Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Neurotrauma ; 38(5): 628-645, 2021 03.
Article in English | MEDLINE | ID: mdl-33203303

ABSTRACT

Glibenclamide (GLY) is the sixth drug tested by the Operation Brain Trauma Therapy (OBTT) consortium based on substantial pre-clinical evidence of benefit in traumatic brain injury (TBI). Adult Sprague-Dawley rats underwent fluid percussion injury (FPI; n = 45), controlled cortical impact (CCI; n = 30), or penetrating ballistic-like brain injury (PBBI; n = 36). Efficacy of GLY treatment (10-µg/kg intraperitoneal loading dose at 10 min post-injury, followed by a continuous 7-day subcutaneous infusion [0.2 µg/h]) on motor, cognitive, neuropathological, and biomarker outcomes was assessed across models. GLY improved motor outcome versus vehicle in FPI (cylinder task, p < 0.05) and CCI (beam balance, p < 0.05; beam walk, p < 0.05). In FPI, GLY did not benefit any other outcome, whereas in CCI, it reduced 21-day lesion volume versus vehicle (p < 0.05). On Morris water maze testing in CCI, GLY worsened performance on hidden platform latency testing versus sham (p < 0.05), but not versus TBI vehicle. In PBBI, GLY did not improve any outcome. Blood levels of glial fibrillary acidic protein and ubiquitin carboxyl terminal hydrolase-1 at 24 h did not show significant treatment-induced changes. In summary, GLY showed the greatest benefit in CCI, with positive effects on motor and neuropathological outcomes. GLY is the second-highest-scoring agent overall tested by OBTT and the only drug to reduce lesion volume after CCI. Our findings suggest that leveraging the use of a TBI model-based phenotype to guide treatment (i.e., GLY in contusion) might represent a strategic choice to accelerate drug development in clinical trials and, ultimately, achieve precision medicine in TBI.


Subject(s)
Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/drug therapy , Glyburide/therapeutic use , Hypoglycemic Agents/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
2.
Mol Neurobiol ; 55(3): 2454-2470, 2018 03.
Article in English | MEDLINE | ID: mdl-28365875

ABSTRACT

Traumatic brain injury (TBI) triggers endoplasmic reticulum (ER) stress and impairs autophagic clearance of damaged organelles and toxic macromolecules. In this study, we investigated the effects of the post-TBI administration of docosahexaenoic acid (DHA) on improving hippocampal autophagy flux and cognitive functions of rats. TBI was induced by cortical contusion injury in Sprague-Dawley rats, which received DHA (16 mg/kg in DMSO, intraperitoneal administration) or vehicle DMSO (1 ml/kg) with an initial dose within 15 min after the injury, followed by a daily dose for 3 or 7 days. First, RT-qPCR reveals that TBI induced a significant elevation in expression of autophagy-related genes in the hippocampus, including SQSTM1/p62 (sequestosome 1), lysosomal-associated membrane proteins 1 and 2 (Lamp1 and Lamp2), and cathepsin D (Ctsd). Upregulation of the corresponding autophagy-related proteins was detected by immunoblotting and immunostaining. In contrast, the DHA-treated rats did not exhibit the TBI-induced autophagy biogenesis and showed restored CTSD protein expression and activity. T2-weighted images and diffusion tensor imaging (DTI) of ex vivo brains showed that DHA reduced both gray matter and white matter damages in cortical and hippocampal tissues. DHA-treated animals performed better than the vehicle control group on the Morris water maze test. Taken together, these findings suggest that TBI triggers sustained stimulation of autophagy biogenesis, autophagy flux, and lysosomal functions in the hippocampus. Swift post-injury DHA administration restores hippocampal lysosomal biogenesis and function, demonstrating its therapeutic potential.


Subject(s)
Autophagy/physiology , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Docosahexaenoic Acids/therapeutic use , Hippocampus/metabolism , Lysosomes/physiology , Animals , Autophagy/drug effects , Brain Injuries, Traumatic/pathology , Docosahexaenoic Acids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Hippocampus/drug effects , Hippocampus/pathology , Lysosomes/drug effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
3.
J Neurotrauma ; 33(6): 553-66, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26671075

ABSTRACT

Operation Brain Trauma Therapy (OBTT) is a consortium of investigators using multiple pre-clinical models of traumatic brain injury (TBI) to bring acute therapies to clinical trials. To screen therapies, we used three rat models (parasagittal fluid percussion injury [FPI], controlled cortical impact [CCI], and penetrating ballistic-like brain injury [PBBI]). We report results of the third therapy (cyclosporin-A; cyclosporine; [CsA]) tested by OBTT. At each site, rats were randomized to treatment with an identical regimen (TBI + vehicle, TBI + CsA [10 mg/kg], or TBI + CsA [20 mg/kg] given intravenously at 15 min and 24 h after injury, and sham). We assessed motor and Morris water maze (MWM) tasks over 3 weeks after TBI and lesion volume and hemispheric tissue loss at 21 days. In FPI, CsA (10 mg/kg) produced histological protection, but 20 mg/kg worsened working memory. In CCI, CsA (20 mg/kg) impaired MWM performance; surprisingly, neither dose showed benefit on any outcome. After PBBI, neither dose produced benefit on any outcome, and mortality was increased (20 mg/kg) partly caused by the solvent vehicle. In OBTT, CsA produced complex effects with histological protection at the lowest dose in the least severe model (FPI), but only deleterious effects as model severity increased (CCI and PBBI). Biomarker assessments included measurements of glial fibrillary acidic protein (GFAP) and ubiquitin C-terminal hydrolase-L1 (UCH-L1) in blood at 4 or 24 h after injury. No positive treatment effects were seen on biomarker levels in any of the models, whereas significant increases in 24 h UCH-L1 levels were seen with CsA (20 mg/kg) after CCI and 24 h GFAP levels in both CsA treated groups in the PBBI model. Lack of behavioral protection in any model, indicators of toxicity, and a narrow therapeutic index reduce enthusiasm for clinical translation.


Subject(s)
Brain Injuries, Traumatic , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Recovery of Function/drug effects , Animals , Biomarkers/blood , Disease Models, Animal , Glial Fibrillary Acidic Protein/blood , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Ubiquitin Thiolesterase/blood
4.
J Neurotrauma ; 33(6): 606-14, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26671284

ABSTRACT

Operation Brain Trauma Therapy (OBTT) is a fully operational, rigorous, and productive multicenter, pre-clinical drug and circulating biomarker screening consortium for the field of traumatic brain injury (TBI). In this article, we synthesize the findings from the first five therapies tested by OBTT and discuss both the current work that is ongoing and potential future directions. Based on the results generated from the first five therapies tested within the exacting approach used by OBTT, four (nicotinamide, erythropoietin, cyclosporine A, and simvastatin) performed below or well below what was expected based on the published literature. OBTT has identified, however, the early post-TBI administration of levetiracetam as a promising agent and has advanced it to a gyrencephalic large animal model--fluid percussion injury in micropigs. The sixth and seventh therapies have just completed testing (glibenclamide and Kollidon VA 64), and an eighth drug (AER 271) is in testing. Incorporation of circulating brain injury biomarker assessments into these pre-clinical studies suggests considerable potential for diagnostic and theranostic utility of glial fibrillary acidic protein in pre-clinical studies. Given the failures in clinical translation of therapies in TBI, rigorous multicenter, pre-clinical approaches to therapeutic screening such as OBTT may be important for the ultimate translation of therapies to the human condition.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Drug Evaluation, Preclinical/trends , Neuroprotective Agents/therapeutic use , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Male , Neurology/methods , Neurology/trends , Rats , Rats, Sprague-Dawley
5.
ASN Neuro ; 7(6)2015.
Article in English | MEDLINE | ID: mdl-26685193

ABSTRACT

We investigated the effects of the administration of docosahexaenoic acid (DHA) post-traumatic brain injury (TBI) on reducing neuroinflammation. TBI was induced by cortical contusion injury in Sprague Dawley rats. Either DHA (16 mg/kg in dimethyl sulfoxide) or vehicle dimethyl sulfoxide (1 ml/kg) was administered intraperitonially at 5 min after TBI, followed by a daily dose for 3 to 21 days. TBI triggered activation of microglia or macrophages, detected by an increase of Iba1 positively stained microglia or macrophages in peri-lesion cortical tissues at 3, 7, and 21 days post-TBI. The inflammatory response was further characterized by expression of the proinflammatory marker CD16/32 and the anti-inflammatory marker CD206 in Iba1(+) microglia or macrophages. DHA-treated brains showed significantly fewer CD16/32(+) microglia or macrophages, but an increased CD206(+) phagocytic microglial or macrophage population. Additionally, DHA treatment revealed a shift in microglial or macrophage morphology from the activated, amoeboid-like state into the more permissive, surveillant state. Furthermore, activated Iba1(+) microglial or macrophages were associated with neurons expressing the endoplasmic reticulum (ER) stress marker CHOP at 3 days post-TBI, and the administration of DHA post-TBI concurrently reduced ER stress and the associated activation of Iba1(+) microglial or macrophages. There was a decrease in nuclear translocation of activated nuclear factor kappa-light-chain-enhancer of activated B cells protein at 3 days in DHA-treated tissue and reduced neuronal degeneration in DHA-treated brains at 3, 7, and 21 days after TBI. In summary, our study demonstrated that TBI mediated inflammatory responses are associated with increased neuronal ER stress and subsequent activation of microglia or macrophages. DHA administration reduced neuronal ER stress and subsequent association with microglial or macrophage polarization after TBI, demonstrating its therapeutic potential to ameliorate TBI-induced cellular pathology.


Subject(s)
Brain Injuries/drug therapy , Docosahexaenoic Acids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Macrophages/drug effects , Microglia/drug effects , Neuroprotective Agents/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Macrophage Activation/drug effects , Macrophage Activation/physiology , Macrophages/metabolism , Macrophages/pathology , Male , Microglia/metabolism , Microglia/pathology , NF-kappa B/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Time Factors , Treatment Outcome
6.
Restor Neurol Neurosci ; 33(2): 95-104, 2015.
Article in English | MEDLINE | ID: mdl-25624423

ABSTRACT

PURPOSE: In previous studies, collagen based matrices have been implanted into the site of lesion in different models of brain injury. We hypothesized that semisynthetic collagen matrix can have neuroprotective function in the setting of traumatic brain injury. METHODS: Rats were subjected to sham injury or controlled cortical impact. They either received extracellular matrix graft (DuraGen) over the injury site or did not receive any graft and underwent beam balance/beam walking test at post injury days 1-5 and Morris water maze at post injury days 14-18. Animals were sacrificed at day 18 for tissue analysis. RESULTS: Collagen matrix implantation in injured rats did not affect motor function (beam balance test: p = 0.627, beam walking test: p = 0.921). However, injured group with collagen matrix had significantly better spatial memory acquisition (p < 0.05). There was a significant reduction in lesion volume, as well as neuronal loss in CA1 (p < 0.001) and CA3 (p < 0.05) regions of the hippocampus in injured group with collagen matrix (p < 0.05). CONCLUSIONS: Collagen matrix reduces contusional lesion volume, neuronal loss, and cognitive deficit after traumatic brain injury. Further studies are needed to demonstrate the mechanisms of neuroprotection by collagen matrix.


Subject(s)
Brain Injuries/therapy , Collagen/pharmacology , Neuroprotective Agents/pharmacology , Tissue Scaffolds , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/pathology , CA3 Region, Hippocampal/physiopathology , Disease Models, Animal , Male , Maze Learning , Motor Activity , Neurons/drug effects , Neurons/physiology , Neurosurgical Procedures , Rats, Sprague-Dawley , Spatial Memory , Treatment Outcome
7.
J Neurosci ; 34(10): 3743-55, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24599472

ABSTRACT

In this study, we investigated the development of endoplasmic reticulum (ER) stress after traumatic brain injury (TBI) and the efficacy of post-TBI administration of docosahexaenoic acid (DHA) in reducing ER stress. TBI was induced by cortical contusion injury in Sprague-Dawley rats. Either DHA (16 mg/kg in DMSO) or vehicle DMSO (1 ml/kg) was administered intraperitoneally at 5 min after TBI, followed by a daily dose for 3-21 d. TBI triggered sustained expression of the ER stress marker proteins including phosphorylated eukaryotic initiation factor-2α, activating transcription factor 4, inositol requiring kinase 1, and C/EBP homologous protein in the ipsilateral cortex at 3-21 d after TBI. The prolonged ER stress was accompanied with an accumulation of abnormal ubiquitin aggregates and increased expression of amyloid precursor protein (APP) and phosphorylated tau (p-Tau) in the frontal cortex after TBI. The ER stress marker proteins were colocalized with APP accumulation in the soma. Interestingly, administration of DHA attenuated all ER stress marker proteins and reduced the accumulation of both ubiquitinated proteins and APP/p-Tau proteins. In addition, the DHA-treated animals exhibited early recovery of their sensorimotor function after TBI. In summary, our study demonstrated that TBI induces a prolonged ER stress, which is positively correlated with abnormal APP accumulation. The sustained ER stress may play a role in chronic neuronal damage after TBI. Our findings illustrate that post-TBI administration of DHA has therapeutic potentials in reducing ER stress, abnormal protein accumulation, and neurological deficits.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain Injuries/metabolism , Docosahexaenoic Acids/therapeutic use , Endoplasmic Reticulum Stress/physiology , Neurons/metabolism , tau Proteins/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Brain Injuries/drug therapy , Brain Injuries/pathology , Docosahexaenoic Acids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Frontal Lobe/pathology , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , tau Proteins/antagonists & inhibitors
8.
Brain Res ; 1548: 63-72, 2014 Feb 22.
Article in English | MEDLINE | ID: mdl-24389074

ABSTRACT

The interaction between the phosphatase calcineurin and transcription factor nuclear factor of activated T cells (NFAT) plays an important role numerous signaling and the regulatory events. Although NFAT is mostly known for its transcription function in the immune system, NFAT also has essential functions even in the central nervous system (CNS). The effects of traumatic brain injury (TBI) on NFAT are currently unknown. To determine if there is an alteration in NFAT after TBI, we examined NFATc3 and c4 levels at 6 h, 1 day, 1 week, 2 weeks and 4 weeks post injury. Rats were anesthetized and surgically prepared for controlled cortical impact (CCI) injury or sham surgery. Semi-quantitative measurements of NFATc3 and c4 in the hippocampal homogenates from injured and sham rats sacrificed at the appropriate time after injury were assessed using Western blot analysis. After TBI insult, in the hippocampus ipsilateral to the injury, NFATc3 expression levels were decreased both in the cytoplasmic and nuclear fractions. However, NFATc4 expression levels were increased in the cytoplasmic fraction but decreased in the nuclear fraction. Double labeling (with NeuN and GFAP) immunohistochemistry revealed that NFATc3 was expressed in subset of astrocytes and NFATc4 was expressed primarily in neurons. These differential responses in NFATc3 and c4 expression after TBI insult may indicate long-term changes in hippocampal excitability and may contribute to behavioral deficits. Further study is warranted to illustrate the role of NFATc3 and c4 in the setting of TBI.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Male , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley
9.
J Neurotrauma ; 30(11): 920-37, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23496248

ABSTRACT

Abstract Explosive blast-induced traumatic brain injury (TBI) is the signature insult in modern combat casualty care and has been linked to post-traumatic stress disorder, memory loss, and chronic traumatic encephalopathy. In this article we report on blast-induced mild TBI (mTBI) characterized by fiber-tract degeneration and axonal injury revealed by cupric silver staining in adult male rats after head-only exposure to 35 psi in a helium-driven shock tube with head restraint. We now explore pathways of secondary injury and repair using biochemical/molecular strategies. Injury produced ∼25% mortality from apnea. Shams received identical anesthesia exposure. Rats were sacrificed at 2 or 24 h, and brain was sampled in the hippocampus and prefrontal cortex. Hippocampal samples were used to assess gene array (RatRef-12 Expression BeadChip; Illumina, Inc., San Diego, CA) and oxidative stress (OS; ascorbate, glutathione, low-molecular-weight thiols [LMWT], protein thiols, and 4-hydroxynonenal [HNE]). Cortical samples were used to assess neuroinflammation (cytokines, chemokines, and growth factors; Luminex Corporation, Austin, TX) and purines (adenosine triphosphate [ATP], adenosine diphosphate, adenosine, inosine, 2'-AMP [adenosine monophosphate], and 5'-AMP). Gene array revealed marked increases in astrocyte and neuroinflammatory markers at 24 h (glial fibrillary acidic protein, vimentin, and complement component 1) with expression patterns bioinformatically consistent with those noted in Alzheimer's disease and long-term potentiation. Ascorbate, LMWT, and protein thiols were reduced at 2 and 24 h; by 24 h, HNE was increased. At 2 h, multiple cytokines and chemokines (interleukin [IL]-1α, IL-6, IL-10, and macrophage inflammatory protein 1 alpha [MIP-1α]) were increased; by 24 h, only MIP-1α remained elevated. ATP was not depleted, and adenosine correlated with 2'-cyclic AMP (cAMP), and not 5'-cAMP. Our data reveal (1) gene-array alterations similar to disorders of memory processing and a marked astrocyte response, (2) OS, (3) neuroinflammation with a sustained chemokine response, and (4) adenosine production despite lack of energy failure-possibly resulting from metabolism of 2'-3'-cAMP. A robust biochemical/molecular response occurs after blast-induced mTBI, with the body protected from blast and the head constrained to limit motion.


Subject(s)
Blast Injuries/metabolism , Brain Injuries/metabolism , Transcriptome , Animals , Blast Injuries/genetics , Blast Injuries/physiopathology , Brain Injuries/genetics , Brain Injuries/physiopathology , Disease Models, Animal , Gene Expression Profiling , Male , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Regeneration/physiology , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley
10.
J Neurotrauma ; 30(4): 259-70, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23094804

ABSTRACT

Experimental investigations into the effects of traumatic brain injury (TBI) have demonstrated significant alterations in dopaminergic systems. Dopaminergic fibers originating within the substantia nigra and ventral tegmental area (VTA) are important for reward learning, addiction, movement, and behavior. However, little is known about the effect of TBI on substantia nigra and VTA function. Environmental enrichment (EE) has been shown to improve functional outcome after TBI, and a number of studies suggest that it may exert some benefits via dopaminergic signaling. To better understand the role of dopamine in chronic TBI pathophysiology and the effect of EE, we examined the mRNA expression profile within the substantia nigra and VTA at 4 weeks post-injury. Specifically, three comparisons were made: 1) TBI versus sham, 2) sham+EE versus sham+standard (STD) housing, and 3) TBI+EE versus TBI+STD. There were differential expressions of 25, 4, and 40 genes in these comparisons, respectively. Chronic alterations in genes post-injury within the substantia nigra and VTA included genes important for cellular membrane homeostasis and transcription. EE-induced gene alterations after TBI included genes important for signal transduction, in particular calcium signaling pathways, membrane homeostasis, and metabolism. Elucidation of these alterations in gene expression within the substantia nigra and VTA provides new insights into chronic changes in dopamine signaling post-TBI, and the potential role of EE in TBI rehabilitation.


Subject(s)
Brain Injuries/genetics , Substantia Nigra/physiopathology , Transcriptome , Animals , Blotting, Western , Brain Injuries/physiopathology , Brain Injuries/rehabilitation , Calcium Signaling/physiology , Disease Models, Animal , Dopamine/biosynthesis , Dopamine/genetics , Environment , Housing, Animal , In Situ Hybridization , Male , Motor Activity/physiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
Exp Neurol ; 229(2): 300-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376040

ABSTRACT

Traumatic brain injury (TBI) causes persistent neurologic deficits. Current therapies, predominantly focused upon cortical and hippocampal cellular survival, have limited benefit on cognitive outcomes. Striatal damage is associated with deficits in executive function, learning, and memory. Dopamine and cAMP regulated phosphoprotein 32 (DARPP-32) is expressed within striatal medium spiny neurons and regulates striatal function. We found that controlled cortical impact injury in rats produces a chronic decrease in DARPP-32 phosphorylation at threonine-34 and an increase in protein phosphatase-1 activity. There is no effect of injury on threonine-75 phosphorylation or on DARPP-32 protein. Amantadine, shown to be efficacious in treating post-TBI cognitive deficits, given daily for two weeks is able to restore the loss of DARPP-32 phosphorylation and reduce protein phosphatase-1 activity. Amantadine also decreases the phosphorylation of threonine-75 consistent with activity as a partial N-methyl-D-aspartate (NMDA) receptor antagonist and partial dopamine agonist. These data demonstrate that targeting the DARPP-32 signaling cascade represents a promising novel therapeutic approach in the treatment of persistent deficits following a TBI.


Subject(s)
Brain Injuries/metabolism , Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Neurons/metabolism , Signal Transduction/physiology , Amantadine/pharmacology , Animals , Blotting, Western , Corpus Striatum/drug effects , Dopamine Agents/pharmacology , Fluorescent Antibody Technique , Immunohistochemistry , Male , Neurons/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
Brain Res ; 1358: 211-20, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20713027

ABSTRACT

Calcineurin subunit isoforms are implicated in long term potentiation, long term depression, and structural plasticity. Calcineurin inhibitors benefit axonal damage, cellular dysfunction, and cognitive outcomes in animal models of traumatic brain injury (TBI). Distribution of the catalytic calcineurin A subunit is altered and calcineurin activity increased following fluid percussion injury. Alterations in calcineurin subunit A isoform distribution within the hippocampus also occur post controlled cortical impact (CCI) demonstrating a reduction in catalytic subunit distribution in CA1-2 dendritic fields. Furthermore the effect of TBI on the regulatory subunit, calcineurin B, is unknown. Understanding the role of both subunits is necessary to effectively target alterations in calcineurin signaling as current calcineurin inhibitors, such as cyclosporin A and FK-506, rely upon binding sites on both subunits for complete inhibition. The effect of moderate CCI on the expression and distribution of calcineurin B isoforms within the hippocampus was examined at 2h and 2weeks post injury. Calcineurin B isoforms showed increased expression throughout the CA1 and CA2 while there was a decrease in expression within the ipsilateral dentate gyrus. Alterations in CnB isoform expression within the CA1, CA1-2, and dentate gyrus have significant implications for persistent hippocampal dysfunction following TBI. Regional changes in regulatory subunit expression may alter the effect of calcineurin inhibitors regionally following a traumatic brain injury.


Subject(s)
Brain Injuries/pathology , Calcineurin/metabolism , Gene Expression Regulation/physiology , Hippocampus/metabolism , Animals , Disease Models, Animal , Functional Laterality/physiology , Hippocampus/pathology , Male , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors
13.
J Neurotrauma ; 27(6): 1091-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20560755

ABSTRACT

Resveratrol (3,5,4'-trihydroxystilbene) is a plant-derived small molecule that is protective against multiple neurological and systemic insults. To date, no studies have explored the potential for resveratrol to provide behavioral protection in adult animals in the setting of traumatic brain injury (TBI). Using 50 male Sprague-Dawley rats, we employed the controlled cortical impact (CCI) model to ascertain whether post-injury administration of resveratrol would reduce the severity of the well-described cognitive and motor deficits associated with the model. Contusion volumes and hippocampal neuronal numbers were also measured to characterize the tissue and neuronal-sparing properties, respectively, of resveratrol. We found that 100 mg/kg, but not 10 mg/kg, of intraperitoneal resveratrol administered after injury provides significant behavioral protection in rats sustaining CCI. Specifically, rodents treated with 100 mg/kg of resveratrol showed improvements in motor performance (beam balance and beam walking) and testing of visuospatial memory (Morris water maze). Behavioral protection was correlated with significantly reduced contusion volumes, preservation of CA1 and CA3 hippocampal neurons, and protection from overt hippocampal loss as a result of incorporation into the overlying cortical contusion in resveratrol-treated animals. Although the mechanisms by which resveratrol mediates its neuroprotection is unclear, the current study adds to the growing literature identifying resveratrol as a potential therapy for human brain injury.


Subject(s)
Brain Injuries/drug therapy , Cerebral Cortex/drug effects , Hippocampus/drug effects , Maze Learning/drug effects , Motor Activity/drug effects , Neurons/drug effects , Stilbenes/pharmacology , Analysis of Variance , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Count , Cell Death/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Dose-Response Relationship, Drug , Hippocampus/pathology , Hippocampus/physiopathology , Male , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Resveratrol , Stilbenes/therapeutic use
14.
J Neurotrauma ; 27(1): 109-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19751097

ABSTRACT

Calcineurin (CaN) is a calcium/calmodulin-dependent phosphatase directly activated by calcium as a result of neuronal activation that is important for neuronal function. CaN subunit isoforms are implicated in long-term potentiation (LTP), long-term depression (LTD), and structural plasticity. CaN inhibitors are also beneficial to cognitive outcomes in animal models of traumatic brain injury (TBI). There are known changes in the CaN A (CnA) subunit following fluid percussion injury (FPI). The CnA subunit has two isoforms: CnAalpha and CnAbeta. The effect of moderate controlled cortical impact (CCI) on distribution of CnA isoforms was examined at 2 h and 2 weeks post-injury. CnA distribution was assayed by immunohistochemistry and graded for non-parametric analysis. Acutely CnA isoforms showed reduced immunoreactivity in stratum radiatum processes of the ipsilateral CA1 and CA1-2. There was also a significant alteration in the immunoreactivity of both CnA isoforms in the ipsilateral dentate gyrus, predominantly within the hidden blade. Alterations in CnA isoform regional distribution within the CA1, CA1-2, and dentate gyrus may have significant implications for persistent hippocampal dysfunction following TBI, including dysfunction in hippocampal plasticity. Understanding alterations in CnA isoform distribution may help improve the targeting of current therapeutic interventions and/or the development of new treatments for TBI.


Subject(s)
Brain Injuries/enzymology , Calcineurin/metabolism , Hippocampus/enzymology , Hippocampus/injuries , Animals , Brain Injuries/physiopathology , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/injuries , CA1 Region, Hippocampal/physiopathology , Calcium/metabolism , Calcium Signaling/physiology , Dentate Gyrus/enzymology , Dentate Gyrus/injuries , Dentate Gyrus/physiopathology , Down-Regulation/physiology , Hippocampus/physiopathology , Immunohistochemistry , Long-Term Potentiation/physiology , Male , Neuronal Plasticity/physiology , Neurons/enzymology , Protein Isoforms/metabolism , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley
15.
Crit Care Med ; 31(8): 2222-7, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12973183

ABSTRACT

OBJECTIVE: To test the hypothesis that etomidate treatment improves functional, cognitive, and histologic outcome after experimental traumatic brain injury. DESIGN: Controlled animal study. SETTING: University research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Traumatic brain injury was produced by controlled cortical impact injury (4 m/sec, 2.6 mm of tissue deformation). Etomidate (2 mg/kg) was administered intravenously immediately before injury (n = 13) or 5 mins after injury (n = 12). Additional rats received saline treatment 5 mins after injury (n = 12) or served as sham controls (n = 10). MEASUREMENTS AND MAIN RESULTS: Rats were evaluated on beam balance and beam walk tasks on postoperative days 1-5 and then trained in the Morris water maze on postoperative days 14-18. On day 28, the rats were killed, and hippocampal CA1 and CA3 neuron counts and cortical lesion volume were measured in histologic brain sections. Preinjury etomidate attenuated beam balance deficits, water maze deficits, hippocampal CA3 neuronal loss, and cortical tissue loss but did not attenuate beam walk deficits or hippocampal CA1 neuronal loss. Postinjury etomidate attenuated water maze deficits, but it did not affect any other outcome measure. CONCLUSIONS: Administration of etomidate both before and after injury attenuates secondary injury resulting from traumatic brain injury, but the effect is more pronounced with pretreatment. The ineffectiveness of postinjury etomidate on motor and histologic tasks suggests a brief therapeutic treatment window in rats. However, the treatment window in humans is unknown. Lastly, postinjury etomidate did not exacerbate neurologic or histologic outcome.


Subject(s)
Brain Injuries/physiopathology , Cognition/drug effects , Etomidate/pharmacology , Hypnotics and Sedatives/pharmacology , Animals , Disease Models, Animal , Etomidate/administration & dosage , Hypnotics and Sedatives/administration & dosage , Infusions, Intravenous , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
16.
Brain Res ; 937(1-2): 22-31, 2002 May 24.
Article in English | MEDLINE | ID: mdl-12020858

ABSTRACT

Traumatic injury to the central nervous system initiates inflammatory processes such as the synthesis of proinflammatory mediators that contribute to secondary tissue damage. Hence, administration of anti-inflammatory cytokines, such as interleukin-10 (IL-10) may be neuroprotective. Moderate hypothermia (30-32 degrees C) also decreases the pro-inflammatory response to traumatic brain injury (TBI). Thus, we hypothesized that the combination of IL-10 and hypothermia would provide synergistic neuroprotective effects after TBI. To test this hypothesis, fifty isoflurane-anesthetized rats underwent a controlled cortical impact (2.7 mm tissue deformation at 4 m/s) or sham injury and then were randomly assigned to one of five conditions (TBI/VEH Normothermia (37 degrees C), TBI/VEH Hypothermia (32 degrees C for 3 h), TBI/IL-10 Normothermia, TBI/IL-10 Hypothermia, and Sham/VEH Normothermia). Human IL-10 (5 microg) or VEH was administered (i.p.) 30 min after surgery. Function was assessed by established motor and cognitive tests on post-operative days 1-5 and 14-18, respectively. Cortical lesion volume and hippocampal CA(1)/CA(3) cell survival were quantified at 4 weeks. Brain sections from 15 additional rats were immunohistochemically assessed (MoAB RP-3) to determine neutrophil accumulation at 5 h after TBI. The administration of IL-10 after TBI produced an approximately 75% reduction in the number of RP-3-positive cells in both the normothermic and hypothermic groups vs. the normothermic vehicle-treated group (P<0.05), but did not improve functional outcome. In contrast, hypothermia alone enhanced both motor and cognitive function and increased CA(3) neuronal survival after TBI. Contrary to our hypothesis, systemic administration of IL-10 combined with hypothermia did not provide synergistic neuroprotective effects after TBI. Rather, IL-10 administration suppressed the beneficial effects produced by hypothermia alone after TBI. The mechanism(s) for the negative effects of IL-10 combined with hypothermia after TBI remain to be determined.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Brain Injuries/therapy , Hypothermia, Induced , Interleukin-10/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain Injuries/pathology , Cell Survival , Cerebral Cortex/drug effects , Cerebral Cortex/injuries , Cerebral Cortex/pathology , Chemotaxis, Leukocyte/drug effects , Hippocampus/drug effects , Hippocampus/injuries , Hippocampus/pathology , Interleukin-10/pharmacology , Interleukin-10/physiology , Male , Maze Learning/drug effects , Motor Activity/drug effects , Neurons/pathology , Postural Balance/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Recombinant Proteins/toxicity , Swimming , Vestibular Function Tests
17.
Restor Neurol Neurosci ; 14(4): 285-294, 1999.
Article in English | MEDLINE | ID: mdl-12671249

ABSTRACT

Amantadine, a dopamine agonist, is reported to have beneficial effects on the neurobehavioral sequelae of clinical brain injury. However, there are currently no published laboratory reports on its use in the assessment of functional or histopathological outcome following experimental traumatic brain injury (TBI). To this end, we examined the effects of daily amantadine treatment on functional recovery (motor and Morris water maze performance) and hippocampal neuronal survival following controlled cortical impact (CCI) injury (4 meters/sec, 2.7 mm tissue deformation). Male Sprague-Dawley rats were pretrained on motor performance tasks (beam balance and beam walking) one day prior to injury and tested on post-operative days 1-5. Additionally, all subjects were trained on the Morris water maze on post-operative days 14-18. Beginning one day after CCI injury or sham surgery, animals were injected once daily for 18 days with either amantadine (10 mg/kg, i.p.) or saline. The amantadine treatment regimen was ineffective in promoting motor recovery and increasing survival of hippocampal neurons in both the CA1 and CA3 regions following TBI, but did show improved swim latencies during the five days of water maze testing (Day 14 vs. Day 18, p < 0.01) when compared to saline controls. Mean (+/- SE) swim latencies on Day 18 were 15.12 +/- 2.8, 13.25 +/- 4.18, 70.83 +/- 11.1, and 38.5 +/- 3.55 sec for the sham/saline, sham/amantadine, injured/saline, and injured/amantadine treatment groups, respectively. Thus, while the daily administration of amantadine exhibited a neutral effect on motor behavior, it produced a modest attenuation of water maze performance deficits. This latter finding is consistent with published clinical data suggesting a beneficial effect on functional outcome with amantadine therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...