Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Molecules ; 26(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546250

ABSTRACT

As abnormal angiogenesis is associated with exacerbation of various diseases, precise control over angiogenesis is imperative. Vascular endothelial growth factor (VEGF), the most well-known angiogenic factor, binds to VEGF receptor (VEGFR), activates various signaling pathways, and mediates angiogenesis. Therefore, blocking the VEGF-induced angiogenic response-related signaling pathways may alleviate various disease symptoms through inhibition of angiogenesis. Ulmus davidiana is a safe natural product that has been traditionally consumed, but its effects on endothelial cells (ECs) and the underlying mechanism of action are unclear. In the present study, we focused on the effect of a 60% edible ethanolic extract of U. davidiana (U60E) on angiogenesis. U60E inhibited the VEGF-mediated proliferation, tube formation, and migration ability of ECs. Mechanistically, U60E inhibited endothelial nitric oxide synthase activation and nitric oxide production by blocking the protein kinase B signaling pathway activated by VEGF and consequently inhibiting proliferation, tube formation, and migration of ECs. These results suggest that U60E could be a potential and safe therapeutic agent capable of suppressing proangiogenic diseases by inhibiting VEGF-induced angiogenesis.


Subject(s)
Angiogenesis Inhibitors , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Pathologic/drug therapy , Plant Extracts , Ulmus/chemistry , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Ethanol/chemistry , Human Umbilical Vein Endothelial Cells/pathology , Humans , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
2.
Neurochem Res ; 45(12): 2991-3002, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33064252

ABSTRACT

A growing body evidence suggests that selenium (Se) deficiency is associated with an increased risk of developing Alzheimer's disease (AD). Se-dependent glutathione peroxidase-1 (GPx-1) of a major antioxidant enzyme, and the most abundant isoform of GPx in the brain. In the present study, we investigated whether GPx-1 is protective against memory impairments induced by beta-amyloid (Aß) (1-42) in mice. As the alteration of protein kinase C (PKC)-mediated ERK activation was recognized in the early stage of AD, we examined whether the GPx-1 gene modulates Aß (1-42)-induced changes in PKC and ERK levels. We observed that Aß (1-42) treatment (400 pmol, i.c.v.) significantly decreased PKC ßII expression in the hippocampus of mice. Aß (1-42)-induced neurotoxic changes [i.e., oxidative stress (i.e., reactive oxygen species, 4-hydroxy-2-noneal, and protein carbonyl), reduced PKC ßII and phospho-ERK expressions, and memory impairment under Y-maze and passive avoidance test] were more pronounced in GPx-1 knockout than in wild type mice. Importantly, exposure to a GPx-1 gene-encoded adenovirus vector (Adv-GPx-1) significantly increased GPx-1 mRNA and GPx activity in the hippocampus of GPx-1 knockout mice. Adv-GPx-1 exposure also significantly blocked the neurotoxic changes induced by Aß (1-42) in GPx-1 knockout mice. Treatment with ERK inhibitor U0126 did not significantly change Adv-GPx-1-mediated attenuation in PKC ßII expression. In contrast, treatment with PKC inhibitor chelerythrine (CHE) reversed Adv-GPx-1-mediated attenuation in ERK phosphorylation, suggesting that PKC ßII-mediated ERK signaling is important for Adv-GPx-1-mediated potentials against Aß (1-42) insult. Our results suggest that treatment with the antioxidant gene GPx-1 rescues Aß (1-42)-induced memory impairment via activating PKC ßII-mediated ERK signaling.


Subject(s)
Glutathione Peroxidase/deficiency , Glutathione Peroxidase/pharmacology , MAP Kinase Signaling System/drug effects , Memory Disorders/enzymology , Memory/drug effects , Protein Kinase C beta/metabolism , Adenoviridae/genetics , Amyloid beta-Peptides , Animals , Gene Expression/drug effects , Genetic Therapy , Glutathione Peroxidase/genetics , Hippocampus/enzymology , Hippocampus/metabolism , Male , Memory Disorders/chemically induced , Memory Disorders/genetics , Memory Disorders/therapy , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments , Glutathione Peroxidase GPX1
3.
J Pharmacol Sci ; 132(2): 166-170, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27802908

ABSTRACT

Dextromethorphan (DM) administered at supra-antitussive doses produce psychotoxic and neurotoxic effects in humans. We administered DM (80 mg/kg) to rats intraperitoneally to determine the ultrastructural change induced by DM, because intraperitoneal route is sensitive for the behavioral responses. Treatment with DM resulted in mitochondrial dysfunction and formation of myelinoid bodies in the hippocampus. MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate] attenuated DM-induced cytosolic oxidative burdens. However, neither MK-801 nor naloxone affected DM-induced mitochondrial dysfunction and formation of myelinoid bodies, indicating that the neurotoxic mechanism needs to be further elucidated. Therefore, the spectrum of toxicological effects associated with DM need to be reassessed.


Subject(s)
Antitussive Agents/toxicity , Dextromethorphan/toxicity , Hippocampus/drug effects , Hippocampus/ultrastructure , Myelin Sheath/ultrastructure , Animals , Antitussive Agents/administration & dosage , Cytosol/drug effects , Cytosol/pathology , Cytosol/ultrastructure , Dextromethorphan/administration & dosage , Hippocampus/pathology , Injections, Intraperitoneal , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/ultrastructure , Myelin Sheath/pathology , Rats
4.
Arch Toxicol ; 90(4): 937-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25895139

ABSTRACT

We investigated whether protein kinase C (PKC) is involved in trimethyltin (TMT)-induced neurotoxicity. TMT treatment (2.8 mg/kg, i.p.) significantly increased PKCδ expression out of PKC isozymes (i.e., α, ßI, ßII, δ, and ς) in the hippocampus of wild-type (WT) mice. Consistently, treatment with TMT resulted in significant increases in cleaved PKCδ expression. Genetic or pharmacological inhibition (PKCδ knockout or rottlerin) was less susceptible to TMT-induced seizures than WT mice. TMT treatment increased glutathione oxidation, lipid peroxidation, protein oxidation, and levels of reactive oxygen species. These effects were more pronounced in the WT mice than in PKCδ knockout mice. In addition, the ability of TMT to induce nuclear translocation of Nrf2, Nrf2 DNA-binding activity, and upregulation of γ-glutamylcysteine ligase was significantly increased in the PKCδ knockout mice and rottlerin (10 or 20 mg/kg, p.o. × 6)-treated WT mice. Furthermore, neuronal degeneration (as shown by nuclear chromatin clumping and TUNEL staining) in WT mice was most pronounced 2 days after TMT. At the same time, TMT-induced inhibition of phosphoinositol 3-kinase (PI3K)/Akt signaling was evident, thereby decreasing phospho-Bad, expression of Bcl-xL and Bcl-2, and the interaction between phospho-Bad and 14-3-3 protein, and increasing Bax expression and caspase-3 cleavage were observed. Rottlerin or PKCδ knockout significantly protected these changes in anti- and pro-apoptotic factors. Importantly, treatment of the PI3K inhibitor LY294002 (0.8 or 1.6 µg, i.c.v.) 4 h before TMT counteracted protective effects (i.e., Nrf-2-dependent glutathione induction and pro-survival phenomenon) of rottlerin. Therefore, our results suggest that down-regulation of PKCδ and up-regulations of Nrf2-dependent glutathione defense mechanism and PI3K/Akt signaling are critical for attenuating TMT neurotoxicity.


Subject(s)
Glutathione/metabolism , Hippocampus/drug effects , Neurotoxicity Syndromes/etiology , Protein Kinase C-delta/metabolism , Trimethyltin Compounds/toxicity , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Dose-Response Relationship, Drug , Hippocampus/metabolism , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Knockout , Neurotoxicity Syndromes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase Inhibitors/pharmacology , Seizures/chemically induced , Seizures/drug therapy
5.
J Appl Toxicol ; 35(8): 927-44, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25523949

ABSTRACT

Recently, we have demonstrated that ginsenoside Re protects methamphetamine (MA)-induced dopaminergic toxicity in mice via genetic inhibition of PKCδ and attenuation of mitochondrial stress. In addition, we have reported that induction of mitochondrial glutathione peroxidase (GPx) is also important for neuroprotection mediated by ginsenoside Re. To extend our knowledge, we examined the effects of ginsenoside Re against MA toxicity in vitro condition using SH-SY5Y neuroblastoma cells. Treatment with ginsenoside Re resulted in significant attenuations against a decrease in the activity of GPx and an increase in the activity of superoxide dismutase (SOD) in the cytosolic and mitochondrial fraction. The changes in glutathione (GSH) paralleled those in GPx in the same experimental condition. Consistently, ginsenoside Re treatment exhibited significant protections against cytosolic and mitochondrial oxidative damage (i.e. lipid peroxidation and protein oxidation), mitochondrial translocation of PKCδ, mitochondrial dysfunction (mitochondrial transmembrane potential and intra-mitochondrial Ca(2+)), apoptotic events [i.e., cytochrome c release from mitochondria, cleavage of caspase-3 and poly(ADP-ribose)polymerase-1, nuclear condensation, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive apoptotic cells], and a reduction in the tyrosine hydroxylase (TH) expression and TH activity induced by MA in SH-SY5Y neuroblastoma cells. These protective effects of ginsenoside Re were comparable to those of PKCδ antisense oligonucleotide (ASO). However, ginsenoside Re did not significantly provide additional protective effects mediated by genetic inhibition of PKCδ. Our results suggest that PKCδ is a specific target for ginsenoside Re-mediated protective activity against MA toxicity in SH-SY5Y neuroblastoma cells.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/toxicity , Ginsenosides/pharmacology , Methamphetamine/antagonists & inhibitors , Methamphetamine/toxicity , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Protein Kinase C-delta/genetics , Antioxidants/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytosol/drug effects , Dopaminergic Neurons/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Tyrosine 3-Monooxygenase/metabolism
6.
Int J Neuropsychopharmacol ; 18(6)2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25550330

ABSTRACT

BACKGROUND: We demonstrated that oxidative stress plays a crucial role in cognitive impairment in klotho mutant mice, a genetic model of aging. Since down-regulation of melatonin due to aging is well documented, we used this genetic model to determine whether the antioxidant property of melatonin affects memory impairment. METHODS: First, we examined the effects of melatonin on hippocampal oxidative parameters and the glutathione/oxidized glutathione (GSH/GSSG) ratio and memory dysfunction of klotho mutant mice. Second, we investigated whether a specific melatonin receptor is involved in the melatonin-mediated pharmacological response by application with melatonin receptor antagonists. Third, we examined phospho-extracellular-signal-regulated kinase (ERK) expression, nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, Nrf2 DNA binding activity, and glutamate-cysteine ligase (GCL) mRNA expression. Finally, we examined effects of the ERK inhibitor SL327 in response to antioxidant efficacy and memory enhancement mediated by melatonin. RESULTS: Treatment with melatonin resulted in significant attenuations of oxidative damage, a decrease in the GSH/GSSG ratio, and a significant amelioration of memory impairment in this aging model. These effects of melatonin were significantly counteracted by the selective MT2 receptor antagonist 4-P-PDOT. Importantly, 4-P-PDOT or SL327 also counteracted melatonin-mediated attenuation in response to the decreases in phospho-ERK expression, Nrf2 nuclear translocation, Nrf2 DNA-binding activity, and GCL mRNA expression in the hippocampi of klotho mutant mice. SL327 also counteracted the up-regulation of the GSH/GSSG ratio and the memory enhancement mediated by melatonin in klotho mutant mice. CONCLUSIONS: Melatonin attenuates oxidative stress and the associated memory impairment induced by klotho deficiency via signaling interaction between the MT2 receptor and ERK- and Nrf2-related antioxidant potential.


Subject(s)
Antioxidants/pharmacology , Behavior, Animal/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucuronidase/deficiency , Hippocampus/drug effects , Melatonin/pharmacology , Memory Disorders/prevention & control , Memory/drug effects , NF-E2-Related Factor 2/metabolism , Nootropic Agents/pharmacology , Oxidative Stress/drug effects , Receptor, Melatonin, MT2/agonists , Signal Transduction/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Glucuronidase/genetics , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Klotho Proteins , Memory Disorders/enzymology , Memory Disorders/genetics , Memory Disorders/physiopathology , Memory Disorders/psychology , Mice, Inbred C3H , Mice, Knockout , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Receptor, Melatonin, MT2/antagonists & inhibitors , Receptor, Melatonin, MT2/metabolism
7.
Front Endocrinol (Lausanne) ; 14: 1138676, 2023.
Article in English | MEDLINE | ID: mdl-37234799

ABSTRACT

Diabetic retinopathy (DR) is a disease that causes visual deficiency owing to vascular leakage or abnormal angiogenesis. Pericyte apoptosis is considered one of the main causes of vascular leakage in diabetic retina, but there are few known therapeutic agents that prevent it. Ulmus davidiana is a safe natural product that has been used in traditional medicine and is attracting attention as a potential treatment for various diseases, but its effect on pericyte loss or vascular leakage in DR is not known at all. In the present study, we investigated on the effects of 60% edible ethanolic extract of U. davidiana (U60E) and catechin 7-O-ß-D-apiofuranoside (C7A), a compound of U. davidiana, on pericyte survival and endothelial permeability. U60E and C7A prevented pericyte apoptosis by inhibiting the activation of p38 and JNK induced by increased glucose and tumor necrosis factor alpha (TNF-α) levels in diabetic retina. Moreover, U60E and C7A reduced endothelial permeability by preventing pericyte apoptosis in co-cultures of pericytes and endothelial cells. These results suggest that U60E and C7A could be a potential therapeutic agent for reducing vascular leakage by preventing pericyte apoptosis in DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Ulmus , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/pathology , Pericytes , Endothelial Cells/pathology , Apoptosis , Diabetes Mellitus/pathology
8.
J Ginseng Res ; 47(4): 561-571, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37397414

ABSTRACT

Background: Escalating evidence shows that ginseng possesses an antiaging potential with cognitive enhancing activity. As mountain cultivated ginseng (MCG) is cultivated without agricultural chemicals, MCG has emerged as a popular herb medicine. However, little is known about the MCG-mediated pharmacological mechanism on brain aging. Methods: As we demonstrated that glutathione peroxidase (GPx) is important for enhancing memory function in the animal model of aging, we investigated the role of MCG as a GPx inducer using GPx-1 (a major type of GPx) knockout (KO) mice. We assessed whether MCG modulates redox and cholinergic parameters, and memory function in aged GPx-1 knockout KOmice. Results: Redox burden of aged GPx-1 KO mice was more evident than that of aged wild-type (WT) mice. Alteration of Nrf2 DNA binding activity appeared to be more evident than that of NFκB DNA binding activity in aged GPx-1 KO mice. Alteration in choline acetyltransferase (ChAT) activity was more evident than that in acetylcholine esterase activity. MCG significantly attenuated reductions in Nrf2 system and ChAT level. MCG significantly enhanced the co-localization of Nrf2-immunoreactivity and ChAT-immunoreactivity in the same cell population. Nrf2 inhibitor brusatol significantly counteracted MCG-mediated up-regulation in ChAT level and ChAT inhibition (by k252a) significantly reduced ERK phosphorylation by MCG, suggesting that MCG might require signal cascade of Nrf2/ChAT/ERK to enhance cognition. Conclusion: GPx-1 depletion might be a prerequisite for cognitive impairment in aged animals. MCG-mediated cognition enhancement might be associated with the activations of Nrf2, ChAT, and ERK signaling cascade.

9.
Toxics ; 10(4)2022 Apr 16.
Article in English | MEDLINE | ID: mdl-35448455

ABSTRACT

We examined the protective effects of esculetin and fucoidan against the neurotoxicity of ZnO NPs in rats. Ninety rats were divided into nine groups and pre-treated with esculetin or fucoidan 1 h before ZnO NP administration on a daily basis for 2 weeks. Serum and brain homogenates were examined by enzyme-linked immunosorbent assay (ELISA), and neurons, microglia, and astrocytes in the hippocampal region were examined with immunohistochemical analysis. The serum levels of interleukin-1-beta (IL-1ß), 3-nitrotyrosine (3-NT), superoxide dismutase (SOD), and 8-hydroxy-2'-deoxyguanosine (8-OHdG) were altered in the ZnO NP treatment groups. Brain IL-1ß and TNF-α levels were elevated after ZnO NP administration, and these effects were inhibited by esculetin and fucoidan. SOD, 8-OHdG, and acetylcholinesterase (AChE) levels in the brain were decreased after ZnO NP administration. The brain levels of beclin-1 and caspase-3 were elevated after ZnO NP treatment, and these effects were significantly ameliorated by esculetin and fucoidan. The number of reactive astrocytes measured by counting glial fibrillary acidic protein (GFAP)-positive cells, but not microglia, increased following ZnO NP treatment, and esculetin and fucoidan ameliorated the changes. Esculetin and fucoidan may be beneficial for preventing ZnO NP-mediated autophagy and apoptosis by the modulation of reactive astrocyte and proinflammatory cytokines in the rat brain.

10.
Brain Sci ; 11(9)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34573138

ABSTRACT

Trimethyltin (TMT) is an environmental neurotoxin that mediates dopaminergic neuronal injury in the brain. In this study, we characterized the toxic mechanism and possible protective compounds against TMT-induced neurotoxicity in human dopaminergic neuroblastoma SH-SY5Y cells. Antioxidants such as melatonin, N-acetylcysteine (NAC), α-tocopherol, and allopurinol alleviated TMT toxicity. Apoptosis induced by TMT was identified by altered expression of cleaved caspase-3, Bax, Bcl-2, and Bcl-xL through Western blot analysis. The iron chelator deferoxamine ameliorated the alteration of apoptosis-related proteins through TMT exposure. TMT also induced delayed ultrastructural necrotic features such as mitochondrial swelling and cytoplasmic membrane rupture; NAC reduced these necrotic injuries. Esculetin, meloxicam, celecoxib, and phenidone decreased TMT toxicity. Elevation of the pro-inflammatory cytokines IL-1ß, TNF-α, and NF-ĸB and reduction of the antioxidant enzymes catalase and glutathione peroxidase-1 (GPx-1) were induced by TMT and ameliorated by inhibitors of LOX and COX-2 enzymes. Both NMDA and non-NMDA antagonists attenuated TMT toxicity. The free calcium ion modulators nimodipine and BAPTA/AM contributed to neuronal survival against TMT toxicity. Inhibitors of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin pathway, an autophagy regulator, decreased TMT toxicity. These results imply that TMT neurotoxicity is the chief participant in LOX- and COX-2-mediated apoptosis, partly via necrosis and autophagy in SH-SY5Y cells.

11.
Phytother Res ; 24(3): 399-403, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19655293

ABSTRACT

We examined whether fucoidan affected the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in rats. EAE was induced in Lewis rats that were immunized with guinea-pig myelin basic protein (MBP) and complete Freund's adjuvant. Fucoidan (50 mg/kg, daily) was administered to rats with EAE intraperitoneally, either in the EAE induction phase from either 1 day before immunization to day 7 post-immunization (PI), or the effector phase from day 8 to 14 PI, to test which phase of rat EAE is affected by fucoidan treatment.The onset, severity and duration of EAE paralysis in the fucoidan-treated group in the days 8-14 PI-treated rats, but not in days -1-7 PI-treated rats, were significantly delayed, suppressed and reduced, respectively, compared with the vehicle-treated controls. Treatment with fucoidan reduced the encephalitogenic response and TNF-alpha production during EAE. Moreover, the clinical amelioration coincided with decreased infiltration of inflammatory cells in the EAE-affected spinal cord. The ameliorative effect of fucoidan on clinical paralysis in EAE-affected rats may be mediated, in part, by the suppression of the autoreactive T cell response and inflammatory cytokine production.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Polysaccharides/therapeutic use , Animals , Female , Freund's Adjuvant , Lymphocyte Activation/drug effects , Male , Myelin Basic Protein , Rats , Rats, Inbred Lew , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
J Neurosci Res ; 87(3): 710-22, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18816793

ABSTRACT

We previously demonstrated that kainic acid (KA)-mediated mitochondrial oxidative stress contributed to hippocampal degeneration and that ginsenosides attenuated KA-induced neurotoxicity and neuronal degeneration. Here, we examined whether ginsenosides affected KA-induced mitochondrial dysfunction and oxidative stress in the rat hippocampus. Treatment with ginsenosides attenuated KA-induced convulsive behavior dose-dependently. KA treatment increased lipid peroxidation and protein oxidation and decreased the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio to a greater degree in the mitochondrial fraction than in the hippocampal homogenate. KA treatment resulted in decreased Mn-superoxide dismutase expression and diminished the mitochondrial membrane potential. Furthermore, KA treatment increased intramitochondrial Ca(2+) and promoted ultrastructural degeneration in hippocampal mitochondria. Treatment with ginsenosides dose-dependently attenuated convulsive behavior and the KA-induced mitochondrial effects. Protection appeared to be more evident in mitochondria than in tissue homogenates. Collectively, the results suggest that ginsenosides prevent KA-induced neurotoxicity by attenuating mitochondrial oxidative stress and mitochondrial dysfunction.


Subject(s)
Ginsenosides/therapeutic use , Hippocampus/drug effects , Kainic Acid/toxicity , Mitochondria/drug effects , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Seizures/drug therapy , Animals , Anticonvulsants/therapeutic use , Antioxidants/therapeutic use , Calcium/metabolism , Glutathione/metabolism , Hippocampus/physiology , Hippocampus/ultrastructure , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Motor Activity/drug effects , Nerve Degeneration/drug therapy , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/pathology , Seizures/physiopathology , Superoxide Dismutase/metabolism
13.
Cell Signal ; 20(4): 645-58, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18248956

ABSTRACT

We have demonstrated that kainate (KA) induces a reduction in mitochondrial Mn-superoxide dismutase (Mn-SOD) expression in the rat hippocampus and that KA-induced oxidative damage is more prominent in senile-prone (SAM-P8) than senile-resistant (SAM-R1) mice. To extend this, we examined whether KA seizure sensitivity contributed to mitochondrial degeneration in these mouse strains. KA-induced seizure susceptibility in SAM-P8 mice paralleled prominent increases in lipid peroxidation and protein oxidation and was accompanied by significant impairment in glutathione homeostasis in the hippocampus. These findings were more pronounced in the mitochondrial fraction than in the hippocampal homogenate. Consistently, KA-induced decreases in Mn-SOD protein expression, mitochondrial transmembrane potential, and uncoupling protein (UCP)-2 expression were more prominent in SAM-P8 than SAM-R1 mice. Marked release of cytochrome c from mitochondria into the cytosol and a higher level of caspase-3 cleavage were observed in KA-treated SAM-P8 mice. Additionally, electron microscopic evaluation indicated that KA-induced increases in mitochondrial damage and lipofuscin-like substances were more pronounced in SAM-P8 than SAM-R1 animals. These results suggest that KA-mediated mitochondrial oxidative stress contributed to hippocampal degeneration in the senile-prone mouse.


Subject(s)
Aging, Premature/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Oxidative Stress , Aging, Premature/genetics , Aging, Premature/pathology , Animals , Caspase 3/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Enzyme Activation , Glutathione/metabolism , Glutathione Disulfide/metabolism , Hippocampus/enzymology , Hippocampus/ultrastructure , Ion Channels/metabolism , Kainic Acid , Lipid Peroxidation/drug effects , Lipofuscin/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/enzymology , Neurons/ultrastructure , Oxidation-Reduction , Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Seizures/chemically induced , Seizures/genetics , Seizures/metabolism , Seizures/pathology , Superoxide Dismutase/metabolism , Time Factors , Uncoupling Protein 2
14.
Anat Cell Biol ; 52(4): 369-377, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31949974

ABSTRACT

Citrus species contain significant amounts of flavonoids that possess antioxidant activities; furthermore, dietary citrus is not associated with adverse effects or cytotoxicity in healthy individuals. Hesperidin, which is an abundant flavanone glycoside in the peel of citrus fruits, possesses a variety of biological capabilities that include antioxidant and anti-inflammatory actions. Over the last few decades, many studies have been investigated the biological actions of hesperidin and its aglycone, hesperetin, as well as their underlying mechanisms. Due to the antioxidant effects of hesperidin and its derivatives, the cardioprotective and anti-cancer effects of these compounds have been widely reviewed. Although the biological activities of hesperidin in neurodegenerative diseases have been evaluated, its potential involvement in a variety of central nervous system (CNS) disorders, including autoimmune demyelinating disease, requires further investigation in terms of the underlying mechanisms. Thus, the present review will focus on the potential role of hesperidin in diverse models of CNS neuroinflammation, including experimental autoimmune encephalomyelitis, with special consideration given to its antioxidant and anti-inflammatory effects in neurodegenerative disease models. Additionally, current evidence provides information regarding the nutraceutical use of hesperidin to prevent various CNS disorders.

15.
Nanomaterials (Basel) ; 9(7)2019 Jul 21.
Article in English | MEDLINE | ID: mdl-31330912

ABSTRACT

The present study examined the potential toxic concentrations of zinc oxide nanoparticles (ZnO NPs) and associated autophagy and apoptosis-related injuries in primary neocortical astrocyte cultures. Concentrations of ZnO NPs ≥3 µg/mL induced significant toxicity in the astrocytes. At 24 h after exposure to the ZnO NPs, transmission electron microscopy revealed swelling of the endoplasmic reticulum (ER) and increased numbers of autophagolysosomes in the cultured astrocytes, and increased levels of LC3 (microtubule-associated protein 1 light chain 3)-mediated autophagy were identified by flow cytometry. Apoptosis induced by ZnO NP exposure was confirmed by the elevation of caspase-3/7 activity and 4',6'-diamidino-2-phenylindole (DAPI) staining. Significant (p < 0.05) changes in the levels of glutathione peroxidase, superoxide dismutase, tumor necrosis factor (TNF-α), and interleukin-6 were observed by enzyme-linked immunoassay (ELISA) assay following the exposure of astrocyte cultures to ZnO NPs. Phosphatidylinositol 3-kinase (PI3K)/mitogen-activated protein kinase (MAPK) dual activation was induced by ZnO NPs in a dose-dependent manner. Additionally, the Akt (protein kinase B) inhibitor BML257 and the mTOR (mammalian target of rapamycin) inhibitor rapamycin contributed to the survival of astrocytes. Inhibitors of cyclooxygenase-2 and lipoxygenase attenuated ZnO NP-induced toxicity. Calcium-modulating compounds, antioxidants, and zinc/iron chelators also decreased ZnO NP-induced toxicity. Together, these results suggest that ZnO NP-induced autophagy and apoptosis may be associated with oxidative stress and the inflammatory process in primary astrocyte cultures.

16.
Toxicol Res ; 35(1): 83-91, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30766660

ABSTRACT

Nanoparticles (NPs) have been recognized as both useful tools and potentially toxic materials in various industrial and medicinal fields. Previously, we found that zinc oxide (ZnO) NPs that are neurotoxic to human dopaminergic neuroblastoma SH-SY5Y cells are mediated by lipoxygenase (LOX), not cyclooxygenase-2 (COX-2). Here, we examined whether human bone marrow-derived mesenchymal stem cells (MSCs), which are different from neuroblastoma cells, might exhibit COX-2- and/or LOX-dependent cytotoxicity of ZnO NPs. Additionally, changes in annexin V expression, caspase-3/7 activity, and mitochondrial membrane potential (MMP) induced by ZnO NPs and ZnO were compared at 12 hr and 24 hr after exposure using flow cytometry. Cytotoxicity was measured based on lactate dehydrogenase activity and confirmed by trypan blue staining. Rescue studies were executed using zinc or iron chelators. ZnO NPs and ZnO showed similar dose-dependent and significant cytotoxic effects at concentrations ≥ 15 µg/mL, in accordance with annexin V expression, caspase-3/7 activity, and MMP results. Human MSCs exhibited both COX-2 and LOX-mediated cytotoxicity after exposure to ZnO NPs, which was different from human neuroblastoma cells. Zinc and iron chelators significantly attenuated ZnO NPs-induced toxicity. Conclusively, these results suggest that ZnO NPs exhibit both COX-2- and LOX-mediated apoptosis by the participation of mitochondrial dysfunction in human MSC cultures.

17.
FEBS Lett ; 582(6): 925-30, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18294966

ABSTRACT

We have investigated the radioprotective efficacy of eckol, a component of brown seaweed Ecklonia cava, against the gamma ray-induced damage in vivo. Our results showed that eckol significantly decreased the mortality of lethally irradiated mice. The mechanisms of eckol's protection were found to include: an improvement in hematopoietic recovery, the repair of damaged DNA in immune cells and an enhancement of their proliferation, which had been severely suppressed by ionizing radiation. Thus, we propose eckol as a candidate for adjuvant therapy to alleviate radiation-induced injuries to cancer patients.


Subject(s)
DNA Damage/drug effects , Dioxins/pharmacology , Hematopoiesis/drug effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/radiation effects , Cell Proliferation/drug effects , Dioxins/chemistry , Dioxins/therapeutic use , Gamma Rays , Hematopoiesis/radiation effects , Mice , Mice, Inbred Strains , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/therapeutic use , Spleen/drug effects , Spleen/radiation effects , T-Lymphocytes/drug effects , T-Lymphocytes/radiation effects
18.
Neurochem Int ; 52(6): 1134-47, 2008 May.
Article in English | MEDLINE | ID: mdl-18226427

ABSTRACT

Oxidative stress may contribute to epileptogenicity in genetic models of epilepsy. To address this, we examined the enzymatic activity of cytosolic Cu/Zn superoxide dismutase (SOD-1), mitochondrial Mn superoxide dismutase (SOD-2), and glutathione peroxidase (GPx) in the developing hippocampus of genetically epilepsy-prone rats (GEPR-9s). We also measured changes in the GSH/GSSG ratio, lipid peroxidation, and protein oxidation at post-natal days (PD) 7, 30, and 90, respectively. Compared with control Sprague-Dawley (SD) rats, GEPR-9s showed similar SOD-1 and SOD-2 activity but lower GPx activity. Epilepsy-prone rats also showed lower GSH/GSSG ratios than controls, and more lipid peroxidation (as measured by malondialdehyde levels) and protein oxidation (as measured by carbonyl levels). Treatment with kainic acid (KA) resulted in more pronounced seizures, less GPx activity, and lower GSH/GSSG ratios in GEPR-9s than in controls, but KA did not significantly affect SOD-1 or SOD-2 activity, suggesting that GEPR-9s do not compensate for reduced GPx activity by increasing SOD. Moreover, KA treatment resulted in significantly a lower GSH/GSSG ratio and GPx-like immunoreactivity and higher malondialdehyde and carbonyl levels in GEPR-9s than in controls. These findings were more evident in GEPR-9s at PD 90 than at PD 30, indicating that oxidative stress is age-dependent. Double-labeling immunocytochemical analysis demonstrated co-localization of GPx-immunoreactive glia-like cells and reactive astrocytes, as labeled by glial fibrillary acidic protein (GFAP). This suggests that mobilization of astroglial cells for synthesis of GPx protein is a response to KA insult, intended to decrease the neurotoxicity induced by peroxides. These responses were more pronounced in control SD rats than in GEPR-9s. Our results suggest that impairment of the GPx (including glutathione)-mediated antioxidant system contributed to epileptogenesis in GEPR-9s.


Subject(s)
Epilepsy/enzymology , Glutathione Peroxidase/metabolism , Hippocampus/enzymology , Oxidative Stress/genetics , Aging/metabolism , Animals , Antioxidants/metabolism , Drug Resistance/drug effects , Drug Resistance/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Epilepsy/chemically induced , Epilepsy/genetics , Excitatory Amino Acid Agonists/toxicity , Genetic Predisposition to Disease/genetics , Glutathione/metabolism , Glutathione Peroxidase/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Kainic Acid/toxicity , Lipid Peroxidation/drug effects , Lipid Peroxidation/genetics , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neuroglia/metabolism , Neurotoxins/toxicity , Oxidative Stress/drug effects , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
19.
Neurosci Lett ; 445(1): 1-6, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18760329

ABSTRACT

Anti-inflammatory drugs such as ibuprofen appear to prevent the development of Parkinson's disease (PD); however, long-term use has undesirable side-effects. A new strategy for anti-inflammatory drug therapy is using a dual inhibitor of COX and lipooxygenase (LOX). Here, we compared the dopaminergic neuroprotective property of phenidone (a dual COX and LOX inhibitor) with COX or LOX inhibitors including SC-560 (a COX-1 inhibitor), aspirin (a COX-1/2 inhibitor), meloxicam (a preferential COX-2 inhibitor), caffeic acid (a 5-LOX inhibitor), and esculetin (a 5, 12-LOX inhibitor) in our lipopolysaccharide (LPS)-induced PD animal model. Our results show that COX-2 and 5-LOX play a major role in LPS-induced dopaminergic neurotoxicity, as meloxicam and phenidone attenuated LPS-induced oxidative stress and meloxicam, phenidone, and caffeic acid attenuated dopaminergic neurodegeneration, while SC-560, aspirin, and esculetin did not. In addition, phenidone was superior in attenuating LPS-induced dopaminergic neurodegeneration and microglia activation, probably as a result of dual inhibition of COX-2 and LOX. Therefore, dual inhibition of COX and LOX with phenidone represents a promising new candidate for anti-inflammatory drug therapy, and may provide a novel therapeutic approach for inflammation-related neurodegenerative diseases including PD.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Dopamine/metabolism , Neurons/drug effects , Neurotoxicity Syndromes , Pyrazoles/pharmacology , Substantia Nigra/pathology , Analysis of Variance , Animals , CD11b Antigen/metabolism , Disease Models, Animal , Drug Interactions , Lipopolysaccharides/toxicity , Male , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/prevention & control , Rats , Rats, Sprague-Dawley
20.
FASEB J ; 20(14): 2496-511, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17142799

ABSTRACT

We investigated the neuroprotective property of analogs of dextromethorphan (DM) in lipopolysaccharide (LPS) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) models to identify neuroprotective drugs for Parkinson's disease (PD). In vivo studies showed that daily injections with DM analogs protected dopamine (DA) neurons in substantia nigra pars compacta and restored DA levels in striatum using two different models for PD. Of the five analogs studied, 3-hydroxymorphinan (3-HM), a metabolite of DM, was the most potent, and restored DA neuronal loss and DA depletion up to 90% of the controls. Behavioral studies showed an excellent correlation between potency for preventing toxin-induced decrease in motor activities and neuroprotective effects among the DM analogs studied, of which 3-HM was the most potent in attenuating behavioral damage. In vitro studies revealed two glia-dependent mechanisms for the neuroprotection by 3-HM. First, astroglia mediated the 3-HM-induced neurotrophic effect by increasing the gene expression of neurotrophic factors, which was associated with the increased acetylation of histone H3. Second, microglia participated in 3-HM-mediated neuroprotection by reducing MPTP-elicited reactive microgliosis as evidenced by the decreased production of reactive oxygen species. In summary, we show the potent neuroprotection by 3-HM in LPS and MPTP PD models investigated. With its high efficacy and low toxicity, 3-HM may be a novel therapy for PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Dextromethorphan/analogs & derivatives , Dextromethorphan/metabolism , Parkinson Disease/drug therapy , Parkinsonian Disorders/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Behavior, Animal/drug effects , Dextromethorphan/chemistry , Dextromethorphan/therapeutic use , Dopamine/metabolism , Dose-Response Relationship, Drug , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , Molecular Structure , Parkinsonian Disorders/chemically induced , Rats , Reactive Oxygen Species , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
SELECTION OF CITATIONS
SEARCH DETAIL