Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Neurotrauma ; 28(7): 1185-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506725

ABSTRACT

Traumatic axonal injury (TAI) is a major feature of traumatic brain injury (TBI) and is associated with much of its morbidity. To date, significant insight has been gained into the initiating pathogenesis of TAI. However, the nature of TAI within the injured brain precludes the consistent evaluation of its specific anterograde and retrograde sequelae. To overcome this limitation, we used the relatively organized optic nerve in a central fluid percussion injury (cFPI) model. To improve the visualization of TAI, we utilized mice expressing yellow fluorescent protein (YFP) in their visual pathways. Through this approach, we consistently generated TAI in the optic nerve and qualitatively and quantitatively evaluated its progression over a 48-h period in YFP axons via confocal microscopy and electron microscopy. In this model, delayed axonal swelling with subsequent disconnection were the norm, together with the fact that once disconnected, both the proximal and distal axonal segments revealed significant dieback, with the proximal swellings showing regression and reorganization, while the distal swellings persisted, although showing signs of impending degeneration. When antibodies targeting the C-terminus of amyloid precursor protein (APP), a routine marker of TAI were employed, they mapped exclusively to the proximal axonal segments without distal targeting, regardless of the survival time. Concomitant with this evolving axonal pathology, focal YFP fluorescence quenching occurred and mapped precisely to immunoreactive loci positive for Texas-Red-conjugated-IgG, indicating that blood-brain barrier disruption and its attendant edema contributed to this phenomenon. This was confirmed through the use of antibodies targeting endogenous YFP, which demonstrated the retention of intact immunoreactive axons despite YFP fluorescence quenching. Collectively, the results of this study within the injured optic nerve provide unprecedented insight into the evolving pathobiology associated with TAI.


Subject(s)
Axons/pathology , Brain Injuries/pathology , Nerve Regeneration/physiology , Optic Nerve Injuries/pathology , Wallerian Degeneration/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Bacterial Proteins/metabolism , Biomarkers/metabolism , Blood-Brain Barrier/injuries , Blood-Brain Barrier/physiopathology , Brain Injuries/physiopathology , Disease Models, Animal , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve Injuries/physiopathology , Wallerian Degeneration/physiopathology
2.
J Biomed Sci ; 17: 38, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20482789

ABSTRACT

BACKGROUND: Excitatory amino acid release and subsequent biochemical cascades following traumatic brain injury (TBI) have been well documented, especially glutamate-related excitotoxicity. The effects of TBI on the essential functions of inhibitory GABA-A receptors, however, are poorly understood. METHODS: We used Western blot procedures to test whether in vivo TBI in rat altered the protein expression of hippocampal GABA-A receptor subunits alpha1, alpha2, alpha3, alpha5, beta3, and gamma2 at 3 h, 6 h, 24 h, and 7 days post-injury. We then used pre-injury injections of MK-801 to block calcium influx through the NMDA receptor, diltiazem to block L-type voltage-gated calcium influx, or diazepam to enhance chloride conductance, and re-examined the protein expressions of alpha1, alpha2, alpha3, and gamma2, all of which were altered by TBI in the first study and all of which are important constituents in benzodiazepine-sensitive GABA-A receptors. RESULTS: Western blot analysis revealed no injury-induced alterations in protein expression for GABA-A receptor alpha2 or alpha5 subunits at any time point post-injury. Significant time-dependent changes in alpha1, alpha3, beta3, and gamma2 protein expression. The pattern of alterations to GABA-A subunits was nearly identical after diltiazem and diazepam treatment, and MK-801 normalized expression of all subunits 24 hours post-TBI. CONCLUSIONS: These studies are the first to demonstrate that GABA-A receptor subunit expression is altered by TBI in vivo, and these alterations may be driven by calcium-mediated cascades in hippocampal neurons. Changes in GABA-A receptors in the hippocampus after TBI may have far-reaching consequences considering their essential importance in maintaining inhibitory balance and their extensive impact on neuronal function.


Subject(s)
Brain Injuries/metabolism , Diazepam/pharmacology , Diltiazem/pharmacology , Dizocilpine Maleate/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Receptors, GABA-A/metabolism , Animals , Blotting, Western , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Chlorides/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Protein Subunits , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
3.
J Neurotrauma ; 26(4): 527-37, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19245307

ABSTRACT

Traumatic brain injury (TBI) has been demonstrated to induce cerebral vascular dysfunction that is reflected in altered responses to various vasodilators. While previous reports have focused primarily on the short-term vascular alterations, few have examined these vascular changes for more than 7 days, or have attempted to correlate these alterations with any persisting behavioral changes or potential therapeutic modulation. Accordingly, we evaluated the long-term microvascular and behavioral consequences of experimental TBI and their therapeutic modulation via hypothermia. In this study, one group was injured with no treatment, another group was injured and 1 h later was treated with 120 min of hypothermia followed by slow rewarming, and a third group was non-injured. Animals equipped with cranial windows for visualization of the pial microvasculature were challenged with various vasodilators, including acetylcholine, hypercapnia, adenosine, pinacidil, and sodium nitroprusside, at either 1 or 3 weeks post-TBI. In addition, all animals were tested for vestibulomotor tasks at 1 week post-TBI, and animals surviving for 3 weeks post-TBI were tested in a Morris water maze (MWM). The results of this investigation demonstrated that TBI resulted in long-term vascular dysfunction in terms of altered vascular reactivity to various vasodilators, which was significantly improved with the use of a delayed 120-min hypothermic treatment. In contrast, data from the MWM task indicated that injured animals revealed persistent deficits in the spatial memory test performance, with hypothermia exerting no protective effects. Collectively, these data illustrate that TBI can evoke long-standing brain vascular and spatial memory dysfunction that manifest different responses to hypothermic intervention. These findings further illustrate the complexity of TBI and highlight the fact that the chosen hypothermic intervention may not necessarily exert a global protective response.


Subject(s)
Brain Injuries/physiopathology , Brain Injuries/therapy , Cerebrovascular Disorders/physiopathology , Cerebrovascular Disorders/therapy , Hypothermia, Induced/methods , Microcirculation/physiology , Animals , Body Temperature/physiology , Brain/blood supply , Brain/pathology , Brain/physiopathology , Brain Injuries/complications , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Cerebrovascular Disorders/etiology , Craniotomy , Cytoprotection/physiology , Disease Models, Animal , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/physiopathology , Memory Disorders/therapy , Microcirculation/drug effects , Pia Mater/blood supply , Pia Mater/drug effects , Pia Mater/physiopathology , Rats , Rewarming/methods , Time , Time Factors , Treatment Outcome , Vasodilator Agents/pharmacology
4.
Neurosurgery ; 63(4): 799-806; discussion 806-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18981892

ABSTRACT

OBJECTIVE: Perfluorocarbon emulsions have been shown to improve outcomes in stroke models. This study examined the effect of Oxycyte, a third-generation perfluorocarbon emulsion (04RD33; Synthetic Blood International, Inc., Costa Mesa, CA) treatment on cognitive recovery and mitochondrial oxygen consumption after a moderate lateral fluid percussion injury (LFPI). METHODS: Adult male Sprague-Dawley rats (Harlan Bioproducts for Science, Indianapolis, IN) were allocated to 4 groups: 1) LFPI treated with a lower dose of Oxycyte (4.5 mL/kg); 2) LFPI with a higher dose of Oxycyte (9.0 mL/kg); 3) LFPI with saline infusion; and 4) sham animals treated with saline. Fifteen minutes after receiving moderate LFPI or sham surgery, animals were infused intravenously with Oxycyte or saline within 30 minutes while breathing 100% O2. Animals breathed 100% O2 continuously for a total of 4 hours after injury. At 11 to 15 days after LFPI, animals were assessed for cognitive deficits using the Morris water maze test. They were sacrificed at Day 15 after injury for histology to assess hippocampal neuronal cell loss. In a parallel study, mitochondrial oxygen consumption values were measured by the Cartesian diver microrespirometer method. RESULTS: We found that injured animals treated with a lower or higher dose of Oxycyte had significant improvement in cognitive function when compared with injured saline-control animals (P < 0.05). Moreover, injured animals that received either dose of Oxycyte had significantly less neuronal cell loss in the hippocampal CA3 region compared with saline-treated animals (P < 0.05). Furthermore, a lower dose of Oxycyte significantly improved mitochondrial oxygen consumption levels (P < 0.05). CONCLUSION: The current study demonstrates that Oxycyte can improve cognitive recovery and reduce CA3 neuronal cell loss after traumatic brain injury in rats.


Subject(s)
Brain Injuries/drug therapy , Cognition Disorders/drug therapy , Fluorocarbons/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Blood Pressure/drug effects , Brain Injuries/complications , Brain Injuries/physiopathology , Cell Count , Cognition Disorders/etiology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/injuries , Hippocampus/pathology , Male , Maze Learning/drug effects , Neurons/drug effects , Neurons/pathology , Oxygen Consumption/drug effects , Oxygen Inhalation Therapy , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Wounds, Nonpenetrating
5.
Neurosci Lett ; 441(1): 115-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18583041

ABSTRACT

Traumatic brain injury (TBI) survivors often suffer from a post-traumatic syndrome with deficits in learning and memory. Calcium (Ca(2+)) has been implicated in the pathophysiology of TBI-induced neuronal death. However, the role of long-term changes in neuronal Ca(2+) function in surviving neurons and the potential impact on TBI-induced cognitive impairments are less understood. Here we evaluated neuronal death and basal free intracellular Ca(2+) ([Ca(2+)](i)) in acutely isolated rat CA3 hippocampal neurons using the Ca(2+) indicator, Fura-2, at seven and thirty days after moderate central fluid percussion injury. In moderate TBI, cognitive deficits as evaluated by the Morris Water Maze (MWM), occur after injury but resolve after several weeks. Using MWM paradigm we compared alterations in [Ca(2+)](i) and cognitive deficits. Moderate TBI did not cause significant hippocampal neuronal death. However, basal [Ca(2+)](i) was significantly elevated when measured seven days post-TBI. At the same time, these animals exhibited significant cognitive impairment (F(2,25)=3.43, p<0.05). When measured 30 days post-TBI, both basal [Ca(2+)](i) and cognitive functions had returned to normal. Pretreatment with MK-801 blocked this elevation in [Ca(2+)](i) and also prevented MWM deficits. These studies provide evidence for a link between elevated [Ca(2+)](i) and altered cognition. Since no significant neuronal death was observed, the alterations in Ca(2+) homeostasis in the traumatized, but surviving neurons may play a role in the pathophysiology of cognitive deficits that manifest in the acute setting after TBI and represent a novel target for therapeutic intervention following TBI.


Subject(s)
Brain Injuries/complications , Calcium/metabolism , Cognition Disorders , Hippocampus/pathology , Neurons/metabolism , Analysis of Variance , Animals , Cell Count/methods , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Maze Learning/physiology , Rats , Time Factors
6.
Brain Res ; 1213: 69-77, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18455704

ABSTRACT

Memory impairment is common following traumatic brain injury. However, the specific processes underlying the impairments remain unknown. Traumatic brain injury may interfere with several of the stages of the learning and memory process. In two separate experiments, we examined the specific nature of both anterograde and retrograde memory dysfunction following fluid percussion brain injury in rats. In Experiment 1, we examined the retention of spatial memory in the MWM after equating initial learning between sham and injured animals. Animals were trained to criterion and then tested for retention 4, 8, or 24 h post-training. Although injured animals displayed deficits in task acquisition, retention performance was not significantly different between groups. In Experiment 2, we examined the effects of injury on the retention of retrograde spatial memories in the MWM. Animals were injured either 1 or 14 days post-training and then received retention probe trials followed by a reminding procedure and second probe trial 14 days post-injury. All injured animals displayed retention deficits in the probe trials 14 days post-injury. However, after the reminding procedure, injured animals displayed sham-level performance during the second probe trial. The results of these experiments suggest that with anterograde memory impairment induced by traumatic brain injury, the primary deficit lies in task acquisition, not the retention of information within long-term memory. Retrograde memory impairment following injury appears to be mediated primarily by deficits in memory retrieval.


Subject(s)
Amnesia, Anterograde/etiology , Amnesia, Retrograde/etiology , Brain Injuries/complications , Animals , Disease Models, Animal , Male , Multivariate Analysis , Rats , Rats, Sprague-Dawley , Time Factors
7.
Eur J Neurosci ; 27(7): 1659-72, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18371074

ABSTRACT

Traumatic brain injury (TBI) survivors often suffer chronically from significant morbidity associated with cognitive deficits, behavioral difficulties and a post-traumatic syndrome and thus it is important to understand the pathophysiology of these long-term plasticity changes after TBI. Calcium (Ca2+) has been implicated in the pathophysiology of TBI-induced neuronal death and other forms of brain injury including stroke and status epilepticus. However, the potential role of long-term changes in neuronal Ca2+ dynamics after TBI has not been evaluated. In the present study, we measured basal free intracellular Ca2+ concentration ([Ca2+](i)) in acutely isolated CA3 hippocampal neurons from Sprague-Dawley rats at 1, 7 and 30 days after moderate central fluid percussion injury. Basal [Ca2+](i) was significantly elevated when measured 1 and 7 days post-TBI without evidence of neuronal death. Basal [Ca2+](i) returned to normal when measured 30 days post-TBI. In contrast, abnormalities in Ca2+ homeostasis were found for as long as 30 days after TBI. Studies evaluating the mechanisms underlying the altered Ca2+ homeostasis in TBI neurons indicated that necrotic or apoptotic cell death and abnormalities in Ca2+ influx and efflux mechanisms could not account for these changes and suggested that long-term changes in Ca2+ buffering or Ca2+ sequestration/release mechanisms underlie these changes in Ca2+ homeostasis after TBI. Further elucidation of the mechanisms of altered Ca2+ homeostasis in traumatized, surviving neurons in TBI may offer novel therapeutic interventions that may contribute to the treatment and relief of some of the morbidity associated with TBI.


Subject(s)
Brain Injuries/metabolism , Calcium/metabolism , Hippocampus/metabolism , Homeostasis/physiology , Intracellular Fluid/physiology , Neurons/metabolism , Animals , Brain Injuries/pathology , Calcium/physiology , Cell Survival/physiology , Hippocampus/cytology , Intracellular Fluid/metabolism , Male , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Time
8.
J Neurotrauma ; 25(3): 248-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18352838

ABSTRACT

Catecholaminergic neurotransmission is regionally altered following injury, and drugs aimed at these systems offer promising avenues for post-traumatic brain injury (TBI) pharmacotherapies. Atomoxetine is a selective norepinephrine transporter (NET) inhibitor currently indicated for treatment of attention-deficit hyperactivity disorder (ADHD). The current study was designed to test the efficacy of atomoxetine in treating cognitive deficits following experimental TBI in animals and to determine an optimal dose and therapeutic window for drug treatment. Sprague-Dawley rats were subjected to lateral fluid-percussion injury (L-FPI) of moderate severity (2.08 atm +/- 0.05). Two experiments were performed. In the first study, atomoxetine (0.3, 1, 3, or 9 mg/kg) or vehicle was administered daily on post-injury days (PID) 1-15. Cognitive assessment was performed using the Morris water maze on PID 11-15. L-FPI resulted in significant cognitive impairment when compared to Sham-Injury. Treatment with lower doses of atomoxetine (0.3, 1, and 3 mg/kg) significantly attenuated the cognitive deficits in injured animals. Treatment with the higher dosage (9 mg/kg) of atomoxetine resulted in animals that were not significantly different than injured-vehicle treated animals. The optimal response was achieved using 1 mg/kg atomoxetine. In the second study, treatment with atomoxetine (1 mg/kg) or vehicle was delayed for 11 days post-injury. Rats were administered atomoxetine daily for 15 days, and cognitive assessment was performed on PID 25-29. In this study, treatment with atomoxetine (1 mg/kg) did not result in improved cognitive performance. In conclusion, this is the first study to show low-dose atomoxetine initiated early after experimental TBI results in improved cognition.


Subject(s)
Brain Injuries/complications , Brain Injuries/drug therapy , Brain/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Propylamines/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Atomoxetine Hydrochloride , Brain/metabolism , Brain/physiopathology , Brain Injuries/physiopathology , Catecholamines/metabolism , Cognition/drug effects , Cognition/physiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Maze Learning/drug effects , Maze Learning/physiology , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Propylamines/therapeutic use , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Time Factors , Treatment Outcome
9.
Neurosci Lett ; 436(1): 27-30, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18355962

ABSTRACT

Experimental traumatic brain injury (TBI) results in marked neurochemical and metabolic changes. Research has demonstrated that after the initial insult the brain undergoes an immediate state of hypermetabolism followed by a sustained period of hypometabolism. The altered extra- and intracellular environment can compromise neuronal performance and limit functional recovery. If brain metabolism is depressed chronically after TBI, then interventions that are designed to increase metabolism may be beneficial to outcome. Glucose treatment has been shown to improve cognition in many populations, particularly those with cognitive deficits. The following experiments examined the effects of delayed postinjury glucose supplementation on cognitive function following TBI. Male Sprague-Dawley rats received either sham or lateral fluid-percussion (LFP) injury. Cognitive functioning was assessed with the Morris water maze (MWM) on postinjury days 11-15. In the first experiment, saline or 100mg/kg glucose was administered 10 min before cognition assessment. Injured animals treated with glucose displayed significantly shorter latencies to reach the goal platform compared to injured saline-treated animals. Glucose had no effect on sham-injured rats. In the second experiment, injured rats were given daily injections of saline or 100mg/kg glucose for 10 days beginning 24h after injury. Rats were then tested in the MWM on days 11-15 without glucose or saline treatment. In this experiment, glucose treatment did not affect MWM performance. These data provide evidence that the chronic energy supplementation after TBI improves outcome when administered shortly before cognitive assessment.


Subject(s)
Brain Injuries/drug therapy , Brain/drug effects , Cognition Disorders/drug therapy , Glucose/administration & dosage , Recovery of Function/drug effects , Animals , Brain/metabolism , Brain Injuries/complications , Cognition Disorders/etiology , Drug Administration Schedule , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley
10.
J Neurosurg ; 106(4): 687-94, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17432723

ABSTRACT

OBJECT: Hyperbaric oxygen (HBO2) has been shown to improve outcome after severe traumatic brain injury, but its underlying mechanisms are unknown. Following lateral fluid-percussion injury (FPI), the authors tested the effects of HBO2 treatment as well as enhanced normobaric oxygenation on mitochondrial function, as measured by both cognitive recovery and cellular adenosine triphosphate (ATP) levels. METHODS: Adult male Sprague-Dawley rats were subjected to moderate lateral FPI or sham injury and were allocated to one of four treatment groups: 1) FPI treated with 4 hours of normobaric 30% O2; 2) FPI treated with 4 hours of normobaric 100% O2; 3) FPI treated with 1 hour of HBO2 plus 3 hours of normobaric 100% O2; and 4) sham-injured treated with normobaric 30% O2. Cognitive outcome was assessed using the Morris water maze (MWM) on Days 11 to 15 after injury. Animals were then killed 21 days postinjury to assess hippocampal neuronal loss. Adenosine triphosphate was extracted from the neocortex and measured using high-performance liquid chromatography. The results showed that injured animals treated with HBO2 or normobaric 100% O2 alone had significantly higher levels of cerebral ATP as compared with animals treated using normobaric 30% O2 (p < or = 0.05). The injured animals treated with HBO2 had significant improvements in cognitive recovery, as characterized by a shorter latency in MWM performance (p < or = 0.05), and decreased neuronal loss in the CA2/3 and hilar regions as compared with those treated with 30% or 100% O2, (p < or = 0.05). CONCLUSIONS: Both hyperbaric and normobaric hyperoxia increased cerebral ATP levels after lateral FPI. In addition, HBO2 treatment improved cognitive recovery and reduced hippocampal neuronal cell loss after brain injury in the rat.


Subject(s)
Brain Injuries/therapy , Cognition/physiology , Hyperbaric Oxygenation , Mitochondria/physiology , Adenosine Triphosphate/metabolism , Animals , Brain Injuries/metabolism , Brain Injuries/psychology , Chromatography, High Pressure Liquid , Free Radicals/metabolism , Hippocampus/pathology , Male , Maze Learning , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology
11.
Brain Inj ; 21(3): 259-74, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17453754

ABSTRACT

PRIMARY OBJECTIVE: We provide a review of recent chronic and delayed rehabilitative pharmacological treatments examined in experimental models of traumatic brain injury. There is a specific emphasis on studies aiming to enhance cognitive recovery. MAIN OUTCOMES AND RESULTS: Decreased neuronal activity is believed to contribute to persistent cognitive disabilities. Neurotransmitter based rehabilitative treatments that increase neuronal activity may assist in the recovery of cognitive function. However, timing and dosage of drug treatment are influential in cognitive enhancement. Drug treatments that affect single and multiple neurotransmitter systems have the ability to significantly influence recovery of function following brain injury. CONCLUSIONS: Understanding the relationship between neural disturbances and functional deficits following brain injury is challenging. Cognitive impairment may be the result of a single event or multiple events that occur after the initial insult. Increasing neuronal activity during the chronic phase of injury seems to be an effective treatment strategy for facilitating cognitive recovery. Pharmacological agents do not necessarily display the same effects in an injured brain as in a non-injured brain. Thus, further research is needed to establish the effectiveness of rehabilitative drug treatments.


Subject(s)
Brain Injuries/drug therapy , Cognition Disorders/prevention & control , Animals , Brain Injuries/psychology , Brain Injuries/rehabilitation , Cognition Disorders/etiology , Disease Models, Animal , Rats , Recovery of Function
12.
J Neurotrauma ; 23(10): 1529-34, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17020487

ABSTRACT

Memory impairment following traumatic brain injury (TBI) is common in both humans and animals. A noteworthy feature of memory dysfunction in human TBI is impaired memory performance that is dependent on the delay between initial learning and recall of information. However, previous studies of TBI-induced memory impairment in animals have failed to control for the initial amount of learning between sham and injured animals. The present study demonstrates that experimental TBI in rats produces delay-dependent memory impairment, even when the initial degree of learning is controlled for. Animals were injured at a moderate level of lateral fluid percussion (LFP) injury (n = 10) or received a sham injury (n = 9), and then trained in a water T-maze version of the delayed-non-matching-to-place (DNMP) task beginning 10 days post-injury. Acquisition training consisted of 15 trials per day on post-injury days 11-15 using a minimal (7-sec) delay between the sample and choice phases of the task. Following acquisition, the delay between the sample and choice phases of the task was progressively increased to 15, 30, and 120 sec. Injured animals acquired the task at the same rate as sham animals and performed equally well at the 15-sec delay (p > 0.05). However, as the delay increased to 30 and 120 sec, the performance of the injured animals deteriorated (p < 0.05). These results indicate that LFP injury produces delay-dependent memory impairments in rats. This is therefore a valid model of an important feature of memory impairment in human TBI, and should be a useful addition to the available methods for assessing memory impairment and the effect of therapeutic interventions after TBI.


Subject(s)
Brain Injuries/psychology , Maze Learning/physiology , Memory Disorders/etiology , Animals , Conditioning, Operant/physiology , Disease Models, Animal , Male , Memory, Short-Term/physiology , Rats , Rats, Sprague-Dawley , Time Factors
13.
J Neurotrauma ; 23(8): 1233-40, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928181

ABSTRACT

Chronic cognitive impairment is an enduring aspect of traumatic brain injury (TBI) in both humans and animals. Treating cognitive impairment in the post-traumatic stages of injury often involves the delivery of pharmacologic agents aimed at specific neurotransmitter systems. The current investigation examined the effects of the nootropoic drug aniracetam on cognitive recovery following TBI in rats. Three experiments were performed to determine (1) the optimal dose of aniracetam for treating cognitive impairment, (2) the effect of delaying drug treatment for a period of days following TBI, and (3) the effect of terminating drug treatment before cognitive assessment. In experiment 1, rats were administered moderate fluid percussion injury and treated with vehicle, 25, or 50 mg/kg aniracetam for 15 days. Both doses of aniracetam effectively reduced injury-induced deficits in the Morris water maze (MWM) as measured on postinjury days 11-15. In experiment 2, injured rats were treated with 50 mg/kg aniracetam or vehicle beginning on day 11 postinjury and continuing for 15 days. MWM performance, assessed on days 26-30, indicates that aniracetam-treated animals performed as well as sham-injured controls. In experiment 3, animals were injured and treated with aniracetam for 15 days. Drug treatment was terminated during MWM testing on postinjury days 16-20. In this experiment, aniracetam-treated rats did not perform better than vehicle-treated rats. The results of these experiments indicate that aniracetam is an effective treatment for cognitive impairment induced by TBI, even when treatment is delayed for a period of days following injury.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/psychology , Cognition/physiology , Nootropic Agents/therapeutic use , Psychomotor Performance/physiology , Pyrrolidinones/therapeutic use , Animals , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Maze Learning/physiology , Nootropic Agents/administration & dosage , Pyrrolidinones/administration & dosage , Rats , Rats, Sprague-Dawley , Time Factors
14.
J Neurotrauma ; 23(6): 962-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16774480

ABSTRACT

Severe traumatic brain injury (TBI) leads to an immunocompromised state responsible for an increased morbidity and mortality. Our understanding of the mechanisms responsible for this brain damage is incomplete. Damage maybe mediated by a complex cascade of neuroinflammation, and cytokine activation. In addition, translocation and accumulation of T cells in the brain parenchyma could take place and be related to detrimental effects. Our aims in this prospective randomized pilot study, were to detect the early effect of severe TBI upon cell-mediated immunity, to verify if early immunologic impairment correlates with neurologic outcome, and finally, to test the effect of early administration of iv infusion of cyclosporin A upon cell-mediated immunologic function. Forty-nine patients with severe TBI were studied. Thirty-six of these patients received a 24-h intravenous infusion of Cyclosporin A, or two 24-h infusions of the drug. 10 patients were in the placebo group. Three patients, not enrolled in the cyclosporin trial, were studied only for the relationship between cellular immunity, neurological outcome, and infection rate. T cell counts and microbiological cultures were performed in all patients. Sixty-five percent of patients demonstrated reduced T lymphocyte counts on admission. Furthermore, reduction of T cell numbers was related with significantly worse neurologic outcome and an increase in pulmonary infection. There was no significant difference between the placebo and CsA treated patients for the studied immunological parameters, or for incidence of infection. We also observed sequestration/diapedesis of T cells into the brain parenchyma, around contusions, after human TBI and we speculate that this could be responsible for further brain damage.


Subject(s)
Brain Injuries/immunology , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Count , T-Lymphocytes/immunology , Adult , Aging/immunology , Aging/physiology , Brain/pathology , Brain Injuries/pathology , CD3 Complex/immunology , CD4 Lymphocyte Count , CD4-CD8 Ratio , CD8-Positive T-Lymphocytes/immunology , Female , Glasgow Coma Scale , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Male , Pilot Projects , Prospective Studies , T-Lymphocytes/drug effects , T-Lymphocytes/microbiology
15.
J Neurotrauma ; 22(6): 645-55, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15941374

ABSTRACT

Evidence of injury-induced neurogenesis in the adult hippocampus suggests that an endogenous repair mechanism exists for cognitive dysfunction following traumatic brain injury (TBI). One factor that may be associated with this restoration is S100B, a neurotrophic/mitogenic protein produced by astrocytes, which has been shown to improve memory function. Therefore, we examined whether an intraventricular S100B infusion enhances neurogenesis within the hippocampus following experimental TBI and whether the biological response can be associated with a measurable cognitive improvement. Following lateral fluid percussion or sham injury in male rats (n = 60), we infused S100B (50 ng/h) or vehicle into the lateral ventricle for 7 days using an osmotic micro-pump. Cell proliferation was assessed by injecting the mitotic marker bromodeoxyuridine (BrdU) on day 2 postinjury. Quantification of BrdU-immunoreactive cells in the dentate gyrus revealed an S100B-enhanced proliferation as assessed on day 5 post-injury (p < 0.05), persisting up to 5 weeks (p < 0.05). Using cell-specific markers, we determined the relative numbers of these progenitor cells that became neurons or glia and found that S100B profoundly increased hippocampal neurogenesis 5 weeks after TBI (p < 0.05). Furthermore, spatial learning ability, as assessed by the Morris water maze on day 30-34 post-injury, revealed an improved cognitive performance after S100B infusion (p < 0.05). Collectively, our findings indicate that an intraventricular S100B infusion induces neurogenesis within the hippocampus, which can be associated with an enhanced cognitive function following experimental TBI. These observations provide compelling evidence for the therapeutic potential of S100B in improving functional recovery following TBI.


Subject(s)
Brain Injuries/drug therapy , Cell Differentiation/drug effects , Nerve Growth Factors/pharmacology , Nerve Regeneration/drug effects , Neuronal Plasticity/drug effects , Recovery of Function/drug effects , S100 Proteins/pharmacology , Animals , Biomarkers/metabolism , Brain Injuries/physiopathology , Brain Injuries/psychology , Bromodeoxyuridine , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Disease Models, Animal , Injections, Intraventricular , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/physiopathology , Nerve Growth Factors/therapeutic use , Nerve Regeneration/physiology , Neuroglia/drug effects , Neuroglia/metabolism , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/therapeutic use , Stem Cells/drug effects , Stem Cells/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
16.
Brain Res ; 1048(1-2): 153-60, 2005 Jun 28.
Article in English | MEDLINE | ID: mdl-15919062

ABSTRACT

Calcineurin, a neuronally enriched, calcium-stimulated phosphatase, is an important modulator of many neuronal processes, including several that are physiologically related to the pathology of traumatic brain injury. The effect of moderate, central fluid percussion injury on the subcellular distribution of this important neuronal enzyme was examined. Animals were sacrificed at several time points post-injury and calcineurin distribution in subcellular fractions was assayed by Western blot analysis and immunohistochemistry. A persistent increase in calcineurin concentration was observed in crude synaptoplasmic membrane-containing fractions. In cortical fractions, calcineurin immunoreactivity remained persistently increased for 2 weeks post-injury. In hippocampal homogenates, calcineurin immunoreactivity remained increased for up to 4 weeks. Finally, immunohistochemical analysis of hippocampal slices revealed increased staining in the apical dendrites of CA1 neurons. The increased staining was greatest in magnitude 24 h post-injury; however, staining was still more intense than control 4 weeks post-injury. The data support the conclusion that fluid percussion injury results in redistribution of the enzyme in the rat forebrain. These changes have broad physiological implications, possibly resulting in altered cellular excitability or a greater likelihood of neuronal cell death.


Subject(s)
Brain Injuries/metabolism , Calcineurin/metabolism , Neurons/metabolism , Percussion/methods , Animals , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Brain Injuries/etiology , Densitometry/methods , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Time Factors
17.
J Neurotrauma ; 22(4): 476-90, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15853464

ABSTRACT

Calcineurin, a neuronally enriched, calcium-stimulated phosphatase, is an important modulator of many neuronal processes, including several that are physiologically related to the pathology of traumatic brain injury. This study examined the effects of moderate, central fluid percussion injury on the activity of this important neuronal enzyme. Animals were sacrificed at several time-points postinjury and cortical, hippocampal, and cerebellar homogenates were assayed for calcineurin activity by dephosphorylation of p-nitrophenol phosphate. A significant brain injury-dependent increase was observed in both hippocampal and cortical homogenates under both basal and maximally-stimulated reaction conditions. This increase persisted 2-3 weeks post-injury. Brain injury did not alter substrate affinity, but did induce a significant increase in the apparent maximal dephosphorylation rate. Unlike the other brain regions, no change in calcineurin activity was observed in the cerebellum following brain injury. No brain region tested displayed a significant change in calcineurin enzyme levels as determined by Western blot, demonstrating that increased enzyme synthesis was not responsible for the observed increase in activity. The data support the conclusion that fluid percussion injury results in increased calcineurin activity in the rat forebrain. This increased activity has broad physiological implications, possibly resulting in altered cellular excitability or a greater likelihood of neuronal cell death.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/physiopathology , Brain/metabolism , Brain/physiopathology , Calcineurin/metabolism , Up-Regulation/physiology , Animals , Brain/pathology , Brain Injuries/pathology , Cell Death/physiology , Cerebellum/metabolism , Cerebellum/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Enzyme Activation/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Kinetics , Male , Nerve Degeneration/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Time Factors
18.
J Neurotrauma ; 21(5): 541-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15165362

ABSTRACT

Elevated serum S100B levels have been shown to be a predictor of poor outcome after traumatic brain injury (TBI). Experimental data, on the other hand, demonstrate a neuroprotective and neurotrophic effect of this calcium-binding protein. The purpose of this study was to examine the role of increased S100B levels on functional outcome after TBI. Following lateral fluid percussion or sham injury in male Sprague Dawley rats (n = 56), we infused S100B (50 ng/h) or vehicle into the cerebrospinal fluid of the ipsilateral ventricle for 7 days using an osmotic mini-pump. Assessment of cognitive performance by the Morris water maze on days 30-34 after injury revealed an improved performance of injured animals after S100B infusion (p < 0.05), when compared to vehicle infusion. Blood samples for analysis of clinical markers of brain damage, S100B and neuron specific enolase, taken at 30 min, 3 h, 4 h, 2 days, or 5 days showed a typical peak 3 h after injury (p < 0.01), and higher serum levels correlated significantly with an impaired cognitive recovery (p < 0.01). The correlation of higher serum S100B levels with poor water maze performance may result from injury induced opening of the blood-brain barrier, allowing the passage of S100B into serum. Thus while higher serum levels of S100B seem to reflect the degree of blood-brain barrier opening and severity of injury, a beneficial effect of intraventricular S100B administration on long-term functional recovery after TBI has been demonstrated for the first time. The exact mechanism by which S100B exerts its neuroprotective or neurotrophic influence remains unknown and needs to be elucidated by further investigation.


Subject(s)
Brain Injuries/drug therapy , Cognition/drug effects , Nerve Growth Factors/administration & dosage , Neuroprotective Agents/therapeutic use , S100 Proteins/administration & dosage , Animals , Brain Injuries/blood , Injections, Intraventricular , Male , Maze Learning/drug effects , Nerve Growth Factors/blood , Phosphopyruvate Hydratase/blood , Rats , Rats, Sprague-Dawley , Recovery of Function , S100 Calcium Binding Protein beta Subunit , S100 Proteins/blood
19.
Am J Phys Med Rehabil ; 82(11): 871-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14566156

ABSTRACT

OBJECTIVE: Traumatic brain injury can cause a variety of impairments, including persistent alterations in personality, mood, and cognition. Antipsychotic agents are frequently used to treat pathologic behaviors in traumatic brain injury patients, but the influence of prolonged administration of such drugs on cognition after injury is unknown. The effects of two antipsychotic drugs on cognitive recovery after traumatic brain injury were assessed using the fluid percussion model in rats. DESIGN: The typical antipsychotic, haloperidol, and the third-generation antipsychotic, olanzapine, were administered via intraperitoneal injection beginning 24 hr after injury and continuing daily for the duration of the study. Morris water maze performance was assessed on days 11-15 postinjury. RESULTS: Haloperidol, an antagonist acting on D2-like dopamine receptors, exacerbated the cognitive deficits induced by injury, as injured rats treated with 0.30 mg/kg haloperidol performed worse in the Morris water maze than injured rats treated with vehicle. CONCLUSIONS: Our results demonstrate the importance of the D2 receptor in cognitive recovery after traumatic brain injury. Also, the data illustrate that some classes of antipsychotic drugs may influence cognitive recovery, and further research is needed to determine the optimal pharmacologic treatment of aggression, agitation, and other pathologic behaviors in patients with traumatic brain injury.


Subject(s)
Antipsychotic Agents/pharmacology , Brain Injuries/complications , Cognition Disorders/drug therapy , Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacology , Analysis of Variance , Animals , Benzodiazepines , Cognition Disorders/etiology , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Olanzapine , Rats , Rats, Sprague-Dawley , Swimming
20.
J Neurotrauma ; 19(7): 829-41, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12184853

ABSTRACT

Traumatic brain injury (TBI) triggers a complex pathophysiological cascade, leading to cell death. A major factor in the pathogenesis of TBI is neuronal overloading with calcium, causing the opening of mitochondrial permeability transition pores (MPTP), which consequently inhibit normal mitochondrial function. The immunosuppressant Cyclosporin A (CsA) has been shown to block MPTPs, and to be neuroprotective in ischemia and TBI. However, the translation of these effects on mitochondrial function, into behavioral endpoints has not been investigated thoroughly. Therefore, we tested the effect of a low, clinically evaluated, CsA dose of 0.125 mg/kg (infused for 3 h) and a higher "known" neuroprotective dose of 18.75 mg/kg on brain tissue O(2) consumption, and on motor and cognitive performance following lateral fluid percussion injury (FPI) in rats. CsA at both concentrations abolished the 25% decrease in O(2) consumption (VO(2)), seen in saline-treated animals at 5 h post-FPI. Furthermore, the lower dose of CsA also ameliorated acute motor deficits (days 1-5 post-FPI) and learning and memory impairments in a Morris water maze test on days 11-15 post-FPI. Although, the higher dose of CsA improved cognitive performance, it worsened acute motor functional recovery. These results suggest, that the CsA-induced preservation of mitochondrial function, as assessed by tissue O(2) consumption, directly translated into improvements in motor and cognitive behavior.


Subject(s)
Brain Injuries/drug therapy , Brain/drug effects , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Learning/drug effects , Oxygen Consumption/drug effects , Animals , Brain/metabolism , Brain Injuries/metabolism , Cognition/drug effects , Cognition/physiology , Learning/physiology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen Consumption/physiology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...