Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Neuroscience ; 163(2): 540-51, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19555742

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.


Subject(s)
Aging , Brain Injuries/physiopathology , Myelin Proteins/deficiency , Recovery of Function/physiology , Amyloid beta-Peptides/metabolism , Animals , Brain Injuries/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuropsychological Tests , Nogo Proteins , Organ Size , Random Allocation , Time Factors , Treatment Outcome
2.
Clin Neuropathol ; 25(6): 255-64, 2006.
Article in English | MEDLINE | ID: mdl-17140155

ABSTRACT

BACKGROUND: Brain injury after trauma is an important cause of mortality and morbidity in society. There is evidence in both man and laboratory animals that in addition to necrosis, cell loss may occur as a result of programmed cell death (PCD). The cellular and molecular responses after head injury are partly influenced by genetic polymorphisms of apolipoprotein E and the pro-inflammatory cytokine IL-I. AIM: The principal aim of this study was to determine whether the presence of the ApoE epsilon4, IL- 1 alpha2 or IL- 1beta2 allele types influenced the amounts of PCD after head injury compared with controls. METHODS: Paraffin sections from the hippocampus of 38 patients (32 M : 6 F, aged 15 - 75, mean 38 years, survival 7- 576 hours; mean 36 hours) who died after a head injury were stained by Tunel histochemistry and quantified, and genotyping was undertaken by PCR "blind" to clinical detail. RESULTS: There were more Tunel+ cells (neurons and glia) after head injury than in controls with statistically increased numbers in all sectors of the hippocampus including the dentate fascia. However, there was no correlation between ApoEepsilon4, IL- 1 alpha allele 2 and IL- 1beta allele 2 and the amount of Tunel positivity. CONCLUSION: Given that both the ApoE and IL-1 influence outcome after various forms of acute brain injury, further work will be required to determine the mechanism underlying this relationship.


Subject(s)
Apolipoprotein E4/genetics , Apoptosis/genetics , Brain Injuries/genetics , Genetic Predisposition to Disease/genetics , Interleukin-1/genetics , Nerve Degeneration/genetics , Adolescent , Adult , Aged , Biomarkers/metabolism , Brain Injuries/immunology , Brain Injuries/metabolism , Cell Count , DNA Mutational Analysis , Female , Gene Frequency , Genetic Testing , Genotype , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , In Situ Nick-End Labeling , Interleukin-1alpha/genetics , Interleukin-1beta/genetics , Male , Middle Aged , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Polymorphism, Genetic/genetics
3.
Neuroscience ; 140(2): 685-97, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16650603

ABSTRACT

Although traumatic brain injury is a major cause of symptomatic epilepsy, the mechanism by which it leads to recurrent seizures is unknown. An animal model of posttraumatic epilepsy that reliably reproduces the clinical sequelae of human traumatic brain injury is essential to identify the molecular and cellular substrates of posttraumatic epileptogenesis, and perform preclinical screening of new antiepileptogenic compounds. We studied the electrophysiologic, behavioral, and structural features of posttraumatic epilepsy induced by severe, non-penetrating lateral fluid-percussion brain injury in rats. Data from two independent experiments indicated that 43% to 50% of injured animals developed epilepsy, with a latency period between 7 weeks to 1 year. Mean seizure frequency was 0.3+/-0.2 seizures per day and mean seizure duration was 113+/-46 s. Behavioral seizure severity increased over time in the majority of animals. Secondarily-generalized seizures comprised an average of 66+/-37% of all seizures. Mossy fiber sprouting was increased in the ipsilateral hippocampus of animals with posttraumatic epilepsy compared with those subjected to traumatic brain injury without epilepsy. Stereologic cell counts indicated a loss of dentate hilar neurons ipsilaterally following traumatic brain injury. Our data suggest that posttraumatic epilepsy occurs with a frequency of 40% to 50% after severe non-penetrating fluid-percussion brain injury in rats, and that the lateral fluid percussion model can serve as a clinically-relevant tool for pathophysiologic and preclinical studies.


Subject(s)
Brain Concussion/complications , Brain Concussion/physiopathology , Brain/physiopathology , Epilepsy/etiology , Epilepsy/physiopathology , Animals , Apnea/etiology , Apnea/physiopathology , Brain/pathology , Brain Concussion/pathology , Cell Death/physiology , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Disease Models, Animal , Electroencephalography , Epilepsy/pathology , Growth Cones/pathology , Growth Cones/physiology , Male , Mossy Fibers, Hippocampal/pathology , Mossy Fibers, Hippocampal/physiopathology , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley , Time Factors
4.
Eur J Neurosci ; 23(5): 1089-102, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16553773

ABSTRACT

Traumatic brain injury (TBI) causes selective hippocampal cell death, which is believed to be associated with cognitive impairment observed both in clinical and experimental settings. Although neurotrophin administration has been tested as a strategy to prevent cell death following TBI, the potential neuroprotective role of neurotrophin-4/5 (NT-4/5) in TBI remains unknown. We hypothesized that NT-4/5 would offer neuroprotection for selectively vulnerable hippocampal neurons following TBI. Measurements of NT-4/5 in rats subjected to lateral fluid percussion (LFP) TBI revealed two-threefold increases in the injured cortex and hippocampus in the acute period (1-3 days) following brain injury. Subsequently, the response of NT-4/5 knockout (NT-4/5(-/-)) mice to controlled-cortical impact TBI was investigated. NT-4/5(-/-) mice were more susceptible to selective pyramidal cell loss in Ahmon's corn (CA) subfields of the hippocampus following TBI, and showed impaired motor recovery when compared with their brain-injured wild-type controls (NT-4/5(wt)). Additionally, we show that acute, prolonged administration of recombinant NT-4/5 (5 microg/kg/day) prevented up to 50% of the hippocampal CA pyramidal cell death following LFP TBI in rats. These results suggest that post-traumatic increases in endogenous NT-4/5 may be part of an adaptive neuroprotective response in the injured brain, and that administration of this neurotrophic factor may be useful as a therapeutic strategy following TBI.


Subject(s)
Brain Injuries , Hippocampus , Nerve Growth Factors/metabolism , Neuroprotective Agents/metabolism , Pyramidal Cells/metabolism , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Gene Deletion , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Knockout , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Neuroprotective Agents/administration & dosage , Pyramidal Cells/cytology , Pyramidal Cells/pathology , Random Allocation , Rats
5.
Neuroscience ; 136(4): 971-89, 2005.
Article in English | MEDLINE | ID: mdl-16242846

ABSTRACT

Approximately 4000 human beings experience a traumatic brain injury each day in the United States ranging in severity from mild to fatal. Improvements in initial management, surgical treatment, and neurointensive care have resulted in a better prognosis for traumatic brain injury patients but, to date, there is no available pharmaceutical treatment with proven efficacy, and prevention is the major protective strategy. Many patients are left with disabling changes in cognition, motor function, and personality. Over the past two decades, a number of experimental laboratories have attempted to develop novel and innovative ways to replicate, in animal models, the different aspects of this heterogenous clinical paradigm to better understand and treat patients after traumatic brain injury. Although several clinically-relevant but different experimental models have been developed to reproduce specific characteristics of human traumatic brain injury, its heterogeneity does not allow one single model to reproduce the entire spectrum of events that may occur. The use of these models has resulted in an increased understanding of the pathophysiology of traumatic brain injury, including changes in molecular and cellular pathways and neurobehavioral outcomes. This review provides an up-to-date and critical analysis of the existing models of traumatic brain injury with a view toward guiding and improving future research endeavors.


Subject(s)
Brain Injuries , Disease Models, Animal , Animals , Behavior, Animal , Brain Injuries/classification , Brain Injuries/physiopathology , Humans , Mice
6.
Neuroscience ; 134(3): 1047-56, 2005.
Article in English | MEDLINE | ID: mdl-15979242

ABSTRACT

Traumatic brain injury causes long-term neurological motor and cognitive deficits, often with limited recovery. The inability of CNS axons to regenerate following traumatic brain injury may be due, in part, to inhibitory molecules associated with myelin. One of these myelin-associated proteins, Nogo-A, inhibits neurite outgrowth in vitro, and inhibition of Nogo-A in vivo enhances axonal outgrowth and sprouting and improves outcome following experimental CNS insults. However, the involvement of Nogo-A in the neurobehavioral deficits observed in experimental traumatic brain injury remains unknown and was evaluated in the present study using the 11C7 monoclonal antibody against Nogo-A. Anesthetized, male Sprague-Dawley rats were subjected to either lateral fluid percussion brain injury of moderate severity (2.5-2.6 atm) or sham injury. Beginning 24 h post-injury, monoclonal antibody 11C7 (n=17 injured, n=6 shams included) or control Ab (IgG) (n=16 injured, n=5 shams included) was infused at a rate of 5 microl/h over 14 days into the ipsilateral ventricle using osmotic minipumps connected to an implanted cannula. Rats were assessed up to 4 weeks post-injury using tests for neurological motor function (composite neuroscore, and sensorimotor test of adhesive paper removal) and, at 4 weeks, cognition was assessed using the Morris water maze. Hippocampal CA3 pyramidal neuron damage and corticospinal tract sprouting, using an anterograde tracer (biotinylated dextran amine), were also evaluated. Brain injury significantly increased sprouting from the uninjured corticospinal tract but treatment with monoclonal antibody 11C7 did not further increase the extent of sprouting nor did it alter the extent of CA3 cell damage. Animals treated with 11C7 showed no improvement in neurologic motor deficits but did show significantly improved cognitive function at 4 weeks post-injury when compared with brain-injured, IgG-treated animals. To our knowledge, the present findings are the first to suggest that (1) traumatic brain injury induces axonal sprouting in the corticospinal tract and this sprouting may be independent of myelin-associated inhibitory factors and (2) that post-traumatic inhibition of Nogo-A may promote cognitive recovery unrelated to sprouting in the corticospinal tract or neuroprotective effects on hippocampal cell loss following experimental traumatic brain injury.


Subject(s)
Axons/physiology , Brain Injuries/physiopathology , Cognition/physiology , Motor Activity/physiology , Myelin Proteins/physiology , Analysis of Variance , Animals , Antibodies/pharmacology , Behavior, Animal , Biotin/analogs & derivatives , Biotin/metabolism , Dextrans/metabolism , Disease Models, Animal , Functional Laterality , Hippocampus/pathology , Male , Myelin Proteins/immunology , Nogo Proteins , Rats , Rats, Sprague-Dawley , Reaction Time , Recovery of Function/physiology , Time Factors
7.
J Neurosci ; 23(27): 9046-58, 2003 Oct 08.
Article in English | MEDLINE | ID: mdl-14534238

ABSTRACT

Mitochondria are localized to regions of the cell where ATP consumption is high and are dispersed according to changes in local energy needs. In addition to motion directed by molecular motors, mitochondrial distribution in neuronal cells appears to depend on the docking of mitochondria to microtubules and neurofilaments. We examined interactions between mitochondria and neurofilaments using fluorescence microscopy, dynamic light scattering, atomic force microscopy, and sedimentation assays. Mitochondria-neurofilament interactions depend on mitochondrial membrane potential, as revealed by staining with a membrane potential sensitive dye (JC-1) in the presence of substrates/ADP or uncouplers (valinomycin/carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone) and are affected by the phosphorylation status of neurofilaments and neurofilament sidearms. Antibodies against the neurofilament heavy subunit disrupt binding between mitochondria and neurofilaments, and isolated neurofilament sidearms alone interact with mitochondria, suggesting that they mediate the interactions between the two structures. These data suggest that specific and regulated mitochondrial-neurofilament interactions occur in situ and may contribute to the dynamic distribution of these organelles within the cytoplasm of neurons.


Subject(s)
Cytoskeleton/chemistry , Mitochondria/chemistry , Animals , Antibodies/metabolism , Binding, Competitive/physiology , Cattle , Cytoskeleton/immunology , Cytoskeleton/metabolism , Electrophoresis, Polyacrylamide Gel , Internet , Membrane Potentials/physiology , Microscopy, Atomic Force , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurofilament Proteins/chemistry , Neurofilament Proteins/metabolism , Phosphorylation , Protein Binding/physiology , Spinal Cord/chemistry , Video Recording
8.
Curr Opin Pharmacol ; 3(1): 27-32, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12550738

ABSTRACT

The intensity of experimental and clinical research to identify a neuroprotective drug for the treatment of traumatic brain injury is motivated by the devastating morbidity and mortality of this condition. Encouraging experimental work has led so far to disappointing clinical trials and the identification of new potential therapeutic targets is critically dependent on a better understanding of the chronic pathophysiology triggered by the initial insult. Future advances in the pharmacological treatment of traumatic brain injury are likely to include the evaluation of sequentially timed therapies combining multiple and targeted agents, and manipulation of the newly discovered neurogenic potential of the adult brain together with the refinement of traditional interventions to block specific cytotoxic cascades.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/metabolism , Animals , Brain Injuries/physiopathology , Cannabinoids/therapeutic use , Enzyme Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/metabolism , Humans
9.
Clin Neuropathol ; 21(4): 156-62, 2002.
Article in English | MEDLINE | ID: mdl-12143927

ABSTRACT

Paraffin sections from the hippocampus of 12 head-injured patients (Group A, aged between 4 and 12 years n = 6 and Group B, aged between 64 and 89 years n = 6) and associated age-matched controls were stained by the terminal deoxynucleotidyl transferase-mediated biotinylated deoxyuridine triphosphate nick end labeling (TUNEL) technique for evidence of in-situ DNA fragmentation. TUNEL+ cells were of 2 Types: I (non-apoptotic) and II (apoptotic). In addition sections stained H&E, combined Luxol Fast Blue/Cresyl Violet and by immunohistochemistry for astrocytes (GFAP) and macrophages (CD68) were used to characterize the lesions. Small numbers of Type I TUNEL+ cells were seen in all sectors of the hippocampus except CA2 of both Groups A and B. Type II TUNEL+ cells were mainly found in the white matter. They constituted less than 1% of all TUNEL+ cells. There were similar or fewer TUNEL+ cells in the corresponding areas in the controls compared with the head-injured patients. However, in the dentate fascia and the CA4 sector of the Group B cases, larger numbers of TUNEL+ cells were seen in controls than after trauma. In the grey matter most TUNEL+ cells had the morphology ofnecrosis that corresponded with foci of selective neuronal damage. Only a few TUNEL+ cells were seen in white matter. The occasional Type I TUNEL+ cells were seen in grey matter. It is concluded that the amount and distribution of DNA fragmentation in children and adults is similar and therefore at least in the hippocampus does not provide an explanation for age as an independent variable of outcome after traumatic brain injury in childhood.


Subject(s)
Brain Injuries/genetics , Brain Injuries/pathology , DNA Damage/genetics , DNA Fragmentation/genetics , Hippocampus/injuries , Hippocampus/pathology , Age Factors , Aged , Aged, 80 and over , Brain Injuries/mortality , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Predictive Value of Tests , Trauma Severity Indices
10.
Neuroscience ; 110(4): 605-16, 2002.
Article in English | MEDLINE | ID: mdl-11934469

ABSTRACT

Although mild traumatic brain injury is associated with behavioral dysfunction and histopathological alterations, few studies have assessed the temporal pattern of regional apoptosis following mild brain injury. Anesthetized rats were subjected to mild lateral fluid-percussion brain injury (1.1-1.3 atm), and brains were evaluated for the presence of in situ DNA fragmentation (terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling, TUNEL) and morphologic characteristics of apoptotic cell death (nuclear and cytoplasmic condensation, presence of apoptotic bodies). Significant numbers of apoptotic TUNEL(+) cells were observed in the injured parietal cortex and underlying white matter up to 72 h post-injury (P<0.05 compared to sham-injured-injured), with maximal numbers present at 24 h. Apoptosis was confirmed by the presence of 180-200 bp nuclear DNA fragments in tissue homogenates. The appearance of apoptotic TUNEL(+) cells in the injured cortex was preceded by a marked decrease in immunoreactivity for the anti-cell death protein, Bcl-2, as early as 2 h post-injury. This decrease in cellular Bcl-2 staining was not accompanied by a concomitant loss of staining for the pro-cell death Bax protein, suggesting that post-traumatic neuronal death in the cortex may be dependent on altered cellular ratios of Bcl-2:Bax. In the hippocampus, no significant increase in apoptotic TUNEL(+) cells was observed compared to sham-injured-injured animals. However, selective neuronal loss was evident in the CA3 region at 24 h post-injury, that was preceded by an overt loss of neuronal Bcl-2 immunoreactivity at 6 h. No changes in either cellular Bcl-2 or Bax expression were observed in the thalamus or white matter at any time post-injury. Taken together from these data, we suggest that apoptosis contributes to cell death in both gray and white matter, and that decreases in cellular Bcl-2 may, in part, be associated with both apoptotic and non-apoptotic cell death following mild brain trauma.


Subject(s)
Apoptosis/physiology , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Down-Regulation/physiology , Hippocampus/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Count , Cell Size/physiology , Cerebral Cortex/injuries , Cerebral Cortex/pathology , DNA Fragmentation/physiology , Hippocampus/injuries , Hippocampus/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Nerve Degeneration/pathology , Neurons/pathology , Proto-Oncogene Proteins/metabolism , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein
11.
J Cereb Blood Flow Metab ; 21(11): 1241-58, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11702040

ABSTRACT

The mechanisms underlying secondary cell death after traumatic brain injury (TBI) are poorly understood. Animal models of TBI recapitulate many clinical and pathologic aspects of human head injury, and the development of genetically engineered animals has offered the opportunity to investigate the specific molecular and cellular mechanisms associated with cell dysfunction and death after TBI, allowing for the evaluation of specific cause-effect relations and mechanistic hypotheses. This article represents a compendium of the current literature using genetically engineered mice in studies designed to better understand the posttraumatic inflammatory response, the mechanisms underlying DNA damage, repair, and cell death, and the link between TBI and neurodegenerative diseases.


Subject(s)
Brain Injuries/genetics , Brain Injuries/physiopathology , Disease Models, Animal , Mice, Transgenic , Animals , Mice
12.
J Neurosurg ; 95(5): 859-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11702878

ABSTRACT

OBJECT: Mild, traumatic repetitive head injury (RHI) leads to neurobehavioral impairment and is associated with the early onset of neurodegenerative disease. The authors developed an animal model to investigate the behavioral and pathological changes associated with RHI. METHODS: Adult male C57BL/6 mice were subjected to a single injury (43 mice), repetitive injury (two injuries 24 hours apart; 49 mice), or no impact (36 mice). Cognitive function was assessed using the Morris water maze test, and neurological motor function was evaluated using a battery of neuroscore, rotarod, and rotating pole tests. The animals were also evaluated for cardiovascular changes, blood-brain barrier (BBB) breakdown, traumatic axonal injury, and neurodegenerative and histopathological changes between 1 day and 56 days after brain trauma. No cognitive dysfunction was detected in any group. The single-impact group showed mild impairment according to the neuroscore test at only 3 days postinjury, whereas RHI caused pronounced deficits at 3 days and 7 days following the second injury. Moreover, RHI led to functional impairment during the rotarod and rotating pole tests that was not observed in any animal after a single impact. Small areas of cortical BBB breakdown and axonal injury. observed after a single brain injury, were profoundly exacerbated after RHI. Immunohistochemical staining for microtubule-associated protein-2 revealed marked regional loss of immunoreactivity only in animals subjected to RHI. No deposits of beta-amyloid or tau were observed in any brain-injured animal. CONCLUSIONS: On the basis of their results, the authors suggest that the brain has an increased vulnerability to a second traumatic insult for at least 24 hours following an initial episode of mild brain trauma.


Subject(s)
Brain Concussion/etiology , Brain/physiopathology , Craniocerebral Trauma/complications , Craniocerebral Trauma/physiopathology , Animals , Brain/pathology , Cardiovascular System/physiopathology , Cognition , Craniocerebral Trauma/pathology , Craniocerebral Trauma/psychology , Disease Susceptibility , Male , Mice , Mice, Inbred C57BL , Movement Disorders/etiology , Recurrence , Severity of Illness Index
13.
Restor Neurol Neurosci ; 18(1): 1-8, 2001.
Article in English | MEDLINE | ID: mdl-11673665

ABSTRACT

PURPOSE: Cyclosporin A (CsA) is widely used in clinical situations to attenuate graft rejection following organ and central nervous system transplantation. Previous studies demonstrated that CsA administration is neuroprotective in models of traumatic brain injury (TBI). However, no studies, to date, have evaluated the influence of post-injury CsA administration on behavioral recovery after TBI. METHODS: Rats (n = 33) were anesthetized and subjected to severe, lateral fluid percussion brain injury. Fifteen minutes thereafter, animals were randomized to receive the first of 28 daily injections of either CsA (10 mg/kg, ip) or saline. Sham-operated animals (n = 14) were anesthetized and surgically prepared without injury and treated daily either with CsA or saline. Motor and sensorimotor functions were assessed at one day before and two days after injury, and weekly thereafter up to 4 wks post-injury. Cognition was assessed at 1 and 4 wks post-injury using a Morris Water Maze test. RESULTS: Injured animals showed significant impairments in motor, sensorimotor and cognitive function over the 4-week post-injury period. Injured animals treated with CsA showed a significant improvement in motor function assessed using a composite neuroscore (at day 28) and in sensorimotor function assessed using a sticky paper test (at days 2, 14, and 28) (p < 0.05, when compared to vehicle treated, injured animals). No beneficial effects on cognitive function were observed following CSA administration. CONCLUSION: These data suggest that daily post-injury treatment with CsA improves certain aspects of motor and sensorimotor function following experimental TBI.


Subject(s)
Brain Injuries/drug therapy , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Recovery of Function/drug effects , Animals , Behavior, Animal , Brain Injuries/mortality , Cognition , Male , Maze Learning , Motor Activity , Motor Neurons/physiology , Neurons, Afferent/physiology , Rats , Rats, Sprague-Dawley
14.
Curr Pharm Des ; 7(15): 1505-16, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562295

ABSTRACT

Considerable effort has led to an increased interest in emerging preclinical and clinical data regarding the pathophysiological changes in the posttraumatic brain. It is widely believed that delayed cell damage and death contributes to behavioral impairment following traumatic brain injury. However, no drug therapy to attenuate this process is available at present, and the development of new therapeutic regimen is urgently warranted. This manuscript represents a compendium of recent preclinical work undertaken to evaluate new pharmacologic strategies in the experimental setting as a first step towards the development of a therapeutic armamentarium directed to improve functional recovery in head-injured patients.


Subject(s)
Brain Injuries/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Glycoproteins/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Caspases/metabolism , Cations/metabolism , Cytoskeleton/drug effects , Drug Therapy/methods , Free Radical Scavengers/therapeutic use , Homeostasis/physiology , Humans , Immunosuppressive Agents/therapeutic use , Kallikrein-Kinin System/drug effects , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Nerve Growth Factors/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism
15.
J Neurotrauma ; 18(8): 821-32, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11526988

ABSTRACT

Reactive oxygen species (ROS) are thought to contribute to the secondary injury process after traumatic brain injury (TBI). ROS scavenging compounds have shown neuroprotective properties in various models of experimental brain injury, including TBI. Administration of nitrone radical scavengers has emerged as a promising pharmacological concept in focal experimental ischemia due to their low toxicity and neuroprotective properties, with a time window of several hours. The aim of this study was to test the neuroprotective efficacy of two nitrones, the readily blood-brain barrier (BBB) penetrating alpha-phenyl-N-tert-butyl nitrone (PBN) and the poorly BBB penetrating sulfo-derivative, 2-sulfo-phenyl-N-tert-butyl nitrone (S-PBN) after moderate (2.20-2.45 atm) lateral fluid percussion injury (FPI) in rats. Twenty-six rats received a 24-h intravenous infusion (30 mg/kg/h) of saline, PBN, or an equimolar dose of S-PBN beginning 30 min after FPI. Eight sham-operated animals were used as controls. Cognitive function was assessed using the Morris Water Maze at day 11-15 after TBI, neurological status at day 1, 4, and 8 and morphological outcome at day 15. PBN and S-PBN treatment significantly reduced the loss of ipsilateral hemispheric tissue whereas only S-PBN tended to reduce the cortical lesion volume. PBN treatment caused a significant improvement in the neurological score as compared to saline-treated animals, while S-PBN alone attenuated the cognitive deficit. Our results suggest that nitrone radical scavengers are neuroprotective when administered 30 min after FPI in rats. Differences in pharmacokinetics may account for the observed individual neuroprotective profiles of the two nitrones.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/metabolism , Free Radical Scavengers/pharmacology , Nitrogen Oxides/pharmacology , Recovery of Function/drug effects , Animals , Cyclic N-Oxides , Infusions, Intravenous , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
16.
J Neurosurg ; 94(5): 765-74, 2001 May.
Article in English | MEDLINE | ID: mdl-11354408

ABSTRACT

OBJECT: Immortalized neural progenitor cells derived from embryonic rat hippocampus (HiB5), were transduced ex vivo with the gene for mouse nerve growth factor (NGF) to secrete NGF (NGF-HiB5) at 2 ng/hr/10(5) cells in culture. METHODS: Fifty-nine male Wistar rats weighing 300 to 370 g each were anesthetized with 60 mg/kg sodium pentobarbital and subjected to lateral fluid-percussion brain injury of moderate severity (2.3-2.4 atm, 34 rats) or sham injury (25 rats). At 24 hours postinjury, 2 microl (150,000 cells/microl) of [3H]thymidine-labeled NGF-HiB5 cells were transplanted stereotactically into three individual sites in the cerebral cortex adjacent to the injury site (14 rats). Separate groups of brain-injured rats received nontransfected (naive [n])-HiB5 cells (12 animals) or cell suspension vehicle (eight animals). One week postinjury, animals underwent neurological evaluation for motor function and cognition (Morris water maze) and were killed for histological, autoradiographic, and immunocytochemical analysis. Viable HiB5 cell grafts were identified in all animals, together with reactive microglia and macrophages located throughout the periinjured parenchyma and grafts (OX-42 immunohistochemistry). Brain-injured animals transplanted with either NGF-HiB5 or n-HiB5 cells displayed significantly improved neuromotor function (p < 0.05) and spatial learning behavior (p < 0.005) compared with brain-injured animals receiving microinjections of vehicle alone. A significant reduction in hippocampal CA3 cell death was observed in brain-injured animals receiving transplants of NGF-HiB5 cells compared with those receiving n-HiB5 cells or vehicle (p < 0.025). CONCLUSIONS: This study demonstrates that immortalized neural stem cells that have been retrovirally transduced to produce NGF can markedly improve cognitive and neuromotor function and rescue hippocampal CA3 neurons when transplanted into the injured brain during the acute posttraumatic period.


Subject(s)
Antigens, CD , Antigens, Neoplasm , Antigens, Surface , Avian Proteins , Blood Proteins , Brain Injuries/therapy , Brain Tissue Transplantation , Genetic Therapy/methods , Nerve Growth Factor/genetics , Neurons/transplantation , Stem Cell Transplantation , Animals , Basigin , Behavior, Animal , Brain Injuries/surgery , Cell Line, Transformed/transplantation , Cerebral Cortex/cytology , Cerebral Cortex/surgery , Cognition , Conditioning, Psychological , Gene Expression , Graft Survival , Hippocampus/cytology , Male , Membrane Glycoproteins/analysis , Memory , Motor Activity , Neurologic Examination , Neurons/chemistry , Neurons/cytology , Rats , Rats, Wistar , Recovery of Function , Stem Cells/chemistry , Stem Cells/cytology
17.
J Neurotrauma ; 18(4): 369-76, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11336438

ABSTRACT

The nuclear enzyme poly(ADP-ribose) polymerase (PARP), which has been shown to be activated following experimental traumatic brain injury (TBI), binds to DNA strand breaks and utilizes nicotinamide adenine dinucleotide (NAD) as a substrate. Since consumption of NAD may be deleterious to recovery in the setting of CNS injury, we examined the effect of a potent PARP inhibitor, GPI 6150, on histological outcome following TBI in the rat. Rats (n = 16) were anesthetized, received a preinjury dose of GPI 6150 (30 min; 15 mg/kg, i.p.), subjected to lateral fluid percussion (FP) brain injury of moderate severity (2.5-2.8 atm), and then received a second dose 3 h postinjury (15 mg/kg, i.p.). Lesion area was examined using Nissl staining, while DNA fragmentation and apoptosis-associated cell death was assessed with terminal deoxynucleotidyl-transferase-mediated biotin-dUTP nick end labeling (TUNEL) with stringent morphological evaluation. Twenty-four hours after brain injury, a significant cortical lesion and number of TUNEL-positive/nonapoptotic cells and TUNEL-positive/apoptotic cells in the injured cortex of vehicle-treated animals were observed as compared to uninjured rats. The size of the trauma-induced lesion area was significantly attenuated in the GPI 6150-treated animals versus vehicle-treated animals (p < 0.05). Treatment of GPI 6150 did not significantly affect the number of TUNEL-positive apoptotic cells in the injured cortex. The observed neuroprotective effects on lesion size, however, offer a promising option for further evaluation of PARP inhibition as a means to reduce cellular damage associated with TBI.


Subject(s)
Benzopyrans/pharmacology , Brain Injuries/enzymology , Brain Injuries/pathology , Enzyme Inhibitors/pharmacology , Isoquinolines/pharmacology , Neuroprotective Agents/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Apoptosis/drug effects , Brain Injuries/genetics , DNA Fragmentation/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , Rats , Rats, Sprague-Dawley
18.
J Neurotrauma ; 18(3): 275-85, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11284548

ABSTRACT

Endothelial-derived prostacyclin is an important regulator of microvascular function, and its main actions are inhibition of platelet/leukocyte aggregation and adhesion, and vasodilation. Disturbances in endothelial integrity following traumatic brain injury (TBI) may result in insufficient prostacyclin production and participate in the pathophysiological sequelae of brain injury. The objective of this study was to evaluate the potential therapeutic effects of a low-dose prostacyclin infusion on cortical lesion volume, CA3 neuron survival and functional outcome following TBI in the rat. Anesthetized animals (sodium pentobarbital, 60 mg/kg, i.p.) were subjected to a lateral fluid percussion brain injury (2.5 atm) or sham injury. Following TBI, animals were randomized to receive a constant infusion of either prostacyclin (1 ng/kg x min(-1) i.v.) or vehicle over 48 h. All sham animals received vehicle (n = 6). Evaluation of neuromotor function, lesion volume, and CA3 neuronal loss was performed blindly. By 7 days postinjury, cortical lesion volume was significantly reduced by 43% in the prostacyclin-treated group as compared to the vehicle treated group (p < 0.01; n = 12 prostacyclin, n = 12 vehicle). No differences were observed in neuromotor function (48 h and 7 days following TBI), or in hippocampal cell loss (7 days following TBI) between the prostacyclin- and vehicle-treated groups. We conclude that prostacyclin in a low dose reduces loss of neocortical neurons following TBI and may be a potential clinical therapeutic agent to reduce neuronal cell death associated with brain trauma.


Subject(s)
Brain Injuries/drug therapy , Epoprostenol/administration & dosage , Neocortex/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Animals , Brain Injuries/pathology , Male , Neocortex/injuries , Neocortex/pathology , Rats , Rats, Sprague-Dawley
19.
J Cereb Blood Flow Metab ; 21(4): 396-403, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11323525

ABSTRACT

Large-conductance, calcium-activated potassium (maxi-K) channels regulate neurotransmitter release and neuronal excitability, and openers of these channels have been shown to be neuroprotective in models of cerebral ischemia. The authors evaluated the effects of postinjury systemic administration of the maxi-K channel opener, BMS-204352, on behavioral and histologic outcome after lateral fluid percussion (FP) traumatic brain injury (TBI) in the rat. Anesthetized Sprague-Dawley rats (n = 142) were subjected to moderate FP brain injury (n = 88) or surgery without injury (n = 54) and were randomized to receive a bolus of 0.1 mg/kg BMS-204352 (n = 26, injured; n = 18, sham), 0.03 mg/kg BMS-204352 (n = 25, injured; n = 18, sham), or 2% dimethyl sulfoxide (DMSO) in polyethylene glycol (vehicle, n = 27, injured; n = 18, sham) at 10 minutes postinjury. One group of rats was tested for memory retention (Morris water maze) at 42 hours postinjury, then killed for evaluation of regional cerebral edema. A second group of injured/sham rats was assessed for neurologic motor function from 48 hours to 2 weeks postinjury and cortical lesion area. Administration of 0.1 mg/kg BMS-204352 improved neurologic motor function at 1 and 2 weeks postinjury (P < 0.05) and reduced the extent of cerebral edema in the ipsilateral hippocampus, thalamus, and adjacent cortex (P < 0.05). Administration of 0.03 mg/kg BMS-204352 significantly reduced cerebral edema in the ipsilateral thalamus (P < 0.05). No effects on cognitive function or cortical tissue loss were observed with either dose. These results suggest that the novel maxi-K channel opener BMS-204352 may be selectively beneficial in the treatment of experimental TBI.


Subject(s)
Brain Edema/drug therapy , Brain Injuries/drug therapy , Indoles/pharmacology , Ion Channel Gating/drug effects , Potassium Channels, Calcium-Activated , Potassium Channels/metabolism , Animals , Brain Chemistry/drug effects , Brain Edema/mortality , Brain Edema/pathology , Brain Injuries/mortality , Brain Injuries/pathology , Cognition Disorders/drug therapy , Cognition Disorders/mortality , Cognition Disorders/pathology , Disease Models, Animal , Large-Conductance Calcium-Activated Potassium Channels , Male , Maze Learning/drug effects , Motor Neurons/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
20.
J Neuropathol Exp Neurol ; 60(2): 183-94, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11273006

ABSTRACT

Traumatic brain injury results in a profound decline in intracellular magnesium ion levels that may jeopardize critical cellular functions. We examined the consequences of preinjury magnesium deficiency and post-traumatic magnesium treatment on injury-induced cytoskeletal damage and cell death at 24 h after injury. Adult male rats were fed either a normal (n = 24) or magnesium-deficient diet (n = 16) for 2 wk prior to anesthesia and lateral fluid percussion brain injury (n = 31) or sham injury (n = 9). Normally fed animals were then randomized to receive magnesium chloride (125 micromol, i.v., n = 10) or vehicle solution (n = 11) at 10 min postinjury. Magnesium treatment reduced cortical cell loss (p < 0.05), cortical alterations in microtubule-associated protein-2 (MAP-2) (p < 0.05), and both cortical and hippocampal calpain-mediated spectrin breakdown (p < 0.05 for each region) when compared to vehicle treatment. Conversely, magnesium deficiency prior to brain injury led to a greater area of cortical cell loss (p < 0.05 compared to vehicle treatment). Moreover, brain injury to magnesium-deficient rats resulted in cytoskeletal alterations within the cortex and hippocampus that were not observed in vehicle- or magnesium-treated animals. These data suggest that cortical cell death and cytoskeletal disruptions in cortical and hippocampal neurons may be sensitive to magnesium status after experimental brain injury, and may be mediated in part through modulation of calpains.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Cell Death/drug effects , Cytoskeleton/metabolism , Magnesium Deficiency/complications , Magnesium/pharmacology , Neurons/metabolism , Animals , Brain/drug effects , Brain/pathology , Brain Injuries/drug therapy , Brain Injuries/pathology , Calpain/drug effects , Calpain/metabolism , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytoskeleton/drug effects , Cytoskeleton/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Magnesium/metabolism , Magnesium Deficiency/pathology , Magnesium Deficiency/physiopathology , Male , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Spectrin/drug effects , Spectrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...