Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Behav Brain Res ; 281: 358-63, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25523029

ABSTRACT

BACKGROUND: Depression is a common and important cause of morbidity, and results in a significant economic burden. Recent human studies have demonstrated that that depression is contagious, and depression in family and friends might cumulatively increase the likelihood that a person will exhibit depressive behaviors. The mechanisms underlying contagion depression are poorly understood, and there are currently no animal models for this condition. METHODS: Rats were divided into 3 groups: depression group, contagion group, and control group. After induction of depression by 5 weeks of chronic unpredictable stress, rats from the contagion group were housed with the depressed rats (1 naïve rat with 2 depressed rats) for 5 weeks. Rats were then subjected to sucrose preference, open field, and forced swim tests. RESULTS: The sucrose preference was significantly reduced in the depressed rats (p<0.01) and contagion depression rats (p<0.01). Climbing time during forced swim test was reduced in the depression and contagion depression groups (p<0.001), whereas immobility time was significantly prolonged in only the depression group (p<0.001). Rats in both the depression (p<0.05) and depression contagion group (p<0.005) had decreased total travel distance and decreased mean velocity in the open field test, whereas the time spent in the central part was significantly shorter in only the depression group (p<0.001). CONCLUSIONS: In this study, for the first time we demonstrated depression contagion in an animal model. A reliable animal model may help better understand the underlying mechanisms of contagion depression, and may allow for future investigations of the studying therapeutic modalities.


Subject(s)
Depression/psychology , Interpersonal Relations , Restraint, Physical/psychology , Stress, Psychological/psychology , Animals , Behavior, Animal , Disease Models, Animal , Food Preferences/psychology , Locomotion , Male , Motor Activity , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Sucrose , Swimming/psychology , Time Factors
2.
J Neural Transm (Vienna) ; 121(8): 971-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24623040

ABSTRACT

It is well known that abnormally elevated glutamate levels in the brain are associated with secondary brain injury following acute and chronic brain insults. As such, a tight regulation of brain glutamate concentrations is of utmost importance in preventing the neurodegenerative effects of excess glutamate. There has been much effort in recent years to better understand the mechanisms by which glutamate is reduced in the brain to non-toxic concentrations, and in how to safely accelerate these mechanisms. Blood glutamate scavengers such as oxaloacetate, pyruvate, glutamate-oxaloacetate transaminase, and glutamate-pyruvate transaminase have been shown to reduce blood glutamate concentrations, thereby increasing the driving force of the brain to blood glutamate efflux and subsequently reducing brain glutamate levels. In the past decade, blood glutamate scavengers have gained increasing international interest, and its uses have been applied to a wide range of experimental contexts in animal models of traumatic brain injury, ischemic stroke, subarachnoid hemorrhage, epilepsy, migraine, and malignant gliomas. Although glutamate scavengers have not yet been used in humans, there is increasing evidence that their use may provide effective and exciting new therapeutic modalities. Here, we review the laboratory evidence for the use of blood glutamate scavengers. Other experimental neuroprotective treatments thought to scavenge blood glutamate, including estrogen and progesterone, beta-adrenergic activation, hypothermia, insulin and glucagon, and hemodialysis and peritoneal dialysis are also discussed. The evidence reviewed here will hopefully pave the way for future clinical trials.


Subject(s)
Brain Diseases/therapy , Brain/drug effects , Brain/metabolism , Glutamic Acid/blood , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Animals , Brain Diseases/drug therapy , Humans , Neuroprotective Agents/therapeutic use
3.
Gynecol Endocrinol ; 29(10): 912-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23862584

ABSTRACT

The purpose of this study was to examine whether changes in estrogen and progesterone levels observed during normal pregnancy influence blood glutamate levels. One-hundred and sixteen pregnant women were divided into three groups based on gestational age: group 1 included women in their first trimester, group 2 included women in their second trimester, and group 3 included women in their third trimester. A single venous blood sample was collected and analyzed for concentrations of estrogen, progesterone, glutamate-pyruvate transaminase (GPT), glutamate-oxaloacetate transaminase (GOT), and glutamate. Concentrations of blood glutamate were significantly lower during the second trimester (p < 0.001) and third trimester (p < 0.001). Blood glutamate levels were inversely correlated with levels of estrogen and progesterone throughout pregnancy (p < 0.001). Levels of GOT and GPT remained stable during the course of pregnancy, apart from a moderate reduction in GPT during the third trimester. Increases in estrogen and progesterone levels during advanced stages of pregnancy were inversely correlated with maternal blood glutamate concentrations. Once a maximal blood glutamate-reducing effect was achieved, any additional estrogen and progesterone had a negligible effect on blood glutamate. This study demonstrates the glutamate-reducing effects of estrogen and progesterone, which is most likely not mediated by a GOT/GPT conversion mechanism.


Subject(s)
Estrogens/blood , Glutamic Acid/blood , Pregnancy/blood , Progesterone/blood , Adolescent , Adult , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Blood Glucose/analysis , Female , Humans , Pregnancy Trimester, First/blood , Pregnancy Trimester, Second/blood , Pregnancy Trimester, Third/blood , Young Adult
4.
Cogn Affect Behav Neurosci ; 13(4): 847-59, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23761136

ABSTRACT

Poststroke depression (PSD) is the most frequent psychological sequela following stroke. While previous studies describe the impact of age on brain infarct volume, brain edema, and blood-brain barrier (BBB) breakdown following ischemia, the role of age on PSD has yet to be described. Here, we examine the influence of age on PSD progression in a rat model of PSD by middle cerebral artery occlusion (MCAO). One hundred forty-three rats were divided into three groups. 48 rats 20 weeks of age underwent a sham procedure, 51 rats 20 weeks of age had MCAO, and 44 rats 22-26 months of age had MCAO. Groups were further divided into two subgroups. The first subgroup was used to measure infarct lesion volume, brain edema, and BBB breakdown at 24 h. In the second subgroup at 3 weeks after MCAO, rats were subjected to a sucrose preference test, two-way shuttle avoidance task, forced swimming test, and a brain-derived neurotrophic factor (BDNF) protein level measurement. Total and striatal infarct volume, brain edema, and BBB breakdown in the striatum were increased in older rats, as compared with younger rats. While both old and young rats exhibited depressive-like behaviors on each of the behavioral tests and lower BDNF levels post-MCAO, as compared with control rats, there were no differences between old and young rats. Although older rats suffered from larger infarct volumes, increased brain edema and more BBB disruption following MCAO, the lack of behavioral differences between young and old rats suggests that there was no effect of rat age on the incidence of PSD.


Subject(s)
Aging , Depression/etiology , Infarction, Middle Cerebral Artery/complications , Age Factors , Animals , Avoidance Learning/physiology , Brain/metabolism , Brain Edema/etiology , Brain Infarction/etiology , Brain Infarction/pathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Food Preferences , Infarction, Middle Cerebral Artery/pathology , Male , Neurologic Examination , Rats , Rats, Sprague-Dawley , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
5.
J Neurosurg Anesthesiol ; 25(3): 262-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23752045

ABSTRACT

BACKGROUND: Previous study has demonstrated the efficacy of hemodialysis in reducing blood glutamate levels. The purpose of the present study is to investigate whether peritoneal dialysis (PD) may be effective in lowering blood glutamate levels, which may serve as a potential tool for improving neurological function after brain injury. METHODS: Two liters of dialysis solution were infused over 10 minutes into 18 patients with stage V chronic kidney disease. Blood samples were collected immediately before initiation of PD, and hourly for a total of 5 blood samples. Blood samples were sent for determination of glutamate, creatinine, urea, glucose, glutamate oxaloacetate transaminase, and glutamate pyruvate transaminase. PD samples were collected and analyzed for glutamate, creatinine, urea, and glucose at the same time points as the blood samples. RESULTS: Blood glutamate concentrations were significantly reduced by 60 minutes after the infusion of dialysis solution (P<0.0001), whereas levels of glutamate in the dialysis solution were increased significantly by 60 minutes (P<0.0001). CONCLUSIONS: We demonstrated that PD is an effective modality in reducing blood glutamate concentrations. This method may be potentially utilized for the treatment of acute and chronic brain disorders that are accompanied by elevated glutamate in the brain's extracellular fluid. Considering the rapid saturation of the PD solution with glutamate, we recommend frequent dwelling of the PD solution in order to maintain low concentrations of blood glutamate.


Subject(s)
Glutamic Acid/blood , Nervous System Diseases/prevention & control , Peritoneal Dialysis , Adult , Aged , Aged, 80 and over , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Bicarbonates/blood , Blood Glucose/metabolism , Blood Urea Nitrogen , Creatinine/blood , Female , Humans , Hydrogen-Ion Concentration , Male , Middle Aged
6.
J Neurosurg Anesthesiol ; 25(2): 174-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23295267

ABSTRACT

BACKGROUND: The exact mechanism of hypothermia-induced neuroprotection has not been determined yet; however, we hypothesized that it may be mediated by a blood glutamate-scavenging effect. Here, we examine the effect of hypothermic conditions (mild, moderate, and deep) on blood glutamate levels in naive rats. To identify the mechanism of hypothermia-induced glutamate reduction, we also measured concentrations of glutamate oxaloacetate transaminase (GOT) and glutamate pyruvate transaminase (GPT), the primary regulators of glutamate concentration in blood. METHODS: Rats were anesthetized with isoflurane, and their rectal temperature was maintained for 6 hours at 36 to 37°C, 33 to 36°C, 30 to 32°C, 18 to 22°C, or was not maintained artificially. At 6 hours, active cooling was discontinued and rats were allowed to rewarm. There were 12 rats in each group for a total of 60 rats. Blood samples were drawn at 0, 3, 6, 12, 24, and 48 hours for the determination of blood glutamate, GOT, and GPT levels. RESULTS: A strong correlation between body temperature and blood glutamate levels was observed (P<0.001). Mild (33 to 36°C) and moderate (30 to 32°C) hypothermia led to reduced blood glutamate levels (P<0.001). Deep hypothermia (18 to 22°C) was associated with significant elevations in blood glutamate levels (P<0.001). Hypothermia, irrespective of the degree, led to elevations in GOT in plasma (P<0.001). CONCLUSIONS: Mild and moderate hypothermia led to a reduction in blood glutamate levels in rats, whereas deep hypothermia was associated with a significant elevation in blood glutamate levels. We further demonstrated an elevation of GOT and GPT levels, supporting their involvement in reducing blood glutamate by the conversion of glutamate to 2-ketoglutarate. We suggest that the neuroprotective properties of hypothermia may be partially because of a blood glutamate-scavenging mechanism.


Subject(s)
Glutamic Acid/blood , Hypothermia, Induced , Alanine Transaminase/blood , Anesthesia, General , Anesthetics, Inhalation , Animals , Aspartate Aminotransferases/blood , Body Temperature/drug effects , Body Temperature/physiology , Isoflurane , Male , Rats , Rats, Sprague-Dawley
7.
Brain Res ; 1491: 109-16, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23123210

ABSTRACT

Despite significant advancements in the understanding of the pathophysiological mechanisms of subarachnoid hemorrhage (SAH), little is known about the emotional consequences. The primary goal of this study was to describe the locomotor and behavioral patterns in rats following both a single-injection and double-injection model of SAH. In 48 rats, SAH was induced by injecting 0.3 ml of autologous arterial blood into the cisterna magnum (single-hemorrhagic model). In 24 of these rats, post-SAH vasospasm was induced by a repeated injection of blood into the cisterna magnum 24h later (double-hemorrhagic model). In 24 additional rats, 0.3 ml of saline was injected into the cisterna magnum (sham group). Neurological performance was assessed at 24, 48 h, 1, 2 and 3 weeks after SAH. Four behavioral tests were performed for 3 weeks after SAH for the duration of 6 consequent days, in the following order: open field test, sucrose preference test, elevated plus maze test and forced swimming test. Following both, a single and double-hemorrhagic models of SAH, rats were found to have significant behavioral abnormalities on the open field test, sucrose preference test, elevated plus maze test, and forced swimming test. A more prominent disability was found in rats that underwent the double-hemorrhagic model of SAH than rats that underwent the single-hemorrhagic model. Both a single and double injection model of rats SAH are associated with significant behavioral disturbances including locomotor abnormalities, depressive behavior and increased anxiety, even as early as 3 weeks after SAH.


Subject(s)
Behavior, Animal/physiology , Nervous System Diseases/pathology , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/psychology , Animals , Anxiety/psychology , Blood Transfusion , Cisterna Magna/physiology , Data Interpretation, Statistical , Depression/psychology , Exploratory Behavior/physiology , Food Preferences/physiology , Food Preferences/psychology , Locomotion/physiology , Male , Motor Activity/physiology , Rats , Rats, Sprague-Dawley , Sucrose , Swimming/psychology , Vasospasm, Intracranial/etiology , Vasospasm, Intracranial/psychology
8.
Hepatol Int ; 7(2): 721-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-26201806

ABSTRACT

PURPOSE: Finding an optimal biomarker for the noninvasive evaluation of acute liver injury (ALI) may be of great value in predicting clinical outcomes and investigating potential treatments. We investigated cell-free DNA (CFD) as a potential biomarker to predict carbon tetrachloride-induced ALI in rats. METHODS: Forty-five Sprague-Dawley rats were randomly assigned to three groups. ALI was induced by carbon tetrachloride via a nasogastric tube at 1, 2.5, or 5 ml/kg of a 50 % solution. Fifteen additional rats underwent a sham procedure. Blood samples were drawn at time t which was 0 (baseline), 3, 6, 12, 24, 48, 72, 96, and 120 h for the measurements of CFD, glutamate-pyruvate transaminase (GPT), glutamate-oxaloacetate transaminase (GOT), and total bilirubin. Prothrombin time and histology were examined at 24 and 120 h following injection of 5 ml/kg carbon tetrachloride in 18 additional rats and in 10 control rats. RESULTS: CFD levels in rats subjected to carbon tetrachloride-induced ALI were significantly increased in all blood samples starting at 12 h after the induction of ALI (p < 0.001), reaching peak levels at 24 h. Blood GOT, GPT, and total bilirubin were elevated in all blood samples starting at 3 h after the induction of ALI (p < 0.0001), reaching peak levels by 48 h. A positive correlation was demonstrated between CFD levels and GOT (R (2) = 0.92), GPT (R (2) = 0.92), and total bilirubin (R (2) = 0.76). CFD levels correlated with liver damage seen on histological examination, as well as predicted liver damage, at 24 h after ALI. CONCLUSIONS: CFD may be a useful biomarker for the prediction and measurement of ALI. There is no evidence to suggest that CFD is superior to other available noninvasive biomarkers.

9.
Shock ; 38(6): 630-4, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143053

ABSTRACT

Treatment of combined traumatic brain injury and hypovolemic shock poses a particular challenge due to the possible conflicting consequences. While restoring diminished volume is the treatment goal for hypovolemia, maintaining and adequate cerebral perfusion pressure and avoidance of secondary damage remain a treatment goal for the injured brain. Various treatment modalities have been proposed, but the optimal resuscitation fluid and goals have not yet been clearly defined. In this study, we investigate the physiological and neurological outcomes in a rat model of combined traumatic brain injury and hypovolemic shock, submitted to treatment with varying amounts of fresh blood. Forty-eight male Lewis rats were divided into control and treatment groups. Traumatic brain injury was inflicted by a free-falling rod on the exposed cranium. Hypovolemia was induced by controlled hemorrhage of 30% blood volume. Treatment groups were treated by fresh whole blood with varying volumes, reaching resuscitation goals of a mean arterial blood pressure (MAP) of 80, 100, and 120 mmHg at 15 min. Mean arterial blood pressure was assessed at 60 min and neurological outcomes and mortality in the subsequent 48 h. At 60 min, MAP was highest for the group resuscitated most aggressively. Neurological outcomes and mortality inversely correlated with the aggressiveness of resuscitation. In this study, we find that mild resuscitation with goals of restoring MAP to 80 mmHg (which is lower than baseline) provided best results when considering hemodynamic stability, survival, and neurological outcomes. An aggressive resuscitation may be detrimental, inducing processes that eventually cause a significant decrease in survival.


Subject(s)
Arterial Pressure , Brain Injuries/physiopathology , Brain Injuries/therapy , Resuscitation , Animals , Blood Transfusion , Brain Injuries/complications , Disease Models, Animal , Male , Nervous System Diseases/etiology , Nervous System Diseases/physiopathology , Nervous System Diseases/therapy , Rats , Rats, Inbred Lew , Shock/complications , Shock/physiopathology , Shock/therapy , Trauma Severity Indices
10.
J Crit Care ; 27(6): 743.e1-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23084134

ABSTRACT

PURPOSE: The purpose of the present study is to investigate whether hemodialysis (HD) is effective in lowering blood glutamate levels. In addition, we examined the effect of HD on glutamate oxaloacetate transaminase (GOT) and glutamate pyruvate transaminase (GPT) levels in the blood and described the rate and pattern of blood glutamate clearance during HD. MATERIALS AND METHODS: Blood samples were taken from 45 patients with stage V chronic kidney disease immediately after initiation of HD and hourly, for a total of 5 blood samples. Samples were sent for determination of glutamate, glucose, GOT, GPT, hemoglobin, hematocrit, urea, and creatinine levels. A blood sample from 25 healthy volunteers without chronic renal failure was used as a control for the determination of baseline blood levels of glutamate, GOT, and GPT. RESULTS: Glutamate and GPT levels in patients on HD were higher at baseline compared with healthy controls (P < .001). In the first 3 hours after HD, there was a decrease in blood glutamate levels compared with baseline levels (P < .00001). At the fourth hour, there was an increase in blood glutamate levels compared with the third hour (P < .05). CONCLUSIONS: Hemodialysis may be a promising method of reducing blood glutamate levels.


Subject(s)
Glutamic Acid/blood , Kidney Failure, Chronic/therapy , Renal Dialysis , Adult , Aged , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Female , Humans , Male , Middle Aged
11.
Int J Mol Sci ; 13(8): 10041-10066, 2012.
Article in English | MEDLINE | ID: mdl-22949847

ABSTRACT

Brain insults are characterized by a multitude of complex processes, of which glutamate release plays a major role. Deleterious excess of glutamate in the brain's extracellular fluids stimulates glutamate receptors, which in turn lead to cell swelling, apoptosis, and neuronal death. These exacerbate neurological outcome. Approaches aimed at antagonizing the astrocytic and glial glutamate receptors have failed to demonstrate clinical benefit. Alternatively, eliminating excess glutamate from brain interstitial fluids by making use of the naturally occurring brain-to-blood glutamate efflux has been shown to be effective in various animal studies. This is facilitated by gradient driven transport across brain capillary endothelial glutamate transporters. Blood glutamate scavengers enhance this naturally occurring mechanism by reducing the blood glutamate concentration, thus increasing the rate at which excess glutamate is cleared. Blood glutamate scavenging is achieved by several mechanisms including: catalyzation of the enzymatic process involved in glutamate metabolism, redistribution of glutamate into tissue, and acute stress response. Regardless of the mechanism involved, decreased blood glutamate concentration is associated with improved neurological outcome. This review focuses on the physiological, mechanistic and clinical roles of blood glutamate scavenging, particularly in the context of acute and chronic CNS injury. We discuss the details of brain-to-blood glutamate efflux, auto-regulation mechanisms of blood glutamate, natural and exogenous blood glutamate scavenging systems, and redistribution of glutamate. We then propose different applied methodologies to reduce blood and brain glutamate concentrations and discuss the neuroprotective role of blood glutamate scavenging.


Subject(s)
Glutamic Acid/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Neuroprotective Agents/therapeutic use , Animals , Humans
12.
Anim Sci J ; 83(9): 656-62, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22943533

ABSTRACT

Several motor-function scales have been developed to assess neurological function in animal models of stroke, subarachnoid hemorrhage and closed head injury. We hypothesize that the location of arterial and venous catheters, even in the absence of brain injury, may impact rats' motor performance. Our study examined the effect of catheter location, rate of infection and the time required for catheter placement. We further describe an original technique of tail artery cannulation without exposure of the artery. Sixty-one rats were anesthetized and randomly assigned to one of seven groups, including no catheter, tail artery or artery + vein catheters, or femoral artery or artery + vein catheters. A neurological severity score (NSS) was determined at 1 h, 24 h and 48 h after surgical preparation or catheter placement. NSS at 1 h after placement of unilateral and bilateral femoral catheters was higher than the NSS observed at 1 h after placement of tail arterial and venous catheters (P < 0.01). The NSS also was higher at 24 h in the bilateral femoral catheter groups as compared with the tail catheter groups (P < 0.05). There were no differences in the NSS observed between the groups that had tail catheters and the sham group at 1 h, 24 h or 48 h. Infection rate at the site of catheter placement and the time required for catheter placement was also higher in the femoral catheter groups (P < 0.001). Thus, we propose that the line location may bias a study's results and lead to deceptive interpretations of neurological assessment following rat head injury. Compared to femoral vessels, tail blood vessels are preferable locations for lines placement.


Subject(s)
Catheterization, Peripheral/methods , Disease Models, Animal , Motor Activity/physiology , Animals , Arteries , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Stroke/physiopathology , Tail/blood supply , Veins
13.
Neurochem Res ; 37(10): 2198-205, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22846966

ABSTRACT

Traumatic brain injury (TBI) and stroke lead to elevated levels of glutamate in the brain that negatively affect the neurological outcomes in both animals and humans. Intravenous administration of glutamate-oxaloacetate transaminase (GOT) and glutamate-pyruvate transaminase (GPT) enzymes can be used to lower the blood glutamate levels and to improve the neurological outcome following TBI and stroke. The objective of this study was to analyze the pharmacokinetics and to determine the glutamate-lowering effects of GOT and GPT enzymes in naïve rats. We determined the time course of serum GOT, GPT, and glutamate levels following a single intravenous administration of two different doses of each one of the studied enzymes. Forty-six male rats were randomly assigned into one of 5 treatment groups: saline (control), human GOT at dose 0.03 and 0.06 mg/kg and porcine GPT at dose 0.6 and 1.2 mg/kg. Blood samples were collected at baseline, 5 min, and 2, 4, 8, 12, and 24 h after the drug injection and GOT, GPT and glutamate levels were determined. The pharmacokinetics of both GOT and GPT followed one-compartment model, and both enzymes exhibited substantial glutamate-lowering effects following intravenous administration. Analysis of the pharmacokinetic data indicated that both enzymes were distributed predominantly in the blood (central circulation) and did not permeate to the peripheral organs and tissues. Several-hour delay was present between the time course of the enzyme levels and the glutamate-lowering effects (leading to clock-wise hysteresis on concentration-effect curves), apparently due to the time that is required to affect the pool of serum glutamate. We conclude that the interaction between the systemically-administered enzymes (GOT and GPT) and the glutamate takes place in the central circulation. Thus, glutamate-lowering effects of GOT and GPT apparently lead to redistribution of the excess glutamate from the brain's extracellular fluid into the blood and can reduce secondary brain injury due to glutamate neurotoxicity. The outcomes of this study regarding the pharmacokinetic and pharmacodynamic properties of the GOT and GPT enzymes will be subsequently verified in clinical studies that can lead to design of effective neuroprotective treatment strategies in patients with traumatic brain diseases and stroke.


Subject(s)
Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Glutamates/blood , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Male , Rats , Rats, Sprague-Dawley
14.
Neurotherapeutics ; 9(3): 649-57, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22711471

ABSTRACT

Blood glutamate scavengers have been shown to effectively reduce blood glutamate concentrations and improve neurological outcome after traumatic brain injury and stroke in rats. This study investigates the efficacy of blood glutamate scavengers oxaloacetate and pyruvate in the treatment of subarachnoid hemorrhage (SAH) in rats. Isotonic saline, 250 mg/kg oxaloacetate, or 125 mg/kg pyruvate was injected intravenously in 60 rats, 60 minutes after induction of SAH at a rate of 0.1 ml/100 g/min for 30 minutes. There were 20 additional rats that were used as a sham-operated group. Blood samples were collected at baseline and 90 minutes after SAH. Neurological performance was assessed at 24 h after SAH. In half of the rats, glutamate concentrations in the cerebrospinal fluid were measured 24 h after SAH. For the remaining half, the blood brain barrier permeability in the frontal and parieto-occipital lobes was measured 48 h after SAH. Blood glutamate levels were reduced in rats treated with oxaloacetate or pyruvate at 90 minutes after SAH (p < 0.001). Cerebrospinal fluid glutamate was reduced in rats treated with pyruvate (p < 0.05). Neurological performance was significantly improved in rats treated with oxaloacetate (p < 0.05) or pyruvate (p < 0.01). The breakdown of the blood brain barrier was reduced in the frontal lobe in rats treated with pyruvate (p < 0.05) and in the parieto-occipital lobes in rats treated with either pyruvate (p < 0.01) or oxaloacetate (p < 0.01). This study demonstrates the effectiveness of blood glutamate scavengers oxaloacetate and pyruvate as a therapeutic neuroprotective strategy in a rat model of SAH.


Subject(s)
Antioxidants/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Oxaloacetic Acid/therapeutic use , Pyruvic Acid/therapeutic use , Subarachnoid Hemorrhage/complications , Animals , Antioxidants/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Disease Models, Animal , Glutamic Acid/blood , Glutamic Acid/cerebrospinal fluid , Male , Nervous System Diseases/blood , Nervous System Diseases/cerebrospinal fluid , Oxaloacetic Acid/blood , Pyruvic Acid/blood , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Subarachnoid Hemorrhage/blood , Subarachnoid Hemorrhage/cerebrospinal fluid , Subarachnoid Hemorrhage/drug therapy , Time Factors
15.
Early Hum Dev ; 88(9): 773-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22633534

ABSTRACT

BACKGROUND: Excess glutamate in the brain is thought to be implicated in the pathophysiology of fetal anoxic brain injury, yet little is known about the mechanisms by which glutamate is regulated in the fetal brain. This study examines whether there are differences between maternal and fetal glutamate concentrations, and whether a correlation between them exists. METHODS: 10 ml of venous blood was extracted from 87 full-term (>37 weeks gestation) pregnant women in active labor. Immediately after delivery of the neonate, 10 ml of blood from the umbilical artery and vein was extracted. Samples were analyzed for levels of glutamate, glutamate-oxaloacetate transaminase (GOT), and glutamate pyruvate transaminase (GPT). RESULTS: Fetal blood glutamate concentrations in both the umbilical artery and vein were found to be significantly higher than maternal blood (p<0.001). Similarly, fetal serum GOT levels in the umbilical artery and vein were found to be significantly higher than maternal GOT levels (p<0.001). The difference in GPT levels between maternal and fetal serum was not statistically significant. There was no difference in fetal glutamate, GOT or GPT between the umbilical artery and vein. There was an association observed between glutamate levels in maternal blood and glutamate levels in both venous (R=0.32, p<0.01) and arterial (R=0.33, p<0.05) fetal blood. CONCLUSIONS: This study demonstrated that higher baseline concentrations of blood glutamate are present in fetal blood compared with maternal blood, and this was associated with elevated GOT, but not GPT levels. An association was observed between maternal and fetal blood glutamate levels.


Subject(s)
Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Fetal Blood/chemistry , Glutamic Acid/blood , Adult , Clinical Enzyme Tests , Female , Fetal Distress/blood , Humans , Infant, Newborn , Male , Pregnancy
16.
Acta Neurobiol Exp (Wars) ; 72(4): 385-96, 2012.
Article in English | MEDLINE | ID: mdl-23377269

ABSTRACT

Excessive concentrations of L-glutamate (glutamate) have been found to posses neurotoxic properties. This study investigates how stress induced by strong physical exercise effects blood glutamate, 2-ketoglutarate, Alanine aminotransferase (ALT) and Aspartate Aminotransferase (AST) levels. The relationship between muscle damage caused by strong physical exercise and blood glutamate levels was also examined. Twenty-two healthy volunteers engaged in intense veloergometry ("spinning") for a duration of 60 minutes. Two 10 minute peaks of extremely intense exercise were performed at 10 minutes and 50 minutes after the start of exercise. After 60 minutes of exercise, volunteers were monitored for an additional 180 minutes in resting conditions. Blood samples for determination of glutamate and 2-ketoglutarate levels were collected prior to exercise and then every 30 min for entire experiment. Blood samples were also taken at those time points to measure glutamate, 2-ketoglutarate, AST, ALT, creatine phosphokinase (CPK), myoglobin, lactate and venous blood gas levels. Blood glutamate levels were significantly elevated throughout the exercise session (P less than 0.001) and then returned to baseline levels at the cessation of exercise. 2-ketoglutarate, a product of glutamate metabolism, reached significantly elevated levels at 30 minutes (P less than 0.01) from the start of exercise and remained elevated up to 240 minutes post exercise initiation (P less than 0.001). AST and ALT levels were elevated at 60 minutes when compared to baseline. AST levels remained elevated at 240 minutes, unlike ALT levels which returned to baseline values at 240 minutes. Strong physical exercise leads to a significant elevation in blood glutamate, most likely as a result of skeletal muscle damage. 2-ketoglutarate was also found to be elevated for long periods of time, reflecting an ongoing process of glutamate breakdown. Elevated concentrations of AST and ALT in plasma reflect the importance of these enzymes in the maintenance of stable blood glutamate concentrations.


Subject(s)
Exercise , Glutamic Acid/blood , Ketoglutaric Acids/blood , Adult , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Blood Gas Analysis , Blood Glucose , Blood Pressure/physiology , Body Temperature , Heart Rate/physiology , Humans , Lactic Acid/blood , Male , Myoglobin/blood , Time Factors , Young Adult
17.
J Neurosurg Anesthesiol ; 24(1): 30-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21979171

ABSTRACT

BACKGROUND: Isoflurane-anesthetized rats subjected to traumatic brain injury (TBI) show a transient reduction in blood L-glutamate levels. Having previously observed that isoproterenol produces a sustained decrease in blood glutamate levels in naive rats, we investigated the possible effects of nonselective and selective ß1 and ß2 adrenergic agonists and antagonists both on blood glutamate levels and on the neurological outcomes of rats subjected to TBI. METHODS: Rats received either 10 mL/kg of isotonic saline 1 hour after TBI, 50 µg/kg of isoproterenol pretreatment 30 minutes before TBI, 10 mg/kg of propranolol pretreatment 60 minutes before TBI, 10 mg/kg of metoprolol pretreatment 60 minutes before TBI, or 10 mg/kg of butaxamine pretreatment 40 minutes before TBI and 10 minutes before pretreatment with 50 µg/kg isoproterenol or 10 mg/kg of propranolol 60 minutes after TBI. A neurological severity score (NSS) was measured at 1, 24, and 48 hours after TBI. Blood glutamate, blood glucose, mean arterial blood pressure, and heart rate were measured at the time of drug injection, at the time of TBI, 60 minutes after TBI, and 90 minutes after TBI. RESULTS: Blood glutamate levels decreased spontaneously by 60 minutes after TBI in the control group (P<0.05), reverting to baseline levels by 90 minutes after TBI. A pretreatment with either 10 mg/kg of metoprolol 60 minutes before TBI or with 50 µg/kg of isoproterenol 30 minutes before TBI also reduced blood glutamate levels (P<0.05) both at 90 minutes after TBI and improved the NSS measured 24 and 48 hours after TBI in comparison with the control saline-treated group. However, a 10-mg/kg butoxamine pretreatment 40 minutes before TBI and 10 minutes before pretreatment with 50 µg/kg of isoproterenol or 10 mg/kg of propranolol 60 minutes before TBI neither affected blood glutamate levels across time after TBI nor caused any significant change in the NSS measured 24 and 48 hours after TBI in comparison with the control saline-treated group. A strong correlation (r(2)=0.73) was demonstrated between the percent decrease in blood glutamate levels at 90 minutes after TBI and the percent improvement of NSS measured 24 hours after TBI. CONCLUSIONS: The results suggest that the transient blood glutamate reduction seen after TBI is the result of a stress response and of the activation of the sympathetic nervous system through the ß2 adrenergic receptors, causing an increase of the brain-to-blood efflux of glutamate observed with excess brain glutamate levels after a brain insult. This strongly correlates with the neurological improvement observed 24 hours after TBI.


Subject(s)
Brain Injuries/blood , Glutamic Acid/blood , Nervous System Diseases/prevention & control , Receptors, Adrenergic, beta-2/physiology , Adrenergic beta-1 Receptor Agonists/therapeutic use , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Adrenergic beta-2 Receptor Agonists/therapeutic use , Adrenergic beta-2 Receptor Antagonists/therapeutic use , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Blood Glucose/metabolism , Blood Pressure/drug effects , Brain Injuries/complications , Butoxamine/therapeutic use , Head Injuries, Closed/blood , Head Injuries, Closed/complications , Heart Rate/drug effects , Hemodynamics/drug effects , Hemodynamics/physiology , Isoproterenol/therapeutic use , Male , Metoprolol/therapeutic use , Movement/drug effects , Movement/physiology , Nervous System Diseases/etiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
18.
J Neurotrauma ; 29(2): 261-7, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22149927

ABSTRACT

Traumatic brain injury (TBI) is a major cause of morbidity and mortality, and early predictors of neurological outcomes are of great clinical importance. Cell free DNA (CFD), a biomarker used for the diagnosis and monitoring of several diseases, has been implicated as a possible prognostic indicator after TBI. The purpose of this study was to determine the pattern and timing of CFD levels after TBI, and whether a relationship exists between the level of CFD and brain edema and neurological outcomes. Thirty-nine Sprague-Dawley rats were randomly assigned to two groups: rats in group 1 (sham group) were anesthetized and had a scalp incision without TBI, and rats in group 2 were anesthetized and had a scalp incision with TBI, which was induced by using a weight drop model that causes diffuse brain injury. A neurological severity score (NSS) was assessed at 1, 24, and 48 h after TBI. CFD was measured via blood samples drawn at t=0 (baseline), 12, 24, 48, 72, and 120 h after TBI. At 48 h after TBI, brain edema was determined in a subgroup of 11 rats by calculating the difference between rats' wet and dry brain weight. The significance of comparisons between and within groups (CFD levels, brain water content, and NSS) were determined using the Kruskal-Wallis, Mann-Whitney and Student t test. The correlation between CFD levels and the NSS, as well as between CFD levels and the extent of brain edema, was calculated using the Spearman and Pearson tests, respectively. Compared with baseline levels, the CFD levels in rats subjected to TBI were significantly increased at 24 and 48 h after TBI (p<0.01 and p<0.05, respectively). A positive correlation was demonstrated between CFD levels 24 h following TBI and the extent of brain edema (r=0.63, p<0.05), as well as between CFD levels and the NSS (r=0.79, p<0.005). In this study, we demonstrated an increase in CFD levels after TBI, as well as a correlation between CFD levels and brain edema and NSS. CFD levels may provide a quick, reliable, and simple prognostic indicator of neurological outcome in animals after TBI. Its role in humans has not been clearly elucidated, but has potentially significant clinical implications.


Subject(s)
Brain Damage, Chronic/blood , Brain Edema/blood , Brain Injuries/blood , DNA/blood , Animals , Biomarkers/blood , Brain Damage, Chronic/diagnosis , Brain Damage, Chronic/pathology , Brain Edema/diagnosis , Brain Edema/pathology , Brain Injuries/diagnosis , Brain Injuries/pathology , Cell-Free System/metabolism , Cell-Free System/pathology , Disease Models, Animal , Male , Predictive Value of Tests , Rats , Rats, Sprague-Dawley
19.
Anesthesiology ; 116(1): 73-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22129535

ABSTRACT

BACKGROUND: Decreasing blood glutamate concentrations after traumatic brain injury accelerates brain-to-blood glutamate efflux, leading to improved neurologic outcomes. The authors hypothesize that treatment with blood glutamate scavengers should reduce neuronal cell loss, whereas administration of glutamate should worsen outcomes. The authors performed histologic studies of neuronal survival in the rat hippocampus after traumatic brain injury and treatment with blood glutamate scavengers. METHODS: Traumatic brain injury was induced on anesthetized male Sprague-Dawley rats by a standardized weight drop. Intravenous treatment groups included saline (control), oxaloacetate, pyruvate, and glutamate. Neurologic outcome was assessed using a Neurological Severity Score at 1 h, and 1, 2, 7, 14, 21, 28 days. Blood glutamate was determined at baseline and 90 min. Four weeks after traumatic brain injury, a histologic analysis of surviving neurons was performed. RESULTS: Oxaloacetate and pyruvate treatment groups demonstrated increased neuronal survival (oxaloacetate 2,200 ± 37, pyruvate 2,108 ± 137 vs. control 1,978 ± 46, P < 0.001, mean ± SD). Glutamate treatment revealed decreased neuronal survival (1,715 ± 48, P < 0.001). Treatment groups demonstrated favorable neurologic outcomes at 24 and 48 h (Neurological Severity Score at 24 and 48 h: 5.5 (1-8.25), 5 (1.75-7.25), P = 0.02 and 3(1-6.5), 4 (1.75-4.5), P = 0.027, median ± corresponding interquartile range). Blood glutamate concentrations were decreased in the oxaloacetate and pyruvate treatment groups. Administration of oxaloacetate and pyruvate was not shown to have any adverse effects. CONCLUSIONS: The authors demonstrate that the blood glutamate scavengers oxaloacetate and pyruvate provide neuroprotection after traumatic brain injury, expressed both by reduced neuronal loss in the hippocampus and improved neurologic outcomes. The findings of this study may bring about new therapeutic possibilities in a variety of clinical settings.


Subject(s)
Brain Injuries/pathology , Glutamic Acid/blood , Glutamic Acid/pharmacology , Hippocampus/injuries , Hippocampus/pathology , Oxaloacetic Acid/pharmacology , Pyruvic Acid/pharmacology , Animals , Behavior, Animal/physiology , Blood Gas Analysis , Blood Glucose/metabolism , Brain/pathology , Cell Survival/drug effects , Hemodynamics/physiology , Hemoglobins/metabolism , Linear Models , Male , Neurologic Examination , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Treatment Outcome
20.
Intensive Care Med ; 38(1): 137-44, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22124768

ABSTRACT

PURPOSE: Estrogen has been shown to possess neuroprotective properties both in vitro and in vivo. Traumatic brain injury (TBI) in ovulating females results in favorable neurological outcomes when compared to males with similar insults. The brain-to-blood glutamate gradient removes excess glutamate from brain extracellular fluids (ECF). Enhancing this gradient leads to improved neurological outcomes following TBI. In this study we investigate the effect of female gonadal steroids on blood glutamate levels and neurological outcomes. METHODS: Forty male Sprague-Dawley rats were assigned to one of five groups: (1) sham, (2) Premarin treatment, (3) TBI, (4) TBI + Premarin treatment, and (5) TBI + Premarin pretreatment. TBI was induced, and estrogen and glutamate levels were determined at 0, 60, 120, 135, and 150 min. Neurological recovery was evaluated using the Neurological Severity Score (NSS) at 1 h and reassessed at 24 h post TBI. RESULTS: Premarin treatment groups demonstrated a decline in blood glutamate levels by 60 min. This decline was found to be more pronounced in the TBI + Premarin group, which maintained the decline throughout the experiment. At 120 min, the difference between groups was most pronounced (TBI + Premarin 99 ± 36 µM/l vs. control 200 ± 46 µM/l, p < 0.01). Neurological recovery was significantly better in the Premarin treatment group (NSS at 24 h 6 ± 1 vs. control 11 ± 1). CONCLUSIONS: Premarin injected into male rats significantly decreases blood glutamate levels in rats suffering TBI. This decrease is associated with improved neurological outcomes, thus implicating the role of estrogen in neuroprotection.


Subject(s)
Brain Injuries/drug therapy , Estrogens, Conjugated (USP)/pharmacology , Estrogens/pharmacology , Glutamates/blood , Animals , Brain Injuries/metabolism , Dose-Response Relationship, Drug , Estrogens/administration & dosage , Estrogens, Conjugated (USP)/administration & dosage , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...