Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 766112, 2021.
Article in English | MEDLINE | ID: mdl-34938290

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global health concern. The development of vaccines with high immunogenicity and safety is crucial for controlling the global COVID-19 pandemic and preventing further illness and fatalities. Here, we report the development of a SARS-CoV-2 vaccine candidate, Nanocovax, based on recombinant protein production of the extracellular (soluble) portion of the spike (S) protein of SARS-CoV-2. The results showed that Nanocovax induced high levels of S protein-specific IgG and neutralizing antibodies in three animal models: BALB/c mouse, Syrian hamster, and a non-human primate (Macaca leonina). In addition, a viral challenge study using the hamster model showed that Nanocovax protected the upper respiratory tract from SARS-CoV-2 infection. Nanocovax did not induce any adverse effects in mice (Mus musculus var. albino) and rats (Rattus norvegicus). These preclinical results indicate that Nanocovax is safe and effective.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19 Vaccines/toxicity , COVID-19/prevention & control , Immunogenicity, Vaccine/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cricetinae , Macaca , Mice , Rats , SARS-CoV-2 , Vaccines, Synthetic/immunology , Vaccines, Synthetic/toxicity
2.
Food Chem Toxicol ; 136: 110986, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31760073

ABSTRACT

It is recognized that d-amphetamine (AMPH)-induced hyperactivity is thought to be a valid animal model of mania. In the present study, we investigated whether a proinflammatory oxidative gene indoleamine-2,3-dioxygenase (IDO) is involved in AMPH-induced mitochondrial burden, and whether mood stabilizers (i.e., lithium and valproate) modulate IDO to protect against AMPH-induced mania-like behaviors. AMPH-induced IDO-1 expression was significantly greater than IDO-2 expression in the prefrontal cortex of wild type mice. IDO-1 expression was more pronounced in the mitochondria than in the cytosol. AMPH treatment activated intra-mitochondrial Ca2+ accumulation and mitochondrial oxidative burden, while inhibited mitochondrial membrane potential and activity of the mitochondrial complex (I > II), mitochondrial glutathione peroxidase, and superoxide dismutase, indicating that mitochondrial burden might be contributable to mania-like behaviors induced by AMPH. The behaviors were significantly attenuated by lithium, valproate, or IDO-1 knockout, but not in IDO-2 knockout mice. Lithium, valproate administration, or IDO-1 knockout significantly attenuated mitochondrial burden. Neither lithium nor valproate produced additive effects above the protective effects observed in IDO-1 KO in mice. Collectively, our results suggest that mood stabilizers attenuate AMPH-induced mania-like behaviors via attenuation of IDO-1-dependent mitochondrial stress, highlighting IDO-1 as a novel molecular target for the protective potential of mood stabilizers.


Subject(s)
Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lithium/therapeutic use , Valproic Acid/therapeutic use , Animals , Bipolar Disorder/chemically induced , Bipolar Disorder/metabolism , Calcium/metabolism , Dextroamphetamine , Glutathione Peroxidase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Locomotion/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Prefrontal Cortex/pathology , Superoxide Dismutase/metabolism
3.
J Psychopharmacol ; 32(11): 1233-1251, 2018 11.
Article in English | MEDLINE | ID: mdl-30207504

ABSTRACT

BACKGROUND: Oxidative stress and mitochondrial dysfunction have been implicated in the pathophysiology of schizophrenia. AIMS: We investigated whether antipsychotic clozapine modulates nicotinamide adenine dinucleotide phosphate oxidase and mitochondrial burdens induced by phencyclidine in mice. METHODS: We examined the effect of clozapine on nicotinamide adenine dinucleotide phosphate oxidase activation, mitochondrial burdens (i.e. oxidative stress and mitochondrial dysfunction), and activities of enzymatic antioxidant in the prefrontal cortex, and subsequent abnormal behaviors induced by repeated treatment with phencyclidine. p47 phox Knockout mice and LY294002, a phosphoinositide 3-kinase inhibitor, were employed to elucidate the pharmacological mechanism of clozapine. RESULTS: Phencyclidine treatment resulted in an early increase nicotinamide adenine dinucleotide phosphate oxidase activity, membrane translocation of p47 phox, interaction between p-Akt and p47 phox, and mitochondrial burdens in wild-type mice. Although these increases returned to near control level four days post-phencyclidine, mitochondrial superoxide dismutase and glutathione peroxidase activities were decreased at that time. Clozapine, LY294002, or p47 phox knockout significantly ameliorated social withdrawal and recognition memory deficits produced by phencyclidine. Importantly, LY294002 did not significantly alter the effects of clozapine against abnormal behaviors and the interaction between p-Akt and p47 phox induced by phencyclidine. Furthermore, neither LY294002 nor clozapine exhibited any additive effects to the protection afforded by p47 phox knockout against phencyclidine insult. CONCLUSION: Our results suggest that p47 phox gene mediates phencyclidine-induced mitochondrial burdens and abnormal behaviors, and that the interactive modulation between p47 phox and phosphoinositide 3-kinase/Akt is important for the understanding on the pharmacological mechanism of clozapine.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Mitochondria/drug effects , NADPH Oxidases/genetics , Animals , Antioxidants/metabolism , Behavior, Animal/drug effects , Chromones/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Morpholines/pharmacology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Phencyclidine/toxicity , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Signal Transduction/drug effects
4.
Mol Neurobiol ; 55(5): 4136-4159, 2018 May.
Article in English | MEDLINE | ID: mdl-28597397

ABSTRACT

Accumulating evidence has suggested that repeated treatment with methamphetamine (MA) resulted in cognitive impairments. Importantly, we show that selective upregulation of protein kinase Cδ (PKCδ) in the prefrontal cortex (PFC) of wild-type mice persisted for 28 days post withdrawal of MA. On day 28, the MA-induced increase in phospho-PKCδ expression and decrease in phospho-ERK1/2 expression were significantly attenuated by both the Src inhibitor PP2 and the dopamine D1 receptor antagonist SCH 23390. However, neither protein kinase A inhibitor H89 nor calmodulin-dependent protein kinase II inhibitor KN93 attenuated MA-induced alterations in phospho-PKCδ expression and phospho-ERK1/2 expression. Since PKCδ knockout (KO) significantly increased the expression of glutathione peroxidase (GPx)-1, we also utilized GPx-1 KO and GPx-1-overexpressing transgenic (GPx-1 TG) mice. Repeated MA treatment induced cognitive impairment, as assessed by the novel object recognition test. Moreover, the extent of cognitive impairment correlated with the extent of increased phospho-PKCδ expression and decreased GPx1 expression. In the absence of MA, exposure to novel objects increased phospho-ERK1/2 and GPx-1 expression in the PFC; however, these expression levels were decreased in the presence of MA. PKCδ KO and GPx-1 TG mice each exhibited significantly attenuated MA-induced decreases in phospho-ERK1/2 and GPx-1 expression. Consistently, PKCδ inhibition induces GPx/GSH-dependent antioxidant systems. More importantly, the antipsychotic drug clozapine significantly protected against cognitive impairment and was associated with alterations in phospho-ERK1/2 and phospho-PKCδ expression. However, GPx-1 KO potentiated MA-induced cognitive deficits and alterations in phospho-ERK1/2 and phospho-PKCδ expression. These results suggest that MA induces cognitive impairment by inhibiting ERK1/2 signaling, activating PKCδ, and inactivating GPx-1 by upregulating Src kinase or the D1 receptor. They also suggest that clozapine requires activation of ERK1/2 signaling via positive modulation between the phospho-PKCδ and GPx-1 genes to restore cognitive function.


Subject(s)
Gene Deletion , Glutathione Peroxidase/genetics , MAP Kinase Signaling System , Memory Disorders/enzymology , Protein Kinase C-delta/genetics , Up-Regulation/genetics , Animals , Behavior, Animal , Butadienes/pharmacology , Clozapine/pharmacology , Cognition Disorders/enzymology , Cognition Disorders/physiopathology , DNA-Binding Proteins , Glutathione Disulfide/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Isoquinolines/pharmacology , Male , Memory Disorders/physiopathology , Methamphetamine , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Nerve Tissue Proteins/metabolism , Nitriles/pharmacology , Nuclear Proteins/metabolism , Phosphorylation , Prefrontal Cortex/enzymology , Prefrontal Cortex/pathology , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Up-Regulation/drug effects , Glutathione Peroxidase GPX1
5.
Brain Res Bull ; 137: 71-78, 2018 03.
Article in English | MEDLINE | ID: mdl-29122692

ABSTRACT

Accumulating evidence suggests that neuroinflammation is one of the important etiologic factors of abusive and neuropsychiatric disorders. Platelet-activating factor (PAF) is potent proinflammatory lipid mediat1or and plays a pivotal role in neuroinflammatory disorders through the specific PAF receptor (PAF-R). Phencyclidine (PCP) induces a psychotomimetic state that closely resembles schizophrenia. Here, we investigated the role of PAF-R in the abnormal behaviors induced by PCP in mice. Repeated treatment with PCP resulted in a significant increase in PAF-R gene expression in the prefrontal cortex (PFC) and in the hippocampus. This increase was more pronounced in the PFC than hippocampus. Treatment with PCP resulted in a significant increase in nuclear translocation of the nuclear factor kappa beta (NF-κB) p65 and DNA binding activity, indicating that the proinflammatory molecule NF-κB was increased through up-regulation of PAF-R. Consistently, NF-κB activation was significantly protected by the PAF-R antagonist, ginkgolide B (Gink B), in PAF-R knockout mice and by the NF-κB inhibitor, pyrrolidine dithiocarbamate (PDTC). In addition, PCP-induced abnormal behaviors (i.e., reduced sociability, depression, cognitive impairment, and behavioral sensitization) were significantly attenuated by Gink B, in PAF-R knockout mice, and by PDTC. Importantly, PDTC did not significantly alter the attenuations observed in Gink B-treated mice or PAF-R knockout mice, indicating that NF-κB is a critical target for neuropsychotoxic modulation of PAF-R. Therefore, the results suggest that PAF-R mediates PCP-induced neuropsychotoxicity via a NF-κB-dependent mechanism, and that up-regulation of PAF-R may be associated with schizophrenia-like behavior in animal models.


Subject(s)
Antipsychotic Agents/pharmacology , Ginkgolides/pharmacology , Lactones/pharmacology , NF-kappa B/metabolism , Phencyclidine/toxicity , Platelet Membrane Glycoproteins/antagonists & inhibitors , Psychoses, Substance-Induced/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Disease Models, Animal , Down-Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Psychoses, Substance-Induced/metabolism , Psychoses, Substance-Induced/pathology , Psychoses, Substance-Induced/psychology , Pyrrolidines/pharmacology , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Thiocarbamates/pharmacology
6.
Food Chem Toxicol ; 110: 300-315, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29037473

ABSTRACT

We investigated whether ginsenoside Re (Re) modulates phencyclidine (PCP)-induced sociability deficits and recognition memory impairments to extend our recent finding. We examined the role of GPx-1 gene in the pharmacological activity of Re against mitochondrial dysfunction induced by PCP in the dorsolateral cortex of mice. Since mitochondrial oxidative stress activates NADPH oxidase (PHOX), we applied PHOX inhibitor apocynin for evaluating interactive modulation between GPx-1 and PHOX against PCP neurotoxicity. Sociability deficits and recognition memory impairments induced by PCP were more pronounced in GPx-1 knockout (KO) than in wild type (WT) mice. PCP-induced mitochondrial oxidative stress, mitochondrial dysfunction, and membrane translocation of p47phox were more evident in GPx-1 KO than in WT. Re treatment significantly attenuated PCP-induced neurotoxic changes. Re also significantly attenuated PCP-induced sociability deficits and recognition memory impairments. The attenuation by Re was comparable to that by apocynin. The attenuation was more obvious in GPx-1 KO than in WT. Importantly, apocynin did not show any additional positive effects on the neuroprotective activity of Re, indicating that PHOX is a molecular target for therapeutic activity of Re. Our results suggest that Re requires interactive modulation between GPx activity and PHOX (p47phox) to exhibit neuroprotective potentials against PCP insult.


Subject(s)
Cerebral Cortex/enzymology , Ginsenosides/administration & dosage , Glutathione Peroxidase/metabolism , Mitochondria/drug effects , NADPH Oxidases/metabolism , Panax/chemistry , Phencyclidine/adverse effects , Protective Agents/administration & dosage , Schizophrenia/drug therapy , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , NADPH Oxidases/genetics , Oxidative Stress/drug effects , Schizophrenia/enzymology , Schizophrenia/genetics , Schizophrenic Psychology , Glutathione Peroxidase GPX1
7.
Toxicol Appl Pharmacol ; 334: 158-166, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28916251

ABSTRACT

Dextromethorphan (DM) is a dextrorotatory isomer of levorphanol, a typical morphine-like opioid. When administered at supra-antitussive doses, DM produces psychotoxic and neurotoxic effects in humans. Although DM abuse has been well-documented, few studies have examined the effects of high-dose DM. The present study aimed to explore the effects of a single high dose of DM on mortality and seizure occurrence. After intraperitoneal administration with a high dose of DM (80mg/kg), Sprague-Dawley rats showed increased seizure occurrence and intensity. Hippocampal expression levels of N-methyl-d-aspartate (NMDA) receptor subunits (GluN1

Subject(s)
Dextromethorphan/toxicity , Dizocilpine Maleate/pharmacology , Naloxone/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/chemically induced , Seizures/drug therapy , Animals , Antitussive Agents/pharmacology , Dextromethorphan/administration & dosage , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Narcotic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics
8.
Arch Pharm Res ; 40(4): 403-428, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28243833

ABSTRACT

Clinical and preclinical studies have indicated that chronic methamphetamine (MA) use is associated with extensive neurodegeneration, psychosis, and cognitive impairment. Evidence from animal models has suggested a considerable role of excess dopamine or glutamate, oxidative stress, neuroinflammation, and apoptosis in MA-induced neurotoxicity, and that protein kinase Cδ might mediate the interaction among these factors. In addition, the relatively long-lasting and recurrent nature of MA psychosis has been reproduced in animals treated with various dosing regimens of MA, which have shown behavioral sensitization, sociability deficits, and impaired prepulse inhibition. Genetic predisposition as well as dopaminergic and glutamatergic alterations might be important in the development of MA psychosis. Neuroimaging studies have identified functional and morphological changes related to the cognitive dysfunction shown in chronic MA users. Failure in the task-evoked phosphorylation of extracellular signal-related kinase likely underlies MA-induced memory impairment. Recent progress has suggested certain roles of oxidative stress and neuroinflammation in the psychosis and cognitive deficits induced by repeated low doses of MA. This review provides a comprehensive description of pertinent findings from human and animal studies, with an emphasis on the current understanding of the underlying mechanisms of MA neuropsychotoxicity and its relevance to Parkinson's disease or schizophrenia.


Subject(s)
Central Nervous System Stimulants/adverse effects , Central Nervous System Stimulants/metabolism , Dopamine/metabolism , Methamphetamine/adverse effects , Methamphetamine/metabolism , Animals , Central Nervous System Stimulants/administration & dosage , Humans , Methamphetamine/administration & dosage
9.
Mol Neurobiol ; 54(9): 7042-7062, 2017 11.
Article in English | MEDLINE | ID: mdl-27796745

ABSTRACT

Escalating evidence suggests that the impairment of glutathione (GSH)-dependent systems is one of the etiologic factors of schizophrenia. GSH is an important substrate of glutathione peroxidase (GPx). Among GPx isozymes, selenium-dependent GPx (GPx-1) is recognized as a major type, and therefore, this study investigates the role of the GPx-1 gene in abnormal behaviors induced by repeated phencyclidine (PCP) treatment using GPx-1 knockout (KO) and overexpressing transgenic (GPx-1 TG) mice. PCP-induced abnormal behaviors were more pronounced in GPx-1 KO mice than abnormal behaviors in wild-type (WT) mice, and the abnormal behaviors were less pronounced in GPx-1 TG mice than abnormal behaviors in non-TG mice. PCP treatment significantly reduced GSH levels and enhanced oxidative burdens in the prefrontal cortex of the test animals. In addition, PCP treatment significantly upregulated the nuclear translocations of nuclear factor erythroid-2-related factor 2 (Nrf2) and nuclear factor kappa-B (NF-κB) p65, as well as their DNA binding activities and γ-glutamylcysteine (GCL) mRNA expression in WT and non-TG mice. However, GPx-1 KO abolished this upregulation system. In contrast, genetic overexpression of GPx-1 further upregulated Nrf2-dependent GSH synthetic system, but downregulated NF-κB p65 activity in the presence of PCP. Clozapine, an antipsychotic, significantly upregulated GPx-1 and Nrf2-dependent GSH synthetic systems in the presence of PCP, but failed to affect NF-κB p65 activity. Our results suggest that interactive modulations between the GPx-1 gene and Nrf2-dependent GSH induction are critical for attenuating PCP-induced abnormal behaviors in mice.


Subject(s)
Behavior, Animal , Glutathione Peroxidase/genetics , Neuroprotective Agents/metabolism , Animals , Antioxidants/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Clozapine/pharmacology , DNA/metabolism , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Reductase/metabolism , Haloperidol/pharmacology , Lipid Peroxidation/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Phencyclidine , Prefrontal Cortex/enzymology , Prefrontal Cortex/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Social Behavior , Superoxide Dismutase/metabolism , Time Factors , Transcription Factor RelA/metabolism , Glutathione Peroxidase GPX1
10.
J Med Food ; 19(10): 961-969, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27668757

ABSTRACT

Escalating evidence indicates that ginseng treatment protects against psychotoxic behaviors and memory impairment. Although the underlying mechanism of schizophrenia remains elusive, recent investigations proposed that downregulation of glutathione (GSH) can be involved in the pathogenesis of this disorder. Since little is known about the effects of ginseng in a schizophrenia-like animal model, we selected mountain-cultivated ginseng (MG) from a variety of ginseng extracts to investigate the effect of ginseng on the psychosis induced by phencyclidine (PCP) in mice. PCP (10 mg/kg/day, s.c.) was administered for 14 consecutive days. Novel object recognition, forced swimming, and social interaction tests were performed during the withdrawal period of 7 days. In addition, behavioral sensitization to an acute challenge of PCP was evaluated. The parameters of the GSH-dependent system in the prefrontal cortex (PFC) were examined. MG (200 mg/kg, i.p./day) or antipsychotic clozapine (10 mg/kg, p.o./day) was administered for seven consecutive days after the final PCP treatment. PCP significantly produced abnormal behaviors, followed by increases in Nrf2 nuclear translocation, its DNA binding activity, and glutamate-cysteine ligase (GCL) mRNA expression in the PFC. PCP treatment significantly decreased GSH/glutathione disulfide (GSSG) ratio and glutathione peroxidase (GPx) activity. MG significantly attenuated abnormal behaviors and the decreases in GSH/GSSG ratio and GPx activity induced by PCP. MG attenuated the increases in Nrf2 activity and GCL expression caused by PCP. The protective potentials of MG were comparable to those of clozapine. MG ameliorates PCP-induced schizophrenia-like psychosis in mice through the positive modulation of the glutathione system.

11.
Mol Neurobiol ; 53(9): 6251-6269, 2016 11.
Article in English | MEDLINE | ID: mdl-26563498

ABSTRACT

Selegiline is a monoamine oxidase-B (MAO-B) inhibitor with anti-Parkinsonian effects, but it is metabolized to amphetamines. Since another MAO-B inhibitor N-Methyl, N-propynyl-2-phenylethylamine (MPPE) is not metabolized to amphetamines, we examined whether MPPE induces behavioral side effects and whether MPPE affects dopaminergic toxicity induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Multiple doses of MPPE (2.5 and 5 mg/kg/day) did not show any significant locomotor activity and conditioned place preference, whereas selegiline (2.5 and 5 mg/kg/day) significantly increased these behavioral side effects. Treatment with MPPE resulted in significant attenuations against decreases in mitochondrial complex I activity, mitochondrial Mn-SOD activity, and expression induced by MPTP in the striatum of mice. Consistently, MPPE significantly attenuated MPTP-induced oxidative stress and MPPE-mediated antioxidant activity appeared to be more pronounced in mitochondrial-fraction than in cytosolic-fraction. Because MPTP promoted mitochondrial p53 translocation and p53/Bcl-xL interaction, it was also examined whether mitochondrial p53 inhibitor pifithrin-µ attenuates MPTP neurotoxicity. MPPE, selegiline, or pifithrin-µ significantly attenuated mitochondrial p53/Bcl-xL interaction, impaired mitochondrial transmembrane potential, cytosolic cytochrome c release, and cleaved caspase-3 in wild-type mice. Subsequently, these compounds significantly ameliorated MPTP-induced motor impairments. Neuroprotective effects of MPPE appeared to be more prominent than those of selegiline. MPPE or selegiline did not show any additional protective effects against the attenuation by p53 gene knockout, suggesting that p53 gene is a critical target for these compounds. Our results suggest that MPPE possesses anti-Parkinsonian potentials with guaranteed behavioral safety and that the underlying mechanism of MPPE requires inhibition of mitochondrial oxidative stress, mitochondrial translocation of p53, and pro-apoptotic process.


Subject(s)
Apoptosis/drug effects , Behavior, Animal , Dopaminergic Neurons/pathology , Mitochondria/metabolism , Phenethylamines/pharmacology , Selegiline/analogs & derivatives , Tumor Suppressor Protein p53/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Caspase 3/metabolism , Conditioning, Psychological , Cytochromes c/metabolism , Cytosol/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Electron Transport Complex I , Locomotion/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Monoamine Oxidase/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Phenethylamines/chemistry , Protein Binding/drug effects , Selegiline/chemistry , Selegiline/pharmacology , Sulfonamides/pharmacology , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/metabolism , Uncoupling Protein 2/metabolism , bcl-X Protein/metabolism
12.
Free Radic Biol Med ; 84: 355-372, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25843655

ABSTRACT

To determine the role of ceruloplasmin (Cp) in epileptic seizures, we used a kainate (KA) seizure animal model and examined hippocampal samples from epileptic patients. Treatment with KA resulted in a time-dependent decrease in Cp protein expression in the hippocampus of rats. Cp-positive cells were colocalized with neurons or reactive astrocytes in KA-treated rats and epileptic patient samples. KA-induced seizures, initial oxidative stress (i.e., hydroxyl radical formation, lipid peroxidation, protein oxidation, and synaptosomal reactive oxygen species), altered iron status (increasing Fe(2+) accumulation and L-ferritin-positive reactive microglial cells and decreasing H-ferritin-positive neurons), and impaired glutathione homeostasis and neurodegeneration (i.e., Fluoro-Nissl and Fluoro-Jade B staining analyses) were more pronounced in Cp antisense oligonucleotide (ASO)- than in Cp sense oligonucleotide-treated rats. Consistently, Cp ASO facilitated KA-induced lactate dehydrogenase (LDH) release, Fe(2+) accumulation, and glutathione loss in neuron-rich and mixed cultures. However, Cp ASO did not alter KA-induced LDH release or Fe(2+) accumulation in the astroglial culture, but did facilitate impairment in glutathione homeostasis in the same culture. Importantly, treatment with human Cp protein resulted in a significant attenuation against these neurotoxicities induced by Cp ASO. Our results suggest that Cp-mediated neuroprotection occurs via the inhibition of seizure-associated oxidative damage (including impairment in glutathione homeostasis), Fe(2+) accumulation, and alterations in ferritin immunoreactivity. Moreover, interactive modulation between neurons and glia was found to be important for Cp upregulation in the attenuation of epileptic damage in both animals and humans.


Subject(s)
Ceruloplasmin/physiology , Epilepsy/metabolism , Oxidative Stress , Adolescent , Adult , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Case-Control Studies , Epilepsy/chemically induced , Female , Glutathione/metabolism , Humans , Hydroxyl Radical/metabolism , Kainic Acid , Lipid Peroxidation , Male , Middle Aged , Rats, Sprague-Dawley , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...