101
|
Generating level-dependent models of cervical and thoracic spinal cord injury: Exploring the interplay of neuroanatomy, physiology, and function. Neurobiol Dis 2017; 105:194-212. [PMID: 28578003 DOI: 10.1016/j.nbd.2017.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/10/2017] [Accepted: 05/29/2017] [Indexed: 01/01/2023] Open
Abstract
The majority of spinal cord injuries (SCI) occur at the cervical level, which results in significant impairment. Neurologic level and severity of injury are primary endpoints in clinical trials; however, how level-specific damages relate to behavioural performance in cervical injury is incompletely understood. We hypothesized that ascending level of injury leads to worsening forelimb performance, and correlates with loss of neural tissue and muscle-specific neuron pools. A direct comparison of multiple models was made with injury realized at the C5, C6, C7 and T7 vertebral levels using clip compression with sham-operated controls. Animals were assessed for 10weeks post-injury with numerous (40) outcome measures, including: classic behavioural tests, CatWalk, non-invasive MRI, electrophysiology, histologic lesion morphometry, neuron counts, and motor compartment quantification, and multivariate statistics on the total dataset. Histologic staining and T1-weighted MR imaging revealed similar structural changes and distinct tissue loss with cystic cavitation across all injuries. Forelimb tests, including grip strength, F-WARP motor scale, Inclined Plane, and forelimb ladder walk, exhibited stratification between all groups and marked impairment with C5 and C6 injuries. Classic hindlimb tests including BBB, hindlimb ladder walk, bladder recovery, and mortality were not different between cervical and thoracic injuries. CatWalk multivariate gait analysis showed reciprocal and progressive changes forelimb and hindlimb function with ascending level of injury. Electrophysiology revealed poor forelimb axonal conduction in cervical C5 and C6 groups alone. The cervical enlargement (C5-T2) showed progressive ventral horn atrophy and loss of specific motor neuron populations with ascending injury. Multivariate statistics revealed a robust dataset, rank-order contribution of outcomes, and allowed prediction of injury level with single-level discrimination using forelimb performance and neuron counts. Level-dependent models were generated using clip-compression SCI, with marked and reliable differences in forelimb performance and specific neuron pool loss.
Collapse
|
102
|
Callahan A, Anderson KD, Beattie MS, Bixby JL, Ferguson AR, Fouad K, Jakeman LB, Nielson JL, Popovich PG, Schwab JM, Lemmon VP. Developing a data sharing community for spinal cord injury research. Exp Neurol 2017; 295:135-143. [PMID: 28576567 DOI: 10.1016/j.expneurol.2017.05.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/24/2017] [Accepted: 05/29/2017] [Indexed: 01/20/2023]
Abstract
The rapid growth in data sharing presents new opportunities across the spectrum of biomedical research. Global efforts are underway to develop practical guidance for implementation of data sharing and open data resources. These include the recent recommendation of 'FAIR Data Principles', which assert that if data is to have broad scientific value, then digital representations of that data should be Findable, Accessible, Interoperable and Reusable (FAIR). The spinal cord injury (SCI) research field has a long history of collaborative initiatives that include sharing of preclinical research models and outcome measures. In addition, new tools and resources are being developed by the SCI research community to enhance opportunities for data sharing and access. With this in mind, the National Institute of Neurological Disorders and Stroke (NINDS) at the National Institutes of Health (NIH) hosted a workshop on October 5-6, 2016 in Bethesda, MD, in collaboration with the Open Data Commons for Spinal Cord Injury (ODC-SCI) titled "Preclinical SCI Data: Creating a FAIR Share Community". Workshop invitees were nominated by the workshop steering committee (co-chairs: ARF and VPL; members: AC, KDA, MSB, KF, LBJ, PGP, JMS), to bring together junior and senior level experts including preclinical and basic SCI researchers from academia and industry, data science and bioinformatics experts, investigators with expertise in other neurological disease fields, clinical researchers, members of the SCI community, and program staff representing federal and private funding agencies. The workshop and ODC-SCI efforts were sponsored by the International Spinal Research Trust (ISRT), the Rick Hansen Institute, Wings for Life, the Craig H. Neilsen Foundation and NINDS. The number of attendees was limited to ensure active participation and feedback in small groups. The goals were to examine the current landscape for data sharing in SCI research and provide a path to its future. Below are highlights from the workshop, including perspectives on the value of data sharing in SCI research, workshop participant perspectives and concerns, descriptions of existing resources and actionable directions for further engaging the SCI research community in a model that may be applicable to many other areas of neuroscience. This manuscript is intended to share these initial findings with the broader research community, and to provide talking points for continued feedback from the SCI field, as it continues to move forward in the age of data sharing.
Collapse
Affiliation(s)
- Alison Callahan
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford 94305, CA, USA.
| | - Kim D Anderson
- Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami 33136, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami 33136, FL, USA
| | - Michael S Beattie
- UCSF Brain and Spinal Injury Center, University of California, San Francisco 94110, CA, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami 33136, FL, USA; Center for Computational Science, University of Miami, Coral Gables 33146, FL, USA; Department of Cellular and Molecular Pharmacology, University of Miami School of Medicine, Miami 33136, FL, USA
| | - Adam R Ferguson
- UCSF Brain and Spinal Injury Center, University of California, San Francisco 94110, CA, USA; San Francisco VA Medical Center, San Francisco 94121, CA, USA
| | - Karim Fouad
- Department of Physical Therapy, Neuroscience and Mental Health Institute, University of Alberta, Edmonton T6G2G4, Alberta, Canada
| | - Lyn B Jakeman
- National Institute of Neurological Disorders and Stroke, The National Institutes of Health, Rockville 20852, MD, USA
| | - Jessica L Nielson
- UCSF Brain and Spinal Injury Center, University of California, San Francisco 94110, CA, USA; San Francisco VA Medical Center, San Francisco 94121, CA, USA
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, The Neurological Institute, Ohio State University Wexner Medical Center, Columbus 43210, OH, USA; Department of Neuroscience, The Neurological Institute, Ohio State University Wexner Medical Center, Columbus 43210, OH, USA
| | - Jan M Schwab
- Department of Neuroscience, The Neurological Institute, Ohio State University Wexner Medical Center, Columbus 43210, OH, USA; Department of Neurology, The Neurological Institute, Ohio State University Wexner Medical Center, Columbus 43210, OH, USA; Department of Physical Medicine and Rehabilitation, The Neurological Institute, Ohio State University Wexner Medical Center, Columbus 43210, OH, USA
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami 33136, FL, USA; Center for Computational Science, University of Miami, Coral Gables 33146, FL, USA.
| | | |
Collapse
|
103
|
Noristani HN, Gerber YN, Sabourin JC, Le Corre M, Lonjon N, Mestre-Frances N, Hirbec HE, Perrin FE. RNA-Seq Analysis of Microglia Reveals Time-Dependent Activation of Specific Genetic Programs following Spinal Cord Injury. Front Mol Neurosci 2017; 10:90. [PMID: 28420963 PMCID: PMC5376598 DOI: 10.3389/fnmol.2017.00090] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/15/2017] [Indexed: 12/18/2022] Open
Abstract
Neurons have inherent competence to regrow following injury, although not spontaneously. Spinal cord injury (SCI) induces a pronounced neuroinflammation driven by resident microglia and infiltrating peripheral macrophages. Microglia are the first reactive glial population after SCI and participate in recruitment of monocyte-derived macrophages to the lesion site. Both positive and negative influence of microglia and macrophages on axonal regeneration had been reported after SCI, raising the issue whether their response depends on time post-lesion or different lesion severity. We analyzed molecular alterations in microglia at several time-points after different SCI severities using RNA-sequencing. We demonstrate that activation of microglia is time-dependent post-injury but is independent of lesion severity. Early transcriptomic response of microglia after SCI involves proliferation and neuroprotection, which is then switched to neuroinflammation at later stages. Moreover, SCI induces an autologous microglial expression of astrocytic markers with over 6% of microglia expressing glial fibrillary acidic protein and vimentin from as early as 72 h post-lesion and up to 6 weeks after injury. We also identified the potential involvement of DNA damage and in particular tumor suppressor gene breast cancer susceptibility gene 1 (Brca1) in microglia after SCI. Finally, we established that BRCA1 protein is specifically expressed in non-human primate spinal microglia and is upregulated after SCI. Our data provide the first transcriptomic analysis of microglia at multiple stages after different SCI severities. Injury-induced microglia expression of astrocytic markers at RNA and protein levels demonstrates novel insights into microglia plasticity. Finally, increased microglia expression of BRCA1 in rodents and non-human primate model of SCI, suggests the involvement of oncogenic proteins after CNS lesion.
Collapse
Affiliation(s)
- Harun N Noristani
- MMDN, University of Montpellier; EPHE, Institut National de la Santé et de la Recherche Médicale U1198Montpellier, France.,Institut National de la Santé et de la Recherche Médicale U1051Montpellier, France
| | - Yannick N Gerber
- MMDN, University of Montpellier; EPHE, Institut National de la Santé et de la Recherche Médicale U1198Montpellier, France.,Institut National de la Santé et de la Recherche Médicale U1051Montpellier, France.,"Integrative Biology of Neurodegeneration", IKERBASQUE Basque Foundation for Science and Neuroscience Department, University of the Basque CountryBilbao, Spain
| | - Jean-Charles Sabourin
- "Integrative Biology of Neurodegeneration", IKERBASQUE Basque Foundation for Science and Neuroscience Department, University of the Basque CountryBilbao, Spain
| | - Marine Le Corre
- Institut National de la Santé et de la Recherche Médicale U1051Montpellier, France.,Department of Neurosurgery, Gui de Chauliac HospitalMontpellier, France
| | - Nicolas Lonjon
- MMDN, University of Montpellier; EPHE, Institut National de la Santé et de la Recherche Médicale U1198Montpellier, France.,Department of Neurosurgery, Gui de Chauliac HospitalMontpellier, France
| | - Nadine Mestre-Frances
- MMDN, University of Montpellier; EPHE, Institut National de la Santé et de la Recherche Médicale U1198Montpellier, France
| | - Hélène E Hirbec
- Institute for Functional Genomics, CNRS UMR5203, Institut National de la Santé et de la Recherche Médicale U1191Montpellier, France
| | - Florence E Perrin
- MMDN, University of Montpellier; EPHE, Institut National de la Santé et de la Recherche Médicale U1198Montpellier, France.,Institut National de la Santé et de la Recherche Médicale U1051Montpellier, France.,"Integrative Biology of Neurodegeneration", IKERBASQUE Basque Foundation for Science and Neuroscience Department, University of the Basque CountryBilbao, Spain
| |
Collapse
|
104
|
Haefeli J, Ferguson AR, Bingham D, Orr A, Won SJ, Lam TI, Shi J, Hawley S, Liu J, Swanson RA, Massa SM. A data-driven approach for evaluating multi-modal therapy in traumatic brain injury. Sci Rep 2017; 7:42474. [PMID: 28205533 PMCID: PMC5311970 DOI: 10.1038/srep42474] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/11/2017] [Indexed: 01/22/2023] Open
Abstract
Combination therapies targeting multiple recovery mechanisms have the potential for additive or synergistic effects, but experimental design and analyses of multimodal therapeutic trials are challenging. To address this problem, we developed a data-driven approach to integrate and analyze raw source data from separate pre-clinical studies and evaluated interactions between four treatments following traumatic brain injury. Histologic and behavioral outcomes were measured in 202 rats treated with combinations of an anti-inflammatory agent (minocycline), a neurotrophic agent (LM11A-31), and physical therapy consisting of assisted exercise with or without botulinum toxin-induced limb constraint. Data was curated and analyzed in a linked workflow involving non-linear principal component analysis followed by hypothesis testing with a linear mixed model. Results revealed significant benefits of the neurotrophic agent LM11A-31 on learning and memory outcomes after traumatic brain injury. In addition, modulations of LM11A-31 effects by co-administration of minocycline and by the type of physical therapy applied reached statistical significance. These results suggest a combinatorial effect of drug and physical therapy interventions that was not evident by univariate analysis. The study designs and analytic techniques applied here form a structured, unbiased, internally validated workflow that may be applied to other combinatorial studies, both in animals and humans.
Collapse
Affiliation(s)
- Jenny Haefeli
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, United States
| | - Adam R. Ferguson
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, United States
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Deborah Bingham
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Adrienne Orr
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Seok Joon Won
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Tina I. Lam
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Jian Shi
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Sarah Hawley
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Jialing Liu
- Brain and Spinal Injury Center (BASIC), Department of Neurological Surgery, University of California, San Francisco, CA, United States
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| | - Raymond A. Swanson
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Stephen M. Massa
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Neurology, University of California, San Francisco, CA, United States
| |
Collapse
|
105
|
Schomberg DT, Miranpuri GS, Chopra A, Patel K, Meudt JJ, Tellez A, Resnick DK, Shanmuganayagam D. Translational Relevance of Swine Models of Spinal Cord Injury. J Neurotrauma 2017; 34:541-551. [DOI: 10.1089/neu.2016.4567] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Dominic T. Schomberg
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| | - Gurwattan S. Miranpuri
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Abhishek Chopra
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kush Patel
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jennifer J. Meudt
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| | | | - Daniel K. Resnick
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Dhanansayan Shanmuganayagam
- Biomedical and Genomic Research Group, Department of Animal Sciences, University of Wisconsin–Madison, Wisconsin
| |
Collapse
|
106
|
Slotkin JR, Pritchard CD, Luque B, Ye J, Layer RT, Lawrence MS, O'Shea TM, Roy RR, Zhong H, Vollenweider I, Edgerton VR, Courtine G, Woodard EJ, Langer R. Biodegradable scaffolds promote tissue remodeling and functional improvement in non-human primates with acute spinal cord injury. Biomaterials 2017; 123:63-76. [PMID: 28167393 DOI: 10.1016/j.biomaterials.2017.01.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 12/08/2016] [Accepted: 01/22/2017] [Indexed: 12/30/2022]
Abstract
Tissue loss significantly reduces the potential for functional recovery after spinal cord injury. We previously showed that implantation of porous scaffolds composed of a biodegradable and biocompatible block copolymer of Poly-lactic-co-glycolic acid and Poly-l-lysine improves functional recovery and reduces spinal cord tissue injury after spinal cord hemisection injury in rats. Here, we evaluated the safety and efficacy of porous scaffolds in non-human Old-World primates (Chlorocebus sabaeus) after a partial and complete lateral hemisection of the thoracic spinal cord. Detailed analyses of kinematics and muscle activity revealed that by twelve weeks after injury fully hemisected monkeys implanted with scaffolds exhibited significantly improved recovery of locomotion compared to non-implanted control animals. Twelve weeks after injury, histological analysis demonstrated that the spinal cords of monkeys with a hemisection injury implanted with scaffolds underwent appositional healing characterized by a significant increase in remodeled tissue in the region of the hemisection compared to non-implanted controls. The number of glial fibrillary acidic protein immunopositive astrocytes was diminished within the inner regions of the remodeled tissue layer in treated animals. Activated macrophage and microglia were present diffusely throughout the remodeled tissue and concentrated at the interface between the preserved spinal cord tissue and the remodeled tissue layer. Numerous unphosphorylated neurofilament H and neuronal growth associated protein positive fibers and myelin basic protein positive cells may indicate neural sprouting inside the remodeled tissue layer of treated monkeys. These results support the safety and efficacy of polymer scaffolds in a primate model of acute spinal cord injury. A device substantially similar to the device described here is the subject of an ongoing human clinical trial.
Collapse
Affiliation(s)
| | - Christopher D Pritchard
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brian Luque
- InVivo Therapeutics Corporation, Cambridge, MA, USA
| | - Janice Ye
- InVivo Therapeutics Corporation, Cambridge, MA, USA
| | | | | | - Timothy M O'Shea
- Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roland R Roy
- Brain Research Institute, University of California, Los Angeles, CA, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Hui Zhong
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Isabel Vollenweider
- Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - V Reggie Edgerton
- Brain Research Institute, University of California, Los Angeles, CA, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA; Departments of Neurobiology and Neurology, University of California, Los Angeles, CA, USA
| | - Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Eric J Woodard
- Department of Neurosurgery, New England Baptist Hospital, Boston, MA, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
107
|
Animal models of spinal cord injury: a systematic review. Spinal Cord 2017; 55:714-721. [PMID: 28117332 DOI: 10.1038/sc.2016.187] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 11/08/2016] [Accepted: 11/27/2016] [Indexed: 02/06/2023]
Abstract
STUDY DESIGN PRISMA-guided systematic review. OBJECTIVES To provide a comprehensive framework of the current animal models for investigating spinal cord injury (SCI) and categorize them based on the aims, patterns and levels of injury, and outcome measurements as well as animal species. SETTING Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran. METHODS An electronic search of the Medline database for literature describing animal models of SCI was performed on 1 January 2016 using the following keywords: 'spinal cord injuries' and 'animal models'. The search retrieved 2870 articles. Reviews and non-original articles were excluded. Data extraction was independently performed by two reviewers. RESULTS Among the 2209 included studies, testing the effects of drug's or growth factor's interventions was the most common aim (36.6%) followed by surveying pathophysiologic changes (30.2%). The most common spinal region involved was thoracic (81%). Contusion was the most common pattern of injury (41%) followed by transection (32.5%) and compression (19.4%). The most common species involved in animal models of SCI was the rat (72.4%). Two or more types of outcome assessments were used in the majority of the studies, and the most common assessment method was biological plus behavioral (50.8%). CONCLUSIONS Prior to choosing an animal model, the objectives of the proposed study must precisely be defined. Contusion and compression models better simulate the biomechanics and neuropathology of human injury, whereas transection models are valuable to study anatomic regeneration. Rodents are the most common and probably best-suited species for preliminary SCI studies.
Collapse
|
108
|
Mosberger AC, Miehlbradt JC, Bjelopoljak N, Schneider MP, Wahl AS, Ineichen BV, Gullo M, Schwab ME. Axotomized Corticospinal Neurons Increase Supra-Lesional Innervation and Remain Crucial for Skilled Reaching after Bilateral Pyramidotomy. Cereb Cortex 2017; 28:625-643. [DOI: 10.1093/cercor/bhw405] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/20/2016] [Indexed: 12/11/2022] Open
|
109
|
Contribution of propriospinal neurons to recovery of hand dexterity after corticospinal tract lesions in monkeys. Proc Natl Acad Sci U S A 2017; 114:604-609. [PMID: 28049844 DOI: 10.1073/pnas.1610787114] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The direct cortico-motoneuronal connection is believed to be essential for the control of dexterous hand movements, such as precision grip in primates. It was reported, however, that even after lesion of the corticospinal tract (CST) at the C4-C5 segment, precision grip largely recovered within 1-3 mo, suggesting that the recovery depends on transmission through intercalated neurons rostral to the lesion, such as the propriospinal neurons (PNs) in the midcervical segments. To obtain direct evidence for the contribution of PNs to recovery after CST lesion, we applied a pathway-selective and reversible blocking method using double viral vectors to the PNs in six monkeys after CST lesions at C4-C5. In four monkeys that showed nearly full or partial recovery, transient blockade of PN transmission after recovery caused partial impairment of precision grip. In the other two monkeys, CST lesions were made under continuous blockade of PN transmission that outlasted the entire period of postoperative observation (3-4.5 mo). In these monkeys, precision grip recovery was not achieved. These results provide evidence for causal contribution of the PNs to recovery of hand dexterity after CST lesions; PN transmission is necessary for promoting the initial stage recovery; however, their contribution is only partial once the recovery is achieved.
Collapse
|
110
|
Capogrosso M, Milekovic T, Borton D, Wagner F, Moraud EM, Mignardot JB, Buse N, Gandar J, Barraud Q, Xing D, Rey E, Duis S, Jianzhong Y, Ko WKD, Li Q, Detemple P, Denison T, Micera S, Bezard E, Bloch J, Courtine G. A brain-spine interface alleviating gait deficits after spinal cord injury in primates. Nature 2016; 539:284-288. [PMID: 27830790 PMCID: PMC5108412 DOI: 10.1038/nature20118] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022]
Abstract
Spinal cord injury disrupts the communication between the brain and the spinal circuits that orchestrate movement. To bypass the lesion, brain–computer interfaces1–3 have directly linked cortical activity to electrical stimulation of muscles, which have restored grasping abilities after hand paralysis1,4. Theoretically, this strategy could also restore control over leg muscle activity for walking5. However, replicating the complex sequence of individual muscle activation patterns underlying natural and adaptive locomotor movements poses formidable conceptual and technological challenges6,7. Recently, we showed in rats that epidural electrical stimulation of the lumbar spinal cord can reproduce the natural activation of synergistic muscle groups producing locomotion8–10. Here, we interfaced leg motor cortex activity with epidural electrical stimulation protocols to establish a brain–spinal interface that alleviated gait deficits after a spinal cord injury in nonhuman primates. Rhesus monkeys were implanted with an intracortical microelectrode array into the leg area of motor cortex; and a spinal cord stimulation system composed of a spatially selective epidural implant and a pulse generator with real-time triggering capabilities. We designed and implemented wireless control systems that linked online neural decoding of extension and flexion motor states with stimulation protocols promoting these movements. These systems allowed the monkeys to behave freely without any restrictions or constraining tethered electronics. After validation of the brain–spinal interface in intact monkeys, we performed a unilateral corticospinal tract lesion at the thoracic level. As early as six days post-injury and without prior training of the monkeys, the brain–spinal interface restored weight-bearing locomotion of the paralyzed leg on a treadmill and overground. The implantable components integrated in the brain–spinal interface have all been approved for investigational applications in similar human research, suggesting a practical translational pathway for proof-of-concept studies in people with spinal cord injury.
Collapse
Affiliation(s)
- Marco Capogrosso
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Center for Neuroprosthetics and Institute of Bioengineering, School of Bioengineering, EPFL, Lausanne, Switzerland
| | - Tomislav Milekovic
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - David Borton
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Fabien Wagner
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Eduardo Martin Moraud
- Center for Neuroprosthetics and Institute of Bioengineering, School of Bioengineering, EPFL, Lausanne, Switzerland
| | - Jean-Baptiste Mignardot
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | | - Jerome Gandar
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Quentin Barraud
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - David Xing
- School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Elodie Rey
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Simone Duis
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | | | | - Qin Li
- Motac Neuroscience Ltd, Manchester, UK.,Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Peter Detemple
- Mainz Institute for Microtechnology, Fraunhofer Institute for Chemical Technology (ICT-IMM), Mainz, Germany
| | | | - Silvestro Micera
- Center for Neuroprosthetics and Institute of Bioengineering, School of Bioengineering, EPFL, Lausanne, Switzerland.,The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Erwan Bezard
- Motac Neuroscience Ltd, Manchester, UK.,Institute of Lab Animal Sciences, China Academy of Medical Sciences, Beijing, China.,Institut des Maladies Neurodégénératives, University of Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Jocelyne Bloch
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
111
|
Côté MP, Murray M, Lemay MA. Rehabilitation Strategies after Spinal Cord Injury: Inquiry into the Mechanisms of Success and Failure. J Neurotrauma 2016; 34:1841-1857. [PMID: 27762657 DOI: 10.1089/neu.2016.4577] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Body-weight supported locomotor training (BWST) promotes recovery of load-bearing stepping in lower mammals, but its efficacy in individuals with a spinal cord injury (SCI) is limited and highly dependent on injury severity. While animal models with complete spinal transections recover stepping with step-training, motor complete SCI individuals do not, despite similarly intensive training. In this review, we examine the significant differences between humans and animal models that may explain this discrepancy in the results obtained with BWST. We also summarize the known effects of SCI and locomotor training on the muscular, motoneuronal, interneuronal, and supraspinal systems in human and non-human models of SCI and address the potential causes for failure to translate to the clinic. The evidence points to a deficiency in neuronal activation as the mechanism of failure, rather than muscular insufficiency. While motoneuronal and interneuronal systems cannot be directly probed in humans, the changes brought upon by step-training in SCI animal models suggest a beneficial re-organization of the systems' responsiveness to descending and afferent feedback that support locomotor recovery. The literature on partial lesions in humans and animal models clearly demonstrate a greater dependency on supraspinal input to the lumbar cord in humans than in non-human mammals for locomotion. Recent results with epidural stimulation that activates the lumbar interneuronal networks and/or increases the overall excitability of the locomotor centers suggest that these centers are much more dependent on the supraspinal tonic drive in humans. Sensory feedback shapes the locomotor output in animal models but does not appear to be sufficient to drive it in humans.
Collapse
Affiliation(s)
- Marie-Pascale Côté
- 1 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Marion Murray
- 1 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Michel A Lemay
- 2 Department of Bioengineering, Temple University , Philadelphia, Pennsylvania
| |
Collapse
|
112
|
Tsintou M, Dalamagkas K, Makris N. Advancing research in regeneration and repair of the motor circuitry: non-human primate models and imaging scales as the missing links for successfully translating injectable therapeutics to the clinic. ACTA ACUST UNITED AC 2016; 3. [PMID: 29600289 DOI: 10.23937/2469-570x/1410042] [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] [Indexed: 12/15/2022]
Abstract
Regeneration and repair is the ultimate goal of therapeutics in trauma of the central nervous system (CNS). Stroke and spinal cord injury (SCI) are two highly prevalent CNS disorders that remain incurable, despite numerous research studies and the clinical need for effective treatments. Neural engineering is a diverse biomedical field, that addresses these diseases using new approaches. Research in the field involves principally rodent models and biologically active, biodegradable hydrogels. Promising results have been reported in preclinical studies of CNS repair, demonstrating the great potential for the development of new treatments for the brain, spinal cord and peripheral nerve injury. Several obstacles stand in the way of clinical translation of neuroregeneration research. There seems to be a key gap in the translation of research from rodent models to human applications, namely non-human primate models, which constitute a critical bridging step. Applying injectable therapeutics and multimodal neuroimaging in stroke lesions using experimental rhesus monkey models is an avenue that a few research groups have begun to embark on. Understanding and assessing the changes that the injured brain or spinal cord undergoes after an intervention with biodegradable hydrogels in non-human primates seem to represent critical preclinical research steps. Existing innovative models in non-human primates allow us to evaluate the potential of neural engineering and injectable hydrogels. The results of these preliminary studies will pave the way for translating this research into much needed clinical therapeutic approaches. Cutting edge imaging technology using Connectome scanners represents a tremendous advancement, enabling the in vivo, detailed, high-resolution evaluation of these therapeutic interventions in experimental animals. Most importantly, they also allow quantifiable and clinically meaningful correlations with humans, increasing the translatability of these innovations to the bedside.
Collapse
Affiliation(s)
- Magdalini Tsintou
- Psychiatry Neuroimaging Laboratory, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Center for Neural Systems Investigations, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129
| | - Kyriakos Dalamagkas
- Psychiatry Neuroimaging Laboratory, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Nikos Makris
- Psychiatry Neuroimaging Laboratory, Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115.,Center for Neural Systems Investigations, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129.,Center for Morphometric Analysis, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129
| |
Collapse
|
113
|
O'Donovan KJ. Intrinsic Axonal Growth and the Drive for Regeneration. Front Neurosci 2016; 10:486. [PMID: 27833527 PMCID: PMC5081384 DOI: 10.3389/fnins.2016.00486] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/10/2016] [Indexed: 02/01/2023] Open
Abstract
Following damage to the adult nervous system in conditions like stroke, spinal cord injury, or traumatic brain injury, many neurons die and most of the remaining spared neurons fail to regenerate. Injured neurons fail to regrow both because of the inhibitory milieu in which they reside as well as a loss of the intrinsic growth capacity of the neurons. If we are to develop effective therapeutic interventions that promote functional recovery for the devastating injuries described above, we must not only better understand the molecular mechanisms of developmental axonal growth in hopes of re-activating these pathways in the adult, but at the same time be aware that re-activation of adult axonal growth may proceed via distinct mechanisms. With this knowledge in hand, promoting adult regeneration of central nervous system neurons can become a more tractable and realistic therapeutic endeavor.
Collapse
Affiliation(s)
- Kevin J O'Donovan
- Department of Chemistry and Life Science, United States Military Academy West Point, NY, USA
| |
Collapse
|
114
|
Reed JL, Liao CC, Qi HX, Kaas JH. Plasticity and Recovery After Dorsal Column Spinal Cord Injury in Nonhuman Primates. J Exp Neurosci 2016; 10:11-21. [PMID: 27578996 PMCID: PMC4991577 DOI: 10.4137/jen.s40197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 12/15/2022] Open
Abstract
Here, we review recent work on plasticity and recovery after dorsal column spinal cord injury in nonhuman primates. Plasticity in the adult central nervous system has been established and studied for the past several decades; however, capacities and limits of plasticity are still under investigation. Studies of plasticity include assessing multiple measures before and after injury in animal models. Such studies are particularly important for improving recovery after injury in patients. In summarizing work by our research team and others, we suggest how the findings from plasticity studies in nonhuman primate models may affect therapeutic interventions for conditions involving sensory loss due to spinal cord injury.
Collapse
Affiliation(s)
- Jamie L Reed
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - Chia-Chi Liao
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - Hui-Xin Qi
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| | - Jon H Kaas
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
115
|
Re-Establishment of Cortical Motor Output Maps and Spontaneous Functional Recovery via Spared Dorsolaterally Projecting Corticospinal Neurons after Dorsal Column Spinal Cord Injury in Adult Mice. J Neurosci 2016; 36:4080-92. [PMID: 27053214 DOI: 10.1523/jneurosci.3386-15.2016] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Motor cortical plasticity contributes to spontaneous recovery after incomplete spinal cord injury (SCI), but the pathways underlying this remain poorly understood. We performed optogenetic mapping of motor cortex in channelrhodopsin-2 expressing mice to assess the capacity of the cortex to re-establish motor output longitudinally after a C3/C4 dorsal column SCI that bilaterally ablated the dorsal corticospinal tract (CST) containing ∼96% of corticospinal fibers but spared ∼3% of CST fibers that project via the dorsolateral funiculus. Optogenetic mapping revealed extensive early deficits, but eventual reestablishment of motor cortical output maps to the limbs at the same latency as preoperatively by 4 weeks after injury. Analysis of skilled locomotion on the horizontal ladder revealed early deficits followed by partial spontaneous recovery by 6 weeks after injury. To dissociate between the contributions of injured dorsal projecting versus spared dorsolateral projecting corticospinal neurons, we established a transient silencing approach to inactivate spared dorsolaterally projecting corticospinal neurons specifically by injecting adeno-associated virus (AAV)-expressing Cre-dependent DREADD (designer receptor exclusively activated by designer drug) receptor hM4Di in sensorimotor cortex and AAV-expressing Cre in C7/C8 dorsolateral funiculus. Transient silencing uninjured dorsolaterally projecting corticospinal neurons via activation of the inhibitory DREADD receptor hM4Di abrogated spontaneous recovery and resulted in a greater change in skilled locomotion than in control uninjured mice using the same silencing approach. These data demonstrate the pivotal role of a minor dorsolateral corticospinal pathway in mediating spontaneous recovery after SCI and support a focus on spared corticospinal neurons as a target for therapy. SIGNIFICANCE STATEMENT Spontaneous recovery can occur after incomplete spinal cord injury (SCI), but the pathways underlying this remain poorly understood. We performed optogenetic mapping of motor cortex after a cervical SCI that interrupts most corticospinal transmission but results in partial recovery on a horizontal ladder task of sensorimotor function. We demonstrate that the motor cortex can reestablish output to the limbs longitudinally. To dissociate the roles of injured and uninjured corticospinal neurons in mediating recovery, we transiently silenced the minor dorsolateral corticospinal pathway spared by our injury. This abrogated spontaneous recovery and resulted in a greater change in skilled locomotion than in uninjured mice using the same approach. Therefore, uninjured corticospinal neurons substantiate remarkable motor cortical plasticity and partial recovery after SCI.
Collapse
|
116
|
Filippi M, Boido M, Pasquino C, Garello F, Boffa C, Terreno E. Successful in vivo MRI tracking of MSCs labeled with Gadoteridol in a Spinal Cord Injury experimental model. Exp Neurol 2016; 282:66-77. [DOI: 10.1016/j.expneurol.2016.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/26/2016] [Accepted: 05/18/2016] [Indexed: 01/02/2023]
|
117
|
Mabray MC, Talbott JF, Whetstone WD, Dhall SS, Phillips DB, Pan JZ, Manley GT, Bresnahan JC, Beattie MS, Haefeli J, Ferguson AR. Multidimensional Analysis of Magnetic Resonance Imaging Predicts Early Impairment in Thoracic and Thoracolumbar Spinal Cord Injury. J Neurotrauma 2016; 33:954-62. [PMID: 26414451 PMCID: PMC4876497 DOI: 10.1089/neu.2015.4093] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Literature examining magnetic resonance imaging (MRI) in acute spinal cord injury (SCI) has focused on cervical SCI. Reproducible systems have been developed for MRI-based grading; however, it is unclear how they apply to thoracic SCI. Our hypothesis is that MRI measures will group as coherent multivariate principal component (PC) ensembles, and that distinct PCs and individual variables will show discriminant validity for predicting early impairment in thoracic SCI. We undertook a retrospective cohort study of 25 patients with acute thoracic SCI who underwent MRI on admission and had American Spinal Injury Association Impairment Scale (AIS) assessment at hospital discharge. Imaging variables of axial grade, sagittal grade, length of injury, thoracolumbar injury classification system (TLICS), maximum canal compromise (MCC), and maximum spinal cord compression (MSCC) were collected. We performed an analytical workflow to detect multivariate PC patterns followed by explicit hypothesis testing to predict AIS at discharge. All imaging variables loaded positively on PC1 (64.3% of variance), which was highly related to AIS at discharge. MCC, MSCC, and TLICS also loaded positively on PC2 (22.7% of variance), while variables concerning cord signal abnormality loaded negatively on PC2. PC2 was highly related to the patient undergoing surgical decompression. Variables of signal abnormality were all negatively correlated with AIS at discharge with the highest level of correlation for axial grade as assessed with the Brain and Spinal Injury Center (BASIC) score. A multiple variable model identified BASIC as the only statistically significant predictor of AIS at discharge, signifying that BASIC best captured the variance in AIS within our study population. Our study provides evidence of convergent validity, construct validity, and clinical predictive validity for the sampled MRI measures of SCI when applied in acute thoracic and thoracolumbar SCI.
Collapse
Affiliation(s)
- Marc C. Mabray
- Department of Radiology and Biomedical Imaging, University of California San Francisco and San Francisco General Hospital, San Francisco, California
| | - Jason F. Talbott
- Department of Radiology and Biomedical Imaging, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - William D. Whetstone
- Department of Emergency Medicine, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Sanjay S. Dhall
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - David B. Phillips
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Jonathan Z. Pan
- Department of Anesthesia and Perioperative Care, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Geoffrey T. Manley
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Jacqueline C. Bresnahan
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Michael S. Beattie
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Jenny Haefeli
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
| | - Adam R. Ferguson
- Department of Neurological Surgery, University of California San Francisco and San Francisco General Hospital, San Francisco, California
- Brain and Spinal Injury Center, San Francisco General Hospital, San Francisco, California
- San Francisco Veteran's Affairs Medical Center, San Francisco, California
| |
Collapse
|
118
|
He Z, Jin Y. Intrinsic Control of Axon Regeneration. Neuron 2016; 90:437-51. [DOI: 10.1016/j.neuron.2016.04.022] [Citation(s) in RCA: 337] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/10/2016] [Accepted: 04/13/2016] [Indexed: 01/12/2023]
|
119
|
Sparrey CJ, Salegio EA, Camisa W, Tam H, Beattie MS, Bresnahan JC. Mechanical Design and Analysis of a Unilateral Cervical Spinal Cord Contusion Injury Model in Non-Human Primates. J Neurotrauma 2016; 33:1136-49. [PMID: 26670940 DOI: 10.1089/neu.2015.3974] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Non-human primate (NHP) models of spinal cord injury better reflect human injury and provide a better foundation to evaluate potential treatments and functional outcomes. We combined finite element (FE) and surrogate models with impact data derived from in vivo experiments to define the impact mechanics needed to generate a moderate severity unilateral cervical contusion injury in NHPs (Macaca mulatta). Three independent variables (impactor displacement, alignment, and pre-load) were examined to determine their effects on tissue level stresses and strains. Mechanical measures of peak force, peak displacement, peak energy, and tissue stiffness were analyzed as potential determinants of injury severity. Data generated from FE simulations predicted a lateral shift of the spinal cord at high levels of compression (>64%) during impact. Submillimeter changes in mediolateral impactor position over the midline increased peak impact forces (>50%). Surrogate cords established a 0.5 N pre-load protocol for positioning the impactor tip onto the dural surface to define a consistent dorsoventral baseline position before impact, which corresponded with cerebrospinal fluid displacement and entrapment of the spinal cord against the vertebral canal. Based on our simulations, impactor alignment and pre-load were strong contributors to the variable mechanical and functional outcomes observed in in vivo experiments. Peak displacement of 4 mm after a 0.5N pre-load aligned 0.5-1.0 mm over the midline should result in a moderate severity injury; however, the observed peak force and calculated peak energy and tissue stiffness are required to properly characterize the severity and variability of in vivo NHP contusion injuries.
Collapse
Affiliation(s)
- Carolyn J Sparrey
- 1 Mechatronic Systems Engineering, Simon Fraser University , Surrey, British Columbia, Canada .,2 International Collaboration on Repair Discoveries (ICORD) , Vancouver, British Columbia, Canada
| | - Ernesto A Salegio
- 3 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco , San Francisco, California
| | - William Camisa
- 4 Taylor Collaboration, St Mary's Medical Center , San Francisco, California
| | - Horace Tam
- 1 Mechatronic Systems Engineering, Simon Fraser University , Surrey, British Columbia, Canada
| | - Michael S Beattie
- 3 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco , San Francisco, California
| | - Jacqueline C Bresnahan
- 3 Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco , San Francisco, California
| |
Collapse
|
120
|
Doulames VM, Plant GW. Induced Pluripotent Stem Cell Therapies for Cervical Spinal Cord Injury. Int J Mol Sci 2016; 17:530. [PMID: 27070598 PMCID: PMC4848986 DOI: 10.3390/ijms17040530] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/17/2016] [Accepted: 03/28/2016] [Indexed: 02/07/2023] Open
Abstract
Cervical-level injuries account for the majority of presented spinal cord injuries (SCIs) to date. Despite the increase in survival rates due to emergency medicine improvements, overall quality of life remains poor, with patients facing variable deficits in respiratory and motor function. Therapies aiming to ameliorate symptoms and restore function, even partially, are urgently needed. Current therapeutic avenues in SCI seek to increase regenerative capacities through trophic and immunomodulatory factors, provide scaffolding to bridge the lesion site and promote regeneration of native axons, and to replace SCI-lost neurons and glia via intraspinal transplantation. Induced pluripotent stem cells (iPSCs) are a clinically viable means to accomplish this; they have no major ethical barriers, sources can be patient-matched and collected using non-invasive methods. In addition, the patient’s own cells can be used to establish a starter population capable of producing multiple cell types. To date, there is only a limited pool of research examining iPSC-derived transplants in SCI—even less research that is specific to cervical injury. The purpose of the review herein is to explore both preclinical and clinical recent advances in iPSC therapies with a detailed focus on cervical spinal cord injury.
Collapse
Affiliation(s)
- Vanessa M Doulames
- Stanford Partnership for Spinal Cord Injury and Repair, Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive Stanford, California, CA 94305, USA.
| | - Giles W Plant
- Stanford Partnership for Spinal Cord Injury and Repair, Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive Stanford, California, CA 94305, USA.
| |
Collapse
|
121
|
Moraud EM, Capogrosso M, Formento E, Wenger N, DiGiovanna J, Courtine G, Micera S. Mechanisms Underlying the Neuromodulation of Spinal Circuits for Correcting Gait and Balance Deficits after Spinal Cord Injury. Neuron 2016; 89:814-28. [PMID: 26853304 DOI: 10.1016/j.neuron.2016.01.009] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/11/2015] [Accepted: 12/26/2015] [Indexed: 01/24/2023]
Abstract
Epidural electrical stimulation of lumbar segments facilitates standing and walking in animal models and humans with spinal cord injury. However, the mechanisms through which this neuromodulation therapy engages spinal circuits remain enigmatic. Using computer simulations and behavioral experiments, we provide evidence that epidural electrical stimulation interacts with muscle spindle feedback circuits to modulate muscle activity during locomotion. Hypothesis-driven strategies emerging from simulations steered the design of stimulation protocols that adjust bilateral hindlimb kinematics throughout gait execution. These stimulation strategies corrected subject-specific gait and balance deficits in rats with incomplete and complete spinal cord injury. The conservation of muscle spindle feedback circuits across mammals suggests that the same mechanisms may facilitate motor control in humans. These results provide a conceptual framework to improve stimulation protocols for clinical applications.
Collapse
Affiliation(s)
| | - Marco Capogrosso
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland; BioRobotics Institute, Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Emanuele Formento
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Nikolaus Wenger
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland; Department of Neurology and Experimental Neurology, University of Berlin, 10098 Berlin, Germany
| | - Jack DiGiovanna
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Grégoire Courtine
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland.
| | - Silvestro Micera
- Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland; BioRobotics Institute, Scuola Superiore Sant'Anna, 56127 Pisa, Italy.
| |
Collapse
|