Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36293002

ABSTRACT

The mechanisms involved in the interaction of PrP 106-126, a peptide corresponding to the prion protein amyloidogenic region, with the blood-brain barrier (BBB) were studied. PrP 106-126 treatment that was previously shown to impair BBB function, reduced cAMP levels in cultured brain endothelial cells, increased nitric oxide (NO) levels, and changed the activation mode of the small GTPases Rac1 (inactivation) and RhoA (activation). The latter are well established regulators of endothelial barrier properties that act via cytoskeletal elements. Indeed, liquid chromatography-mass spectrometry (LC-MS)-based proteomic profiling study revealed extensive changes in expression of cytoskeleton-related proteins. These results shed light on the nature of the interaction between the prion peptide PrP 106-126 and the BBB and emphasize the importance of the cytoskeleton in endothelium response to prion- induced stress.


Subject(s)
Monomeric GTP-Binding Proteins , Prions , Blood-Brain Barrier/metabolism , Prions/metabolism , Endothelial Cells/metabolism , Prion Proteins/metabolism , Nitric Oxide/metabolism , Proteomics , Endothelium/metabolism , Cytoskeleton/metabolism , Monomeric GTP-Binding Proteins/metabolism , Peptides/pharmacology , Peptides/metabolism
2.
Neurodegener Dis ; 15(4): 233-42, 2015.
Article in English | MEDLINE | ID: mdl-26113413

ABSTRACT

BACKGROUND/AIM: The sporadic form of the disease affects the majority of amyotrophic lateral sclerosis (ALS) patients. The role of glutamate (Glu) excitotoxicity in ALS has been extensively documented and remains one of the prominent hypotheses of ALS pathogenesis. In light of this evidence, the availability of a method to remove excess Glu from brain and spinal cord extracellular fluids without the need to deliver drugs across the blood-brain barrier and with minimal or no adverse effects may provide a major therapeutic asset, which is the primary aim of this study. METHODS: The therapeutic efficacy of the combined treatment with recombinant Glu-oxaloacetate-transaminase (rGOT) and its co-factor oxaloacetic acid (OxAc) has been tested in an animal model of sporadic ALS. RESULTS: We found that OxAc/rGOT treatment provides significant neuroprotection to spinal cord motor neurons. It also slows down the development of motor weakness and prolongs survival. CONCLUSION: In this study we bring evidence that the administration of Glu scavengers to rats with sporadic ALS inhibited the massive death of spinal cord motor neurons, slowed the onset of motor weakness and prolonged survival. This treatment may be of high clinical significance for the future treatment of chronic neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Aspartate Aminotransferases/administration & dosage , Neuroprotective Agents/administration & dosage , Oxaloacetic Acid/administration & dosage , Animals , Aspartate Aminotransferases/pharmacokinetics , Disease Models, Animal , Drug Therapy, Combination , Kaplan-Meier Estimate , Male , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Neuroprotective Agents/pharmacokinetics , Oxaloacetic Acid/pharmacokinetics , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Rotarod Performance Test , Spinal Cord/drug effects , Spinal Cord/pathology
3.
J Cereb Blood Flow Metab ; 34(2): 221-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24149933

ABSTRACT

Organophosphate-induced brain damage is an irreversible neuronal injury, likely because there is no pharmacological treatment to prevent or block secondary damage processes. The presence of free glutamate (Glu) in the brain has a substantial role in the propagation and maintenance of organophosphate-induced seizures, thus contributing to the secondary brain damage. This report describes for the first time the ability of blood glutamate scavengers (BGS) oxaloacetic acid in combination with glutamate oxaloacetate transaminase to reduce the neuronal damage in an animal model of paraoxon (PO) intoxication. Our method causes a rapid decrease of blood Glu levels and creates a gradient that leads to the efflux of the excess brain Glu into the blood, thus reducing neurotoxicity. We demonstrated that BGS treatment significantly prevented the peripheral benzodiazepine receptor (PBR) density elevation, after PO exposure. Furthermore, we showed that BGS was able to rescue neurons in the piriform cortex of the treated rats. In conclusion, these results suggest that treatment with BGS has a neuroprotective effect in the PO intoxication. This is the first time that this approach is used in PO intoxication and it may be of high clinical significance for the future treatment of the secondary neurologic damage post organophosphates exposure.


Subject(s)
Aspartate Aminotransferases/pharmacology , Brain Injuries , Cholinesterase Inhibitors/adverse effects , Oxaloacetic Acid/pharmacology , Paraoxon/adverse effects , Animals , Brain Injuries/blood , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/pathology , Carrier Proteins/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Hep G2 Cells , Humans , Male , Neurons/metabolism , Neurons/pathology , Paraoxon/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism
4.
J Biol Chem ; 287(53): 44676-83, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23150670

ABSTRACT

Most chemotherapeutic agents are blood-brain barrier (BBB) impermeants. HIV-1-derived TAT protein variants contain a transmembrane domain, which may enable them to cross the BBB and reach the brain. Here we synthesized CAYGRKKRRQRRR, a peptide containing a cysteine moiety attached to the N terminus of the transmembrane domain (C-TAT peptide), and studied its effects in an in vitro BBB model, which we found to reflect penetration by a receptor-independent pathway. Incubation of the brain capillary endothelial cell monolayer with 0.3-0.6 µmol/ml of this C-TAT peptide, for a period of 1-2 h, destabilizes brain capillary endothelial cell monolayer and introduces the ability of impermeant therapeutic agents including high molecular weight proteins to penetrate it substantially. The cysteinyl moiety at position 1 of the C-TAT peptide contributes largely to the destabilizing potency and the penetration efficacy of impermeant substances. The destabilizing effect was reversed using heparin. In summary, experimental conditions allowing a significant increase in entry of impermeant low and high molecular weight substances from the luminal (blood) to the abluminal side (brain) were found in an in vitro BBB model reflecting in vivo protein penetrability by a receptor-independent pathway.


Subject(s)
Endothelial Cells/metabolism , HIV Infections/metabolism , HIV-1/metabolism , Peptides/metabolism , Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Blood-Brain Barrier , Brain/metabolism , HIV Infections/virology , HIV-1/genetics , Humans , Models, Biological , Molecular Weight , Peptides/genetics , Permeability , Protein Transport , Proteins/chemistry , Swine , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics
5.
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
6.
Invest New Drugs ; 30(6): 2226-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22392507

ABSTRACT

L-Glutamate (Glu) plays a crucial role in the growth of malignant gliomas. We have established the feasibility of accelerating a naturally occurring brain to-blood Glu efflux by decreasing blood Glu levels with intravenous oxaloacetate, the respective Glu co-substrate of the blood resident enzyme humane glutamate­oxaloacetate transaminase(hGOT). We wished to demonstrate that blood Glu scavenging provides neuroprotection in the case of glioma.We now describe the neuroprotective effects of blood Glu scavenging in a fatal condition such as brain-implanted C6 glioma in rats and brain-implanted human U87 MG glioma in nude mice. Rat (C-6) or human (U87) glioma cells were grafted stereotactically in the brain of rats or mice. After development of tumors, the animals were drinking oxaloacetate with or without injections of hGOT. In addition, mice were treated with combination treatment, which included drinking oxaloacetate with intracutaneous injections of hGOT and intraperitoneal injection of Temozolomide. Animals drinking oxaloacetate with or without injections of hGOT displayed a smaller tumor volume, reduced invasiveness and prolonged survival than control animals drinking saline. These effects were significantly enhanced by Temozolomide in mice, which increased survival by 237%. This is the first demonstration of blood Glu scavenging in brain cancer, and because of its safety, is likely to be of clinical significance for the future treatment of human gliomas. As we demonstrated, the blood glutamate scavenging treatment in combination with TMZ could be a good candidate or as an alternative treatment to the patients that do not respond to TMZ.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Aspartate Aminotransferases/administration & dosage , Dacarbazine/analogs & derivatives , Glutamic Acid/blood , Oxaloacetic Acid/administration & dosage , Animals , Brain , Brain Neoplasms/blood , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Dacarbazine/administration & dosage , Glioma/blood , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Temozolomide , Tumor Burden/drug effects
7.
Invest New Drugs ; 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22297683

ABSTRACT

L-Glutamate (Glu) plays a crucial role in the growth of malignant gliomas. We have established the feasibility of accelerating a naturally occurring brain to-blood Glu efflux by decreasing blood Glu levels with intravenous oxaloacetate, the respective Glu co-substrate of the blood resident enzyme humane glutamate-oxaloacetate transaminase (hGOT). We wished to demonstrate that blood Glu scavenging provides neuroprotection in the case of glioma. We now describe the neuroprotective effects of blood Glu scavenging in a fatal condition such as brain-implanted C6 glioma in rats and brain-implanted human U87 MG glioma in nude mice. Rat (C-6) or human (U87) glioma cells were grafted stereotactically in the brain of rats or mice. After development of tumors, the animals were drinking oxaloacetate with or without injections of hGOT. In addition, mice were treated with combination treatment, which included drinking oxaloacetate with intracutaneous injections of hGOT and intraperitoneal injection of Temozolomide. Animals drinking oxaloacetate with or without injections of hGOT displayed a smaller tumor volume, reduced invasiveness and prolonged survival than control animals drinking saline. These effects were significantly enhanced by Temozolomide in mice, which increased survival by 237%. This is the first demonstration of blood Glu scavenging in brain cancer, and because of its safety, is likely to be of clinical significance for the future treatment of human gliomas. As we demonstrated, the blood glutamate scavenging treatment in combination with TMZ could be a good candidate or as an alternative treatment to the patients that do not respond to TMZ.

8.
J Cereb Blood Flow Metab ; 32(1): 177-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21915136

ABSTRACT

At high concentrations, glutamate (Glu) exerts potent neurotoxic properties, leading to irreversible brain damages found in numerous neurological disorders. The accepted notion that Glu homeostasis in brain interstitial fluid is maintained primarily through the activity of Glu transporters present on glial cells does not take into account the possible contribution of endothelial cells constituting the blood-brain barrier (BBB) to this process. Here, we present evidence for the presence of the Glu transporters, excitatory amino-acid transporters (EAATs) 1 to 3, in porcine brain endothelial cells (PBECs) and show their participation in Glu uptake into PBECs. Moreover, transport of Glu across three in vitro models of the BBB is investigated for the first time, and evidence for Glu transport across the BBB in both directions is presented. Our results provide evidence that the BBB can function in the efflux mode to selectively remove Glu, via specific transporters, from the abluminal side (brain) into the luminal compartment (blood). Furthermore, we found that glial cells lining the BBB have an active role in the efflux process by taking up Glu and releasing it, through hemichannels, anion channels, and possibly the reversal of its EAATs, in close proximity to ECs, which in turn take up Glu and release it to the blood.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Glutamic Acid/metabolism , Neuroglia/metabolism , Animals , Animals, Newborn , Biological Transport , Blood-Brain Barrier/cytology , Blotting, Western , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Flow Cytometry , Glutamate Plasma Membrane Transport Proteins/metabolism , Homeostasis , Immunohistochemistry , Models, Biological , Neuroglia/cytology , Rats , Rats, Wistar , Swine
9.
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
10.
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
11.
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
12.
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
13.
Eur J Neurosci ; 34(9): 1432-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21936878

ABSTRACT

In previous studies, we have shown that by increasing the brain-to-blood glutamate efflux upon scavenging blood glutamate with either oxaloacetate or pyruvate, one achieves highly significant neuroprotection particularly in the context of traumatic brain injury. The current study examines, for the first time, how the blood glutamate scavenging properties of glutamate-pyruvate transaminase (GPT), alone or in combination with pyruvate, may contribute to the spectrum of its neuroprotective mechanisms and improve the outcome of rats exposed to brain ischemia, as they do after head trauma. Rats that were exposed to permanent middle cerebral artery occlusion (MCAO) and treated with intravenous 250 mg/kg pyruvate had a smaller volume of infarction and reduced brain edema, resulting in an improved neurological outcome and reduced mortality compared to control rats treated with saline. Intravenous pyruvate at the low dose of 31.3 mg/kg did not demonstrate any neuroprotection. However, when combined with 0.6 mg/kg of GPT there was a similar neuroprotection observed as seen with pyruvate at 250 mg/kg. Animals treated with 1.69 g/kg glutamate had a worse neurological outcome and a larger extent of brain edema. The decrease in mortality, infarcted brain volume and edema, as well as the improved neurological outcome following MCAO, was correlated with a decrease in blood glutamate levels. We therefore suggest that the blood glutamate scavenging activity of GPT and pyruvate contributes to the spectrum of their neuroprotective mechanisms and may serve as a new neuroprotective strategy for the treatment of ischemic stroke.


Subject(s)
Glutamic Acid/blood , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Pyruvic Acid/administration & dosage , Animals , Aspartate Aminotransferases/therapeutic use , Brain Edema/etiology , Brain Edema/prevention & control , Brain Infarction/etiology , Brain Infarction/pathology , Brain Infarction/prevention & control , Disease Models, Animal , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/complications , Male , Motor Activity/drug effects , Neurologic Examination , Oxaloacetic Acid/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors
14.
J Neurosurg Anesthesiol ; 23(4): 323-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21836527

ABSTRACT

BACKGROUND: Elevated levels of glutamate in brain fluids, in the context of several neurodegenerative conditions, are associated with a worsened neurological outcome. Because there is a clear relationship between brain glutamate levels and glutamate levels in the blood, and an association of the latter with stress, the purpose of this study was to investigate the effects of glucose, insulin, and glucagon on rat blood glutamate levels. METHODS: Rats received either 1 mL/100 g of rat body weight (BW) intravenous isotonic saline (control), 150 mg/1 mL/100 g BW intravenous glucose, 75 mg/1 mL/100 g BW intravenous glutamate, 50 g/100 g BW intraparitoneal glucagon, or 0.2 UI/100 g BW intraparitoneal insulin. Blood samples were subsequently drawn at 0, 30, 60, 90, and 120 minutes for determination of blood glutamate and glucose levels. RESULTS: We observed a significant decrease in blood glutamate levels at 30 minutes after injection of glucose (P<0.05), at 30 and 60 minutes after injection of insulin (P<0.05), and at 90 and 120 minutes after injection of glucagon. Plasma glucose levels were elevated after infusion of glutamate and glucose but were decreased after injection of insulin. CONCLUSIONS: The results of this study demonstrate that glucose, insulin, and glucagon significantly reduce blood glutamate levels. The effect of insulin is immediate and transient, whereas the effect of glucagon is delayed but longer lasting, suggesting that the sensitivity of pancreatic glucagon and insulin-secreting cells to glutamate is dependent on glucose concentration. The results of this study provide insight into blood glutamate homeostasis and may assist in the implementation of new therapies for brain neuroprotection from excess glutamate.


Subject(s)
Blood Glucose/metabolism , Glucagon/pharmacology , Glucose/pharmacology , Glutamic Acid/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Animals , Injections, Intraperitoneal , Injections, Intravenous , Insulin/metabolism , Male , Pancreas/drug effects , Rats , Rats, Sprague-Dawley
15.
Methods Mol Biol ; 762: 347-54, 2011.
Article in English | MEDLINE | ID: mdl-21717369

ABSTRACT

Claudins are transmembrane proteins that form the backbone of the tight junctions (TJs) at the blood-brain barrier (BBB). TJs are cellular structures that physically obstruct the inter-endothelial space and restrict the paracellular diffusion of blood-borne substances from the peripheral circulation into the CNS. TJs are also dynamic structures that rapidly respond to external signals that produce changes in BBB permeability. We focus here on the biochemical and immunohistochemical properties of claudin-5 as expressed in three in vitro models of the BBB, and show that the contact co-culture of endothelial cells with glial cells significantly increases claudin-5 expression.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Culture Techniques/methods , Claudins/metabolism , Endothelial Cells/metabolism , Neuroglia/metabolism , Tight Junctions/metabolism , Animals , Blotting, Western/methods , Electrophoresis, Polyacrylamide Gel/methods , Endothelial Cells/physiology , Immunohistochemistry/methods , Neuroglia/physiology , Swine
16.
Neuropharmacology ; 61(5-6): 1026-32, 2011.
Article in English | MEDLINE | ID: mdl-21781978

ABSTRACT

Postconditioning can be induced by a broad range of stimuli within minutes to days after an ischemic cerebral insult. A special form is elicited by pharmacological intervention called second pathophysiological stress. The present study aimed to evaluate the effects of low-dose (5 mg/kg) kainate postconditioning with onsets 0, 24 and 48 h after the ischemic insult on the hippocampal synaptic plasticity in a 2-vessel occlusion model in rat. The hippocampal function was tested by LTP measurements of Schaffer collateral-CA1 pyramidal cell synapses in acute slices and the changes in density of Golgi-Cox-stained apical dendritic spines. Postconditioning 0 and 24 h after ischemia was not protective, whereas 48-h-onset postconditioning resulted in the reappearance of a normal spine density (>100,000 spines) 3 days after ischemia, in parallel with the long-term restoration of the damaged LTP function. Similar, but somewhat less effects were observed after 10 days. Our data clearly demonstrate the onset dependence of postconditioning elicited by a subconvulsant dose of kainate treatment in global ischemia, with restoration of the structural plasticity and hippocampal function.


Subject(s)
CA1 Region, Hippocampal/physiology , Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Long-Term Potentiation/drug effects , Animals , CA1 Region, Hippocampal/drug effects , Drug Evaluation, Preclinical , Electroencephalography , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Ischemic Attack, Transient/mortality , Ischemic Attack, Transient/physiopathology , Long-Term Potentiation/physiology , Male , Molecular Targeted Therapy , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Random Allocation , Rats , Rats, Wistar , Stress, Physiological/drug effects , Stress, Physiological/physiology
17.
J Neurosurg Anesthesiol ; 23(3): 222-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593692

ABSTRACT

BACKGROUND: The animal model of stroke that is most frequently used is a rat model of focal brain ischemia caused by middle cerebral artery occlusion (MCAO). Several studies have reported a link between levels of cell-free DNA (CFD) and neurologic outcome in human stroke. The purpose of this study was to assess brain injury and measure CFD levels in 2 models of MCAO in rats, and to determine whether brain injury correlates with CFD. METHODS: A total of 60 rats were used for this study. Twenty rats underwent a sham procedure, 20 rats had MCAO using a monofilament, and 20 rats had MCAO with a silicon-coated filament. Groups were further divided into 2 subgroups. In 1 subgroup of 10 rats, neurologic performance [measured as a neurologic severity score, (NSS)] was measured at 1 and 24 hours after the procedure, and brain edema and infarct volume were determined at 24 hours. In the second subgroup of 10 rats, CFD was measured at 0, 1, 2, 4, 8, 12, and 24 hours and at 2, 3, 4, and 5 days. Neurologic performance (measured as a NSS) was measured at 1 and 24 hours after the procedure. RESULTS: The main finding was a significant increase in CFD levels observed 24 hours after the onset of MCAO. The correlation between the total infarct volume and CFD levels of the 3 groups was R=0.78, P<0.0001. Brain edema and NSS also were strongly correlated with CFD levels at 24 hours after MCAO (R=0.91, P<0.0001 and R=0.73, P<0.0001, respectively). CONCLUSIONS: We found that CFD levels correlate well with the extent of ischemic injury, brain edema, and neurologic outcome in rats 24 hours post-MCAO. We have also shown that CFD correlates well with the expected temporal progression of ischemic injury. These findings place CFD in a unique place as a biomarker for stroke, both experimentally and possibly clinically.


Subject(s)
Brain Ischemia/blood , Cell-Free System/metabolism , DNA/blood , Stroke/blood , Animals , Biomarkers/blood , Disease Models, Animal , Male , Predictive Value of Tests , Rats , Rats, Sprague-Dawley
18.
Behav Brain Res ; 223(1): 17-23, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21419805

ABSTRACT

Middle cerebral artery occlusion (MCAO) is widely used as a rat model of focal brain ischemia. Evaluation of brain damage often includes the morphological analysis of the injury area, MRI, and various scales which depend on functional tests, commonly known as neurological severity score (NSS). We determined the optimal number of NSS tests and assessed their capacity for non-invasive evaluation of brain ischemic injury in the rat MCAO model. 275 male Sprague-Dawley rats were randomly divided into five groups, given either permanent (p) MCAO or transient (t) MCAO using an uncoated 4-0 monofilament catheter or a silicone-coated monofilament. The rats' neurological status was examined before and at 1 and 24h following MCAO. The size of brain injury was then measured histologically and the extent of right cerebral hemisphere edema was calculated. We established a correlation between these tests and morphological data for brain injury. Adjusted R(2) of the prediction of total histology score was 0.7. The Hosmer-Lemeshow p-value of this model was 0.812 for total brain histology. For the brain edema the adjusted R(2) of the prediction model was 0.48. The Hosmer-Lemeshow p-value of this model was 0.558 for brain edema. Our methods of estimating infarct size produces reliable and well correlated results at 24h and demonstrates to be an easy and quick way to assess infarct size soon after ischemic injury has occurred. The described method for neurological assessment could ultimately aid in assessing various treatment modalities in the early hours following stroke.


Subject(s)
Behavior, Animal/physiology , Brain Edema/pathology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/psychology , Neurologic Examination/methods , Animals , Brain/blood supply , Brain/pathology , Brain Edema/complications , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/physiopathology , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Time Factors
20.
J Neurosurg Anesthesiol ; 23(1): 45-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21248494

ABSTRACT

BACKGROUND: Abnormally high concentrations of glutamate in brain fluids have been shown to be neurotoxic and correlate with a poor neurological outcome following traumatic brain injury (TBI). Since brain fluid glutamate can be reduced by scavenging blood glutamate, the purpose of this study was to investigate factors that may potentially influence levels of blood glutamate, glucose, and the enzymes glutamate-pyruvate transaminase (GPT) and glutamate-oxaloacetate transaminase (GOT) in healthy individuals. METHODS: Factors that were examined included age, gender, time of last meal or drink, and recent consumption of coffee. A total of 112 healthy volunteers between 18 and 70 years of age participated in the study. The average participant was 38 years old, and the sample consisted of 48 males and 64 females. Five milliliters of venous blood was collected from participants' cubital vein and blood glutamate, glucose, GOT and GPT levels were determined. Participants were then asked to complete a questionnaire addressing their gender, age, time of last meal, time of last drink, and whether coffee was consumed within the last 6 hours. RESULTS: Blood glutamate concentrations were significantly higher in males than in females (P < 0.001) and may be due to effects of estrogen and progesterone. Concentrations of GOT were significantly higher in males than in females (P < 0.01). Concentrations of GPT were significantly higher in males than in females (P < 0.01). There were no other significant differences demonstrated. CONCLUSIONS: Understanding the factors that affect blood glutamate levels may give new insight into mechanisms that protect the brain from excess glutamate and result in a better neurological outcome following TBI.


Subject(s)
Glutamic Acid/blood , Adolescent , Adult , Aged , Aging , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Blood Glucose/metabolism , Brain Chemistry , Coffee , Eating/physiology , Female , Glutamic Acid/metabolism , Humans , Male , Middle Aged , Reference Values , Sex Characteristics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...