1
|
Buchl SC, Kim HN, Hur B, Simon WL, Langley MR, Sung J, Scarisbrick IA. Delayed atorvastatin delivery promotes recovery after experimental spinal cord injury. Neurotherapeutics 2025:e00517. [PMID: 39755500 DOI: 10.1016/j.neurot.2024.e00517] [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: 09/06/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025] Open
Abstract
Spinal cord injury (SCI) significantly alters gene expression, potentially impeding functional recovery. This study investigated the effects of atorvastatin, a widely prescribed cholesterol-lowering drug, on gene expression and functional recovery in a chronic murine SCI model. Female C57BL/6J mice underwent moderate 0.25 mm lateral compression SCI and received daily atorvastatin (10 mg/kg) or vehicle-only injections from two weeks post-injury for four weeks. Sensorimotor functions were assessed using the Basso Mouse Scale (BMS), its subscore, and the inclined plane test. RNA sequencing of spinal cord tissues identified robust transcriptomic changes from SCI and a smaller subset from atorvastatin treatment. Atorvastatin enhanced sensorimotor recovery within two weeks of treatment initiation, with effects persisting to the experimental endpoint. Pathway analysis showed atorvastatin enriched neural regeneration processes including Fatty Acid Transport, Axon Guidance, and the Endocannabinoid Developing Neuron Pathway; improved mitochondrial function via increased TCA Cycle II and reduced Mitochondrial Dysfunction; and decreased Inhibition of Matrix Metalloproteases. Key gene drivers included Fabp7, Unc5c, Rest, and Klf4. Together, these results indicate atorvastatin's potential in chronic SCI recovery, especially where already indicated for cardiovascular protection.
Collapse
Affiliation(s)
- Samuel C Buchl
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Benjamin Hur
- Microbiomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA; Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Whitney L Simon
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Monica R Langley
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jaeyun Sung
- Microbiomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA; Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Isobel A Scarisbrick
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Hassan OI, Takamiya S, Asgarihafshejani A, Fehlings MG. Bridging the gap: a translational perspective in spinal cord injury. Exp Biol Med (Maywood) 2024; 249:10266. [PMID: 39391076 PMCID: PMC11464315 DOI: 10.3389/ebm.2024.10266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating and complex condition to treat with no curative options. In the past few decades, rapid advancements in our understanding of SCI pathophysiology as well as the mergence of new treatments has created more optimism. Focusing on clinical translation, this paper provides a comprehensive overview of SCI through its epidemiology, pathophysiology, currently employed management strategies, and emerging therapeutic approaches. Additionally, it emphasizes the importance of addressing the heavy quality of life (QoL) challenges faced by SCI patients and their desires, providing a basis to tailor patient-centric forms of care. Furthermore, this paper discusses the frequently encountered barriers in translation from preclinical models to clinical settings. It also seeks to summarize significant completed and ongoing SCI clinical trials focused on neuroprotective and neuroregenerative strategies. While developing a cohesive regenerative treatment strategy remains challenging, even modest improvements in sensory and motor function can offer meaningful benefits and motivation for patients coping with this highly debilitating condition.
Collapse
Affiliation(s)
- Omar Imad Hassan
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Soichiro Takamiya
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Azam Asgarihafshejani
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Sun S, Li J, Wang S, Li J, Ren J, Bao Z, Sun L, Ma X, Zheng F, Ma S, Sun L, Wang M, Yu Y, Ma M, Wang Q, Chen Z, Ma H, Wang X, Wu Z, Zhang H, Yan K, Yang Y, Zhang Y, Zhang S, Lei J, Teng ZQ, Liu CM, Bai G, Wang YJ, Li J, Wang X, Zhao G, Jiang T, Belmonte JCI, Qu J, Zhang W, Liu GH. CHIT1-positive microglia drive motor neuron ageing in the primate spinal cord. Nature 2023; 624:611-620. [PMID: 37907096 DOI: 10.1038/s41586-023-06783-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Ageing is a critical factor in spinal-cord-associated disorders1, yet the ageing-specific mechanisms underlying this relationship remain poorly understood. Here, to address this knowledge gap, we combined single-nucleus RNA-sequencing analysis with behavioural and neurophysiological analysis in non-human primates (NHPs). We identified motor neuron senescence and neuroinflammation with microglial hyperactivation as intertwined hallmarks of spinal cord ageing. As an underlying mechanism, we identified a neurotoxic microglial state demarcated by elevated expression of CHIT1 (a secreted mammalian chitinase) specific to the aged spinal cords in NHP and human biopsies. In the aged spinal cord, CHIT1-positive microglia preferentially localize around motor neurons, and they have the ability to trigger senescence, partly by activating SMAD signalling. We further validated the driving role of secreted CHIT1 on MN senescence using multimodal experiments both in vivo, using the NHP spinal cord as a model, and in vitro, using a sophisticated system modelling the human motor-neuron-microenvironment interplay. Moreover, we demonstrated that ascorbic acid, a geroprotective compound, counteracted the pro-senescent effect of CHIT1 and mitigated motor neuron senescence in aged monkeys. Our findings provide the single-cell resolution cellular and molecular landscape of the aged primate spinal cord and identify a new biomarker and intervention target for spinal cord degeneration.
Collapse
Affiliation(s)
- Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhaoshi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
| | - Le Sun
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xibo Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Fangshuo Zheng
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Liang Sun
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Min Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Yan Yu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miyang Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Chen
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - He Ma
- MAIS, State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- College of Medicine and Biomedical Information Engineering, Northeastern University, Shenyang, China
| | - Xuebao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zeming Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hui Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yuanhan Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yixin Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sheng Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ge Bai
- The MOE Frontier Research Center of Brain & Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yan-Jiang Wang
- Aging Biomarker Consortium, Beijing, China
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jian Li
- Aging Biomarker Consortium, Beijing, China
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xiaoqun Wang
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
- Clinical Research Center for Epilepsy Capital Medical University, Beijing, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- The Chinese Glioma Genome Atlas, Beijing, China
- Beijing Neurosurgical Institute, Beijing, China
| | | | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
4
|
Niu Y, Liu J, Qin H, Liu Y, Huang N, Jiang J, Chen Y, Chen S, Bai T, Yang C, Cao Y, Liu S, Yuan H. Development of an innovative minimally invasive primate spinal cord injury model: A case report. IBRAIN 2023; 9:349-356. [PMID: 37786753 PMCID: PMC10527794 DOI: 10.1002/ibra.12117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 10/04/2023]
Abstract
Spinal cord injury (SCI) animal models have been widely created and utilized for repair therapy research, but more suitable experimental animals and accurate modeling methodologies are required to achieve the desired results. In this experiment, we constructed an innovative dorsal 1/4 spinal cord transection macaque model that had fewer severe problems, facilitating postoperative care and recovery. In essence, given that monkeys and humans share similar genetics and physiology, the efficacy of this strategy in a nonhuman primate SCI model basically serves as a good basis for its prospective therapeutic use in human SCI.
Collapse
Affiliation(s)
- Yong‐Min Niu
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Jin‐Xiang Liu
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Hao‐Yue Qin
- Department of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Yi‐Fan Liu
- Yunnan Cancer HospitalThe Third Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Ni‐Jiao Huang
- Department of Orthopaedic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Ji‐Li Jiang
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| | - Yan‐Qiu Chen
- School of Preclinical MedicalZunyi Medical UniversityZunyiGuizhouChina
| | - Si‐Jing Chen
- Nursing SchoolZunyi Medical UniversityZunyiGuizhouChina
| | - Tao Bai
- School of Preclinical MedicalKunming Medical UniversityKunmingYunnanChina
| | - Chang‐Wei Yang
- Department of Nuclear MedicineAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yu Cao
- Department of Nuclear MedicineAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Sheng Liu
- Pharmacology InstituteHeidelberg UniversityHeidelbergGermany
| | - Hao Yuan
- Department of Orthopaedic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
5
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Liang F, Yu S, Pang S, Wang X, Jie J, Gao F, Song Z, Li B, Liao WH, Yin M. Non-human primate models and systems for gait and neurophysiological analysis. Front Neurosci 2023; 17:1141567. [PMID: 37188006 PMCID: PMC10175625 DOI: 10.3389/fnins.2023.1141567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Brain-computer interfaces (BCIs) have garnered extensive interest and become a groundbreaking technology to restore movement, tactile sense, and communication in patients. Prior to their use in human subjects, clinical BCIs require rigorous validation and verification (V&V). Non-human primates (NHPs) are often considered the ultimate and widely used animal model for neuroscience studies, including BCIs V&V, due to their proximity to humans. This literature review summarizes 94 NHP gait analysis studies until 1 June, 2022, including seven BCI-oriented studies. Due to technological limitations, most of these studies used wired neural recordings to access electrophysiological data. However, wireless neural recording systems for NHPs enabled neuroscience research in humans, and many on NHP locomotion, while posing numerous technical challenges, such as signal quality, data throughout, working distance, size, and power constraint, that have yet to be overcome. Besides neurological data, motion capture (MoCap) systems are usually required in BCI and gait studies to capture locomotion kinematics. However, current studies have exclusively relied on image processing-based MoCap systems, which have insufficient accuracy (error: ≥4° and 9 mm). While the role of the motor cortex during locomotion is still unclear and worth further exploration, future BCI and gait studies require simultaneous, high-speed, accurate neurophysiological, and movement measures. Therefore, the infrared MoCap system which has high accuracy and speed, together with a high spatiotemporal resolution neural recording system, may expand the scope and improve the quality of the motor and neurophysiological analysis in NHPs.
Collapse
Affiliation(s)
- Fengyan Liang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Department of Rehabilitation Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Shanshan Yu
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Siqi Pang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Xiao Wang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Jing Jie
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Fei Gao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhenhua Song
- Department of Rehabilitation Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Binbin Li
- Department of Rehabilitation Medicine, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Wei-Hsin Liao
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, China
| | - Ming Yin
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- *Correspondence: Ming Yin,
| |
Collapse
|
7
|
Chen C, Xu HH, Liu XY, Zhang YS, Zhong L, Wang YW, Xu L, Wei P, Chen YX, Liu P, Hao CR, Jia XL, Hu N, Wu XY, Gu XS, Chen LQ, Li XH. 3D printed collagen/silk fibroin scaffolds carrying the secretome of human umbilical mesenchymal stem cells ameliorated neurological dysfunction after spinal cord injury in rats. Regen Biomater 2022; 9:rbac014. [PMID: 35480857 PMCID: PMC9036898 DOI: 10.1093/rb/rbac014] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
Abstract
Although implantation of biomaterials carrying mesenchymal stem cells (MSCs) is considered as a promising strategy for ameliorating neural function after spinal cord injury (SCI), there are still some challenges including poor cell survival rate, tumorigenicity and ethics concerns. The performance of the secretome derived from MSCs was more stable, and its clinical transformation was more operable. Cytokine antibody array demonstrated that the secretome of MSCs contained 79 proteins among the 174 proteins analyzed. 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome improved hindlimb locomotor function according to the BBB scores, the inclined-grid climbing test and electrophysiological analysis. Parallel with locomotor function recovery, 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome could further facilitate nerve fiber regeneration, enhance remyelination and accelerate the establishment of synaptic connections at the injury site compared to 3D printed collagen/silk fibroin scaffolds alone group according to magnetic resonance imaging (MRI), diffusion Tensor imaging (DTI), hematoxylin and eosin (HE) staining, Bielschowsky’s silver staining immunofluorescence staining and transmission electron microscopy (TEM). These results indicated the implantation of 3D printed collagen/silk fibroin scaffolds carrying MSCs secretome might be a potential treatment for SCI.
Collapse
Affiliation(s)
- Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People’s Armed Police Forces, Tianjin, 300162, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of People’s Armed Police Forces, Tianjin, 300162, China
| | - Xiao-Yin Liu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Yu-Sheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Lin Zhong
- Department of Hematology, the first affiliated hospital of Chengdu medical college, Chengdu, Sichuan, 610500, China
| | - You-Wei Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lin Xu
- Medical Psychology Section, Hubei General Hospital of Armed Police Force, Wuhan, Hubei, 430071, China
| | - Pan Wei
- Department of Neurosurgery, The First People's Hospital Of Long Quan yi District, Cheng Du 610000, Si Chuan, China
| | - Ya-Xing Chen
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Peng Liu
- Department of Neurosurgery, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chen-Ru Hao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Li Jia
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Yang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, China
| | - Xiao-Song Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
8
|
Anatomical and behavioral outcomes following a graded hemi-contusive cervical spinal cord injury model in mice. Behav Brain Res 2022; 419:113698. [PMID: 34856301 DOI: 10.1016/j.bbr.2021.113698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND A graded hemi-contusion spinal cord injury produces complex anatomical deformation of the spinal cord parenchyma. The relationship between lesion severity and behavioral consequences in a novel contusion mouse model remains unknown. PURPOSE We aimed to establish a graded cervical hemi-contusion spinal cord injury model in mice and investigate the correlation between graded anatomical damage to the spinal cord and resulting behavioral impairments. METHODS Thirty-two mice were divided into groups of 1.2 mm, 1.5 mm and sham. The tip of an impactor with a diameter of 1 mm was utilized to compress the left dorsal cord of C5 by 1.2 mm or 1.5 mm at a speed of 300 mm/s. Forelimb motor function was evaluated using rearing, grooming and grip-strength tests before and after the injuries. Histologically the area of white matter sparing, gray matter sparing and lesion area were quantified at 6-week-post-injury. RESULTS Behavioral assessments showed a more severe forelimb functional deficit in 1.5 mm contusion displacements relative to 1.2 mm contusion displacements after injury. The 1.2 mm hemi-contusion mainly caused damage to the dorsal fasciculus, ventral and dorsal horn, while the 1.5 mm hemi-contusion lead to additional damage extending to ventral fasciculus. Sparing of the gray and white matter at the epicenter was 36.8 ± 2.4% and 12.4 ± 2.6% in the 1.2 mm group, and 27.6 ± 4.0% and 4.1 ± 2.2% in the 1.5 mm group, respectively. Furthermore, the lesion area was 20.8 ± 3.0% and 36.0 ± 2.1% in the 1.2 mm and 1.5 mm groups, respectively. There was a significant correlation between the performance in the grooming test and white matter sparing, and between grip-test strength and gray matter sparing. CONCLUSION The present study demonstrates that a hemi-contusion cervical spinal cord injury in mice can be graded by contusion displacement and that there is a correlation between anatomical and behavioral outcomes. This study provides a means for determining the severity of lesions in a contusion mouse model.
Collapse
|
9
|
Perez JC, Gerber YN, Perrin FE. Dynamic Diversity of Glial Response Among Species in Spinal Cord Injury. Front Aging Neurosci 2021; 13:769548. [PMID: 34899275 PMCID: PMC8662749 DOI: 10.3389/fnagi.2021.769548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/29/2021] [Indexed: 12/11/2022] Open
Abstract
The glial scar that forms after traumatic spinal cord injury (SCI) is mostly composed of microglia, NG2 glia, and astrocytes and plays dual roles in pathophysiological processes induced by the injury. On one hand, the glial scar acts as a chemical and physical obstacle to spontaneous axonal regeneration, thus preventing functional recovery, and, on the other hand, it partly limits lesion extension. The complex activation pattern of glial cells is associated with cellular and molecular crosstalk and interactions with immune cells. Interestingly, response to SCI is diverse among species: from amphibians and fishes that display rather limited (if any) glial scarring to mammals that exhibit a well-identifiable scar. Additionally, kinetics of glial activation varies among species. In rodents, microglia become activated before astrocytes, and both glial cell populations undergo activation processes reflected amongst others by proliferation and migration toward the injury site. In primates, glial cell activation is delayed as compared to rodents. Here, we compare the spatial and temporal diversity of the glial response, following SCI amongst species. A better understanding of mechanisms underlying glial activation and scar formation is a prerequisite to develop timely glial cell-specific therapeutic strategies that aim to increase functional recovery.
Collapse
Affiliation(s)
| | - Yannick N Gerber
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France
| | - Florence E Perrin
- MMDN, Université de Montpellier, EPHE, INSERM, Montpellier, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
10
|
Lai BQ, Zeng X, Han WT, Che MT, Ding Y, Li G, Zeng YS. Stem cell-derived neuronal relay strategies and functional electrical stimulation for treatment of spinal cord injury. Biomaterials 2021; 279:121211. [PMID: 34710795 DOI: 10.1016/j.biomaterials.2021.121211] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 10/09/2021] [Accepted: 10/20/2021] [Indexed: 01/06/2023]
Abstract
The inability of adult mammals to recover function lost after severe spinal cord injury (SCI) has been known for millennia and is mainly attributed to a failure of brain-derived nerve fiber regeneration across the lesion. Potential approaches to re-establishing locomotor function rely on neuronal relays to reconnect the segregated neural networks of the spinal cord. Intense research over the past 30 years has focused on endogenous and exogenous neuronal relays, but progress has been slow and the results often controversial. Treatments with stem cell-derived neuronal relays alone or together with functional electrical stimulation offer the possibility of improved repair of neuronal networks. In this review, we focus on approaches to recovery of motor function in paralyzed patients after severe SCI based on novel therapies such as implantation of stem cell-derived neuronal relays and functional electrical stimulation. Recent research progress offers hope that SCI patients will one day be able to recover motor function and sensory perception.
Collapse
Affiliation(s)
- Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Wei-Tao Han
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Tian Che
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ying Ding
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan, School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
11
|
Züchner M, Escalona MJ, Teige LH, Balafas E, Zhang L, Kostomitsopoulos N, Boulland JL. How to generate graded spinal cord injuries in swine - tools and procedures. Dis Model Mech 2021; 14:dmm049053. [PMID: 34464444 PMCID: PMC8419714 DOI: 10.1242/dmm.049053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a medically, psychologically and socially disabling condition. A large body of our knowledge on the basic mechanisms of SCI has been gathered in rodents. For preclinical validation of promising therapies, the use of animal models that are closer to humans has several advantages. This has promoted the more-intensive development of large-animal models for SCI during the past decade. We recently developed a multimodal SCI apparatus for large animals that generated biomechanically reproducible impacts in vivo. It is composed of a spring-load impactor and support systems for the spinal cord and the vertebral column. We now present the functional outcome of farm pigs and minipigs injured with different lesion strengths. There was a correlation between the biomechanical characteristics of the impact, the functional outcome and the tissue damage observed several weeks after injury. We also provide a detailed description of the procedure to generate such a SCI in both farm pigs and minipigs, in the hope to ease the adoption of the swine model by other research groups.
Collapse
Affiliation(s)
- Mark Züchner
- Department of Neurosurgery, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway
| | - Manuel J. Escalona
- Department for Immunology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway
| | - Lena Hammerlund Teige
- Department for Immunology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway
| | - Evangelos Balafas
- Center of Clinical Experimental Surgery and Translational Research, Biomedical Research Foundation of Academy of Athens, 11527 Athens, Greece
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - Nikolaos Kostomitsopoulos
- Center of Clinical Experimental Surgery and Translational Research, Biomedical Research Foundation of Academy of Athens, 11527 Athens, Greece
| | - Jean-Luc Boulland
- Department for Immunology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway
| |
Collapse
|
12
|
Zhong G, Yang Y, Huang X, Chen J, Feng D, Wei K, Chen J, Chen H. The Serum SIRT1 Protein is Associated with the Severity of Injury and Neurological Recovery in Mice with Traumatic Spinal Cord Injury. Neuroscience 2021; 469:103-109. [PMID: 34171408 DOI: 10.1016/j.neuroscience.2021.06.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
The present study aimed to investigate the association between the serum SIRT1 protein and the severity of spinal cord injury (SCI) as well as the neurological recovery in mice. In this study, the wild-type (WT), Mx1-Cre+ SIRT1loxP/loxP (Mx1), and LCK-Cre+SIRT1loxP/loxP (LCK) mice were subjected to sham surgery, mild, moderate, or severe SCI, respectively. The serum was collected at intervals of 12 h, 1 day (d), 3 d, 5 d, 7 d, 10 d, 14 d, and 21 d after the injury. The locomotor function of all the animals was assessed using the Basso mouse scale (BMS) and the serum SIRT1 proteins were analyzed using enzyme-linked immunosorbent assay (ELISA). The results demonstrated that about 7-10 d after SCI, the levels of SIRT1 protein in the serum correlated significantly with the severity of the injury and at 28 d post-injury, there was a distant neurological recovery (BMS score). The serum SIRT1 concentration in both the Mx1 and LCK mice in the sham group was significantly reduced compared to that in the WT mice, and there was a delayed increase in the serum SIRT1 levels after injury. These findings indicate that the SIRT1 concentrations in the serum of the SCI mice closely correlated with the acute severity and neurological outcome.
Collapse
Affiliation(s)
- Guibin Zhong
- Medical Department, Baoshan Branch Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200444, China; Department of Orthopedics, Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yanqiu Yang
- Medical Department, Baoshan Branch Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200444, China
| | - Xiaodong Huang
- Department of Orthopedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangdong 510150, China
| | - Junling Chen
- Medical Department, Baoshan Branch Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200444, China
| | - Daming Feng
- Medical Department, Baoshan Branch Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200444, China
| | - Ke Wei
- Medical Department, Baoshan Branch Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200444, China
| | - Jianwei Chen
- Department of Orthopedics, Ren Ji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Haihong Chen
- Orthopedic Department, Minhang Center Hospital, Fudan University, Shanghai 201100, China.
| |
Collapse
|
13
|
Baklaushev VP, Durov OV, Kalsin VA, Gulaev EV, Kim SV, Gubskiy IL, Revkova VA, Samoilova EM, Melnikov PA, Karal-Ogly DD, Orlov SV, Troitskiy AV, Chekhonin VP, Averyanov AV, Ahlfors JE. Disease modifying treatment of spinal cord injury with directly reprogrammed neural precursor cells in non-human primates. World J Stem Cells 2021; 13:452-469. [PMID: 34136075 PMCID: PMC8176843 DOI: 10.4252/wjsc.v13.i5.452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/20/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of regenerative therapy for human spinal cord injury (SCI) is dramatically restricted by two main challenges: the need for a safe source of functionally active and reproducible neural stem cells and the need of adequate animal models for preclinical testing. Direct reprogramming of somatic cells into neuronal and glial precursors might be a promising solution to the first challenge. The use of non-human primates for preclinical studies exploring new treatment paradigms in SCI results in data with more translational relevance to human SCI.
AIM To investigate the safety and efficacy of intraspinal transplantation of directly reprogrammed neural precursor cells (drNPCs).
METHODS Seven non-human primates with verified complete thoracic SCI were divided into two groups: drNPC group (n = 4) was subjected to intraspinal transplantation of 5 million drNPCs rostral and caudal to the lesion site 2 wk post injury, and lesion control (n = 3) was injected identically with the equivalent volume of vehicle.
RESULTS Follow-up for 12 wk revealed that animals in the drNPC group demonstrated a significant recovery of the paralyzed hindlimb as well as recovery of somatosensory evoked potential and motor evoked potential of injured pathways. Magnetic resonance diffusion tensor imaging data confirmed the intraspinal transplantation of drNPCs did not adversely affect the morphology of the central nervous system or cerebrospinal fluid circulation. Subsequent immunohistochemical analysis showed that drNPCs maintained SOX2 expression characteristic of multipotency in the transplanted spinal cord for at least 12 wk, migrating to areas of axon growth cones.
CONCLUSION Our data demonstrated that drNPC transplantation was safe and contributed to improvement of spinal cord function after acute SCI, based on neurological status assessment and neurophysiological recovery within 12 wk after transplantation. The functional improvement described was not associated with neuronal differentiation of the allogeneic drNPCs. Instead, directed drNPCs migration to the areas of active growth cone formation may provide exosome and paracrine trophic support, thereby further supporting the regeneration processes.
Collapse
Affiliation(s)
- Vladimir P Baklaushev
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Oleg V Durov
- Department of Neurosurgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, Moscow 115682, Moskva, Russia
| | - Vladimir A Kalsin
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Eugene V Gulaev
- Department of Neurosurgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, Moscow 115682, Moskva, Russia
| | - Sergey V Kim
- Department of Anesthesiology, N.N.Blokhin Russian Cancer Research Centre, Moscow 115478, Moskva, Russia
| | - Ilya L Gubskiy
- Ilya L Gubskiy, Radiology and Clinical Physiology Scientific Research Center, Federal center of brain research and neurotechnologies of the Federal Medical Biological Agency, Moscow 117997, Russia
| | - Veronika A Revkova
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Ekaterina M Samoilova
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Pavel A Melnikov
- Department of Neurobiology, Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119992, Moskva, Russia
| | - Dzhina D Karal-Ogly
- Department of Primatology, Russian Acad Med Sci, Research Institute of Medical Primatology, Sochi 119992, Sochi, Russia
| | - Sergey V Orlov
- Department of Primatology, Russian Acad Med Sci, Research Institute of Medical Primatology, Sochi 119992, Sochi, Russia
| | - Alexander V Troitskiy
- Department of Vascular Surgery, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | - Vladimir P Chekhonin
- Department of Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Center for Psychiatry and Narcology, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University (RNRMU), Moscow 115682, Moskva, Russia
| | - Alexander V Averyanov
- Biomedical Research, Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, Moscow 115682, Moskva, Russia
| | | |
Collapse
|
14
|
Cui HY, Wu YX, Li R, Li GS, Hu Y. A translational study of somatosensory evoked potential time-frequency components in rats, goats, and humans. Neural Regen Res 2021; 16:2269-2275. [PMID: 33818512 PMCID: PMC8354111 DOI: 10.4103/1673-5374.310693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Somatosensory evoked potentials (SEPs) have been widely used to assess neurological function in clinical practice. A good understanding of the association between SEP signals and neurological function is helpful for precise diagnosis of impairment location. Previous studies on SEPs have been reported in animal models. However, few studies have reported the relationships between SEP waveforms in animals and those in humans. In this study, we collected normal SEP waveforms and decomposed them into specific time-frequency components (TFCs). Our results showed three stable TFC distribution regions in intact goats and rats and in humans. After we induced spinal cord injury in the animal models, a greater number of small TFC distribution regions were observed in the injured goat and rat groups than in the normal group. Moreover, there were significant correlations (P < 0.05) and linear relationships between the main SEP TFCs of the human group and those of the goat and rat groups. A stable TFC distribution of SEP components was observed in the human, goat and rat groups, and the TFC distribution modes were similar between the three groups. Results in various animal models in this study could be translated to future clinical studies based on SEP TFC analysis. Human studies were approved by the Institutional Review Board of the University of Hong Kong/Hospital Authority Hong Kong West Cluster (approval No. UM 05-312 T/975) on December 5, 2005. Rat experiments were approved by the Committee on the Use of Live Animals in Teaching and Research of Li Ka Shing Faculty of Medicine of the University of Hong Kong (approval No. CULART 2912-12) on January 28, 2013. Goat experiments were approved by the Animal Ethics Committee of Affiliated Hospital of Guangdong Medical University (approval No. GDY2002132) on March 5, 2018.
Collapse
Affiliation(s)
- Hong-Yan Cui
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yi-Xin Wu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Rong Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Guang-Sheng Li
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region; Spinal Division, Department of Orthopaedics, Affiliated Hospital of Guangdong Medical University, Guangdong Province, China
| | - Yong Hu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin; Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region; Department of Orthopaedics and Traumatology, The University of Hong Kong -Shenzhen Hospital, Shenzhen, Guangdong Province, China
| |
Collapse
|
15
|
Liu Y, Xie JX, Niu F, Xu Z, Tan P, Shen C, Gao H, Liu S, Ma Z, So KF, Wu W, Chen C, Gao S, Xu XM, Zhu H. Surgical intervention combined with weight-bearing walking training improves neurological recoveries in 320 patients with clinically complete spinal cord injury: a prospective self-controlled study. Neural Regen Res 2021; 16:820-829. [PMID: 33229715 PMCID: PMC8178778 DOI: 10.4103/1673-5374.297080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Although a large number of trials in the SCI field have been conducted, few proven gains have been realized for patients. In the present study, we determined the efficacy of a novel combination treatment involving surgical intervention and long-term weight-bearing walking training in spinal cord injury (SCI) subjects clinically diagnosed as complete or American Spinal Injury Association Impairment Scale (AIS) Class A (AIS-A). A total of 320 clinically complete SCI subjects (271 male and 49 female), aged 16–60 years, received early (≤ 7 days, n = 201) or delayed (8–30 days, n = 119) surgical interventions to reduce intraspinal or intramedullary pressure. Fifteen days post-surgery, all subjects received a weight-bearing walking training with the “Kunming Locomotion Training Program (KLTP)” for a duration of 6 months. The neurological deficit and recovery were assessed using the AIS scale and a 10-point Kunming Locomotor Scale (KLS). We found that surgical intervention significantly improved AIS scores measured at 15 days post-surgery as compared to the pre-surgery baseline scores. Significant improvement of AIS scores was detected at 3 and 6 months and the KLS further showed significant improvements between all pair-wise comparisons of time points of 15 days, 3 or 6 months indicating continued improvement in walking scores during the 6-month period. In conclusion, combining surgical intervention within 1 month post-injury and weight-bearing locomotor training promoted continued and statistically significant neurological recoveries in subjects with clinically complete SCI, which generally shows little clinical recovery within the first year after injury and most are permanently disabled. This study was approved by the Science and Research Committee of Kunming General Hospital of PLA and Kunming Tongren Hospital, China and registered at ClinicalTrials.gov (Identifier: NCT04034108) on July 26, 2019.
Collapse
Affiliation(s)
- Yansheng Liu
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Jia-Xin Xie
- Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Fang Niu
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Zhexi Xu
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Pengju Tan
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Caihong Shen
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Hongkun Gao
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Song Liu
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Zhengwen Ma
- Department of Laboratory Animal Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kwok-Fai So
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region; Guangdong-Hongkong-Macau Institute for Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Wutian Wu
- Guangdong-Hongkong-Macau Institute for Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province; Re-Stem Biotechnology, Co., Ltd., Suzhou, Jiangsu Province, China
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hui Zhu
- Kunming International Spine and Spinal Cord Injury Treatment Center, Kunming Tongren Hospital; Clinical Center for Spinal Cord Injury, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| |
Collapse
|
16
|
Boakye M, Morehouse J, Ethridge J, Burke DA, Khattar NK, Kumar C, Manouchehri N, Streijger F, Reed R, Magnuson DS, Sherwood L, Kwon BK, Howland DR. Treadmill-Based Gait Kinematics in the Yucatan Mini Pig. J Neurotrauma 2020; 37:2277-2291. [PMID: 32605423 PMCID: PMC9836690 DOI: 10.1089/neu.2020.7050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Yucatan miniature pigs (YMPs) are similar to humans in spinal cord size as well as physiological and neuroanatomical features, making them a useful model for human spinal cord injury. However, little is known regarding pig gait kinematics, especially on a treadmill. In this study, 12 healthy YMPs were assessed during bipedal and/or quadrupedal stepping on a treadmill at six speeds (1.0, 1.5, 2.0, 2.5, 3.0, and 3.5 km/h). Kinematic parameters, including limb coordination and proximal and distal limb angles, were measured. Findings indicate that YMPs use a lateral sequence footfall pattern across all speeds. Stride and stance durations decreased with increasing speed whereas swing duration showed no significant change. Across all speeds assessed, no significant differences were noted between hindlimb stepping parameters for bipedal or quadrupedal gait with the exception of distal limb angular kinematics. Specifically, significant differences were observed between locomotor tasks during maximum flexion (quadrupedal > bipedal), total excursion (bipedal > quadrupedal), and the phase relationship between the timing of maximum extension between the right and left hindlimbs (bipedal > quadrupedal). Speed also impacted maximum flexion and right-left phase relationships given that significant differences were found between the fastest speed (3.5 km/h) relative to each of the other speeds. This study establishes a methodology for bipedal and quadrupedal treadmill-based kinematic testing in healthy YMPs. The treadmill approach used was effective in recruiting primarily the spinal circuitry responsible for the basic stepping patterns as has been shown in cats. We recommend 2.5 km/h (0.7 m/sec) as a target walking gait for pre-clinical studies using YMPs, which is similar to that used in cats.
Collapse
Affiliation(s)
- Maxwell Boakye
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Johnny Morehouse
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Jay Ethridge
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Darlene A. Burke
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Nicolas K. Khattar
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Chitra Kumar
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Neda Manouchehri
- International Collaboration on Repair Discoveries, Department of Orthopedics, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Femke Streijger
- International Collaboration on Repair Discoveries, Department of Orthopedics, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Robert Reed
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - David S.K. Magnuson
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Leslie Sherwood
- Research Resources Facilities, University of Louisville, Louisville, Kentucky, USA
| | - Brian K. Kwon
- International Collaboration on Repair Discoveries, Department of Orthopedics, University of British Columbia (UBC), Vancouver, British Columbia, Canada
- Vancouver Spine Surgery Institute, Department of Orthopedics, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Dena R. Howland
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Research Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| |
Collapse
|
17
|
Müller HP, Roselli F, Rasche V, Kassubek J. Diffusion Tensor Imaging-Based Studies at the Group-Level Applied to Animal Models of Neurodegenerative Diseases. Front Neurosci 2020; 14:734. [PMID: 32982659 PMCID: PMC7487414 DOI: 10.3389/fnins.2020.00734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
The understanding of human and non-human microstructural brain alterations in the course of neurodegenerative diseases has substantially improved by the non-invasive magnetic resonance imaging (MRI) technique of diffusion tensor imaging (DTI). Animal models (including disease or knockout models) allow for a variety of experimental manipulations, which are not applicable to humans. Thus, the DTI approach provides a promising tool for cross-species cross-sectional and longitudinal investigations of the neurobiological targets and mechanisms of neurodegeneration. This overview with a systematic review focuses on the principles of DTI analysis as used in studies at the group level in living preclinical models of neurodegeneration. The translational aspect from in-vivo animal models toward (clinical) applications in humans is covered as well as the DTI-based research of the non-human brains' microstructure, the methodological aspects in data processing and analysis, and data interpretation at different abstraction levels. The aim of integrating DTI in multiparametric or multimodal imaging protocols will allow the interrogation of DTI data in terms of directional flow of information and may identify the microstructural underpinnings of neurodegeneration-related patterns.
Collapse
Affiliation(s)
| | - Francesco Roselli
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
18
|
Li T, Zhao Z, Wang Y, Xie J, Zhang Y, Bi N, Shi Z, Lu Q, Li Q. A preliminary study of spinal cord blood flow during PVCR with spinal column shortening: A prospective clinic study in severe rigid scoliokyphosis patients. Medicine (Baltimore) 2020; 99:e21579. [PMID: 32769906 PMCID: PMC7593061 DOI: 10.1097/md.0000000000021579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Posterior vertebral column resection (PVCR) was the most powerful technique for treating severe rigid spinal deformity, but it has been plagued with high neurologic deficits risk. The fluctuations of spinal cord blood flow (SCBF) play an important role in secondary spinal cord injury during deformity correction surgery.The objective of this study was to first provide the characteristic of SCBF during PVCR with spinal column shortening in severe rigid spinal deformity.Severe rigid scoliokyphosis patients received PVCR above L1 level were included in this prospective study. Patients with simple kyphosis, intraspinal pathology and any degree of neurologic deficits were excluded. The deformity correction was based on spinal column shortening over the resected gap during PVCR. Laser Doppler flowmetry was used to monitor the SCBF at different surgical stages.There were 12 severe rigid scoliokyphosis patients in the study. The baseline SCBF was 316 ± 86 perfusion unite (PU), and the SCBF decreased to 228 ± 68 PU after VCR (P = .008). The SCBF increased to 296 ± 102 PU after the middle shortening and correction which has a 121% increased comparison to the SCBF after VCR (P = .02). The SCBF will slightly decrease to 271 ± 65 PU at final fixation. The postoperative neural physical examination of all patients was negative, and the MEP and SSEP of all patients did not reach the alarm value during surgery.These results indicate that PVCR is accompanied by a change in SCBF, a proper spinal cord shortening can protect the SCBF and can prevent a secondary spinal cord injury during the surgery.
Collapse
|
19
|
Masood F, Farzana M, Nesathurai S, Abdullah HA. Comparison study of classification methods of intramuscular electromyography data for non-human primate model of traumatic spinal cord injury. Proc Inst Mech Eng H 2020; 234:955-965. [PMID: 32605433 DOI: 10.1177/0954411920935741] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Traumatic spinal cord injury is a serious neurological disorder. Patients experience a plethora of symptoms that can be attributed to the nerve fiber tracts that are compromised. This includes limb weakness, sensory impairment, and truncal instability, as well as a variety of autonomic abnormalities. This article will discuss how machine learning classification can be used to characterize the initial impairment and subsequent recovery of electromyography signals in an non-human primate model of traumatic spinal cord injury. The ultimate objective is to identify potential treatments for traumatic spinal cord injury. This work focuses specifically on finding a suitable classifier that differentiates between two distinct experimental stages (pre-and post-lesion) using electromyography signals. Eight time-domain features were extracted from the collected electromyography data. To overcome the imbalanced dataset issue, synthetic minority oversampling technique was applied. Different ML classification techniques were applied including multilayer perceptron, support vector machine, K-nearest neighbors, and radial basis function network; then their performances were compared. A confusion matrix and five other statistical metrics (sensitivity, specificity, precision, accuracy, and F-measure) were used to evaluate the performance of the generated classifiers. The results showed that the best classifier for the left- and right-side data is the multilayer perceptron with a total F-measure of 79.5% and 86.0% for the left and right sides, respectively. This work will help to build a reliable classifier that can differentiate between these two phases by utilizing some extracted time-domain electromyography features.
Collapse
Affiliation(s)
- Farah Masood
- School of Engineering, University of Guelph, Guelph, ON, Canada.,Department of Biomedical Engineering, Al-Khwarizmi College of Engineering, Baghdad University, Baghdad, Iraq
| | - Maisha Farzana
- School of Engineering, University of Guelph, Guelph, ON, Canada
| | - Shanker Nesathurai
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Division of Physical Medicine and Rehabilitation, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Department of Physical Medicine and Rehabilitation, Hamilton Health Sciences, St Joseph's Hamilton Healthcare, Hamilton, ON, Canada
| | | |
Collapse
|
20
|
Wang X, Zhou T, Maynard GD, Terse PS, Cafferty WB, Kocsis JD, Strittmatter SM. Nogo receptor decoy promotes recovery and corticospinal growth in non-human primate spinal cord injury. Brain 2020; 143:1697-1713. [PMID: 32375169 PMCID: PMC7850069 DOI: 10.1093/brain/awaa116] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/19/2019] [Accepted: 02/23/2020] [Indexed: 12/20/2022] Open
Abstract
After CNS trauma such as spinal cord injury, the ability of surviving neural elements to sprout axons, reorganize neural networks and support recovery of function is severely restricted, contributing to chronic neurological deficits. Among limitations on neural recovery are myelin-associated inhibitors functioning as ligands for neuronal Nogo receptor 1 (NgR1). A soluble decoy (NgR1-Fc, AXER-204) blocks these ligands and provides a means to promote recovery of function in multiple preclinical rodent models of spinal cord injury. However, the safety and efficacy of this reagent in non-human primate spinal cord injury and its toxicological profile have not been described. Here, we provide evidence that chronic intrathecal and intravenous administration of NgR1-Fc to cynomolgus monkey and to rat are without evident toxicity at doses of 20 mg and greater every other day (≥2.0 mg/kg/day), and far greater than the projected human dose. Adult female African green monkeys underwent right C5/6 lateral hemisection with evidence of persistent disuse of the right forelimb during feeding and right hindlimb during locomotion. At 1 month post-injury, the animals were randomized to treatment with vehicle (n = 6) or 0.10-0.17 mg/kg/day of NgR1-Fc (n = 8) delivered via intrathecal lumbar catheter and osmotic minipump for 4 months. One animal was removed from the study because of surgical complications of the catheter, but no treatment-related adverse events were noted in either group. Animal behaviour was evaluated at 6-7 months post-injury, i.e. 1-2 months after treatment cessation. The use of the impaired forelimb during spontaneous feeding and the impaired hindlimb during locomotion were both significantly greater in the treatment group. Tissue collected at 7-12 months post-injury showed no significant differences in lesion size, fibrotic scar, gliosis or neuroinflammation between groups. Serotoninergic raphespinal fibres below the lesion showed no deficit, with equal density on the lesioned and intact side below the level of the injury in both groups. Corticospinal axons traced from biotin-dextran-amine injections in the left motor cortex were equally labelled across groups and reduced caudal to the injury. The NgR1-Fc group tissue exhibited a significant 2-3-fold increased corticospinal axon density in the cervical cord below the level of the injury relative to the vehicle group. The data show that NgR1-Fc does not have preclinical toxicological issues in healthy animals or safety concerns in spinal cord injury animals. Thus, it presents as a potential therapeutic for spinal cord injury with evidence for behavioural improvement and growth of injured pathways in non-human primate spinal cord injury.
Collapse
Affiliation(s)
- Xingxing Wang
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Tianna Zhou
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA
| | | | - Pramod S Terse
- National Center for Translational Sciences, NIH, Rockville, MD, USA
| | - William B Cafferty
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Liu J, Li R, Huang Z, Huang Z, Li Y, Wu X, Lin J, Jiang H, Cheng Y, Kong G, Wu X, Liu Q, Liu Y, Yang Z, Li R, Chen J, Fu J, Ramer MS, Kwon BK, Liu J, Kramer JLK, Tetzlaff W, Hu Y, Zhu Q. A Cervical Spinal Cord Hemi-Contusion Injury Model Based on Displacement Control in Non-Human Primates (Macaca fascicularis). J Neurotrauma 2020; 37:1669-1686. [PMID: 32174266 DOI: 10.1089/neu.2019.6822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Non-human primate (NHP) spinal cord injury (SCI) models can be informative in the evaluation of treatments that show promise in rodent models prior to translation to humans. In the present study, we aimed to establish a cervical spinal hemi-contusion model with controlled displacement and evaluate the abnormalities in behavior, electrophysiology, histology, and magnetic resonance imaging. Twelve adult NHPs were divided into an SCI group (n = 8, 24 and 48 weeks) and a control group (n = 4). An impactor (Φ = 4 mm) was driven to compress the left C5 cord at 800 mm/sec. The contusion displacement and peak force was 4.08 ± 0.17 mm and 19.8 ± 4.6 N. The behavioral assessment showed a consistent dysfunction below the wrist and spontaneous recovery of limb function after injury. Lesion length and lesion area at the epicenter based on T2 hyperintensity were 5.68 ± 0.47 mm and 5.99 ± 0.24 mm2 at 24 weeks post-injury (wpi), and 5.29 ± 0.17 mm and 5.95 ± 0.24 mm2 at 48 wpi. The spared spinal cord area immuno-positive for glial fibrillary acidic protein was significantly reduced, while the staining intensity increased at 24 wpi and 48 wpi, compared with the sham group. Ipsilateral somatosensory and motor evoked potentials were dynamic, increasing in latency and decreasing in amplitude compared with pre-operative values or the contralateral values, and correlated to varying degrees with behavioral outcomes. A shift in size-frequency distribution of sensory neurons of the dorsal root ganglia (DRG) was consistent with a loss of large-diameter cells. The present study demonstrated that the NHP SCI model resulted in consistent unilateral limb dysfunction and potential plasticity in the face of loss of spinal cord and DRG tissue.
Collapse
Affiliation(s)
- Junhao Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rong Li
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zucheng Huang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiping Huang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou, China
| | - Xiaoliang Wu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junyu Lin
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Jiang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongquan Cheng
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ganggang Kong
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiuhua Wu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yapu Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhou Yang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoyao Li
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianting Chen
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Joey Fu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matt S Ramer
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian K Kwon
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Center, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yong Hu
- Department of Orthopedics and Traumatology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Qingan Zhu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Lu A, Baker-Nigh A, Sun P. Operation spinal cord regeneration: Patterning information residing in extracellular matrix glycosaminoglycans. Brain Behav 2020; 10:e01531. [PMID: 31944630 PMCID: PMC7010577 DOI: 10.1002/brb3.1531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Spinal cord injuries are devastating, with many complications beyond paralysis and loss of sensory function. Although spinal cord regeneration can revolutionize treatment for spinal cord injuries, the goal has not yet been achieved. The regenerative mechanism of axolotls demonstrates that the regeneration is a repeat of developmental process that all animals have all the genes, but axolotls have both the genes and the patterning information to do it at the adult stage. METHODS A narrative review was conducted. Relevant studies were collected via an English-language PubMed database search and those known to the authors. RESULTS Research during the past 30 years reveals that growth factors, along with spinal cord extracellular matrix, especially glycosaminoglycans, regulates axonal regrowth. Degrading chondroitin sulfate glycosaminoglycans by injecting the bacterial enzyme chondroitinase improves axonal sprouting and functional recovery after spinal cord injury in both rodents and rhesus monkeys. Furthermore, the brain is one of the first organs to develop during the embryonic period, and heparan sulfate glycosaminoglycans are key molecules required for brain development. CONCLUSIONS Patterning information residing in glycosaminoglycans might be key elements in restricting spinal cord regeneration. A recommended solution is not to edit the human genome, considering the conserved signaling pathways between animals, but to take advantage of the regenerative mechanism of axolotls and the current knowledge about the pattern-forming glycosaminoglycans for successful spinal cord regeneration and clinical applications.
Collapse
Affiliation(s)
- Alexander Lu
- Department of Biology, Saint Louis University, St. Louis, Missouri.,Program in Neuroscience, Saint Louis University, St. Louis, Missouri
| | - Alaina Baker-Nigh
- Department of Biology, Saint Louis University, St. Louis, Missouri.,Program in Neuroscience, Saint Louis University, St. Louis, Missouri
| | - Peng Sun
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Lai B, Che M, Feng B, Bai Y, Li G, Ma Y, Wang L, Huang M, Wang Y, Jiang B, Ding Y, Zeng X, Zeng Y. Tissue-Engineered Neural Network Graft Relays Excitatory Signal in the Completely Transected Canine Spinal Cord. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901240. [PMID: 31763143 PMCID: PMC6864506 DOI: 10.1002/advs.201901240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/19/2019] [Indexed: 05/10/2023]
Abstract
Tissue engineering produces constructs with defined functions for the targeted treatment of damaged tissue. A complete spinal cord injury (SCI) model is generated in canines to test whether in vitro constructed neural network (NN) tissues can relay the excitatory signal across the lesion gap to the caudal spinal cord. Established protocols are used to construct neural stem cell (NSC)-derived NN tissue characterized by a predominantly neuronal population with robust trans-synaptic activities and myelination. The NN tissue is implanted into the gap immediately following complete transection SCI of canines at the T10 spinal cord segment. The data show significant motor recovery of paralyzed pelvic limbs, as evaluated by Olby scoring and cortical motor evoked potential (CMEP) detection. The NN tissue survives in the lesion area with neuronal phenotype maintenance, improves descending and ascending nerve fiber regeneration, and synaptic integration with host neural circuits that allow it to serve as a neuronal relay to transmit excitatory electrical signal across the injured area to the caudal spinal cord. These results suggest that tissue-engineered NN grafts can relay the excitatory signal in the completely transected canine spinal cord, providing a promising strategy for SCI treatment in large animals, including humans.
Collapse
Affiliation(s)
- Bi‐Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Ming‐Tian Che
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
| | - Bo Feng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
| | - Yu‐Rong Bai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
| | - Yuan‐Huan Ma
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
| | - Lai‐Jian Wang
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Meng‐Yao Huang
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Ya‐Qiong Wang
- Department of Electron MicroscopeZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Bin Jiang
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| | - Ying Ding
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
| | - Yuan‐Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat‐sen University)Ministry of EducationGuangzhou510080China
- Department of Histology and EmbryologyZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
- Institute of Spinal Cord InjurySun Yat‐sen UniversityGuangzhou510120China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseZhongshan School of MedicineSun Yat‐sen UniversityGuangzhou510080China
| |
Collapse
|
24
|
Construction of rat spinal cord injury model based on Allen's animal model. Saudi J Biol Sci 2019; 26:2122-2126. [PMID: 31889806 PMCID: PMC6923460 DOI: 10.1016/j.sjbs.2019.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/24/2019] [Accepted: 09/29/2019] [Indexed: 12/11/2022] Open
Abstract
The aim of this study is to explore the construction of rat spinal cord injury model guided by Allen's model. Methods: Male rats aged 4–5 weeks and weighing about 250 g are selected as subjects in the Animal Laboratory Center of XX Hospital. Rats are divided into two groups, which are experimental group 1 and experimental group 2, respectively, so as to construct spinal cord injury model in rats. The first group is given 300 g.cm hitting force of T10 spinal cord, and the second group is given 500 g.cm hitting force of T10 spinal cord. Within 25 days after spinal cord injury in Allen's rats, the survival, neurological function, diet, motor ability, tactile ability and auditory ability of the two groups are monitored and evaluated daily. Results: In terms of survival, the survival rate of rats in group 1 is 85%, while that of rats in group 2 is 21%, and there is a concentrated death phenomenon in group 2. In terms of neurological function recovery, experimental group 1 is stable and gets 7 points and experimental group 2 is stable and gets 3 points. In terms of diet, the experimental group 1 is stable and gets 5 points and the experimental group 2 is stable and gets 2 points. In terms of motor ability, the experimental group 1 is stable and gets 5 points and the experimental group 2 is stable and gets 2 points. In tactile sense, experimental group 1 is stable and gets 17 points and experimental group 2 is stable and gets 12 points. It can be seen that the post-operative recovery ability of the experimental group 1 is better than that of the experimental group 2. Conclusion: Under the guidance of Allen's model, compared with the group 2, the experimental group 1 of the rat spinal cord injury model has better recovery in each index. It can be seen that the smaller impact strength is more beneficial to the recovery of rats after spinal cord injury surgery.
Collapse
|
25
|
Ko CC, Tu TH, Chen YT, Wu JC, Huang WC, Cheng H. Monkey Recovery from Spinal Cord Hemisection: Nerve Repair Strategies for Rhesus Macaques. World Neurosurg 2019; 129:e343-e351. [PMID: 31132502 DOI: 10.1016/j.wneu.2019.05.145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Repair of spinal cord injury (SCI) using peripheral nerve graft (PNG) and acidic fibroblast growth factor (aFGF) has shown promising results in rats and a few human patients, but not in nonhuman primates. The aim of this study was to verify the effective use of PNG and aFGF for repairing incomplete SCI in nonhuman primates. METHODS Six adult rhesus macaques received spinal cord hemisection at T8 level and were grouped into repair and control groups (n = 3 in each). Animals in the repair group underwent nerve repair with autologous PNG plus aFGF immediately after lesioning. The control group received exactly the same operation for lesioning but no treatment. Postoperative behavioral evaluations, electrophysiologic tests (including motor and somatosensory evoked potentials), and magnetic resonance imaging were performed and compared between the 2 groups as well as histologic examination of the spinal cord cephalic to, at, and caudal to the lesion site after sacrifice. RESULTS Animals in the repair group had better motor function in the lower limbs at every observed time point and demonstrated more improvement on electrophysiologic examinations than the control group. The repair group had smaller areas of myelomalacia on magnetic resonance imaging around the lesion compared with the control group, suggesting diminished inflammatory responses with the repair strategy. CONCLUSIONS PNG plus aFGF for SCI in nonhuman primates yielded improvements in clinical behavior, electrophysiologic tests, and magnetic resonance imaging. This study suggests that the repair strategy is feasible and effective for nonhuman primate SCI. Further investigations are warranted to corroborate its effectiveness for clinical application.
Collapse
Affiliation(s)
- Chin-Chu Ko
- Jhong Jheng Spine & Orthopedic Hospital, Kaohsiung, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Hsi Tu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Ya-Tzu Chen
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jau-Ching Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Wen-Cheng Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Henrich Cheng
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
26
|
Gallagher MJ, Hogg FR, Zoumprouli A, Papadopoulos MC, Saadoun S. Spinal Cord Blood Flow in Patients with Acute Spinal Cord Injuries. J Neurotrauma 2019; 36:919-929. [DOI: 10.1089/neu.2018.5961] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mathew J. Gallagher
- Academic Neurosurgery Unit, St. George's, University of London, London, United Kingdom
| | - Florence R.A. Hogg
- Academic Neurosurgery Unit, St. George's, University of London, London, United Kingdom
| | - Argyro Zoumprouli
- Neuro-intensive Care Unit, St. George's Hospital, London, United Kingdom
| | | | - Samira Saadoun
- Academic Neurosurgery Unit, St. George's, University of London, London, United Kingdom
| |
Collapse
|
27
|
Verma R, Virdi JK, Singh N, Jaggi AS. Animals models of spinal cord contusion injury. Korean J Pain 2019; 32:12-21. [PMID: 30671199 PMCID: PMC6333579 DOI: 10.3344/kjp.2019.32.1.12] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 12/03/2022] Open
Abstract
Spinal cord contusion injury is one of the most serious nervous system disorders, characterized by high morbidity and disability. To mimic spinal cord contusion in humans, various animal models of spinal contusion injury have been developed. These models have been developed in rats, mice, and monkeys. However, most of these models are developed using rats. Two types of animal models, i.e. bilateral contusion injury and unilateral contusion injury models, are developed using either a weight drop method or impactor method. In the weight drop method, a specific weight or a rod, having a specific weight and diameter, is dropped from a specific height on to the exposed spinal cord. Low intensity injury is produced by dropping a 5 g weight from a height of 8 cm, moderate injury by dropping 10 g weight from a height of 12.5–25 mm, and high intensity injury by dropping a 25 g weight from a height of 50 mm. In the impactor method, injury is produced through an impactor by delivering a specific force to the exposed spinal cord area. Mild injury is produced by delivering 100 ± 5 kdyn of force, moderate injury by delivering 200 ± 10 kdyn of force, and severe injury by delivering 300 ± 10 kdyn of force. The contusion injury produces a significant development of locomotor dysfunction, which is generally evident from the 0–14th day of surgery and is at its peak after the 28–56th day. The present review discusses different animal models of spinal contusion injury.
Collapse
Affiliation(s)
- Renuka Verma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| | - Jasleen Kaur Virdi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| |
Collapse
|
28
|
Baklaushev VP, Durov OV, Kim SV, Gulaev EV, Gubskiy IL, Konoplyannikov MA, Zabozlaev FG, Zhang C, Agrba VZ, Orlov SV, Lapin BA, Troitskiy AV, Averyanov AV, Ahlfors JE. Development of a motor and somatosensory evoked potentials-guided spinal cord Injury model in non-human primates. J Neurosci Methods 2018; 311:200-214. [PMID: 30393204 DOI: 10.1016/j.jneumeth.2018.10.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
Abstract
Background Nonhuman primates (NHP) may provide the most adequate (in terms of neuroanatomy and neurophysiology) model of spinal cord injury (SCI) for testing regenerative therapies, but bioethical considerations exclude their use in severe SCI. New Method A reproducible model of SCI at the lower thoracic level has been developed in Rhesus macaques. The model comprises surgical resection of 25% of the spinal cord in the projection of the dorsal funiculus and dorsolateral corticospinal pathways, controlled via registration of intraoperative evoked potentials (EPs). The animals were evaluated using the modified Hindlimb score, MRI, SSEP, and MEP over a time period of 8-12 weeks post-SCI, followed by histological examination. Results Complete disappearance of intraoperative EPs from distal hindlimb muscles without restoration within two weeks post-SCI was an indicator for irreversible disruption of the abovementioned pathways. As a result, controlled damage to the spinal cord was achieved in three NHPs, clinically manifested as irreversible lower monoplegia. No significant functional restoration was observed in these NHPs up to 12 weeks post-SCI. Demyelination of the damaged ascending tracts was detected. Disturbances in pelvic organ function were not observed in all animals. Comparison with existing methods The new method of EPs-guided SCI allows a more controlled and irreversible damage to the spinal cord compared with contusion and other transection approaches. Conclusions This method to induce complete SCI in NHP is well tolerated, reproducible and ethically acceptable: these are valuable attributes in a preclinical model that will hopefully help advance testing of new regenerative therapies in SCI.
Collapse
Affiliation(s)
- V P Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia; Institute for Advanced Training, FMBA, Moscow, Russia.
| | - O V Durov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia
| | - S V Kim
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia
| | - E V Gulaev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia
| | - I L Gubskiy
- Research and Education Center for Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - M A Konoplyannikov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia; Institute of Regenerative Medicine, Sechenov Medical University, Moscow, Russia
| | - F G Zabozlaev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia
| | - C Zhang
- Research and Education Center for Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia; Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - V Z Agrba
- Institute of Medicinal Primatology Russian Academy of Science, Sochi, Russia
| | - S V Orlov
- Institute of Medicinal Primatology Russian Academy of Science, Sochi, Russia
| | - B A Lapin
- Institute of Medicinal Primatology Russian Academy of Science, Sochi, Russia
| | - A V Troitskiy
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia; Institute for Advanced Training, FMBA, Moscow, Russia
| | - A V Averyanov
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA, 28 Orekhovy Blvd., 115682 Moscow, Russia
| | | |
Collapse
|
29
|
Le Corre M, Noristani HN, Mestre-Frances N, Saint-Martin GP, Coillot C, Goze-Bac C, Lonjon N, Perrin FE. A Novel Translational Model of Spinal Cord Injury in Nonhuman Primate. Neurotherapeutics 2018; 15:751-769. [PMID: 29181770 PMCID: PMC6095780 DOI: 10.1007/s13311-017-0589-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injuries (SCI) lead to major disabilities affecting > 2.5 million people worldwide. Major shortcomings in clinical translation result from multiple factors, including species differences, development of moderately predictive animal models, and differences in methodologies between preclinical and clinical studies. To overcome these obstacles, we first conducted a comparative neuroanatomical analysis of the spinal cord between mice, Microcebus murinus (a nonhuman primate), and humans. Next, we developed and characterized a new model of lateral spinal cord hemisection in M. murinus. Over a 3-month period after SCI, we carried out a detailed, longitudinal, behavioral follow-up associated with in vivo magnetic resonance imaging (1H-MRI) monitoring. Then, we compared lesion extension and tissue alteration using 3 methods: in vivo 1H-MRI, ex vivo 1H-MRI, and classical histology. The general organization and glial cell distribution/morphology in the spinal cord of M. murinus closely resembles that of humans. Animals assessed at different stages following lateral hemisection of the spinal cord presented specific motor deficits and spinal cord tissue alterations. We also found a close correlation between 1H-MRI signal and microglia reactivity and/or associated post-trauma phenomena. Spinal cord hemisection in M. murinus provides a reliable new nonhuman primate model that can be used to promote translational research on SCI and represents a novel and more affordable alternative to larger primates.
Collapse
Affiliation(s)
- Marine Le Corre
- INSERM U1051, Rue Augustin Fliche, F-34095, Montpellier Cedex 5, France
- CHRU Montpellier, Gui de Chauliac Hospital, F-34095, Montpellier, France
| | - Harun N Noristani
- INSERM U1051, Rue Augustin Fliche, F-34095, Montpellier Cedex 5, France
- INSERM U1198, University of Montpellier, EPHE, Place Eugène Bataillon CC105, F-34095, Montpellier, France
| | - Nadine Mestre-Frances
- INSERM U1198, University of Montpellier, EPHE, PSL Research University, Place Eugène Bataillon CC105, F-34095, Montpellier, France
| | - Guillaume P Saint-Martin
- INSERM U1198, University of Montpellier, EPHE, Place Eugène Bataillon CC105, F-34095, Montpellier, France
- CNRS UMR 5221, University of Montpellier, Place Eugène Bataillon, F-34095, Montpellier, France
| | - Christophe Coillot
- CNRS UMR 5221, University of Montpellier, Place Eugène Bataillon, F-34095, Montpellier, France
| | - Christophe Goze-Bac
- CNRS UMR 5221, University of Montpellier, Place Eugène Bataillon, F-34095, Montpellier, France
| | - Nicolas Lonjon
- CHRU Montpellier, Gui de Chauliac Hospital, F-34095, Montpellier, France
- INSERM U1198, University of Montpellier, EPHE, Place Eugène Bataillon CC105, F-34095, Montpellier, France
| | - Florence E Perrin
- INSERM U1051, Rue Augustin Fliche, F-34095, Montpellier Cedex 5, France.
- INSERM U1198, University of Montpellier, EPHE, Place Eugène Bataillon CC105, F-34095, Montpellier, France.
| |
Collapse
|
30
|
Kroehne V, Tsata V, Marrone L, Froeb C, Reinhardt S, Gompf A, Dahl A, Sterneckert J, Reimer MM. Primary Spinal OPC Culture System from Adult Zebrafish to Study Oligodendrocyte Differentiation In Vitro. Front Cell Neurosci 2017; 11:284. [PMID: 28959189 PMCID: PMC5603699 DOI: 10.3389/fncel.2017.00284] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/31/2017] [Indexed: 01/24/2023] Open
Abstract
Endogenous oligodendrocyte progenitor cells (OPCs) are a promising target to improve functional recovery after spinal cord injury (SCI) by remyelinating denuded, and therefore vulnerable, axons. Demyelination is the result of a primary insult and secondary injury, leading to conduction blocks and long-term degeneration of the axons, which subsequently can lead to the loss of their neurons. In response to SCI, dormant OPCs can be activated and subsequently start to proliferate and differentiate into mature myelinating oligodendrocytes (OLs). Therefore, researchers strive to control OPC responses, and utilize small molecule screening approaches in order to identify mechanisms of OPC activation, proliferation, migration and differentiation. In zebrafish, OPCs remyelinate axons of the optic tract after lysophosphatidylcholine (LPC)-induced demyelination back to full thickness myelin sheaths. In contrast to zebrafish, mammalian OPCs are highly vulnerable to excitotoxic stress, a cause of secondary injury, and remyelination remains insufficient. Generally, injury induced remyelination leads to shorter internodes and thinner myelin sheaths in mammals. In this study, we show that myelin sheaths are lost early after a complete spinal transection injury, but are re-established within 14 days after lesion. We introduce a novel, easy-to-use, inexpensive and highly reproducible OPC culture system based on dormant spinal OPCs from adult zebrafish that enables in vitro analysis. Zebrafish OPCs are robust, can easily be purified with high viability and taken into cell culture. This method enables to examine why zebrafish OPCs remyelinate better than their mammalian counterparts, identify cell intrinsic responses, which could lead to pro-proliferating or pro-differentiating strategies, and to test small molecule approaches. In this methodology paper, we show efficient isolation of OPCs from adult zebrafish spinal cord and describe culture conditions that enable analysis up to 10 days in vitro. Finally, we demonstrate that zebrafish OPCs differentiate into Myelin Basic Protein (MBP)-expressing OLs when co-cultured with human motor neurons differentiated from induced pluripotent stem cells (iPSCs). This shows that the basic mechanisms of oligodendrocyte differentiation are conserved across species and that understanding the regulation of zebrafish OPCs can contribute to the development of new treatments to human diseases.
Collapse
Affiliation(s)
- Volker Kroehne
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Vasiliki Tsata
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Lara Marrone
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Claudia Froeb
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Susanne Reinhardt
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany.,Deep Sequencing Group, Center for Molecular and Cellular Bioengineering (CMCB), Biotechnologisches Zentrum (BIOTEC), Technische Universität DresdenDresden, Germany
| | - Anne Gompf
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Andreas Dahl
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany.,Deep Sequencing Group, Center for Molecular and Cellular Bioengineering (CMCB), Biotechnologisches Zentrum (BIOTEC), Technische Universität DresdenDresden, Germany
| | - Jared Sterneckert
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| | - Michell M Reimer
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität DresdenDresden, Germany
| |
Collapse
|
31
|
Wang-Leandro A, Hobert MK, Alisauskaite N, Dziallas P, Rohn K, Stein VM, Tipold A. Spontaneous acute and chronic spinal cord injuries in paraplegic dogs: a comparative study of in vivo diffusion tensor imaging. Spinal Cord 2017; 55:1108-1116. [DOI: 10.1038/sc.2017.83] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 06/12/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022]
|
32
|
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: 5.0] [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
|
33
|
Wu X, Zhang YP, Qu W, Shields LBE, Shields CB, Xu XM. A Tissue Displacement-based Contusive Spinal Cord Injury Model in Mice. J Vis Exp 2017. [PMID: 28654063 PMCID: PMC5608444 DOI: 10.3791/54988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Producing a consistent and reproducible contusive spinal cord injury (SCI) is critical to minimizing behavioral and histological variabilities between experimental animals. Several contusive SCI models have been developed to produce injuries using different mechanisms. The severity of the SCI is based on the height that a given weight is dropped, the injury force, or the spinal cord displacement. In the current study, we introduce a novel mouse contusive SCI device, the Louisville Injury System Apparatus (LISA) impactor, which can create a displacement-based SCI with high injury velocity and accuracy. This system utilizes laser distance sensors combined with advanced software to produce graded and highly-reproducible injuries. We performed a contusive SCI at the 10th thoracic vertebral (T10) level in mice to demonstrate the step-by-step procedure. The model can also be applied to the cervical and lumbar spinal levels.
Collapse
Affiliation(s)
- Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman and Campbell Brain and Spine, Indiana University School of Medicine
| | | | - Wenrui Qu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman and Campbell Brain and Spine, Indiana University School of Medicine
| | | | - Christopher B Shields
- Norton Neuroscience Institute, Norton Healthcare; Department of Anatomical Sciences and Neurobiology, University of Louisville;
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery and Goodman and Campbell Brain and Spine, Indiana University School of Medicine;
| |
Collapse
|
34
|
Moore SA, Granger N, Olby NJ, Spitzbarth I, Jeffery ND, Tipold A, Nout-Lomas YS, da Costa RC, Stein VM, Noble-Haeusslein LJ, Blight AR, Grossman RG, Basso DM, Levine JM. Targeting Translational Successes through CANSORT-SCI: Using Pet Dogs To Identify Effective Treatments for Spinal Cord Injury. J Neurotrauma 2017; 34:2007-2018. [PMID: 28230415 DOI: 10.1089/neu.2016.4745] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Translation of therapeutic interventions for spinal cord injury (SCI) from laboratory to clinic has been historically challenging, highlighting the need for robust models of injury that more closely mirror the human condition. The high prevalence of acute, naturally occurring SCI in pet dogs provides a unique opportunity to evaluate expeditiously promising interventions in a population of animals that receive diagnoses and treatment clinically in a manner similar to persons with SCI, while adhering to National Institutes of Health guidelines for scientific rigor and transparent reporting. In addition, pet dogs with chronic paralysis are often maintained long-term by their owners, offering a similarly unique population for study of chronic SCI. Despite this, only a small number of studies have used the clinical dog model of SCI. The Canine Spinal Cord Injury Consortium (CANSORT-SCI) was recently established by a group of veterinarians and basic science researchers to promote the value of the canine clinical model of SCI. The CANSORT-SCI group held an inaugural meeting November 20 and 21, 2015 to evaluate opportunities and challenges to the use of pet dogs in SCI research. Key challenges identified included lack of familiarity with the model among nonveterinary scientists and questions about how and where in the translational process the canine clinical model would be most valuable. In light of these, we review the natural history, outcome, and available assessment tools associated with canine clinical SCI with emphasis on their relevance to human SCI and the translational process.
Collapse
Affiliation(s)
- Sarah A Moore
- 1 Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine , Columbus Ohio.,2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI)
| | - Nicolas Granger
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,3 Faculty of Health Sciences, University of Bristol , Langford, North Somerset, United Kingdom
| | - Natasha J Olby
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,4 Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh, North Carolina.,5 Comparative Medicine Institute, North Carolina State University , Raleigh, North Carolina
| | - Ingo Spitzbarth
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,6 Department of Pathology, University of Veterinary Medicine , Hannover, Germany .,7 Center for Systems Neuroscience , Hannover, Germany
| | - Nick D Jeffery
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,8 Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University , College Station, Texas
| | - Andrea Tipold
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,7 Center for Systems Neuroscience , Hannover, Germany.,9 Department of Small Animal Medicine and Surgery, University of Veterinary Medicine , Hannover, Germany
| | - Yvette S Nout-Lomas
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,10 College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Fort Collins, Colorado
| | - Ronaldo C da Costa
- 1 Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine , Columbus Ohio.,2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI)
| | - Veronika M Stein
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,11 Department of Clinical Veterinary Sciences, University of Bern , Bern, Switzerland
| | - Linda J Noble-Haeusslein
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,12 Departments of Physical Therapy and Rehabilitation Sciences and Neurological Surgery, University of California , San Francisco, San Francisco, California
| | - Andrew R Blight
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,13 Acorda Therapeutics, Inc. Ardsley, New York
| | - Robert G Grossman
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,14 Department of Neurosurgery, Houston Methodist Neurological Institute , Houston, Texas
| | - D Michele Basso
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,15 School of Health and Rehabilitation Sciences, The Ohio State University , Columbus, Ohio
| | - Jonathan M Levine
- 2 The Canine Spinal Cord Injury Consortium (CANSORT-SCI).,8 Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University , College Station, Texas
| |
Collapse
|