Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Behav Neurosci ; 16: 863195, 2022.
Article in English | MEDLINE | ID: mdl-35747840

ABSTRACT

Blast traumatic brain injury (bTBI) presents a serious threat to military personnel and often results in psychiatric conditions related to limbic system dysfunction. In this study, the functional outcomes for anxiety- and depressive-like behaviors and neuronal activation were evaluated in male and female mice after exposure to an Advanced Blast Simulator (ABS) shock wave. Mice were placed in a ventrally exposed orientation inside of the ABS test section and received primary and tertiary shock wave insults of approximately 15 psi peak pressure. Evans blue staining indicated cases of blood-brain barrier breach in the superficial cerebral cortex four, but not 24 h after blast, but the severity was variable. Behavioral testing with the elevated plus maze (EPM) or elevated zero maze (EZM), sucrose preference test (SPT), and tail suspension test (TST) or forced swim test (FST) were conducted 8 days-3.5 weeks after shock wave exposure. There was a sex difference, but no injury effect, for distance travelled in the EZM where female mice travelled significantly farther than males. The SPT and FST did not indicate group differences; however, injured mice were less immobile than sham mice during the TST; possibly indicating more agitated behavior. In a separate cohort of animals, the expression of the immediate early gene, c-Fos, was detected 4 h after undergoing bTBI or sham procedures. No differences in c-Fos expression were found in the cerebral cortex, but female mice in general displayed enhanced c-Fos activation in the paraventricular nucleus of the thalamus (PVT) compared to male mice. In the amygdala, more c-Fos-positive cells were observed in injured animals compared to sham mice. The observed sex differences in the PVT and c-Fos activation in the amygdala may correlate with the reported hyperactivity of females post-injury. This study demonstrates, albeit with mild effects, behavioral and neuronal activation correlates in female rodents after blast injury that could be relevant to the incidence of increased post-traumatic stress disorder in women.

2.
J Neurotrauma ; 39(11-12): 784-799, 2022 06.
Article in English | MEDLINE | ID: mdl-35243900

ABSTRACT

The consequences of forceful rotational acceleration on the central nervous system are not fully understood. While traumatic brain injury (TBI) research primarily has focused on effects related to the brain parenchyma, reports of traumatic meningeal enhancement in TBI patients may possess clinical significance. The objective of this study was to evaluate the meninges and brain for changes in dynamic contrast enhancement (DCE) magnetic resonance imaging (MRI) following closed-head impact model of engineered rotational acceleration (CHIMERA)-induced cerebral insult. Adult male and female mice received one (1 × ; n = 19 CHIMERA, n = 19 Sham) or four (4 × one/day; n = 18 CHIMERA, n = 12 Sham) injuries. Each animal underwent three MRI scans: 1 week before injury, immediately after the final injury, and 1 week post-injury. Compared with baseline readings and measures in sham animals, meningeal DCE in males was increased after single impact and repetitive injury. In female mice, DCE was elevated relative to their baseline level after a single impact. One week after CHIMERA, the meningeal enhancement returned to below baseline for single injured male mice, but compared with uninjured mice remained elevated in both sexes in the multiple impact groups. Pre-DCE meningeal T2-weighted relaxation time was increased only after 1 × CHIMERA in injured mice. Since vision is impaired after CHIMERA, visual pathway regions were analyzed through imaging and glial fibrillary acidic protein (GFAP) histology. Initial DCE in the lateral geniculate nucleus (LGN) and superior colliculus (SC) and T2 increases in the optic tract (OPT) and LGN were observed after injury with decreases in DCE and T2 1 week later. Astrogliosis was apparent in the OPT and SC with increased GFAP staining 7 days post-injury. To our knowledge, this is the first study to examine meningeal integrity after CHIMERA in both male and female rodents. DCE-MRI may serve as a useful approach for pre-clinical models of meningeal injury that will enable further evaluation of the underlying mechanisms.


Subject(s)
Brain Injuries, Traumatic , Visual Pathways , Animals , Female , Humans , Male , Mice , Acceleration , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Magnetic Resonance Imaging , Meninges/diagnostic imaging , Mice, Inbred C57BL , Visual Pathways/pathology
3.
J Neurotrauma ; 38(11): 1585-1606, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33622092

ABSTRACT

Cognitive dysfunction is a common, often long-term complaint following acquired traumatic brain injury (TBI). Cognitive deficits suggest dysfunction in hippocampal circuits. The goal of the studies described here is to phenotype in both male and female mice the hippocampal-dependent learning and memory deficits resulting from TBI sustained by the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) device-a model that delivers both a contact-concussion injury as well as unrestrained rotational head movement. Mice sustained either sham procedures or four injuries (0.7 J, 24-h intervals). Spatial learning and memory skills assessed in the Morris water maze (MWM) approximately 3 weeks following injuries were significantly impaired by brain injuries; however, slower swimming speeds and poor performance on visible platform trials suggest that measurement of cognitive impairment with this test is confounded by injury-induced motor and/or visual impairments. A separate experiment confirmed hippocampal-dependent cognitive deficits with trace fear conditioning (TFC), a behavioral test less dependent on motor and visual function. Male mice had greater injury-induced deficits on both the MWM and TFC tests than female mice. Pathologically, the injury was characterized by white matter damage as observed by silver staining and glial fibrillary acidic protein (astrogliosis) in the optic tracts, with milder damage seen in the corpus callosum, and fimbria and brainstem (cerebral peduncles) of some animals. No changes in the density of GABAergic parvalbumin-expressing cells in the hippocampus, amygdala, or parietal cortex were found. This experiment confirmed significant sexually dimorphic cognitive impairments following a repeated, diffuse brain injury.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/psychology , Cognitive Dysfunction/etiology , Hippocampus/physiopathology , Memory/physiology , Spatial Learning/physiology , Animals , Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/psychology , Conditioning, Psychological , Disease Models, Animal , Fear , Female , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Sex Factors
4.
J Neurotrauma ; 38(11): 1551-1571, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33605175

ABSTRACT

In civilian and military settings, mild traumatic brain injury (mTBI) is a common consequence of impacts to the head, sudden blows to the body, and exposure to high-energy atmospheric shockwaves from blast. In some cases, mTBI from blast exposure results in long-term emotional and cognitive deficits and an elevated risk for certain neuropsychiatric diseases. Here, we tested the effects of mTBI on various forms of auditory-cued fear learning and other measures of cognition in male C57BL/6J mice after single or repeated blast exposure (blast TBI; bTBI). bTBI produced an abnormality in the temporal organization of cue-induced freezing behavior in a conditioned trace fear test. Spatial working memory, evaluated by the Y-maze task performance, was also deleteriously affected by bTBI. Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis for glial markers indicated an alteration in the expression of myelin-related genes in the hippocampus and corpus callosum 1-8 weeks after bTBI. Immunohistochemical and ultrastructural analyses detected bTBI-related myelin and axonal damage in the hippocampus and corpus callosum. Together, these data suggest a possible link between blast-induced mTBI, myelin/axonal injury, and cognitive dysfunction.


Subject(s)
Blast Injuries/pathology , Blast Injuries/psychology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Cognitive Dysfunction/etiology , Myelin Sheath/pathology , Animals , Disease Models, Animal , Fear , Male , Mice , Mice, Inbred C57BL , Spatial Memory
5.
Brain Res ; 1750: 147147, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33091394

ABSTRACT

The frontal lobes are among the most vulnerable sites in traumatic brain injuries. In the current study, a balanced 2 × 2 × 2 design (n = 18 mice/group), female and male C57Bl/6J mice received repeated bilateral frontal concussive brain injury (frCBI) and underwent fear conditioning (FC) to assess how injured mice respond to adverse conditions. Shocks received during FC impacted behavior on all subsequent tests except the tail suspension test. FC resulted in more freezing behavior in all mice that received foot shocks when evaluated in subsequent context and cue tests and induced hypoactivity in the open field (OF) and elevated zero maze (EZM). Mice that sustained frCBI learned the FC association between tone and shock. Injured mice froze less than sham controls during context and cue tests, which could indicate memory impairment, but could also suggest that frCBI resulted in hyperactivity that overrode the rodent's natural freezing response to threat, as injured mice were also more active in the OF and EZM. There were notable sex differences, where female mice exhibited more freezing behavior than male mice during FC context and cue tests. The findings suggest frCBI impaired, but did not eliminate, FC retention and resulted in an overall increase in general activity. The injury was characterized pathologically by increased inflammation (CD11b staining) in cortical regions underlying the injury site and in the optic tracts. The performance of male and female mice after injury suggested the complexity of possible sex differences for neuropsychiatric symptoms.


Subject(s)
Brain Injuries, Traumatic/psychology , Fear/physiology , Animals , Behavior, Animal , Brain/physiopathology , Brain Concussion/psychology , Brain Injuries, Traumatic/physiopathology , Conditioning, Classical/physiology , Conditioning, Operant/physiology , Fear/psychology , Female , Frontal Lobe/physiopathology , Male , Memory Disorders , Mice , Mice, Inbred C57BL , Sex Factors
6.
PLoS One ; 14(9): e0222153, 2019.
Article in English | MEDLINE | ID: mdl-31487322

ABSTRACT

There is strong evidence to suggest a link between repeated head trauma and cognitive and emotional disorders, and Repetitive concussive brain injuries (rCBI) may also be a risk factor for depression and anxiety disorders. Animal models of brain injury afford the opportunity for controlled study of the effects of injury on functional outcomes. In this study, male and cycling female C57BL/6J mice sustained rCBI (3x) at 24-hr intervals and were tested in a context and cued fear conditioning paradigm, open field (OF), elevated zero maze and tail suspension test. All mice with rCBI showed less freezing behavior than sham control mice during the fear conditioning context test. Injured male, but not female mice also froze less in response to the auditory cue (tone). Injured mice were hyperactive in an OF environment and spent more time in the open quadrants of the elevated zero maze, suggesting decreased anxiety, but there were no differences between injured mice and sham-controls in depressive-like activity on the tail suspension test. Pathologically, injured mice showed increased astrogliosis in the injured cortex and white matter tracts (optic tracts and corpus callosum). There were no changes in the number of parvalbumin-positive interneurons in the cortex or amygdala, but injured male mice had fewer parvalbumin-positive neurons in the hippocampus. Parvalbumin-reactive interneurons of the hippocampus have been previously demonstrated to be involved in hippocampal-cortical interactions required for memory consolidation, and it is possible memory changes in the fear-conditioning paradigm following rCBI are the result of more subtle imbalances in excitation and inhibition both within the amygdala and hippocampus, and between more widespread brain regions that are injured following a diffuse brain injury.


Subject(s)
Anxiety/pathology , Brain Concussion/complications , Cues , Depression/pathology , Fear/psychology , Hippocampus/metabolism , Parvalbumins/metabolism , Animals , Anxiety/etiology , Behavior, Animal , Brain Concussion/psychology , Cell Count , Depression/etiology , Female , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Sex Factors
7.
Brain Res ; 1723: 146400, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31445032

ABSTRACT

Changes in 18F-fluorodeoxyglucose ([18F]FDG) measured by positron emission tomography (PET) can be used for the noninvasive detection of metabolic dysfunction following mild traumatic brain injury (mTBI). This study examined the time course of metabolic changes induced by primary blast injury by measuring regional [18F]FDG uptake. Adult, male rats were exposed to blast overpressure (15 psi) or sham injury, and [18F]FDG uptake was measured before injury and again at 1-3 h and 7 days post-injury, using both volume-of-interest (VOI) and voxel-based analysis. VOI analysis revealed significantly increased [18F]FDG uptake in corpus callosum and amygdala at both 1-3 h and 7 days following blast, while a transient decrease in uptake was observed in the midbrain at 1-3 h only. Voxel-based analysis revealed similar significant differences in uptake between sham and blast-injured rats at both time points. At 1-3 h post-injury, clusters of increased uptake were found in the amygdala, somatosensory cortex, and corpus callosum, while regions of decreased uptake were observed in midbrain structures (inferior colliculus, ventrolateral tegmental area) and dorsal auditory cortex. At day 7, a region of increased uptake in blast-injured rats was found in a cluster centered on the cortex-amygdala transition zone, while no regions of decreased uptake were observed. These results suggest that a relatively mild primary blast injury results in altered brain metabolism in multiple brain regions and that post-injury time of assessment is an important factor in observing regional changes in [18F]FDG uptake.


Subject(s)
Brain Concussion/diagnostic imaging , Brain Concussion/physiopathology , Fluorodeoxyglucose F18/metabolism , Amygdala/metabolism , Animals , Blast Injuries/physiopathology , Brain/metabolism , Brain Injuries/metabolism , Corpus Callosum/metabolism , Male , Positron-Emission Tomography/methods , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley
8.
Front Neurol ; 10: 509, 2019.
Article in English | MEDLINE | ID: mdl-31178814

ABSTRACT

Traumatic brain injury (TBI) resulting from repeated head trauma is frequently characterized by diffuse axonal injury and long-term motor, cognitive and neuropsychiatric symptoms. Given the delay, often decades, between repeated head traumas and the presentation of symptoms in TBI patients, animal models of repeated injuries should be studied longitudinally to properly assess the longer-term effects of multiple concussive injuries on functional outcomes. In this study, male and cycling female C57BL/6J mice underwent repeated (three) concussive brain injuries (rCBI) delivered via a Leica ImpactOne cortical impact device and were assessed chronically on motor (open field and rotarod), cognitive (y-maze and active place avoidance), and neuropsychiatric (marble-burying, elevated zero maze and tail suspension) tests. Motor deficits were significant on the rotarod on the day following the injuries, and slight impairment remained for up to 6 months. All mice that sustained rCBI had significant cognitive deficits on the active place avoidance test and showed greater agitation (less immobility) in the tail suspension test. Only injured male mice were significantly hyperactive in the open field, and had increased time spent in the open quadrants of the elevated zero maze. One year after the injuries, mice of both sexes exhibited persistent pathological changes by the presence of Prussian blue staining (indication of prior microbleeds), primarily in the cortex at the site of the injury, and increased GFAP staining in the perilesional cortex and axonal tracts (corpus callosum and optic tracts). These data demonstrate that a pathological phenotype with motor, cognitive, and neuropsychiatric symptoms can be observed in an animal model of rCBI for at least one year post-injury, providing a pre-clinical setting for the study of the link between multiple brain injuries and neurodegenerative disorders. Furthermore, this is the first study to include both sexes in a pre-clinical long-term rCBI model, and female mice are less impaired functionally than males.

9.
Brain Res ; 1700: 138-151, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30176241

ABSTRACT

The employment of explosive weaponry in modern warfare exposes populations to shock wave-induced and impact-related brain injuries. Among the most common clinical complaints resulting from traumatic brain injury (TBI) are sleep-wake disturbances. The current study assessed the acute effects of mild concussive brain injury (CBI) and mild blast wave-induced brain injury (BTBI) on mouse behavior and orexin-A expression. Male C57BL/6J mice were exposed to CBI, BTBI, or sham procedures. Injured animals and their shams were further divided into the following subgroups: 24-h survival in standard group (SG) housing, 72-h survival in SG housing, and 72-h survival in Any-Maze cages (AMc). AMc enabled continuous monitoring of home cage activities. BTBI caused significant but transient decreases in wheel running and ingestive behaviors 24 h post-injury (PI), while CBI transiently decreased running and water intake. BTBI resulted in general hypoactivity in the open field (OF) at both PI time points for SG-housed animals. In contrast, CBI did not cause hypoactivity. Mice subjected to CBI traveled more in the center of the OF at both time points PI, suggesting that CBI caused reduced anxiety in mice. Increased activity in the center of the OF was also seen at 24 h PI after BTBI. CBI treatment caused increased CD11b immunostaining. However, neither injury was accompanied by an alteration in the number of orexin-A hypothalamic neurons. Taken together, shock wave exposure and concussive injury transiently reduced mouse activities, but some differences between the two injuries were seen.


Subject(s)
Blast Injuries/metabolism , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/metabolism , Motor Activity/physiology , Orexins/metabolism , Animals , Behavior, Animal/physiology , Blast Injuries/pathology , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Housing, Animal , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Random Allocation
10.
J Neurosci Res ; 96(4): 501-511, 2018 04.
Article in English | MEDLINE | ID: mdl-28675644

ABSTRACT

As a consequence of their commercial availability, ease of use, and reproducibility, controlled cortical impact (CCI) devices have attained significant prevalence in preclinical traumatic brain injury research. With a CCI, the severity of injury is controlled by varying the impact depth, velocity, and duration, but the actual performance of the device is not well appreciated, partly because of the velocity and short travel distance to impact. This study used a high-speed video digital camera to investigate the performance of five electromagnetically driven CCI devices of the same model. Videography indicated that the impactor tip made a series of distinctive vertical advances and retractions before it attained the desired preset depth; this was also observed in male mouse CCI tests. The impactor tip was also observed to move in the horizontal direction by .8-1.6 mm. On the first advance, the tip extended a distance that was shorter than the preset depth and the velocity of impactor tip was slightly faster than the preset values for three of the five machines. One of the devices was evaluated on four separate occasions over a 14-month period and was found to operate consistently over time. Overall, differences in impact depth and velocity between the devices were modest, suggesting that comparisons of experimental results from different laboratories will generally be informative, particularly if reports provide relevant descriptions of neuropathology. However, the repetitive extension and retraction and horizontal movement of the tip suggests caution in modeling CCI as a single injurious event.


Subject(s)
Brain Injuries, Traumatic , Equipment Design/instrumentation , Animals , Cerebral Cortex , Disease Models, Animal , Mice , Mice, Inbred C57BL , Reproducibility of Results , Video Recording
11.
Behav Brain Res ; 324: 115-124, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28214540

ABSTRACT

In contact sports, repetitive concussive brain injury (rCBI) is the prevalent form of head injury seen in athletes. The need for effective treatment is urgent as rCBI has been associated with a host of cognitive, behavioral and neurological complaints. There has been a growing trend in the use of female animals in pre-clinical research, but few studies have investigated possible sex differences following rCBI. The goal of the current study was to determine any differences between male and female C57BL/6J mice on assessments of learning and memory after repetitive concussive injury. Following rCBI by impact to the scalp, male mice exhibited longer righting reflexes during acute recovery. In both sexes, there were no evident histopathological changes observed in the underlying cerebral cortex or hippocampus. Reactive astrogliosis was elevated in the corpus callosum and optic tract, and astrogliosis was slightly less in the optic tract of female mice. rCBI mice exhibited impairment during the learning phase of the Morris water maze (MWM), but female mice, in comparison to male mice, were observed to have superior spatial memory during standard MWM probe trials. Female mice were overall more active, evidenced by greater distances traveled in the y-maze and greater swim speeds in the MWM. The results of this study demonstrate sex differences in cognitive performance following rCBI and support previous research suggesting the neuroprotective role of sex in brain injury.


Subject(s)
Brain Concussion/physiopathology , Cognition Disorders/physiopathology , Animals , Brain Concussion/complications , Brain Concussion/pathology , Cerebral Cortex/pathology , Cognition/physiology , Cognition Disorders/etiology , Cognition Disorders/pathology , Corpus Callosum/pathology , Female , Gliosis/pathology , Male , Maze Learning/physiology , Mice, Inbred C57BL , Motor Activity , Sex Characteristics , Spatial Memory/physiology
12.
J Neurotrauma ; 34(4): 890-905, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27149139

ABSTRACT

Psychiatric symptoms such as anxiety and depression are frequent and persistent complaints following traumatic brain injury (TBI). Modeling these symptoms in animal models of TBI affords the opportunity to determine mechanisms underlying behavioral pathologies and to test potential therapeutic agents. However, testing these symptoms in animal models of TBI has yielded inconsistent results. The goal of the current study was to employ a battery of tests to measure multiple anxiety- and depressive-like symptoms following TBI in C57BL/6J mice, and to determine if male and female mice are differentially affected by the injury. Following controlled cortical impact (CCI) at a parietal location, neither male nor female mice showed depressive-like symptoms as measured by the Porsolt forced-swim test and sucrose preference test. Conclusions regarding anxiety-like behaviors were dependent upon the assay employed; CCI-induced thigmotaxis in the open field suggested an anxiogenic effect of the injury; however, results from the elevated zero maze, light-dark box, and marble-burying tests indicated that CCI reduced anxiety-like behaviors. Fewer anxiety-like behaviors were also associated with the female sex. Increased levels of activity were also measured in female mice and injured mice in these tests, and conclusions regarding anxiety should be taken with caution when experimental manipulations induce changes in baseline activity. These results underscore the irreconcilability of results from studies attempting to model TBI-induced neuropsychiatric symptoms. Changes in injury models or better attempts to replicate the clinical syndrome may improve the translational applicability of rodent models of TBI-induced anxiety and depression.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Brain Injuries, Traumatic/physiopathology , Depression/physiopathology , Disease Models, Animal , Animals , Female , Male , Mice , Mice, Inbred C57BL
13.
J Neurotrauma ; 33(9): 880-94, 2016 05 01.
Article in English | MEDLINE | ID: mdl-25951234

ABSTRACT

To date, clinical trials have failed to find an effective therapy for victims of traumatic brain injury (TBI) who live with motor, cognitive, and psychiatric complaints. Pre-clinical investigators are now encouraged to include male and female subjects in all translational research, which is of particular interest in the field of neurotrauma given that circulating female hormones (progesterone and estrogen) have been demonstrated to exert neuroprotective effects. To determine whether behavior of male and female C57BL6/J mice is differentially impaired by TBI, male and cycling female mice were injured by controlled cortical impact and tested for several weeks with functional assessments commonly employed in pre-clinical research. We found that cognitive and motor impairments post-TBI, as measured by the Morris water maze (MWM) and rotarod, respectively, were largely equivalent in male and female animals. However, spatial working memory, assessed by the y-maze, was poorer in female mice. Female mice were generally more active, as evidenced by greater distance traveled in the first exposure to the open field, greater distance in the y-maze, and faster swimming speeds in the MWM. Statistical analysis showed that variability in all behavioral data was no greater in cycling female mice than it was in male mice. These data all suggest that with careful selection of tests, procedures, and measurements, both sexes can be included in translational TBI research without concern for effect of hormones on functional impairments or behavioral variability.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Cognition/physiology , Maze Learning/physiology , Motor Activity/physiology , Psychomotor Performance/physiology , Animals , Biomedical Research , Brain Injuries, Traumatic/psychology , Female , Male , Mice , Mice, Inbred C57BL , Random Allocation , Rotarod Performance Test/methods , Rotarod Performance Test/psychology , Sex Factors
14.
Neuropharmacology ; 85: 427-39, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24937045

ABSTRACT

Traumatic brain injury (TBI) is the leading cause of death in young adults in the United States, but there is still no effective agent for treatment. N-arachidonoylethanolamine (anandamide, AEA) is a major endocannabinoid in the brain. Its increase after brain injury is believed to be protective. However, the compensatory role of AEA is transient due to its rapid hydrolysis by the fatty acid amide hydrolase (FAAH). Thus, inhibition of FAAH can boost the endogenous levels of AEA and prolong its protective effect. Using a TBI mouse model, we found that post-injury chronic treatment with PF3845, a selective and potent FAAH inhibitor, reversed TBI-induced impairments in fine motor movement, hippocampus dependent working memory and anxiety-like behavior. Treatment with PF3845 inactivated FAAH activity and enhanced the AEA levels in the brain. It reduced neurodegeneration in the dentate gyrus, and up-regulated the expression of Bcl-2 and Hsp70/72 in both cortex and hippocampus. PF3845 also suppressed the increased production of amyloid precursor protein, prevented dendritic loss and restored the levels of synaptophysin in the ipsilateral dentate gyrus. Furthermore, PF3845 suppressed the expression of inducible nitric oxide synthase and cyclooxygenase-2 and enhanced the expression of arginase-1 post-TBI, suggesting a shift of microglia/macrophages from M1 to M2 phenotype. The effects of PF3845 on TBI-induced behavioral deficits and neurodegeneration were mediated by activation of cannabinoid type 1 and 2 receptors and might be attributable to the phosphorylation of ERK1/2 and AKT. These results suggest that selective inhibition of FAAH is likely to be beneficial for TBI treatment.


Subject(s)
Brain Injuries/drug therapy , Neuroimmunomodulation/drug effects , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Recovery of Function/drug effects , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Animals , Anxiety/drug therapy , Anxiety/pathology , Anxiety/physiopathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain Injuries/psychology , Caspase 3/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Neuroimmunomodulation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Recovery of Function/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...