Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neurotrauma ; 30(13): 1161-9, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23409698

ABSTRACT

The goal of this project was to determine whether biochemical markers of brain damage can be used to diagnose and assess the severity of injury in a rat model of penetrating ballistic-like brain injury (PBBI). To determine the relationship between injury magnitude and biomarker levels, rats underwent three discrete PBBI severity levels defined by the magnitude of the ballistic component of the injury, calibrated to equal 5%, 10%, or 12.5% of total rat brain volume. Cortex, cerebrospinal fluid (CSF), and blood were collected at multiple time points. Levels of three biomarkers (αII-spectrin breakdown product [SBDP150], glial fibrillary acidic protein [GFAP], and ubiquitin C-terminal hydrolase-L1 [UCH-L1]), were measured using quantitative immunoblotting and/or enzyme-linked immunosorbent assays. In injured cortex, SBDP150 and GFAP levels were increased significantly over controls. Cortical SBDP150 was elevated at 1 day but not 7 days, and GFAP at 7 days but not 1 day. At their respective time points, mean levels of SBDP150 and GFAP biomarkers in the cortex rose stepwise as injury magnitude increased. In the CSF, increasing severity of PBBI was associated with increasing concentrations of both neuronal and glial biomarkers acutely at 1 day after injury, but no trends were observed at 7 days. In plasma, SBDP150 was elevated at 5 min after 10% PBBI and at 6 h after 12.5% PBBI. UCH-L1 levels in plasma were elevated acutely at 5 min post-injury reflecting injury severity and rapidly decreased within 2 h. Overall, our results support the conclusion that biomarkers are effective indicators of brain damage after PBBI and may also aid in the assessment of injury magnitude.


Subject(s)
Biomarkers/analysis , Glial Fibrillary Acidic Protein/analysis , Head Injuries, Penetrating/metabolism , Spectrin/analysis , Ubiquitin Thiolesterase/analysis , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Male , Rats , Rats, Sprague-Dawley
2.
Electrophoresis ; 33(24): 3693-704, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23161467

ABSTRACT

Proteomics and systems biology have significantly contributed to biomarker discovery in the field of brain injury. This study utilized 2D-DIGE-PMF-MS as a preliminary screen to detect biomarkers in a rat model of penetrating ballistic-like brain injury (PBBI). Brain-specific systems biology analysis of brain tissue identified 386 proteins having a fold change of more than 2, of which 321 proteins were increased and 65 were decreased 24 h after PBBI compared to sham controls. The majority of upregulated proteins were cytoskeletal (10.5%), nucleic acid binding (9.3%), or kinases (8.9%). Most proteins were involved in protein metabolism (22.7%), signal transduction (20.4%), and development (9.6%). Pathway analysis indicated that these proteins were involved in neurite outgrowth and cell differentiation. Semiquantitative Western blotting of 6, 24, 48, and 72 h after PBBI indicated ubiquitin carboxyl-terminal hydrolase isozyme L1 (a proposed traumatic brain injury biomarker in human clinical trials), tyrosine hydroxylase, and syntaxin-6 were found to be consistently elevated in brain tissue and cerebral spinal fluid after PBBI compared to sham controls. Combining proteomics and brain-specific systems biology can define underlying mechanisms of traumatic brain injury and provide valuable information in biomarker discovery that, in turn, may lead to novel therapeutic targets.


Subject(s)
Head Injuries, Penetrating/metabolism , Proteome/analysis , Proteomics/methods , Animals , Biomarkers/analysis , Biomarkers/chemistry , Brain Chemistry , Databases, Protein , Disease Models, Animal , Head Injuries, Penetrating/pathology , Histocytochemistry , Male , Proteins/analysis , Proteome/chemistry , Rats , Rats, Sprague-Dawley , Systems Biology/methods
3.
J Neurotrauma ; 28(10): 2185-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21644814

ABSTRACT

This study evaluated the injury severity profile of unilateral, frontal penetrating ballistic-like brain injury (PBBI) on neurofunctional outcome, blood-brain barrier (BBB) permeability, and brain edema formation. The degree of injury severity was determined by the delivery of a water-pressure pulse designed to produce a temporary cavity by rapid (<40 ms) expansion of the probe's elastic balloon calibrated to equal 5%, 10%, 12.5%, or 15% of total rat brain volume (control groups consisted of sham surgery or insertion of the probe only). Neurofunctional assessments revealed motor and cognitive deficits related to the degree of injury severity, with the most clear-cut profile of PBBI injury severity depicted by the Morris water maze (MWM) results. A biphasic pattern of BBB leakage was detected in the injured hemisphere at all injury severity levels at 4 h post-injury, and again at 48-72 h post-injury, which remained evident out to 7 days post-PBBI in the 10% and 12.5% PBBI groups. Likewise, significant brain edema was detected in the injured hemisphere by 4 h post-injury and remained elevated out to 7 days post-injury in the 10% and 12.5% PBBI groups. However, following 5% PBBI, significant levels of edema were only detected from 24 h to 48h post-injury. These results identify an injury severity profile of BBB permeability, brain edema, and neurofunctional impairment that provides sensitive and clinically relevant outcome metrics for studying potential therapeutics.


Subject(s)
Blood-Brain Barrier/physiology , Brain Edema/etiology , Head Injuries, Penetrating/pathology , Nervous System Diseases/etiology , Animals , Behavior, Animal/physiology , Blood-Brain Barrier/pathology , Brain/pathology , Brain Edema/pathology , Extravasation of Diagnostic and Therapeutic Materials , Forelimb/physiology , Functional Laterality , Head Injuries, Penetrating/complications , Head Injuries, Penetrating/surgery , Intracranial Pressure/physiology , Male , Maze Learning/physiology , Nervous System Diseases/pathology , Neurosurgical Procedures , Permeability , Postural Balance/physiology , Rats , Rats, Sprague-Dawley , Recovery of Function
4.
J Neurotrauma ; 28(7): 1237-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21463155

ABSTRACT

Brain edema formation associated with trauma-induced intracerebral hemorrhage (ICH) is a clinical complication with high mortality. Studies have shown that heme oxygenase-1 (HO-1) plays an important role in ICH-induced brain edema. In order to understand the role of HO-1 in the protective effect of selective brain cooling (SBC), we investigated the time course of HO-1 changes following penetrating ballistic-like brain injury (PBBI) in rats. Samples were collected from injured and control animals at 6, 24, 48, and 72 h, and 7 days post-injury to evaluate HO-1 expression, heme concentration, brain water content, and immunohistochemistry (IHC). Following a 10% frontal PBBI, HO-1 mRNA and protein was increased at all time points studied, reaching maximum expression levels at 24-48 h post-injury. An increase in the heme concentration and the development of brain edema coincided with the upregulation of HO-1 mRNA and protein during the 7-day post-injury period. SBC significantly decreased PBBI-induced heme concentration, attenuated HO-1 upregulation, and concomitantly reduced brain water content. These results suggest that the neuroprotective effects of SBC may be partially mediated by reducing the heme accumulation, which reduced injury-mediated upregulation of HO-1, and in turn ameliorated edema formation. Collectively, these results suggest a potential value of HO-1 as a diagnostic and/or therapeutic biomarker in hemorrhagic brain injury.


Subject(s)
Brain Edema/enzymology , Brain Edema/therapy , Cerebral Hemorrhage/enzymology , Cerebral Hemorrhage/therapy , Head Injuries, Penetrating/enzymology , Head Injuries, Penetrating/therapy , Heme Oxygenase (Decyclizing)/physiology , Hypothermia, Induced/methods , Animals , Body Water/metabolism , Brain Edema/physiopathology , Cerebral Hemorrhage/physiopathology , Disease Models, Animal , Down-Regulation/physiology , Head Injuries, Penetrating/complications , Heme/antagonists & inhibitors , Heme/metabolism , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/genetics , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
5.
J Neurotrauma ; 27(10): 1837-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20698760

ABSTRACT

Diagnosis and treatment of stroke and traumatic brain injury remain significant health care challenges to society. Patient care stands to benefit from an improved understanding of the interactive biochemistry underlying neurotrauma pathobiology. In this study, we assessed the power of neuroproteomics to contrast biochemical responses following ischemic and traumatic brain injuries in the rat. A middle cerebral artery occlusion (MCAO) model was employed in groups of 30-min and 2-h focal neocortical ischemia with reperfusion. Neuroproteomes were assessed via tandem cation-anion exchange chromatography-gel electrophoresis, followed by reversed-phase liquid chromatography-tandem mass spectrometry. MCAO results were compared with those from a previous study of focal contusional brain injury employing the same methodology to characterize homologous neocortical tissues at 2 days post-injury. The 30-min MCAO neuroproteome depicted abridged energy production involving pentose phosphate, modulated synaptic function and plasticity, and increased chaperone activity and cell survival factors. The 2-h MCAO data indicated near complete loss of ATP production, synaptic dysfunction with degraded cytoarchitecture, more conservative chaperone activity, and additional cell survival factors than those seen in the 30-min MCAO model. The TBI group exhibited disrupted metabolism, but with retained malate shuttle functionality. Synaptic dysfunction and cytoarchitectural degradation resembled the 2-h MCAO group; however, chaperone and cell survival factors were more depressed following TBI. These results underscore the utility of neuroproteomics for characterizing interactive biochemistry for profiling and contrasting the molecular aspects underlying the pathobiological differences between types of brain injuries.


Subject(s)
Brain Injuries/metabolism , Brain Ischemia/metabolism , Reperfusion Injury/metabolism , Analysis of Variance , Animals , Blotting, Western , Chromatography, Ion Exchange , Chromatography, Liquid , Disease Models, Animal , Male , Proteomics , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry
6.
J Neuroinflammation ; 6: 19, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19656406

ABSTRACT

BACKGROUND: Inflammatory cytokines play a crucial role in the pathophysiology of traumatic brain injury (TBI), exerting either deleterious effects on the progression of tissue damage or beneficial roles during recovery and repair. NNZ-2566, a synthetic analogue of the neuroprotective tripeptide Glypromate, has been shown to be neuroprotective in animal models of brain injury. The goal of this study was to determine the effects of NNZ-2566 on inflammatory cytokine expression and neuroinflammation induced by penetrating ballistic-like brain injury (PBBI) in rats. METHODS: NNZ-2566 or vehicle (saline) was administered intravenously as a bolus injection (10 mg/kg) at 30 min post-injury, immediately followed by a continuous infusion of NNZ-2566 (3 mg/kg/h), or equal volume of vehicle, for various durations. Inflammatory cytokine gene expression from the brain tissue of rats exposed to PBBI was evaluated using microarray, quantitative real time PCR (QRT-PCR), and enzyme-linked immunosorbent assay (ELISA) array. Histopathology of the injured brains was examined using hematoxylin and eosin (H&E) and immunocytochemistry of inflammatory cytokine IL-1beta. RESULTS: NNZ-2566 treatment significantly reduced injury-mediated up-regulation of IL-1beta, TNF-alpha, E-selectin and IL-6 mRNA during the acute injury phase. ELISA cytokine array showed that NZ-2566 treatment significantly reduced levels of the pro-inflammatory cytokines IL-1beta, TNF-alpha and IFN-gamma in the injured brain, but did not affect anti-inflammatory cytokine IL-6 levels. CONCLUSION: Collectively, these results suggest that the neuroprotective effects of NNZ-2566 may, in part, be functionally attributed to the compound's ability to modulate expression of multiple neuroinflammatory mediators in the injured brain.


Subject(s)
Brain Injuries/drug therapy , Cytokines/drug effects , Encephalitis/drug therapy , Head Injuries, Penetrating/drug therapy , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain Injuries/complications , Brain Injuries/physiopathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Encephalitis/etiology , Encephalitis/physiopathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Head Injuries, Penetrating/complications , Head Injuries, Penetrating/physiopathology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Treatment Outcome
7.
J Neurotrauma ; 26(8): 1295-305, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19317603

ABSTRACT

To gain additional insights into the pathogenic cellular and molecular mechanisms underlying different types of brain injury (e.g., trauma versus ischemia), recently attention has focused on the discovery and study of protein biomarkers. In previous studies, using a high-throughput immunoblotting (HTPI) technique, we reported changes in 29 out of 998 proteins following acute injuries to the rat brain (penetrating traumatic versus focal ischemic). Importantly, we discovered that one protein, endothelial monocyte-activating polypeptide II precursor (p43/pro-EMAPII), was differentially expressed between these two types of brain injury. Among other functions, p43/pro-EMAPII is a known pro-inflammatory cytokine involved in the progression of apoptotic cell death. Our current objective was to verify the changes in p43/pro-EMAPII expression, and to evaluate the potentially important implications that the differential regulation of this protein has on injury development. At multiple time points following either a penetrating ballistic-like brain injury (PBBI), or a transient middle cerebral artery occlusion (MCAo) brain injury, tissue samples (6-72 h), CSF samples (24 h), and blood samples (24 h) were collected from rats for analysis. Changes in protein expression were assessed by Western blot analysis and immunohistochemistry. Our results indicated that p43/pro-EMAPII was significantly increased in brain tissues, CSF, and plasma following PBBI, but decreased after MCAo injury compared to their respective sham control samples. This differential expression of p43/pro-EMAPII may be a useful injury-specific biomarker associated with the underlying pathologies of traumatic versus ischemic brain injury, and provide valuable information for directing injury-specific therapeutics.


Subject(s)
Brain Injuries/diagnosis , Brain Ischemia/diagnosis , Cytokines/metabolism , Neoplasm Proteins/metabolism , Protein Precursors/metabolism , RNA-Binding Proteins/metabolism , Animals , Biomarkers/metabolism , Brain Injuries/metabolism , Brain Ischemia/metabolism , Cell Count , Immunoblotting , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley
8.
J Neurotrauma ; 26(1): 141-54, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19119917

ABSTRACT

Glycine-proline-glutamate (GPE) is an N-terminal tripeptide endogenously cleaved from insulin-like growth factor-1 in the brain and is neuroprotective against hypoxic-ischemic brain injury and neurodegeneration. NNZ-2566 is an analog of GPE designed to have improved bioavailability. In this study, we tested NNZ-2566 in a rat model of penetrating ballistic-type brain injury (PBBI) and assessed its effects on injury-induced histopathology, behavioral deficits, and molecular and cellular events associated with inflammation and apoptosis. In the initial dose-response experiments, NNZ-2566 (0.01-3 mg/kg/h x 12 h intravenous infusion) was given at 30 min post-injury and the therapeutic time window was established by delaying treatments 2-4 h post-injury, but with the addition of a 10- or 30-mg/kg bolus dose. All animals survived 72 h. Neuroprotection was evaluated by balance beam testing and histopathology. The effects of NNZ-2566 on injury-induced changes in Bax and Bcl-2 proteins, activated microgliosis, neutrophil infiltration, and astrocyte reactivity were also examined. Behavioral results demonstrated that NNZ-2566 dose-dependently reduced foot faults by 19-66% after acute treatments, and 35-55% after delayed treatments. Although gross lesion volume was not affected, NNZ-2566 treatment significantly attenuated neutrophil infiltration and reduced the number of activated microglial cells in the peri-lesion regions of the PBBI. PBBI induced a significant upregulation in Bax expression (36%) and a concomitant downregulation in Bcl-2 expression (33%), both of which were significantly reversed by NNZ-2566. Collectively, these results demonstrated that NNZ-2566 treatment promoted functional recovery following PBBI, an effect related to the modulation of injury-induced neural inflammatory and apoptotic mechanisms.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Encephalitis/drug therapy , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Recovery of Function/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalitis/metabolism , Encephalitis/physiopathology , Gliosis/drug therapy , Gliosis/physiopathology , Gliosis/prevention & control , Injections, Intravenous , Microglia/drug effects , Microglia/pathology , Movement Disorders/drug therapy , Movement Disorders/etiology , Movement Disorders/physiopathology , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/therapeutic use , Oligopeptides/agonists , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Treatment Outcome
9.
Methods Mol Biol ; 566: 25-40, 2009.
Article in English | MEDLINE | ID: mdl-20058162

ABSTRACT

Protein changes induced by traumatic or ischemic brain injury can serve as diagnostic markers as well as therapeutic targets for neuroprotection. The focus of this chapter is to provide a representative overview of preclinical brain injury and proteomics analysis protocols for evaluation and discovery of novel biomarkers. Detailed surgical procedures have been provided for inducing MCAo and implantation of chronic indwelling cannulas for drug delivery. Sample collection and tissue processing techniques for collection of blood, CSF, and brain are also described including standard biochemical methodology for the proteomic analysis of these tissues.The dynamics of proteomic analysis is a multistep process comprising sample preparation, separation, quantification, and identification of proteins. Our approach is to separate proteins first by two-dimensional gel electrophoresis according to charge and molecular mass. Proteins are then fragmented and analyzed using matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS). Identification of proteins can be achieved by comparing the mass-to-charge data to protein sequences in respective databases.


Subject(s)
Brain Ischemia/physiopathology , Nerve Tissue Proteins/analysis , Proteomics/methods , Animals , Brain/pathology , Brain/physiology , Humans , Infarction, Middle Cerebral Artery , Proteome/analysis , Rats
10.
Brain Inj ; 22(10): 723-32, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18720098

ABSTRACT

PRIMARY OBJECTIVE: Recent efforts have been aimed at developing a panel of protein biomarkers for the diagnosis/prognosis of the neurological damage associated with acute brain injury. METHODS AND PROCEDURES: This study utilized high-throughput immunoblotting (HTPI) technology to compare changes between two animal models of acute brain injury: penetrating ballistic-like brain injury (PBBI) which mimics the injury created by a gunshot wound and transient middle cerebral artery occlusion (MCAo) which is a model of stroke. Brain and blood were collected at 24-hours post-injury. MAIN OUTCOMES AND RESULTS: This study identified the changes in 18 proteins following PBBI and 17 proteins following MCAo out of a total of 998 screened proteins. Distinct differences were observed between the two models: five proteins were up- or down-regulated in both models, 23 proteins changed in only one model and one protein was differentially expressed. Western blots were used to verify HTPI results for selected proteins with measurable changes observed in both blood and brain for the proteins STAT3, Tau, PKA RII beta, 14-3-3 epsilon and p43/EMAPII. CONCLUSIONS: These results suggest distinct post-injury protein profiles between brain injury types (traumatic vs. ischemic) that will facilitate strategies aimed at the differential diagnosis and prognosis of acute brain injury.


Subject(s)
Cerebral Hemorrhage, Traumatic/metabolism , Infarction, Middle Cerebral Artery/metabolism , Membrane Proteins/analysis , Wounds, Gunshot/metabolism , Animals , Biomarkers/analysis , Blotting, Western , Brain Chemistry , Cerebral Hemorrhage, Traumatic/pathology , Immunoblotting/methods , Infarction, Middle Cerebral Artery/pathology , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Wounds, Gunshot/pathology
11.
Neurosci Lett ; 422(1): 64-7, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17600621

ABSTRACT

In an earlier study, we demonstrated that PAN-811 (3-aminopyridine-2-carboxaldehyde thiosemicarbazone), a novel neuroprotectant, provides protection against glutamate, staurosporine, veratridine, or hypoxia/hypoglycemia toxicities in primary cortical neuronal cultures by upregulating Bcl-2 expression [R.-W. Chen, C. Yao, X.C. Lu, Z.-G. Jiang, R. Whipple, Z. Liao, H.A. Ghanbari, B. Almassian, F.C. Tortella, J.R. Dave. PAN-811 (3-aminopyridine-2-carboxaldehyde thiosemicarbazone), a novel neuroprotectant, elicits its function in primary neuronal cultures by upregulating Bcl-2 expression. Neuroscience 135 (2005) 191-201]. Both JNK (c-Jun N-terminal kinase) and p38 MAP (mitogen-activated protein) kinase activation have a direct inhibitory action on Bcl-2 by phosphorylation. In the present study, we continued to explore the mechanism of PAN-811 neuroprotection. Our results indicate that treatment of cultured cortical neurons with glutamate (100 microM) induces phosphorylation of both JNK and p38 MAPK. Specifically, pretreatment of neurons with 10 microM PAN-811 (an optimal neuroprotective concentration) for 1h, 4h, or 24h significantly suppresses glutamate-mediated activation of both JNK and p38 MAPK. Furthermore, the p38 MAPK-specific inhibitor SB203580 and the JNK-specific inhibitor SP600125 prevented glutamate-induced neuronal death in these primary cultures. Our results demonstrate that glutamate-induced phosphorylation of JNK and p38 MAPK is suppressed by PAN-811, which might contribute to Bcl-2 upregulation and PAN-811 neuroprotection.


Subject(s)
Excitatory Amino Acid Antagonists , Glutamic Acid/toxicity , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Pyridines/pharmacology , Thiosemicarbazones/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Anthracenes/pharmacology , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Female , Genes, bcl-2/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Tetrazolium Salts , Thiazoles
12.
Neurosci Lett ; 418(2): 165-9, 2007 May 17.
Article in English | MEDLINE | ID: mdl-17398001

ABSTRACT

Cellular injury can involve the aberrant stimulation of cell cycle proteins in part through activation of phosphodiesterases (PDEs) and downstream expression of cell-cycle components such as cyclin D1. In mature non-proliferating cells activation of the cell cycle can lead to the induction of programmed cell death. In the present study, we investigated the in vitro neuroprotective efficacy and mechanism of action of vinpocetine (PDE1 inhibitor), trequinsin (PDE3 inhibitor), and rolipram (PDE4 inhibitor) in four mechanistically-distinct models of injury to primary rat cortical neurons as related to cell cycle regulation and apoptosis. Cellular injury was induced by hypoxia/hypoglycemia, veratridine (10 microM), staurosporine (1 microM), or glutamate (100 microM), resulting in average neuronal cell death rates of 43-48% as determined by MTT assay. Treatment with each PDE inhibitor (PDEI) resulted in a similar concentration-dependent neuroprotection profile with maximal effective concentrations of 5-10 microM (55-77% neuroprotection) in all four neurotoxicity models. Direct cytotoxicity due to PDE inhibition alone was not observed at concentrations below 100 microM. Further studies indicated that PDEIs can suppress the excitotoxic upregulation of cyclin D1 similar to the effects of flavopiridol, a cyclin-dependent kinase inhibitor, including suppression of pro-apoptotic caspase-3 activity. Overall, these data indicate that PDEIs are broad-spectrum neuroprotective agents acting through modulation of cell cycle elements and may offer a novel mode of therapy against acute injury to the brain.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle Proteins/drug effects , Nerve Degeneration/drug therapy , Neurons/drug effects , Phosphodiesterase Inhibitors/pharmacology , Animals , Apoptosis/physiology , Caspase 3/drug effects , Cell Cycle Proteins/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Cyclin D1/drug effects , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/drug effects , Phosphoric Diester Hydrolases/metabolism , Rats , Rats, Sprague-Dawley
13.
J Neurosci Res ; 77(6): 843-57, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15334602

ABSTRACT

Temporal changes in gene expression were measured using DNA microarrays after 30-min or 2-hr transient middle cerebral artery occlusion (MCAo) in rats. Total RNA was extracted from the injured hemisphere at 30 min, 4 hr, 8 hr, 24 hr, 3 days, and 7 days after MCAo for GeneChip analysis using Affymetrix U34 Rat Neurobiology arrays (1,322 functional genes). In total, 267 genes were expressed differentially: 166 genes were upregulated, 94 genes were downregulated, and 7 genes were biphasically up- and downregulated. Among all differentially expressed genes, 88 were newly identified as associated with ischemic brain injury. Most affected genes were distributed among 12 functional categories. Immediate early genes, transcription factors, and heat shock proteins were upregulated as early as 30 min after MCAo, followed by the upregulation of inflammation, apoptosis, cytoskeletal, and metabolism genes, which peaked within 4-24 hr of injury. Neurotrophic growth factors exhibited a sustained upregulation beginning 24 hr after MCAo and persisting through 7 days post-injury. Three classes of genes were downregulated with distinct temporal patterns: ion channel genes and neurotransmitter receptor genes were downregulated between 8-24 hr after injury, whereas synaptic proteins genes were downregulated between 3-7 days after MCAo. Downregulation of synaptic protein gene expression after ischemic injury is of particular interest because of its conspicuously delayed pattern as a functional group, which has not been reported previously and may play a role in post-injury recovery.


Subject(s)
Brain Ischemia/genetics , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Reperfusion Injury/genetics , Animals , Brain Ischemia/metabolism , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism
14.
Neurotox Res ; 5(3): 213-20, 2003.
Article in English | MEDLINE | ID: mdl-12835125

ABSTRACT

This study investigated the effects of veratridine-induced neuronal toxicity on sodium channel gene (NaCh) expression in primary forebrain cultures enriched in neurons, and its reversal by a novel sodium channel blocker, RS100642. Using quantitative RT-PCR, our findings demonstrated the expression ratio of NaCh genes in normal fetal rat forebrain neurons to be Na(v)1.2 > Na(v)1.3 > Na(v)1.8 > Na(v)1.1 > Na(v)1.7 (rBII > rBIII > PN3 > rBI > PN1). Veratridine treatment of neuronal cells produced neurotoxicity in a dose-dependent manner (0.25-20 micro M). Neuronal injury caused by a dose of veratridine producing 80% cell death (2.5 micro M) significantly, and exclusively down-regulated the Na(v)1.1 gene. However, treatment of neurons with RS100642 (200 micro M) reversed the down-regulation of the Na(v)1.1 gene expression caused by veratridine. Our findings document for the first time quantitative and relative changes in the expression of various NaCh genes in neurons following injury produced by selective activation of voltage-gated sodium channels, and suggest that the Na(v)1.1 sodium channel gene may play a key role in the neuronal injury/recovery process.


Subject(s)
Mexiletine/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Veratridine/pharmacology , Animals , Cell Death , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , Embryo, Mammalian/metabolism , Female , Mexiletine/analogs & derivatives , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Neurons/physiology , Prosencephalon/embryology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channels/genetics , Veratridine/administration & dosage
15.
J Cereb Blood Flow Metab ; 22(9): 1068-79, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12218412

ABSTRACT

Ischemia-reperfusion brain injury initiates an inflammatory response involving the expression of adhesion molecules and cytokines, some of which are regulated by the nuclear transcription factor NF-kappaB. In this study the authors examined mRNA expression levels for several important genes associated with inflammation at five time points (3, 6, 12, 24, and 72 hours) after transient middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats. A sensitive and quantitative technique (TaqMan real-time QRT-PCR) was used to simultaneously measure mRNA levels for key cell adhesion molecules and inflammatory cytokines. Gene expression increased significantly in the injured hemisphere for interleukin (IL)-1beta (12-fold increase at 24 hours), IL-6 (25-fold increase at 6 hours) and ICAM-1 (4-fold increase at 24 hours), and the interhemispheric differences for these genes were significant for every time point examined (P < 0.05 for all values). Tumor necrosis factor-alpha mRNA was upregulated in the injured versus uninjured hemisphere from 3 to 24 hours (5-fold increase at 6 hours), while E-selectin showed a significant increase in mRNA levels from 6 to 24 hours after MCAO (10-fold increase at 6 hours) (P < 0.05 for all values). VCAM-1 mRNA levels did not respond differentially to injury at any time point between the two brain hemispheres. At all time points examined, activated NF-kappaB immunoreactivity was observed in cells throughout the infarct-damaged tissue. These results are consistent with the proinflammatory properties of the induced molecules, which are involved in the initiation of the inflammatory cascade, and may thus contribute to secondary cellular responses that lead to further brain damage.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/genetics , Ischemic Attack, Transient/genetics , Reperfusion Injury/genetics , Tumor Necrosis Factor-alpha/genetics , Vascular Cell Adhesion Molecule-1/genetics , Animals , Base Sequence , Corpus Striatum/immunology , Corpus Striatum/pathology , DNA Primers , E-Selectin/genetics , Ischemic Attack, Transient/physiopathology , Male , Middle Cerebral Artery , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...