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










Publication year range
1.
J Neuroinflammation ; 13(1): 270, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27737716

ABSTRACT

BACKGROUND: Epilepsy patients have distinct immune/inflammatory cell profiles and inflammatory mediator levels in the blood. Although the neural origin of inflammatory cells and mediators has been implied, few studies have measured these inflammatory components in the human brain itself. This study examines the brain levels of chemokines (8), cytokines (14), and vascular injury mediators (3) suspected of being altered in epilepsy. METHODS: Soluble protein extracts of fresh frozen resected hippocampus, entorhinal cortex, and temporal cortex from 58 medically refractory mesial temporal lobe epilepsy subjects and 4 nonepileptic neurosurgical subjects were assayed for 25 inflammation-related mediators using ultrasensitive low-density arrays. RESULTS: Brain mediator levels were compared between regions and between epileptic and nonepileptic cases, showing a number of regional and possible epilepsy-associated differences. Eotaxin, interferon-γ, interleukin (IL)-2, IL-4, IL-12 p70, IL-17A, tumor necrosis factor-α, and intercellular adhesion molecule (ICAM)-1 levels were highest in the hippocampus, the presumptive site of epileptogenesis. Surprisingly, IL-1ß and IL-1α were lowest in the hippocampus, compared to cortical regions. In the temporal cortex, IL-1ß, IL-8, and MIP-1α levels were highest, compared to the entorhinal cortex and the hippocampus. The most pronounced epilepsy-associated differences were decreased levels of eotaxin, IL-1ß, C-reactive protein, and vascular cell adhesion molecule (VCAM)-1 and increased IL-12 p70 levels. Caution must be used in interpreting these results, however, because nonepileptic subjects were emergent neurosurgical cases, not a control group. Correlation analyses of each mediator in each brain region yielded valuable insights into the regulation of these mediator levels in the brain. Over 70 % of the associations identified were between different mediators in a single brain region, providing support for local control of mediator levels. Correlations of different mediators in different brain regions suggested more distributed control mechanisms, particularly in the hippocampus. Interestingly, only four mediators showed robust correlations between the brain regions, yet levels in three of these were significantly different between regions, indicating both global and local controls for these mediators. CONCLUSIONS: Both brain region-specific and epilepsy-associated changes in inflammation-related mediators were detected. Correlations in mediator levels within and between brain regions indicated local and global regulation, respectively. The hippocampus showed the majority of interregional associations, suggesting a focus of inflammatory control between these regions.


Subject(s)
Brain/metabolism , Drug Resistant Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Inflammation Mediators/metabolism , Adult , Brain/pathology , Brain/surgery , Drug Resistant Epilepsy/pathology , Drug Resistant Epilepsy/surgery , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Female , Humans , Male
2.
BMC Genomics ; 14: 303, 2013 May 04.
Article in English | MEDLINE | ID: mdl-23642095

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) induces arachidonic acid (ArA) release from cell membranes. ArA metabolites form a class of over 50 bioactive eicosanoids that can induce both adaptive and/or maladaptive brain responses. The dynamic metabolism of ArA to eicosanoids, and how they affect the injured brain, is poorly understood due to their diverse activities, trace levels, and short half-lives. The eicosanoids produced in the brain postinjury depend upon the enzymes present locally at any given time. Eicosanoids are synthesized by heme-containing enzymes, including cyclooxygenases, lipoxygenases, and arachidonate monoxygenases. The latter comprise a subset of the cytochrome P450 "Cyp" gene family that metabolize fatty acids, steroids, as well as endogenous and exogenous toxicants. However, for many of these genes neither baseline neuroanatomical nor injury-related temporal expression have been studied in the brain.In a rat model of parietal cortex TBI, Cyp and eicosanoid-related mRNA levels were determined at 6 h, 24 h, 3d, and 7d postinjury in parietal cortex and hippocampus, where dynamic changes in eicosanoids have been observed. Quantitative real-time polymerase chain reaction with low density arrays were used to assay 62 rat Cyps, 37 of which metabolize ArA or other unsaturated fatty acids; 16 eicosanoid-related enzymes that metabolize ArA or its metabolites; 8 eicosanoid receptors; 5 other inflammatory- and recovery-related genes, plus 2 mouse Cyps as negative controls and 3 highly expressed "housekeeping" genes. RESULTS: Sixteen arachidonate monoxygenases, 17 eicosanoid-related genes, and 12 other Cyps were regulated in the brain postinjury (p < 0.05, Tukey HSD). Discrete tissue levels and distinct postinjury temporal patterns of gene expression were observed in hippocampus and parietal cortex. CONCLUSIONS: The results suggest complex regulation of ArA and other lipid metabolism after TBI. Due to the temporal nature of brain injury-induced Cyp gene induction, manipulation of each gene (or its products) at a given time after TBI will be required to assess their contributions to secondary injury and/or recovery. Moreover, a better understanding of brain region localization and cell type-specific expression may be necessary to deduce the role of these eicosanoid-related genes in the healthy and injured brain.


Subject(s)
Brain Injuries/genetics , Cytochrome P-450 Enzyme System/metabolism , Eicosanoids/metabolism , Transcriptome , Animals , Brain Injuries/enzymology , Brain Injuries/physiopathology , Cytochrome P-450 Enzyme System/genetics , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice , Parietal Lobe/metabolism , Parietal Lobe/physiopathology , Polymerase Chain Reaction , Rats , Recovery of Function , Steroids/metabolism
3.
Prostaglandins Other Lipid Mediat ; 104-105: 18-24, 2013.
Article in English | MEDLINE | ID: mdl-22922090

ABSTRACT

After traumatic brain injury (TBI), arachidonic acid (ArA) is released from damaged cell membranes and metabolized to many bioactive eicosanoids, including several epoxyeicosatrienoic acids (EETs). Soluble epoxide hydrolase (Ephx2, sEH) appears to be the predominant pathway for EET metabolism to less active dihydroxyeicosatrienoates (DHETs). Prior studies indicate that brain levels of EETs increase transiently after TBI and EETs have antiinflammatory and neuroprotective activities which may benefit the injured brain. If the net effect of increased EET levels in the injured brain is beneficial to recovery, then Ephx2 gene disruption would be expected to enhance elevated EET levels and improve recovery in the injured brain. Thus, Ephx2-KO (Ephx2(-/-) bred onto pure C57Bl/6 background) mice were compared to wild-type controls in a unilateral controlled cortical impact model of TBI. Before injury, animals behaved comparably in open field activity and neurologic reflexes. Interestingly, the Ephx2-KO mice showed improved motor coordination on a beam walk task, yet showed indications of defective learning in a test of working spatial memory. After surgery, brain-injured Ephx2-KO mice again had less of a deficit in the beam walk than wild-type, and the difference in latency (post-pre) showed a trend of protection for Ephx2-KO mice after TBI. Brain-injured mice showed no genotype differences in working memory. Surprisingly, sham-operated Ephx2-KO mice exhibited an injured phenotype for working memory, compared to sham-operated wild-type mice. Brain eicosanoid levels were measured using liquid chromatography with tandem mass spectrometry. Of the 20 eicosanoids evaluated, only 8,9-EET was elevated in the Ephx2-KO cerebral cortex (37 d post-surgery, in both sham and injured). Tissue DHET levels were below the limit of quantification. These results reflect a significant contribution of sEH deficiency in coordination of ambulatory movements and working spatial memory in the mouse. Further investigation of differential sEH expression and EET levels at earlier time points and across other brain regions may shed light on these behavioral differences.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Behavior, Animal , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Epoxide Hydrolases/deficiency , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Brain Chemistry , Brain Injuries/genetics , Brain Injuries/physiopathology , Cerebral Cortex/physiopathology , Epoxide Hydrolases/genetics , Female , Gene Expression , Genotype , Male , Memory , Mice , Mice, Knockout , Phenotype
4.
Neurobiol Aging ; 33(9): 1857-73, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22088680

ABSTRACT

This proteomic study investigates the widely observed clinical phenomenon, that after comparable brain injuries, geriatric patients fare worse and recover less cognitive and neurologic function than younger victims. Utilizing a rat traumatic brain injury model, sham surgery or a neocortical contusion was induced in 3 age groups. Geriatric (21 months) rats performed worse on behavioral measures than young adults (12-16 weeks) and juveniles (5-6 weeks). Motor coordination and certain cognitive deficits showed age-dependence both before and after injury. Brain proteins were analyzed using silver-stained two-dimensional electrophoresis gels. Spot volume changes (>2-fold change, p<0.01) were identified between age and injury groups using computer-assisted densitometry. Sequences were determined by mass spectrometry of tryptic peptides. The 19 spots identified represented 13 different genes that fell into 4 general age- and injury-dependent expression patterns. Fifteen isoforms changed differentially with respect to both age and injury (p<0.05). Further investigations into the nature and function of these isoforms may yield insights into the vulnerability of older patients and resilience of younger patients in recovery after brain injuries.


Subject(s)
Aging , Brain Injuries/pathology , Gene Expression Regulation/physiology , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Age Factors , Analysis of Variance , Animals , Brain Injuries/complications , Cognition Disorders/diagnosis , Cognition Disorders/etiology , Electrophoresis, Gel, Two-Dimensional , Male , Mass Spectrometry , Maze Learning/physiology , Motor Activity/physiology , Nervous System Diseases/etiology , Proteomics/methods , Rats , Rats, Sprague-Dawley
6.
J Neurotrauma ; 27(3): 515-25, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19968558

ABSTRACT

Polyamines spermine and spermidine are highly regulated, ubiquitous aliphatic cations that maintain DNA structure and function as immunomodulators and as antioxidants. Polyamine homeostasis is disrupted after brain injuries, with concomitant generation of toxic metabolites that may contribute to secondary injuries. To test the hypothesis of increased brain polyamine catabolism after traumatic brain injury (TBI), we determined changes in catabolic enzymes and polyamine levels in the rat brain after lateral controlled cortical impact TBI. Spermine oxidase (SMO) catalyzes the degradation of spermine to spermidine, generating H2O2 and aminoaldehydes. Spermidine/spermine-N(1)-acetyltransferase (SSAT) catalyzes acetylation of these polyamines, and both are further oxidized in a reaction that generates putrescine, H2O2, and aminoaldehydes. In a rat cortical impact model of TBI, SSAT mRNA increased subacutely (6-24 h) after TBI in ipsilateral cortex and hippocampus. SMO mRNA levels were elevated late, from 3 to 7 days post-injury. Polyamine catabolism increased as well. Spermine levels were normal at 6 h and decreased slightly at 24 h, but were normal again by 72 h post-injury. Spermidine levels also decreased slightly (6-24 h), then increased by approximately 50% at 72 h post-injury. By contrast, normally low putrescine levels increased up to sixfold (6-72 h) after TBI. Moreover, N-acetylspermidine (but not N-acetylspermine) was detectable (24-72 h) near the site of injury, consistent with increased SSAT activity. None of these changes were seen in the contralateral hemisphere. Immunohistochemical confirmation indicated that SSAT and SMO were expressed throughout the brain. SSAT-immunoreactivity (SSAT-ir) increased in both neuronal and nonneuronal (likely glial) populations ipsilateral to injury. Interestingly, bilateral increases in cortical SSAT-ir neurons occurred at 72 h post-injury, whereas hippocampal changes occurred only ipsilaterally. Prolonged increases in brain polyamine catabolism are the likely cause of loss of homeostasis in this pathway. The potential for simple therapeutic interventions (e.g., polyamine supplementation or inhibition of polyamine oxidation) is an exciting implication of these studies.


Subject(s)
Biogenic Polyamines/metabolism , Brain Chemistry/physiology , Brain Injuries/metabolism , Brain/metabolism , Metabolism/physiology , Acetyltransferases/analysis , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Biomarkers/metabolism , Brain/physiopathology , Brain Injuries/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Functional Laterality/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Immunohistochemistry , Neurons/metabolism , Organ Culture Techniques , Oxidoreductases Acting on CH-NH Group Donors/analysis , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spermidine/analysis , Spermidine/metabolism , Spermine/analysis , Spermine/metabolism , Time Factors , Up-Regulation/physiology , Polyamine Oxidase
7.
J Neurosci Methods ; 168(2): 431-42, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18164073

ABSTRACT

A refined battery of neurological tests, SNAP (Simple Neuroassessment of Asymmetric Impairment), was developed and validated to efficiently assess neurological deficits induced in a mouse model of traumatic brain injury. Four to 7-month old mice were subjected to unilateral controlled cortical impact or sham injury (craniectomy only). Several behavioral tests (SNAP, beam walk, foot fault, and water maze) were used to assess functional deficits. SNAP was unique among these in that it required no expensive equipment and was performed in less than 5 min per mouse. SNAP demonstrated a high level of sensitivity and specificity as determined by receiver-operator characteristics curve analysis. Interrater reliability was good, as determined by Cohen's Kappa method and by comparing the sensitivity and specificity across various raters. SNAP detected deficits in proprioception, visual fields, and motor strength in brain-injured mice at 3 days, and was sensitive enough to detect magnitude and recovery of injury. The contribution of individual battery components changed as a function of time after injury, however, each was important to the overall SNAP score. SNAP provided a sensitive, reliable, time-efficient and cost-effective means of assessing neurological deficits in mice after unilateral brain injury.


Subject(s)
Brain Injuries/psychology , Cerebral Cortex/injuries , Animals , Behavior, Animal/physiology , Brain Injuries/genetics , Brain Injuries/pathology , Cerebral Cortex/pathology , Chronic Disease , Data Interpretation, Statistical , Functional Laterality/physiology , Genotype , Hand Strength/physiology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Neurologic Examination , Observer Variation , Postural Balance/physiology , Psychomotor Performance/physiology , Reproducibility of Results , Videotape Recording
8.
Brain Behav Immun ; 22(3): 285-98, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17996418

ABSTRACT

Overexpression of COX2 appears to be both a marker and an effector of neural damage after a variety of acquired brain injuries, and in natural or pathological aging of the brain. COX2 inhibitors may be neuroprotective in the brain by reducing prostanoid and free radical synthesis, or by directing arachidonic acid down alternate metabolic pathways. The arachidonic acid shunting hypothesis proposes that COX2 inhibitors' neuroprotective effects may be mediated by increased formation of potentially beneficial eicosanoids. Under conditions where COX2 activity is inhibited, arachidonic acid accumulates or is converted to eicosanoids via lipoxygenases and cytochrome P450 (CYP) epoxygenases. Several P450 eicosanoids have been demonstrated to have beneficial effects in the brain and/or periphery. We suspect that arachidonic acid shunting may be as important to functional recovery after brain injuries as altered prostanoid formation per se. Thus, COX2 inhibition and arachidonic acid shunting have therapeutic implications beyond the suppression of prostaglandin synthesis and free radical formation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Injuries/metabolism , Brain/drug effects , Brain/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Animals , Arachidonic Acid/metabolism , Eicosanoids/metabolism , Free Radicals/antagonists & inhibitors , Humans , Prostaglandins/metabolism
9.
J Pharm Biomed Anal ; 43(3): 1122-34, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17125954

ABSTRACT

A sensitive, specific, and robust liquid chromatography/mass spectrometric (LC/MS) method was developed and validated that allows simultaneous analysis of arachidonic acid (AA) and its cyclooxygenase, cytochrome P450, and lipoxygenase pathway metabolites prostaglandins (PGs), dihydroxyeicosatrienoic acids (DiHETrEs), hydroxyeicosatetraenoic acids (HETEs) and epoxyeicosatrienoic acids (EETs), including PGF(2alpha), PGE(2), PGD(2), PGJ(2), 14,15-DiHETrE, 11,12-DiHETrE, 8,9-DiHETrE, 5,6-DiHETrE, 20-HETE, 15-HETE, 12-HETE, 9-HETE, 8-HETE, 5-HETE, 14,15-EET, 11,12-EET, 8,9-EET, and 5,6-EET in rat brain tissues. Deuterium labeled PGF(2alpha)-d(4), PGD(2)-d(4), 15(S)-HETE-d(8), 14,15-EET-d(8), 11,12-EET-d(8), 8,9-EET-d(8), and AA-d(8) were used as internal standards. Solid phase extraction was used for sample preparation. A gradient LC/MS method using a C18 column and electrospray ionization source under negative ion mode was optimized for the best sensitivity and separation within 35 min. The method validation, including LC/MS instrument qualification, specificity, calibration model, accuracy, precision (without brain matrix and with brain matrix), and extraction efficiency were performed. The linear ranges of the calibration curves were 2-1000 pg for PGs, DiHETrEs, HETEs, and EETs, 10-2400 pg for PGE(2) and PGD(2), and 20-2000 ng for AA, respectively.


Subject(s)
Arachidonic Acid/analysis , Arachidonic Acids/analysis , Brain Chemistry/drug effects , Eicosanoids/analysis , Hydroxyeicosatetraenoic Acids/analysis , Prostaglandins/analysis , Animals , Calibration , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , Lipoxygenase/metabolism , Male , Mass Spectrometry , Prostaglandin-Endoperoxide Synthases/metabolism , Quality Control , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results
10.
J Neurosci Res ; 80(1): 104-13, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15744743

ABSTRACT

N-Methyl-D-aspartate (NMDA) at a subtoxic concentration (100 microM) promotes neuronal survival against glutamate-mediated excitotoxicity via a brain-derived neurotrophic factor (BDNF) autocrine loop in cultured cerebellar granule cells. The signal transduction mechanism(s) underlying NMDA neuroprotection, however, remains elusive. The mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3 kinase (PI3-K) pathways alter gene expression and are involved in synaptic plasticity and neuronal survival. This study tested whether neuroprotective activation of NMDA receptors, together with TrkB receptors, coactivated the MAPK or PI3-K pathways to protect rat cerebellar neurons. NMDA receptor activation caused a concentration- and time-dependent activation of MAPK lasting 24 hr. This activation was blocked by the NMDA receptor antagonist MK-801 but was attenuated only partially by the tyrosine kinase inhibitor k252a, suggesting that activation of both NMDA and TrkB receptors are required for maximal neuroprotection. The MAPK kinase (MEK) inhibitor U0126 (10 microM) partially blocked NMDA neuroprotection, whereas LY294002, a selective inhibitor of the PI3-K pathway, did not affect the neuroprotective activity of NMDA. Glutamate excitotoxicity decreased bcl-2, bcl-X(L), and bax mRNA levels,. NMDA increases Bcl-2 and Bcl-X(L) protein levels and decreases Bax protein levels. NMDA and TrkB receptor activation thus converge on the extracellular signal-regulated kinase (ERK) 1/2 signaling pathway to protect neurons against glutamate-mediated excitotoxicity. By increasing antiapoptotic proteins of the Bcl-2 family, NMDA receptor activation may also promote neuronal survival by preventing apoptosis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Glutamic Acid/metabolism , N-Methylaspartate/pharmacology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Receptor, trkB/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Carbazoles/pharmacology , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Expression/drug effects , Glutamic Acid/pharmacology , Indole Alkaloids , Mitogen-Activated Protein Kinase Kinases/drug effects , Morpholines/pharmacology , Neurons/drug effects , Nitriles/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Signal Transduction , bcl-2-Associated X Protein , bcl-X Protein
11.
Neurosurgery ; 56(3): 590-604, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15730585

ABSTRACT

OBJECTIVE: Increases in brain cyclooxygenase-2 (COX2) are associated with the central inflammatory response and with delayed neuronal death, events that cause secondary insults after traumatic brain injury. A growing literature supports the benefit of COX2-specific inhibitors in treating brain injuries. METHODS: DFU [5,5-dimethyl-3(3-fluorophenyl)-4(4-methylsulfonyl)phenyl-2(5)H)-furanone] is a third-generation, highly specific COX2 enzyme inhibitor. DFU treatments (1 or 10 mg/kg intraperitoneally, twice daily for 3 d) were initiated either before or after traumatic brain injury in a lateral cortical contusion rat model. RESULTS: DFU treatments initiated 10 minutes before injury or up to 6 hours after injury enhanced functional recovery at 3 days compared with vehicle-treated controls. Significant improvements in neurological reflexes and memory were observed. DFU initiated 10 minutes before injury improved histopathology and altered eicosanoid profiles in the brain. DFU 1 mg/kg reduced the rise in prostaglandin E2 in the brain at 24 hours after injury. DFU 10 mg/kg attenuated injury-induced COX2 immunoreactivity in the cortex (24 and 72 h) and hippocampus (6 and 72 h). This treatment also decreased the total number of activated caspase-3-immunoreactive cells in the injured cortex and hippocampus, significantly reducing the number of activated caspase-3-immunoreactive neurons at 72 hours after injury. DFU 1 mg/kg amplified potentially anti-inflammatory epoxyeicosatrienoic acid levels by more than fourfold in the injured brain. DFU 10 mg/kg protected the levels of 2-arachidonoyl glycerol, a neuroprotective endocannabinoid, in the injured brain. CONCLUSION: These improvements, particularly when treatment began up to 6 hours after injury, suggest exciting neuroprotective potential for COX2 inhibitors in the treatment of traumatic brain injury and support the consideration of Phase I/II clinical trials.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries/drug therapy , Cyclooxygenase Inhibitors/therapeutic use , Furans/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arachidonic Acids/analysis , Ataxia/drug therapy , Ataxia/etiology , Brain Chemistry , Brain Injuries/complications , Brain Injuries/psychology , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cyclooxygenase 2 , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/analysis , Drug Administration Schedule , Drug Evaluation, Preclinical , Eicosanoids/analysis , Endocannabinoids , Enzyme Induction , Exploratory Behavior/drug effects , Furans/administration & dosage , Furans/pharmacology , Glycerides/analysis , Male , Maze Learning/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Premedication , Prostaglandin-Endoperoxide Synthases/biosynthesis , Rats , Rats, Sprague-Dawley , Recovery of Function , Reflex, Abnormal/drug effects
12.
Neurotox Res ; 6(4): 333-42, 2004.
Article in English | MEDLINE | ID: mdl-15545017

ABSTRACT

Cell death/survival following traumatic brain injury (TBI) may be a result of alterations in the intracellular ratio of death and survival factors. Bcl-2 family genes mediate both cell survival and the initiation of cell death. Using lysate RNase protection assays, mRNA expression of the anti-cell death genes Bcl-2 and Bcl-xL, and the pro-cell death gene Bax, was evaluated following experimental brain injuries in adult male Sprague-Dawley rats. Both the lateral fluid-percussion (LFP) and the lateral controlled cortical impact (LCI) models of TBI showed similar patterns of gene expression. Anti-cell death bcl-2 and bcl-xL mRNAs were attenuated early and tended to remain depressed for at least 3 days after injury in the cortex and hippocampus ipsilateral to injury. Pro-cell death bax mRNA was elevated in these areas, usually following the decrease in anti-cell death genes. These common patterns of gene expression suggest an important role for Bcl-2 genes in cell death and survival in the injured brain. Understanding the regulation of these genes may facilitate the development of new therapeutic strategies for a condition that currently has no proven pharmacologic treatments.


Subject(s)
Brain Injuries/genetics , Brain Injuries/metabolism , Disease Models, Animal , Multigene Family , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Gene Expression Regulation/physiology , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Time Factors , bcl-2-Associated X Protein , bcl-X Protein
13.
J Neurosci ; 24(47): 10763-72, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15564594

ABSTRACT

Recent studies suggest that postmitotic neurons can reenter the cell cycle as a prelude to apoptosis after brain injury. However, most dying neurons do not pass the G1/S-phase checkpoint to resume DNA synthesis. The specific factors that trigger abortive DNA synthesis are not characterized. Here we show that the combination of hypoxia and ischemia induces adult rodent neurons to resume DNA synthesis as indicated by incorporation of bromodeoxyuridine (BrdU) and expression of G1/S-phase cell cycle transition markers. After hypoxia-ischemia, the majority of BrdU- and neuronal nuclei (NeuN)-immunoreactive cells are also terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL)-stained, suggesting that they undergo apoptosis. BrdU+ neurons, labeled shortly after hypoxia-ischemia, persist for >5 d but eventually disappear by 28 d. Before disappearing, these BrdU+/NeuN+/TUNEL+ neurons express the proliferating cell marker Ki67, lose the G1-phase cyclin-dependent kinase (CDK) inhibitors p16INK4 and p27Kip1 and show induction of the late G1/S-phase CDK2 activity and phosphorylation of the retinoblastoma protein. This contrasts to kainic acid excitotoxicity and traumatic brain injury, which produce TUNEL-positive neurons without evidence of DNA synthesis or G1/S-phase cell cycle transition. These findings suggest that hypoxia-ischemia triggers neurons to reenter the cell cycle and resume apoptosis-associated DNA synthesis in brain. Our data also suggest that the demonstration of neurogenesis after brain injury requires not only BrdU uptake and mature neuronal markers but also evidence showing absence of apoptotic markers. Manipulating the aberrant apoptosis-associated DNA synthesis that occurs with hypoxia-ischemia and perhaps neurodegenerative diseases could promote neuronal survival and neurogenesis.


Subject(s)
Apoptosis/physiology , Brain/physiopathology , DNA/biosynthesis , Hypoxia-Ischemia, Brain/physiopathology , Neurons/physiology , S Phase/physiology , Adrenalectomy , Age Factors , Animals , Brain/metabolism , Brain/pathology , Brain Injuries/physiopathology , Bromodeoxyuridine , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , In Situ Nick-End Labeling , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley
14.
Article in English | MEDLINE | ID: mdl-15063335

ABSTRACT

Arachidonic acid (AA) is metabolized to prostaglandins (PGs) via cyclooxygenases (COX) catalysis, and to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatrienoic acids (DiHETrEs), and hydroxyeicosatetraenoic acids (HETEs) via cytochrome P450 (CYP450) enzymes. A reliable and robust fluorescence based HPLC method for these eicosanoids was developed. A new selective reverse-phase solid phase extraction (SPE) procedure was developed for PG, DiHETrEs, HETE, and EETs of interest from rat cortical brain tissue. The eicosanoids were derivatized with 2-(2,3-naphthalimino)ethyl-trifluoromethanesulphonate (NE-OTf), followed by separation and quantification at high sensitivity using reverse-phase HPLC with fluorescent detection, and further identified via LC/MS. The derivatization was studied and optimized to obtain reproducible reactions. Various PGs, DiHETrEs, HETEs, EETs, and AA were sensitively detected and baseline resolved simultaneously. LC/MS under positive electrospray ionization selected ion monitoring (SIM) mode was developed to further identify the peaks of these eicosanoids in cortical brain tissue. The method was applied in the traumatic brain injured rat brain.


Subject(s)
Brain Chemistry , Chromatography, High Pressure Liquid/methods , Eicosanoids/analysis , Spectrometry, Fluorescence/methods , Animals , Calibration , Male , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
15.
NeuroRx ; 1(1): 26-35, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15717005

ABSTRACT

Animals exposed to brief periods of moderate hypoxia (8% to 10% oxygen for 3 hours) are protected against cerebral and cardiac ischemia between 1 and 2 days later. This hypoxia preconditioning requires new RNA and protein synthesis. The mechanism of this hypoxia-induced tolerance correlates with the induction of the hypoxia-inducible factor (HIF), a transcription factor heterodimeric complex composed of inducible HIF-1alpha and constitutive HIF-1beta proteins that bind to the hypoxia response elements in a number of HIF target genes. Our recent studies show that HIF-1alpha correlates with hypoxia induced tolerance in neonatal rat brain. HIF target genes, also induced following hypoxia-induced tolerance, include vascular endothelial growth factor, erythropoietin, glucose transporters, glycolytic enzymes, and many other genes. Some or all of these genes may contribute to hypoxia-induced protection against ischemia. HIF induction of the glycolytic enzymes accounts in part for the Pasteur effect in brain and other tissues. Hypoxia-induced tolerance is not likely to be equivalent to treatment with a single HIF target gene protein since other transcription factors including Egr-1 (NGFI-A) have been implicated in hypoxia regulation of gene expression. Understanding the mechanisms and genes involved in hypoxic tolerance may provide new therapeutic targets to treat ischemic injury and enhance recovery.


Subject(s)
Brain Ischemia/physiopathology , Ischemic Preconditioning , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Brain/metabolism , Brain/pathology , Brain/physiopathology , Brain Ischemia/genetics , DNA-Binding Proteins/metabolism , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit , Rats , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism
16.
Ann N Y Acad Sci ; 993: 134-45; discussion 159-60, 2003 May.
Article in English | MEDLINE | ID: mdl-12853306

ABSTRACT

Delineating the mechanisms of survival pathways that exist in neurons will provide important insight into how neurons utilize intracellular proteins as neuroprotectants against the causes of acute neurodegeneration. We have employed cultured rat cerebellar granule cells as a model for determining the mechanisms of these intraneuronal survival pathways. Glutamate has long been known to kill neurons by an N-methyl-d-aspartate (NMDA) receptor-mediated mechanism. Paradoxically, subtoxic concentrations of NMDA protect neurons against glutamate-mediated excitotoxicity. Because NMDA protects neurons in physiologic concentrations of glucose and oxygen, we refer to this phenomenon as physiologic preconditioning. One of the major mechanisms of NMDA neuroprotection involves the activation of NMDA receptors leading to the rapid release of brain-derived neurotrophic factor (BDNF). BDNF then binds to and activates its cognate receptor, receptor tyrosine kinase B (TrkB). The efficient utilization of these two receptors confers remarkable resistance against millimolar concentrations of glutamate that kill more than eighty percent of the neurons in the absence of preconditioning the neurons with a subtoxic concentration of NMDA. Exactly how the neurons mediate neuroprotection by activation of both receptors is just beginning to be understood. Both NMDA and TrkB receptors activate nuclear factor kappaB (NF-kappaB), a transcription factor known to be involved in protecting neurons against many different kinds of toxic insults. By converging on survival transcription factors, such as NF-kappaB, NMDA and TrkB receptors protect neurons. Thus, crosstalk between these very different receptors provides a rapid means of neuronal communication to upregulate survival proteins through release and transcriptional activation of messenger RNA.


Subject(s)
Cerebellum/metabolism , Ischemic Preconditioning/methods , Neurons/metabolism , Neuroprotective Agents/metabolism , Receptor, trkB/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Autocrine Communication , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Genes, bcl-2 , Glutamic Acid/toxicity , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Oligonucleotides, Antisense/metabolism
17.
J Neurotrauma ; 20(5): 421-35, 2003 May.
Article in English | MEDLINE | ID: mdl-12803975

ABSTRACT

Cell death/survival following CNS injury may be a result of alterations in the intracellular ratio of death and survival factors. Using immunohistochemistry, Western analysis and in situ hybridization, the expression of the anti-cell death protein, Bcl-2, and the pro-cell death protein, Bax, was evaluated following lateral fluid-percussion (FP) brain injury of moderate severity (2.3-2.6 atm) in adult male Sprague-Dawley rats. By 2 h post-injury, a marked reduction of cellular Bcl-2-immunoreactivity (IR) and a mild decrease in cellular Bax IR were observed in the temporal and occipital cortices, and in the hippocampal CA3 ipsilateral to the site of impact. These decreases in Bcl-2 and Bax IR appeared to precede the overt cell loss in these regions that was evident at 24 h. Immunoblot analysis supported the immunohistochemical data, with a modest but significant reduction in the intensities of both the Bcl-2 and Bax protein bands at 2 h (p < 0.05 compared to sham levels). However, the Bax:Bcl-2 ratio increased significantly at 2 h (2.28 +/- 0.13) and remained elevated up to 7 days (2.05 +/- 0.13) post-injury compared to sham-injured control tissue (1.62 +/- 0.10, p < 0.05). Furthermore, cortical, but not hippocampal, levels of Bax protein increased by 25% (p < 0.05 compared to sham-injured controls) at 24 h post-injury, and returned to control levels by 7 days. In situ hybridization analysis of Bax mRNA revealed increased cellular grain density in the injured cortex (p < 0.05 compared to sham-injured brains), but not in the CA3 region of the injured hippocampus. No injury-induced changes in the expression of Bcl-2 mRNA were observed in any brain region. Taken together, these data suggest that the association between regional post-traumatic cell death and alterations in the cellular ratio of Bcl-2 and Bax may be, in part, due to alterations in mRNA and/or protein expression of the Bcl-2 family of proteins.


Subject(s)
Brain Injuries/metabolism , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Animals , Blotting, Western , Brain Injuries/pathology , Functional Laterality , Gene Expression , Immunohistochemistry , In Situ Hybridization , Male , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Time Factors , bcl-2-Associated X Protein
18.
J Neurotrauma ; 20(5): 463-76, 2003 May.
Article in English | MEDLINE | ID: mdl-12803978

ABSTRACT

The purpose of this study was to investigate the efficacy of a novel steroid, fluasterone (DHEF, a dehydroepiandrosterone (DHEA) analog), at improving functional recovery in a rat model of traumatic brain injury (TBI). The lateral cortical impact model was utilized in two studies of efficacy and therapeutic window. DHEF was given (25 mg/kg, intraperitoneally) at the initial time point and once a day for 2 more days. Study A included four groups: sham injury, vehicle treated (n = 22); injured, vehicle treated (n = 30); injured, pretreated (5-10 min prior to injury, n = 24); and injured, posttreated (initial dose 30 min postinjury, n = 15). Study B (therapeutic window) included five groups: sham injury, vehicle treated (n = 17); injured, vehicle treated (n = 26); and three posttreatment groups: initial dose at 30 min (n = 18), 2 h (n = 23), or 12 h (n = 16) postinjury. Three criteria were used to grade functional recovery. In study A, DHEF improved beam walk performance both with pretreatment (79%) and 30-min posttreatment group (54%; p < 0.01, Dunnett vs. injured vehicle). In study B, the 12-h posttreatment group showed a 97% improvement in beam walk performance (p < 0.01, Dunnett). The 30-min and 12-h posttreatment groups showed a decreased incidence of falls from the beam, which reached statistical significance (p < 0.05, Dunnett). Tests of memory (Morris water maze) and neurological reflexes both revealed significant improvements in all DHEF treatment groups. In cultured rat mesangial cells, DHEF (and DHEA) potently inhibited interleukin-1beta-induced cyclooxygenase-2 (COX2) mRNA and prostaglandin (PGE2) production. In contrast, DHEF treatment did not alter injury-induced COX2 mRNA levels in the cortex or hippocampus. However, DHEF (and DHEA) relaxed ex vivo bovine middle cerebral artery preparations by about 30%, with an IC(50) approximately 40 microM. This was a direct effect on the vascular smooth muscle, independent of the endothelial cell layer. Fluasterone (DHEF) treatments improved functional recovery in a rat TBI model. Possible mechanisms of action for this novel DHEA analog are discussed. These findings suggest an exciting potential use for this agent in the clinical treatment of traumatic brain injury.


Subject(s)
Brain Injuries/drug therapy , Brain/drug effects , Dehydroepiandrosterone/analogs & derivatives , Dehydroepiandrosterone/pharmacology , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Animals , Brain/metabolism , Cells, Cultured , Cyclooxygenase 2 , Dehydroepiandrosterone/administration & dosage , Dinoprostone/metabolism , Glomerular Mesangium/drug effects , Isoenzymes/drug effects , Isoenzymes/metabolism , Male , Maze Learning/drug effects , Middle Cerebral Artery/drug effects , Muscle, Smooth, Vascular/drug effects , Neuroprotective Agents/administration & dosage , Organ Culture Techniques , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Time Factors
19.
J Neurotrauma ; 19(5): 503-57, 2002 May.
Article in English | MEDLINE | ID: mdl-12042091

ABSTRACT

Traumatic brain injury (TBI) remains a major public health problem globally. In the United States the incidence of closed head injuries admitted to hospitals is conservatively estimated to be 200 per 100,000 population, and the incidence of penetrating head injury is estimated to be 12 per 100,000, the highest of any developed country in the world. This yields an approximate number of 500,000 new cases each year, a sizeable proportion of which demonstrate significant long-term disabilities. Unfortunately, there is a paucity of proven therapies for this disease. For a variety of reasons, clinical trials for this condition have been difficult to design and perform. Despite promising pre-clinical data, most of the trials that have been performed in recent years have failed to demonstrate any significant improvement in outcomes. The reasons for these failures have not always been apparent and any insights gained were not always shared. It was therefore feared that we were running the risk of repeating our mistakes. Recognizing the importance of TBI, the National Institute of Neurological Disorders and Stroke (NINDS) sponsored a workshop that brought together experts from clinical, research, and pharmaceutical backgrounds. This workshop proved to be very informative and yielded many insights into previous and future TBI trials. This paper is an attempt to summarize the key points made at the workshop. It is hoped that these lessons will enhance the planning and design of future efforts in this important field of research.


Subject(s)
Brain Injuries/therapy , Clinical Trials as Topic/methods , Humans
20.
J Neurotrauma ; 19(5): 627-38, 2002 May.
Article in English | MEDLINE | ID: mdl-12042097

ABSTRACT

Primary insults to the brain can initiate glutamate release that may result in excitotoxicity followed by neuronal cell death. This secondary process is mediated by both N-methyl-D-aspartate (NMDA) and non-NMDA receptors in vivo and requires new gene expression. Neuronal cyclooxygenase-2 (COX2) expression is upregulated following brain insults, via glutamatergic and inflammatory mechanisms. The products of COX2 are bioactive prostanoids and reactive oxygen species that may play a role in neuronal survival. This study explores the role of neuronal COX2 in glutamate excitotoxicity using cultured cerebellar granule neurons (day 8 in vitro). Treatment with excitotoxic concentrations of glutamate or kainate transiently induced COX2 mRNA (two- and threefold at 6 h, respectively, p < 0.05, Dunnett) and prostaglandin production (five- and sixfold at 30 min, respectively, p < 0.05, Dunnett). COX2 induction peaked at toxic concentrations of these excitatory amino acids. Surprisingly, NMDA, L-quisqualate, and trans-ACPD did not induce COX2 mRNA at any concentration tested. The glutamate receptor antagonist NBQX (5 microM, AMPA/kainate receptor) completely inhibited kainate-induced COX2 mRNA and partially inhibited glutamate-induced COX2 (p < 0.05, Dunnett). Other glutamate receptor antagonists, such as MK-801 (1 microM, NMDA receptor) or MCPG (500 microM, class 1 metabotropic receptors), partially attenuated glutamate-induced COX2 mRNA. These antagonists all reduced steady-state COX2 mRNA (p < 0.05, Dunnett). To determine whether COX2 might be an effector of excitotoxic cell death, cerebellar granule cells were pretreated (24 h) with the COX2-specific enzyme inhibitor, DFU (5,5-dimethyl-3-(3-fluorophenyl)-4-(4-methylsulphonyl) phenyl-2((5)H)-furanone) prior to glutamate challenge. DFU (1 to 1000 nM) completely protected cultured neurons from glutamate-mediated neurotoxicity. Approximately 50% protection from NMDA-mediated neurotoxicity, and no protection from kainate-mediated neurotoxicity was observed. Therefore, glutamate-mediated COX2 induction contributes to excitotoxic neuronal death. These results suggest that glutamate, NMDA, and kainate neurotoxicity involve distinct excitotoxic pathways, and that the glutamate and NMDA pathways may intersect at the level of COX2.


Subject(s)
Cell Death/drug effects , Isoenzymes/antagonists & inhibitors , Neurons/cytology , Neurons/enzymology , Animals , Cells, Cultured , Cerebellum/cytology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Furans/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glutamic Acid/pharmacology , Isoenzymes/genetics , Kainic Acid/pharmacology , Neurotoxins/pharmacology , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...