Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Drug Chem Toxicol ; : 1-8, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38726980

ABSTRACT

The neurodegenerative mechanisms of Alzheimer's disease (AD) are not fully understood, but it is believed that amyloid beta (Aß) peptide causes oxidative stress, neuroinflammation, and disrupts metabotropic glutamate receptor 5 (mGluR5) signaling by interacting with cholesterol and caveolin-1 (Cav-1) in pathogenic lipid rafts. This study examined the effect of 2-hydroxypropyl-ß-cyclodextrin (HP-CD) on cholesterol, oxidative stress (total oxidant status), neuroinflammation (TNF-α), and mGluR5 signaling molecules such as PKCß1, PKCß2, ERK1/2, CREB, BDNF, and NGF in Aß (1-42)-induced neurotoxicity. The Sprague-Dawley rats were divided into four groups: control (saline), Aß (1-42), HP-CD (100 mg/kg), and Aß (1-42) + HP-CD (100 mg/kg). All groups received bilateral stereotaxic injections of Aß (1-42) or saline into the hippocampus. After surgery, HP-CD was administered intraperitoneally (ip) for 7 days. Cholesterol, TNF-α, and TOS levels were measured in synaptosomes isolated from hippocampus tissue using spectrophotometry, fluorometry, and enzyme immunoassay, respectively. The gene expressions of Cav-1, mGluR5, PKCß1, PKCß2, ERK1/2, CREB, BDNF, and NGF in hippocampus tissue were evaluated using reverse transcription PCR after real-time PCR analysis. Treatment with Aß (1-42) significantly elevated cholesterol, TOS, TNF-α, Cav-1, PKCß2, and ERK1/2 levels. Additionally, mGluR5, CREB, and BDNF levels were shown to be lowered. HP-CD reduced cholesterol, TOS, and TNF-α levels while increasing mGluR5, CREB, and BDNF in response to Aß (1-42) treatment. These findings indicate that HP-CD may have neuroprotective activity due to the decreased levels of cholesterol, oxidative stress, and neuroinflammation, as well as upregulated levels of mGluR5, CREB, and BDNF.

2.
ACS Omega ; 9(16): 18366-18374, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38680345

ABSTRACT

Antithrombotic agents and anticoagulant drugs, such as those from the heparin family, are employed in clinical settings for the prevention and treatment of clotting, thromboembolism, and wound healing. The potency assessment of antithrombotic agents is typically conducted using antifactor IIa assay with manual systems which are time-consuming and often lack repeatability. Here, we present a novel automated system that significantly enhances assay repeatability, attaining an outstandingly low relative standard deviation (RSD) % of only 0.6% for repeatability. This system has been applied to a pharmaceutical gel formulation for wound healing developed by Abdi Ibrahim Pharmaceuticals R&D Center as a case study for validation. The automated system demonstrated substantial improvements over manual systems in linearity (R2 = 0.9927), precision, accuracy, specificity, and robustness. The system aligns with the European Pharmacopoeia specifications, promising to enhance quality control across pharmaceutical formulations and conduct absorbance-based end-point assays within the pharmaceutical industry while offering increased throughput and cost-effectiveness.

3.
J Pharm Sci ; 105(8): 2372-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27353207

ABSTRACT

ß-Amyloid (Aß) plaques are the key neurotoxic assemblies in Alzheimer disease. It has been suggested that an interaction occurs between membrane cholesterol and Aß aggregation in the brain. Cyclodextrins can remove cholesterol from cell membranes and change receptor function. This study aimed to investigate the effect of hydroxypropyl-ß-cyclodextrin (HP-CD) polymeric microspheres, based on chitosan or sodium alginate, on the levels of lipid peroxidation, reactive oxygen species production, and mitochondrial function in brain synaptosomes. The effect of microspheres on DNA fragmentation, the expression of Bcl-2, Bax, and Apex1 mRNAs in rat hippocampus after Aß(1-42) peptide-induced neurotoxicity was also evaluated. Comparison with HP-CD raw material was performed. Aß(1-42) treatment significantly decreased the mitochondrial activity of Apex1 and Bcl-2 mRNAs, induced DNA fragmentation, and increased mRNA levels of Bax. Treatment with HP-CD microspheres against Aß(1-42) significantly reduced DNA fragmentation and increased the Bcl-2/Bax mRNA ratio and mitochondrial function. In addition, HP-CD microspheres used against Aß(1-42) decreased the levels of lipid peroxidation and reactive oxygen species production. These results indicate that nasally administered spray-dried HP-CD microspheres are able to provide protection against Aß(1-42)-induced neurotoxicity, due to the suppressed levels of oxidative stress and apoptotic signals in the rat hippocampus.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/therapeutic use , Amyloid beta-Peptides/toxicity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/prevention & control , Peptide Fragments/toxicity , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Administration, Intranasal , Animals , DNA Fragmentation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Lipid Peroxidation/drug effects , Microspheres , Neuroprotective Agents/administration & dosage , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology
4.
J Cereb Blood Flow Metab ; 35(12): 2043-51, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26174325

ABSTRACT

Oxidative stress is a major brain injury mechanism after ischemic stroke. 12/15-lipoxygenase (12/15-LOX) is a key mediator of oxidative stress, contributing to neuronal cell death and vascular leakage. Nonetheless, the mechanism leading to its upregulation is currently unknown. We show here that Signal Transducers and Activators of Transcription (STATs), specifically STAT6 and possibly STAT1, increase transcription of 12/15-LOX in neuronal cells. Both p-STAT6 and -1 bound to specific STAT binding sites in the mouse 12/15-LOX promoter. Small interfering RNA (siRNA) knockdown showed STAT6 to be the dominant regulator, reducing 12/15-LOX promoter activation and cell death in oxidatively stressed HT22 cells. STAT6 siRNA efficiently prevented the increase of 12/15-LOX in murine primary neurons, both after induction of oxidative stress and after oxygen-glucose deprivation. Early activation of STAT6 and STAT1 in mice was consistent with a role in regulating 12/15-LOX in focal ischemia. Brains of human stroke patients showed increased p-STAT6 and p-STAT1 in the peri-infarct region, along with 12/15-LOX and markers of apoptosis. These results link STAT6 and STAT1 to the 12/15-LOX damage pathway and suggest disregulation of STAT-dependent transcription as injury mechanism in stroke. Selectively targeting STATs may thus be a novel therapeutic approach to reducing brain injury after a stroke.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/biosynthesis , Neurons/pathology , STAT Transcription Factors/metabolism , Stroke/enzymology , Aged , Animals , Apoptosis , Female , Gene Knockdown Techniques , Glucose/deficiency , Humans , Hypoxia, Brain/enzymology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oxidative Stress , RNA, Small Interfering , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Stroke/pathology , Up-Regulation
5.
Cell Biochem Funct ; 32(7): 557-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25111857

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and is characterized by the presence of senile plaques and neurofibrillary tangles, along with synaptic loss. The underlying mechanisms of AD are not clarified yet, but oxidative stress and mitochondrial dysfunction are important factors. Overactivation of poly(adenosine diphosphate ribose) polymerase-1 (PARP-1) enzyme has been known to cause neuroinflammation and cell death in neurodegenerative processes. The aim of the present study was to investigate the protective effects of the PARP-1 inhibitors, 3-aminobenzamide (3-AB) and nicotinamide (NA), against amyloid ß peptide (1-42) (Aß(1-42))-induced oxidative damage and mitochondrial reduction capacity on isolated synaptosomes. Rats were injected intraperitoneally with 3-AB (30-100 mg kg(-1)), NA (100-500 mg kg(-1)) or with saline for 7 days. Synaptosomes were incubated with 10-30 µM Aß(1-42) or saline for 6 h at 37 °C. Ex vivo Aß(1-42) treatment significantly induced oxidative stress and mitochondrial dysfunction in synaptosomes of the saline group, while synaptosomes of 3-AB and NA groups showed significant decreases in lipid peroxidation, reactive oxygen species production and protein oxidation. Moreover, both NA and 3-AB were able to improve the mitochondrial reduction capacity against Aß(1-42). These data suggest that NA and 3-AB may have protective effects in neurodegenerative processes because of the reduced levels of oxidative stress and the improvement of mitochondrial function.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzamides/pharmacology , Brain/drug effects , Neuroprotective Agents/pharmacology , Niacinamide/pharmacology , Peptide Fragments/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Synaptosomes/drug effects , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Animals , Brain/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Rats, Sprague-Dawley , Synaptosomes/pathology
6.
Comb Chem High Throughput Screen ; 16(9): 695-701, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23782036

ABSTRACT

Glutathione (GSH) is a major endogenous antioxidant highly active in human tissues and plays a key role in controlling cellular thiol redox system, maintaining the immune and detoxification system. The determination of GSH levels in tissue is important to estimate endogenous defenses against oxidative stress. In our study, the multi-walled carbon nanotube modified screen-printed electrodes (MWCNT-SPEs) were used to determine the levels of GSH in trichloroacetic acid (TCA)-treated or untreated samples of rat plasma. It was found that the deproteinization of samples with TCA improved the electrochemical detection of GSH particularly in plasma. The oxidation of GSH was measured by using differential pulse voltammetry (DPV) method in combination with MWCNT-SPE (n=3), and the detection limit of GSH was found to be 0.47 µM (S/N=3). The GSH levels in plasma samples were also measured spectrophotometrically in order to compare the effectiveness of electrochemical method and we obtained a high correlation between the two methods (R(2)=0.976).


Subject(s)
Electrochemical Techniques/instrumentation , Glutathione/blood , Nanotubes, Carbon/chemistry , Animals , Electrodes , Equipment Design , Limit of Detection , Rats
7.
Eur J Pharm Sci ; 49(4): 603-13, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23643737

ABSTRACT

Given the fundamentally multifactorial character of Alzheimer's disease (AD), addressing more than one target for disease modification or therapy is expected to be highly advantageous. Here, following the cholinergic hypothesis, we aimed to inhibit both acetyl- and butyrylcholinesterase (AChE and BuChE) in order to increase the concentration of acetylcholine in the synaptic cleft. In addition, the formation of the amyloid ß fibrils should be inhibited and already preformed fibrils should be destroyed. Based on a recently identified AChE inhibitor with a 1,4-substituted 4-(1H)-pyridylene-hydrazone skeleton, a substance library has been generated and tested for inhibition of AChE, BuChE, and fibril formation. Blood-brain barrier mobility was ensured by a transwell assay. Whereas the p-nitrosubstituted compound 18C shows an anti-AChE activity in the nanomolar range of concentration (IC50=90 nM), the bisnaphthyl substituted compound 20L was found to be the best overall inhibitor of AChE/BuChE and enhances the fibril destruction.


Subject(s)
Acetylcholinesterase/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Hydrazones/pharmacology , Animals , Blood-Brain Barrier/metabolism , Endothelial Cells , HEK293 Cells , Humans , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
8.
Epilepsy Res ; 105(1-2): 13-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23337899

ABSTRACT

The opioid/nociceptin receptors are involved in many neurological disorders such as Alzheimer's disease, Parkinson's disease and epilepsy. Kainic acid (KA) is an analog of the excitatory amino acid transmitter glutamate and the systemic administration of KA induces status epilepticus (SE) in rodents. In this study, we examined the alterations in the G-protein activity and the gene expression levels of mu, kappa, delta opioid and nociceptin receptors (MOPr, KOPr, DOPr and NOPr) as well as PNOC, the precursor polypeptide of nociceptin-OFQ (N/OFQ) in KA-induced seizures in the rat brain cortex. KA was used to create seizures with the dose of 10 mg/kg body weight i.p. Following the KA administration, the rats were observed for 3 h to assess seizure activity. Seizures occurred approximately 45 min after the KA injection. Only rats exhibiting full limbic seizures, forelimb clonus with rearing, were used in this study. All animals were decapitated 4 h after the administration of KA. Our [(35)S]GTPγS binding results showed that there was a significant difference in both the affinity and efficacy particularly one of NOPr stimulation following KA treatment. Slight, but significant increase was observed for MOPr. Moreover PNOC, NOPr and MOPr mRNA levels were increased by KA treatment but there were no significant changes in the levels of DOPr and KOPr mRNAs. These results show that the activities of opioid/nociceptin receptors can be modified by KA-treatment, and MOPr, PNOC and NOPr are the most responsive to KA-induced seizures in the rat brain cortex.


Subject(s)
Cerebral Cortex/metabolism , GTP-Binding Proteins/biosynthesis , Kainic Acid/toxicity , RNA, Messenger/biosynthesis , Receptors, Opioid/biosynthesis , Seizures/metabolism , Animals , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Gene Expression Regulation , Male , Opioid Peptides/metabolism , Opioid Peptides/pharmacology , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Nociceptin Receptor
9.
Neural Regen Res ; 8(6): 485-95, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-25206691

ABSTRACT

Resveratrol, a natural phenolic compound, has been shown to prevent cardiovascular diseases and cancer and exhibit neuroprotective effects. In this study, we examined the neuroprotective and antioxidant effects of resveratrol against hydrogen peroxide in embryonic neural stem cells. Hydrogen peroxide treatment alone increased catalase and glutathione peroxidase activities but did not change superoxide dismutase levels compared with hydrogen peroxide + resveratrol treatment. Nitric oxide synthase activity and concomitant nitric oxide levels increased in response to hydrogen peroxide treatment. Conversely, resveratrol treatment decreased nitric oxide synthase activity and nitric oxide levels. Resveratrol also attenuated hydrogen peroxide-induced nuclear or mitochondrial DNA damage. We propose that resveratrol may be a promising agent for protecting embryonic neural stem cells because of its potential to decrease oxidative stress by inducing higher activity of antioxidant enzymes, decreasing nitric oxide production and nitric oxide synthase activity, and alleviating both nuclear and mitochondrial DNA damage.

10.
Drug Chem Toxicol ; 35(4): 393-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22486999

ABSTRACT

Inflammation is deleterious for organs with reduced capacity of regeneration, such as the brain. Recently, studies have focused on investigating the therapeutic effects of nonsteroidal anti-inflammatory drugs (NSAIDs) in Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. Excitotoxicity is the pathological process when receptors for the excitatory neurotransmitter glutamate, such as the N-methyl-D-aspartate (NMDA), receptors are overactivated. This process may be involved in neurodegenerative diseases. D-serine is one of the coagonist of NMDA receptors, and increased levels of D-serine are associated with excitotoxicity. In our study, the potential neuroprotective effects of mefenamic acid, acetaminophen, and naproxen sodium were investigated against D-serine-induced oxidative stress in the rat brain in vitro. To show their potential neuroprotective properties, NSAIDs were incubated with D-serine and reactive oxygen species (ROS), malondialdehyde, and protein carbonyl content of the brain after different treatments were measured. Our results demostrate that NSAIDs used in the present study significantly reduced ROS production, lipid peroxidation, and protein oxidation against D-serine treatment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Acetaminophen/pharmacology , Animals , Brain/drug effects , Brain/pathology , Inflammation/pathology , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Mefenamic Acid/pharmacology , Naproxen/pharmacology , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/toxicity
11.
Neurochem Int ; 60(6): 555-64, 2012 May.
Article in English | MEDLINE | ID: mdl-22382076

ABSTRACT

Excitotoxicity is a contributing factor to the pathogenesis of acute or chronic neurodegenerative disease states. Kainic acid (KA) is an excitotoxic substance and the administration of it to rodents induces seizure activity (status epilepticus, SE) and leads to neurodegeneration. In this study the effect of KA-induced excitotoxicity on the G-protein activations and the gene expression levels of the opioid/nociceptin system receptors as MOPr, KOPr, DOPr, ORL-1, and PNOC (N/OFQ) were investigated, and the regulator effect of naloxone (Nal) on the gene expressions of the opioid system receptors against KA-induced seizures in the rat hippocampus was tested. In addition, the expression levels of stress-toxicity genes were assessed in the hippocampus following KA-induced excitotoxicity in order to determine the potential genetic targets which can be helpful for neuroprotective interventions. Our results indicate that the KA-induced excitotoxicity increased the mRNA levels of MOPr, DOPr, KOPr, PNOC, and ORL-1. However, G-protein activations of MOPr, DOPr, and KOPr remained relatively unchanged while both the potency and efficacy of N/OFQ were significantly increased. The PCR array data showed that KA-induced excitotoxicity altered the expression levels of genes in the cellular stress or toxicity pathways. Our data suggests that the induction of the opioid/nociceptin system may be involved in the cellular stress response following a neurodegenerative insult and that the genes modulated by the KA-treatment in the stress-toxicity pathways may be evaluated as targets of potential neuroprotective interventions.


Subject(s)
Hippocampus/drug effects , Kainic Acid/toxicity , Opioid Peptides/physiology , Receptors, Opioid/physiology , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Animals , Excitatory Amino Acid Agonists/toxicity , Hippocampus/physiology , Male , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid/genetics , Status Epilepticus/drug therapy , Nociceptin
12.
Free Radic Res ; 46(6): 726-39, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22369458

ABSTRACT

Mefenamic acid, a non-steroidal antiinflammatory drug (NSAID), directly and dose-dependently exhibits neuroprotective activity. In our study, we investigated the effects of mefenamic acid against d-serine on oxidative stress in the hippocampus, cortex and cerebellum of rats. Furthermore, the potential inflammatory and apoptotic effects of d-serine and potential protective effect of mefenamic acid were determined at mRNA and protein levels of TNF-α, IL-1ß, Bcl-2 and Bax. We found that d-serine significantly increased oxidative stress, levels of inflammation- and apoptosis-related molecules in a region specific manner. Mefenamic acid treatment provided significant protection against the elevation of lipid peroxidation, protein oxidation, levels of TNF-α, IL-1ß and Bax. As a conclusion, we suggest that d-serine, as a potential neurodegenerative agent, may have a pivotal role in the regulation of oxidative stress, inflammation and apoptosis; and NSAIDs, such as mefenamic acid, may assist other therapeutics in treating disorders where d-serine-induced neurotoxic mechanisms are involved in.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/drug effects , Mefenamic Acid/pharmacology , Serine/pharmacology , Animals , Apoptosis/drug effects , Brain/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Serine/metabolism
13.
Hippocampus ; 22(2): 122-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21049484

ABSTRACT

Mu opioid receptors (MOR) are known to be involved in seizure activity. The main goal of the present study was to characterize the MOR mRNA expression, binding, as well as G protein activation mediated by these receptors in epileptic hippocampus of patients with pharmacoresistant mesial temporal lobe epilepsy (TLE). In contrast with autopsy samples, hippocampus obtained from patients with mesial TLE demonstrated enhanced MOR mRNA expression (116%). Saturation binding experiments revealed significantly higher (60%) B(max) values for the mesial TLE group, whereas the K(d) values were not statistically different. Although mesial TLE group demonstrated high levels of basal binding for the G proteins (136%), DAMGO-stimulated [(35)S]GTPγS binding did not demonstrate significant alterations. In conclusion, our present data provide strong evidence that the epileptic hippocampus of patients with pharmacoresistant mesial TLE presents significant alterations in MOR. Such changes may represent adaptive mechanisms to compensate for other as yet unknown alterations.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , GTP-Binding Proteins/metabolism , Hippocampus/metabolism , RNA, Messenger/analysis , Receptors, Opioid, mu/metabolism , Adolescent , Adult , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
14.
Epilepsy Res ; 99(1-2): 64-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22079489

ABSTRACT

It has been known for centuries that exogenous cannabinoids, such as tetrahydrocannabinol have anticonvulsant activity. Recent studies have advanced our understanding of the endogenous cannabinoid system and renewed the interest in cannabinoids as a potential treatment for epilepsy. The endogenous cannabinoid system is rapidly activated after seizure activity but still little is known about the molecular mechanisms underlying the role of the cannabinoid system in epilepsy. In this study epileptiform activity was induced by kainic acid (KA) and effects of the CB1 receptor agonists N-(2-Chloroethyl)-5Z,8Z,11Z,14Z-eicosatetraenamide (ACEA) on G-protein signaling using the agonist-stimulated [(35)S]GTPγS binding assay were evaluated. Control and KA treated rat hippocampus and cortex membranes were used. Our results showed that the ACEA displayed a high potency and efficacy in stimulating the G-proteins and when compared to the control animals, significant enhancements were observed in tissues from the KA treated animals. Potency and efficacy values were in particular increased in the hippocampus tissues. Furthermore, gene expression levels of the cannabinoid receptor 1 (CB1) receptor and cannabinoid receptor interacting protein 1 (CRIP1) were measured by RT-PCR, where both CB1 and CRIP1 expressions were found to be elevated in the KA treated animals.


Subject(s)
Carrier Proteins/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Kainic Acid/toxicity , Receptor, Cannabinoid, CB1/metabolism , Seizures/metabolism , Animals , Arachidonic Acids/pharmacology , Cannabinoids/biosynthesis , Carrier Proteins/agonists , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Seizures/chemically induced
15.
J Neurochem ; 118(2): 248-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21534958

ABSTRACT

Oxidative stress is a pathway of injury that is common to almost all neurological conditions. Hence, methods to scavenge radicals have been extensively tested for neuroprotection. However, saving neurons alone may not be sufficient in treating CNS disease. In this study, we tested the cytoprotective actions of the glutathione precursor gamma-glutamylcysteine ethyl ester (GCEE) in brain endothelium. First, oxidative stress was induced in a human brain microvascular endothelial cell line by exposure to H(2)O(2). Addition of GCEE significantly reduced formation of reactive oxygen species, restored glutathione levels which were reduced in the presence of H(2)O(2), and decreased cell death during H(2)O(2)-mediated injury. Next, we asked whether GCEE can also protect brain endothelial cells against oxygen-glucose deprivation (OGD). As expected, OGD disrupted mitochondrial membrane potentials. GCEE was able to ameliorate these mitochondrial effects. Concomitantly, GCEE significantly decreased endothelial cell death after OGD. Lastly, our in vivo experiments using a mouse model of brain trauma show that post-trauma (10 min after controlled cortical impact) administration of GCEE by intraperitoneal injection results in a decrease in acute blood-brain barrier permeability. These data suggest that the beneficial effects of GCEE on brain endothelial cells and microvessels may contribute to its potential efficacy as a neuroprotective agent in traumatic brain injury.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Capillary Permeability/physiology , Dipeptides/therapeutic use , Endothelial Cells/metabolism , Neuroprotective Agents/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Brain Injuries/prevention & control , Capillary Permeability/drug effects , Cell Line , Dipeptides/pharmacology , Endothelial Cells/drug effects , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/metabolism
16.
Drug Chem Toxicol ; 34(2): 129-38, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21314463

ABSTRACT

D-serine plays a significant role in neuronal activity, including learning, memory, neuronal migration at developmental stages, and cell-death signaling. It has been also suggested that D-serine can potantiate the neurotoxicity induced by N-methyl-D-aspartate (NMDA) receptor activation due to its coagonist function. However, little is known about the role of D-serine in oxidative stress mechanisms. The aim of this study was to determine the possible neurotoxic or oxidative effects of the dose- (50-200 mg/kg) and time-dependent (2 or 6 hours) D-serine administration on lipid, protein, DNA, mitochondrial integrity (i.e., function), levels of antioxidant enzyme activities (e.g., catalase, glutathione peroxidase, and superoxide dismutase), and glutathione (GSH) in the rat brain. Our results showed that D-serine significantly increases the levels of lipid peroxidation, protein carbonyls, and DNA damage. In addition, D-serine treatment changes cellular antioxidant status due to the decreased levels of antioxidant enzymes, GSH, and mitochondrial function. Therefore, it is concluded that the regulation of D-serine levels in the brain may be an important target for the development of neuroprotective strategies against neurodegenerative processes where excitotoxicity is involved.


Subject(s)
Brain/drug effects , Oxidants/toxicity , Oxidative Stress/drug effects , Serine/toxicity , Animals , Brain/metabolism , Cell Fractionation , DNA/drug effects , DNA Damage , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/physiology , Oxidoreductases/metabolism , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects
17.
J Pharm Pharmacol ; 62(8): 1010-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20663035

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effect of gamma-glutamylcysteine ethylester (GCEE), a precursor of glutathione biosynthesis, on the levels of glutathione, formation of reactive oxygen species and c-fos mRNA expression in rat hippocampus and cortex in kainic acid-induced excitotoxicity. METHODS: Sprague-Dawley rats were used and divided into four groups: control, kainic acid (10 mg/kg), GCEE (10 mg/kg) and kainic acid (10 mg/kg) + GCEE (10 mg/kg). Kainic acid and GCEE were administered to the rats intraperitoneally. The levels of glutathione and the expressions of c-fos mRNA in hippocampus and cortex tissues were determined using spectrophotometric and reverse transcription followed real-time PCR methods, respectively. Formation of reactive oxygen species was determined using dichlorofluorescin fluorescence in brain synaptosomes treated with kainic acid or GCEE in vitro. KEY FINDINGS: Kainic acid treatment significiantly upregulated the expression of c-fos mRNA in the hippocampus and cortex when compared to the control group. GCEE treatment significantly decreased the levels of c-fos mRNA in the cortex when compared to the kainic acid-treated group. GCEE treatment against kainic acid significantly increased the levels of glutathione in the cortex and hippocampus, and decreased the levels of formation of reactive oxygen species when compared to kainic acid-treated synaptosomes. CONCLUSIONS: The increased levels of glutathione and the reduced levels of reactive oxygen species formation lead us to conclude that GCEE may be beneficial as a potential antioxidant against neurodegenerative processes where excitotoxicity is involved.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/drug effects , Dipeptides/pharmacology , Genes, fos , Glutathione/metabolism , Hippocampus/drug effects , Kainic Acid/toxicity , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/administration & dosage , Cerebral Cortex/metabolism , Dipeptides/administration & dosage , Epilepsy, Tonic-Clonic/chemically induced , Hippocampus/metabolism , Injections, Intraperitoneal , Kainic Acid/administration & dosage , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry , Synaptosomes/drug effects , Synaptosomes/metabolism , Time Factors
18.
Free Radic Res ; 44(5): 513-21, 2010 May.
Article in English | MEDLINE | ID: mdl-20214503

ABSTRACT

The aim of this study was to investigate the effect of gamma-Glutamylcysteine Ethyl Ester (GCEE) on the levels of GSH, caspase-3 activity, DNA damage and the expressions of Bcl-2, Bax and p53 mRNAs in rat hippocampus after status epilepticus (SE) induced by systemic kainic acid (KA). The male rats were divided into four groups as controls, KA (10 mg/kg), GCEE (10 mg/kg) and KA+GCEE. Glutathione (GSH) levels and caspase-3 activity were determined spectrophotometrically and colourimetrically, respectively. DNA damage and Bcl-2, Bax and p53 mRNA expressions were quantified by comet assay and reverse transcription followed by RT-PCR, respectively. KA treatment significantly depleted GSH levels, induced DNA damage, caspase-3 activity and the expressions of p53 and Bax mRNA. GCEE treatment protected GSH levels, decreased DNA damage and the levels of p53 and Bax/Bcl-2 mRNA against KA injection. These results indicate that GCEE treatment at the dose of 10 mg/kg is capable to protect the depleted levels of GSH and shows an anti-apoptotic activity due to the decreased levels of apoptotic biomarkers in the rat hippocampus after SE induced by KA.


Subject(s)
Apoptosis/drug effects , Dipeptides/pharmacology , Hippocampus/drug effects , Kainic Acid/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , DNA Damage , Hippocampus/metabolism , Hippocampus/pathology , Male , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
19.
AAPS PharmSciTech ; 10(1): 104-12, 2009.
Article in English | MEDLINE | ID: mdl-19184450

ABSTRACT

In this study, we investigated the in vitro characteristics of mefenamic acid (MA) microparticles as well as their effects on DNA damage. MA-loaded chitosan and alginate beads were prepared by the ionotropic gelation process. Microsponges containing MA and Eudragit RS 100 were prepared by quasi-emulsion solvent diffusion method. The microparticles were characterized in terms of particle size, surface morphology, encapsulation efficiency, and in vitro release profiles. Most of the formulation variables manifested an influence on the physical characteristics of the microparticles at varying degrees. We also studied the effects of MA, MA-loaded microparticles, and three different polymers on rat brain cortex DNA damage. Our results showed that DNA damage was higher in MA-loaded Eudragit microsponges than MA-loaded biodegradable chitosan or alginate microparticles.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cerebral Cortex/drug effects , DNA Damage , Mefenamic Acid/pharmacology , Neuroprotective Agents/pharmacology , Acrylic Resins/chemistry , Alginates/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cerebral Cortex/pathology , Chemistry, Pharmaceutical , Chitosan/chemistry , Dosage Forms , Drug Carriers , Drug Compounding , Excitatory Amino Acid Agonists/toxicity , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Kainic Acid/toxicity , Kinetics , Mefenamic Acid/chemistry , Neuroprotective Agents/chemistry , Particle Size , Rats , Rats, Sprague-Dawley , Solubility , Technology, Pharmaceutical/methods
20.
Int J Neurosci ; 119(2): 227-39, 2009.
Article in English | MEDLINE | ID: mdl-19125376

ABSTRACT

The selective estrogen receptor modulators (SERMs) are compounds that activate the estrogen receptors with different estrogenic and antiestrogenic tissue-specific effects. The similar effects of SERMs on estrogen encourage the efforts in the research of neuroprotective effects of SERMs. In our study, the potential neuroprotective effects of raloxifene were investigated on the brain cortex of ovariectomized rats after kainic acid-induced oxidative stress. To show the neuroprotective effect of raloxifene against a neurodegenerative agent, kainic acid, expression of Bcl-2, total glutathione (GSH), and nitrite-nitrate levels were investigated in the rat brain cortex. Our results demostrate that raloxifene treatment against oxidative stress significantly increases the expression of Bcl-2 and the level of GSH in the brain cortex.


Subject(s)
Glutathione/metabolism , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Disease Models, Animal , Female , Kainic Acid/antagonists & inhibitors , Kainic Acid/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Ovariectomy , Oxidative Stress/physiology , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...