1
|
Danner SM, Shepard CT, Hainline C, Shevtsova NA, Rybak IA, Magnuson DSK. Spinal control of locomotion before and after spinal cord injury. Exp Neurol 2023; 368:114496. [PMID: 37499972 PMCID: PMC10529867 DOI: 10.1016/j.expneurol.2023.114496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/10/2023] [Accepted: 07/23/2023] [Indexed: 07/29/2023]
Abstract
Thoracic spinal cord injury affects long propriospinal neurons that interconnect the cervical and lumbar enlargements. These neurons are crucial for coordinating forelimb and hindlimb locomotor movements in a speed-dependent manner. However, recovery from spinal cord injury is usually studied over a very limited range of speeds that may not fully expose circuitry dysfunction. To overcome this limitation, we investigated overground locomotion in rats trained to move over an extended distance with a wide range of speeds both pre-injury and after recovery from thoracic hemisection or contusion injuries. In this experimental context, intact rats expressed a speed-dependent continuum of alternating (walk and trot) and non-alternating (canter, gallop, half-bound gallop, and bound) gaits. After a lateral hemisection injury, rats recovered the ability to locomote over a wide range of speeds but lost the ability to use the highest-speed gaits (half-bound gallop and bound) and predominantly used the limb contralateral to the injury as lead during canter and gallop. A moderate contusion injury caused a greater reduction in maximal speed, loss of all non-alternating gaits, and emergence of novel alternating gaits. These changes resulted from weak fore-hind coupling together with appropriate control of left-right alternation. After hemisection, animals expressed a subset of intact gaits with appropriate interlimb coordination even on the side of the injury, where the long propriospinal connections were severed. These observations highlight how investigating locomotion over the full range of speeds can reveal otherwise hidden aspects of spinal locomotor control and post-injury recovery.
Collapse
Affiliation(s)
- Simon M Danner
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA.
| | - Courtney T Shepard
- Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| | - Casey Hainline
- Speed School of Engineering, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| | - Natalia A Shevtsova
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Ilya A Rybak
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - David S K Magnuson
- Department of Neurological Surgery, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA; Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Health Sciences Campus, Louisville, KY, USA
| |
Collapse
|
2
|
Shibata T, Tashiro S, Nakamura M, Okano H, Nagoshi N. A Review of Treatment Methods Focusing on Human Induced Pluripotent Stem Cell-Derived Neural Stem/Progenitor Cell Transplantation for Chronic Spinal Cord Injury. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1235. [PMID: 37512047 PMCID: PMC10384869 DOI: 10.3390/medicina59071235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
Cell transplantation therapy using human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NS/PCs) has attracted attention as a regenerative therapy for spinal cord injury (SCI), and its efficacy in treating the subacute phase of SCI has been reported in numerous studies. However, few studies have focused on treatment in the chronic phase, which accounts for many patients, suggesting that there are factors that are difficult to overcome in the treatment of chronic SCI. The search for therapeutic strategies that focus on chronic SCI is fraught with challenges, and the combination of different therapies is thought to be the key to a solution. In addition, many issues remain to be addressed, including the investigation of therapeutic approaches for more severe injury models of chronic SCI and the acquisition of practical motor function. This review summarizes the current progress in regenerative therapy for SCI and discusses the prospects for regenerative medicine, particularly in animal models of chronic SCI.
Collapse
Affiliation(s)
- Takahiro Shibata
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Syoichi Tashiro
- Department of Rehabilitation Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
3
|
Danner SM, Shepard CT, Hainline C, Shevtsova NA, Rybak IA, Magnuson DS. Spinal control of locomotion before and after spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533794. [PMID: 36993490 PMCID: PMC10055332 DOI: 10.1101/2023.03.22.533794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Thoracic spinal cord injury affects long propriospinal neurons that interconnect the cervical and lumbar enlargements. These neurons are crucial for coordinating forelimb and hindlimb locomotor movements in a speed-dependent manner. However, recovery from spinal cord injury is usually studied over a very limited range of speeds that may not fully expose circuitry dysfunction. To overcome this limitation, we investigated overground locomotion in rats trained to move over an extended distance with a wide range of speeds both pre-injury and after recovery from thoracic hemisection or contusion injuries. In this experimental context, intact rats expressed a speed-dependent continuum of alternating (walk and trot) and non-alternating (canter, gallop, half-bound gallop, and bound) gaits. After a lateral hemisection injury, rats recovered the ability to locomote over a wide range of speeds but lost the ability to use the highest-speed gaits (half-bound gallop and bound) and predominantly used the limb contralateral to the injury as lead during canter and gallop. A moderate contusion injury caused a greater reduction in maximal speed, loss of all non-alternating gaits, and emergence of novel alternating gaits. These changes resulted from weak fore-hind coupling together with appropriate control of left-right alternation. After hemisection, animals expressed a subset of intact gaits with appropriate interlimb coordination even on the side of the injury, where the long propriospinal connections were severed. These observations highlight how investigating locomotion over the full range of speeds can reveal otherwise hidden aspects of spinal locomotor control and post-injury recovery.
Collapse
Affiliation(s)
- Simon M. Danner
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Courtney T. Shepard
- Interdisciplinary Program in Translational Neuroscience, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| | - Casey Hainline
- Speed School of Engineering, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| | - Natalia A. Shevtsova
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Ilya A. Rybak
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - David S.K. Magnuson
- Department of Neurological Surgery, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Health Sciences Campus, Louisville, Kentucky, USA
| |
Collapse
|
4
|
Coyoy-Salgado A, Orozco-Barrios C, Sánchez-Torres S, Olayo MG, Cruz GJ, Morales-Corona J, Olayo R, Diaz-Ruiz A, Ríos C, Alvarez-Mejia L, Mondragón-Lozano R, Morales-Guadarrama A, Alonso-García AL, Fabela-Sánchez O, Salgado-Ceballos H. Gene expression and locomotor recovery in adult rats with spinal cord injury and plasma-synthesized polypyrrole/iodine application combined with a mixed rehabilitation scheme. Front Neurol 2023; 14:1124245. [PMID: 37288064 PMCID: PMC10243140 DOI: 10.3389/fneur.2023.1124245] [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: 12/14/2022] [Accepted: 04/14/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Spinal cord injury (SCI) can cause paralysis, for which effective therapeutic strategies have not been developed yet. The only accepted strategy for patients is rehabilitation (RB), although this does not allow complete recovery of lost functions, which makes it necessary to combine it with strategies such as plasma-synthesized polypyrrole/iodine (PPy/I), a biopolymer with different physicochemical properties than PPy synthesized by conventional methods. After SCI in rats, PPy/I promotes functional recovery. Therefore, the purpose of this study was to increase the beneficial effects of both strategies and identify which genes activate PPy/I when applied alone or in combination with a mixed scheme of RB by swimming and enriched environment (SW/EE) in rats with SCI. Methods Microarray analysis was performed to identify mechanisms of action underlying the effects of PPy/I and PPy/I+SW/EE on motor function recovery as evaluated by the BBB scale. Results Results showed robust upregulation by PPy/I in genes related to the developmental process, biogenesis, synapse, and synaptic vesicle trafficking. In addition, PPy/I+SW/EE increased the expression of genes related to proliferation, biogenesis, cell development, morphogenesis, cell differentiation, neurogenesis, neuron development, and synapse formation processes. Immunofluorescence analysis showed the expression of β-III tubulin in all groups, a decreased expression of caspase-3 in the PPy/I group and GFAP in the PPy/I+SW/EE group (p < 0.05). Better preservation of nerve tissue was observed in PPy/I and PPy/SW/EE groups (p < 0.05). In the BBB scale, the control group scored 1.72 ± 0.41, animals with PPy/I treatment scored 4.23 ± 0.33, and those with PPy/I+SW/EE scored 9.13 ± 0.43 1 month after follow-up. Conclusion Thus, PPy/I+SW/EE could represent a therapeutic alternative for motor function recovery after SCI.
Collapse
Affiliation(s)
- Angélica Coyoy-Salgado
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Carlos Orozco-Barrios
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Stephanie Sánchez-Torres
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - María Guadalupe Olayo
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Guillermo Jesus Cruz
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Juan Morales-Corona
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Roberto Olayo
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Araceli Diaz-Ruiz
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Camilo Ríos
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Laura Alvarez-Mejia
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Rodrigo Mondragón-Lozano
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Axayacatl Morales-Guadarrama
- Electrical Engineering Department, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
- National Center for Research in Imaging and Medical Instrumentation, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | | | - Omar Fabela-Sánchez
- Researchers for Mexico CONACyT-Centro de Investigación en Química Aplicada, Department of Chemistry Macromolecules and Nanomaterials, Saltillo, Mexico
| | - Hermelinda Salgado-Ceballos
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| |
Collapse
|
5
|
Moderate-Intensity Treadmill Exercise Promotes mTOR-Dependent Motor Cortical Neurotrophic Factor Expression and Functional Recovery in a Murine Model of Crush Spinal Cord Injury (SCI). Mol Neurobiol 2023; 60:960-978. [PMID: 36385234 DOI: 10.1007/s12035-022-03117-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022]
Abstract
Treadmill exercise is widely considered an effective strategy for restoration of skilled motor function after spinal cord injury (SCI). However, the specific exercise intensity that optimizes recovery and the underlying mechanistic basis of this recovery remain unclear. To that end, we sought to investigate the effect of different treadmill exercise intensities on cortical mTOR activity, a key regulator of functional recovery following CNS trauma, in an animal model of C5 crush spinal cord injury (SCI). Following injury, animals were subjected to treadmill exercise for 4 consecutive weeks at three different intensities (low intensity [LEI]; moderate intensity [MEI]; and high intensity [HEI]). Motor function recovery was assessed by horizontal ladder test, cylinder rearing test, and electrophysiology, while neurotrophic factors and cortical mechanistic target of rapamycin (mTOR) pathway-related proteins were assessed by Western blotting. The activation of the cortical mTOR pathway and axonal sprouting was evaluated by immunofluorescence and the changes of plasticity in motor cortex neurons were assessed by Golgi staining. In keeping with previous studies, we found that 4 weeks of treadmill training resulted in improved skilled motor function, enhanced nerve conduction capability, increased neuroplasticity, and axonal sprouting. Importantly, we also demonstrated that when compared with the LEI group, MEI and HEI groups demonstrated elevated expression of brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), phosphorylated ribosomal S6 protein (p-S6), and protein kinase B (p-Akt), consistent with an intensity-dependent activation of the mTOR pathway and neurotrophic factor expression in the motor cortex. We also observed impaired exercise endurance and higher mortality during training in the HEI group than in the LEI and MEI groups. Collectively, our findings suggest that treadmill exercise following SCI is an effective means of promoting recovery and highlight the importance of the cortical mTOR pathway and neurotrophic factors as mediators of this effect. Importantly, our findings also demonstrate that excessive exercise can be detrimental, suggesting that moderation may be the optimal strategy. These findings provide an important foundation for further investigation of treadmill training as a modality for recovery following spinal cord injury and of the underlying mechanisms.
Collapse
|
6
|
Bazanovich SA, Yadgarov MY, Zvyagintseva MA, Ryabov SI, Grin' AA, Smirnov VA. A Method of Assessment of the Motor Function of Hind Limbs by Swim Test in Spinal Cord Injury Models. Bull Exp Biol Med 2022; 172:499-503. [PMID: 35175478 DOI: 10.1007/s10517-022-05422-5] [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: 07/23/2021] [Indexed: 10/19/2022]
Abstract
In most studies, various load tests are used to assess the recovery of functions after spinal cord injury in animals. However, the existing methods of assessing the movement in animals are not sufficiently accurate and objective. We developed a new method for assessing motor activity of laboratory animals that allows objective and highly accurate evaluation of movements in animals with serious neurological disorders caused by spinal cord injury. The swimming test was used as the main load test. Motor activity of swimming animals was assessed by measuring angles relative to the axis of motion, and the degree of angle spread for each joint and limb was estimated using the dispersion parameters depending on the values of the angles of the joints and the dispersion depending on the amplitudes of the angles. In Sprague-Dawley rats, contusion of the spinal cord at the Th9 level was modeled. In the swimming test, healthy control animals showed stability of both variance indicators over 6 weeks. In rats with spinal cord injury, motor activity of the hind limbs tended to increase from the first to the third weeks and remained at this level from the third to sixth weeks. The results suggest that the proposed method can become a good analogue of modern methods for assessing motor activity.
Collapse
Affiliation(s)
- S A Bazanovich
- Laboratory of Stem Cells, National Medical Research Center of Cardiology, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - M Ya Yadgarov
- Laboratory of Stem Cells, National Medical Research Center of Cardiology, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - M A Zvyagintseva
- Laboratory of Stem Cells, National Medical Research Center of Cardiology, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - S I Ryabov
- Laboratory of Stem Cells, National Medical Research Center of Cardiology, Ministry of the Health of the Russian Federation, Moscow, Russia
| | - A A Grin'
- Department of Neurosurgery, N. V. Sklifosovsky Research Institute of Emergency Care, Moscow Healthcare Department, Russian Federation, Moscow, Russia
| | - V A Smirnov
- Laboratory of Stem Cells, National Medical Research Center of Cardiology, Ministry of the Health of the Russian Federation, Moscow, Russia. .,Department of Neurosurgery, N. V. Sklifosovsky Research Institute of Emergency Care, Moscow Healthcare Department, Russian Federation, Moscow, Russia.
| |
Collapse
|
7
|
Shepard CT, Pocratsky AM, Brown BL, Van Rijswijck MA, Zalla RM, Burke DA, Morehouse JR, Riegler AS, Whittemore SR, Magnuson DSK. Silencing long ascending propriospinal neurons after spinal cord injury improves hindlimb stepping in the adult rat. eLife 2021; 10:e70058. [PMID: 34854375 PMCID: PMC8639151 DOI: 10.7554/elife.70058] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
Long ascending propriospinal neurons (LAPNs) are a subpopulation of spinal cord interneurons that directly connect the lumbar and cervical enlargements. Previously we showed, in uninjured animals, that conditionally silencing LAPNs disrupted left-right coordination of the hindlimbs and forelimbs in a context-dependent manner, demonstrating that LAPNs secure alternation of the fore- and hindlimb pairs during overground stepping. Given the ventrolateral location of LAPN axons in the spinal cord white matter, many likely remain intact following incomplete, contusive, thoracic spinal cord injury (SCI), suggesting a potential role in the recovery of stepping. Thus, we hypothesized that silencing LAPNs after SCI would disrupt recovered locomotion. Instead, we found that silencing spared LAPNs post-SCI improved locomotor function, including paw placement order and timing, and a decrease in the number of dorsal steps. Silencing also restored left-right hindlimb coordination and normalized spatiotemporal features of gait such as stance and swing time. However, hindlimb-forelimb coordination was not restored. These data indicate that the temporal information carried between the spinal enlargements by the spared LAPNs post-SCI is detrimental to recovered hindlimb locomotor function. These findings are an illustration of a post-SCI neuroanatomical-functional paradox and have implications for the development of neuronal- and axonal-protective therapeutic strategies and the clinical study/implementation of neuromodulation strategies.
Collapse
Affiliation(s)
- Courtney T Shepard
- Interdisciplinary Program in Translational Neuroscience, School of Interdisciplinary and Graduate Studies, University of LouisvilleLouisvilleUnited States
- Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Amanda M Pocratsky
- Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Brandon L Brown
- Interdisciplinary Program in Translational Neuroscience, School of Interdisciplinary and Graduate Studies, University of LouisvilleLouisvilleUnited States
- Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Morgan A Van Rijswijck
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
| | - Rachel M Zalla
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Johnny R Morehouse
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Amberley S Riegler
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Scott R Whittemore
- Interdisciplinary Program in Translational Neuroscience, School of Interdisciplinary and Graduate Studies, University of LouisvilleLouisvilleUnited States
- Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - David SK Magnuson
- Interdisciplinary Program in Translational Neuroscience, School of Interdisciplinary and Graduate Studies, University of LouisvilleLouisvilleUnited States
- Department of Anatomical Sciences and Neurobiology, University of Louisville, LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| |
Collapse
|
8
|
Development of a Spinal Cord Injury Model Permissive to Study the Cardiovascular Effects of Rehabilitation Approaches Designed to Induce Neuroplasticity. BIOLOGY 2021; 10:biology10101006. [PMID: 34681105 PMCID: PMC8533334 DOI: 10.3390/biology10101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 11/24/2022]
Abstract
Simple Summary People living with high-level spinal cord injury experience worse cardiovascular health than the general population. In most spinal cord injuries, there are some remaining functioning pathways leading from the brain through the spinal cord to the organs and muscles, but not enough to sustain normal levels of function. Recently, therapies that aim to increase the strength of connections in these remaining pathways have shown great potential in restoring walking, hand, and breathing function in the spinal cord injured population. In order to test these therapies for their effects on cardiovascular function, we developed a new type of spinal cord injury rat model that spares enough pathways for these therapies to act upon but still produces measurable reductions in heart and blood vessel function that can be targeted with interventions/treatments. Abstract As primary medical care for spinal cord injury (SCI) has improved over the last decades there are more individuals living with neurologically incomplete (vs. complete) cervical injuries. For these individuals, a number of promising therapies are being actively researched in pre-clinical settings that seek to strengthen the remaining spinal pathways with a view to improve motor function. To date, few, if any, of these interventions have been tested for their effectiveness to improve autonomic and cardiovascular (CV) function. As a first step to testing such therapies, we aimed to develop a model that has sufficient sparing of descending sympathetic pathways for these interventions to target yet induces robust CV impairment. Twenty-six Wistar rats were assigned to SCI (n = 13) or naïve (n = 13) groups. Animals were injured at the T3 spinal segment with 300 kdyn of force. Fourteen days post-SCI, left ventricular (LV) and arterial catheterization was performed to assess in vivo cardiac and hemodynamic function. Spinal cord lesion characteristics along with sparing in catecholaminergic and serotonergic projections were determined via immunohistochemistry. SCI produced a decrease in mean arterial pressure of 17 ± 3 mmHg (p < 0.001) and left ventricular contractility (end-systolic elastance) of 0.7 ± 0.1 mmHg/µL (p < 0.001). Our novel SCI model produced significant decreases in cardiac and hemodynamic function while preserving 33 ± 9% of white matter at the injury epicenter, which we believe makes it a useful pre-clinical model of SCI to study rehabilitation approaches designed to induce neuroplasticity.
Collapse
|
9
|
Treadmill training based on the overload principle promotes locomotor recovery in a mouse model of chronic spinal cord injury. Exp Neurol 2021; 345:113834. [PMID: 34370998 DOI: 10.1016/j.expneurol.2021.113834] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/16/2021] [Accepted: 08/02/2021] [Indexed: 11/21/2022]
Abstract
Rehabilitative treatment, including treadmill training, is considered an important strategy for restoring motor function after spinal cord injury (SCI). However, many unexplained problems persist regarding the appropriate rehabilitative method and the mechanism underlying the beneficial effects of rehabilitation. Moreover, only a few preclinical studies have been performed on rehabilitative interventions for chronic SCI, although most patients have chronic injuries. In fact, several preclinical studies reported that rehabilitative training was less effective when applied during the chronic phase than when applied sooner. While numerous studies have examined the effects of treadmill training during the subacute phase, the training conditions vary considerably among preclinical reports. Therefore, establishing a standard training protocol is essential for achieving beneficial rehabilitation effects at the chronic stage. Since the difficulty of applying an appropriate training load hinders training at constant speeds, it is important to adjust the training intensity in accordance with the exercise tolerance of an individual animal to provide further functional recovery benefits. Here, we created a novel quadrupedal treadmill training protocol based on the overload principle for mice with incomplete thoracic SCI. We subjected SCI model mice to rehabilitative training according to the protocol for two consecutive weeks starting at 42 days after injury. We examined the treadmill speeds at which the mice were able to run based on the severity of paresis and investigated the impact of the protocol on functional recovery. Assessment of running speed changes during the treadmill training period revealed faster treadmill speeds for mice with mild paresis than for those with severe paresis. The training parameters, including the speed and distance traveled, were positively correlated with the changes in motor function. These results suggest that the most suitable running speed during treadmill training differs according to the level of motor dysfunction and that running longer distances has a positive impact on motor functional recovery. Based on this established protocol, we compared functional and histological results between the chronic SCI groups with and without rehabilitation. The gait analyses showed significantly better functional improvement in the rehabilitation group than in the nonrehabilitation group. Histological analyses revealed that the BDNF- and VGLUT1-positive areas of lumbar enlargement were significantly increased in the rehabilitation group. These findings implied that rehabilitation promoted not only motor performance but also motor control, including forelimb-hindlimb coordination, even in chronic SCI, resulting in functional improvement by treadmill training alone. Therefore, rehabilitative training based on the overload principle appears to be one of the appropriate treatment options for incomplete thoracic SCI, and evidence of its efficacy exists in actual clinical settings.
Collapse
|
10
|
Fouad K, Ng C, Basso DM. Behavioral testing in animal models of spinal cord injury. Exp Neurol 2020; 333:113410. [PMID: 32735871 PMCID: PMC8325780 DOI: 10.1016/j.expneurol.2020.113410] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 01/08/2023]
Abstract
This review is based on a lecture presented at the Craig H. Neilsen Foundation sponsored Spinal Cord Injury Training Program at Ohio State University. We discuss the advantages and challenges of injury models in rodents and theory relation to various behavioral outcome measures. We offer strategies and advice on experimental design, behavioral testing, and on the challenges, one will encounter with animal testing. This review is designed to guide those entering the field of spinal cord injury and/or involved with in vivo animal testing.
Collapse
Affiliation(s)
- K Fouad
- University of Alberta, Faculty of Rehabilitation Medicine, Dept of Physical Therapy, 3-48 Corbett Hall, Edmonton T6G 2G4, Canada; University of Alberta, Neuroscience and Mental Health Institute, 2-132 Li Ka Shing, Edmonton T6G 2E1, Canada.
| | - C Ng
- University of Alberta, Neuroscience and Mental Health Institute, 2-132 Li Ka Shing, Edmonton T6G 2E1, Canada
| | - D M Basso
- Ohio State University, College of Medicine, School of Health and Rehabilitation Sciences, 106A Atwell Hall, 453 W. 10th Ave, Columbus, OH 43210, USA
| |
Collapse
|
11
|
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: 9] [Impact Index Per Article: 2.3] [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
|
12
|
Li M, Jiang WT, Li J, Ji WC. Exercise protects against spinal cord injury through miR-21-mediated suppression of PDCD4. Am J Transl Res 2020; 12:5708-5718. [PMID: 33042450 PMCID: PMC7540147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
Spinal cord injury (SCI) can lead to different levels of paraplegia. Studies have shown that exercise exerts wide protective effects against various diseases, and microRNAs (miRNAs) are involved in its beneficial effects. However, the specific role of miRNAs in the protective effects of exercise against SCI remains unclear. Here, we showed that exercise exerted protective effects against SCI as evidenced by increased locomotor activity and spinal cord cell survival in rats with SCI. Exercise upregulated circulating miR-21, detected by miRNA microarray, in rats with SCI. Treating SCI rats with agomiR-21 upregulated circulating miR-21 and exerted protective effects against SCI. Additionally, downregulating miR-21 using antagomir-21 abolished the protective effects of exercise on SCI. Programmed cell death protein 4 (PDCD4) was found to be the target of miR-21. These results suggested that exercise protects against SCI, at least partly, through miR-21-mediated suppression of PDCD4.
Collapse
Affiliation(s)
- Meng Li
- Department of Orthopaedics, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Wan-Ting Jiang
- Department of Ultrasound Diagnosis, The Fourth Hospital of Xi’anXi’an 710004, China
| | - Jia Li
- Department of Orthopaedics, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| | - Wen-Chen Ji
- Department of Orthopaedics, The First Affiliated Hospital of Xi’an Jiaotong UniversityXi’an 710061, China
| |
Collapse
|
13
|
Pocratsky AM, Shepard CT, Morehouse JR, Burke DA, Riegler AS, Hardin JT, Beare JE, Hainline C, States GJR, Brown BL, Whittemore SR, Magnuson DSK. Long ascending propriospinal neurons provide flexible, context-specific control of interlimb coordination. eLife 2020; 9:e53565. [PMID: 32902379 PMCID: PMC7527236 DOI: 10.7554/elife.53565] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 09/08/2020] [Indexed: 11/13/2022] Open
Abstract
Within the cervical and lumbar spinal enlargements, central pattern generator (CPG) circuitry produces the rhythmic output necessary for limb coordination during locomotion. Long propriospinal neurons that inter-connect these CPGs are thought to secure hindlimb-forelimb coordination, ensuring that diagonal limb pairs move synchronously while the ipsilateral limb pairs move out-of-phase during stepping. Here, we show that silencing long ascending propriospinal neurons (LAPNs) that inter-connect the lumbar and cervical CPGs disrupts left-right limb coupling of each limb pair in the adult rat during overground locomotion on a high-friction surface. These perturbations occurred independent of the locomotor rhythm, intralimb coordination, and speed-dependent (or any other) principal features of locomotion. Strikingly, the functional consequences of silencing LAPNs are highly context-dependent; the phenotype was not expressed during swimming, treadmill stepping, exploratory locomotion, or walking on an uncoated, slick surface. These data reveal surprising flexibility and context-dependence in the control of interlimb coordination during locomotion.
Collapse
Affiliation(s)
- Amanda M Pocratsky
- Department of Anatomical Sciences and Neurobiology, University of LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Courtney T Shepard
- Department of Anatomical Sciences and Neurobiology, University of LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Johnny R Morehouse
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Amberley S Riegler
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - Josiah T Hardin
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
| | - Jason E Beare
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Cardiovascular Innovation Institute, Department of Physiology and Biophysics, University of LouisvilleLouisvilleUnited States
| | - Casey Hainline
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
| | - Gregory JR States
- Department of Anatomical Sciences and Neurobiology, University of LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Brandon L Brown
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
| | - Scott R Whittemore
- Department of Anatomical Sciences and Neurobiology, University of LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
| | - David SK Magnuson
- Department of Anatomical Sciences and Neurobiology, University of LouisvilleLouisvilleUnited States
- Kentucky Spinal Cord Injury Research Center, University of LouisvilleLouisvilleUnited States
- Department of Neurological Surgery, University of LouisvilleLouisvilleUnited States
- Speed School of Engineering, University of LouisvilleLouisvilleUnited States
| |
Collapse
|
14
|
Sánchez-Torres S, Díaz-Ruíz A, Ríos C, Olayo MG, Cruz GJ, Olayo R, Morales J, Mondragón-Lozano R, Fabela-Sánchez O, Orozco-Barrios C, Coyoy-Salgado A, Orozco-Suárez S, González-Ruiz C, Álvarez-Mejía L, Morales-Guadarrama A, Buzoianu-Anguiano V, Damián-Matsumura P, Salgado-Ceballos H. Recovery of motor function after traumatic spinal cord injury by using plasma-synthesized polypyrrole/iodine application in combination with a mixed rehabilitation scheme. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:58. [PMID: 32607849 DOI: 10.1007/s10856-020-06395-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/15/2020] [Indexed: 06/11/2023]
Abstract
Traumatic spinal cord injury (TSCI) can cause paralysis and permanent disability. Rehabilitation (RB) is currently the only accepted treatment, although its beneficial effect is limited. The development of biomaterials has provided therapeutic possibilities for TSCI, where our research group previously showed that the plasma-synthesized polypyrrole/iodine (PPy/I), a biopolymer with different physicochemical characteristics than those of the PPy synthesized by conventional methods, promotes recovery of motor function after TSCI. The present study evaluated if the plasma-synthesized PPy/I applied in combination with RB could increase its beneficial effects and the mechanisms involved. Adult rats with TSCI were divided into no treatment (control); biopolymer (PPy/I); mixed RB by swimming and enriched environment (SW/EE); and combined treatment (PPy/I + SW/EE) groups. Eight weeks after TSCI, the general health of the animals that received any of the treatments was better than the control animals. Functional recovery evaluated by two scales was better and was achieved in less time with the PPy/I + SW/EE combination. All treatments significantly increased βIII-tubulin (nerve plasticity) expression, but only PPy/I increased GAP-43 (nerve regeneration) and MBP (myelination) expression when were analyzed by immunohistochemistry. The expression of GFAP (glial scar) decreased in treated groups when determined by histochemistry, while morphometric analysis showed that tissue was better preserved when PPy/I and PPy/I + SW/EE were administered. The application of PPy/I + SW/EE, promotes the preservation of nervous tissue, and the expression of molecules related to plasticity as βIII-tubulin, reduces the glial scar, improves general health and allows the recovery of motor function after TSCI. The implant of the biomaterial polypyrrole/iodine (PPy/I) synthesized by plasma (an unconventional synthesis method), in combination with a mixed rehabilitation scheme with swimming and enriched environment applied after a traumatic spinal cord injury, promotes expression of GAP-43 and βIII-tubulin (molecules related to plasticity and nerve regeneration) and reduces the expression of GFAP (molecule related to the formation of the glial scar). Both effects together allow the formation of nerve fibers, the reconnection of the spinal cord in the area of injury and the recovery of lost motor function. The figure shows the colocalization (yellow) of βIII-tubilin (red) and GAP-43 (green) in fibers crossing the epicenter of the injury (arrowheads) that reconnect the rostral and caudal ends of the injured spinal cord and allowed recovery of motor function.
Collapse
Affiliation(s)
- Stephanie Sánchez-Torres
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Av. Cuauhtémoc 330, Col. Doctores, México City, CP, 06720, México
- Doctorate in Biological and Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, Mexico City, CP, 09340, Mexico
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
| | - Araceli Díaz-Ruíz
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez S.S.A, Mexico city, CP, 14269, Mexico
| | - Camilo Ríos
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez S.S.A, Mexico city, CP, 14269, Mexico
| | - María G Olayo
- Departamento de Física, Instituto Nacional de Investigaciones Nucleares. Carretera Mexico-Toluca, km 36.5, Ocoyoacac, State of Mexico, CP, 52750, Mexico
| | - Guillermo J Cruz
- Departamento de Física, Instituto Nacional de Investigaciones Nucleares. Carretera Mexico-Toluca, km 36.5, Ocoyoacac, State of Mexico, CP, 52750, Mexico
| | - Roberto Olayo
- Departamento de Física, Universidad Autónoma Metropolitana, Mexico City, CP, 09340, Mexico
| | - Juan Morales
- Departamento de Física, Universidad Autónoma Metropolitana, Mexico City, CP, 09340, Mexico
| | - Rodrigo Mondragón-Lozano
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- CONACyT-Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Mexico City, Mexico
| | - Omar Fabela-Sánchez
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana, San Rafael Atlixco 186, 09340, Iztapalapa, CDMX, México
- Catedrático CONACyT-Centro de Investigación en Química Aplicada, Enrique Reyna H. No. 140, San José de los Cerritos, Saltillo, Coahuila, 25294, México
| | - Carlos Orozco-Barrios
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- CONACyT-Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Mexico City, Mexico
| | - Angélica Coyoy-Salgado
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- CONACyT-Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Mexico City, Mexico
| | - Sandra Orozco-Suárez
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Av. Cuauhtémoc 330, Col. Doctores, México City, CP, 06720, México
| | - Cristian González-Ruiz
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- Escuela Superior de Medicina-Instituto Politécnico Nacional, Mexico City, Mexico
| | - Laura Álvarez-Mejía
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Av. Cuauhtémoc 330, Col. Doctores, México City, CP, 06720, México
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico
- Departamento de Física, Instituto Nacional de Investigaciones Nucleares. Carretera Mexico-Toluca, km 36.5, Ocoyoacac, State of Mexico, CP, 52750, Mexico
| | | | - Vinnitsa Buzoianu-Anguiano
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Av. Cuauhtémoc 330, Col. Doctores, México City, CP, 06720, México
| | - Pablo Damián-Matsumura
- Doctorate in Biological and Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, Mexico City, CP, 09340, Mexico
| | - Hermelinda Salgado-Ceballos
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI. Av. Cuauhtémoc 330, Col. Doctores, México City, CP, 06720, México.
- Proyecto Camina A.C. Research Center, Mexico City, CP, 14050, Mexico.
| |
Collapse
|
15
|
Gallegos C, Carey M, Zheng Y, He X, Cao QL. Reaching and Grasping Training Improves Functional Recovery After Chronic Cervical Spinal Cord Injury. Front Cell Neurosci 2020; 14:110. [PMID: 32536855 PMCID: PMC7266985 DOI: 10.3389/fncel.2020.00110] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022] Open
Abstract
Previous studies suggest locomotion training could be an effective non-invasive therapy after spinal cord injury (SCI) using primarily acute thoracic injuries. However, the majority of SCI patients have chronic cervical injuries. Regaining hand function could significantly increase their quality of life. In this study, we used a clinically relevant chronic cervical contusion to study the therapeutic efficacy of rehabilitation in forelimb functional recovery. Nude rats received a moderate C5 unilateral contusive injury and were then divided into two groups with or without Modified Montoya Staircase (MMS) rehabilitation. For the rehabilitation group, rats were trained 5 days a week starting at 8 weeks post-injury (PI) for 6 weeks. All rats were assessed for skilled forelimb functions with MMS test weekly and for untrained gross forelimb locomotion with grooming and horizontal ladder (HL) tests biweekly. Our results showed that MMS rehabilitation significantly increased the number of pellets taken at 13 and 14 weeks PI and the accuracy rates at 12 to 14 weeks PI. However, there were no significant differences in the grooming scores or the percentage of HL missteps at any time point. Histological analyses revealed that MMS rehabilitation significantly increased the number of serotonergic fibers and the amount of presynaptic terminals around motor neurons in the cervical ventral horns caudal to the injury and reduced glial fibrillary acidic protein (GFAP)-immunoreactive astrogliosis in spinal cords caudal to the lesion. This study shows that MMS rehabilitation can modify the injury environment, promote axonal sprouting and synaptic plasticity, and importantly, improve reaching and grasping functions in the forelimb, supporting the therapeutic potential of task-specific rehabilitation for functional recovery after chronic SCI.
Collapse
Affiliation(s)
- Chrystine Gallegos
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Matthew Carey
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Summer Undergraduate Research Program, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yiyan Zheng
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiuquan He
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qi Lin Cao
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
16
|
Torres-Espín A, Beaudry E, Fenrich K, Fouad K. Rehabilitative Training in Animal Models of Spinal Cord Injury. J Neurotrauma 2019; 35:1970-1985. [PMID: 30074874 DOI: 10.1089/neu.2018.5906] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rehabilitative motor training is currently one of the most widely used approaches to promote moderate recovery following injuries of the central nervous system. Such training is generally applied in the clinical setting, whereas it is not standard in preclinical research. This is a concern as it is becoming increasingly apparent that neuroplasticity enhancing treatments require training or some form of activity as a co-therapy to promote functional recovery. Despite the importance of training and the many open questions regarding its mechanistic consequences, its use in preclinical animal models is rather limited. Here we review approaches, findings and challenges when training is applied in animal models of spinal cord injury, and we suggest recommendations to facilitate the integration of training using an appropriate study design, into pre-clinical studies.
Collapse
Affiliation(s)
- Abel Torres-Espín
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| | - Eric Beaudry
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| | | | - Karim Fouad
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta , Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Chariker JH, Ohri SS, Gomes C, Brabazon F, Harman KA, DeVeau KM, Magnuson DSK, Hetman M, Petruska JC, Whittemore SR, Rouchka EC. Activity/exercise-induced changes in the liver transcriptome after chronic spinal cord injury. Sci Data 2019; 6:88. [PMID: 31197156 PMCID: PMC6565704 DOI: 10.1038/s41597-019-0087-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023] Open
Abstract
Multi-organ dysfunction is a major complication after spinal cord injury (SCI). In addition to local injury within the spinal cord, SCI causes major disruption to the peripheral organ innervation and regulation. The liver contains sympathetic, parasympathetic, and small sensory axons. The bi-directional signaling of sensory dorsal root ganglion (DRG) neurons that provide both efferent and afferent information is of key importance as it allows sensory neurons and peripheral organs to affect each other. SCI-induced liver inflammation precedes and may exacerbate intraspinal inflammation and pathology after SCI, which may be modulated by activity and exercise. In this study, we collected comprehensive gene expression data through RNA sequencing of liver tissue from rats with chronic SCI to determine the effects of activity and exercise on those expression patterns. The sequenced data are of high quality and show a high alignment rate to the Rn6 genome. Gene expression is demonstrated for genes associated with known liver pathologies. UCSC Genome Browser expression tracks are provided with the data to facilitate exploration of the samples.
Collapse
Affiliation(s)
- Julia H Chariker
- Department of Neuroscience Training, University of Louisville, 522 East Gray St., Louisville, KY, 40202, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, 522 East Gray St., Louisville, Kentucky, 40202, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - Cynthia Gomes
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Fiona Brabazon
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Wiley Publishing, Hoboken, NJ, 07030, USA
| | - Kathryn A Harman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Health & Sport Sciences, University of Louisville, 2100 South Floyd Street, Louisville, KY, 40208, USA
| | - Kathryn M DeVeau
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - David S K Magnuson
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Jeffrey C Petruska
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA.
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
| | - Eric C Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, 522 East Gray St., Louisville, Kentucky, 40202, USA.
- Department of Computer Engineering and Computer Science, Speed School of Engineering, University of Louisville, Duthie Center for Engineering, 2301 South 3rd St., Louisville, Kentucky, 40292, USA.
| |
Collapse
|
18
|
Chariker JH, Gomes C, Brabazon F, Harman KA, Ohri SS, Magnuson DSK, Whittemore SR, Petruska JC, Rouchka EC. Transcriptome of dorsal root ganglia caudal to a spinal cord injury with modulated behavioral activity. Sci Data 2019; 6:83. [PMID: 31175296 PMCID: PMC6555821 DOI: 10.1038/s41597-019-0088-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating clinical condition resulting in significant disabilities. Apart from local injury within the spinal cord, SCI patients develop a myriad of complications including multi-organ dysfunction. Some of the dysfunctions may be directly or indirectly related to the sensory neurons of the dorsal root ganglia (DRG), which signal to both the spinal cord and the peripheral organs. After SCI, some classes of DRG neurons exhibit sensitization and undergo axonal sprouting both peripherally and centrally. Such physiological and anatomical re-organization after SCI contributes to both adaptive and maladaptive plasticity processes, which may be modulated by activity and exercise. In this study, we collected comprehensive gene expression data in whole DRG below the levels of the injury to compare the effects of SCI with and without two different forms of exercise in rats.
Collapse
Affiliation(s)
- Julia H Chariker
- Department of Neuroscience Training, University of Louisville, 522 East Gray Street, Louisville, Kentucky, 40202, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, 522 East Gray Street, Louisville, Kentucky, 40202, USA
| | - Cynthia Gomes
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
| | - Fiona Brabazon
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
- Wiley Publishing, Hoboken, NJ, 07030, USA
| | - Kathryn A Harman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Health & Sport Sciences, University of Louisville, 2100 South Floyd Street, Louisville, KY, 40208, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - David S K Magnuson
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - Scott R Whittemore
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA
| | - Jeffrey C Petruska
- Department of Anatomical Sciences and Neurobiology, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, 511 South Floyd St., Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Suite 1500, Louisville, KY, 40202, USA.
| | - Eric C Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, 522 East Gray Street, Louisville, Kentucky, 40202, USA.
- Department of Computer Engineering and Computer Science, Speed School of Engineering, University of Louisville, Duthie Center for Engineering, 2301 South 3rd St., Louisville, Kentucky, 40292, USA.
| |
Collapse
|
19
|
Keller AV, Hainline C, Rees K, Krupp S, Prince D, Wood BD, Shum-Siu A, Burke DA, Petruska JC, Magnuson DSK. Nociceptor-dependent locomotor dysfunction after clinically-modeled hindlimb muscle stretching in adult rats with spinal cord injury. Exp Neurol 2019; 318:267-276. [PMID: 30880143 DOI: 10.1016/j.expneurol.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 01/10/2023]
Abstract
In the course of investigating how common clinical treatments and adaptive technologies affect recovery after spinal cord injury (SCI), we discovered that a clinically-modeled hindlimb stretching protocol dramatically, but transiently, reduces locomotor function. Nociceptive sensory input is capable of altering motor output at the spinal level, and nociceptive neurons are sensitized after SCI. Here we tested the hypotheses that stretch-induced locomotor deficits are dependent on nociceptive afferents by depleting TRPV1+ sensory afferents using capsaicin injections in neonatal rats. Following maturation, animals received 25g-cm contusive SCI at T10. After plateau of locomotor recovery at 6 weeks, daily stretching was performed for 3 weeks, followed by 2 weeks without stretch, and again for two additional weeks. Animals were sacrificed 2 h after the last stretching session for histological assessments. Consistent with previous findings, stretch-induced drops in locomotor function were observed in nociceptor-intact animals but were nearly absent in nociceptor-depleted animals. These functional changes were accompanied by corresponding increases in the number of c-Fos+ nuclei throughout the lumbar enlargement. As expected, nociceptor-depleted animals had very little CGRP+ axonal innervation of the dorsal horn. Nociceptor-intact stretched animals had significantly higher levels of CGRP+ as compared to non-stretched SCI rats, suggesting that stretching promoted intraspinal CGRP+ sprouting. These results indicate that stretch-induced locomotor dysfunction in animals with incomplete SCI involves C-fibers, adding a negative post-SCI role to their adaptive roles (e.g., bladder control), and suggesting that the clinical use of muscle stretching to combat contractures and spasticity may be unintentionally detrimental to locomotor function.
Collapse
Affiliation(s)
- Anastasia V Keller
- Department of Physiology, University of Louisville, School of Medicine, HSC A 1115, 500 South Preston Street, Louisville, KY 40292, USA
| | - Casey Hainline
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Kathleen Rees
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Sarah Krupp
- Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA
| | - Daniella Prince
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Brittney D Wood
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Alice Shum-Siu
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA
| | - Jeffrey C Petruska
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA
| | - David S K Magnuson
- Kentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville, 220 Abraham Flexner Way, Louisville, KY 40202, USA; Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, 511 South Floyd, Room 111, Louisville, KY 40202, USA; Department of Physiology, University of Louisville, School of Medicine, HSC A 1115, 500 South Preston Street, Louisville, KY 40292, USA.
| |
Collapse
|
20
|
Loy K, Bareyre FM. Rehabilitation following spinal cord injury: how animal models can help our understanding of exercise-induced neuroplasticity. Neural Regen Res 2019; 14:405-412. [PMID: 30539806 PMCID: PMC6334617 DOI: 10.4103/1673-5374.245951] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spinal cord injury is a devastating condition that is followed by long and often unsuccessful recovery after trauma. The state of the art approach to manage paralysis and concomitant impairments is rehabilitation, which is the only strategy that has proven to be effective and beneficial for the patients over the last decades. How rehabilitation influences the remodeling of spinal axonal connections in patients is important to understand, in order to better target these changes and define the optimal timing and onset of training. While clinically the answers to these questions remain difficult to obtain, rodent models of rehabilitation like bicycling, treadmill training, swimming, enriched environments or wheel running that mimic clinical rehabilitation can be helpful to reveal the axonal changes underlying motor recovery. This review will focus on the different animal models of spinal cord injury rehabilitation and the underlying changes in neuronal networks that are improved by exercise and rehabilitation.
Collapse
Affiliation(s)
- Kristina Loy
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich, Germany
| | - Florence M Bareyre
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
21
|
Spiess MR, Steenbrink F, Esquenazi A. Getting the Best Out of Advanced Rehabilitation Technology for the Lower Limbs: Minding Motor Learning Principles. PM R 2018; 10:S165-S173. [PMID: 30269803 DOI: 10.1016/j.pmrj.2018.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/04/2018] [Accepted: 06/09/2018] [Indexed: 01/12/2023]
Abstract
Advanced technology, including gait-training devices, is increasingly being integrated into neurorehabilitation. However, to use gait-training devices to their optimal potential, it is important that they are applied in accordance with motor learning and locomotor training principles. In this article, we outline the most important principles and explain how advanced gait-training devices are best used to improve therapy outcome.
Collapse
|
22
|
Bhupal PK, Anderson KA, Shall GP, Lynn JD, Hoolsema KS, Rossignol J, Dunbar GL, Sandstrom MI. Behavioral and neurochemical responses derived from dopaminergic intrastriatal grafts in hemiparkinsonian rats engaged in a novel motor task. J Neurosci Methods 2018; 307:149-163. [PMID: 29924980 DOI: 10.1016/j.jneumeth.2018.06.005] [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: 12/05/2017] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Putative treatments derived from in vivo stem cell transplant-derived dopamine (DA) in hemiparkinsonian rats have been assessed via DA-agonist-induced rotations involving imbalanced intra-hemispheric striatal DA receptor stimulation. However, such tests obscure the natural responses of grafts to sensory stimuli, and drug-induced plasticity can modify the circuit being tested. Thus, we propose an alternative testing strategy using a novel water tank swimming apparatus. NEW METHOD Microdialysis was used to compare striatal DA levels when rats were: (1) in a rest-phase within a bowl-shaped apparatus, or (2) in an active forced-swim phase within a specially-equipped water tank. Resting-phase DA release levels were compared with active-phase levels obtained while rats were required to swim in the water-tank task. Behavioral variables such as asymmetric circling while swimming (rotations), front-limb strokes, and front-limb reaches were captured by a camera for analysis. RESULTS AND COMPARISON WITH EXISTING METHODS Transplanted cells had a very modest effect on percentage of contralateral front-limb strokes, but did not reduce lesion-induced rotational asymmetry in the swim task. Neither striatal DA levels, nor their breakdown products, were significantly different between transplanted and sham-transplanted groups. Our new behavioral test eliminates the need for pharmacological stimulation, enabling simultaneous assessment of DA released in resting and active phases to explore graft control. CONCLUSIONS Our new method allows for accurate assessments of stem cell therapy for PD as an alternative to "rotation" tests. Use of natural motivations to engage in sensory-driven motor tasks provides more accurate insights into ongoing graft-derived behavioral support.
Collapse
Affiliation(s)
- Parnit K Bhupal
- Neuroscience Graduate Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Kevin A Anderson
- Department of Psychology, Experimental Psychology Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Gabrielle P Shall
- Neuroscience Graduate Program, Central Michigan University, Mount Pleasant, MI, United States
| | - Jonathan D Lynn
- Department of Psychiatry, Wayne State University, Detroit, MI, United States
| | | | - Julien Rossignol
- Neuroscience Graduate Program, Central Michigan University, Mount Pleasant, MI, United States; College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Gary L Dunbar
- Neuroscience Graduate Program, Central Michigan University, Mount Pleasant, MI, United States; Department of Psychology, Experimental Psychology Program, Central Michigan University, Mount Pleasant, MI, United States; Field Neuroscience Institute, Saginaw, MI, United States
| | - Michael I Sandstrom
- Neuroscience Graduate Program, Central Michigan University, Mount Pleasant, MI, United States; Department of Psychology, Experimental Psychology Program, Central Michigan University, Mount Pleasant, MI, United States.
| |
Collapse
|
23
|
Electromyographic patterns of the rat hindlimb in response to muscle stretch after spinal cord injury. Spinal Cord 2018; 56:560-568. [PMID: 29459656 PMCID: PMC5984666 DOI: 10.1038/s41393-018-0069-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 01/06/2023]
Abstract
Study Design Experimental Study Objectives To characterize the specific hindlimb electromyographic (EMG) patterns in response to muscle stretch and to measure the applied forces during stretching in the rat model of moderate SCI. Setting Kentucky Spinal Cord Injury Research Center, Louisville, KY, USA Methods Female Sprague Dawley rats (n=4) were instrumented for telemetry-based EMG recording (right Rectus Femoris and Biceps Femoris) and received a moderate T10 spinal cord injury (SCI). The major hindlimb muscle groups were stretched using our clinically modeled protocol. The EMG responses were recorded biweekly for 8 weeks. The forces applied during stretching were measured using a custom-designed glove. Locomotor function was assessed using the BBB Open Field Locomotor Scale, 3D kinematics and gait analysis. Results Three main EMG patterns in response to stretch were identified: clonic-like, air-stepping and spasms. Torques applied during stretching ranged from 0.8–6 N*cm, and did not change significantly over the weeks of stretching. Two stretching sessions a week did not result in a significant disruption to locomotor function. Conclusions Stretching evokes EMG patterns in rats similar to those reported in humans including clonus and spasms. The torques used during stretching are comparable, based on the ratio of torque to body weight, to the few previously published studies that measured the forces and/or torques applied by physical therapists when stretching patients. Future studies are warranted to fully explore the impact of muscle stretch on spinal cord function after injury. Sponsorship DoD, KSCHIRT, NIH
Collapse
|
24
|
Reversible silencing of lumbar spinal interneurons unmasks a task-specific network for securing hindlimb alternation. Nat Commun 2017; 8:1963. [PMID: 29213073 PMCID: PMC5719045 DOI: 10.1038/s41467-017-02033-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/02/2017] [Indexed: 12/05/2022] Open
Abstract
Neural circuitry in the lumbar spinal cord governs two principal features of locomotion, rhythm and pattern, which reflect intra- and interlimb movement. These features are functionally organized into a hierarchy that precisely controls stepping in a stereotypic, speed-dependent fashion. Here, we show that a specific component of the locomotor pattern can be independently manipulated. Silencing spinal L2 interneurons that project to L5 selectively disrupts hindlimb alternation allowing a continuum of walking to hopping to emerge from the otherwise intact network. This perturbation, which is independent of speed and occurs spontaneously with each step, does not disrupt multi-joint movements or forelimb alternation, nor does it translate to a non-weight-bearing locomotor activity. Both the underlying rhythm and the usual relationship between speed and spatiotemporal characteristics of stepping persist. These data illustrate that hindlimb alternation can be manipulated independently from other core features of stepping, revealing a striking freedom in an otherwise precisely controlled system. Intra- and interlimb coordination during locomotion is governed by hierarchically organized lumbar spinal networks. Here, the authors show that reversible silencing of spinal L2–L5 interneurons specifically disrupts hindlimb alternation leading to a continuum of walking to hopping.
Collapse
|
25
|
DeVeau KM, Harman KA, Squair JW, Krassioukov AV, Magnuson DSK, West CR. A comparison of passive hindlimb cycling and active upper-limb exercise provides new insights into systolic dysfunction after spinal cord injury. Am J Physiol Heart Circ Physiol 2017; 313:H861-H870. [PMID: 28710067 PMCID: PMC9925118 DOI: 10.1152/ajpheart.00046.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/05/2017] [Accepted: 07/12/2017] [Indexed: 12/24/2022]
Abstract
Active upper-limb and passive lower-limb exercise are two interventions used in the spinal cord injury (SCI) population. Although the global cardiac responses have been previously studied, it is unclear how either exercise influences contractile cardiac function. Here, the cardiac contractile and volumetric responses to upper-limb (swim) and passive lower-limb exercise were investigated in rodents with a severe high-thoracic SCI. Animals were divided into control (CON), SCI no exercise (NO-EX), SCI passive hindlimb cycling (PHLC), or SCI swim (SWIM) groups. Severe contusion SCI was administered at the T2 level. PHLC and SWIM interventions began on day 8 postinjury and lasted 25 days. Echocardiography and dobutamine stress echocardiography were performed before and after injury. Cardiac contractile indexes were assessed in vivo at study termination via a left ventricular pressure-volume conductance catheter. Stroke volume was reduced after SCI (91 µl in the NO-EX group vs. 188 µl in the CON group, P < 0.05) and was reversed at study termination in the PHLC (167 µl) but not SWIM (90 µl) group. Rates of contraction were reduced in NO-EX versus CON groups (6,079 vs. 9,225 mmHg, respectively, P < 0.05) and were unchanged by PHLC and SWIM training. Similarly, end-systolic elastance was reduced in the NO-EX versus CON groups (0.67 vs. 1.37 mmHg/µl, respectively, P < 0.05) and was unchanged by PHLC or SWIM training. Dobutamine infusion normalized all pressure indexes in each SCI group (all P < 0.05). In conclusion, PHLC improves flow-derived cardiac indexes, whereas SWIM training displayed no cardiobeneficial effect. Pressure-derived deficits were corrected only with dobutamine, suggesting that reduced β-adrenergic stimulation is principally responsible for the impaired cardiac contractile function after SCI.NEW & NOTEWORTHY This is the first direct comparison between the cardiac changes elicited by active upper-limb or passive lower-limb exercise after spinal cord injury. Here, we demonstrate that lower-limb exercise positively influences flow-derived cardiac indexes, whereas upper-limb exercise does not. Furthermore, neither intervention corrects the cardiac contractile dysfunction associated with spinal cord injury.
Collapse
Affiliation(s)
- Kathryn M. DeVeau
- 1International Collaboration on Repair Discoveries, Vancouver, British Columbia, Canada; ,2Kentucky Spinal Cord Injury Research Institute, University of Louisville, Louisville, Kentucky;
| | - Kathryn A. Harman
- 1International Collaboration on Repair Discoveries, Vancouver, British Columbia, Canada; ,2Kentucky Spinal Cord Injury Research Institute, University of Louisville, Louisville, Kentucky;
| | - Jordan W. Squair
- 1International Collaboration on Repair Discoveries, Vancouver, British Columbia, Canada; ,3MD/PhD Training Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada;
| | - Andrei V. Krassioukov
- 4Division of Physical Medicine and Rehabilitation, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; ,5GF Strong Rehabilitation Centre, Vancouver Health Authority, Vancouver, British Columbia, Canada; and
| | - David S. K. Magnuson
- 2Kentucky Spinal Cord Injury Research Institute, University of Louisville, Louisville, Kentucky;
| | - Christopher R. West
- 1International Collaboration on Repair Discoveries, Vancouver, British Columbia, Canada; ,6School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Saunders NR, Dziegielewska KM, Whish SC, Hinds LA, Wheaton BJ, Huang Y, Henry S, Habgood MD. A bipedal mammalian model for spinal cord injury research: The tammar wallaby. F1000Res 2017; 6:921. [PMID: 28721206 PMCID: PMC5497825 DOI: 10.12688/f1000research.11712.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Most animal studies of spinal cord injury are conducted in quadrupeds, usually rodents. It is unclear to what extent functional results from such studies can be translated to bipedal species such as humans because bipedal and quadrupedal locomotion involve very different patterns of spinal control of muscle coordination. Bipedalism requires upright trunk stability and coordinated postural muscle control; it has been suggested that peripheral sensory input is less important in humans than quadrupeds for recovery of locomotion following spinal injury. Methods: We used an Australian macropod marsupial, the tammar wallaby
(Macropuseugenii), because tammars exhibit an upright trunk posture, human-like alternating hindlimb movement when swimming and bipedal over-ground locomotion. Regulation of their muscle movements is more similar to humans than quadrupeds. At different postnatal (P) days (P7–60) tammars received a complete mid-thoracic spinal cord transection. Morphological repair, as well as functional use of hind limbs, was studied up to the time of their pouch exit. Results: Growth of axons across the lesion restored supraspinal innervation in animals injured up to 3 weeks of age but not in animals injured after 6 weeks of age. At initial pouch exit (P180), the young injured at P7-21 were able to hop on their hind limbs similar to age-matched controls and to swim albeit with a different stroke. Those animals injured at P40-45 appeared to be incapable of normal use of hind limbs even while still in the pouch. Conclusions: Data indicate that the characteristic over-ground locomotion of tammars provides a model in which regrowth of supraspinal connections across the site of injury can be studied in a bipedal animal. Forelimb weight-bearing motion and peripheral sensory input appear not to compensate for lack of hindlimb control, as occurs in quadrupeds. Tammars may be a more appropriate model for studies of therapeutic interventions relevant to humans.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Katarzyna M Dziegielewska
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Sophie C Whish
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Lyn A Hinds
- Health and Biosecurity Business Unit, Commonwealth Science and Industrial Research Organisation (CSIRO), Canberra, ACT, 2601, Australia
| | - Benjamin J Wheaton
- Centre for Evolutionary and Theoretical Immunology, The University of New Mexico, Albuquerque, NM, 87131, USA
| | - Yifan Huang
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Steve Henry
- Health and Biosecurity Business Unit, Commonwealth Science and Industrial Research Organisation (CSIRO), Canberra, ACT, 2601, Australia
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| |
Collapse
|
27
|
Liao CF, Yang TY, Chen YH, Yao CH, Way TD, Chen YS. Effects of swimming exercise on nerve regeneration in a rat sciatic nerve transection model. Biomedicine (Taipei) 2017; 7:3. [PMID: 28474579 PMCID: PMC5439339 DOI: 10.1051/bmdcn/2017070103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 12/28/2022] Open
Abstract
Background: Swimming is commonly considered to be an efficient rehabilitation exercise to treat peripheral nerve injury. However, the most effective resistance level and exercise duration is still unclear. We investigated the effects and mechanisms of swimming at various exertion levels in a rat sciatic nerve transection model. Methods: Sciatic nerve transection rats were randomized into the following four groups based on swimming duration (from the 7th day to the 28th day post-surgery): sedentary control group (SC), S10 group (10 min/3 times/week), S20 group (20 min/3 times/week), and S30 group (30 min/3 times/week) (n = 10 each). Axon regeneration, electrophysiological properties, muscular weights, macrophage infiltration, and nerve repair associated maker, calcitonin gene-related peptide (CGRP), were measured. Results: Dramatic higher successful percentages of nerve regeneration across the 10-mm gaps in swimming groups compared to the SC group. Total area of nerve regeneration significantly improved in the S10 group; however, electrophysiological properties, muscular weights, and macrophage infiltration in the regenerated nerves of rats did not differ significantly between the various exercise groups. CGRP expression was significantly increased in the spinal cord of rats in the S20 group. Conclusions: Our data indicated that CGRP-related axonal regeneration improved significantly with moderate swimming. These results should inspire new studies in physiotherapeutic practice for related human treatment.
Collapse
Affiliation(s)
- Chien-Fu Liao
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
| | - Tse-Yen Yang
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 404, Taiwan - Department of Psychology, Asia University, Wufeng District, Taichung 413, Taiwan
| | - Chun-Hsu Yao
- Biomaterials Translational Research Center, China Medical University Hospital, Taichung 404, Taiwan - Department of Biomedical Informatics, Asia University, Wufeng District, Taichung 413, Taiwan
| | - Tzong-Der Way
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan
| | - Yueh-Sheng Chen
- Biomaterials Translational Research Center, China Medical University Hospital, Taichung 404, Taiwan - Department of Biomedical Informatics, Asia University, Wufeng District, Taichung 413, Taiwan - Lab of Biomaterials, School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
28
|
Ryu Y, Ogata T, Nagao M, Kitamura T, Morioka K, Ichihara Y, Doi T, Sawada Y, Akai M, Nishimura R, Fujita N. The swimming test is effective for evaluating spasticity after contusive spinal cord injury. PLoS One 2017; 12:e0171937. [PMID: 28182676 PMCID: PMC5300247 DOI: 10.1371/journal.pone.0171937] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/28/2017] [Indexed: 12/27/2022] Open
Abstract
Spasticity is a frequent chronic complication in individuals with spinal cord injury (SCI). However, the severity of spasticity varies in patients with SCI. Therefore, an evaluation method is needed to determine the severity of spasticity. We used a contusive SCI model that is suitable for clinical translation. In this study, we examined the feasibility of the swimming test and an EMG for evaluating spasticity in a contusive SCI rat model. Sprague-Dawley rats received an injury at the 8th thoracic vertebra. Swimming tests were performed 3 to 6 weeks after SCI induction. We placed the SCI rats into spasticity-strong or spasticity-weak groups based on the frequency of spastic behavior during the swimming test. Subsequently, we recorded the Hoffman reflex (H-reflex) and examined the immunoreactivity of serotonin (5-HT) and its receptor (5-HT2A) in the spinal tissues of the SCI rats. The spasticity-strong group had significantly decreased rate-dependent depression of the H-reflex compared to the spasticity-weak group. The area of 5-HT2A receptor immunoreactivity was significantly increased in the spasticity-strong group. Thus, both electrophysiological and histological evaluations indicate that the spasticity-strong group presented with a more severe upper motor neuron syndrome. We also observed the groups in their cages for 20 hours. Our results suggest that the swimming test provides an accurate evaluation of spasticity in this contusive SCI model. We believe that the swimming test is an effective method for evaluating spastic behaviors and developing treatments targeting spasticity after SCI.
Collapse
Affiliation(s)
- Youngjae Ryu
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Toru Ogata
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
- * E-mail:
| | - Motoshi Nagao
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Taku Kitamura
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Kazuhito Morioka
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
- Department of Neurosurgery, Brain and Spinal Injury Center, University of California, San Francisco, California, United States of America
| | - Yoshinori Ichihara
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Toru Doi
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Yasuhiro Sawada
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
| | - Masami Akai
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center, Saitama, Japan
- Graduate School, International University of Health and Welfare, Tokyo, Japan
| | - Ryohei Nishimura
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoki Fujita
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Challenging cardiac function post-spinal cord injury with dobutamine. Auton Neurosci 2016; 209:19-24. [PMID: 28065654 DOI: 10.1016/j.autneu.2016.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/28/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022]
Abstract
There is general consensus that spinal cord injuries (SCI) above T6 result in altered sympathetic control of the heart, which negatively influences cardiac structure and function. To by-pass disrupted circuitry and investigate cardiac responses under enhanced sympathetic activity we utilized dobutamine (DOB) stress echocardiography. Animals were divided into a T2, 25g-cm contusive SCI (SCI) or an uninjured control (CON) group. Echocardiography was performed pre-SCI and at 1, 2 and 6weeks post-SCI. Increasing doses of DOB (5, 10 & 20μg/min/kg) were infused intravenously pre-SCI and at 1 and 6weeks post-SCI. Parasternal-short axis images were used to compare group differences in systolic function and track changes in response to SCI and DOB over time. One week post-SCI, stroke volume (SV), end diastolic volume (EDV), cardiac output (CO) and ejection fraction (EF) were all reduced compared to CON and these deficits persisted to 6weeks. We also found an increase in collagen deposition at 6weeks post SCI. Pre-SCI, DOB elicited a decrease in EDV and increases in CO, EF and HR but not SV. At 6weeks following SCI, in addition to increases in CO, EF and HR, DOB also induced increases in SV. This is the first report, to our knowledge, of DOB responses in a contusive SCI model with persistent cardiac impairments. The return of CO to pre-SCI levels and the substantial increase in SV at low DOB dosages shows that impaired descending control of the heart is directly contributing to reduced resting SV after SCI.
Collapse
|
30
|
Zehr EP, Barss TS, Dragert K, Frigon A, Vasudevan EV, Haridas C, Hundza S, Kaupp C, Klarner T, Klimstra M, Komiyama T, Loadman PM, Mezzarane RA, Nakajima T, Pearcey GEP, Sun Y. Neuromechanical interactions between the limbs during human locomotion: an evolutionary perspective with translation to rehabilitation. Exp Brain Res 2016; 234:3059-3081. [PMID: 27421291 PMCID: PMC5071371 DOI: 10.1007/s00221-016-4715-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 06/27/2016] [Indexed: 11/10/2022]
Abstract
During bipedal locomotor activities, humans use elements of quadrupedal neuronal limb control. Evolutionary constraints can help inform the historical ancestry for preservation of these core control elements support transfer of the huge body of quadrupedal non-human animal literature to human rehabilitation. In particular, this has translational applications for neurological rehabilitation after neurotrauma where interlimb coordination is lost or compromised. The present state of the field supports including arm activity in addition to leg activity as a component of gait retraining after neurotrauma.
Collapse
Affiliation(s)
- E P Zehr
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1.
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada.
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| | - Trevor S Barss
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| | - Katie Dragert
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
| | - Alain Frigon
- Department of Pharmacology-physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Erin V Vasudevan
- Department of Physical Therapy, SUNY Stony Brook University, Stony Brook, NY, USA
| | - Carlos Haridas
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
| | - Sandra Hundza
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
- Motion and Mobility Rehabilitation Laboratory, University of Victoria, Victoria, BC, Canada
| | - Chelsea Kaupp
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| | - Taryn Klarner
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| | - Marc Klimstra
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
- Motion and Mobility Rehabilitation Laboratory, University of Victoria, Victoria, BC, Canada
| | - Tomoyoshi Komiyama
- Division of Sports and Health Science, Chiba University, Chiba, Japan
- The United Graduate School of Education, Tokyo Gakugei University, Tokyo, Japan
| | - Pamela M Loadman
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| | - Rinaldo A Mezzarane
- Laboratory of Signal Processing and Motor Control, College of Physical Education, Universidade de Brasília-UnB, Brasília, Brazil
| | - Tsuyoshi Nakajima
- Department of Integrative Physiology, Kyorin University School of Medicine, Tokyo, Japan
| | - Gregory E P Pearcey
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| | - Yao Sun
- Rehabilitation Neuroscience Laboratory, University of Victoria, PO Box 3010 STN CSC, Victoria, BC, Canada, V8W 3P1
- Human Discovery Science, International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
31
|
Keller AVP, Wainwright G, Shum-Siu A, Prince D, Hoeper A, Martin E, Magnuson DSK. Disruption of Locomotion in Response to Hindlimb Muscle Stretch at Acute and Chronic Time Points after a Spinal Cord Injury in Rats. J Neurotrauma 2016; 34:661-670. [PMID: 27196003 DOI: 10.1089/neu.2015.4227] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
After spinal cord injury (SCI) muscle contractures develop in the plegic limbs of many patients. Physical therapists commonly use stretching as an approach to avoid contractures and to maintain the extensibility of soft tissues. We found previously that a daily stretching protocol has a negative effect on locomotor recovery in rats with mild thoracic SCI. The purpose of the current study was to determine the effects of stretching on locomotor function at acute and chronic time points after moderately severe contusive SCI. Female Sprague-Dawley rats with 25 g-cm T10 contusion injuries received our standard 24-min stretching protocol starting 4 days (acutely) or 10 weeks (chronically) post-injury (5 days/week for 5 or 4 weeks, respectively). Locomotor function was assessed using the BBB (Basso, Beattie, and Bresnahan) Open Field Locomotor Scale, video-based kinematics, and gait analysis. Locomotor deficits were evident in the acute animals after only 5 days of stretching and increasing the perceived intensity of stretching at week 4 resulted in greater impairment. Stretching initiated chronically resulted in dramatic decrements in locomotor function because most animals had BBB scores of 0-3 for weeks 2, 3, and 4 of stretching. Locomotor function recovered to control levels for both groups within 2 weeks once daily stretching ceased. Histological analysis revealed no apparent signs of overt and persistent damage to muscles undergoing stretching. The current study extends our observations of the stretching phenomenon to a more clinically relevant moderately severe SCI animal model. The results are in agreement with our previous findings and further demonstrate that spinal cord locomotor circuitry is especially vulnerable to the negative effects of stretching at chronic time points. While the clinical relevance of this phenomenon remains unknown, we speculate that stretching may contribute to the lack of locomotor recovery in some patients.
Collapse
Affiliation(s)
- Anastasia V P Keller
- 1 Kentucky Spinal Cord Injury Research Center, University of Louisville , Louisville, Kentucky.,2 Department of Physiology and Biophysics, University of Louisville , Louisville, Kentucky
| | - Grace Wainwright
- 5 Department of J.B. Speed School of Engineering, University of Louisville , Louisville, Kentucky
| | - Alice Shum-Siu
- 1 Kentucky Spinal Cord Injury Research Center, University of Louisville , Louisville, Kentucky.,3 Department of Neurological Surgery, University of Louisville , Louisville, Kentucky
| | - Daniella Prince
- 1 Kentucky Spinal Cord Injury Research Center, University of Louisville , Louisville, Kentucky.,3 Department of Neurological Surgery, University of Louisville , Louisville, Kentucky
| | - Alyssa Hoeper
- 5 Department of J.B. Speed School of Engineering, University of Louisville , Louisville, Kentucky
| | - Emily Martin
- 5 Department of J.B. Speed School of Engineering, University of Louisville , Louisville, Kentucky
| | - David S K Magnuson
- 1 Kentucky Spinal Cord Injury Research Center, University of Louisville , Louisville, Kentucky.,3 Department of Neurological Surgery, University of Louisville , Louisville, Kentucky.,4 Department of Anatomical Sciences and Neurobiology, University of Louisville , Louisville, Kentucky
| |
Collapse
|
32
|
Koehn LM, Noor NM, Dong Q, Er SY, Rash LD, King GF, Dziegielewska KM, Saunders NR, Habgood MD. Selective inhibition of ASIC1a confers functional and morphological neuroprotection following traumatic spinal cord injury. F1000Res 2016; 5:1822. [PMID: 28105306 PMCID: PMC5200949 DOI: 10.12688/f1000research.9094.2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Tissue loss after spinal trauma is biphasic, with initial mechanical/haemorrhagic damage at the time of impact being followed by gradual secondary expansion into adjacent, previously unaffected tissue. Limiting the extent of this secondary expansion of tissue damage has the potential to preserve greater residual spinal cord function in patients. The acute tissue hypoxia resulting from spinal cord injury (SCI) activates acid-sensing ion channel 1a (ASIC1a). We surmised that antagonism of this channel should provide neuroprotection and functional preservation after SCI. We show that systemic administration of the spider-venom peptide PcTx1, a selective inhibitor of ASIC1a, improves locomotor function in adult Sprague Dawley rats after thoracic SCI. The degree of functional improvement correlated with the degree of tissue preservation in descending white matter tracts involved in hind limb locomotor function. Transcriptomic analysis suggests that PcTx1-induced preservation of spinal cord tissue does not result from a reduction in apoptosis, with no evidence of down-regulation of key genes involved in either the intrinsic or extrinsic apoptotic pathways. We also demonstrate that trauma-induced disruption of blood-spinal cord barrier function persists for at least 4 days post-injury for compounds up to 10 kDa in size, whereas barrier function is restored for larger molecules within a few hours. This temporary loss of barrier function provides a “
treatment window” through which systemically administered drugs have unrestricted access to spinal tissue in and around the sites of trauma. Taken together, our data provide evidence to support the use of ASIC1a inhibitors as a therapeutic treatment for SCI. This study also emphasizes the importance of objectively grading the functional severity of initial injuries (even when using standardized impacts) and we describe a simple scoring system based on hind limb function that could be adopted in future studies.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Natassya M Noor
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Qing Dong
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Sing-Yan Er
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Lachlan D Rash
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Glenn F King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | | | - Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| |
Collapse
|
33
|
Koehn LM, Noor NM, Dong Q, Er SY, Rash LD, King GF, Dziegielewska KM, Saunders NR, Habgood MD. Selective inhibition of ASIC1a confers functional and morphological neuroprotection following traumatic spinal cord injury. F1000Res 2016; 5:1822. [PMID: 28105306 PMCID: PMC5200949 DOI: 10.12688/f1000research.9094.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2016] [Indexed: 11/10/2023] Open
Abstract
Tissue loss after spinal trauma is biphasic, with initial mechanical/haemorrhagic damage at the time of impact being followed by gradual secondary expansion into adjacent, previously unaffected tissue. Limiting the extent of this secondary expansion of tissue damage has the potential to preserve greater residual spinal cord function in patients. The acute tissue hypoxia resulting from spinal cord injury (SCI) activates acid-sensing ion channel 1a (ASIC1a). We surmised that antagonism of this channel should provide neuroprotection and functional preservation after SCI. We show that systemic administration of the spider-venom peptide PcTx1, a selective inhibitor of ASIC1a, improves locomotor function in adult Sprague Dawley rats after thoracic SCI. The degree of functional improvement correlated with the degree of tissue preservation in descending white matter tracts involved in hind limb locomotor function. Transcriptomic analysis suggests that PcTx1-induced preservation of spinal cord tissue does not result from a reduction in apoptosis, with no evidence of down-regulation of key genes involved in either the intrinsic or extrinsic apoptotic pathways. We also demonstrate that trauma-induced disruption of blood-spinal cord barrier function persists for at least 4 days post-injury for compounds up to 10 kDa in size, whereas barrier function is restored for larger molecules within a few hours. This temporary loss of barrier function provides a " treatment window" through which systemically administered drugs have unrestricted access to spinal tissue in and around the sites of trauma. Taken together, our data provide evidence to support the use of ASIC1a inhibitors as a therapeutic treatment for SCI. This study also emphasizes the importance of objectively grading the functional severity of initial injuries (even when using standardized impacts) and we describe a simple scoring system based on hind limb function that could be adopted in future studies.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Natassya M Noor
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Qing Dong
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Sing-Yan Er
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - Lachlan D Rash
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Glenn F King
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | | | - Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Australia
| |
Collapse
|
34
|
Training-Induced Functional Gains following SCI. Neural Plast 2016; 2016:4307694. [PMID: 27403345 PMCID: PMC4926009 DOI: 10.1155/2016/4307694] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/27/2016] [Indexed: 12/30/2022] Open
Abstract
We previously demonstrated that daily, hour-long training sessions significantly improved both locomotor (limb kinematics, gait, and hindlimb flexor-extensor bursting patterns) and nonlocomotor (bladder function and at-level mechanical allodynia) functions following a moderate contusive spinal cord injury. The amount of training needed to achieve this recovery is unknown. Furthermore, whether this recovery is induced primarily by neuronal activity below the lesion or other aspects related to general exercise is unclear. Therefore, the current study objectives were to (1) test the efficacy of 30 minutes of step training for recovery following a clinically relevant contusion injury in male Wistar rats and (2) test the efficacy of training without hindlimb engagement. The results indicate that as little as 30 minutes of step training six days per week enhances overground locomotion in male rats with contusive spinal cord injury but does not alter allodynia or bladder function. Thirty minutes of forelimb-only exercise did not alter locomotion, allodynia, or bladder function, and neither training protocol altered the amount of in-cage activity. Taken together, locomotor improvements were facilitated by hindlimb step training for 30 minutes, but longer durations of training are required to affect nonlocomotor systems.
Collapse
|
35
|
Battistuzzo CR, Rank MM, Flynn JR, Morgan DL, Callister R, Callister RJ, Galea MP. Gait recovery following spinal cord injury in mice: Limited effect of treadmill training. J Spinal Cord Med 2016; 39:335-43. [PMID: 26781526 PMCID: PMC5073763 DOI: 10.1080/10790268.2015.1133017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Several studies in rodents with complete spinal cord transections have demonstrated that treadmill training improves stepping movements. However, results from studies in incomplete spinal cord injured animals have been conflicting and questions regarding the training dosage after injury remain unresolved. OBJECTIVES To assess the effects of treadmill-training regimen (20 minutes daily, 5 days a week) for 3, 6 or 9 weeks on the recovery of locomotion in hemisected SCI mice. METHODS A randomized and blinded controlled experimental trial used a mouse model of incomplete spinal cord injury (SCI). After a left hemisection at T10, adult male mice were randomized to trained or untrained groups. The trained group commenced treadmill training one week after surgery and continued for 3, 6 or 9 weeks. Quantitative kinematic gait analysis was used to assess the spatiotemporal characteristics of the left hindlimb prior to injury and at 1, 4, 7 and 10 weeks post-injury. RESULTS One week after injury there was no movement of the left hindlimb and some animals dragged their foot. Treadmill training led to significant improvements in step duration, but had limited effect on the hindlimb movement pattern. Locomotor improvements in trained animals were most evident at the hip and knee joints whereas recovery of ankle movement was limited, even after 9 weeks of treadmill training. CONCLUSION These results demonstrate that treadmill training may lead to only modest improvement in recovery of hindlimb movement after incomplete spinal cord injury in mice.
Collapse
Affiliation(s)
- Camila R. Battistuzzo
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, VIC, Australia,Corresponding author: Camila R. Battistuzzo. Department of Medicine, Royal Melbourne Hospital, Royal Park Campus, Royal Parade, 34–54 Poplar Road, 3052, VIC, Australia. E-mail:
| | - Michelle M. Rank
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Jamie R. Flynn
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - David L. Morgan
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, VIC, Australia
| | - Robin Callister
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Robert J. Callister
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Mary P. Galea
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, VIC, Australia
| |
Collapse
|
36
|
Smilde HA, Vincent JA, Baan GC, Nardelli P, Lodder JC, Mansvelder HD, Cope TC, Maas H. Changes in muscle spindle firing in response to length changes of neighboring muscles. J Neurophysiol 2016; 115:3146-55. [PMID: 27075540 DOI: 10.1152/jn.00937.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/05/2016] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle force can be transmitted to the skeleton, not only via its tendons of origin and insertion but also through connective tissues linking the muscle belly to surrounding structures. Through such epimuscular myofascial connections, length changes of a muscle may cause length changes within an adjacent muscle and hence, affect muscle spindles. The aim of the present study was to investigate the effects of epimuscular myofascial forces on feedback from muscle spindles in triceps surae muscles of the rat. We hypothesized that within an intact muscle compartment, muscle spindles not only signal length changes of the muscle in which they are located but can also sense length changes that occur as a result of changing the length of synergistic muscles. Action potentials from single afferents were measured intra-axonally in response to ramp-hold release (RHR) stretches of an agonistic muscle at different lengths of its synergist, as well as in response to synergist RHRs. A decrease in force threshold was found for both soleus (SO) and lateral gastrocnemius afferents, along with an increase in length threshold for SO afferents. In addition, muscle spindle firing could be evoked by RHRs of the synergistic muscle. We conclude that muscle spindles not only signal length changes of the muscle in which they are located but also local length changes that occur as a result of changing the length and relative position of synergistic muscles.
Collapse
Affiliation(s)
- Hiltsje A Smilde
- Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio
| | - Jake A Vincent
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio
| | - Guus C Baan
- Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Paul Nardelli
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; School of Applied Physiology and Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia; and
| | - Johannes C Lodder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tim C Cope
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio; School of Applied Physiology and Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia; and
| | - Huub Maas
- Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands;
| |
Collapse
|
37
|
Bernabei M, van Dieën JH, Maas H. Altered mechanical interaction between rat plantar flexors due to changes in intermuscular connectivity. Scand J Med Sci Sports 2016; 27:177-187. [PMID: 26773332 DOI: 10.1111/sms.12644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2015] [Indexed: 01/20/2023]
Abstract
Connective tissue formation following muscle injury and remedial surgery may involve changes in the stiffness and configuration of the connective tissues linking adjacent muscles. We investigated changes in mechanical interaction of muscles by implanting either a tissue-integrating mesh (n = 8) or an adhesion barrier (n = 8) to respectively increase or decrease the intermuscular connectivity between soleus muscle (SO) and the lateral gastrocnemius and plantaris complex (LG+PL) of the rat. As a measure of mechanical interaction, changes in SO tendon forces and proximal-distal LG+PL force differences in response to lengthening LG+PL proximally were assessed 1 and 2 weeks post-surgery. The extent of mechanical interaction was doubled 1 week post-implantation of the tissue-integrating mesh compared to an unaffected compartment (n = 8), and was more than four times higher 2 weeks post-surgery. This was found only for maximally activated muscles, but not when passive. Implanting the adhesion barrier did not result in a reduction of the mechanical interaction between these muscles. Our findings indicate that the ratio of force transmitted via myofascial, rather than myotendinous pathways, can increase substantially when the connectivity between muscles is enhanced. This improves our understanding of the consequences of connective tissue formation at the muscle boundary on skeletal muscle function.
Collapse
Affiliation(s)
- M Bernabei
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - J H van Dieën
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - H Maas
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Sandrow-Feinberg HR, Houlé JD. Exercise after spinal cord injury as an agent for neuroprotection, regeneration and rehabilitation. Brain Res 2015; 1619:12-21. [PMID: 25866284 PMCID: PMC4540698 DOI: 10.1016/j.brainres.2015.03.052] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a traumatic event from which there is limited recovery of function, despite the best efforts of many investigators to devise realistic therapeutic treatments. Partly this is due to the multifaceted nature of SCI, where there is considerable disarray and dysfunction secondary to the initial injury. Contributing to this secondary degeneration is neurotoxicity, vascular dysfunction, glial scarring, neuroinflammation, apoptosis and demyelination. It seems logical that addressing the need for neuroprotection, regeneration and rehabilitation will require different treatment strategies that may be applied at varied stages of the post-injury response. Here we focus on a single strategy, exercise/physical training, which appears to have multiple applications and benefits for an acute or chronic SCI. Exercise has been demonstrated to be advantageous at cellular and biochemical levels, as well as being of benefit for the whole animal or human subject. Data from our lab and others will be discussed to further elucidate the many positive aspects of implementing exercise following injury and to suggest that rehabilitation is not the sole target of a training regimen following SCI. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Harra R Sandrow-Feinberg
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine, 2900 Queen Lane, PA 19129, Philadelphia, United States
| | - John D Houlé
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine, 2900 Queen Lane, PA 19129, Philadelphia, United States.
| |
Collapse
|
39
|
May Z, Fouad K, Shum-Siu A, Magnuson DSK. Challenges of animal models in SCI research: Effects of pre-injury task-specific training in adult rats before lesion. Behav Brain Res 2015; 291:26-35. [PMID: 25975172 DOI: 10.1016/j.bbr.2015.04.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/28/2015] [Accepted: 04/30/2015] [Indexed: 11/19/2022]
Abstract
A rarely explored subject in animal research is the effect of pre-injury variables on behavioral outcome post-SCI. Low reporting of such variables may underlie some discrepancies in findings between laboratories. Particularly, intensive task-specific training before a SCI might be important, considering that sports injuries are one of the leading causes of SCI. Thus, individuals with SCI often underwent rigorous training before their injuries. In the present study, we asked whether training before SCI on a grasping task or a swimming task would influence motor recovery in rats. Swim pre-training impaired recovery of swimming 2 and 4 weeks post-injury. This result fits with the idea of motor learning interference, which posits that learning something new may disrupt learning of a new task; in this case, learning strategies to compensate for functional loss after SCI. In contrast to swimming, grasp pre-training did not influence grasping ability after SCI at any time point. However, grasp pre-trained rats attempted to grasp more times than untrained rats in the first 4 weeks post-injury. Also, lesion volume of grasp pre-trained rats was greater than that of untrained rats, a finding which may be related to stress or activity. The increased participation in rehabilitative training of the pre-trained rats in the early weeks post-injury may have potentiated spontaneous plasticity in the spinal cord and counteracted the deleterious effect of interference and bigger lesions. Thus, our findings suggest that pre-training plays a significant role in recovery after CNS damage and needs to be carefully controlled for.
Collapse
Affiliation(s)
- Zacnicte May
- Faculty of Rehabilitation Research, University of Alberta, Edmonton, AB, Canada
| | - Karim Fouad
- Faculty of Rehabilitation Research, University of Alberta, Edmonton, AB, Canada
| | - Alice Shum-Siu
- Department of Neurological Surgery, Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA
| | - David S K Magnuson
- Department of Neurological Surgery, Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
40
|
Bernabei M, van Dieën JH, Baan GC, Maas H. Significant mechanical interactions at physiological lengths and relative positions of rat plantar flexors. J Appl Physiol (1985) 2015; 118:427-36. [DOI: 10.1152/japplphysiol.00703.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In situ studies involving supraphysiological muscle lengths and relative positions have shown that connective tissue linkages connecting adjacent muscles can transmit substantial forces, but the physiological significance is still subject to debate. The present study investigates effects of such epimuscular myofascial force transmission in the rat calf muscles. Unlike previous approaches, we quantified the mechanical interaction between the soleus (SO) and the lateral gastrocnemius and plantaris complex (LG+PL) applying a set of muscle lengths and relative positions corresponding to the range of knee and ankle angles occurring during normal movements. In nine deeply anesthetized Wistar rats, the superficial posterior crural compartment was exposed, and distal and proximal tendons of LG+PL and the distal SO tendon were severed and connected to force transducers. The target muscles were excited simultaneously. We found that SO active and passive tendon force was substantially affected by proximally lengthening of LG+PL mimicking knee extension (10% and 0.8% of maximal active SO force, respectively; P < 0.05). Moreover, SO relative position significantly changed the LG+PL length-force relationship, resulting in nonunique values for passive slack-length and optimum-length estimates. We conclude that also, for physiological muscle conditions, isometric force of rat triceps surae muscles is determined by its muscle-tendon unit length as well as by the length and relative position of its synergists. This has implications for understanding the neuromechanics of skeletal muscle in normal and pathological conditions, as well as for studies relying on the assumption that muscles act as independent force actuators.
Collapse
Affiliation(s)
- Michel Bernabei
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, The Netherlands
| | - Jaap H. van Dieën
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, The Netherlands
| | - Guus C. Baan
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, The Netherlands
| | - Huub Maas
- Research Institute MOVE, Faculty of Human Movement Sciences, VU University Amsterdam, The Netherlands
| |
Collapse
|
41
|
Caudle KL, Atkinson DA, Brown EH, Donaldson K, Seibt E, Chea T, Smith E, Chung K, Shum-Siu A, Cron CC, Magnuson DSK. Hindlimb stretching alters locomotor function after spinal cord injury in the adult rat. Neurorehabil Neural Repair 2014; 29:268-77. [PMID: 25106555 DOI: 10.1177/1545968314543500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Stretching is a widely accepted standard-of-care therapy following spinal cord injury (SCI) that has not been systematically studied in animal models. OBJECTIVE To investigate the influence of a daily stretch-based physical therapy program on locomotor recovery in adult rats with moderate T9 contusive SCI. METHODS A randomized treatment and control study of stretching in an animal model of acute SCI. Moderate SCIs were delivered with the NYU Impactor. Daily stretching (30 min/day, 5 days/wk for 8 weeks) was provided by a team of animal handlers. Hindlimb function was assessed using the BBB Open Field Locomotor Scale and kinematically. Passive range-of-motion for each joint was determined weekly using a goniometer. RESULTS Declines in hindlimb function during overground stepping were observed for the first 4 weeks for stretched animals. BBB scores improved weeks 5 to 10 but remained below the control group. Stretched animals had significant deficits in knee passive range of motion starting at week 4 and for the duration of the study. Kinematic assessment showed decreased joint excursion during stepping that partially recovered beginning at week 5. CONCLUSION Stretch-based therapy significantly impaired functional recovery in adult rats with a moderate contusive SCI at T10. The negative impact on function was greatest acutely but persisted even after the stretching ceased at 8 weeks postinjury.
Collapse
Affiliation(s)
| | | | | | | | - Erik Seibt
- University of Louisville, Louisville KY, USA
| | - Tim Chea
- University of Louisville, Louisville KY, USA
| | - Erin Smith
- University of Louisville, Louisville KY, USA
| | | | | | | | | |
Collapse
|
42
|
Ward PJ, Herrity AN, Smith RR, Willhite A, Harrison BJ, Petruska JC, Harkema SJ, Hubscher CH. Novel multi-system functional gains via task specific training in spinal cord injured male rats. J Neurotrauma 2014; 31:819-33. [PMID: 24294909 DOI: 10.1089/neu.2013.3082] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Locomotor training (LT) after spinal cord injury (SCI) is a rehabilitative therapy used to enhance locomotor recovery. There is evidence, primarily anecdotal, also associating LT with improvements in bladder function and reduction in some types of SCI-related pain. In the present study, we determined if a step training paradigm could improve outcome measures of locomotion, bladder function, and pain/allodynia. After a T10 contusive SCI trained animals (adult male Wistar rats), trained animals began quadrupedal step training beginning 2 weeks post-SCI for 1 h/day. End of study experiments (3 months of training) revealed significant changes in limb kinematics, gait, and hindlimb flexor-extensor bursting patterns relative to non-trained controls. Importantly, micturition function, evaluated with terminal transvesical cystometry, was significantly improved in the step trained group (increased voiding efficiency, intercontraction interval, and contraction amplitude). Because both SCI and LT affect neurotrophin signaling, and neurotrophins are involved with post-SCI plasticity in micturition pathways, we measured bladder neurotrophin mRNA. Training regulated the expression of nerve growth factor (NGF) but not BDNF or NT3. Bladder NGF mRNA levels were inversely related to bladder function in the trained group. Monitoring of overground locomotion and neuropathic pain throughout the study revealed significant improvements, beginning after 3 weeks of training, which in both cases remained consistent for the study duration. These novel findings, improving non-locomotor in addition to locomotor functions, demonstrate that step training post-SCI could contribute to multiple quality of life gains, targeting patient-centered high priority deficits.
Collapse
Affiliation(s)
- Patricia J Ward
- 1 Department of Anatomical Sciences and Neurobiology, University of Louisville , Louisville, Kentucky
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Elevated MMP-9 in the lumbar cord early after thoracic spinal cord injury impedes motor relearning in mice. J Neurosci 2013; 33:13101-11. [PMID: 23926264 DOI: 10.1523/jneurosci.1576-13.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spinal cord injury results in distant pathology around putative locomotor networks that may jeopardize the recovery of locomotion. We previously showed that activated microglia and increased cytokine expression extend at least 10 segments below the injury to influence sensory function. Matrix metalloproteinase-9 (MMP-9) is a potent regulator of acute neuroinflammation. Whether MMP-9 is produced remote to the injury or influences locomotor plasticity remains unexamined. Therefore, we characterized the lumbar enlargement after a T9 spinal cord injury in C57BL/6 (wild-type [WT]) and MMP-9-null (knock-out [KO]) mice. Within 24 h, resident microglia displayed an activated phenotype alongside increased expression of progelatinase MMP-3 in WT mice. By 7 d, increases in active MMP-9 around lumbar vasculature and production of proinflammatory TNF-α were evident. Deletion of MMP-9 attenuated remote microglial activation and restored TNF-α expression to homeostatic levels. To determine whether MMP-9 impedes locomotor plasticity, we delivered lumbar-focused treadmill training in WT and KO mice during early (2-9 d) or late (35-42 d) phases of recovery. Robust behavioral improvements were observed by 7 d, when only trained KO mice stepped in the open field. Locomotor improvements were retained for 4 weeks as identified using state of the art mouse kinematics. Neither training nor MMP-9 depletion alone promoted recovery. The same intervention delivered late was ineffective, suggesting that lesion site sparing is insufficient to facilitate activity-based training and recovery. Our work suggests that by attenuating remote mechanisms of inflammation, acute treadmill training can harness endogenous spinal plasticity to promote robust recovery.
Collapse
|
44
|
Harel NY, Yigitkanli K, Fu Y, Cafferty WBJ, Strittmatter SM. Multimodal exercises simultaneously stimulating cortical and brainstem pathways after unilateral corticospinal lesion. Brain Res 2013; 1538:17-25. [PMID: 24055330 DOI: 10.1016/j.brainres.2013.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/05/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022]
Abstract
In the context of injury to the corticospinal tract (CST), brainstem-origin circuits may provide an alternative system of descending motor influence. However, subcortical circuits are largely under subconscious control. To improve volitional control over spared fibers after CST injury, we hypothesized that a combination of physical exercises simultaneously stimulating cortical and brainstem pathways above the injury would strengthen corticobulbar connections through Hebbian-like mechanisms. We sought to test this hypothesis in mice with unilateral CST lesions. Ten days after pyramidotomy, mice were randomized to four training groups: (1) postural exercises designed to stimulate brainstem pathways (BS); (2) distal limb-grip exercises preferentially stimulating CST pathways (CST); (3) simultaneous multimodal exercises (BS+CST); or (4) no training (NT). Behavioral and anatomical outcomes were assessed after 20 training sessions over 4 weeks. Mice in the BS+CST training group showed a trend toward greater improvements in skilled limb performance than mice in the other groups. There were no consistent differences between training groups in gait kinematics. Anatomically, multimodal BS+CST training neither increased corticobulbar fiber density of the lesioned CST rostral to the lesion nor collateral sprouting of the unlesioned CST caudal to the lesion. Further studies should incorporate electrophysiological assessment to gauge changes in synaptic strength of direct and indirect pathways between the cortex and spinal cord in response to multimodal exercises.
Collapse
Affiliation(s)
- Noam Y Harel
- Department of Neurology, and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, P.O. Box 208018, New Haven, CT 06520, USA.
| | | | | | | | | |
Collapse
|
45
|
Sheets AL, Lai PL, Fisher LC, Basso DM. Quantitative evaluation of 3D mouse behaviors and motor function in the open-field after spinal cord injury using markerless motion tracking. PLoS One 2013; 8:e74536. [PMID: 24058586 PMCID: PMC3776828 DOI: 10.1371/journal.pone.0074536] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/02/2013] [Indexed: 12/20/2022] Open
Abstract
Thousands of scientists strive to identify cellular mechanisms that could lead to breakthroughs in developing ameliorative treatments for debilitating neural and muscular conditions such as spinal cord injury (SCI). Most studies use rodent models to test hypotheses, and these are all limited by the methods available to evaluate animal motor function. This study’s goal was to develop a behavioral and locomotor assessment system in a murine model of SCI that enables quantitative kinematic measurements to be made automatically in the open-field by applying markerless motion tracking approaches. Three-dimensional movements of eight naïve, five mild, five moderate, and four severe SCI mice were recorded using 10 cameras (100 Hz). Background subtraction was used in each video frame to identify the animal’s silhouette, and the 3D shape at each time was reconstructed using shape-from-silhouette. The reconstructed volume was divided into front and back halves using k-means clustering. The animal’s front Center of Volume (CoV) height and whole-body CoV speed were calculated and used to automatically classify animal behaviors including directed locomotion, exploratory locomotion, meandering, standing, and rearing. More detailed analyses of CoV height, speed, and lateral deviation during directed locomotion revealed behavioral differences and functional impairments in animals with mild, moderate, and severe SCI when compared with naïve animals. Naïve animals displayed the widest variety of behaviors including rearing and crossing the center of the open-field, the fastest speeds, and tallest rear CoV heights. SCI reduced the range of behaviors, and decreased speed (r = .70 p<.005) and rear CoV height (r = .65 p<.01) were significantly correlated with greater lesion size. This markerless tracking approach is a first step toward fundamentally changing how rodent movement studies are conducted. By providing scientists with sensitive, quantitative measurement methods, subjectivity and human error is reduced, potentially providing insights leading to breakthroughs in treating human disease.
Collapse
Affiliation(s)
- Alison L. Sheets
- Department of Mechanical and Aeronautical Engineering, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Po-Lun Lai
- Civil, Environmental and Geodetic Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Lesley C. Fisher
- School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio, United States of America
| | - D. Michele Basso
- School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
46
|
Kuypers NJ, James KT, Enzmann GU, Magnuson DSK, Whittemore SR. Functional consequences of ethidium bromide demyelination of the mouse ventral spinal cord. Exp Neurol 2013; 247:615-22. [PMID: 23466931 DOI: 10.1016/j.expneurol.2013.02.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/19/2013] [Accepted: 02/25/2013] [Indexed: 02/08/2023]
Abstract
Ethidium bromide (EB) has been extensively used in the rat as a model of spinal cord demyelination. However, this lesion has not been addressed in the adult mouse, a model with unlimited genetic potential. Here we characterize behavioral function, inflammation, myelin status and axonal viability following bilateral injection of 0.20 mg/mL ethidium bromide or saline into the ventral white matter (VWM) of female C57Bl/6 mice. EB-induced VWM demyelination significantly reduced spared VWM and Basso Mouse Scale (BMS) scores persisting out to 2 months. Chronic hindlimb dysfunction was accompanied by a persistent inflammatory response (demonstrated by CD45(+) immunofluorescence) and axonal loss (demonstrated by NF-M immunofluorescence and electron microscopy; EM). These cellular responses differ from the rat where inflammation resolves by 3-4 weeks and axon loss is minimal following EB demyelination. As these data suggest that EB-injection in the mouse spinal cord is a non-remyelinating lesion, we sought to ask whether wheel running could promote recovery by enhancing plasticity of local lumbar circuitry independent of remyelination. This did not occur as BMS and Treadscan assessment revealed no significant effect of wheel running on recovery. However, this study defines the importance of descending ventral motor pathways to locomotor function in the mouse as VWM loss results in a chronic hindlimb deficit.
Collapse
Affiliation(s)
- Nicholas J Kuypers
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40292, USA
| | | | | | | | | |
Collapse
|
47
|
Weishaupt N, Vavrek R, Fouad K. Training following unilateral cervical spinal cord injury in rats affects the contralesional forelimb. Neurosci Lett 2013; 539:77-81. [DOI: 10.1016/j.neulet.2013.01.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/02/2013] [Accepted: 01/19/2013] [Indexed: 10/27/2022]
|
48
|
The pig model of chronic paraplegia: A challenge for experimental studies in spinal cord injury. Prog Neurobiol 2012; 97:288-303. [DOI: 10.1016/j.pneurobio.2012.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 01/22/2012] [Accepted: 04/17/2012] [Indexed: 12/27/2022]
|
49
|
Shah PK, Gerasimenko Y, Shyu A, Lavrov I, Zhong H, Roy RR, Edgerton VR. Variability in step training enhances locomotor recovery after a spinal cord injury. Eur J Neurosci 2012; 36:2054-62. [PMID: 22591277 DOI: 10.1111/j.1460-9568.2012.08106.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Performance of a motor task is improved by practicing a specific task with added 'challenges' to a training regimen. We tested the hypothesis that, in the absence of brain control, the performance of a motor task is enhanced by training using specific variations of that task. We utilized modifications of step performance training to improve the ability of spinal rats to forward step. After a complete thoracic spinal cord transection, 20 adult rats were divided randomly to bipedally step on a treadmill in the forward, sideward, or backward direction for 28 sessions (20 min, 5 days/week) and subsequently tested for their ability to step in the forward direction. Although the animals from all trained groups showed improvement, the rats in the sideward-trained and backward-trained groups had greater step consistency and coordination along with higher peak amplitudes and total integrated activity of the rectified electromyographic signals from selected hindlimb muscles per step during forward stepping than the rats in the forward-trained group. Our results demonstrate that, by retaining the fundamental features of a motor task (bipedal stepping), the ability to perform that motor task can be enhanced by the addition of specific contextual variations to the task (direction of stepping). Our data suggest that the forward stepping neuronal locomotor networks are partially complemented by synchronous activation of interneuronal/motoneuronal populations that are also a part of the sideward or backward stepping locomotor networks. Accordingly, the overlap and interaction of neuronal elements may play a critical role in positive task transference.
Collapse
Affiliation(s)
- Prithvi K Shah
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Battistuzzo CR, Callister RJ, Callister R, Galea MP. A systematic review of exercise training to promote locomotor recovery in animal models of spinal cord injury. J Neurotrauma 2012; 29:1600-13. [PMID: 22401139 DOI: 10.1089/neu.2011.2199] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In the early 1980s experiments on spinalized cats showed that exercise training on the treadmill could enhance locomotor recovery after spinal cord injury (SCI). In this review, we summarize the evidence for the effectiveness of exercise training aimed at promoting locomotor recovery in animal models of SCI. We performed a systematic search of the literature using Medline, Web of Science, and Embase. Of the 362 studies screened, 41 were included. The adult female rat was the most widely used animal model. The majority of studies (73%) reported that exercise training had a positive effect on some aspect of locomotor recovery. Studies employing a complete SCI were less likely to have positive outcomes. For incomplete SCI models, contusion was the most frequently employed method of lesion induction, and the degree of recovery depended on injury severity. Positive outcomes were associated with training regimens that involved partial weight-bearing activity, commenced within a critical period of 1-2 weeks after SCI, and maintained training for at least 8 weeks. Considerable heterogeneity in training paradigms and methods used to assess or quantify recovery was observed. A 13-item checklist was developed and employed to assess the quality of reporting and study design; only 15% of the studies had high methodological quality. We recommend that future studies include control groups, randomize animals to groups, conduct blinded assessments, report the extent of the SCI lesion, and report sample size calculations. A small battery of objective assessment methods including assessment of over-ground stepping should also be developed and routinely employed. This would allow future meta-analyses of the effectiveness of exercise interventions on locomotor recovery.
Collapse
Affiliation(s)
- Camila R Battistuzzo
- Department of Physiotherapy, Melbourne School of Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|