1
|
Conditional RAC1 knockout in motor neurons restores H-reflex rate-dependent depression after spinal cord injury. Sci Rep 2021; 11:7838. [PMID: 33837249 PMCID: PMC8035187 DOI: 10.1038/s41598-021-87476-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/30/2021] [Indexed: 12/27/2022] Open
Abstract
A major complication with spinal cord injury (SCI) is the development of spasticity, a clinical symptom of hyperexcitability within the spinal H-reflex pathway. We have previously demonstrated a common structural motif of dendritic spine dysgenesis associated with hyperexcitability disorders after injury or disease insults to the CNS. Here, we used an adeno-associated viral (AAV)-mediated Cre-Lox system to knockout Rac1 protein expression in motor neurons after SCI. Three weeks after AAV9-Cre delivery into the soleus/gastrocnemius of Rac1-“floxed” adult mice to retrogradely infect spinal alpha-motor neurons, we observed significant restoration of RDD and reduced H-reflex excitability in SCI animals. Additionally, viral-mediated Rac1 knockdown reduced presence of dendritic spine dysgenesis on motor neurons. In control SCI animals without Rac1 knockout, we continued to observe abnormal dendritic spine morphology associated with hyperexcitability disorder, including an increase in mature, mushroom dendritic spines, and an increase in overall spine length and spine head size. Taken together, our results demonstrate that viral-mediated disruption of Rac1 expression in ventral horn motor neurons can mitigate dendritic spine morphological correlates of neuronal hyperexcitability, and reverse hyperreflexia associated with spasticity after SCI. Finally, our findings provide evidence of a putative mechanistic relationship between motor neuron dendritic spine dysgenesis and SCI-induced spasticity.
Collapse
|
2
|
Thompson AK, Sinkjær T. Can Operant Conditioning of EMG-Evoked Responses Help to Target Corticospinal Plasticity for Improving Motor Function in People With Multiple Sclerosis? Front Neurol 2020; 11:552. [PMID: 32765389 PMCID: PMC7381136 DOI: 10.3389/fneur.2020.00552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/15/2020] [Indexed: 11/25/2022] Open
Abstract
Corticospinal pathway and its function are essential in motor control and motor rehabilitation. Multiple sclerosis (MS) causes damage to the brain and descending connections, and often diminishes corticospinal function. In people with MS, neural plasticity is available, although it does not necessarily remain stable over the course of disease progress. Thus, inducing plasticity to the corticospinal pathway so as to improve its function may lead to motor control improvements, which impact one's mobility, health, and wellness. In order to harness plasticity in people with MS, over the past two decades, non-invasive brain stimulation techniques have been examined for addressing common symptoms, such as cognitive deficits, fatigue, and spasticity. While these methods appear promising, when it comes to motor rehabilitation, just inducing plasticity or having a capacity for it does not guarantee generation of better motor functions. Targeting plasticity to a key pathway, such as the corticospinal pathway, could change what limits one's motor control and improve function. One of such neural training methods is operant conditioning of the motor-evoked potential that aims to train the behavior of the corticospinal-motoneuron pathway. Through up-conditioning training, the person learns to produce the rewarded neuronal behavior/state of increased corticospinal excitability, and through iterative training, the rewarded behavior/state becomes one's habitual, daily motor behavior. This minireview introduces operant conditioning approach for people with MS. Guiding beneficial CNS plasticity on top of continuous disease progress may help to prolong the duration of maintained motor function and quality of life in people living with MS.
Collapse
Affiliation(s)
- Aiko K Thompson
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Thomas Sinkjær
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.,Lundbeck Foundation, Copenhagen, Denmark
| |
Collapse
|
3
|
Thompson AK, Cote RH, Sniffen JM, Brangaccio JA. Operant conditioning of the tibialis anterior motor evoked potential in people with and without chronic incomplete spinal cord injury. J Neurophysiol 2018; 120:2745-2760. [PMID: 30207863 DOI: 10.1152/jn.00362.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The activity of corticospinal pathways is important in movement control, and its plasticity is essential for motor skill learning and re-learning after central nervous system (CNS) injuries. Therefore, enhancing the corticospinal function may improve motor function recovery after CNS injuries. Operant conditioning of stimulus-induced muscle responses (e.g., reflexes) is known to induce the targeted plasticity in a targeted pathway. Thus, an operant conditioning protocol to target the corticospinal pathways may be able to enhance the corticospinal function. To test this possibility, we investigated whether operant conditioning of the tibialis anterior (TA) motor evoked potential (MEP) to transcranial magnetic stimulation can enhance corticospinal excitability in people with and without chronic incomplete spinal cord injury (SCI). The protocol consisted of 6 baseline and 24 up-conditioning/control sessions over 10 wk. In all sessions, TA MEPs were elicited at 10% above active MEP threshold while the sitting participant provided a fixed preset level of TA background electromyographic activity. During baseline sessions, MEPs were simply measured. During conditioning trials of the conditioning sessions, the participant was encouraged to increase MEP and was given immediate feedback indicating whether MEP size was above a criterion. In 5/8 participants without SCI and 9/10 with SCI, over 24 up-conditioning sessions, MEP size increased significantly to ~150% of the baseline value, whereas the silent period (SP) duration decreased by ~20%. In a control group of participants without SCI, neither MEP nor SP changed. These results indicate that MEP up-conditioning can facilitate corticospinal excitation, which is essential for enhancing motor function recovery after SCI. NEW & NOTEWORTHY We investigated whether operant conditioning of the motor evoked potential (MEP) to transcranial magnetic stimulation can systematically increase corticospinal excitability for the ankle dorsiflexor tibialis anterior (TA) in people with and without chronic incomplete spinal cord injury. We found that up-conditioning can increase the TA MEP while reducing the accompanying silent period (SP) duration. These findings suggest that MEP up-conditioning produces the facilitation of corticospinal excitation as targeted, whereas it suppresses inhibitory mechanisms reflected in SP.
Collapse
Affiliation(s)
- Aiko K Thompson
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina , Charleston, South Carolina
| | - Rachel H Cote
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina , Charleston, South Carolina
| | - Janice M Sniffen
- Department of Physical Therapy, School of Health Technology and Management, Stony Brook University , Stony Brook, New York
| | - Jodi A Brangaccio
- Helen Hayes Hospital, New York State Department of Health, West Haverstraw, New York
| |
Collapse
|
4
|
Abstract
Recent advances in neuroscience and devices are ushering in a new generation of medical treatments. Engineered biodevices are demonstrating the potential to create long-term changes in neural circuits, termed neuroplasticity. Thus, the approach of engineering neuroplasticity is rapidly expanding, building on recent demonstrations of improved quality of life for people with movement disorders, epilepsy, and spinal cord injury. In addition, discovering the fundamental mechanisms of engineered neuroplasticity by leveraging anatomically well-documented systems like the spinal cord is likely to provide powerful insights into solutions for other neurotraumas, such as stroke and traumatic brain injury, as well as neurodegenerative disorders, such as Alzheimer's, Parkinson disease, and multiple sclerosis. Now is the time for advancing both the experimental neuroscience, device development, and pioneering human trials to reap the benefits of engineered neuroplasticity as a therapeutic approach for improving quality of life after spinal cord injury.
Collapse
Affiliation(s)
- Chet T Moritz
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA.
- Department of Physiology & Biophysics, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
- UW Institute of Neuroengineering (UWIN), University of Washington, Seattle, WA, USA.
- Washington Spinal Cord Injury Consortium, University of Washington, Seattle, WA, USA.
- Center for Sensorimotor Neural Engineering, Seattle, WA, USA.
- Department of Electrical Engineering, University of Washington , Box 356490, Seattle, WA, 98195, USA.
| |
Collapse
|
5
|
Rowe RK, Harrison JL, Zhang H, Bachstetter AD, Hesson DP, O'Hara BF, Greene MI, Lifshitz J. Novel TNF receptor-1 inhibitors identified as potential therapeutic candidates for traumatic brain injury. J Neuroinflammation 2018; 15:154. [PMID: 29789012 PMCID: PMC5964690 DOI: 10.1186/s12974-018-1200-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/13/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) begins with the application of mechanical force to the head or brain, which initiates systemic and cellular processes that are hallmarks of the disease. The pathological cascade of secondary injury processes, including inflammation, can exacerbate brain injury-induced morbidities and thus represents a plausible target for pharmaceutical therapies. We have pioneered research on post-traumatic sleep, identifying that injury-induced sleep lasting for 6 h in brain-injured mice coincides with increased cortical levels of inflammatory cytokines, including tumor necrosis factor (TNF). Here, we apply post-traumatic sleep as a physiological bio-indicator of inflammation. We hypothesized the efficacy of novel TNF receptor (TNF-R) inhibitors could be screened using post-traumatic sleep and that these novel compounds would improve functional recovery following diffuse TBI in the mouse. METHODS Three inhibitors of TNF-R activation were synthesized based on the structure of previously reported TNF CIAM inhibitor F002, which lodges into a defined TNFR1 cavity at the TNF-binding interface, and screened for in vitro efficacy of TNF pathway inhibition (IκB phosphorylation). Compounds were screened for in vivo efficacy in modulating post-traumatic sleep. Compounds were then tested for efficacy in improving functional recovery and verification of cellular mechanism. RESULTS Brain-injured mice treated with Compound 7 (C7) or SGT11 slept significantly less than those treated with vehicle, suggesting a therapeutic potential to target neuroinflammation. SGT11 restored cognitive, sensorimotor, and neurological function. C7 and SGT11 significantly decreased cortical inflammatory cytokines 3 h post-TBI. CONCLUSIONS Using sleep as a bio-indicator of TNF-R-dependent neuroinflammation, we identified C7 and SGT11 as potential therapeutic candidates for TBI.
Collapse
Affiliation(s)
- Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA. .,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA. .,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.
| | - Jordan L Harrison
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hongtao Zhang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, Spinal Cord and Brain Injury Research Center, and Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - David P Hesson
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bruce F O'Hara
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Mark I Greene
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA.,Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA.,Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
6
|
Estrada-Rojo F, Morales-Gomez J, Coballase-Urrutia E, Martinez-Vargas M, Navarro L. Diurnal variation of NMDA receptor expression in the rat cerebral cortex is associated with traumatic brain injury damage. BMC Res Notes 2018; 11:150. [PMID: 29467028 PMCID: PMC5822486 DOI: 10.1186/s13104-018-3258-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/14/2018] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVE Data from our laboratory suggest that recovery from a traumatic brain injury depends on the time of day at which it occurred. In this study, we examined whether traumatic brain injury -induced damage is related to circadian variation in N-methyl-D-aspartate receptor expression in rat cortex. RESULTS We confirmed that traumatic brain injury recovery depended on the time of day at which the damage occurred. We also found that motor cortex N-methyl-D-aspartate receptor subunit NR1 expression exhibited diurnal variation in both control and traumatic brain injury-subjected rats. However, this rhythm is more pronounced in traumatic brain injury-subjected rats, with minimum expression in those injured during nighttime hours. These findings suggest that traumatic brain injury occurrence times should be considered in future clinical studies and when designing neuroprotective strategies for patients.
Collapse
Affiliation(s)
- Francisco Estrada-Rojo
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico.,Programa de Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de México, Mexico City, Mexico
| | - Julio Morales-Gomez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | | | - Marina Martinez-Vargas
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Luz Navarro
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico.
| |
Collapse
|
7
|
Noain D, Büchele F, Schreglmann SR, Valko PO, Gavrilov YV, Morawska MM, Imbach LL, Baumann CR. Increased Sleep Need and Reduction of Tuberomammillary Histamine Neurons after Rodent Traumatic Brain Injury. J Neurotrauma 2018; 35:85-93. [DOI: 10.1089/neu.2017.5067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Daniela Noain
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Fabian Büchele
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Sebastian R. Schreglmann
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Philipp O. Valko
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Yuri V. Gavrilov
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Marta M. Morawska
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas L. Imbach
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Christian R. Baumann
- Department of Neurology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Albayram O, Kondo A, Mannix R, Smith C, Tsai CY, Li C, Herbert MK, Qiu J, Monuteaux M, Driver J, Yan S, Gormley W, Puccio AM, Okonkwo DO, Lucke-Wold B, Bailes J, Meehan W, Zeidel M, Lu KP, Zhou XZ. Cis P-tau is induced in clinical and preclinical brain injury and contributes to post-injury sequelae. Nat Commun 2017; 8:1000. [PMID: 29042562 PMCID: PMC5645414 DOI: 10.1038/s41467-017-01068-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 08/16/2017] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is characterized by acute neurological dysfunction and associated with the development of chronic traumatic encephalopathy (CTE) and Alzheimer's disease. We previously showed that cis phosphorylated tau (cis P-tau), but not the trans form, contributes to tau pathology and functional impairment in an animal model of severe TBI. Here we found that in human samples obtained post TBI due to a variety of causes, cis P-tau is induced in cortical axons and cerebrospinal fluid and positively correlates with axonal injury and clinical outcome. Using mouse models of severe or repetitive TBI, we showed that cis P-tau elimination with a specific neutralizing antibody administered immediately or at delayed time points after injury, attenuates the development of neuropathology and brain dysfunction during acute and chronic phases including CTE-like pathology and dysfunction after repetitive TBI. Thus, cis P-tau contributes to short-term and long-term sequelae after TBI, but is effectively neutralized by cis antibody treatment.Induction of the cis form of phosphorylated tau (cis P-tau) has previously been shown to occur in animal models of traumatic brain injury (TBI), and blocking this form of tau using antibody was beneficial in a rodent model of severe TBI. Here the authors show that cis P-tau induction is a feature of several different forms of TBI in humans, and that administration of cis P-tau targeting antibody to rodents reduces or delays pathological features of TBI.
Collapse
Affiliation(s)
- Onder Albayram
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
| | - Asami Kondo
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
| | - Rebekah Mannix
- Division of Emergency Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Colin Smith
- Department of Neuropathology, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Cheng-Yu Tsai
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
| | - Chenyu Li
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
| | - Megan K Herbert
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
| | - Jianhua Qiu
- Division of Emergency Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Michael Monuteaux
- Division of Emergency Medicine, Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jane Driver
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA
- Geriatric Research Education and Clinical Center, VA Boston Healthcare System, Harvard Medical School, 150S Huntington Ave, Boston, MA, 02130, USA
| | - Sandra Yan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - William Gormley
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Ava M Puccio
- Department of Neurosurgery, University of Pittsburgh Medical Center, 200 Lothrop St, Pittsburgh, PA, 15213, USA
| | - David O Okonkwo
- Department of Neurosurgery, University of Pittsburgh Medical Center, 200 Lothrop St, Pittsburgh, PA, 15213, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, West Virginia University, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV, 26506, USA
| | - Julian Bailes
- Department of Neurosurgery, NorthShore University Health System, University of Chicago, Pritzker School of Medicine, 3rd Floor Kellogg, Evanston, IL, 60637, USA
| | - William Meehan
- Micheli Center for Sports Injury Prevention, Division of Sports Medicine, Children's Hospital Boston, Harvard Medical School, 319 Longwood Avenue, Boston, MA, 02115, USA
| | - Mark Zeidel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA.
| | - Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS 0408, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
Protective Role of Apocynin via Suppression of Neuronal Autophagy and TLR4/NF-κB Signaling Pathway in a Rat Model of Traumatic Brain Injury. Neurochem Res 2017; 42:3296-3309. [DOI: 10.1007/s11064-017-2372-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 06/14/2017] [Accepted: 08/01/2017] [Indexed: 10/19/2022]
|
10
|
Why New Spinal Cord Plasticity Does Not Disrupt Old Motor Behaviors. J Neurosci 2017; 37:8198-8206. [PMID: 28743726 DOI: 10.1523/jneurosci.0767-17.2017] [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: 03/21/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 12/28/2022] Open
Abstract
When new motor learning changes the spinal cord, old behaviors are not impaired; their key features are preserved by additional compensatory plasticity. To explore the mechanisms responsible for this compensatory plasticity, we transected the spinal dorsal ascending tract before or after female rats acquired a new behavior-operantly conditioned increase or decrease in the right soleus H-reflex-and examined an old behavior-locomotion. Neither spinal dorsal ascending tract transection nor H-reflex conditioning alone impaired locomotion. Nevertheless, when spinal dorsal ascending tract transection and H-reflex conditioning were combined, the rats developed a limp and a tilted posture that correlated in direction and magnitude with the H-reflex change. When the right H-reflex was increased by conditioning, the right step lasted longer than the left and the right hip was higher than the left; when the right H-reflex was decreased by conditioning, the opposite occurred. These results indicate that ascending sensory input guides the compensatory plasticity that normally prevents the plasticity underlying H-reflex change from impairing locomotion. They support the concept of the state of the spinal cord as a negotiated equilibrium that reflects the concurrent influences of all the behaviors in an individual's repertoire; and they support the new therapeutic strategies this concept introduces.SIGNIFICANCE STATEMENT The spinal cord provides a reliable final common pathway for motor behaviors throughout life. Until recently, its reliability was explained by the assumption that it is hardwired; but it is now clear that the spinal cord changes continually as new behaviors are acquired. Nevertheless, old behaviors are preserved. This study shows that their preservation depends on sensory feedback from the spinal cord to the brain: if feedback is removed, the acquisition of a new behavior may disrupt an old behavior. In sum, when a new behavior changes the spinal cord, sensory feedback to the brain guides further change that preserves old behaviors. This finding contributes to a new understanding of spinal cord function and to development of new rehabilitation therapies.
Collapse
|
11
|
Paterno R, Folweiler KA, Cohen AS. Pathophysiology and Treatment of Memory Dysfunction After Traumatic Brain Injury. Curr Neurol Neurosci Rep 2017; 17:52. [PMID: 28500417 PMCID: PMC5861722 DOI: 10.1007/s11910-017-0762-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Memory is fundamental to everyday life, and cognitive impairments resulting from traumatic brain injury (TBI) have devastating effects on TBI survivors. A contributing component to memory impairments caused by TBI is alteration in the neural circuits associated with memory function. In this review, we aim to bring together experimental findings that characterize behavioral memory deficits and the underlying pathophysiology of memory-involved circuits after TBI. While there is little doubt that TBI causes memory and cognitive dysfunction, it is difficult to conclude which memory phase, i.e., encoding, maintenance, or retrieval, is specifically altered by TBI. This is most likely due to variation in behavioral protocols and experimental models. Additionally, we review a selection of experimental treatments that hold translational potential to mitigate memory dysfunction following injury.
Collapse
Affiliation(s)
- Rosalia Paterno
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA.
| | - Kaitlin A Folweiler
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| | - Akiva S Cohen
- Department of Anesthesiology and Critical Care Medicine, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Abramson Research Center, Rm. 816-h, Philadelphia, PA, 19104, USA
| |
Collapse
|
12
|
Ezaki J, Shimada R, Shibuya M, Kibayashi K. Hippocampal neuronal degeneration in the traumatic brain injury mouse: non-trivial effect of scalp incision. Neurol Res 2016; 38:994-1002. [PMID: 27615406 DOI: 10.1080/01616412.2016.1228746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES In experimental models of traumatic brain injury (TBI), posttraumatic hippocampal neuronal degeneration in the cornu ammonis 1 (CA1), and/or the cornu ammonis 3 (CA3) regions are regarded as the most notable phenotypic appearances relating to the pathophysiology of human post-concussion syndrome. However, these morphological changes are often also seen in subjects without TBI, namely 'sham' groups. The frequencies and reasons of appearance of hippocampal neuronal degeneration in mice with TBI and/or sham are not clear. METHODS We compared the frequencies of hippocampal neuronal degeneration among three groups: TBI (mice with external force impact performed by Marmarou's weight drop model after scalp incision), sham (mice with scalp incision alone), and control (mice with neither external force impact nor scalp incision), using hematoxylin and eosin stain in day 6 (n = 5 in each group.) Isoflurane was used for anesthesia in all mice. RESULTS The frequencies were 80, 100, and 20% in CA1, and 20, 40, and 60% in CA3, for TBI, sham, and control, respectively. In CA1, a significant difference of the frequency was observed between sham and control (p = 0.048), but not, between TBI and sham (p = 1.000) in Fisher's exact test. In CA3, no significant difference in the frequency was observed between the three groups. CONCLUSION Scalp incision, rather than external impact force, might affect the CA1 hippocampal neuronal degeneration in mice with TBI. In addition, factor(s) other than external impact force or scalp incision may also cause hippocampal neuronal degeneration in both CA1 and CA3. Careful interpretation is needed concerning hippocampal neuronal degeneration induced by a weight drop device observed in mice with TBI.
Collapse
Affiliation(s)
- Jiro Ezaki
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Ryo Shimada
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Misato Shibuya
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| | - Kazuhiko Kibayashi
- a Department of Legal Medicine , School of Medicine, Tokyo Women's Medical University , Tokyo , Japan
| |
Collapse
|
13
|
Israel I, Ohsiek A, Al-Momani E, Albert-Weissenberger C, Stetter C, Mencl S, Buck AK, Kleinschnitz C, Samnick S, Sirén AL. Combined [(18)F]DPA-714 micro-positron emission tomography and autoradiography imaging of microglia activation after closed head injury in mice. J Neuroinflammation 2016; 13:140. [PMID: 27266706 PMCID: PMC4897946 DOI: 10.1186/s12974-016-0604-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 05/30/2016] [Indexed: 11/27/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of death and disability. Neuroinflammation contributes to acute damage after TBI and modulates long-term evolution of degenerative and regenerative responses to injury. The aim of the present study was to evaluate the relationship of microglia activation to trauma severity, brain energy metabolism, and cellular reactions to injury in a mouse closed head injury model using combined in vivo PET imaging, ex vivo autoradiography, and immunohistochemistry. Methods A weight-drop closed head injury model was used to produce a mixed diffuse and focal TBI or a purely diffuse mild TBI (mTBI) in C57BL6 mice. Lesion severity was determined by evaluating histological damage and functional outcome using a standardized neuroscore (NSS), gliosis, and axonal injury by immunohistochemistry. Repeated intra-individual in vivo μPET imaging with the specific 18-kDa translocator protein (TSPO) radioligand [18F]DPA-714 was performed on day 1, 7, and 16 and [18F]FDG-μPET imaging for energy metabolism on days 2–5 after trauma using freshly synthesized radiotracers. Immediately after [18F]DPA-714-μPET imaging on days 7 and 16, cellular identity of the [18F]DPA-714 uptake was confirmed by exposing freshly cut cryosections to film autoradiography and successive immunostaining with antibodies against the microglia/macrophage marker IBA-1. Results Functional outcome correlated with focal brain lesions, gliosis, and axonal injury. [18F]DPA-714-μPET showed increased radiotracer uptake in focal brain lesions on days 7 and 16 after TBI and correlated with reduced cerebral [18F]FDG uptake on days 2–5, with functional outcome and number of IBA-1 positive cells on day 7. In autoradiography, [18F]DPA-714 uptake co-localized with areas of IBA1-positive staining and correlated strongly with both NSS and the number of IBA1-positive cells, gliosis, and axonal injury. After mTBI, numbers of IBA-1 positive cells with microglial morphology increased in both brain hemispheres; however, uptake of [18F]DPA-714 was not increased in autoradiography or in μPET imaging. Conclusions [18F]DPA-714 uptake in μPET/autoradiography correlates with trauma severity, brain metabolic deficits, and microglia activation after closed head TBI.
Collapse
Affiliation(s)
- Ina Israel
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Andrea Ohsiek
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Ehab Al-Momani
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christiane Albert-Weissenberger
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany.,Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christian Stetter
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Stine Mencl
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, 97080, Würzburg, Germany.,Department of Neurology, University Hospital Essen, 45147, Essen, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Anna-Leena Sirén
- Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
14
|
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability for people under 45 years of age. Clinical TBI is often the result of disparate forces resulting in heterogeneous injuries. Preclinical modeling of TBI is a vital tool for studying the complex cascade of metabolic, cellular, and molecular post-TBI events collectively termed secondary injury. Preclinical models also provide an important platform for studying therapeutic interventions. However, modeling TBI in the preclinical setting is challenging, and most models replicate only certain aspects of clinical TBI. This chapter details the most widely used models of preclinical TBI, including the controlled cortical impact, fluid percussion, blast, and closed head models. Each of these models replicates particular critical aspects of clinical TBI. Prior to selecting a preclinical TBI model, it is important to address what aspect of human TBI is being sought to evaluate.
Collapse
|
15
|
Gupta RK, Prasad S. Age-Dependent Alterations in the Interactions of NF-κB and N-myc with GLT-1/EAAT2 Promoter in the Pericontusional Cortex of Mice Subjected to Traumatic Brain Injury. Mol Neurobiol 2015; 53:3377-3388. [PMID: 26081154 DOI: 10.1007/s12035-015-9287-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is one of the major risk factors of dementia, aging, and cognitive impairments, etc. We have previously reported that expression of the astrocytic glutamate transporter GLT-1/EAAT2 is downregulated in the pericontusional cortex of adult and old mice in post-TBI time-dependent manner, and the process of decline starts before in old than in adult TBI mice. However, relationship between age- and TBI-dependent alterations in GLT-1/EAAT2 expression and interactions of transcription factors NF-κB and N-myc with their cognate GLT-1/EAAT2 promoter sequences, an important step of its transcriptional control, is not known. To understand this, we developed TBI mouse model by modified chronic head injury (CHI) method, analyzed expression of GFAP, TNF-α, and AQP4 by RT-PCR for its validation, and analyzed interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences by electrophoretic mobility shift assay (EMSA). Our EMSA data revealed that interactions of NF-κB and N-myc with GLT-1/EAAT2 promoter sequences was significantly elevated in the ipsi-lateral cortex of both adult and old TBI mice in post-TBI time-dependent manner; however, these interactions started immediately in the old compared to that in adult TBI mice, which could be attributed to our previously reported age- and post-TBI time-dependent differential expression of GLT-1/EAAT2 in the pericontusional cortex.
Collapse
Affiliation(s)
- Rajaneesh K Gupta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, UP, India
| | - S Prasad
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, UP, India.
| |
Collapse
|
16
|
Xenon improves neurologic outcome and reduces secondary injury following trauma in an in vivo model of traumatic brain injury. Crit Care Med 2015; 43:149-158. [PMID: 25188549 DOI: 10.1097/ccm.0000000000000624] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES To determine the neuroprotective efficacy of the inert gas xenon following traumatic brain injury and to determine whether application of xenon has a clinically relevant therapeutic time window. DESIGN Controlled animal study. SETTING University research laboratory. SUBJECTS Male C57BL/6N mice (n = 196). INTERVENTIONS Seventy-five percent xenon, 50% xenon, or 30% xenon, with 25% oxygen (balance nitrogen) treatment following mechanical brain lesion by controlled cortical impact. MEASUREMENTS AND MAIN RESULTS Outcome following trauma was measured using 1) functional neurologic outcome score, 2) histological measurement of contusion volume, and 3) analysis of locomotor function and gait. Our study shows that xenon treatment improves outcome following traumatic brain injury. Neurologic outcome scores were significantly (p < 0.05) better in xenon-treated groups in the early phase (24 hr) and up to 4 days after injury. Contusion volume was significantly (p < 0.05) reduced in the xenon-treated groups. Xenon treatment significantly (p < 0.05) reduced contusion volume when xenon was given 15 minutes after injury or when treatment was delayed 1 or 3 hours after injury. Neurologic outcome was significantly (p < 0.05) improved when xenon treatment was given 15 minutes or 1 hour after injury. Improvements in locomotor function (p < 0.05) were observed in the xenon-treated group, 1 month after trauma. CONCLUSIONS These results show for the first time that xenon improves neurologic outcome and reduces contusion volume following traumatic brain injury in mice. In this model, xenon application has a therapeutic time window of up to at least 3 hours. These findings support the idea that xenon may be of benefit as a neuroprotective treatment in patients with brain trauma.
Collapse
|
17
|
Chen Y, Chen L, Wang Y, Wolpaw JR, Chen XY. Persistent beneficial impact of H-reflex conditioning in spinal cord-injured rats. J Neurophysiol 2014; 112:2374-81. [PMID: 25143542 DOI: 10.1152/jn.00422.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Operant conditioning of a spinal cord reflex can improve locomotion in rats and humans with incomplete spinal cord injury. This study examined the persistence of its beneficial effects. In rats in which a right lateral column contusion injury had produced asymmetric locomotion, up-conditioning of the right soleus H-reflex eliminated the asymmetry while down-conditioning had no effect. After the 50-day conditioning period ended, the H-reflex was monitored for 100 [±9 (SD)] (range 79-108) more days and locomotion was then reevaluated. After conditioning ended in up-conditioned rats, the H-reflex continued to increase, and locomotion continued to improve. In down-conditioned rats, the H-reflex decrease gradually disappeared after conditioning ended, and locomotion at the end of data collection remained as impaired as it had been before and immediately after down-conditioning. The persistence (and further progression) of H-reflex increase but not H-reflex decrease in these spinal cord-injured rats is consistent with the fact that up-conditioning improved their locomotion while down-conditioning did not. That is, even after up-conditioning ended, the up-conditioned H-reflex pathway remained adaptive because it improved locomotion. The persistence and further enhancement of the locomotor improvement indicates that spinal reflex conditioning protocols might supplement current therapies and enhance neurorehabilitation. They may be especially useful when significant spinal cord regeneration becomes possible and precise methods for retraining the regenerated spinal cord are needed.
Collapse
Affiliation(s)
- Yi Chen
- Wadsworth Center, New York State Department of Health, Albany, New York
| | - Lu Chen
- Wadsworth Center, New York State Department of Health, Albany, New York
| | - Yu Wang
- Wadsworth Center, New York State Department of Health, Albany, New York
| | - Jonathan R Wolpaw
- Wadsworth Center, New York State Department of Health, Albany, New York; Department of Biomedical Sciences, State University of New York, Albany, New York; and Department of Neurology, Neurological Institute, Columbia University, New York, New York
| | - Xiang Yang Chen
- Wadsworth Center, New York State Department of Health, Albany, New York; Department of Biomedical Sciences, State University of New York, Albany, New York; and
| |
Collapse
|
18
|
Umschweif G, Liraz-Zaltsman S, Shabashov D, Alexandrovich A, Trembovler V, Horowitz M, Shohami E. Angiotensin receptor type 2 activation induces neuroprotection and neurogenesis after traumatic brain injury. Neurotherapeutics 2014; 11:665-78. [PMID: 24957202 PMCID: PMC4121449 DOI: 10.1007/s13311-014-0286-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Angiotensin II receptor type 2 (AT(2)) agonists have been shown to limit brain ischemic insult and to improve its outcome. The activation of AT(2) was also linked to induced neuronal proliferation and differentiation in vitro. In this study, we examined the therapeutic potential of AT(2) activation following traumatic brain injury (TBI) in mice, a brain pathology that displays ischemia-like secondary damages. The AT(2) agonist CGP42112A was continuously infused immediately after closed head injury (CHI) for 3 days. We have followed the functional recovery of the injured mice for 35 days post-CHI, and evaluated cognitive function, lesion volume, molecular signaling, and neurogenesis at different time points after the impact. We found dose-dependent improvement in functional recovery and cognitive performance after CGP42112A treatment that was accompanied by reduced lesion volume and induced neurogenesis in the neurogenic niches of the brain and also in the injury region. At the cellular/molecular level, CGP42112A induced early activation of neuroprotective kinases protein kinase B (Akt) and extracellular-regulated kinases ½ (ERK½), and the neurotrophins nerve growth factor and brain-derived neurotrophic factor; all were blocked by treatment with the AT(2) antagonist PD123319. Our results suggest that AT(2) activation after TBI promotes neuroprotection and neurogenesis, and may be a novel approach for the development of new drugs to treat victims of TBI.
Collapse
Affiliation(s)
- Gali Umschweif
- />Department of Pharmacology, The Hebrew University, Jerusalem, Israel
- />Laboratory of Environmental Physiology, The Hebrew University, Jerusalem, Israel
| | | | - Dalia Shabashov
- />Department of Pharmacology, The Hebrew University, Jerusalem, Israel
| | | | | | - Michal Horowitz
- />Laboratory of Environmental Physiology, The Hebrew University, Jerusalem, Israel
| | - Esther Shohami
- />Department of Pharmacology, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
19
|
Harrison JL, Rowe RK, O’Hara BF, Adelson PD, Lifshitz J. Acute over-the-counter pharmacological intervention does not adversely affect behavioral outcome following diffuse traumatic brain injury in the mouse. Exp Brain Res 2014; 232:2709-19. [DOI: 10.1007/s00221-014-3948-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/03/2014] [Indexed: 01/28/2023]
|
20
|
Kumar A, Jaggi AS, Singh N. Pharmacological investigations on possible role of Src kinases in neuroprotective mechanism of ischemic postconditioning in mice. Int J Neurosci 2014; 124:777-86. [DOI: 10.3109/00207454.2013.879869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
21
|
Role of P2X7 purinoceptors in neuroprotective mechanism of ischemic postconditioning in mice. Mol Cell Biochem 2014; 390:161-73. [DOI: 10.1007/s11010-014-1967-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 01/21/2014] [Indexed: 01/23/2023]
|
22
|
Chen Y, Chen L, Liu R, Wang Y, Chen XY, Wolpaw JR. Locomotor impact of beneficial or nonbeneficial H-reflex conditioning after spinal cord injury. J Neurophysiol 2013; 111:1249-58. [PMID: 24371288 DOI: 10.1152/jn.00756.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
When new motor learning changes neurons and synapses in the spinal cord, it may affect previously learned behaviors that depend on the same spinal neurons and synapses. To explore these effects, we used operant conditioning to strengthen or weaken the right soleus H-reflex pathway in rats in which a right spinal cord contusion had impaired locomotion. When up-conditioning increased the H-reflex, locomotion improved. Steps became longer, and step-cycle asymmetry (i.e., limping) disappeared. In contrast, when down-conditioning decreased the H-reflex, locomotion did not worsen. Steps did not become shorter, and asymmetry did not increase. Electromyographic and kinematic analyses explained how H-reflex increase improved locomotion and why H-reflex decrease did not further impair it. Although the impact of up-conditioning or down-conditioning on the H-reflex pathway was still present during locomotion, only up-conditioning affected the soleus locomotor burst. Additionally, compensatory plasticity apparently prevented the weaker H-reflex pathway caused by down-conditioning from weakening the locomotor burst and further impairing locomotion. The results support the hypothesis that the state of the spinal cord is a "negotiated equilibrium" that serves all the behaviors that depend on it. When new learning changes the spinal cord, old behaviors undergo concurrent relearning that preserves or improves their key features. Thus, if an old behavior has been impaired by trauma or disease, spinal reflex conditioning, by changing a specific pathway and triggering a new negotiation, may enable recovery beyond that achieved simply by practicing the old behavior. Spinal reflex conditioning protocols might complement other neurorehabilitation methods and enhance recovery.
Collapse
Affiliation(s)
- Yi Chen
- Wadsworth Center, New York State Department of Health, Albany, New York
| | | | | | | | | | | |
Collapse
|
23
|
Operant conditioning of a spinal reflex can improve locomotion after spinal cord injury in humans. J Neurosci 2013; 33:2365-75. [PMID: 23392666 DOI: 10.1523/jneurosci.3968-12.2013] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Operant conditioning protocols can modify the activity of specific spinal cord pathways and can thereby affect behaviors that use these pathways. To explore the therapeutic application of these protocols, we studied the impact of down-conditioning the soleus H-reflex in people with impaired locomotion caused by chronic incomplete spinal cord injury. After a baseline period in which soleus H-reflex size was measured and locomotion was assessed, subjects completed either 30 H-reflex down-conditioning sessions (DC subjects) or 30 sessions in which the H-reflex was simply measured [unconditioned (UC) subjects], and locomotion was reassessed. Over the 30 sessions, the soleus H-reflex decreased in two-thirds of the DC subjects (a success rate similar to that in normal subjects) and remained smaller several months later. In these subjects, locomotion became faster and more symmetrical, and the modulation of EMG activity across the step cycle increased bilaterally. Furthermore, beginning about halfway through the conditioning sessions, all of these subjects commented spontaneously that they were walking faster and farther in their daily lives, and several noted less clonus, easier stepping, and/or other improvements. The H-reflex did not decrease in the other DC subjects or in any of the UC subjects; and their locomotion did not improve. These results suggest that reflex-conditioning protocols can enhance recovery of function after incomplete spinal cord injuries and possibly in other disorders as well. Because they are able to target specific spinal pathways, these protocols could be designed to address each individual's particular deficits, and might thereby complement other rehabilitation methods.
Collapse
|
24
|
HLA-DR3-DQ2 Mice Do Not Develop Ataxia in the Presence of High Titre Anti-gliadin Antibodies. THE CEREBELLUM 2012; 12:370-6. [PMID: 23086706 DOI: 10.1007/s12311-012-0425-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Lapchak PA. Transcranial near-infrared laser therapy applied to promote clinical recovery in acute and chronic neurodegenerative diseases. Expert Rev Med Devices 2012; 9:71-83. [PMID: 22145842 DOI: 10.1586/erd.11.64] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One of the most promising methods to treat neurodegeneration is noninvasive transcranial near-infrared laser therapy (NILT), which appears to promote acute neuroprotection by stimulating mitochondrial function, thereby increasing cellular energy production. NILT may also promote chronic neuronal function restoration via trophic factor-mediated plasticity changes or possibly neurogenesis. Clearly, NILT is a treatment that confers neuroprotection or neurorestoration using pleiotropic mechanisms. The most advanced application of NILT is for acute ischemic stroke based upon extensive preclinical and clinical studies. In laboratory settings, NILT is also being developed to treat traumatic brain injury, Alzheimer's disease and Parkinson's disease. There is some intriguing data in the literature that suggests that NILT may be a method to promote clinical improvement in neurodegenerative diseases where there is a common mechanistic component, mitochondrial dysfunction and energy impairment. This article will analyze and review data supporting the continued development of NILT to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul A Lapchak
- Cedars-Sinai Medical Center, Department of Neurology, Los Angeles, CA 90048, USA.
| |
Collapse
|
26
|
Albert-Weißenberger C, Várrallyay C, Raslan F, Kleinschnitz C, Sirén AL. An experimental protocol for mimicking pathomechanisms of traumatic brain injury in mice. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:1. [PMID: 22300472 PMCID: PMC3305492 DOI: 10.1186/2040-7378-4-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/02/2012] [Indexed: 01/30/2023]
Abstract
Traumatic brain injury (TBI) is a result of an outside force causing immediate mechanical disruption of brain tissue and delayed pathogenic events. In order to examine injury processes associated with TBI, a number of rodent models to induce brain trauma have been described. However, none of these models covers the entire spectrum of events that might occur in TBI. Here we provide a thorough methodological description of a straightforward closed head weight drop mouse model to assess brain injuries close to the clinical conditions of human TBI.
Collapse
|
27
|
Xu S, Zhuo J, Racz J, Shi D, Roys S, Fiskum G, Gullapalli R. Early microstructural and metabolic changes following controlled cortical impact injury in rat: a magnetic resonance imaging and spectroscopy study. J Neurotrauma 2011; 28:2091-102. [PMID: 21761962 DOI: 10.1089/neu.2010.1739] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Understanding tissue alterations at an early stage following traumatic brain injury (TBI) is critical for injury management and limiting severe consequences from secondary injury. We investigated the early microstructural and metabolic profiles using in vivo diffusion tensor imaging (DTI) and proton magnetic resonance spectroscopy ((1)H MRS) at 2 and 4 h following a controlled cortical impact injury in the rat brain using a 7.0 Tesla animal MRI system and compared profiles to baseline. Significant decrease in mean diffusivity (MD) and increased fractional anisotropy (FA) was found near the impact site (hippocampus and bilateral thalamus; p<0.05) immediately following TBI, suggesting cytotoxic edema. Although the DTI parameters largely normalized on the contralateral side by 4 h, a large inter-individual variation was observed with a trend towards recovery of MD and FA in the ipsilateral hippocampus and a sustained elevation of FA in the ipsilateral thalamus (p<0.05). Significant reduction in metabolite to total creatine ratios of N-acetylaspartate (NAA, p=0.0002), glutamate (p=0.0006), myo-inositol (Ins, p=0.04), phosphocholine and glycerophosphocholine (PCh+GPC, p=0.03), and taurine (Tau, p=0.009) were observed ipsilateral to the injury as early as 2 h, while glutamine concentration increased marginally (p=0.07). These metabolic alterations remained sustained over 4 h after TBI. Significant reductions of Ins (p=0.024) and Tau (p=0.013) and marginal reduction of NAA (p=0.06) were also observed on the contralateral side at 4 h after TBI. Overall our findings suggest significant microstructural and metabolic alterations as early as 2 h following injury. The tendency towards normalization at 4 h from the DTI data and no further metabolic changes at 4 h from MRS suggest an optimal temporal window of about 3 h for interventions that might limit secondary damage to the brain. Results indicate that early assessment of TBI patients using DTI and MRS may provide valuable information on the available treatment window to limit secondary brain damage.
Collapse
Affiliation(s)
- Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
PRIMARY OBJECTIVE A volumetric blood surge (rapid physical movement/displacement of blood) is hypothesized to cause the non-impact, mild TBI and battlefield PTSD induced by a blast over-pressure wave. RESEARCH DESIGN Systematic review of the literature. METHODS AND PROCEDURES Articles relating to the fields of blast injury, brain injury and relevant disorders were searched between the years 1968-2010 for keywords such as 'brain injury', 'post-traumatic stress disorder' and 'blast pressure wave'. Articles found through journal and Internet databases were cross-referenced. MAIN OUTCOMES AND RESULTS The blood surge, which is driven by elevated overall pressure in the ventral body cavity after exposure of the torso to blast wave, may move through blood vessels to the low-pressure cranial cavity from the high-pressure ventral body cavity. It dramatically increases cerebral perfusion pressure and causes damage to both tiny cerebral blood vessels and the BBB. CONCLUSIONS Three factors may be critical to the induction of blast-induced brain injuries: (1) the difference in pressure between the ventral body cavity and cranial cavity; (2) blood that acts as a transmission medium to propagate a pressure wave to the brain; and (3) the vulnerability of cerebral blood vessels and the BBB to a sudden fluctuation in perfusion pressure.
Collapse
Affiliation(s)
- Yun Chen
- Tripler Army Medical Center, Honolulu, HI, USA.
| | | |
Collapse
|
29
|
Thompson AK, Lapallo B, Duffield M, Abel BM, Pomerantz F. Repetitive common peroneal nerve stimulation increases ankle dorsiflexor motor evoked potentials in incomplete spinal cord lesions. Exp Brain Res 2011; 210:143-52. [PMID: 21360230 DOI: 10.1007/s00221-011-2607-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 02/16/2011] [Indexed: 11/29/2022]
Abstract
Plasticity of corticospinal tract (CST) activity likely plays a key role in motor function recovery after central nervous system (CNS) lesions. In non-injured adults, 30 min of repetitive common peroneal nerve stimulation (rCPnS) increases CST excitability by 40-50% and the effect persists for at least 30 min. The present study evaluated with transcranial magnetic stimulation (TMS) the changes in CST excitability after 30 min of rCPnS in people with foot drop due to incomplete SCI. Suprathreshold rCPnS (25 Hz, alternating 1 s on 1 s off stimulation cycle) was given for two 15-min periods, while the subject sat at rest with ankle and knee joints fixed. Before, between, and after the periods of stimulation, the tibialis anterior (TA) motor evoked potentials (MEPs) to TMS were measured at a TMS intensity that originally produced a half-maximum MEP (typically 10-20% above threshold) while the sitting subject provided 25-30% maximum voluntary TA contraction. In 10 subjects with SCI, the peak-to-peak TA MEP increased by 14 ± 3% after rCPnS and the peak increase (+21 ± 7%) occurred 15 min after the cessation of rCPnS. The TA H-reflex, measured in separate experiments in 7 subjects, did not increase after rCPnS. The results indicate that rCPnS can increase CST excitability for the TA in people with incomplete SCI, although its effects appear smaller and shorter lasting than those found in non-injured control subjects. Such short-term plasticity in the CST excitability induced by rCPnS may contribute to long-term therapeutic effects of functional electrical stimulation previously reported in people with CNS lesions.
Collapse
Affiliation(s)
- Aiko K Thompson
- Translational Neuroscience Research Program, Helen Hayes Hospital, New York State Department of Health, Route 9W, West Haverstraw, NY 10993, USA.
| | | | | | | | | |
Collapse
|
30
|
Albert-Weissenberger C, Sirén AL. Experimental traumatic brain injury. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:16. [PMID: 20707892 PMCID: PMC2930598 DOI: 10.1186/2040-7378-2-16] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 08/13/2010] [Indexed: 12/03/2022]
Abstract
Traumatic brain injury, a leading cause of death and disability, is a result of an outside force causing mechanical disruption of brain tissue and delayed pathogenic events which collectively exacerbate the injury. These pathogenic injury processes are poorly understood and accordingly no effective neuroprotective treatment is available so far. Experimental models are essential for further clarification of the highly complex pathology of traumatic brain injury towards the development of novel treatments. Among the rodent models of traumatic brain injury the most commonly used are the weight-drop, the fluid percussion, and the cortical contusion injury models. As the entire spectrum of events that might occur in traumatic brain injury cannot be covered by one single rodent model, the design and choice of a specific model represents a major challenge for neuroscientists. This review summarizes and evaluates the strengths and weaknesses of the currently available rodent models for traumatic brain injury.
Collapse
|
31
|
Fanne RA, Nassar T, Mazuz A, Waked O, Heyman SN, Hijazi N, Goelman G, Higazi AAR. Neuroprotection by glucagon: role of gluconeogenesis. J Neurosurg 2010; 114:85-91. [PMID: 20509730 DOI: 10.3171/2010.4.jns10263] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The severity of neurological impairment following traumatic brain injury (TBI) is exacerbated by several endogenous processes, including hyperglycemia, hypotension, and the generation of glutamate. However, in addition to controlling hyperglycemia, insulin has pleiotropic effects on tissue metabolism, which include reducing the concentration of the neurotoxic amino acid glutamate, making it unclear whether insulin's beneficial effects are attributable to the establishment of euglycemia per se. In the present study, the authors asked if reducing glutamate via approaches that do not lower glucose levels would improve neurological outcome following TBI. METHODS Glucagon activates gluconeogenesis by increasing the hepatic uptake of amino acids such as glutamate and facilitating their conversion to glucose. Glucagon was administered as a single intraperitoneal injection before or after closed head injury (CHI). Neurological function, brain histological features, blood glutamate and glucose levels, and CSF glutamate concentrations were measured. RESULTS A single intraperitoneal injection of glucagon (25 μg) into mice 10 minutes before or after CHI reduced lesion size by about 60% (p < 0.0001) and accelerated neurological recovery. The neuroprotective effect of glucagon was related to gluconeogenesis by decreasing the concentration of the neuroexcitatory amino acid glutamate in the circulation from 207 ± 32.1 μmol/L in untreated mice to 101.11 ± 21.6 μmol/L in treated mice (p < 0.001); a similar effect occurred in the CSF. The neuroprotective effect of glucagon was seen notwithstanding the attendant increase in blood glucose, the final substrate of gluconeogenesis. CONCLUSIONS Glucagon exerts a marked neuroprotective effect post-TBI by decreasing CNS glutamate. Glucagon was beneficial despite increasing blood glucose. Favorable effects also occurred when glucagon was given prior to TBI, suggesting its involvement in the preconditioning process. Thus, glucagon may be of value in providing neuroprotection when administered after TBI or prior to certain neurosurgical or cardiac interventions in which the incidence of perioperative ischemia is high.
Collapse
Affiliation(s)
- Rami Abu Fanne
- Department of Clinical Biochemistry, MRI/MRS Lab of the Human Biology Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Szczygielski J, Mautes AE, Schwerdtfeger K, Steudel WI. The effects of selective brain hypothermia and decompressive craniectomy on brain edema after closed head injury in mice. ACTA NEUROCHIRURGICA. SUPPLEMENT 2010; 106:225-229. [PMID: 19812954 DOI: 10.1007/978-3-211-98811-4_42] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Intractable brain edema remains one of the main causes of death after traumatic brain injury (TBI). Brain hypothermia and decompressive craniectomy have been considered as potential therapies. The goal of our experimental study was to determine if selective hypothermia in combination with craniectomy could modify the development of posttraumatic brain edema. Male CD-1 mice were anesthetized with halothane and randomly assigned into the following groups: sham-operated (n = 5), closed head injury (CHI) alone (n = 5), CHI followed by craniectomy at 1 h post-TBI (n = 5) and CHI + craniectomy and selective hypothermia (focal brain cooling using cryosurgery device) maintained for 5 h (n = 5). Animals were sacrificed at 7 h posttrauma and brains were removed, sagittally dissected and dried. The brain water content of separate hemispheres was calculated from the weight difference before and after drying. In the CHI alone group there was no significant increase in brain water content in both the ipsi- and contralateral hemispheres (80.59 +/- 1% and 78.74 +/- 0.9% in the CHI group vs. 79.31 +/- 0.7% and 79.01 +/- 0.3% in the sham group, respectively). Brain edema was significantly increased ipsilaterally in the trauma + craniectomy group (82.11 +/- 0.6%, p < 0.05), but not in the trauma + craniectomy + hypothermia group (81.52 +/- 1.1%, p > 0.05) as compared to the sham group (79.31 +/- 0.7%). These data suggest that decompressive craniectomy leads to an increase in brain water content after CHI. Additional focal hypothermia may be an effective approach in the treatment of posttraumatic brain edema.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Hospital, Kirrberger Strasse, Homburg, Saar, Germany.
| | | | | | | |
Collapse
|
33
|
Degos V, Lescot T, Puybasset L. Quantitative CT Scan and CT-Estimated Brain Specific Gravity in TBI. Intensive Care Med 2010. [DOI: 10.1007/978-1-4419-5562-3_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Zhao TZ, Xia YZ, Li L, Li J, Zhu G, Chen S, Feng H, Lin JK. Bovine serum albumin promotes IL-1beta and TNF-alpha secretion by N9 microglial cells. Neurol Sci 2009; 30:379-83. [PMID: 19696964 DOI: 10.1007/s10072-009-0123-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 07/29/2009] [Indexed: 12/30/2022]
Abstract
Bovine serum albumin (BSA) is generally used in biomedical experiments. In the solution of some reagents, BSA is necessary to maintain the stability and concentration of the effective component. Therefore, the potential impact of BSA on experimental results should not be neglected when BSA is used. In this study, we observed that BSA induced significant upregulation of mRNA expression and release of pro-inflammatory cytokines, IL-1beta, and TNF-alpha, by N9 microglial cells. Our results suggest that the effects of BSA should be taken into account in experiments on microglia or the central nervous system when BSA is used. In light of the high similarity and homology among mammalian albumins, our findings also indicate that serum albumin may be a potent trigger of cytokine release by microglia.
Collapse
Affiliation(s)
- Tian-zhi Zhao
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing, 400038, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Crack PJ, Gould J, Bye N, Ross S, Ali U, Habgood MD, Morganti-Kossman C, Saunders NR, Hertzog PJ. The genomic profile of the cerebral cortex after closed head injury in mice: effects of minocycline. J Neural Transm (Vienna) 2008; 116:1-12. [PMID: 19018450 DOI: 10.1007/s00702-008-0145-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Accepted: 10/20/2008] [Indexed: 12/15/2022]
Abstract
Microarray analysis was used to delineate gene expression patterns and profile changes following traumatic brain injury (TBI) in mice. A parallel microarray analysis was carried out in mice with TBI that were subsequently treated with minocycline, a drug proposed as a neuroprotectant in other neurological disorders. The aim of this comparison was to identify pathways that may be involved in secondary injury processes following TBI and potential specific pathways that could be targeted with second generation therapeutics for the treatment of neurotrauma patients. Gene expression profiles were measured with the compugen long oligo chip and real-time PCR was used to validate microarray findings. A pilot study of effect of minocycline on gene expression following TBI was also carried out. Gene ontology comparison analysis of sham TBI and minocycline treated brains revealed biological pathways with more genes differentially expressed than predicted by chance. Among 495 gene ontology categories, the significantly different gene ontology groups included chemokines, genes involved in cell surface receptor-linked signal transduction and pro-inflammatory cytokines. Expression levels of some key genes were validated by real-time quantitative PCR. This study confirms that multiple regulatory pathways are affected following brain injury and demonstrates for the first time that specific genes and molecular networks are affected by minocycline following brain injury.
Collapse
Affiliation(s)
- Peter J Crack
- Department of Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Degos V, Pereira AR, Lescot T, Sanchez-Peña P, Daoudi M, Zouaoui A, Coriat P, Puybasset L. Does brain swelling increase estimated specific gravity? Neurocrit Care 2008; 9:338-43. [PMID: 18818888 DOI: 10.1007/s12028-008-9131-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 07/21/2008] [Indexed: 11/25/2022]
Abstract
OBJECTIVE At the acute phase of traumatic brain injury (TBI), brain swelling contributes substantially to the development of secondary neurological lesions. Elucidating the pathophysiology of brain swelling is crucial to improve TBI management. In a previous study, specific gravity (SG) of the noncontused hemisphere, as estimated by computed tomography (CT), was higher in patients with high Marshall CT scores and severe brain swelling. The aim of this study was to investigate the relationship between estimated specific gravity (eSG) and clinical variable suggestive of brain swelling. DESIGN Retrospective study of data from a prospectively established database. SETTING Neurology ICU in a teaching hospital in Paris, France. PARTICIPANTS We studied 20 patients with severe traumatic brain injury (TBI), 20 patients with high-grade subarachnoid hemorrhage (SAH) presenting similar brain-swelling criteria, 20 patients with low-grade SAH, and 20 healthy controls. INTERVENTIONS None. MEASUREMENTS AND RESULTS Estimated brain specific gravity was acquired from CT images obtained at ICU admission. eSG was estimated in the overall intracerebral content and in a region-of-interest composed of white matter and the diencephalon. eSG in the region of interest was significantly higher in the TBI patients than in the high-grade SAH patients (1.0350 +/- 0.0041 vs. 1.0310 +/- 0.0019 g/ml, P < 0.05). eSG was similar in the high-grade SAH, low-grade SAH, and control groups. CONCLUSIONS Our findings do not support a causal link between brain swelling and eSG elevation. The eSG increase in severe TBI patients is not due to brain swelling.
Collapse
Affiliation(s)
- Vincent Degos
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Teaching Hospital, Assistance Publique-Hôpitaux de Paris, Pierre and Marie Curie Paris 6 University, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Shein NA, Tsenter J, Alexandrovich AG, Horowitz M, Shohami E. Akt phosphorylation is required for heat acclimation-induced neuroprotection. J Neurochem 2007; 103:1523-9. [PMID: 17725578 DOI: 10.1111/j.1471-4159.2007.04862.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Long-term heat exposure, known as heat acclimation (HA; 30 days at 34 +/- 1 degrees C) is neuroprotective against traumatic brain injury. Acclimated mice were previously found to display improved functional recovery as well as an increase in the levels of the specific erythropoietin receptor. As the activation of this receptor is known to facilitate functional recovery on one hand and the phosphorylation and activation of Akt, an intracellular kinase which regulates anti-apoptotic pathways on the other, in this study we investigated whether HA affects Akt phosphorylation prior to and following injury and whether this step is required for development of HA-induced neuroprotection. Akt phosphorylation was blocked using Triciribine (TCN), a compound shown to block the phosphorylation process without affecting upstream effectors of this kinase, and several post-injury functional end-point measures were subsequently evaluated. Acclimation led to a post-injury increase in the levels of phosphorylated Akt, resulting in higher levels when compared with normothermic controls at 4 h post-injury (63.6 +/- 5.2% and 42.7 +/- 3.7%, respectively, p </= 0.05). This increase was diminished following TCN administration. Post-injury TCN treatment abolished the HA-induced functional benefits, including effects on motor and cognitive functions as well as the attenuation of edema formation. We therefore suggest that Akt phosphorylation is essential for HA-induced neuroprotection after traumatic brain injury.
Collapse
Affiliation(s)
- Na'ama A Shein
- Department of Pharmacology, The Hebrew University, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
38
|
Chen XY, Chen Y, Chen L, Tennissen AM, Wolpaw JR. Corticospinal tract transection permanently abolishes H-reflex down-conditioning in rats. J Neurotrauma 2007; 23:1705-12. [PMID: 17115915 DOI: 10.1089/neu.2006.23.1705] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that corticospinal tract (CST) transection, but not transection of other major spinal cord tracts, prevents down-conditioning of the H-reflex, the electrical analog of the spinal stretch reflex. This study set out to determine whether the loss of the capacity for H-reflex down-conditioning caused by CST transection is permanent. Female Sprague-Dawley rats received CST, lateral column (LC), or dorsal column ascending tract (DA) transection at T8-9; 9-10 months later, they were exposed to the H-reflex down-conditioning protocol for 50 days. In the LC and DA rats, H-reflex size fell to 60 (+/- 9 SEM)% and 60 (+/- 19)%, respectively, of its initial size. This down-conditioning was comparable to that of normal rats. In contrast, H-reflex size in the CST rats rose to 170 (+/- 42)% of its initial size. A similar rise does not occur in rats exposed to down-conditioning shortly after CST transection. These results indicate that CST transection permanently eliminates the capacity for H-reflex down-conditioning and has gradual long-term effects on sensorimotor cortex function. They imply that H-reflex down-conditioning can be a reliable measure of CST function for long-term studies of the effects of spinal cord injury and/or for evaluations of the efficacy of experimental therapeutic procedures, such as those intended to promote CST regeneration. The results also suggest that the role of sensorimotor cortex in down-conditioning extends beyond generation of the essential CST activity.
Collapse
Affiliation(s)
- Xiang Yang Chen
- Laboratory of Nervous System Disorder, Wadsworth Center, New York State Department of Health and State University of New York, Albany, New York 12201-0509, USA.
| | | | | | | | | |
Collapse
|
39
|
Chen Y, Chen XY, Jakeman LB, Chen L, Stokes BT, Wolpaw JR. Operant conditioning of H-reflex can correct a locomotor abnormality after spinal cord injury in rats. J Neurosci 2006; 26:12537-43. [PMID: 17135415 PMCID: PMC6674902 DOI: 10.1523/jneurosci.2198-06.2006] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study asked whether operant conditioning of the H-reflex can modify locomotion in spinal cord-injured rats. Midthoracic transection of the right lateral column of the spinal cord produced a persistent asymmetry in the muscle activity underlying treadmill locomotion. The rats were then either exposed or not exposed to an H-reflex up-conditioning protocol that greatly increased right soleus motoneuron response to primary afferent input, and locomotion was reevaluated. H-reflex up-conditioning increased the right soleus burst and corrected the locomotor asymmetry. In contrast, the locomotor asymmetry persisted in the control rats. These results suggest that appropriately selected reflex conditioning protocols might improve function in people with partial spinal cord injuries. Such protocols might be especially useful when significant regeneration becomes possible and precise methods for reeducating the regenerated spinal cord neurons and synapses are needed for restoring effective function.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health and State University of New York, Albany, New York 12201, and
- Department of Physiology and Cell Biology, Ohio State University, Columbus, Ohio 43210
| | - Xiang Yang Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health and State University of New York, Albany, New York 12201, and
| | - Lyn B. Jakeman
- Department of Physiology and Cell Biology, Ohio State University, Columbus, Ohio 43210
| | - Lu Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health and State University of New York, Albany, New York 12201, and
| | - Bradford T. Stokes
- Department of Physiology and Cell Biology, Ohio State University, Columbus, Ohio 43210
| | - Jonathan R. Wolpaw
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health and State University of New York, Albany, New York 12201, and
| |
Collapse
|
40
|
Abstract
During the past several years, there has been increasing interest in the role of the blood-brain barrier (BBB) in epilepsy. Advances in neuroradiology have enhanced our ability to image and study the human cerebrovasculature, and further developments in the research of metabolic deficiencies linked to seizure disorders (e.g., GLUT1 deficiency), neuroinflammation, and multiple drug resistance to antiepileptic drugs (AEDs) have amplified the significance of the BBB's relationship to epilepsy. Prior to 1986, BBB research in epilepsy focused on three main areas: ultrastructural studies, brain glucose availability and transport, and clinical uses of AEDs. However, contrast-based imaging techniques and medical procedures such as BBB disruption provided a framework that demonstrated that the BBB could be reversibly disrupted by pathologic or iatrogenic manipulations, with important implications in terms of CNS drug delivery to "multiple drug resistant" brain. This concept of BBB breakdown for therapeutic purposes has also unveiled a previously unrecognized role for BBB failure as a possible etiologic mechanism in epileptogenesis. Finally, a growing body of evidence has shown that inflammatory mechanisms may participate in the pathological changes observed in epileptic brain, with increasing awareness that blood-borne cells or signals may participate in epileptogenesis by virtue of a leaky BBB. In this article we will review the relationships between BBB function and epilepsy. In particular, we will illustrate consensus and divergence between clinical reality and animal studies.
Collapse
Affiliation(s)
- Emily Oby
- Cleveland Clinic Foundation, Department of Neurological Surgery, Cerebrovascular Research, Ohio 44195, USA
| | | |
Collapse
|
41
|
Chen XY, Carp JS, Chen L, Wolpaw JR. Sensorimotor Cortex Ablation Prevents H-Reflex Up-Conditioning and Causes a Paradoxical Response to Down-Conditioning in Rats. J Neurophysiol 2006; 96:119-27. [PMID: 16598062 DOI: 10.1152/jn.01271.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Operant conditioning of the H-reflex, a simple model for skill acquisition, requires the corticospinal tract (CST) and does not require other major descending pathways. To further explore its mechanisms, we assessed the effects of ablating contralateral sensorimotor cortex (cSMC). In 22 Sprague–Dawley rats, the hindlimb area of left cSMC was ablated. EMG electrodes were implanted in the right soleus muscle and a stimulating cuff was placed around the right posterior tibial nerve. When EMG remained in a specified range, nerve stimulation just above the M response threshold elicited the H-reflex. In control mode, no reward occurred. In conditioning mode, reward occurred if H-reflex size was above (HRup mode) or below (HRdown mode) a criterion value. After exposure to the control mode for ≥10 days, each rat was exposed for another 50 days to the control mode, the HRup mode, or the HRdown mode. In control and HRup rats, final H-reflex size was not significantly different from initial H-reflex size. In contrast, in HRdown rats, final H-reflex size was significantly increased to an average of 136% of initial size. Thus like recent CST transection, cSMC ablation greatly impaired up-conditioning. However, unlike recent CST transection, cSMC produced a paradoxical response to down-conditioning: the H-reflex actually increased. These results confirm the critical role of cSMC in H-reflex conditioning and suggest that this role extends beyond producing essential CST activity. Its interactions with ipsilateral SMC or other areas contribute to the complex pattern of spinal and supraspinal plasticity that underlies H-reflex conditioning.
Collapse
Affiliation(s)
- Xiang Yang Chen
- Wadsworth Center, Laboratory of Nervous System Disorders, New York State Department of Health and State University of New York, Albany, NY 12201-0509, USA.
| | | | | | | |
Collapse
|
42
|
Chen Y, Chen XY, Jakeman LB, Schalk G, Stokes BT, Wolpaw JR. The interaction of a new motor skill and an old one: H-reflex conditioning and locomotion in rats. J Neurosci 2006; 25:6898-906. [PMID: 16033899 PMCID: PMC6725342 DOI: 10.1523/jneurosci.1684-05.2005] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
New and old motor skills can interfere with each other or interact in other ways. Because each skill entails a distributed pattern of activity-dependent plasticity, investigation of their interactions is facilitated by simple models. In a well characterized model of simple learning, rats and monkeys gradually change the size of the H-reflex, the electrical analog of the spinal stretch reflex. This study evaluates in normal rats the interactions of this new skill of H-reflex conditioning with the old well established skill of overground locomotion. In rats in which the soleus H-reflex elicited in the conditioning protocol (i.e., the conditioning H-reflex) had been decreased by down-conditioning, the H-reflexes elicited during the stance and swing phases of locomotion (i.e., the locomotor H-reflexes) were also smaller. Similarly, in rats in which the conditioning H-reflex had been increased by up-conditioning, the locomotor H-reflexes were also larger. Soleus H-reflex conditioning did not affect the duration, length, or right/left symmetry of the step cycle. However, the conditioned change in the stance H-reflex was positively correlated with change in the amplitude of the soleus locomotor burst, and the correlation was consistent with current estimates of the contribution of primary afferent input to the burst. Although H-reflex conditioning and locomotion did not interfere with each other, H-reflex conditioning did affect how locomotion was produced: it changed soleus burst amplitude and may have induced compensatory changes in the activity of other muscles. These results illustrate and clarify the subtlety and complexity of skill interactions. They also suggest that H-reflex conditioning might be used to improve the abnormal locomotion produced by spinal cord injury or other disorders of supraspinal control.
Collapse
Affiliation(s)
- Yi Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health, State University of New York, Albany, New York 12201, USA
| | | | | | | | | | | |
Collapse
|
43
|
Frigon A, Rossignol S. Functional plasticity following spinal cord lesions. PROGRESS IN BRAIN RESEARCH 2006; 157:231-260. [PMID: 17167915 DOI: 10.1016/s0079-6123(06)57016-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Spinal cord injury results in marked modification and reorganization of several reflex pathways caudal to the injury. The sudden loss or disruption of descending input engenders substantial changes at the level of primary afferents, interneurons, and motoneurons thus dramatically influencing sensorimotor interactions in the spinal cord. As a general rule reflexes are initially depressed following spinal cord injury due to severe reductions in motoneuron excitability but recover and in some instances become exaggerated. It is thought that modified inhibitory connections and/or altered transmission in some of these reflex pathways after spinal injury as well as the recovery and enhancement of membrane properties in motoneurons underlie several symptoms such as spasticity and may explain some characteristics of spinal locomotion observed in spinally transected animals. Indeed, after partial or complete spinal lesions at the last thoracic vertebra cats recover locomotion when the hindlimbs are placed on a treadmill. Although some deficits in spinal locomotion are related to lesion of specific descending motor pathways, other characteristics can also be explained by changes in the excitability of reflex pathways mentioned above. Consequently it may be the case that to reestablish a stable walking pattern that modified afferent inflow to the spinal cord incurred after injury must be normalized to enable a more normal re-expression of locomotor rhythm generating networks. Indeed, recent evidence demonstrates that step training, which has extensively been shown to facilitate and ameliorate locomotor recovery in spinal animals, directly influences transmission in simple reflex pathways after complete spinal lesions.
Collapse
Affiliation(s)
- Alain Frigon
- Center and Group for Neurological Sciences, CIHR Group in Neurological Sciences, CIHR Regenerative Medicine and Nanomedicine Team, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
44
|
Levi O, Lütjohann D, Devir A, von Bergmann K, Hartmann T, Michaelson DM. Regulation of hippocampal cholesterol metabolism by apoE and environmental stimulation. J Neurochem 2005; 95:987-97. [PMID: 16190879 DOI: 10.1111/j.1471-4159.2005.03441.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease is associated with genetic risk factors, of which the allele E4 of apolipoprotein E (apoE4) is the most prevalent, and it is also affected by environmental factors such as early life education. We have recently shown, utilizing apoE-deficient and apoE transgenic mice, that synaptogenesis in the hippocampus following environmental stimulation is affected by apoE. In view of the pivotal role of cholesterol in synaptic plasticity, and of its suggested role in synaptogenesis, we presently examined the effects of apoE and environmental stimulation on brain cholesterol homeostasis. The hippocampal levels of cholesterol and its precursors and metabolites in control mice were not affected by exposure to environmental stimulation. In contrast, the hippocampal levels of cholesterol and its precursors lathosterol and desmosterol and metabolite 24S-hydroxycholesterol were lower in apoE-deficient mice that were maintained in a regular environmental than those of corresponding control mice, whereas they were markedly elevated following environmental stimulation. Histological and immunohistochemical experiments revealed that the combined stimulatory effects of apoE deficiency and environmental stimulation on cholesterol metabolism were associated with marked activation of hippocampal astrocytes and with the abnormal accumulation of cholesterol in neurons and astrocytes. These effects were rescued similarly in apoE3 and apoE4 transgenic mice. These findings suggest that apoE plays an important role in the translocation of cholesterol from astrocytes to neurons in vivo and in the regulation and homeostasis of this process.
Collapse
Affiliation(s)
- Ofir Levi
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
45
|
Chen XY, Chen L, Wolpaw JR. Conditioned H-reflex increase persists after transection of the main corticospinal tract in rats. J Neurophysiol 2003; 90:3572-8. [PMID: 12917382 DOI: 10.1152/jn.00264.2003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The brain shapes spinal cord function throughout life. Operant conditioning of the H-reflex, the electrical analog of the spinal stretch reflex (SSR), is a relatively simple model for exploring the spinal cord plasticity underlying this functional change and may provide a new method for modifying spinal cord reflexes after spinal cord injury. In response to an operant conditioning protocol, rats can gradually increase (i.e., up-training mode) or decrease (i.e., down-training mode) the soleus H-reflex. This study explored the effects of midthoracic transection of the ipsilateral lateral column (LC) (rubrospinal, vestibulospinal, and reticulospinal tracts), the dorsal column corticospinal tract (CST), or the dorsal column ascending tract (DA) on maintenance of an H-reflex increase that has already occurred. Rats were implanted with EMG electrodes in the right soleus muscle and a nerve-stimulating cuff on the right posterior tibial nerve. After initial (i.e., control) H-reflex size was determined, the rats were exposed for 50 days to the up-training mode, in which reward was given when the H-reflex was above a criterion value. H-reflex size gradually rose to 168 +/- 12% (mean +/- SE) of its initial value. Each rat then received an LC, CST, or DA transection and continued under the up-training mode for 50 more days. None of the transections abolished the H-reflex increase. H-reflex size increased further to 197 +/- 19% of its initial value and did not differ significantly among LC, CST, and DA rats (P > 0.78 by ANOVA). Although earlier studies show that the main CST is needed for acquisition of H-reflex up-training and down-training and for maintenance of down-training, this study shows that it is not needed for maintenance of up-training. It adds to the evidence that H-reflex conditioning changes the spinal cord and that the spinal cord plasticity associated with up-training is different from that associated with down-training.
Collapse
Affiliation(s)
- Xiang Yang Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health, Albany, New York 12201, USA.
| | | | | |
Collapse
|
46
|
Leker RR, Shohami E, Constantini S. Experimental models of head trauma. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 83:49-54. [PMID: 12442621 DOI: 10.1007/978-3-7091-6743-4_9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Traumatic brain injury is one of the most common causes for chronic disability in young people. Despite this there are currently no widely available modes of therapy that would limit the extent of brain damage secondary to trauma. Therefore, new insights into the pathological mechanisms involved in head trauma possibly leading to the identification of new therapeutic targets are urgently needed. In order to attain these goals adequate animal models for traumatic brain injury are needed. In the following paper the authors will review the various animal models for head trauma and emphasize their potential strengths and weaknesses.
Collapse
Affiliation(s)
- R R Leker
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School, Hadassah University Hospital, Jerusalem, Israel
| | | | | |
Collapse
|
47
|
McCarron RM, Shohami E, Panikashvili D, Chen Y, Golech S, Strasser A, Mechoulam R, Spatz M. Antioxidant properties of the vasoactive endocannabinoid, 2-arachidonoyl glycerol (2-AG). ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 86:271-5. [PMID: 14753451 DOI: 10.1007/978-3-7091-0651-8_59] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Reactive oxygen species (ROS) were shown to play a role in altering blood-brain barrier (BBB) permeability and formation of brain edema induced by trauma and/or ischemia. 2-arachidonoyl glycerol (2-AG), a novel, potent vasodilatory and cytoprotective endocannabinoid has been implicated to act as an antioxidative agent. This study examines: 1) the possible 2-AG modulation of BBB injury and edema formation induced by closed head injury (CHI); and 2) comparable effects between 2-AG and 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (TPL), a known antioxidant nitroxide on endothelial Ca2+ and cytoskeletal responses to H2O2 (ROS). 2-AG treatment reduced the CHI-induced increase in BBB permeability and brain edema. The endothelial H2O2-stimulated Ca2+ mobilization and cytoskeleton (vimentin) rearrangement was modified by either 2-AG or TPL. These findings provide evidence of 2-AG antioxidant activity and are consistent with the involvement of ROS in the pathomechanism of CHI-induced BBB injury and brain edema.
Collapse
Affiliation(s)
- R M McCarron
- Resuscitative Medicine Department, Naval Medical Research Center, Forest Glen, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen XY, Chen L, Wolpaw JR, Jakeman LB. Corticospinal tract transection reduces H-reflex circadian rhythm in rats. Brain Res 2002; 942:101-8. [PMID: 12031858 DOI: 10.1016/s0006-8993(02)02702-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In freely moving rats and monkeys, H-reflex amplitude displays a marked circadian variation without change in background motoneuron tone. In rats, the H-reflex is largest around noon and smallest around midnight. The present study evaluated in rats the effects on this rhythm of calibrated contusions of mid-thoracic spinal cord and mid-thoracic transection of specific spinal cord pathways. In 33 control rats, rhythm amplitude averaged 29.0(+/-2.6 S.E.)% of H-reflex amplitude. Contusion injuries at T8-9 that destroyed 53-88% of the white matter significantly reduced the rhythm to 18.9(+/-2.4)% of H-reflex amplitude. Transection of the ipsilateral lateral column, which contains the rubrospinal, vestibulospinal, and reticulospinal tracts, or bilateral transection of the dorsal column ascending tract did not affect rhythm amplitude or phase. In contrast, bilateral transection of the main corticospinal tract significantly reduced the rhythm to 14.7(+/-6.6)%. These results indicate that the H-reflex circadian rhythm depends in part on descending influence from the brain and that this influence is conveyed by the main corticospinal tract.
Collapse
Affiliation(s)
- Xiang Yang Chen
- Wadsworth Center, New York State Department of Health and State University of New York, P.O. Box 509, Albany, NY 12201-0509, USA.
| | | | | | | |
Collapse
|
49
|
Chen XY, Wolpaw JR. Probable corticospinal tract control of spinal cord plasticity in the rat. J Neurophysiol 2002; 87:645-52. [PMID: 11826033 DOI: 10.1152/jn.00391.2001] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Descending activity from the brain shapes spinal cord reflex function throughout life, yet the mechanisms responsible for this spinal cord plasticity are poorly understood. Operant conditioning of the H-reflex, the electrical analogue of the spinal stretch reflex, is a simple model for investigating these mechanisms. An earlier study in the Sprague-Dawley rat showed that acquisition of an operantly conditioned decrease in the soleus H-reflex is not prevented by mid-thoracic transection of the ipsilateral lateral column (LC), which contains the rubrospinal, reticulospinal, and vestibulospinal tracts, and is prevented by transection of the dorsal column, which contains the main corticospinal tract (CST) and the dorsal column ascending tract (DA). The present study explored the effects of CST or DA transection on acquisition of an H-reflex decrease, and the effects of LC, CST, or DA transection on maintenance of an established decrease. CST transection prior to conditioning prevented acquisition of H-reflex decrease, while DA transection did not do so. CST transection after H-reflex decrease had been acquired led to gradual loss of the decrease over 10 days, and resulted in an H-reflex that was significantly larger than the original, naive H-reflex. In contrast, LC or DA transection after H-reflex decrease had been acquired did not affect maintenance of the decrease. These results, in combination with the earlier study, strongly imply that in the rat the corticospinal tract (CST) is essential for acquisition and maintenance of operantly conditioned decrease in the H-reflex and that other major spinal cord pathways are not essential. This previously unrecognized aspect of CST function gives insight into the processes underlying acquisition and maintenance of motor skills and could lead to novel methods for inducing, guiding, and assessing recovery of function after spinal cord injury.
Collapse
Affiliation(s)
- Xiang Yang Chen
- Laboratory of Nervous System Disorders, Wadsworth Center, New York State Department of Health and State University of New York, P.O. Box 509, Empire State Plaza, Albany, NY 12201.
| | | |
Collapse
|
50
|
Lynch JR, Pineda JA, Morgan D, Zhang L, Warner DS, Benveniste H, Laskowitz DT. Apolipoprotein E affects the central nervous system response to injury and the development of cerebral edema. Ann Neurol 2002; 51:113-7. [PMID: 11782990 DOI: 10.1002/ana.10098] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Apolipoprotein E has been implicated in modifying neurological outcome after traumatic brain injury, although the mechanisms by which this occurs remain poorly defined. To investigate the role of endogenous apolipoprotein E following acute brain injury, noninvasive magnetic resonance imaging was performed on anesthetized mice following closed head injury. Effacement of the lateral ventricle was used as a radiographic surrogate for cerebral edema. At 24 hours following injury, apolipoprotein E-deficient animals had a greater degree of cerebral edema as compared to matched controls. In addition, the brains of apolipoprotein E-deficient animals had a significantly greater upregulation of tissue necrosis factor alpha messenger ribonucleic acid as compared to controls as early as 1-hr post injury. Thus, modulation of the endogenous central nervous system inflammatory response may be one mechanism by which apolipoprotein E affects outcome following acute brain injury.
Collapse
Affiliation(s)
- John R Lynch
- Multidisciplinary Neuroprotection Laboratory, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|