Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Pharmacol Sci ; 99(1): 117-20, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16141634

ABSTRACT

Accumulating evidence indicates that growth hormone (GH) might be effective at preventing the development of Alzheimer's disease. However, exogenous GH treatment has exhibited side effects for clinical application; thus supplementation with amino acids to promote the release of GH could be a possible alternative treatment. In this study, mice that were fed with a diet of GH-releasing supplements had significantly attenuated memory impairments and hippocampal changes in the acetylcholinesterase activity and acetylcholine level induced by amyloid beta protein (Abeta) (1 - 42). Our results suggest that the use of GH-releasing supplement exerts beneficial effects on the memory impairment induced by Abeta (1 - 42).


Subject(s)
Aging/physiology , Amino Acids/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Growth Hormone/blood , Memory Disorders/prevention & control , Memory/drug effects , Peptide Fragments/antagonists & inhibitors , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Amino Acids/administration & dosage , Animals , Brain/drug effects , Brain/enzymology , Brain Chemistry , Growth Hormone/pharmacology , Male , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Time Factors
2.
Behav Brain Res ; 158(1): 143-57, 2005 Mar 07.
Article in English | MEDLINE | ID: mdl-15680202

ABSTRACT

Current evidence suggests that ginsenosides inhibit methamphetamine (MA)-induced changes in behavior, but the precise mechanisms that underlie this effect are yet to be determined. We examined the role of adenosine receptors in the ginsenoside-induced changes in hyperlocomotion and conditioned place preference (CPP) in mice that occurred in response to administration of MA (2 mg/kg, i.p. x 1 or 2 mg/kg, i.p. x 6). Changes in circling behavior paralleled changes in CPP in the presence of MA. Pre-treatment with ginsenosides (50 or 150 mg/kg, i.p.) attenuated the MA-induced circling behavior and CPP. This attenuation was reversed by the adenosine A2A receptor antagonist 1,3,7-trimethyl-8-(3-chrostyryl)xanthine (CSC; 0.5 and 1.0 mg/kg) in a dose-dependent manner, but neither the adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dimethylxanthine (CPT; 0.5 and 1.0 mg/kg) nor the A2B receptor antagonist alloxazine (ALX; 1.5 and 3.0 mg/kg) had any such effect. MA-induced increases in activator protein (AP)-1 DNA binding activity, Fos-related antigen immunoreactivity (FRA-IR), proenkephalin mRNA expression, and proenkephalin-like immunoreactivity were reduced consistently in the striatum of animals that were pretreated with ginsenosides. These reductions were largely prevented by CSC, but not by CPT or ALX. Our results suggest that the stimulation of A2A receptors by ginsenosides attenuates the changes in behavior and the increases in AP-1 DNA binding activity, FRA-IR, and proenkephalin gene expression in mouse striatum that are induced by MA.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Corpus Striatum/drug effects , Enkephalins/metabolism , Ginsenosides/pharmacology , Methamphetamine/pharmacology , Protein Precursors/metabolism , Receptor, Adenosine A2A/physiology , Transcription Factor AP-1/metabolism , Adenosine A2 Receptor Antagonists , Analysis of Variance , Animals , Blotting, Northern/methods , Cell Count/methods , Conditioning, Operant/drug effects , Corpus Striatum/physiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Electrophoretic Mobility Shift Assay/methods , Enkephalins/genetics , Gene Expression/drug effects , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Oncogene Proteins v-fos/metabolism , Protein Precursors/genetics , RNA, Messenger/metabolism , Stereotyped Behavior/drug effects , Teprotide/pharmacology , Time Factors
3.
Br J Pharmacol ; 144(7): 908-18, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15723099

ABSTRACT

In a previous study, we demonstrated that a dextromethorphan analog, dimemorfan, has neuroprotective effects. Dextromethorphan and dimemorfan are high-affinity ligands at sigma1 receptors. Dextromethorphan has moderate affinities for phencyclidine sites, while dimemorfan has very low affinities for such sites, suggesting that these sites are not essential for the anticonvulsant actions of dimemorfan. Kainate (KA) administration (10 mg kg(-1), i.p.) produced robust convulsions lasting 4-6 h in rats. Pre-treatment with dimemorfan (12 or 24 mg kg(-1)) reduced seizures in a dose-dependent manner. Dimemorfan pre-treatment also attenuated the KA-induced increases in c-fos/c-jun expression, activator protein (AP)-1 DNA-binding activity, and loss of cells in the CA1 and CA3 fields of the hippocampus. These effects of dimemorfan were comparable to those of dextromethorphan. The anticonvulsant action of dextromethorphan or dimemorfan was significantly counteracted by a selective sigma1 receptor antagonist BD 1047, suggesting that the anticonvulsant action of dextromethorphan or dimemorfan is, at least in part, related to sigma1 receptor-activated modulation of AP-1 transcription factors. We asked whether dimemorfan produces the behavioral side effects seen with dextromethorphan or dextrorphan (a phencyclidine-like metabolite of dextromethorphan). Conditioned place preference and circling behaviors were significantly increased in mice treated with phencyclidine, dextrorphan or dextromethorphan, while mice treated with dimemorfan showed no behavioral side effects. Our results suggest that dimemorfan is equipotent to dextromethorphan in preventing KA-induced seizures, while it may lack behavioral effects, such as psychotomimetic reactions.


Subject(s)
Dextromethorphan/analogs & derivatives , Dextromethorphan/therapeutic use , Morphinans/therapeutic use , Receptors, sigma/metabolism , Seizures/drug therapy , Animals , Dextromethorphan/chemistry , Kainic Acid/antagonists & inhibitors , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Morphinans/chemistry , Rats , Rats, Sprague-Dawley , Seizures/metabolism
4.
Life Sci ; 75(23): 2751-64, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15464827

ABSTRACT

A recent investigation indicated that Polygala tenuifolia Willdenow extract (PTE) possesses a potential antipsychotic effect. In this study, we examined the effects of PTE on the cocaine-induced changes in locomotor activity, conditioned place preference (CPP), fos-related antigen-immunoreactivity (FRA-IR), and activator protein (AP)-1 DNA binding activity. Cocaine-induced behavioral effects (hyperlocomotion and CPP) occurred in parallel with increases in FRA-IR and AP-1 DNA binding activity in the nucleus accumbens. These responses induced by cocaine were consistently attenuated by concurrent treatment with PTE (25 mg or 50 mg/kg/day, i.p. x 7). The adenosine A2A receptor antagonist, 1,3,7-trimethyl-8-(3-chlorostyrl)xanthine (0.5 or 1.0 mg/kg, i.p.), reversed the PTE-mediated pharmacological action in a dose related manner; neither the adenosine A(1) receptor antagonist, 8-cyclopentyl-1,3-dimethylxanthine (0.5 or 1.0 mg/kg, i.p.) nor the A2B receptor antagonist, alloxazine (1.5 or 3.0 mg/kg, i.p.) significantly affected this pharmacological action. Our results suggest that PTE prevents cocaine-induced behavioral effects, at least in part, via the activation of the adenosine A2A receptor.


Subject(s)
Cocaine/antagonists & inhibitors , Locomotion/drug effects , Plant Roots/chemistry , Polygala/chemistry , Spatial Behavior/drug effects , Analysis of Variance , Animals , Autoradiography , DNA/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/immunology , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-fos/immunology , Purinergic P1 Receptor Antagonists , Transcription Factor AP-1/metabolism
5.
Behav Brain Res ; 155(2): 185-96, 2004 Dec 06.
Article in English | MEDLINE | ID: mdl-15364477

ABSTRACT

We have demonstrated that oxidative stress is involved, at least in part, in beta-amyloid protein (Abeta)-induced neurotoxicity in vivo [Eur. J. Neurosci. 1999;11:83-90; Neuroscience 2003;119:399-419]. However, mechanistic links between oxidative stress and memory loss in response to Abeta remain elusive. In the present study, we examined whether oxidative stress contributes to the memory deficits induced by intracerebroventricular injection of Abeta (1-42) in mice. Abeta (1-42)-induced memory impairments were observed, as measured by the water maze and passive avoidance tests, although these impairments were not found in Abeta (40-1)-treated mice. Treatment with antioxidant alpha-tocopherol significantly prevented memory impairment induced by Abeta (1-42). Increased activities of the cytosolic Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and mitochondrial Mn-superoxide dismutase (Mn-SOD) were observed in the hippocampus and cerebral cortex of Abeta (1-42)-treated animals, as compared with Abeta (40-1)-treated mice. The induction of Cu,Zn-SOD was more pronounced than that of Mn-SOD after Abeta (1-42) insult. However, the concomitant induction of glutathione peroxidase (GPX) in response to significant increases in SOD activity was not seen in animals treated with Abeta (1-42). Furthermore, glutathione reductase (GRX) activity was only increased at 2h after Abeta (1-42) injection. Production of malondialdehyde (lipid peroxidation) and protein carbonyl (protein oxidation) remained elevated at 10 days post-Abeta (1-42), but the antioxidant alpha-tocopherol significantly prevented these oxidative stresses. Therefore, our results suggest that the oxidative stress contributes to the Abeta (1-42)-induced learning and memory deficits in mice.


Subject(s)
Antioxidants/pharmacology , Cerebral Cortex/drug effects , Hippocampus/drug effects , Memory Disorders/prevention & control , Memory Disorders/physiopathology , Oxidative Stress/drug effects , alpha-Tocopherol/pharmacology , Amyloid beta-Peptides/administration & dosage , Analysis of Variance , Animals , Avoidance Learning/drug effects , Cerebral Cortex/enzymology , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Hippocampus/enzymology , Injections, Intraventricular , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Memory/drug effects , Memory Disorders/chemically induced , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Peptide Fragments/administration & dosage , Superoxide Dismutase/metabolism , Time Factors
6.
Clin Exp Pharmacol Physiol ; 31(5-6): 320-6, 2004.
Article in English | MEDLINE | ID: mdl-15191405

ABSTRACT

The effect of pyrrolidine dithiocarbamate (PDTC) on kainate (KA)-induced neurotoxicity was examined in Sprague-Dawley rats. At 10 mg/kg, i.p., KA produced seizures accompanied by neuronal loss in the hippocampus and increased levels of malondialdehyde (MDA) and protein carbonyl. Pretreatment with PDTC (100 or 200 mg/kg, p.o., every 12 h x 5) blocked KA-induced neurotoxicities (seizures, increases in MDA and protein carbonyl and neuronal losses) in a dose-dependent manner. These effects were counteracted by the adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dimethylxanthine (25 or 50 micro g/kg, i.p.), but not by the A(2A) receptor antagonist 1,3,7-trimethyl-8-(3-chlorostyryl)xanthine (0.5 or 1 mg/kg, i.p.) or the A(2B) receptor antagonist alloxazine (1.5 or 3.0 mg/kg, i.p.). Our results suggest that the anticonvulsant and neuroprotective effects of PDTC are mediated, at least in part, via adenosine A(1) receptor stimulation.


Subject(s)
Hippocampus/drug effects , Kainic Acid/toxicity , Neuroprotective Agents/pharmacology , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/pathology , Male , Rats , Rats, Sprague-Dawley , Seizures/pathology , Seizures/prevention & control
7.
Biol Pharm Bull ; 27(6): 906-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15187444

ABSTRACT

The effect of PAP 9704, a traditional prescription in Korea consisting of Polygala tenuifolia, Acorus gramineus, and Poria cocos at a ratio of 1:1:1 (dry weight), on methamphetamine (MA)-induced hyperlocomotion was examined in mice. The increased locomotor activity induced by MA (1 mg/kg/d, i.p. x 7) was significantly attenuated by co-administration with PAP 9704 (100 or 200 mg/kg/d, p.o. x 7) in a dose dependent manner. Consistently, it was found that the hyperlocomotor activity occurred in parallel with the expression of striatal fos-related antigen immunoreactivity. The adenosine A(2A) receptor antagonist, 1,3,7-trimethyl-8-(3-chlorostyryl)xanthine (0.5 or 1.0 mg/kg, i.p.), significantly reversed the pharmacological action of PAP 9704 in a dose related manner, but the adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dimethylxanthine (0.5 or 1.0 mg/kg, i.p.) and the A(2B) receptor antagonist alloxazine (1.5 or 3.0 mg/kg, i.p.) did not significantly affect this pharmacological action. Our results suggest that PAP 9704 prevents MA-induced hyperlocomotion, at least in part, via the stimulation of the adenosine A(2A) receptor.


Subject(s)
Hyperkinesis/drug therapy , Methamphetamine/toxicity , Plant Preparations/pharmacology , Plants, Medicinal , Receptor, Adenosine A2A/physiology , Animals , Hyperkinesis/chemically induced , Korea , Locomotion/drug effects , Locomotion/physiology , Male , Methamphetamine/antagonists & inhibitors , Mice , Mice, Inbred BALB C
8.
Behav Brain Res ; 151(1-2): 267-76, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15084442

ABSTRACT

A dextromethorphan (3-methoxy-17-methylmorphinan) analog, dimemorfan (3-methyl-N-methylmorphinan) that is not metabolized to dextrorphan [3-hydroxy-17-methylmorphinan, which induces phencyclidine (PCP)-like behavioral effects], attenuates maximal electroshock seizures. However, the pharmacological mechanism of action of dimemorfan remains to be determined. In this study, we assessed the locomotor activity mediated by these morphinans. Circling behavior was pronounced in mice treated with PCP or dextrorphan, while animals treated with dextromethorphan exhibited moderate behaviors. Dimemorfan did not show any significant behavioral effects. We used BAY k-8644 (an L-type Ca2+ channel agonist in the dihydropyridine class) to explore the effects of dextromethorphan and dimemorfan on the convulsant activity regulated by calcium channels. Intracerebroventricular injection of BAY k-8644 (37.5 microg) significantly induced seizures in mice. As with dextromethorphan (6.25 or 12.5 mg/kg), dimemorfan (6.25 or 12.5 mg/kg) pretreatment significantly attenuated BAY k-8644-induced seizures in a dose-dependent manner. BAY k-8644-induced seizure activity paralleled increased expression of c-fos and c-jun, AP-1 DNA binding activity, and fos-related antigen immunoreactivity. Pretreatment with dextromethorphan or dimemorfan significantly attenuated the expression induced by BAY k-8644. Therefore, our results suggest that the anticonvulsant effects of dextromethorphan and dimemorfan are mediated, at least in part, via L-type calcium channel, and that dimemorfan is equipotent to dextromethorphan in preventing BAY k-8644-induced seizures, while it lacks behavioral side effects related to psychotomimetic reactions.


Subject(s)
Anticonvulsants/therapeutic use , Calcium Channels, L-Type/metabolism , Morphinans/analysis , Morphinans/therapeutic use , Seizures/prevention & control , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester , Animals , Behavior, Animal , Blotting, Northern/methods , Blotting, Western/methods , Calcium Channels, L-Type/drug effects , Cell Count/methods , Densitometry/methods , Dose-Response Relationship, Drug , Drug Interactions , Electrophoretic Mobility Shift Assay/methods , Hippocampus/drug effects , Hippocampus/metabolism , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Morphinans/chemistry , Morphinans/pharmacology , Motor Activity/drug effects , Phencyclidine/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA/isolation & purification , RNA/metabolism , Reaction Time/drug effects , Seizures/chemically induced
9.
Neurosci Lett ; 355(3): 231-5, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14732473

ABSTRACT

We examined the effects of the antioxidant propolis on seizures induced by kainic acid (KA). Sprague-Dawley rats received propolis (75 and 150 mg/kg, p.o.) five times at 12 h intervals. KA (10 mg/kg, i.p.) was injected 1 h after the last propolis treatment. Pretreatment with propolis significantly attenuated KA-induced seizures and KA-induced increases in hippocampal AP-1 DNA binding activity in a dose-dependent manner. KA induced increases in the levels of malondialdehyde and protein carbonyl, and a decrease in the ratio of GSH/GSSG. These oxidative stresses and neuronal degenerations were significantly attenuated by pretreatment with propolis. The neuroprotective effects of propolis appeared to be counteracted by adenosine receptor antagonists [A1 antagonist, 8-cyclopentyl-1,3-dimethylxanthine (25 or 50 microg/kg); A2A antagonist, 1,3,7-trimethyl-8-(3-chlorostyryl)xanthine (0.5 or 1 mg/kg); and A2B antagonist, alloxazine (1.5 or 3.0 mg/kg)]. However, this counteraction was most pronounced in the presence of the A1 antagonist. Our results suggest that the protective effect of propolis against KA-induced neurotoxic oxidative damage is, at least in part, via adenosine A1 receptor modulation.


Subject(s)
Antioxidants/pharmacology , Kainic Acid/toxicity , Propolis/pharmacology , Receptor, Adenosine A1/physiology , Adenosine A1 Receptor Antagonists , Animals , Flavins/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiology , Kainic Acid/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley
10.
Life Sci ; 72(16): 1883-95, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12586225

ABSTRACT

Interest in dextromethorphan (DM) has been renewed because of its anticonvulsant and neuroprotective properties. However, DM at supra-antitussive doses can produce psychotomimetic effects in humans. Recently, we demonstrated that DM exerts psychotropic effects in mice [Neurosci. Lett. 288 (2000) 76, Life Sci. 69 (2001) 615]. We synthesized a series of compounds with a modified morphinan ring system, with the intention of developing compounds that retain the anticonvulsant activity with weak psychotropic effects [Bioorg. Med. Chem. Lett. 11 (2001) 1651]. In order to extend our understanding of the pharmacological intervention of these morphinans, we assessed their behavioral effects, and then examined whether they exert protective effects on maximal electroshock convulsions (MES) in mice. Repeated treatment (20 or 40 mg/kg, i.p./day x 7) with DM or dextrorphan (a major metabolite of DM; DX) significantly enhanced locomotor activity in a dose-related manner. This locomotor stimulation was accentuated more in the animals treated with DX, and might be comparable to that of phencyclidine (PCP). By contrast, treatment with a metabolite of DM [3-methoxymorphinan (3MM) or 3-hydroxymorphinan (3HM)], 3-allyloxy-17-methylmorphinan (CPK-5), or 3-cyclopropylmethoxy-17-methylmorphinan (CPK-6) did not significantly alter locomotor activity or patterns. The behavioral effects mediated by these morphinans and PCP paralleled the effects of conditioned place preference. DM, DX, CPK-5, and CPK-6 had anticonvulsant effects against MES, while 3MM and 3HM did not show any anticonvulsant effects. We found that DM, DX, CPK-5 and CPK-6 were high-affinity ligands at sigma(1) receptors, while they all had low affinity at sigma(2) receptors. DX had relatively higher affinity for the PCP sites than DM. By contrast, CPK-5 and CPK-6 had very low affinities for PCP sites, suggesting that PCP sites are not requisites for their anticonvulsant actions. Our results suggest that the new morphinan analogs are promising anticonvulsants that are devoid of PCP-like behavioral side effects, and their anticonvulsant actions may be, in part, mediated via sigma(1) receptors.


Subject(s)
Anticonvulsants/therapeutic use , Dextromethorphan/analogs & derivatives , Electroshock , Morphinans/therapeutic use , Seizures/prevention & control , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/metabolism , Dextromethorphan/therapeutic use , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Morphinans/administration & dosage , Morphinans/metabolism , Radioligand Assay , Receptors, sigma/metabolism , Seizures/etiology
11.
Life Sci ; 72(7): 769-83, 2003 Jan 03.
Article in English | MEDLINE | ID: mdl-12479976

ABSTRACT

The effects of dextromethorphan (DM), and its major metabolite dextrorphan (DX) on kainic acid-induced seizures in mice were examined. Intracerebroventricular DM or DX (5 or 10 microg/0.5 microl) pretreatment significantly attenuated seizures induced by kainic acid (0.07 microg/0.07 microl) in a dose-related manner. DM or DX pretreatment significantly attenuated kainic acid-induced increases in AP-1 DNA-binding activity and fos-related antigen-immunoreactivity as well as neuronal loss in the hippocampus. DM appears to be a more potent neuroprotectant than DX. Since the high-affinity DM binding sites are recognized as being identical to the sigma-1 site, we examined the role of the sigma-1 receptor on the pharmacological action mediated by DM or DX. Pretreatment with the sigma-1 receptor antagonist BD1047 (2.5 or 5 mg/kg, i.p.) blocked the neuroprotection by DM in a dose-related manner. This effect of BD 1047 was more pronounced in the animals treated with DM than in those treated with DX. Combined, our results suggest that metabolism of DM to DX is not essential for DM to exert its effect. They also suggest that DM provides neuroprotection from kainic acid via sigma-1 receptor modulation.


Subject(s)
Anticonvulsants/therapeutic use , Dextromethorphan/therapeutic use , Dextrorphan/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Seizures/drug therapy , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/metabolism , DNA/metabolism , Dextromethorphan/administration & dosage , Dextromethorphan/metabolism , Dextrorphan/administration & dosage , Dextrorphan/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Injections, Intraventricular , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred DBA , Neurons/drug effects , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Opioid, delta/metabolism , Seizures/chemically induced , Transcription Factor AP-1/metabolism
12.
Brain Res Mol Brain Res ; 108(1-2): 60-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12480179

ABSTRACT

In order to evaluate the role of transforming growth factor (TGF)-beta3 in the neurodegenerative process, we examined the levels of mRNA and immunocytochemical distribution for TGF-beta3 in the rat hippocampus after systemic kainic acid (KA) administration. Hippocampal TGF-beta3 mRNA level was reduced 3 h after KA injection. However, the levels of TGF-beta3 mRNA were elevated 1 day post-KA and lasted for at least 30 days. A mild TGF-beta3 immunoreactivity (TGF-beta3-IR) in the Ammon's horn and a moderate TGF-beta3-IR in the dentate granule cells were observed in the normal hippocampus. The CA1 and CA3 neurons lost their TGF-beta3-IR, while TGF-beta3-positive glia-like cells proliferated mainly throughout the CA1 sector and had an intense immunoreactivity at 7, 15 and 30 days after KA. This immunocytochemical distribution of TGF-beta3-positive non-neuronal populations was similar to that of glial fibrillary acidic protein (GFAP)-positive cells. Double labeling immunocytochemical analysis demonstrated colocalization of TGF-beta3- and GFAP-immunoreactivity in the same cells. These findings suggest a compensatory mechanism of astrocytes for the synthesis of TGF-beta3 protein in response to KA-induced neurodegeneration. In addition, exogenous TGF-beta3 (5 or 10 ng/i.c.v.) significantly attenuated KA-induced seizures and neuronal damages in a dose-related manner. Therefore, our results suggest that TGF-beta3 plays an important role in protective mechanisms against KA-induced neurodegeneration.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Kainic Acid/pharmacology , Transforming Growth Factor beta/metabolism , Animals , Behavior, Animal/drug effects , Cell Size , Excitatory Amino Acid Agonists/toxicity , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Immunohistochemistry , Kainic Acid/toxicity , Male , Neuroglia/metabolism , Neuroprotective Agents/metabolism , Rats , Rats, Inbred F344 , Seizures/chemically induced , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta3
13.
Behav Brain Res ; 131(1-2): 211-20, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11844588

ABSTRACT

We have demonstrated that seizures induced by kainic acid (KA) are, at least in part, mediated via oxidative stress in rats [Life. Sci. 61 (1997) PL373; Brain Res. 853 (2000) 215; Brain Res. 874 (2000) 15; Neurosci. Lett. 281 (2000) 65]. In order to extend our findings, we employed the rodent aging model in this study. After KA treatments (once a day for 5 days; 20,20,20,20 and 40 mg/kg, i.p.), several parameters reflecting neurotoxic behaviors, oxidative stress [malondialdehyde (MDA) and protein carbonyl] and aging (lipofuscin-like substances) were compared between senile-prone (P8) and resistant (R1) strains of 9-month-old male senescence-accelerated mice (SAM). KA-induced neurotoxic signs as shown by mortality and seizure activity were more accentuated in the SAM-P8 than in the SAM-R1. Levels of MDA and carbonyl are consistently higher in the hippocampus of SAM-P8 than that of SAM-R1. Significant increases in the values of MDA and carbonyl were observed 4 h or 2 days after the final KA administration. This finding was more pronounced in the SAM-P8 than in the SAM-R1. Although a significant loss of hippocampal neurons was observed 7 days post-KA, at this time the MDA and carbonyl content had returned to near control levels. In contrast, fluorescent lipofuscin-like substances and lipofuscin granules were significantly increased 7 days after KA treatments. Therefore, our data suggests that mice in the senescence model are more susceptible to KA-induced seizures/oxidative damage, and that oxidative damage could be one of the casual factors in the accumulation of lipofuscin.


Subject(s)
Aging/genetics , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/physiology , Kainic Acid/pharmacology , Lipofuscin/metabolism , Animals , Brain Chemistry/drug effects , Cell Count , Excitatory Amino Acid Agonists/administration & dosage , Hippocampus/cytology , Hippocampus/metabolism , Kainic Acid/administration & dosage , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Mice , Mice, Inbred Strains , Microscopy, Electron , Nerve Tissue Proteins/metabolism , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/psychology , Oxidative Stress/drug effects , Seizures/chemically induced , Seizures/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...