Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Myung Bok Wie is active.

Publication


Featured researches published by Myung Bok Wie.


European Journal of Neuroscience | 2005

Fucoidan inhibits cellular and neurotoxic effects of β-amyloid (Aβ) in rat cholinergic basal forebrain neurons

Jack H. Jhamandas; Myung Bok Wie; Kim H. Harris; David MacTavish; Satyabrata Kar

The deposition of β‐amyloid protein (Aβ), a 39–43 amino acid peptide, in the brain and a loss of cholinergic neurons in the basal forebrain are pathological hallmarks of Alzheimers disease (AD). Seaweeds consumed in Asia contain Fucoidan, a sulfated polysaccharide. Fucoidan has been known to exhibit various biological actions, such as an anti‐inflammatory and antioxidant action. In this study, using whole‐cell patch clamp recordings we examined the effects of Fucoidan on Aβ‐induced whole‐cell currents in acutely dissociated rat basal forebrain neurons. We further investigated whether Fucoidan is capable of blocking Aβ neurotoxicity in primary neuronal cultures. In dissociated cells, bath application of Aβ25−35 (1u2003µm) caused a reduction of the whole‐cell currents by 16%. Fucoidan, in a dose‐dependent manner, blocks the Aβ25−35 reduction of whole‐cell currents. Exposure of Aβ25−35 (20u2003µm) or Aβ1−42 (20u2003µm) to rat cholinergic basal forebrain cultures for 48u2003h resulted in 40–60% neuronal death, which was significantly decreased by pretreatment of cultures with Fucoidan (0.1–1.0u2003µm). Fucoidan also attenuated Aβ‐induced down‐regulation of phosphorylated protein kinase C. Aβ1−42‐induced generation of reactive oxygen species was blocked by prior exposure of cultures to Fucoidan. Furthermore, Aβ activation of caspases 9 and 3, which are signaling pathways implicated in apoptotic cell death, is blocked by pretreatment of cultures with Fucoidan. These results show that Fucoidan is able to block Aβ‐induced reduction in whole‐cell currents in basal forebrain neurons and has neuroprotective effects against Aβ‐induced neurotoxicity in basal forebrain neuronal cultures.


FEBS Letters | 2008

Radioprotective properties of eckol against ionizing radiation in mice

Eun-Jin Park; Ginnae Ahn; Nam Ho Lee; Jeong Mi Kim; Jin Seok Yun; Jin Won Hyun; You-Jin Jeon; Myung Bok Wie; Young Jae Lee; Jae Woo Park; Youngheun Jee

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 eckols 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.


Neuroscience Letters | 1998

Postischemic hypothermia induced by eugenol protects hippocampal neurons from global ischemia in gerbils

Moo-Ho Won; Jae-Chul Lee; Yung Hi Kim; Dong Keun Song; Hong Won Suh; Yang Seok Oh; Jung Hoon Kim; Tae kyun Shin; Young Jae Lee; Myung Bok Wie

We studied whether eugenol provides neuroprotection against delayed neuronal death in the hippocampal CA1 region following a 5 min occlusion of the common carotid arteries bilaterally under either free-regulating temperature (TF) or maintained temperature (TM, 37 degrees C) conditions in gerbils. Right after occlusion of the carotid arteries, we injected eugenol intraperitoneally at concentrations of either 50, 100, or 200 mg/kg. There was significant preservation of neuronal cells in the CA1 region in the eugenol-treated groups 7 days after the ischemic insult in the TF condition, with respective survival values of 26, 43, and 68%. In the TM condition, however, significant neuroprotection was only seen with eugenol concentrations of 100 and 200 mg/kg (32% and 52%, respectively). When the rectal temperature was maintained at 38 degrees C for 30 min after occlusion of the carotid arteries, no reduction in CA1 damage was observed with any dose of eugenol. These results suggest that eugenol may provide neuroprotection against ischemic damage by its hypothermic action.


The International Journal of Neuropsychopharmacology | 2015

Melatonin Attenuates Memory Impairment Induced by Klotho Gene Deficiency Via Interactive Signaling Between MT2 Receptor, ERK, and Nrf2-Related Antioxidant Potential

Eun-Joo Shin; Yoon Hee Chung; Hoang-Lan Thi Le; Ji Hoon Jeong; Duy-Khanh Dang; Yunsung Nam; Myung Bok Wie; Seung-Yeol Nah; Yo-ichi Nabeshima; Toshitaka Nabeshima; Hyoung-Chun Kim

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.


Neuroscience Letters | 2004

Antioxidant propolis attenuates kainate-induced neurotoxicity via adenosine A1 receptor modulation in the rat

Yong Soo Kwon; Dae Hun Park; Eun Joo Shin; Myung Sang Kwon; Kwang Ho Ko; Won Ki Kim; Jin Hyeong Jhoo; Wang Kee Jhoo; Myung Bok Wie; Bae Dong Jung; Hyoung Chun Kim

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.


Journal of Applied Toxicology | 2015

Ginsenoside Re protects methamphetamine-induced mitochondrial burdens and proapoptosis via genetic inhibition of protein kinase C δ in human neuroblastoma dopaminergic SH-SY5Y cell lines.

Yunsung Nam; Myung Bok Wie; Eun-Joo Shin; Thuy-Ty Lan Nguyen; Seung-Yeol Nah; Sung Kwon Ko; Ji Hoon Jeong; Choon-Gon Jang; Hyoung-Chun Kim

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 Ca2+), 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. Copyright


Neurochemistry International | 2013

Platelet-activating factor receptor knockout mice are protected from MPTP-induced dopaminergic degeneration.

Beom Keun Kim; Eun Joo Shin; Hyoung Chun Kim; Yoon Hee Chung; Duy Khanh Dang; Bae Dong Jung; Dae Hun Park; Myung Bok Wie; Won Ki Kim; Takao Shimizu; Toshitaka Nabeshima; Ji Hoon Jeong

Platelet-activating factor (PAF), a potent mediator of inflammatory and immune responses, plays various roles in neuronal functions. However, little is known about the role of PAF/platelet-activating factor receptor (PAF-R) in Parkinsons disease. Treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) resulted in significant increases in PAF species in the striatum of wild-type mice. These increases paralleled PAF-R gene expression in wild-type mice. Although nuclear factor kappa B (NF-κB) DNA-binding activity was increased significantly in MPTP-treated wild-type mice, this increase was not significant in PAF-R antagonist ginkgolide B (GB)-treated mice or PAF-R knockout (PAF-R(-/-)) mice. Pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor, significantly ameliorated the dopaminergic deficits induced by MPTP in wild-type mice. MPTP treatment significantly increased oxidative damage, the immunoreactivity of ionized calcium binding adaptor molecule 1 (Iba-1)-positive microglial cells, and microglial differentiation of the M1 type in the striatum of wild-type mice. Consistently, PDTC significantly attenuated MPTP-induced behavioral impairments in wild-type mice. However, dopaminergic deficits, oxidative damage, reactive microglial cells, and behavioral impairments induced by MPTP were not significantly observed in GB-treated mice or PAF-R(-/-) mice. PDTC did not significantly alter the attenuations evident in MPTP-treated PAF-R(-/-) mice, indicating that NF-κB is a critical target for neurotoxic modulation of PAF-R. We propose for the first time that PAF/PAF-R can mediate dopaminergic degeneration via an NF-κB-dependent signaling process.


Epilepsy Research | 2007

Repeated intracerebroventricular infusion of nicotine prevents kainate-induced neurotoxicity by activating the α7 nicotinic acetylcholine receptor

Eun-Joo Shin; Jong Seok Chae; Myung-Eun Jung; Guoying Bing; Kwang Ho Ko; Won-Ki Kim; Myung Bok Wie; Mi-Ae Cheon; Seung-Yeol Nah; Hyoung-Chun Kim

We examined whether (-)-nicotine infusion can affect kainic acid (KA)-induced neurotoxicity in rats. Although treatment with a single nicotine infusion (0.5 or 1.0 microg/side, i.c.v.) failed to attenuate KA-induced neurotoxicity, repeated nicotine infusions (1.0 microg/side/day for 10 days) attenuated the seizures, the severe loss of cells in hippocampal regions CA1 and CA3, the increase in activator protein (AP)-1 DNA binding activity, and mortality after KA administration. alpha-Bungarotoxin and mecamylamine blocked the neuroprotective effects of nicotine. These results suggest that repeated nicotine treatment provides alpha7 nicotinic acetylcholine receptor-mediated neuroprotection against KA toxicity.


Progress in Neuro-psychopharmacology & Biological Psychiatry | 2001

BAPTA/AM, an intracellular calcium chelator, induces delayed necrosis by lipoxygenase-mediated free radicals in mouse cortical cultures.

Myung Bok Wie; Jae Young Koh; Moo-Ho Won; Jae-Chul Lee; Tae kyun Shin; Chang Jong Moon; Hyun Joo Ha; Shin Mi Park; Hyoung Chun Kim

1. Disruption of calcium homeostasis during neurodegenerative diseases is known to trigger apoptotic or necrotic death in neuronal cells. Recently, the authors reported that intracellular calcium restriction by NMDA receptor antagonists induces apoptosis in cortical cultures. To evaluate whether further restriction of intracellular free calcium can induce apoptosis or necrosis, we examined the neurotoxic characterization of BAPTA/AM, a permeable free calcium chelator, in mouse cortical cultures. 2. Exposure of mixed (glia and neuron) cortical cultures (DIV 13-16) to 3-10 microM BAPTA/AM (non-toxic concentration for glial cells) for 24-48 hr resulted in delayed and necrotic neuronal death. The necrotic findings included swelling and loss of mitochondria and endoplasmic reticulum (ER) with neuronal membrane rupture 24 hr after treatment with BAPTA/AM. Simultaneously, we observed a few TUNEL-positive cells in the neuronal subpopulation of the same cultures. 3. The neurotoxicity evoked by BAPTA/AM (10 microM) was significantly attenuated by the addition of 0.5 microM cycloheximide (a protein synthesis inhibitor), 10 microM actinomycin D (an RNA transcription inhibitor), a high extracellular potassium concentration (total 15 mM KCl), 100 microM t-ACPD (a metabotrophic agonist), 100 microM alpha-tocopherol (a free radical scavenger), 100 microM deferoxamine (a ferric ion chelator), 100 microM L-NAME (a nitric oxide synthase (NOS) inhibitor), 50 microM DNQX (a non-NMDA receptor blocker), and 3-30 microM esculetin (a lipoxygenase inhibitor). However, 0.3-3 mM ASA (a cyclooxygenase inhibitor), 100 ng/ml nerve growth factor (NGF), 10 microM MK-801 (a NMDA receptor antagonist), 20 microM zVAD-fmk (caspase inhibitor) and 50 U/ml catalase failed to inhibit the injury. 4. However, NGF and catalase blocked the neurotoxicity induced by BAPTA/AM in young neuronal cells (DIV 6). BAPTA/AM (10 microM) did not alter the expression of inducible nitric oxide synthase (iNOS) on glial cells. 5. These results suggest that the feature of neuronal death induced by BAPTA/AM exhibits predominantly delayed necrosis mediated by lipoxygenase-dependent free radicals.


Neuroscience Letters | 1999

Phenidone attenuates oxygen/glucose deprivation-induced neurotoxicity by antioxidant and antiapoptotic action in mouse cortical cultures.

Myung Bok Wie; Yong Joon Cho; Wang Kee Jhoo; Hyoung Chun Kim

The abrupt elevation in the levels of cyclooxygenase or lipoxygenase metabolites of arachidonic acid during cerebral ischemia contributes to neuronal injury. Recently, evidence has accumulated that both excitotoxic and apoptotic features can coexist in ischemia models in vitro and in vivo. In this study, we evaluated whether phenidone, an inhibitor of both cyclooxygenase and lipoxygenase, can provide protection against excitotoxin- or ischemia-induced neurotoxicity, including the staurosporine apoptosis model, in mouse cortical cultures. We examined the protective effect of phenidone against free radical injuries induced by arachidonic acid, hydrogen peroxide, xanthine/xanthine oxidase, Fe2+/ascorbic acid. Pre- and post-treatment with phenidone (300 microM for 24 h) moderately attenuated the neuronal injury induced by 50 microM kainate and oxygen/glucose deprivation (45 min) by 33% and 50%, respectively. It had no effect on NMDA induced injury (150 microM for 5 min). The maximum dose of phenidone (300 microM) reduced the oxidative injury induced by arachidonic acid (71% inhibition), hydrogen peroxide (95% inhibition), xanthine/xanthine oxidase (57% inhibition), and Fe2+/ascorbic acid (99% inhibition) neurotoxicity. Phenidone (300 microM) decreased staurosporine (100 nM)-induced apoptosis to 30%. These results suggest that phenidone may contribute to neuronal survival by modulating oxidative stress, which is involved in the excitotoxic and apoptotic processes occurring under ischemic conditions.

Collaboration


Dive into the Myung Bok Wie's collaboration.

Top Co-Authors

Avatar

Hyoung Chun Kim

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar

Eun-Joo Shin

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar

Hyoung-Chun Kim

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kwang Ho Ko

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Taekyun Shin

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Bae Dong Jung

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar

Changjong Moon

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Eun Joo Shin

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge