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










Publication year range
1.
J Neurotrauma ; 13(3): 125-37, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8965322

ABSTRACT

This study employed Western blotting and qualitative immunohistochemistry to analyze the effects of cortical impact traumatic brain injury (TBI) on acute changes in MAP2 immunoreactivity in the rat cortex. We employed a lateral cortical impact injury device to induce severe TBI, which is associated with focal cortical contusion and neuronal death at the impact site. Three hours following TBI, Western blotting detected substantial MAP2 loss only in the cortex ipsilateral to the site of injury. Light microscopic studies of MAP2 revealed a prominent loss of MAP2 immunofluorescence in apical dendrites of pyramidal neurons within layers 3 and 5, as well as a loss of fine dendritic arborization within layer 1. These changes in MAP2 immunolabeling were associated with, but not exclusively restricted to, the presence of dark shrunken neurons labeled by hematoxylin and eosin staining, suggesting impending cell death. Alterations in MAP2 immunofluorescence were found both within and beyond areas of focal contusion and necrosis in the ipsilateral cortex. Thus, traumatic brain injury in rats can produce rapid and significant dendritic pathology within sites of contusion. However, immunohistochemical changes in MAP2 labeling outside of contused regions suggests that TBI-induced dendritic damage may not be exclusively associated with acute cell death.


Subject(s)
Brain Injuries/immunology , Cerebral Cortex/immunology , Microtubule-Associated Proteins/immunology , Animals , Blotting, Western , Immunohistochemistry , Rats
2.
J Neuropathol Exp Neurol ; 55(1): 68-80, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8558173

ABSTRACT

Semiquantitative Western blot analyses have shown that traumatic brain injury (TBI) can produce significant loss of cytoskeletal proteins (neurofilament 68 [NF68], neurofilament 200 [NF200] and microtubule associated protein 2 [MAP2]) possibly by calpain-mediated proteolysis. Thus, we employed immunofluorescence (light and confocal microscopy) to study the histopathological correlates of acute neurofilament and MAP2 protein decreases observed 3 hours following unilateral cortical injury in rats. TBI induced dramatic alterations in NF68, NF200, and MAP2 immunolabeling in dendrites within and beyond contusion sites ipsilateral and contralateral to the injury site. Marked changes in immunolabeling were associated with but not exclusively restricted to regions of dark shrunken neurons labeled by hematoxylin and eosin staining, a morphopathological response to injury suggesting impending cell death. Light microscopic studies of NF200 immunofluorescence revealed a prominent fragmented appearance of apical dendrites of pyramidal neurons within layers 3 and 5, as well as a loss of fine dendritic arborization within layer 1. Confocal microscopy detected varying degrees of NF200 disassembly associated with these areas of neurofilament fragmentation. Light microscopic studies of NF68 immunofluorescence detected subtle and less severe structural changes including smaller breaks and focal vacuolization of apical dendrites. Light microscopic immunofluorescence of MAP2 revealed changes similar to those seen for NF200. Acute axonal alterations detected with NF68 were minimal compared to immunofluorescence changes seen in dendritic regions. Therefore, preferential dendritic cytoskeletal derangements may be an early morphological feature of experimental traumatic brain injury in vivo. In addition, these cytoskeletal derangements may not be exclusively restricted to sites of contusion and cell death.


Subject(s)
Brain Injuries/pathology , Cytoskeletal Proteins/metabolism , Neurons/pathology , Animals , Cerebral Cortex/pathology , Fluorescent Antibody Technique , Male , Rats , Rats, Sprague-Dawley , Time Factors
3.
J Neurotrauma ; 11(5): 523-32, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7861445

ABSTRACT

Acute biochemical consequences of moderate traumatic brain injury (TBI) include activation of kinases, including protein kinase C (PKC). To determine the possible consequences of PKC activation at the substrate level, we have examined protein phosphorylation patterns 1 h following injury. Although the phosphorylation of most proteins remained unchanged following injury, we observed a significant increase in the phosphorylation of a 61,000 dalton protein (TBI61) in injured rat hippocampus (121% higher than sham control) in vitro. TBI61 phosphorylation could be enhanced by phosphatidyl serine and diacylglycerol or by addition of exogenous PKC. In addition, TBI61 phosphorylation was inhibited by the PKC inhibitor, staurosporine, suggesting further that this protein may be a PKC substrate. These data suggest that TBI increases the phosphorylation of a 61 kD hippocampal protein in vitro. Increases in the protein level and activity of PKC could contribute to this increased phosphorylation.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Alkaloids/pharmacology , Animals , Brain Injuries/enzymology , Diglycerides/pharmacology , Enzyme Activation , Hippocampus/drug effects , Hippocampus/enzymology , Male , Molecular Weight , Phosphatidylserines/pharmacology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Staurosporine
4.
J Neurotrauma ; 11(5): 533-45, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7861446

ABSTRACT

We have examined the effect of lateral cortical impact injury on the levels of axonal cytoskeletal proteins in adult rats. Traumatic brain injury (TBI) causes a significant decrease in the protein levels of two prominent neurofilament (NF) proteins, NF68 and NF200. We employed quantitative immunoreactivity measurements on Western blots to examine NF68 and NF200 levels in homogenates of hippocampal and cortical tissue taken at several intervals postinjury. Sham injury had no effect on NF protein levels. However, injury was associated with a significant loss of NF68, restricted to the cortex ipsilateral to the injury site. NF68 loss was detectable as early as 3 h and lasted at least 2 weeks postinjury. Similarly, TBI induced a decrease in NF200 protein, although losses were observed both ipsilateral and contralateral to the injury site. No loss of NF68 or NF200 protein was detected in hippocampal samples obtained from the same injured animals. An increase in the presence of lower molecular weight (MW) NF68 immunopositive bands was associated with the decrease of NF68 in the ipsilateral cortex. This NF68 antigenicity pattern suggests the production of NF68 breakdown products caused by the pathologic activation of neuronal proteases, such as calpain. Putative NF68 breakdown products increase significantly until 1 day postinjury, suggesting that NF degradation may be ongoing until that time and indicating that a potential therapeutic window may exist within the first 24 h postinjury. In summary, these data identify specific biochemical alterations of the neuronal cytoskeleton following TBI and lay a foundation for further investigation of postinjury cytoskeletal changes in neuronal processes.


Subject(s)
Brain Injuries/metabolism , Neurofilament Proteins/metabolism , Animals , Axons/metabolism , Blotting, Western , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cytoskeleton/metabolism , Hippocampus/metabolism , Immunohistochemistry , Male , Molecular Weight , Rats , Rats, Sprague-Dawley , Time Factors
5.
J Neurotrauma ; 11(3): 275-87, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7996582

ABSTRACT

Evidence suggests that prolonged memory deficits in several neurodegenerative diseases are attributable to deficits in central cholinergic neurotransmission. In traumatic brain injury (TBI), such cholinergic deficits also may contribute to prolonged memory disturbances. This study determined whether moderate magnitudes of TBI produced by controlled cortical impact and mild magnitudes of experimental TBI produced by a new closed head impact technique in rats would produce an enhanced vulnerability to the memory disruptive effects of scopolamine, a muscarinic cholinergic receptor antagonist. Water maze performance was used to determine changes in cholinergic hippocampal function following TBI. In the first experiment, rats received a moderate level of TBI by means of a controlled cortical impact. A Morris water maze task assessed spatial memory function on days 30-34 postinjury. During the 5 day assessment period, statistical analyses showed a group main effect for swim latency. Subsequent post hoc analyses indicated that injured rats had significantly longer latencies on days 30 and 31 (p < 0.05, injury vs sham controls). By days 32-35, injured rats showed no statistically significant deficits in spatial memory performance. On day 35, scopolamine (1 mg/kg, IP) was injected into injured rats and sham-injured rats 15 min prior to being retested in the maze. Results showed that although the scopolamine had no effects on the performance of the sham-injured rats, the same dose significantly (p < 0.05) increased the latency to find the hidden platform in the injured group. In the second experiment, rats received a mild concussive closed head impact. Water maze performance was assessed on days 8-12 postinjury. No significant water maze performance deficits were observed. On day 13, injured and uninjured rats were pharmacologically challenged with scopolamine (1 mg/kg) and retested. Similar to the first experiment, injured rats manifested a significantly greater (p < 0.05) sensitivity to scopolamine than sham controls. The results from both experiments suggest that concussive and more severe levels of TBI can produce an enhanced vulnerability to disruption of cholinergically mediated memory function, even when memory function appears normal in the absence of secondary challenges. These data demonstrate that covert deficits can persist after the recovery of normal function. These deficits may be attributable to a decrease in the ability of cholinergic neurons to function properly. These data also provide important insights into features of receptor-coupled disturbances that could contribute to the maintenance of enduring cognitive deficits following TBI.


Subject(s)
Brain Injuries/physiopathology , Brain/drug effects , Cerebral Cortex/injuries , Scopolamine/pharmacology , Wounds, Nonpenetrating/physiopathology , Acetylcholine/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Brain Injuries/psychology , Male , Maze Learning/drug effects , Memory/drug effects , Muscarinic Antagonists , Rats , Rats, Sprague-Dawley , Space Perception/drug effects , Synaptic Transmission , Wounds, Nonpenetrating/psychology
6.
J Cereb Blood Flow Metab ; 13(5): 796-802, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8360286

ABSTRACT

Traumatic brain injury (TBI) produces a tissue-specific decrease in protein levels of microtubule-associated protein 2 (MAP2), an important cross-linking component of the neuronal cytoskeleton. Because moderate brain hypothermia (30 degrees C) reduces certain neurobehavioral deficits produced by TBI, we examined the efficacy of moderate hypothermia (30 degrees C) in reversing the TBI-induced loss of MAP2 protein. Naive, sham-injured, and moderate (2.1 atm) fluid percussion-injured rats were assessed for MAP2 protein content 3 h post injury using quantitative immunoreactivity measurements. Parallel groups of sham-injured and fluid percussion-injured animals were maintained in moderate hypothermia (30 degrees C), as measured by temporalis muscle temperature, for MAP2 quantitation 3 h post injury. No difference in MAP2 levels was observed between naive and sham-injured normothermic animals. Hypothermia alone had no effect on soluble MAP2 levels in sham-injured animals compared with normothermic sham-injured controls (88.0 +/- 7.3%; p > 0.10). Fluid percussion injury dramatically reduced MAP2 levels in the normothermic group (44.3 +/- 5.9%; p < 0.0005) compared with normothermic sham-injured controls. No significant reduction of MAP2 was seen in the hypothermic injured group (95.2 +/- 4.6%; compared with hypothermic sham-injured controls, p > 0.20). Although it is premature to infer any causal link, the data suggest that the attenuation of injury-induced MAP2 loss by hypothermia may contribute to its overall neuroprotective action.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , Hypothermia, Induced , Microtubule-Associated Proteins/metabolism , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Immunologic Techniques , Male , Rats , Rats, Sprague-Dawley , Wounds, Nonpenetrating/metabolism
7.
Stroke ; 24(2): 271-7; discussion 277-8, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8093648

ABSTRACT

BACKGROUND AND PURPOSE: Glutamate-induced excitotoxicity has been implicated as a causative factor for selective neuronal loss in ischemia and hypoxia. Toxic exposure of neurons to glutamate results in an extended neuronal depolarization that precedes delayed neuronal death. Because both delayed neuronal death and extended neuronal depolarization are dependent on calcium, we examined the effect of glutamate exposure on extended neuronal depolarization and calcium/calmodulin-dependent protein kinase II (CaM kinase II) activity. METHODS: Three-week-old cortical cell cultures from embryonic rats were exposed to 500 microM glutamate and 10 microM glycine or to control medium for 10 minutes. Cells were examined for neuronal toxicity, electrophysiology, and biochemical alterations. In one set of experiments, whole-cell current clamp recording was performed throughout the experiment. In a parallel experiment, cortical cultures were allowed to recover from glutamate exposure for 1 hour, at which time the cells were homogenized and CaM kinase II activity was assayed using standard techniques. RESULTS: Excitotoxic exposure to glutamate resulted in extended neuronal depolarization, which remained after removal of the glutamate. Glutamate exposure also resulted in delayed neuronal death, which was preceded by significant inhibition of CaM kinase II activity. The excitotoxic inhibition of CaM kinase II correlated with neuronal loss, was N-methyl-D-aspartate receptor-mediated, and was not due to autophosphorylation of the enzyme. CONCLUSIONS: Glutamate-induced delayed neuronal toxicity correlates with extended neuronal depolarization and inhibition of CaM kinase II activity. Because inhibition of CaM kinase II activity significantly preceded the histological loss of neurons, the data suggest that modulation of CaM kinase II activity may be involved in the cascade of events resulting in loss of calcium homeostasis and delayed neuronal death.


Subject(s)
Cerebral Cortex/cytology , Dizocilpine Maleate/pharmacology , Glutamates/toxicity , Neurons/physiology , Protein Kinase Inhibitors , Animals , Calcium-Calmodulin-Dependent Protein Kinases , Cell Death/physiology , Glutamic Acid , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/enzymology , Phosphorylation , Protein Kinases/metabolism , Rats , Time Factors
8.
J Neurotrauma ; 10(3): 287-95, 1993.
Article in English | MEDLINE | ID: mdl-8258841

ABSTRACT

Calcium-dependent excitotoxic processes contribute significantly to pathologic responses to traumatic brain injury (TBI). TBI causes neuronal depolarization and excessive excitatory neurotransmitter release, which may lead to increases in intracellular calcium levels. However, responses of calcium-dependent enzymes such as protein kinase C (PKC) following TBI are poorly understood. Since PKC plays an important role in signal transduction and maintenance of normal neuronal function, we investigated changes in PKC activity and protein levels following fluid percussion brain injury in rats. We observed a 23.1% increase in PKC activity 1 h postinjury and 80.7% increase in PKC activity 3 h postinjury. There was no statistically significant change in PKC activity 5 min and 24 h after injury. PKC immunolabelling studies detected a significant increase in PKC levels in membrane fractions 3 h but not 1 h after injury. Thus PKC activation is transiently increased following TBI and may play an important role in pathophysiologic responses to TBI.


Subject(s)
Brain Injuries/enzymology , Hippocampus/enzymology , Protein Kinase C/metabolism , Wounds, Nonpenetrating/enzymology , Animals , Brain Injuries/physiopathology , Cell Membrane/enzymology , Male , Rats , Rats, Sprague-Dawley , Time Factors , Wounds, Nonpenetrating/physiopathology
9.
J Neurochem ; 59(4): 1221-32, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1328515

ABSTRACT

The activity of multifunctional calcium/calmodulin-dependent protein kinase II (CaM kinase II) has recently been shown to be inhibited by transient global ischemia. To investigate the nature of ischemia-induced inhibition of the enzyme, CaM kinase II was purified to greater than 1,000-fold from brains of control and ischemic gerbils. The characteristics of CaM kinase II from control and ischemic preparations were compared by numerous parameters. Kinetic analysis of purified control and ischemic CaM kinase II was performed for autophosphorylation properties, ATP, magnesium, calcium, and calmodulin affinity, immunoreactivity, and substrate recognition. Ischemia induced a reproducible inhibition of CaM kinase II activity, which could not be overcome by increasing the concentration of any of the reaction parameters. Ischemic CaM kinase II was not different from control enzyme in affinity for calmodulin, Ca2+, Mg2+, or exogenously added substrate or rate of autophosphorylation. CaM kinase II isolated from ischemic gerbils displayed decreased immunoreactivity with a monoclonal antibody (immunoglobulin G3) directed toward the beta subunit of the enzyme. In addition, ischemia caused a significant decrease in affinity of CaM kinase II for ATP when measured by extent of autophosphorylation. To characterize further the decrease in ATP affinity of CaM kinase II, the covalent-binding ATP analog 8-azido-adenosine-5'-[alpha-32P]triphosphate was used. Covalent binding of 25 microM azido-ATP was decreased 40.4 +/-12.3% in ischemic CaM kinase II when compared with control enzyme (n = 5; p less than 0.01 by paired Student's t test). Thus, CaM kinase II levels for ischemia and control fractions were equivalent by protein staining, percent recovery, and calmodulin binding but were significantly different by immunoreactivity and ATP binding. The data are consistent with the hypothesis that ischemia induces a posttranslational modification that alters ATP binding in CaM kinase II and that results in an apparent decrease in enzymatic activity.


Subject(s)
Brain Ischemia/enzymology , Prosencephalon/blood supply , Protein Kinases/metabolism , Protein Processing, Post-Translational , Adenosine Triphosphate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases , Calmodulin/metabolism , Gerbillinae , Kinetics , Male , Phosphorylation , Protein Kinase Inhibitors , Protein Kinases/isolation & purification
10.
J Neurotrauma ; 9(3): 281-90, 1992.
Article in English | MEDLINE | ID: mdl-1474611

ABSTRACT

We examined microtubule-associated protein 2 (MAP2) levels in hippocampal and cortical tissue 3 h following moderate traumatic brain injury (TBI) in the rat. MAP2 levels were assayed by quantitative immunoreactivity in tissue fractions obtained from naive, sham-injured, or fluid percussion-injured animals. Tissues were homogenized in the presence of protease inhibitors (0.3 mM phenylmethylsulfonyl fluoride, PMSF), a specific calpain inhibitors (0.1 mM leupeptin), and chelators (2 mM ethylene glycol-bis-tetraacetic acid, EGTA; 1 mM ethylenedinitrilo-tetraacetic acid, EDTA) to eliminate in vitro MAP2 proteolysis during tissue processing. Compared to naive rats, sham injury had no effect on soluble MAP2 levels in either cortex (105.0 +/- 4.4% of naive value) or hippocampus (106.6 +/- 5.2% of naive value). However, TBI caused a significant (p < 0.005) decrease in hippocampal MAP2 levels (55.7 +/- 5.9% of sham-injured controls). The effect appeared to be regionally selective, since the MAP2 decrease did not occur in cortex (89.1 +/- 1.4%). The degree of MAP2 decrease in hippocampus was similar in both membrane (57.8%) and cytosolic (55.7%) fractions, ruling out the possibility of partitioning artifacts. The data suggest that sublethal alterations of neuronal structure and function caused by MAP2 degradation may play an important role in the development of TBI-induced functional deficits. Since MAP2 is exclusively associated with the cytoskeleton in somal and dendritic compartments of neurons, the pathophysiology of sublethal magnitudes of TBI may also involve dendritic and somal dysfunction.


Subject(s)
Brain Injuries/metabolism , Hippocampus/metabolism , Microtubule-Associated Proteins/metabolism , Animals , Male , Microtubule-Associated Proteins/immunology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/immunology , Subcellular Fractions/metabolism
11.
Stroke ; 21(12): 1715-21, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2264078

ABSTRACT

We used brief bilateral carotid artery occlusion in gerbils to examine the effects of temperature on ischemia-induced inhibition of calcium/calmodulin-dependent protein kinase II activity and neuronal death. In normothermic (36 degrees C) gerbils, ischemia induced a severe loss of hippocampal CA1 pyramidal neurons measured 7 days after ischemia (28.4 neurons/mm, n = 10; control density in 10 naive gerbils 262.1 neurons/mm) and a significant decrease in forebrain calcium/calmodulin-dependent protein kinase II autophosphorylation measured 2 hours after ischemia (12.9 fmol/min, n = 6; control phosphorylation in six naive gerbils 23.5 fmol/min). The effect of temperature on these indicators of ischemic damage was examined by adjusting intracerebral temperature before and during the ischemic insult. Hyperthermic (39 degrees C) gerbils showed almost complete loss of neurons in the CA1 region (3.0 neurons/mm, n = 11) and extension of neuronal death into the CA2, CA3, and CA4 regions. In addition, hyperthermia exacerbated ischemia-induced inhibition of calcium/calmodulin-dependent protein kinase II activity (4.2 fmol/min, n = 6). Hypothermia (32 degrees C) protected against ischemia-induced CA1 pyramidal cell damage (257.0 neurons/mm, n = 20) and inhibition of calcium/calmodulin-dependent protein kinase II activity (26.0 fmol/min, n = 6). Our results are consistent with the hypothesis that loss of calcium/calmodulin-dependent protein kinase II activity may be a critical event in the development of ischemia-induced cell death.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Neurons/physiology , Temperature , Animals , Brain/enzymology , Brain/metabolism , Brain Ischemia/enzymology , Calmodulin-Binding Proteins/metabolism , Cell Survival , Gerbillinae , Hippocampus/pathology
12.
Stroke ; 21(11 Suppl): III112-6, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2173173

ABSTRACT

Cerebral ischemia produces a disruption of calcium homeostasis in neurons. This may explain the extreme sensitivity of these cells to ischemic insult. Prolonged increases in calcium levels may produce irreversible damage to the cell by altering important calcium-dependent enzyme systems such as calcium/calmodulin-dependent protein kinase II. Five minutes of acute forebrain ischemia in the gerbil produced a significant decrease in calcium/calmodulin-dependent protein kinase II activity as early as 10 seconds postischemia and persisting up to 7 days after insult. Because hypothermia protects against ischemia-induced cell death in the gerbil, we examined the effect of ischemia on cell death and calcium/calmodulin-dependent protein kinase II at different intracerebral temperatures: hyperthermia (39 degrees C), normothermia (36 degrees C), and hypothermia (32 degrees C). In ischemic animals, hyperthermia produced severe loss of neurons in CA1 and moderate loss in CA3-CA4 subregions. Normothermia in ischemic animals produced severe loss of neurons in the CA1 subregion. Hypothermic ischemic animals showed no significant loss of neurons in any hippocampal region. Ischemia produced a severe decrease (17 +/- 6% of control) in calcium/calmodulin-dependent kinase II activity in hyperthermic animals, a moderate decrease (55 +/- 15% of control) in normothermic animals, and no decrease of enzyme activity in hypothermic animals. Thus, lowering and raising intracerebral temperature decreased and increased, respectively, the extent of ischemia-induced damage in the gerbil. Because ischemia-induced effects on calcium/calmodulin-dependent protein kinase II activity are rapid and long-lasting, hypothermia may protect through preservation of calcium/calmodulin-dependent protein kinase II activity.


Subject(s)
Brain Ischemia/enzymology , Protein Kinases/metabolism , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Calcium-Calmodulin-Dependent Protein Kinases , Cell Survival , Gerbillinae
13.
Stroke ; 20(11): 1545-52, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2815190

ABSTRACT

We looked at FiO2, choice of anesthetic, nutritional status, and body temperature in a gerbil model of forebrain ischemia to determine their effect on data interpretation, ischemic outcome, and extent of pharmacologic protection. We subjected 484 gerbils to 5 minutes of forebrain ischemia under different experimental conditions. The gerbils were anesthetized with 3% halothane and inspired 21% O2, 37% O2 and 60% N2O, or 97% O2. Six groups of gerbils pretreated with 200 mg/kg phenytoin or 2 ml/kg polyethylene glycol (vehicle) underwent ischemia in the fasted or fed state. Three groups of gerbils receiving no pretreatment underwent ischemia with rectal temperatures of 32-33 degrees C, 34-35 degrees C, or 37 degrees C. We counted intact neurons in the CA1 hippocampal sector in brains fixed on Day 7 after ischemia. t tests of square-root-transformed cell counts were used to assess the effect of hypothermia, and analysis of variance of the transformed data was used to test for the effects of phenytoin, FiO2, and nutritional status. Phenytoin pretreatment provided significant protection from CA1 neuron loss in all groups tested (p less than 0.001), but the degree of protection varied from 20% to 44%. In spite of significantly higher serum glucose concentrations in fed than in fasted gerbils (173 and 118 mg/dl, respectively), we found no significant effect of nutritional status upon neuron loss in phenytoin- or vehicle-pretreated gerbils. An FiO2 of 21% significantly decreased the number of viable neurons in both vehicle- and phenytoin-pretreated groups (p less than 0.03), despite the lack of an effect of hypoxemia on arterial blood gases.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Ischemic Attack, Transient/drug therapy , Phenytoin/therapeutic use , Anesthesia , Animals , Blood Glucose/metabolism , Body Temperature , Carotid Arteries , Constriction , Fasting , Gerbillinae , Halothane , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Neurons/pathology , Oxygen/administration & dosage
14.
Brain Res ; 483(1): 143-8, 1989 Mar 27.
Article in English | MEDLINE | ID: mdl-2706503

ABSTRACT

Brief bilateral carotid occlusion in the gerbil produces forebrain ischemia that results in almost complete neuronal destruction in the CA1 sector of the hippocampus. Treatment with phenytoin (200 mg/kg) blocked the ischemia-induced neuronal death. The average density of CA1 pyramidal neurons (cells/mm CA1) was 253.6 +/- 4.4 in the sham surgery group, 12.3 +/- 3.4 in the ischemia group, and 119.5 +/- 16.6 in the group treated with phenytoin before ischemia. Thus, phenytoin reduced ischemia-produced neuronal loss in hippocampal CA1 by 44.4% (P less than 0.001). The plasma levels of phenytoin that produced this effect ranged from 28.1 to 45.0 mg per liter, with a mean phenytoin level of 34.7 +/- 1.7 mg/l (n = 10). The results suggest that phenytoin may be a clinically useful cerebroprotective agent.


Subject(s)
Hippocampus/physiopathology , Ischemic Attack, Transient/drug therapy , Phenytoin/therapeutic use , Animals , Cell Survival , Dose-Response Relationship, Drug , Gerbillinae , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Male , Phenytoin/pharmacokinetics
15.
J Neurotrauma ; 6(2): 71-81, 1989.
Article in English | MEDLINE | ID: mdl-2769771

ABSTRACT

One hundred and one rats were administered either D, alpha-tocopheryl succinate plus polyethylene glycol (PEG), PEG, or saline 30 min prior to or 5 min after moderate fluid percussion brain injury. Mortality rates, performance on beam balance and beam-walking tasks, and body weight were assessed daily for 10 days. With preinjury administration, mortality rate was reduced from 31% with saline to 9% with PEG and 9% with D, alpha-tocopheryl succinate plus PEG. With postinjury administration, mortality rate was reduced from 36% with saline to 20% with PEG and to 10% with the D, alpha-tocopheryl succinate plus PEG combination. With administration prior to injury, PEG and D, alpha-tocopheryl succinate plus PEG reduced the deficits seen on beam balance testing on days 1-3 after injury. On beam walking, PEG and D, alpha-tocopheryl succinate plus PEG reduced deficits compared to those in saline-injected animals on days 1 and 2 and on day 1 after injury, respectively. A strongly protective effect of PEG and of D, alpha-tocopheryl succinate plus PEG was seen with preinjury administration. With postinjury administration, D, alpha-tocopheryl succinate plus PEG reduced deficits on beam balance testing compared to animals receiving both saline and PEG on days 1-3 after injury. On beam-walking latencies, D, alpha-tocopheryl succinate plus PEG reduced deficits on days 1 and 2 after injury compared to saline and to PEG. Both PEG and D, alpha-tocopheryl succinate plus PEG reduced weight loss after injury compared to saline. The protective effects of these agents and their relatively low toxicity and high lipid solubility give them potential for the treatment of human head injury.


Subject(s)
Brain Injuries/drug therapy , Polyethylene Glycols/therapeutic use , Vitamin E/analogs & derivatives , Animals , Brain Injuries/mortality , Brain Injuries/physiopathology , Drug Therapy, Combination , Male , Rats , Rats, Inbred Strains , Tocopherols , Vitamin E/therapeutic use
16.
Brain Res ; 447(1): 159-63, 1988 Apr 26.
Article in English | MEDLINE | ID: mdl-2838127

ABSTRACT

Acute forebrain ischemia in the gerbil produced a significant decrease in endogenous Ca2+-dependent hippocampal protein phosphorylation in comparison to sham and naive control animals. Inhibition of Ca2+-dependent phosphorylation was demonstrated as early as 2 h post-ischemia and shown to persist up to 7 days following the ischemic insult. The ischemia-induced loss of Ca2+-dependent phosphorylation was only seen in brain regions affected by the localized ischemia and not in non-ischemic brain regions from the same animal. These results suggest that an early event in the development of ischemia-induced neuronal death is a decrease in Ca2+-dependent protein phosphorylation.


Subject(s)
Brain/enzymology , Ischemic Attack, Transient/enzymology , Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases , Gerbillinae , Male , Nerve Tissue Proteins/metabolism , Organ Specificity , Phosphorylation , Reference Values
17.
J Cell Physiol ; 131(2): 226-34, 1987 May.
Article in English | MEDLINE | ID: mdl-3034922

ABSTRACT

A series of benzodiazepines was evaluated for their capacity to induce the differentiation of HL-60 acute promyelocytic leukemia cells. Benzodiazepines were effective initiators of maturation in the concentration range of 50 to 150 microM. The possible involvement of benzodiazepine receptors in mediating the differentiation induced by these agents was investigated. The presence of high affinity, peripheral type benzodiazepine binding sites (KD = 7.3 nM, TB = 14.5 pmol/mg protein with Ro5-4864) was demonstrated in HL-60 membranes. The occupancy of peripheral type high affinity benzodiazepine receptors by various benzodiazepines showed some correlation (r = 0.76) with their differentiation-inducing capabilities, but binding potencies were 1,000-fold higher than the concentrations required to produce differentiation. A class of benzodiazepine receptors with lower binding affinity was also detected in HL-60 membranes (KD = 28.6 microM; TB = 199 pmol/mg protein with diazepam). A higher level of correlation (r = 0.88) was demonstrated between benzodiazepine occupancy of these lower affinity receptors and the capacity to induce maturation. Significantly, benzodiazepine concentrations needed for low affinity binding and induction of differentiation were the same (25-200 microM), suggesting that low affinity benzodiazepine receptors may be involved in the induction process. We have shown that the molecular form responsible for the induction of the differentiation of HL-60 cells to mature forms by 6-thioguanine (TGua) is the free base, TGua, itself [Ishiguro, Schwartz, and Sartorelli (1984) J. Cell. Physiol., 121:383-390]. Since hypoxanthine (Hyp) and inosine (Ino) have been identified as putative endogenous ligands for high affinity benzodiazepine receptors in brain tissue, the potential involvement of benzodiazepine receptors in the differentiation of HL-60 cells by the purines was investigated. Physiological purines such as Hyp and Ino were inactive in displacing the benzodiazepines from their high and low affinity binding sites in HL-60 membranes. In contrast, TGua caused inhibition of benzodiazepine binding to high and low affinity sites. The inhibition of Ro5-4864 binding to high affinity binding sites by TGua appeared to be due to the binding of TGua to membranes through the formation of a mixed disulfide between the 6-thiopurine and protein thiols, since the inhibition was reversed by the presence of 2-mercaptoethanol. The findings suggest a possible relationship between the occupancy of benzodiazepine receptors by TGua and the induction of leukemic cell differentiation.


Subject(s)
Benzodiazepines/pharmacology , Leukemia, Myeloid, Acute/pathology , Purines/pharmacology , Receptors, GABA-A/physiology , Benzodiazepinones/metabolism , Cell Differentiation/drug effects , Cell Line , Humans , Isoquinolines/metabolism , Pyrazoles/metabolism , Pyridazines/metabolism
18.
Yale J Biol Med ; 60(2): 99-106, 1987.
Article in English | MEDLINE | ID: mdl-2437710

ABSTRACT

Calcium is a major second messenger in neurons and modulates many neuronal functions, including protein phosphorylation, phospholipid metabolism, cytoskeletal activity, and neurotransmitter release. These important events, which regulate neuronal activity, are directly dependent on the influx of extracellular calcium through voltage-sensitive calcium channels (VSCCs) in the neuronal membrane. Modulation of VSCC function represents an important strategy for regulating neuronal excitability. Although substantial evidence supports the ability of dihydropyridines to block VSCCs and contractility in cardiovascular tissue, their ability to block the majority of neuronal VSCCs remains controversial. Benzodiazepines, and other anticonvulsants, block depolarization-dependent 45Ca uptake through VSCCs in brain synaptosome preparations. In addition, benzodiazepines reduce voltage-gated calcium conductance as determined by voltage clamp studies of identified invertebrate neurons. Inhibition of VSCC activity may be an important mechanism by which these compounds produce their anticonvulsant and sedative effects. Intrasomal injection of calcium-calmodulin-dependent protein kinase modulates calcium conductance in invertebrate neurons, suggesting that protein phosphorylation may be an endogenous regulatory mechanism of VSCC activity. Developing novel pharmacological approaches to regulating VSCCs and understanding the endogenous regulatory mechanisms may lead to new therapeutic approaches to the treatment of neurological diseases.


Subject(s)
Anticonvulsants/pharmacology , Brain/physiology , Calcium/physiology , Ion Channels/physiology , Animals , Benzodiazepines/pharmacology , Biological Transport/drug effects , Electric Conductivity , Leeches , Membrane Potentials , Phosphoproteins/physiology , Protein Kinases/metabolism , Snails , Synaptosomes/metabolism
19.
Adv Exp Med Biol ; 221: 409-25, 1987.
Article in English | MEDLINE | ID: mdl-2829586

ABSTRACT

A type II calmodulin-dependent protein kinase (CaM kinase II) has been characterized in the synaptic region and may mediate some of the effects of Ca2+ on neuronal excitability. The activity of CaM kinase II is inhibited by anticonvulsant compounds and may be the molecular basis of their neuro-modulatory effects. The direct injection of purified CaM kinase II into invertebrate neurons has demonstrated that this kinase can directly alter specific ion conductances and neuronal activity. A long-lasting decrease in CaM kinase II activity is associated with septal kindling, an experimental model of epilepsy and long-term memory. In summary, CaM kinase II appears to be a central mediator of the effects of Ca2+ on neuronal function. Further investigation of this enzyme and its effects on neuronal activity may provide a molecular insight into an endogenous mechanism for modulating some of the effects of Ca2+ on neuronal excitability and may increase our understanding of the complex regulatory mechanisms that underlie the pathogenesis of seizure discharge and its regulation by anticonvulsant compounds.


Subject(s)
Neurons/physiology , Protein Kinases/physiology , Seizures/physiopathology , Animals , Calcium/physiology , Calcium-Calmodulin-Dependent Protein Kinases , Epilepsy/physiopathology , Homeostasis , Humans
20.
Ann N Y Acad Sci ; 477: 238-46, 1986.
Article in English | MEDLINE | ID: mdl-2433983

ABSTRACT

Anticonvulsants are neuronal stabilizing compounds that exhibit multiple clinical effects, including anticonvulsant, anxiolytic, sedative, and muscle-relaxant properties. This complex therapeutic picture complicates the treatment of seizure disorders in individuals with mental and developmental disorders, and frequently impairs the routine integration into society for these individuals. In order to improve the therapeutic effectiveness of these compounds, it is necessary to identify their precise molecular actions on the neuronal membrane and their effects on neuronal function. We have identified two major classes of low-affinity BZ binding sites that seem to function as generalized anticonvulsant receptors and that may mediate the anticonvulsant and sedative effects produced by these compounds. The identification of these binding sites and their anticonvulsant binding profile may clarify the complex picture of anticonvulsant mechanisms and elucidate the site(s) at which anticonvulsants produce their inhibition of MES-induced seizures and sedative effects. We will continue to examine the physiological changes induced by anticonvulsant binding at these BZ binding sites that may be a foundation for understanding the molecular basis of sedation and MES-induced seizure inhibition. Specifically, we will investigate the specific membrane components associated with the inhibition of Ca2+ channels, Na+ channel rectification, and CaM kinase II. If these goals can be achieved, then model systems could be developed to screen potential anticonvulsant or sedative compounds in the search for more effective therapeutic drugs.


Subject(s)
Anticonvulsants/pharmacology , Brain/drug effects , Animals , Ca(2+) Mg(2+)-ATPase/antagonists & inhibitors , Calcium/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Ion Channels/metabolism , Phosphorylation , Rats , Receptors, GABA-A/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...