1
|
Ruscher K, Michalettos G, Abu Hamdeh S, Clausen F, Nolan AL, Flygt J, Özen I, Marklund N. Persistent increase of Nogo-A-positive cells and dynamic reduction in oligodendroglia lineage cells in white matter regions following experimental and clinical traumatic brain injury. J Neuropathol Exp Neurol 2025; 84:423-435. [PMID: 40085014 PMCID: PMC12012378 DOI: 10.1093/jnen/nlaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
White matter (WM) disruption and atrophy is a consequence of traumatic brain injury (TBI) and contributes to persisting cognitive impairment. An increased expression of the myelin-associated axonal outgrowth inhibitor Nogo-A and oligodendrocyte pathology might be negatively associated with postinjury WM changes. Here, we analyzed brain tissue from severe TBI patients, obtained by surgical decompression in the early postinjury phase and postmortem brain tissue of long-term TBI survivors and observed an increased number of Nogo-A+ cells in WM tracts such as the corpus callosum (CC). Likewise, the number of Nogo-A+ cells in the CC was increased from day 7 postinjury to 6 months postinjury (mpi) following central fluid percussion injury (cFPI) in mice. In addition, the number of Olig2+ cells in the CC and capsula externa remained constant, while the numbers of Olig2+/CC1+ and GST-π+ mature oligodendrocytes declined throughout the observation time of 18 months. A significantly lower number of Olig2+/CC1+ cells was found in cFPI mice compared to controls at 18 mpi. Persistent vulnerability of oligodendrocytes in combination with dynamic alterations of Nogo-A expression may have implications for the WM atrophy and insufficient recovery observed after TBI.
Collapse
Affiliation(s)
- Karsten Ruscher
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Division of Neurosurgery, Laboratory for Experimental Brain Research, Lund University, Lund, Sweden
| | - Georgios Michalettos
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Sami Abu Hamdeh
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Johanna Flygt
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Ilknur Özen
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
2
|
Powers BE, Ton ST, Farrer RG, Chaudhary S, Nockels RP, Kartje GL, Tsai SY. Anti-Nogo-A Antibody Therapy Improves Functional Outcome Following Traumatic Brain Injury. Neurorehabil Neural Repair 2023; 37:682-693. [PMID: 37837331 PMCID: PMC10843026 DOI: 10.1177/15459683231203194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) can cause sensorimotor deficits, and recovery is slow and incomplete. There are no effective pharmacological treatments for recovery from TBI, but research indicates potential for anti-Nogo-A antibody (Ab) therapy. This Ab neutralizes Nogo-A, an endogenous transmembrane protein that inhibits neuronal plasticity and regeneration. OBJECTIVE We hypothesized that anti-Nogo-A Ab treatment following TBI results in disinhibited axonal growth from the contralesional cortex, the establishment of new compensatory neuronal connections, and improved function. METHODS We modeled TBI in rats using the controlled cortical impact method, resulting in focal brain damage and motor deficits like those observed in humans with a moderate cortical TBI. Rats were trained on the skilled forelimb reaching task and the horizontal ladder rung walking task. They were then given a TBI, targeting the caudal forelimb motor cortex, and randomly divided into 3 groups: TBI-only, TBI + Anti-Nogo-A Ab, and TBI + Control Ab. Testing resumed 3 days after TBI and continued for 8 weeks, when rats received an injection of the anterograde neuronal tracer, biotinylated dextran amine (BDA), into the corresponding area contralateral to the TBI. RESULTS We observed significant improvement in rats that received anti-Nogo-A Ab treatment post-TBI compared to controls. Analysis of BDA-positive axons revealed that anti-Nogo-A Ab treatment resulted in cortico-rubral plasticity to the deafferented red nucleus. Conclusions. Anti-Nogo-A Ab treatment may improve functional recovery via neuronal plasticity to brain areas important for skilled movements, and this treatment shows promise to improve outcomes in humans who have suffered a TBI.
Collapse
Affiliation(s)
- Brian E Powers
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| | - Son T Ton
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| | | | | | - Russ P Nockels
- Department of Neurological Surgery, Loyola University Medical Center, Maywood, IL, USA
| | - Gwendolyn L Kartje
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
- Department of Molecular Pharmacology and Neuroscience, Loyola University Health Sciences Division, Maywood, IL, USA
| | - Shih-Yen Tsai
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| |
Collapse
|
3
|
Selective plasticity of callosal neurons in the adult contralesional cortex following murine traumatic brain injury. Nat Commun 2022; 13:2659. [PMID: 35551446 PMCID: PMC9098892 DOI: 10.1038/s41467-022-29992-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) results in deficits that are often followed by recovery. The contralesional cortex can contribute to this process but how distinct contralesional neurons and circuits respond to injury remains to be determined. To unravel adaptations in the contralesional cortex, we used chronic in vivo two-photon imaging. We observed a general decrease in spine density with concomitant changes in spine dynamics over time. With retrograde co-labeling techniques, we showed that callosal neurons are uniquely affected by and responsive to TBI. To elucidate circuit connectivity, we used monosynaptic rabies tracing, clearing techniques and histology. We demonstrate that contralesional callosal neurons adapt their input circuitry by strengthening ipsilateral connections from pre-connected areas. Finally, functional in vivo two-photon imaging demonstrates that the restoration of pre-synaptic circuitry parallels the restoration of callosal activity patterns. Taken together our study thus delineates how callosal neurons structurally and functionally adapt following a contralateral murine TBI. Which contralesional circuits adapt after traumatic brain injury (TBI) is unclear. Here the authors used in vivo imaging, retrograde labeling, rabies tracing, clearing and functional imaging to demonstrate that callosal neurons selectively adapt after TBI in mice.
Collapse
|
4
|
Inhibition of HDAC increases BDNF expression and promotes neuronal rewiring and functional recovery after brain injury. Cell Death Dis 2020; 11:655. [PMID: 32811822 PMCID: PMC7434917 DOI: 10.1038/s41419-020-02897-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022]
Abstract
Brain injury causes serious motor, sensory, and cognitive disabilities. Accumulating evidence has demonstrated that histone deacetylase (HDAC) inhibitors exert neuroprotective effects against various insults to the central nervous system (CNS). In this study, we investigated the effects of the HDAC inhibition on the expression of brain-derived neurotrophic factor (BDNF) and functional recovery after traumatic brain injury (TBI) in mice. Administration of class I HDAC inhibitor increased the number of synaptic boutons in rewiring corticospinal fibers and improved the recovery of motor functions after TBI. Immunohistochemistry results showed that HDAC2 is mainly expressed in the neurons of the mouse spinal cord under normal conditions. After TBI, HDAC2 expression was increased in the spinal cord after 35 days, whereas BDNF expression was decreased after 42 days. Administration of CI-994 increased BDNF expression after TBI. Knockdown of HDAC2 elevated H4K5ac enrichment at the BDNF promoter, which was decreased following TBI. Together, our findings suggest that HDAC inhibition increases expression of neurotrophic factors, and promote neuronal rewiring and functional recovery following TBI.
Collapse
|
5
|
Feter N, Freitas M, Gonzales N, Umpierre D, Cardoso R, Rombaldi A. Effects of physical exercise on myelin sheath regeneration: A systematic review and meta-analysis. Sci Sports 2018. [DOI: 10.1016/j.scispo.2017.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
6
|
Repeated “Day 1” FOB testing in ICH S7A safety assessment protocols: The influence of within- and between-session learning. J Pharmacol Toxicol Methods 2017; 85:61-72. [DOI: 10.1016/j.vascn.2017.02.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/14/2016] [Accepted: 02/10/2017] [Indexed: 11/17/2022]
|
7
|
Shepherd DJ, Tsai SY, O'Brien TE, Farrer RG, Kartje GL. Anti-Nogo-A Immunotherapy Does Not Alter Hippocampal Neurogenesis after Stroke in Adult Rats. Front Neurosci 2016; 10:467. [PMID: 27803646 PMCID: PMC5067305 DOI: 10.3389/fnins.2016.00467] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/28/2016] [Indexed: 12/30/2022] Open
Abstract
Ischemic stroke is a leading cause of adult disability, including cognitive impairment. Our laboratory has previously shown that treatment with function-blocking antibodies against the neurite growth inhibitory protein Nogo-A promotes functional recovery after stroke in adult and aged rats, including enhancing spatial memory performance, for which the hippocampus is critically important. Since spatial memory has been linked to hippocampal neurogenesis, we investigated whether anti-Nogo-A treatment increases hippocampal neurogenesis after stroke. Adult rats were subject to permanent middle cerebral artery occlusion followed 1 week later by 2 weeks of antibody treatment. Cellular proliferation in the dentate gyrus was quantified at the end of treatment, and the number of newborn neurons was determined at 8 weeks post-stroke. Treatment with both anti-Nogo-A and control antibodies stimulated the accumulation of new microglia/macrophages in the dentate granule cell layer, but neither treatment increased cellular proliferation or the number of newborn neurons above stroke-only levels. These results suggest that anti-Nogo-A immunotherapy does not increase post-stroke hippocampal neurogenesis.
Collapse
Affiliation(s)
- Daniel J Shepherd
- Neuroscience Institute, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA; Research Service, Edward Hines Jr. VA HospitalHines, IL, USA
| | - Shih-Yen Tsai
- Research Service, Edward Hines Jr. VA Hospital Hines, IL, USA
| | - Timothy E O'Brien
- Department of Mathematics and Statistics, Loyola University Chicago Chicago, IL, USA
| | - Robert G Farrer
- Research Service, Edward Hines Jr. VA Hospital Hines, IL, USA
| | - Gwendolyn L Kartje
- Neuroscience Institute, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA; Research Service, Edward Hines Jr. VA HospitalHines, IL, USA; Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Health Sciences DivisionMaywood, IL, USA
| |
Collapse
|
8
|
Liu GM, Luo YG, Li J, Xu K. Knockdown of Nogo gene by short hairpin RNA interference promotes functional recovery of spinal cord injury in a rat model. Mol Med Rep 2016; 13:4431-6. [PMID: 27035338 DOI: 10.3892/mmr.2016.5072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 02/01/2016] [Indexed: 11/05/2022] Open
Abstract
The specific myelin component Nogo protein is one of the major inhibitory molecules of spinal cord axonal outgrowth following spinal cord injury. The present study aimed to investigate the effects of silencing Nogo protein with shRNA interference on the promotion of functional recovery in a rat model with spinal cord hemisection. Nogo-A short hairpin RNAs (Nogo shRNAs) were constructed and transfected into rats with spinal cord hemisection by adenovirus-mediated transfection. Reverse transcription‑polymerase chain reaction and western blotting were performed to analyze the expression of Nogo-A and Growth Associated Protein 43 (GAP-43). In addition, Basso Beattie Bresnahan (BBB) scores were used to assess the functional recovery of rats following spinal cord injury. The results demonstrated that expression of the Nogo‑A gene was observed to be downregulated following transfection and GAP‑43 expression was observed to increase. The BBB scores were increased following treatment with Nogo shRNAs, indicating functional recovery of the injured nerves. Thus, Nogo-A shRNA interference can knockdown Nogo gene expression and upregulate GAP-43 to promote the functional recovery of spinal cord injury in rats. This finding may advance progress toward assisting the regeneration of injured neurons through the use of Nogo-A shRNA.
Collapse
Affiliation(s)
- Guo-Min Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yun-Gang Luo
- Department of Stomatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Juan Li
- Department of Preventive Medicine, College of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Kun Xu
- Department of Preventive Medicine, College of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
9
|
Osier ND, Carlson SW, DeSana A, Dixon CE. Chronic Histopathological and Behavioral Outcomes of Experimental Traumatic Brain Injury in Adult Male Animals. J Neurotrauma 2015; 32:1861-82. [PMID: 25490251 PMCID: PMC4677114 DOI: 10.1089/neu.2014.3680] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this review is to survey the use of experimental animal models for studying the chronic histopathological and behavioral consequences of traumatic brain injury (TBI). The strategies employed to study the long-term consequences of TBI are described, along with a summary of the evidence available to date from common experimental TBI models: fluid percussion injury; controlled cortical impact; blast TBI; and closed-head injury. For each model, evidence is organized according to outcome. Histopathological outcomes included are gross changes in morphology/histology, ventricular enlargement, gray/white matter shrinkage, axonal injury, cerebrovascular histopathology, inflammation, and neurogenesis. Behavioral outcomes included are overall neurological function, motor function, cognitive function, frontal lobe function, and stress-related outcomes. A brief discussion is provided comparing the most common experimental models of TBI and highlighting the utility of each model in understanding specific aspects of TBI pathology. The majority of experimental TBI studies collect data in the acute postinjury period, but few continue into the chronic period. Available evidence from long-term studies suggests that many of the experimental TBI models can lead to progressive changes in histopathology and behavior. The studies described in this review contribute to our understanding of chronic TBI pathology.
Collapse
Affiliation(s)
- Nicole D. Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anthony DeSana
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Seton Hill University, Greensburg, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Amy M, Staehlin O, René F, Blasco H, Marouillat S, Daoud H, Vourc'h P, Gordon PH, Camu W, Corcia P, Loeffler JP, Palkovits M, Sommer WH, Andres CR. A common functional allele of the Nogo receptor gene, reticulon 4 receptor (RTN4R), is associated with sporadic amyotrophic lateral sclerosis in a French population. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16:490-6. [PMID: 26083872 DOI: 10.3109/21678421.2015.1051988] [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: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis is sporadic (SALS) in 90% of cases and has complex environmental and genetic influences. Nogo protein inhibits neurite outgrowth and is overexpressed in muscle in ALS. Our aims were to study the reticulon 4 receptor gene RTN4R which encodes Nogo 1 receptor (NgR1) in SALS, to test if the variants were associated with variable expression of the gene and whether NgR1 protein expression was modified in a transgenic mouse model of ALS. We genotyped three single nucleotide polymorphisms (SNPs; rs701421, rs701427, and rs1567871) of the RTN4R gene in 364 SALS French patients and 430 controls. We examined expression of RTN4R mRNA by quantitative PCR in control post mortem human brain tissue. We determined the expression of NgR1 protein in spinal motor neurons from a SOD1 G86R ALS mouse model. We observed significant associations between SALS and RTN4R alleles. Messenger RNA expression from RTN4R in human cortical brain tissue correlated significantly with the genotypes of rs701427. NgR1 protein expression was reduced in Nogo A positive motor neurons from diseased transgenic animals. In conclusion, these observations suggest that a functional RTN4R gene variant is associated with SALS. This variant may act in concert with other genetic variants or environmental influences.
Collapse
Affiliation(s)
- Maïté Amy
- a INSERM U930 , Tours , France.,b Université François Rabelais , Tours , France
| | - Oliver Staehlin
- c Institute of Psychopharmacology at Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg , Mannheim , Germany
| | - Frédérique René
- d INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence , Strasbourg , France.,e Université de Strasbourg, UMRS 1118 , Strasbourg , France
| | - Hélène Blasco
- a INSERM U930 , Tours , France.,b Université François Rabelais , Tours , France.,f Service de Biochimie et Biologie Moléculaire, Hôpital Bretonneau , CHRU de Tours, Tours , France
| | | | | | - Patrick Vourc'h
- a INSERM U930 , Tours , France.,b Université François Rabelais , Tours , France.,f Service de Biochimie et Biologie Moléculaire, Hôpital Bretonneau , CHRU de Tours, Tours , France
| | - Paul H Gordon
- g Northern Navajo Medical Center , Shiprock NM , USA
| | - William Camu
- h ALS Centre, Hôpital Gui de Chauliac, CHU de Montpellier , Montpellier , France
| | - Philippe Corcia
- a INSERM U930 , Tours , France.,b Université François Rabelais , Tours , France.,i ALS Centre, Department of Neurology , CHRU de Tours, France
| | - Jean-Philippe Loeffler
- d INSERM U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence , Strasbourg , France.,e Université de Strasbourg, UMRS 1118 , Strasbourg , France
| | - Miklós Palkovits
- j Laboratory of Neuromorphology, Semmelweis University and the Hungarian Academy of Sciences , Budapest , Hungary
| | - Wolfgang H Sommer
- c Institute of Psychopharmacology at Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg , Mannheim , Germany
| | - Christian R Andres
- a INSERM U930 , Tours , France.,b Université François Rabelais , Tours , France.,f Service de Biochimie et Biologie Moléculaire, Hôpital Bretonneau , CHRU de Tours, Tours , France
| |
Collapse
|
11
|
Hånell A, Marklund N. Structured evaluation of rodent behavioral tests used in drug discovery research. Front Behav Neurosci 2014; 8:252. [PMID: 25100962 PMCID: PMC4106406 DOI: 10.3389/fnbeh.2014.00252] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 07/03/2014] [Indexed: 12/01/2022] Open
Abstract
A large variety of rodent behavioral tests are currently being used to evaluate traits such as sensory-motor function, social interactions, anxiety-like and depressive-like behavior, substance dependence and various forms of cognitive function. Most behavioral tests have an inherent complexity, and their use requires consideration of several aspects such as the source of motivation in the test, the interaction between experimenter and animal, sources of variability, the sensory modality required by the animal to solve the task as well as costs and required work effort. Of particular importance is a test’s validity because of its influence on the chance of successful translation of preclinical results to clinical settings. High validity may, however, have to be balanced against practical constraints and there are no behavioral tests with optimal characteristics. The design and development of new behavioral tests is therefore an ongoing effort and there are now well over one hundred tests described in the contemporary literature. Some of them are well established following extensive use, while others are novel and still unproven. The task of choosing a behavioral test for a particular project may therefore be daunting and the aim of the present review is to provide a structured way to evaluate rodent behavioral tests aimed at drug discovery research.
Collapse
Affiliation(s)
- Anders Hånell
- Department of Neuroscience, Section for Neurosurgery, Uppsala University Uppsala, Sweden ; Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine Richmond, VA, USA
| | - Niklas Marklund
- Department of Neuroscience, Section for Neurosurgery, Uppsala University Uppsala, Sweden
| |
Collapse
|
12
|
Evans TM, Van Remmen H, Purkar A, Mahesula S, Gelfond JA, Sabia M, Qi W, Lin AL, Jaramillo CA, Haskins WE. Microwave & Magnetic (M 2) Proteomics of a Mouse Model of Mild Traumatic Brain Injury. TRANSLATIONAL PROTEOMICS 2014; 3:10-21. [PMID: 26157646 DOI: 10.1016/j.trprot.2014.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Short-term increases in oxidative stress and decreases in motor function, including debilitating effects on balance and motor control, can occur following primary mild traumatic brain injuries (mTBI). However, the long-term effects on motor unit impairment and integrity as well as the molecular mechanisms underlying secondary injuries are poorly understood. We hypothesized that changes in central nervous system-specific protein (CSP) expression might correlate to these long-term effects. To test our hypothesis, we longitudinally assessed a closed-skull mTBI mouse model, vs. sham control, at 1, 7, 30, and 120 days post-injury. Motor impairment was determined by rotarod and grip strength performance measures, while motor unit integrity was determined using electromyography. Relative protein expression was determined by microwave & magnetic (M2) proteomics of ipsilateral brain tissue, as previously described. Isoprostane measurements were performed to confirm a primary oxidative stress response. Decoding the relative expression of 476 ± 56 top-ranked proteins for each specimen revealed statistically significant changes in the expression of two well-known CSPs at 1, 7 and 30 days post-injury: P < 0.001 for myelin basic protein (MBP) and P < 0.05 for myelin associated glycoprotein (MAG). This was confirmed by Western blot. Moreover, MAG, αII-spectrin (SPNA2) and neurofilament light (NEFL) expression at 30 days post-injury were directly related to grip strength (P < 0.05). While higher-powered studies of larger cohorts merit further investigation, this study supports the proof-of-concept that M2 proteomics is a rapid method to quantify putative protein biomarkers and therapeutic targets of mTBI and suggests the feasibility of CSP expression correlations to long-term effects on motor impairment.
Collapse
Affiliation(s)
- Teresa M Evans
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Holly Van Remmen
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA ; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Anjali Purkar
- Pediatric Biochemistry Laboratory, Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Swetha Mahesula
- Pediatric Biochemistry Laboratory, Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas, USA
| | - J Al Gelfond
- Department of Epidemiology & Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Marian Sabia
- South Texas Veterans Health Care System, San Antonio, Texas, USA, Department of Rehabilitation Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Wenbo Qi
- South Texas Veterans Health Care System, San Antonio, Texas, USA, Department of Rehabilitation Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ai-Ling Lin
- Research Imaging Institute, Barshop Institute and Department of Cellular & Structural Biology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, USA
| | - Carlos A Jaramillo
- Polytrauma Rehabilitation Center, South Texas Veterans Health Care System, San Antonio, Texas, USA, Department of Rehabilitation Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - William E Haskins
- Pediatric Biochemistry Laboratory, Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
13
|
Petrasek T, Prokopova I, Sladek M, Weissova K, Vojtechova I, Bahnik S, Zemanova A, Schönig K, Berger S, Tews B, Bartsch D, Schwab ME, Sumova A, Stuchlik A. Nogo-A-deficient Transgenic Rats Show Deficits in Higher Cognitive Functions, Decreased Anxiety, and Altered Circadian Activity Patterns. Front Behav Neurosci 2014; 8:90. [PMID: 24672453 PMCID: PMC3957197 DOI: 10.3389/fnbeh.2014.00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/02/2014] [Indexed: 11/19/2022] Open
Abstract
Decreased levels of Nogo-A-dependent signaling have been shown to affect behavior and cognitive functions. In Nogo-A knockout and knockdown laboratory rodents, behavioral alterations were observed, possibly corresponding with human neuropsychiatric diseases of neurodevelopmental origin, particularly schizophrenia. This study offers further insight into behavioral manifestations of Nogo-A knockdown in laboratory rats, focusing on spatial and non-spatial cognition, anxiety levels, circadian rhythmicity, and activity patterns. Demonstrated is an impairment of cognitive functions and behavioral flexibility in a spatial active avoidance task, while non-spatial memory in a step-through avoidance task was spared. No signs of anhedonia, typical for schizophrenic patients, were observed in the animals. Some measures indicated lower anxiety levels in the Nogo-A-deficient group. Circadian rhythmicity in locomotor activity was preserved in the Nogo-A knockout rats and their circadian period (tau) did not differ from controls. However, daily activity patterns were slightly altered in the knockdown animals. We conclude that a reduction of Nogo-A levels induces changes in CNS development, manifested as subtle alterations in cognitive functions, emotionality, and activity patterns.
Collapse
Affiliation(s)
- Tomas Petrasek
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic ; First Faculty of Medicine, Charles University in Prague , Prague , Czech Republic
| | - Iva Prokopova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Martin Sladek
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Kamila Weissova
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Iveta Vojtechova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Stepan Bahnik
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic ; Social Psychology, Department of Psychology II, University of Würzburg , Würzburg , Germany
| | - Anna Zemanova
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Stefan Berger
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Björn Tews
- Brain Research Institute, University of Zurich , Zurich , Switzerland ; Neurosciences, Department of Biology, Swiss Federal Institute of Technology Zurich , Zurich , Switzerland ; Division of Molecular Mechanisms of Tumor Invasion, German Cancer Research Center , Heidelberg , Germany
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health , Mannheim , Germany
| | - Martin E Schwab
- Brain Research Institute, University of Zurich , Zurich , Switzerland ; Neurosciences, Department of Biology, Swiss Federal Institute of Technology Zurich , Zurich , Switzerland
| | - Alena Sumova
- Department of Neurohumoral Regulations, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Ales Stuchlik
- Department of Neurophysiology of Memory, Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| |
Collapse
|
14
|
LIU FENG, LIAO FAN, LI WEI, HAN YONGFENG, LIAO DAGUANG. Progesterone alters Nogo-A, GFAP and GAP-43 expression in a rat model of traumatic brain injury. Mol Med Rep 2014; 9:1225-31. [DOI: 10.3892/mmr.2014.1967] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 01/31/2014] [Indexed: 11/06/2022] Open
|
15
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Mahmood A, Wu H, Qu C, Mahmood S, Xiong Y, Kaplan DL, Chopp M. Suppression of neurocan and enhancement of axonal density in rats after treatment of traumatic brain injury with scaffolds impregnated with bone marrow stromal cells. J Neurosurg 2014; 120:1147-55. [PMID: 24460490 DOI: 10.3171/2013.12.jns131362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECT Neurocan is a major form of growth-inhibitory molecule (growth-IM) that suppresses axonal regeneration after neural injury. Bone marrow stromal cells (MSCs) have been shown to inhibit neurocan expression in vitro and in animal models of cerebral ischemia. Therefore, the present study was designed to investigate the effects of treatment of MSCs impregnated with collagen scaffolds on neurocan expression after traumatic brain injury (TBI). METHODS Adult male Wistar rats were injured with controlled cortical impact and treated with saline, human MSCs (hMSCs) (3 × 10(6)) alone, or hMSCs (3 × 10(6)) impregnated into collagen scaffolds (scaffold + hMSCs) transplanted into the lesion cavity 7 days after TBI (20 rats per group). Rats were sacrificed 14 days after TBI, and brain tissues were harvested for immunohistochemical studies, Western blot analyses, laser capture microdissections, and quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) to evaluate neurocan protein and gene expressions after various treatments. RESULTS Animals treated with scaffold + hMSCs after TBI showed increased axonal and synaptic densities compared with the other groups. Scaffold + hMSC treatment was associated with reduced TBI-induced neurocan protein expression and upregulated growth-associated protein 43 (GAP-43) and synaptophysin expression in the lesion boundary zone. In addition, animals in the scaffold + hMSC group had decreased neurocan transcription in reactive astrocytes after TBI. Reduction of neurocan expression was significantly greater in the scaffold + hMSC group than in the group treated with hMSCs alone. CONCLUSIONS The results of this study show that transplanting hMSCs with scaffolds enhances the effect of hMSCs on axonal plasticity in TBI rats. This enhanced axonal plasticity may partially be attributed to the downregulation of neurocan expression by hMSC treatment after injury.
Collapse
|
17
|
Mahmood A, Wu H, Qu C, Mahmood S, Xiong Y, Kaplan D, Chopp M. Down-regulation of Nogo-A by collagen scaffolds impregnated with bone marrow stromal cell treatment after traumatic brain injury promotes axonal regeneration in rats. Brain Res 2013; 1542:41-8. [PMID: 24177046 DOI: 10.1016/j.brainres.2013.10.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/23/2013] [Indexed: 10/26/2022]
Abstract
Nogo-A is a major form of growth inhibitory molecule (growth-IM) which inhibits axonal regeneration and neurite regrowth after neural injury. Bone marrow stromal cells (MSCs) have been shown to inhibit Nogo-A expression in vitro and in cerebral ischemic animal models. The present study was designed to investigate the effects of treatment with human MSCs (hMSCs) impregnated into collagen scaffolds on the expression of Nogo-A and axonal plasticity after traumatic brain injury (TBI). Adult male Wistar rats were injured with controlled cortical impact and treated either with saline, hMSCs-alone or hMSCs impregnated into collagen scaffolds (scaffold+hMSC) transplanted into the lesion cavity 7 days after TBI. Rats were sacrificed 14 days after TBI and brain tissues were harvested for immunohistochemical studies, Western blot analysis, laser capture microdissections and qRT-PCR to evaluate axonal density and Nogo-A protein and gene expressions. Our data showed that treatment of TBI with scaffold+hMSC significantly decreased TBI-induced Nogo-A protein expression and increased axonal density compared to saline and hMSC-alone treatments. In addition, scaffold+hMSC transplantation decreased Nogo-A transcription in oligodendrocytes after TBI. Scaffold+hMSC treatment was superior to hMSC-alone treatment in suppressing Nogo-A expression and enhancing axonal regeneration after TBI. Our data suggest that transplanting hMSCs with scaffolds down-regulates Nogo-A transcription and protein expression which may partially contribute to the enhanced axonal regeneration after TBI.
Collapse
Affiliation(s)
- Asim Mahmood
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Hongtao Wu
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Changsheng Qu
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Selina Mahmood
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Ye Xiong
- Department of Neurosurgery, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - David Kaplan
- Department of Biomedical Engineering, Science and Technology Center, Room 251, Tufts University, Boston, MA 02155, USA.
| | - Michael Chopp
- Department of Neurology, 2799W Grand Blvd, Henry Ford Hospital, Detroit, MI 48202, USA; Department of Physics, Oakland University, 2200 North Squirrel Road, Rochester, MI 48309-4401, USA.
| |
Collapse
|
18
|
VanGuilder Starkey HD, Sonntag WE, Freeman WM. Increased hippocampal NgR1 signaling machinery in aged rats with deficits of spatial cognition. Eur J Neurosci 2013; 37:1643-58. [PMID: 23438185 DOI: 10.1111/ejn.12165] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/18/2013] [Accepted: 01/21/2013] [Indexed: 01/31/2023]
Abstract
Myelin-associated inhibitor/NgR1 signaling has important roles in modulation of synaptic plasticity, with demonstrated effects on cognitive function. We have previously demonstrated that NgR1 and its ligands are upregulated in the hippocampus of aged rats with impaired spatial learning and memory, but it is unknown whether increased expression of these proteins indicates a potential increase in pathway signaling because NgR1 requires co-receptors for signal transduction through RhoA. Two co-receptor complexes have been identified to date, comprised of NgR1 and LINGO-1, and either p75 or TROY. In this study, we assessed the expression of LINGO-1, p75 and TROY, and the downstream effector RhoA in mature adult (12 months) and aged (26 months) male Fischer 344/Brown Norway hybrid rats classified as cognitively impaired or cognitively intact by Morris water maze testing. The hippocampal distribution of NgR1 and its co-receptors was assessed to determine whether receptor/co-receptor interaction, and therefore signaling through this pathway, is possible. Protein expression of LINGO-1, p75, TROY and RhoA was significantly elevated in cognitively impaired, but not intact, aged rats compared with mature adults, and expression levels correlated significantly with water maze performance. Co-localization of NgR1 with LINGO-1, p75 and TROY was observed in hippocampal neurons of aged, cognitively impaired rats. Further, expression profiles of NgR1 pathway components were demonstrated to classify rats as cognitively intact or cognitively impaired with high accuracy. Together, this suggests that hippocampal induction of this pathway is a conserved phenomenon in cognitive decline that may impair learning and memory by suppressing neuronal plasticity.
Collapse
Affiliation(s)
- Heather D VanGuilder Starkey
- Department of Pharmacology, R130 Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | |
Collapse
|
19
|
Mahmood A, Wu H, Qu C, Xiong Y, Chopp M. Effects of treating traumatic brain injury with collagen scaffolds and human bone marrow stromal cells on sprouting of corticospinal tract axons into the denervated side of the spinal cord. J Neurosurg 2012. [PMID: 23198801 DOI: 10.3171/2012.11.jns12753] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT This study was designed to investigate how transplantation into injured brain of human bone marrow stromal cells (hMSCs) impregnated in collagen scaffolds affects axonal sprouting in the spinal cord after traumatic brain injury (TBI) in rats. Also investigated was the relationship of axonal sprouting to sensorimotor functional recovery after treatment. METHODS Adult male Wistar rats (n = 24) underwent a controlled cortical impact injury and were divided into three equal groups (8 rats/group). The two treatment groups received either hMSCs (3 × 10(6)) alone or hMSC (3 × 10(6))-impregnated collagen scaffolds transplanted into the lesion cavity. In the control group, saline was injected into the lesion cavity. All treatments were performed 7 days after TBI. On Day 21 after TBI, a 10% solution of biotinylated dextran amine (10,000 MW) was stereotactically injected into the contralateral motor cortex to label the corticospinal tract (CST) originating from this area. Sensorimotor function was tested using the modified neurological severity score (mNSS) and foot-fault tests performed on Days 1, 7, 14, 21, 28, and 35 after TBI. Spatial learning was tested with Morris water maze test on Days 31-35 after TBI. All rats were sacrificed on Day 35 after TBI, and brain and spinal cord (cervical and lumbar) sections were stained immunohistochemically for histological analysis. RESULTS Few biotinylated dextran amine-labeled CST fibers crossing over the midline were found in the contralateral spinal cord transverse sections at both cervical and lumbar levels in saline-treated (control) rats. However, hMSC-alone treatment significantly increased axonal sprouting from the intact CST into the denervated side of the gray matter of both cervical and lumbar levels of the spinal cord (p < 0.05). Also, this axonal sprouting was significantly more in the scaffold+hMSC group compared with the hMSC-alone group (p < 0.05). Sensorimotor functional analysis showed significant improvement of mNSS (p < 0.05) and foot-fault tests (p < 0.05) in hMSC-alone and scaffold+hMSC-treated rats compared with controls (p < 0.05). Functional improvement, however, was significantly greater in the scaffold+hMSC group compared with the hMSC-alone group (p < 0.05). Morris water maze testing also showed significant improvement in spatial learning in scaffold+hMSC and hMSC-alone groups compared with the control group (p < 0.05), with rats in the scaffold+hMSC group performing significantly better than those in the hMSC-alone group (p < 0.05). Pearson correlation data showed significant correlation between the number of crossing CST fibers detected and sensorimotor recovery (p < 0.05). CONCLUSIONS Axonal plasticity plays an important role in neurorestoration after TBI. Transplanting hMSCs with scaffolds enhances the effect of hMSCs on axonal sprouting of CST fibers from the contralateral intact cortex into the denervated side of spinal cord after TBI. This enhanced axonal regeneration may at least partially contribute to the therapeutic benefits of treating TBI with hMSCs.
Collapse
Affiliation(s)
- Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan 48202, USA.
| | | | | | | | | |
Collapse
|
20
|
Schoch KM, Madathil SK, Saatman KE. Genetic manipulation of cell death and neuroplasticity pathways in traumatic brain injury. Neurotherapeutics 2012; 9:323-37. [PMID: 22362424 PMCID: PMC3337028 DOI: 10.1007/s13311-012-0107-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of secondary neurodegenerative mechanisms contributing to cell dysfunction and necrotic and apoptotic cell death. The injured brain responds by activating endogenous reparative processes to counter the neurodegeneration or remodel the brain to enhance functional recovery. A vast array of genetically altered mice provide a unique opportunity to target single genes or proteins to better understand their role in cell death and endogenous repair after TBI. Among the earliest targets for transgenic and knockout studies in TBI have been programmed cell death mediators, such as the Bcl-2 family of proteins, caspases, and caspase-independent pathways. In addition, the role of cell cycle regulatory elements in the posttraumatic cell death pathway has been explored in mouse models. As interest grows in neuroplasticity in TBI, the use of transgenic and knockout mice in studies focused on gliogenesis, neurogenesis, and the balance of growth-promoting and growth-inhibiting molecules has increased in recent years. With proper consideration of potential effects of constitutive gene alteration, traditional transgenic and knockout models can provide valuable insights into TBI pathobiology. Through increasing sophistication of conditional and cell-type specific genetic manipulations, TBI studies in genetically altered mice will be increasingly useful for identification and validation of novel therapeutic targets.
Collapse
Affiliation(s)
- Kathleen M. Schoch
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Sindhu K. Madathil
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| |
Collapse
|
21
|
VanGuilder HD, Bixler GV, Sonntag WE, Freeman WM. Hippocampal expression of myelin-associated inhibitors is induced with age-related cognitive decline and correlates with deficits of spatial learning and memory. J Neurochem 2012; 121:77-98. [PMID: 22269040 PMCID: PMC3341628 DOI: 10.1111/j.1471-4159.2012.07671.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Impairment of cognitive functions including hippocampus-dependent spatial learning and memory affects nearly half of the aged population. Age-related cognitive decline is associated with synaptic dysfunction that occurs in the absence of neuronal cell loss, suggesting that impaired neuronal signaling and plasticity may underlie age-related deficits of cognitive function. Expression of myelin-associated inhibitors (MAIs) of synaptic plasticity, including the ligands myelin-associated glycoprotein, neurite outgrowth inhibitor A, and oligodendrocyte myelin glycoprotein, and their common receptor, Nogo-66 receptor, was examined in hippocampal synaptosomes and Cornu ammonis area (CA)1, CA3 and dentate gyrus subregions derived from adult (12-13 months) and aged (26-28 months) Fischer 344 × Brown Norway rats. Rats were behaviorally phenotyped by Morris water maze testing and classified as aged cognitively intact (n = 7-8) or aged cognitively impaired (n = 7-10) relative to adults (n = 5-7). MAI protein expression was induced in cognitively impaired, but not cognitively intact, aged rats and correlated with cognitive performance in individual rats. Immunohistochemical experiments demonstrated that up-regulation of MAIs occurs, in part, in hippocampal neuronal axons and somata. While a number of pathways and processes are altered with brain aging, we report a coordinated induction of myelin-associated inhibitors of functional and structural plasticity only in cognitively impaired aged rats. Induction of MAIs may decrease stimulus-induced synaptic strengthening and structural remodeling, ultimately impairing synaptic mechanisms of spatial learning and memory and resulting in cognitive decline.
Collapse
Affiliation(s)
- Heather D. VanGuilder
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| | - Georgina V. Bixler
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| | - William E. Sonntag
- Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Science Center, 975 NE 10th Street, BRC-1303, Oklahoma City OK 73104 USA
| | - Willard M. Freeman
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| |
Collapse
|
22
|
Ueno M, Hayano Y, Nakagawa H, Yamashita T. Intraspinal rewiring of the corticospinal tract requires target-derived brain-derived neurotrophic factor and compensates lost function after brain injury. Brain 2012; 135:1253-67. [DOI: 10.1093/brain/aws053] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
23
|
Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here? Br J Pharmacol 2011; 164:1207-29. [PMID: 21175576 PMCID: PMC3229758 DOI: 10.1111/j.1476-5381.2010.01163.x] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in young adults. Survivors of TBI frequently suffer from long-term personality changes and deficits in cognitive and motor performance, urgently calling for novel pharmacological treatment options. To date, all clinical trials evaluating neuroprotective compounds have failed in demonstrating clinical efficacy in cohorts of severely injured TBI patients. The purpose of the present review is to describe the utility of animal models of TBI for preclinical evaluation of pharmacological compounds. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. To successfully develop compounds for clinical TBI, a thorough evaluation in several TBI models and injury severities is crucial. Additionally, brain pharmacokinetics and the time window must be carefully evaluated. Although the search for a single-compound, 'silver bullet' therapy is ongoing, a combination of drugs targeting various aspects of neuroprotection, neuroinflammation and regeneration may be needed. In summary, finding drugs and prove clinical efficacy in TBI is a major challenge ahead for the research community and the drug industry. For a successful translation of basic science knowledge to the clinic to occur we believe that a further refinement of animal models and functional outcome methods is important. In the clinical setting, improved patient classification, more homogenous patient cohorts in clinical trials, standardized treatment strategies, improved central nervous system drug delivery systems and monitoring of target drug levels and drug effects is warranted.
Collapse
Affiliation(s)
- Niklas Marklund
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
24
|
Nogo-A expression in the brain of mice with cerebral malaria. PLoS One 2011; 6:e25728. [PMID: 21980529 PMCID: PMC3183069 DOI: 10.1371/journal.pone.0025728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 09/09/2011] [Indexed: 02/07/2023] Open
Abstract
Cerebral malaria (CM) is associated with a high rate of transient or persistent neurological sequelae. Nogo-A, a protein that is highly expressed in the endoplasmic reticulum (ER) of the mammalian central nervous system (CNS), is involved in neuronal regeneration and synaptic plasticity in the injured CNS. The current study investigates the role of Nogo-A in the course of experimental CM. C57BL/6J mice were infected with Plasmodium berghei ANKA blood stages. Brain homogenates of mice with different clinical severity levels of CM, infected animals without CM and control animals were analyzed for Nogo-A up-regulation by Western blotting and immunohistochemistry. Brain regions with Nogo-A upregulation were evaluated by transmission electron microscopy. Densitometric analysis of Western blots yielded a statistically significant upregulation of Nogo-A in mice showing moderate to severe CM. The number of neurons and oligodendrocytes positive for Nogo-A did not differ significantly between the studied groups. However, mice with severe CM showed a significantly higher number of cells with intense Nogo-A staining in the brain stem. In this region ultrastructural alterations of the ER were regularly observed. Nogo-A is upregulated during the early course of experimental CM. In the brain stem of severely affected animals increased Nogo-A expression and ultrastructural changes of the ER were observed. These data indicate a role of Nogo-A in neuronal stress response during experimental CM.
Collapse
|
25
|
Budde MD, Janes L, Gold E, Turtzo LC, Frank JA. The contribution of gliosis to diffusion tensor anisotropy and tractography following traumatic brain injury: validation in the rat using Fourier analysis of stained tissue sections. ACTA ACUST UNITED AC 2011; 134:2248-60. [PMID: 21764818 DOI: 10.1093/brain/awr161] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diffusion tensor imaging is highly sensitive to the microstructural integrity of the brain and has uncovered significant abnormalities following traumatic brain injury not appreciated through other methods. It is hoped that this increased sensitivity will aid in the detection and prognostication in patients with traumatic injury. However, the pathological substrates of such changes are poorly understood. Specifically, decreases in fractional anisotropy derived from diffusion tensor imaging are consistent with axonal injury, myelin injury or both in white matter fibres. In contrast, in both humans and animal models, increases in fractional anisotropy have been suggested to reflect axonal regeneration and plasticity, but the direct histological evidence for such changes remains tenuous. We developed a method to quantify the anisotropy of stained histological sections using Fourier analysis, and applied the method to a rat controlled cortical impact model to identify the specific pathological features that give rise to the diffusion tensor imaging changes in subacute to chronic traumatic brain injury. A multiple linear regression was performed to relate the histological measurements to the measured diffusion tensor changes. The results show that anisotropy was significantly increased (P < 0.001) in the perilesioned cortex following injury. Cortical anisotropy was independently associated (standardized β = 0.62, P = 0.04) with the coherent organization of reactive astrocytes (i.e. gliosis) and was not attributed to axons. By comparison, a decrease in white matter anisotropy (P < 0.001) was significantly related to demyelination (β = 0.75, P = 0.0015) and to a lesser extent, axonal degeneration (β = -0.48, P = 0.043). Gliosis within the lesioned cortex also influenced diffusion tensor tractography, highlighting the fact that spurious tracts in the injured brain may not necessarily reflect continuous axons and may instead depict glial scarring. The current study demonstrates a novel method to relate pathology to diffusion tensor imaging findings, elucidates the underlying mechanisms of anisotropy changes following traumatic brain injury and significantly impacts the clinical interpretation of diffusion tensor imaging findings in the injured brain.
Collapse
|
26
|
VanGuilder HD, Farley JA, Yan H, Van Kirk CA, Mitschelen M, Sonntag WE, Freeman WM. Hippocampal dysregulation of synaptic plasticity-associated proteins with age-related cognitive decline. Neurobiol Dis 2011; 43:201-12. [PMID: 21440628 PMCID: PMC3096728 DOI: 10.1016/j.nbd.2011.03.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 03/18/2011] [Indexed: 12/16/2022] Open
Abstract
Age-related cognitive decline occurs without frank neurodegeneration and is the most common cause of memory impairment in aging individuals. With increasing longevity, cognitive deficits, especially in hippocampus-dependent memory processes, are increasing in prevalence. Nevertheless, the neurobiological basis of age-related cognitive decline remains unknown. While concerted efforts have led to the identification of neurobiological changes with aging, few age-related alterations have been definitively correlated to behavioral measures of cognitive decline. In this work, adult (12 months) and aged (28 months) rats were categorized by Morris water maze performance as Adult cognitively Intact, Aged cognitively Intact or Aged cognitively Impaired, and protein expression was examined in hippocampal synaptosome preparations. Previously described differences in synaptic expression of neurotransmission-associated proteins (Dnm1, Hpca, Stx1, Syn1, Syn2, Syp, SNAP25, VAMP2 and 14-3-3 eta, gamma, and zeta) were confirmed between Adult and Aged rats, with no further dysregulation associated with cognitive impairment. Proteins related to synaptic structural stability (MAP2, drebrin, Nogo-A) and activity-dependent signaling (PSD-95, 14-3-3θ, CaMKIIα) were up- and down-regulated, respectively, with cognitive impairment but were not altered with increasing age. Localization of MAP2, PSD-95, and CaMKIIα demonstrated protein expression alterations throughout the hippocampus. The altered expression of activity- and structural stability-associated proteins suggests that impaired synaptic plasticity is a distinct phenomenon that occurs with age-related cognitive decline, and demonstrates that cognitive decline is not simply an exacerbation of the aging phenotype.
Collapse
Affiliation(s)
- Heather D. VanGuilder
- Penn State College of Medicine, Department of Pharmacology, Hershey Center for Applied Research, 500 University Drive, Hershey, PA, 17033, USA
| | - Julie A. Farley
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10 Street, BRC-1303, Oklahoma City, OK, 73104, USA
| | - Han Yan
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10 Street, BRC-1303, Oklahoma City, OK, 73104, USA
| | - Colleen A. Van Kirk
- Penn State College of Medicine, Department of Pharmacology, Hershey Center for Applied Research, 500 University Drive, Hershey, PA, 17033, USA
| | - Matthew Mitschelen
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10 Street, BRC-1303, Oklahoma City, OK, 73104, USA
| | - William E. Sonntag
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10 Street, BRC-1303, Oklahoma City, OK, 73104, USA
| | - Willard M. Freeman
- Penn State College of Medicine, Department of Pharmacology, Hershey Center for Applied Research, 500 University Drive, Hershey, PA, 17033, USA
| |
Collapse
|
27
|
Axonal remodeling for motor recovery after traumatic brain injury requires downregulation of γ-aminobutyric acid signaling. Cell Death Dis 2011; 2:e133. [PMID: 21412279 PMCID: PMC3101813 DOI: 10.1038/cddis.2011.16] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Remodeling of the remnant neuronal network after brain injury possibly mediates spontaneous functional recovery; however, the mechanisms inducing axonal remodeling during spontaneous recovery remain unclear. Here, we show that altered γ-aminobutyric acid (GABA) signaling is crucial for axonal remodeling of the contralesional cortex after traumatic brain injury. After injury to the sensorimotor cortex in mice, we found a significant decrease in the expression of GABAAR-α1 subunits in the intact sensorimotor cortex for 2 weeks. Motor functions, assessed by grid walk and cylinder tests, spontaneously improved in 4 weeks after the injury to the sensorimotor cortex. With motor recovery, corticospinal tract (CST) axons from the contralesional cortex sprouted into the denervated side of the cervical spinal cord at 2 and 4 weeks after the injury. To determine the functional implications of the changes in the expression of GABAAR-α1 subunits, we infused muscimol, a GABA R agonist, into the contralesional cortex for a week after the injury. Compared with the vehicle-treated mice, we noted significantly inhibited recovery in the muscimol-treated mice. Further, muscimol infusion greatly suppressed the axonal sprouting into the denervated side of the cervical spinal cord. In conclusion, recovery of motor function and axonal remodeling of the CST following cortical injury requires suppressed GABAAR subunit expression and decreased GABAergic signaling.
Collapse
|
28
|
Zhang Y, Xiong Y, Mahmood A, Meng Y, Liu Z, Qu C, Chopp M. Sprouting of corticospinal tract axons from the contralateral hemisphere into the denervated side of the spinal cord is associated with functional recovery in adult rat after traumatic brain injury and erythropoietin treatment. Brain Res 2010; 1353:249-57. [PMID: 20654589 PMCID: PMC2933297 DOI: 10.1016/j.brainres.2010.07.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/12/2010] [Accepted: 07/14/2010] [Indexed: 12/24/2022]
Abstract
Erythropoietin (EPO) promotes functional recovery after traumatic brain injury (TBI). This study was designed to investigate whether EPO treatment promotes contralateral corticospinal tract (CST) plasticity in the spinal cord in rats after TBI. Biotinylated dextran amine (BDA) was injected into the right sensorimotor cortex to anterogradely label the CST. TBI was induced by controlled cortical impact over the left parietal cortex immediately after BDA injections. EPO (5000 U/kg) or saline was administered intraperitoneally at Days 1, 2, and 3 post-injury. Neurological function was assessed using a modified neurological severity score (mNSS) and footfault tests. Animals were sacrificed 35 days after injury and brain sections stained for histological analysis. Compared to the saline treatment, EPO treatment significantly improved sensorimotor functional outcome (lower mNSS and reduced footfaults) from Days 7 to 35 post-injury. TBI alone significantly stimulated contralateral CST axon sprouting toward the denervated gray matter of the cervical and lumbar spinal cord; however, EPO treatment further significantly increased the axon sprouting in TBI rats although EPO treatment did not significantly affect axon sprouting in sham animals. The contralesional CST sprouting was highly and positively correlated with sensorimotor recovery after TBI. These data demonstrate that CST fibers originating from the contralesional intact cerebral hemisphere are capable of sprouting into the denervated spinal cord after TBI and EPO treatment, which may at least partially contribute to functional recovery.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202
| | - Yuling Meng
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202
| | - Changsheng Qu
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202
- Department of Physics, Oakland University, Rochester, MI 48309
| |
Collapse
|
29
|
Hånell A, Clausen F, Björk M, Jansson K, Philipson O, Nilsson LN, Hillered L, Weinreb PH, Lee D, McIntosh TK, Gimbel DA, Strittmatter SM, Marklund N. Genetic deletion and pharmacological inhibition of Nogo-66 receptor impairs cognitive outcome after traumatic brain injury in mice. J Neurotrauma 2010; 27:1297-309. [PMID: 20486800 PMCID: PMC2942864 DOI: 10.1089/neu.2009.1255] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Functional recovery is markedly restricted following traumatic brain injury (TBI), partly due to myelin-associated inhibitors including Nogo-A, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), that all bind to the Nogo-66 receptor-1 (NgR1). In previous studies, pharmacological neutralization of both Nogo-A and MAG improved outcome following TBI in the rat, and neutralization of NgR1 improved outcome following spinal cord injury and stroke in rodent models. However, the behavioral and histological effects of NgR1 inhibition have not previously been evaluated in TBI. We hypothesized that NgR1 negatively influences behavioral recovery following TBI, and evaluated NgR1(-/-) mice (NgR1(-/-) study) and, in a separate study, soluble NgR1 infused intracerebroventricularly immediately post-injury to neutralize NgR1 (sNgR1 study) following TBI in mice using a controlled cortical impact (CCI) injury model. In both studies, motor function, TBI-induced loss of tissue, and hippocampal beta-amyloid immunohistochemistry were not altered up to 5 weeks post-injury. Surprisingly, cognitive function (as evaluated with the Morris water maze at 4 weeks post-injury) was significantly impaired both in NgR1(-/-) mice and in mice treated with soluble NgR1. In the sNgR1 study, we evaluated hippocampal mossy fiber sprouting using the Timm stain and found it to be increased at 5 weeks following TBI. Neutralization of NgR1 significantly increased mossy fiber sprouting in sham-injured animals, but not in brain-injured animals. Our data suggest a complex role for myelin-associated inhibitors in the behavioral recovery process following TBI, and urge caution when inhibiting NgR1 in the early post-injury period.
Collapse
Affiliation(s)
- Anders Hånell
- Department of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Department of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Maria Björk
- Department of Neurosurgery, Uppsala University, Uppsala, Sweden
| | | | - Ola Philipson
- Department of Public Health and Caring Science, Uppsala University, Uppsala, Sweden
| | - Lars N.G. Nilsson
- Department of Public Health and Caring Science, Uppsala University, Uppsala, Sweden
| | - Lars Hillered
- Department of Neurosurgery, Uppsala University, Uppsala, Sweden
| | | | | | - Tracy K. McIntosh
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A. Gimbel
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neurology, Yale University School of Medicine, New Haven, Connecticut
| | - Niklas Marklund
- Department of Neurosurgery, Uppsala University, Uppsala, Sweden
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Richardson RM, Singh A, Sun D, Fillmore HL, Dietrich DW, Bullock MR. Stem cell biology in traumatic brain injury: effects of injury and strategies for repair. J Neurosurg 2010; 112:1125-38. [PMID: 19499984 DOI: 10.3171/2009.4.jns081087] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Approximately 350,000 individuals in the US are affected annually by severe and moderate traumatic brain injuries (TBI) that may result in long-term disability. This rate of injury has produced approximately 3.3 million disabled survivors in the US alone. There is currently no specific treatment available for TBI other than supportive care, but aggressive prehospital resuscitation, rapid triage, and intensive care have reduced mortality rates. With the recent demonstration that neurogenesis occurs in all mammals (including man) throughout adult life, albeit at a low rate, the concept of replacing neurons lost after TBI is now becoming a reality. Experimental rodent models have shown that neurogenesis is accelerated after TBI, especially in juveniles. Two approaches have been followed in these rodent models to test possible therapeutic approaches that could enhance neuronal replacement in humans after TBI. The first has been to define and quantify the phenomenon of de novo hippocampal and cortical neurogenesis after TBI and find ways to enhance this (for example by exogenous trophic factor administration). A second approach has been the transplantation of different types of neural progenitor cells after TBI. In this review the authors discuss some of the processes that follow after acute TBI including the changes in the brain microenvironment and the role of trophic factor dynamics with regard to the effects on endogenous neurogenesis and gliagenesis. The authors also discuss strategies to clinically harness the factors influencing these processes and repair strategies using exogenous neural progenitor cell transplantation. Each strategy is discussed with an emphasis on highlighting the progress and limiting factors relevant to the development of clinical trials of cellular replacement therapy for severe TBI in humans.
Collapse
Affiliation(s)
- R Mark Richardson
- Department of Neurological Surgery, University of California San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
31
|
Gillani RL, Tsai SY, Wallace DG, O'Brien TE, Arhebamen E, Tole M, Schwab ME, Kartje GL. Cognitive recovery in the aged rat after stroke and anti-Nogo-A immunotherapy. Behav Brain Res 2010; 208:415-24. [PMID: 20035795 PMCID: PMC2831114 DOI: 10.1016/j.bbr.2009.12.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 12/08/2009] [Accepted: 12/12/2009] [Indexed: 01/09/2023]
Abstract
We have previously shown that immunotherapy directed against the protein Nogo-A leads to recovery on a skilled forelimb reaching task in rats after sensorimotor cortex stroke, which correlated with axonal and dendritic plasticity. Here we investigated anti-Nogo-A immunotherapy as an intervention to improve performance on a spatial memory task in aged rats after stroke, and whether cognitive recovery was correlated with structural plasticity. Aged rats underwent a unilateral distal permanent middle cerebral artery occlusion and one week later were treated with an anti-Nogo-A or control antibody. Nine weeks post-stroke, treated rats and normal aged rats were tested on the Morris water maze task. Following testing rats were sacrificed and brains processed for the Golgi-Cox method. Hippocampal CA3 and CA1 pyramidal and dentate gyrus granule cells were examined for dendritic length and number of branch segments, and CA3 and CA1 pyramidal cells were examined for spine density and morphology. Anti-Nogo-A immunotherapy given one week following stroke in aged rats improved performance on the reference memory portion of the Morris water maze task. However, this improved performance was not correlated with structural changes in the hippocampal neurons examined. Our finding of improved performance on the Morris water maze in aged rats after stroke and treatment with anti-Nogo-A immunotherapy demonstrates the promising therapeutic potential for anti-Nogo-A immunotherapy to treat cognitive deficits after stroke. The identification of sites of axonal and dendritic plasticity in the aged brain after stroke and treatment with anti-Nogo-A immunotherapy is still under investigation.
Collapse
Affiliation(s)
- Rebecca L Gillani
- Neuroscience Program, Loyola University Chicago, Maywood, IL 60153, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Marklund N, Morales D, Clausen F, Hånell A, Kiwanuka O, Pitkänen A, Gimbel DA, Philipson O, Lannfelt L, Hillered L, Strittmatter SM, McIntosh TK. Functional outcome is impaired following traumatic brain injury in aging Nogo-A/B-deficient mice. Neuroscience 2009; 163:540-51. [PMID: 19555742 PMCID: PMC2756649 DOI: 10.1016/j.neuroscience.2009.06.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 06/15/2009] [Accepted: 06/16/2009] [Indexed: 12/22/2022]
Abstract
Increasing age is associated with a poor prognosis following traumatic brain injury (TBI). CNS axons may recover poorly following TBI due to expression of myelin-derived inhibitors to axonal outgrowth such as Nogo-A. To study the role of Nogo-A/B in the pathophysiological response of the elderly to TBI, 1-year-old mice deficient in Nogo-A/B (Nogo-A/B homozygous(-/-) mice), Nogo-A/B heterozygous(-/+) mice, and age-matched wild-type (WT) littermate controls were subjected to a controlled cortical impact (CCI) TBI. Sham-injured WT mice (7 months old) and 12 month old naïve Nogo-A/B(-/-) and Nogo-A/B(-/+) served as controls. Neurological motor function was evaluated up to 3 weeks, and cognitive function, hemispheric tissue loss, myelin staining and hippocampal beta-amyloid (A beta) immunohistochemistry were evaluated at 4 weeks post-injury. In WT littermates, TBI significantly impaired learning ability at 4 weeks and neurological motor function up to 2 weeks post-injury and caused a significant loss of hemispheric tissue. Following TBI, Nogo-A/B(-/-) mice showed significantly less recovery from neurological motor and cognitive deficits compared to brain-injured WT mice. Naïve Nogo-A/B(-/-) and Nogo-A/B(-/+) mice quickly learned the MWM task in contrast to brain-injured Nogo-A/B(-/-) mice who failed to learn the MWM task at 4 weeks post-injury. Hemispheric tissue loss and cortical lesion volume were similar among the brain-injured genotypes. Neither TBI nor the absence of NogoA/B caused an increased A beta expression. Myelin staining showed a reduced area and density in the corpus callosum in brain-injured Nogo-A/B(-/-) animals compared to their littermate controls. These novel and unexpected behavioral results demonstrate that the absence of Nogo-A/B may negatively influence outcome, possibly related to hypomyelination, following TBI in mice and suggest a complex role for this myelin-associated axonal growth inhibitor following TBI.
Collapse
Affiliation(s)
- N Marklund
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mueller BK, Mueller R, Schoemaker H. Stimulating neuroregeneration as a therapeutic drug approach for traumatic brain injury. Br J Pharmacol 2009; 157:675-85. [PMID: 19422372 DOI: 10.1111/j.1476-5381.2009.00220.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury, a silent epidemic of modern societies, is a largely neglected area in drug development and no drug is currently available for the treatment of patients suffering from brain trauma. Despite this grim situation, much progress has been made over the last two decades in closely related medical indications, such as spinal cord injury, giving rise to a more optimistic approach to drug development in brain trauma. Fundamental insights have been gained with animal models of central nervous system (CNS) trauma and spinal cord injury. Neuroregenerative drug candidates have been identified and two of these have progressed to clinical development for spinal cord injury patients. If successful, these drug candidates may be used to treat brain trauma patients. Significant progress has also been made in understanding the fundamental molecular mechanism underlying irreversible axonal growth arrest in the injured CNS of higher mammals. From these studies, we have learned that the axonal retraction bulb, previously regarded as a marker for failure of regenerative growth, is not static but dynamic and, therefore, amenable to pharmacotherapeutic approaches. With the development of modified magnetic resonance imaging methods, fibre tracts can be visualised in the living human brain and such imaging methods will soon be used to evaluate the neuroregenerative potential of drug candidates. These significant advances are expected to fundamentally change the often hopeless situation of brain trauma patients and will be the first step towards overcoming the silent epidemic of brain injury.
Collapse
Affiliation(s)
- Bernhard K Mueller
- Neuroscience Research, Abbott GmbH and Company KG, Ludwigshafen, Germany.
| | | | | |
Collapse
|
34
|
Hou ST, Jiang SX, Smith RA. Permissive and repulsive cues and signalling pathways of axonal outgrowth and regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2008; 267:125-81. [PMID: 18544498 DOI: 10.1016/s1937-6448(08)00603-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Successful axonal outgrowth in the adult central nervous system (CNS) is central to the process of nerve regeneration and brain repair. To date, much of the knowledge on axonal guidance and outgrowth comes from studies on neuritogenesis and patterning during development where distal growth cones constantly sample the local environment and respond to specific physical and trophic influences. Opposing permissive (e.g., growth factors) and hostile signals (e.g., repulsive cues) are processed, leading to growth cone remodelling, and a concomitant restructuring of the cytoskeleton, thereby permitting pioneering extension and a potential for establishing synaptic connections. Repulsive cues, such as semaphorins, ephrins and myelin-secreted inhibitory glycoproteins, act through their respective receptors to affect the collapsing or turning of growth cones via several pathways, such as the Rho GTPases signalling which precipitates the cytoskeletal changes. One of the direct modulators of microtubules is the family of brain-specific proteins, collapsin response mediator protein (CRMP). Exciting evidence emerged recently that cleavage of CRMPs in response to injury-activated proteases, such as calpain, signals axonal retraction and neuronal death in adult post-mitotic neurons, while blocking this signal transduction prevents axonal retraction and death following excitotoxic insult and cerebral ischemia. Regeneration is minimal in injured postnatal CNS, albeit the occurrence of some limited remodelling in areas where synaptic plasticity is prevalent. Frequently in the absence of axonal regeneration, there is not only an inevitable loss of functional connections, but also a loss of neurons, such as through the actions of dependence receptors. Deciphering the cues and signalling pathways of axonal guidance and outgrowth may hold the key to fully understanding nerve regeneration and brain repair, thereby opening the way for developing potential therapeutics.
Collapse
Affiliation(s)
- Sheng T Hou
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, K1A 0R6, Canada
| | | | | |
Collapse
|
35
|
Chytrova G, Ying Z, Gomez-Pinilla F. Exercise normalizes levels of MAG and Nogo-A growth inhibitors after brain trauma. Eur J Neurosci 2007; 27:1-11. [DOI: 10.1111/j.1460-9568.2007.05982.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
Marklund N, Bareyre FM, Royo NC, Thompson HJ, Mir AK, Grady MS, Schwab ME, McIntosh TK. Cognitive outcome following brain injury and treatment with an inhibitor of Nogo-A in association with an attenuated downregulation of hippocampal growth-associated protein-43 expression. J Neurosurg 2007; 107:844-53. [PMID: 17937233 PMCID: PMC2366808 DOI: 10.3171/jns-07/10/0844] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECT Central nervous system axons regenerate poorly after traumatic brain injury (TBI), partly due to inhibitors such as the protein Nogo-A present in myelin. The authors evaluated the efficacy of anti-Nogo-A monoclonal antibody (mAb) 7B12 administration on the neurobehavioral and cognitive outcome of rats following lateral fluid-percussion brain injury, characterized the penetration of the 7B12 or control antibodies into target brain regions, and evaluated the effects of Nogo-A inhibition on hemispheric tissue loss and sprouting of uninjured motor tracts in the cervical cord. To elucidate a potential molecular response to Nogo-A inhibition, we evaluated the effects of 7B12 on hippocampal GAP-43 expression. METHODS Beginning 24 hours after lateral fluid-percussion brain injury or sham injury in rats, the mAb 7B12 or control antibody was infused intracerebroventricularly over 14 days, and behavior was assessed over 4 weeks. RESULTS Immunoreactivity for 7B12 or immunoglobulin G was detected in widespread brain regions at 1 and 3 weeks postinjury. The brain-injured animals treated with 7B12 showed improvement in cognitive function (p < 0.05) at 4 weeks but no improvement in neurological motor function from 1 to 4 weeks postinjury compared with brain-injured, vehicle-treated controls. The enhanced cognitive function following inhibition of Nogo-A was correlated with an attenuated postinjury downregulation of hippocampal GAP-43 expression (p < 0.05). CONCLUSIONS Increased GAP-43 expression may be a novel molecular mechanism of the enhanced cognitive recovery mediated by Nogo-A inhibition after TBI in rats.
Collapse
Affiliation(s)
- Niklas Marklund
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Royo NC, LeBold D, Magge SN, Chen I, Hauspurg A, Cohen AS, Watson DJ. Neurotrophin-mediated neuroprotection of hippocampal neurons following traumatic brain injury is not associated with acute recovery of hippocampal function. Neuroscience 2007; 148:359-70. [PMID: 17681695 PMCID: PMC2579330 DOI: 10.1016/j.neuroscience.2007.06.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 06/12/2007] [Accepted: 06/18/2007] [Indexed: 11/23/2022]
Abstract
Traumatic brain injury (TBI) causes selective hippocampal cell death which is believed to be associated with the cognitive impairment observed in both clinical and experimental settings. The endogenous neurotrophin-4/5 (NT-4/5), a TrkB ligand, has been shown to be neuroprotective for vulnerable CA3 pyramidal neurons after experimental brain injury. In this study, infusion of recombinant NT-4/5 increased survival of CA2/3 pyramidal neurons to 71% after lateral fluid percussion brain injury in rats, compared with 55% in vehicle-treated controls. The functional outcome of this NT-4/5-mediated neuroprotection was examined using three hippocampal-dependent behavioral tests. Injury-induced impairment was evident in all three tests, but interestingly, there was no treatment-related improvement in any of these measures. Similarly, injury-induced decreased excitability in the Schaffer collaterals was not affected by NT-4/5 treatment. We propose that a deeper understanding of the factors that link neuronal survival to recovery of function will be important for future studies of potentially therapeutic agents.
Collapse
Affiliation(s)
- N C Royo
- Department of Neurosurgery, 371A Stemmler Hall/6071, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The concept of brain plasticity covers all the mechanisms involved in the capacity of the brain to adjust and remodel itself in response to environmental requirements, experience, skill acquisition, and new challenges including brain lesions. Advances in neuroimaging and neurophysiologic techniques have increased our knowledge of task-related changes in cortical representation areas in the intact and injured human brain. The recognition that neuronal progenitor cells proliferate and differentiate in the subventricular zone and dentate gyrus in the adult mammalian brain has raised the hope that regeneration may be possible after brain lesions. Regeneration will require that new cells differentiate, survive, and integrate into existing neural networks and that axons regenerate. To what extent this will be possible is difficult to predict. Current research explores the possibilities to modify endogenous neurogenesis and facilitate axonal regeneration using myelin inhibitory factors. After apoptotic damage in mice new cortical neurons can form long-distance connections. Progenitor cells from the subventricular zone migrate to cortical and subcortical regions after ischemic brain lesions, apparently directed by signals from the damaged region. Postmortem studies on human brains suggest that neurogenesis may be altered in degenerative diseases. Functional and anatomic data indicate that myelin inhibitory factors, cell implantation, and modification of extracellular matrix may be beneficial after spinal cord lesions. Neurophysiologic data demonstrating that new connections are functioning are needed to prove regeneration. Even if not achieving the goal, methods aimed at regeneration can be beneficial by enhancing plasticity in intact brain regions.
Collapse
Affiliation(s)
- Barbro B Johansson
- Wallenberg Neuroscience Center, Department of Clinical Neuroscience, Lund University, Lund, Sweden.
| |
Collapse
|
39
|
Brody DL, Mac Donald C, Kessens CC, Yuede C, Parsadanian M, Spinner M, Kim E, Schwetye KE, Holtzman DM, Bayly PV. Electromagnetic controlled cortical impact device for precise, graded experimental traumatic brain injury. J Neurotrauma 2007; 24:657-73. [PMID: 17439349 PMCID: PMC2435168 DOI: 10.1089/neu.2006.0011] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Genetically modified mice represent useful tools for traumatic brain injury (TBI) research and attractive preclinical models for the development of novel therapeutics. Experimental methods that minimize the number of mice needed may increase the pace of discovery. With this in mind, we developed and characterized a prototype electromagnetic (EM) controlled cortical impact device along with refined surgical and behavioral testing techniques. By varying the depth of impact between 1.0 and 3.0 mm, we found that the EM device was capable of producing a broad range of injury severities. Histologically, 2.0-mm impact depth injuries produced by the EM device were similar to 1.0-mm impact depth injuries produced by a commercially available pneumatic device. Behaviorally, 2.0-, 2.5-, and 3.0-mm impacts impaired hidden platform and probe trial water maze performance, whereas 1.5-mm impacts did not. Rotorod and visible platform water maze deficits were also found following 2.5- and 3.0-mm impacts. No impairment of conditioned fear performance was detected. No differences were found between sexes of mice. Inter-operator reliability was very good. Behaviorally, we found that we could statistically distinguish between injury depths differing by 0.5 mm using 12 mice per group and between injury depths differing by 1.0 mm with 7-8 mice per group. Thus, the EM impactor and refined surgical and behavioral testing techniques may offer a reliable and convenient framework for preclinical TBI research involving mice.
Collapse
Affiliation(s)
- David L Brody
- Department of Neurology, Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Thompson HJ, Marklund N, LeBold DG, Morales DM, Keck CA, Vinson M, Royo NC, Grundy R, McIntosh TK. Tissue sparing and functional recovery following experimental traumatic brain injury is provided by treatment with an anti-myelin-associated glycoprotein antibody. Eur J Neurosci 2006; 24:3063-72. [PMID: 17156367 PMCID: PMC2377452 DOI: 10.1111/j.1460-9568.2006.05197.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Axonal injury is a hallmark of traumatic brain injury (TBI) and is associated with a poor clinical outcome. Following central nervous system injury, axons regenerate poorly, in part due to the presence of molecules associated with myelin that inhibit axonal outgrowth, including myelin-associated glycoprotein (MAG). The involvement of MAG in neurobehavioral deficits and tissue loss following experimental TBI remains unexplored and was evaluated in the current study using an MAG-specific monoclonal antibody (mAb). Anesthetized rats (n=102) were subjected to either lateral fluid percussion brain injury (n=59) or sham injury (n=43). In surviving animals, beginning at 1 h post-injury, 8.64 microg anti-MAG mAb (n=33 injured, n=21 sham) or control IgG (n=26 injured, n=22 sham) was infused intracerebroventricularly for 72 h. One group of these rats (n=14 sham, n=11 injured) was killed at 72 h post-injury for verification of drug diffusion and MAG immunohistochemistry. All other animals were evaluated up to 8 weeks post-injury using tests for neurologic motor, sensory and cognitive function. Hemispheric tissue loss was also evaluated at 8 weeks post-injury. At 72 h post-injury, increased immunoreactivity for MAG was seen in the ipsilateral cortex, thalamus and hippocampus of brain-injured animals, and anti-MAG mAb was detectable in the hippocampus, fimbria and ventricles. Brain-injured animals receiving anti-MAG mAb showed significantly improved recovery of sensorimotor function at 6 and 8 weeks (P<0.01) post-injury when compared with brain-injured IgG-treated animals. Additionally, at 8 weeks post-injury, the anti-MAG mAb-treated brain-injured animals demonstrated significantly improved cognitive function and reduced hemispheric tissue loss (P<0.05) when compared with their brain-injured controls. These results indicate that MAG may contribute to the pathophysiology of experimental TBI and treatment strategies that target MAG may be suitable for further evaluation.
Collapse
Affiliation(s)
- Hilaire J Thompson
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, The University of Pennsylvania, Philadelphia, PA, USA, and Department of Neurosurgery, Uppsala University Hospital, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lippert-Gruener M, Maegele M, Garbe J, Angelov DN. Late effects of enriched environment (EE) plus multimodal early onset stimulation (MEOS) after traumatic brain injury in rats: Ongoing improvement of neuromotor function despite sustained volume of the CNS lesion. Exp Neurol 2006; 203:82-94. [PMID: 16965773 DOI: 10.1016/j.expneurol.2006.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 07/05/2006] [Accepted: 07/24/2006] [Indexed: 11/30/2022]
Abstract
Recently we showed that the combination between MEOS and EE applied to rats for 7-15 days after traumatic brain injury (TBI) was associated with reduced CNS lesion volume and enhanced reversal of neuromotor dysfunction. In a continuation of this work, we tested whether these effects persisted for longer post-operative periods, e.g. 30 days post-injury (dpi). Rats were subjected to lateral fluid percussion (LFP) or to sham injury. After LFP, one third of the animals (injured and sham) was placed under conditions of standard housing (SH), one third was kept in EE-only, and one third received EE+MEOS. Standardized composite neuroscore (NS) for neurological functions and computerized analysis of the vibrissal motor performance were used to assess post-traumatic neuromotor deficits. These were followed by evaluation of the cortical lesion volume (CLV) after immunostaining for neuron-specific enolase, caspase 3 active, and GFAP. Finally, the volume of cortical lesion containing regeneration-associated proteins (CLV-RAP) was determined in sections stained for GAP-43, MAP2, and neuronal class III beta-tubulin. We found (i) no differences in the vibrissal motor performance; (ii) EE+MEOS rats performed significantly better than SH rats in NS; (iii) EE-only and EE+MEOS animals, but not SH rats, showed better recovery at 30 dpi than at 15 dpi; (iv) no differences among all groups in CLV (larger than that at 15 dpi) and CLV-RAP, despite a clear tendency to reduction in the EE-only and EE+MEOS rats. We conclude that EE+MEOS retards, but cannot prevent the increase of lesion volume. This retardation is sufficient for a continuous restoration of neurological functions.
Collapse
|
42
|
Lee DHS, Seamans KW. The Nogo66 receptor pathway and CNS axon regeneration: new hopes for treating CNS injuries and neurodegeneration. Expert Opin Ther Pat 2006. [DOI: 10.1517/13543776.16.8.1041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Marklund N, Fulp CT, Shimizu S, Puri R, McMillan A, Strittmatter SM, McIntosh TK. Selective temporal and regional alterations of Nogo-A and small proline-rich repeat protein 1A (SPRR1A) but not Nogo-66 receptor (NgR) occur following traumatic brain injury in the rat. Exp Neurol 2006; 197:70-83. [PMID: 16321384 PMCID: PMC2849132 DOI: 10.1016/j.expneurol.2005.08.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 07/26/2005] [Accepted: 08/11/2005] [Indexed: 01/12/2023]
Abstract
Axons show a poor regenerative capacity following traumatic central nervous system (CNS) injury, partly due to the expression of inhibitors of axonal outgrowth, of which Nogo-A is considered the most important. We evaluated the acute expression of Nogo-A, the Nogo-66 receptor (NgR) and the novel small proline-rich repeat protein 1A (SPRR1A, previously undetected in brain), following experimental lateral fluid percussion (FP) brain injury in rats. Immunofluorescence with antibodies against Nogo-A, NgR and SPRR1A was combined with antibodies against the neuronal markers NeuN and microtubule-associated protein (MAP)-2 and the oligodendrocyte marker RIP, while Western blot analysis was performed for Nogo-A and NgR. Brain injury produced a significant increase in Nogo-A expression in injured cortex, ipsilateral external capsule and reticular thalamus from days 1-7 post-injury (P < 0.05) compared to controls. Increased expression of Nogo-A was observed in both RIP- and NeuN positive (+) cells in the ipsilateral cortex, in NeuN (+) cells in the CA3 region of the hippocampus and reticular thalamus and in RIP (+) cells in white matter tracts. Alterations in NgR expression were not observed following traumatic brain injury (TBI). Brain injury increased the extent of SPRR1A expression in the ipsilateral cortex and the CA3 at all post-injury time-points in NeuN (+) cells. The marked increases in Nogo-A and SPRR1A in several important brain regions suggest that although inhibitors of axonal growth may be upregulated, the injured brain is also capable of expressing proteins promoting axonal outgrowth following TBI.
Collapse
Affiliation(s)
- Niklas Marklund
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Buchli AD, Schwab ME. Inhibition of Nogo: a key strategy to increase regeneration, plasticity and functional recovery of the lesioned central nervous system. Ann Med 2005; 37:556-67. [PMID: 16338758 DOI: 10.1080/07853890500407520] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In the adult central nervous system (CNS) myelin and oligodendrocytes, Nogo-A exerts a growth inhibitory function leading to restricted axonal regeneration. After development of different anti-Nogo-A antibodies and other Nogo-A blocking reagents their application has recently been studied in various in vivo animal models of spinal cord injury and stroke. These studies show that intracerebral application of Nogo-A-inactivating reagents leads to enhanced regeneration and compensatory sprouting, structural reorganization or plasticity, and functional recovery as seen in different behavioural analyses.
Collapse
Affiliation(s)
- Anita D Buchli
- Brain Research Institute, University of Zurich and Department of Biology, Swiss Federal Institute of Technology-Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | | |
Collapse
|