neurociencias, Centro de Investigacion Médica Aplicada; Universidad de Navarra; Pamplona 31008 Spain
| | - Jose Manuel Garcia-Verdugo
- Laboratorio de Neurobiología Comparada; Instituto Cavanilles, Universidad de Valencia; CIBERNED Valencia 46980 Spain
| |
Collapse
|
48
|
Su Z, Niu W, Liu ML, Zou Y, Zhang CL. In vivo conversion of astrocytes to neurons in the injured adult spinal cord. Nat Commun 2014; 5:3338. [PMID: 24569435 PMCID: PMC3966078 DOI: 10.1038/ncomms4338] [Citation(s) in RCA: 303] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 01/29/2014] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to irreversible neuronal loss and glial scar formation, which ultimately result in persistent neurological dysfunction. Cellular regeneration could be an ideal approach to replenish the lost cells and repair the damage. However, the adult spinal cord has limited ability to produce new neurons. Here we show that resident astrocytes can be converted to doublecortin (DCX)-positive neuroblasts by a single transcription factor, SOX2, in the injured adult spinal cord. Importantly, these induced neuroblasts can mature into synapse-forming neurons in vivo. Neuronal maturation is further promoted by treatment with a histone deacetylase inhibitor, valproic acid (VPA). The results of this study indicate that in situ reprogramming of endogenous astrocytes to neurons might be a potential strategy for cellular regeneration after SCI.
Collapse
Affiliation(s)
- Zhida Su
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Ministry of Education, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, 800 Xiangyin Rd, Shanghai 200433, China
| | - Wenze Niu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA
| | - Meng-Lu Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA
- Corresponding author Chun-Li Zhang, Ph.D., Tel: 212-648-1670, Fax: 214-648-1488,
| |
Collapse
|
49
|
Omega-3 fatty acids and traumatic neurological injury: from neuroprotection to neuroplasticity? Trends Neurosci 2014; 37:30-8. [DOI: 10.1016/j.tins.2013.10.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 01/21/2023]
|
50
|
Fiorelli R, Cebrian-Silla A, Garcia-Verdugo JM, Raineteau O. The adult spinal cord harbors a population of GFAP-positive progenitors with limited self-renewal potential. Glia 2013; 61:2100-13. [DOI: 10.1002/glia.22579] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Roberto Fiorelli
- Brain Research Institute; University of Zurich/ETHZ; Switzerland
| | - Arantxa Cebrian-Silla
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva; University of Valencia, 46980, CIBERNED; Valencia Spain
| | - Jose-Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biologia Evolutiva; University of Valencia, 46980, CIBERNED; Valencia Spain
| | | |
Collapse
|