Network


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

Hotspot


Dive into the research topics where Jin Won Hyun is active.

Publication


Featured researches published by Jin Won Hyun.


Toxicology Letters | 2011

Silver nanoparticles induce oxidative cell damage in human liver cells through inhibition of reduced glutathione and induction of mitochondria-involved apoptosis.

Mei Jing Piao; Kyoung Ah Kang; In Kyung Lee; Hye Sun Kim; Suhkmann Kim; Jeong Yun Choi; Jinhee Choi; Jin Won Hyun

Silver nanoparticles (AgNPs), which have well-known antimicrobial properties, are extensively used in various medical and general applications. Despite the widespread use of AgNPs, relatively few studies have been undertaken to determine the cytotoxic effects of AgNPs exposure. This study investigates possible molecular mechanisms underlying the cytotoxic effects of AgNPs. Here, we show that AgNPs-induced cytotoxicity was higher compared than that observed when AgNO(3) was used as a silver ion source. AgNPs induced reactive oxygen species (ROS) generation and suppression of reduced glutathione (GSH) in human Chang liver cells. ROS generated by AgNPs resulted in damage to various cellular components, DNA breaks, lipid membrane peroxidation, and protein carbonylation. Upon AgNPs exposure, cell viability decreased due to apoptosis, as demonstrated by the formation of apoptotic bodies, sub-G(1) hypodiploid cells, and DNA fragmentation. AgNPs induced a mitochondria-dependent apoptotic pathway via modulation of Bax and Bcl-2 expressions, resulting in the disruption of mitochondrial membrane potential (Δψ(m)). Loss of Δψ(m) was followed by cytochrome c release from the mitochondria, resulting in the activation of caspases 9 and 3. The apoptotic effect of AgNPs was exerted via the activation of c-Jun NH(2)-terminal kinase (JNK) and was abrogated by the JNK-specific inhibitor, SP600125 and siRNA targeting JNK. In summary, the results suggest that AgNPs cause cytotoxicity by oxidative stress-induced apoptosis and damage to cellular components.


FEBS Letters | 2005

Eckol isolated from Ecklonia cava attenuates oxidative stress induced cell damage in lung fibroblast cells

Kyoung Ah Kang; Kyoung Hwa Lee; Sungwook Chae; Rui Zhang; Myung Sun Jung; Youngki Lee; So Young Kim; Hee-Sun Kim; Hong Gu Joo; Jae Woo Park; Young Min Ham; Nam Ho Lee; Jin Won Hyun

We have investigated the cytoprotective effect of eckol, which was isolated from Ecklonia cava, against oxidative stress induced cell damage in Chinese hamster lung fibroblast (V79‐4) cells. Eckol was found to scavenge 1,1‐diphenyl‐2‐picrylhydrazyl radical, hydrogen peroxide (H2O2), hydroxy radical, intracellular reactive oxygen species (ROS), and thus prevented lipid peroxidation. As a result, eckol reduced H2O2 induced cell death in V79‐4 cells. In addition, eckol inhibited cell damage induced by serum starvation and radiation by scavenging ROS. Eckol was found to increase the activity of catalase and its protein expression. Further, molecular mechanistic study revealed that eckol increased phosphorylation of extracellular signal‐regulated kinase and activity of nuclear factor κ B. Taken together, the results suggest that eckol protects V79‐4 cells against oxidative damage by enhancing the cellular antioxidant activity and modulating cellular signal pathway.


Journal of Cellular Biochemistry | 2006

Cytoprotective effect of phloroglucinol on oxidative stress induced cell damage via catalase activation

Kyoung Ah Kang; Kyoung Hwa Lee; Sungwook Chae; Rui Zhang; Myung Sun Jung; Young Min Ham; Jong Seok Baik; Nam Ho Lee; Jin Won Hyun

We investigated the cytoprotective effect of phloroglucinol, which was isolated from Ecklonia cava (brown alga), against oxidative stress induced cell damage in Chinese hamster lung fibroblast (V79‐4) cells. Phloroglucinol was found to scavenge 1,1‐diphenyl‐2‐picrylhydrazyl (DPPH) radical, hydrogen peroxide (H2O2), hydroxy radical, intracellular reactive oxygen species (ROS), and thus prevented lipid peroxidation. As a result, phloroglucinol reduced H2O2 induced apoptotic cells formation in V79‐4 cells. In addition, phloroglucinol inhibited cell damage induced by serum starvation and radiation through scavenging ROS. Phloroglucinol increased the catalase activity and its protein expression. In addition, catalase inhibitor abolished the protective effect of phloroglucinol from H2O2 induced cell damage. Furthermore, phloroglucinol increased phosphorylation of extracellular signal regulated kinase (ERK). Taken together, the results suggest that phloroglucinol protects V79‐4 cells against oxidative damage by enhancing the cellular catalase activity and modulating ERK signal pathway.


Journal of Pharmacology and Experimental Therapeutics | 2006

Anti-Inflammatory Mechanisms of Isoflavone Metabolites in Lipopolysaccharide-Stimulated Microglial Cells

Jin Sun Park; Moon Sook Woo; Dong-Hyun Kim; Jin Won Hyun; Won Ki Kim; Jae Chul Lee; Hee-Sun Kim

The microglial activation plays an important role in neurodegenerative diseases by producing several proinflammatory cytokines and nitric oxide (NO). We found that three types of isoflavones and their metabolites that are transformed by the human intestinal microflora suppress lipopolysaccharide (LPS)-induced release of NO and tumor necrosis factor (TNF)-α in primary cultured microglia and BV2 microglial cell lines. The inhibitory effect of the isoflavone metabolites (aglycon form) was more potent than that of isoflavones (glycoside form). The RNase protection assay showed that the isoflavone metabolites regulated inducible nitric oxide synthase (iNOS) and the cytokines at either the transcriptional or post-transcriptional level. A further molecular mechanism study was performed for irisolidone, a metabolite of kakkalide, which had the most potent anti-inflammatory effect among the six isoflavones tested. Irisolidone significantly inhibited the DNA binding and transcriptional activity of nuclear factor (NF)-κB and activator protein-1. Moreover, it repressed the LPS-induced extracellular signal-regulated kinase (ERK) phosphorylation without affecting the activity of c-Jun N-terminal kinase or p38 mitogen-activated protein kinase. The level of NF-κB inhibition by irisolidone correlated with the level of iNOS, TNF-α, and interleukin (IL)-1β suppression in LPS-stimulated microglia, whereas the level of ERK inhibition correlated with the level of TNF-α and IL-1β repression. Overall, the repression of proinflammatory cytokines and iNOS gene expression in activated microglia by isoflavones such as irisolidone might have therapeutic potential for various neurodegenerative diseases including ischemic cerebral disease.


Free Radical Research | 2005

Triphlorethol-A from Ecklonia cava protects V79-4 lung fibroblast against hydrogen peroxide induced cell damage

Kyoung Ah Kang; Kyoung Hwa Lee; Sungwook Chae; Young Sang Koh; Byoung-Sam Yoo; Ju Ho Kim; Young Min Ham; Jong Seok Baik; Nam Ho Lee; Jin Won Hyun

In the present study, triphlorethol-A, a phlorotannin, was isolated from Ecklonia cava and its antioxidant properties were investigated. Triphlorethol-A was found to scavenge intracellular reactive oxygen species (ROS) and 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical, and thus prevented lipid peroxidation. The radical scavenging activity of triphlorethol-A protected the Chinese hamster lung fibroblast (V79-4) cells exposed to hydrogen peroxide (H2O2) against cell death, via the activation of ERK protein. Furthermore, triphlorethol-A reduced the apoptotic cells formation induced by H2O2. Triphlorethol-A increased the activities of cellular antioxidant enzymes like, superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx). Hence, from the present study, it is suggestive that triphlorethol-A protects V79-4 cells against H2O2 damage by enhancing the cellular antioxidative activity.


International Journal of Cancer | 2006

Ginseng saponin metabolite suppresses phorbol ester-induced matrix metalloproteinase-9 expression through inhibition of activator protein-1 and mitogen-activated protein kinase signaling pathways in human astroglioma cells

Soo Hyun Jung; Moon Sook Woo; So Young Kim; Won Ki Kim; Jin Won Hyun; Eun-Jin Kim; Dong-Hyun Kim; Hee-Sun Kim

Aberrant expression of matrix metalloproteinase‐9 (MMP‐9) is implicated in the process of invasion and angiogenesis of malignant tumors as well as in inflammatory diseases of the CNS. Therefore, the development of compounds that can inhibit or suppress MMP‐9 is required to treat brain tumors. We investigated the effects of a ginseng saponin metabolite, compound K (20‐O‐(β‐D‐glucopyranosyl)‐20(S)‐protopanaxadiol), on MMP‐9 expression in human astroglioma cells. Compound K significantly inhibited the secretion and protein expression of MMP‐9 induced by PMA. The inhibitory effect of compound K on MMP‐9 expression correlated with decreased MMP‐9 mRNA levels and suppression of MMP‐9 promoter activity. The compound K–mediated inhibition of MMP‐9 gene expression appears to occur via AP‐1 because its DNA‐binding and transcriptional activities were suppressed by the agent. Furthermore, compound K significantly repressed the PMA‐mediated activation of p38 MAPK, ERK and JNK, which are upstream modulators of AP‐1. Finally, compound K inhibited the in vitro invasiveness of glioma cells. Therefore, inhibition of MMP‐9 expression by compound K might have therapeutic potential for controlling the growth and invasiveness of brain tumors.


The International Journal of Biochemistry & Cell Biology | 2012

Endoplasmic reticulum stress signaling is involved in silver nanoparticles-induced apoptosis.

Rui Zhang; Mei Jing Piao; Ki Cheon Kim; Areum Kim; Jeong-Yun Choi; Jinhee Choi; Jin Won Hyun

Although silver nanoparticles (AgNPs) have been reported to exert strong acute toxic effects on various cultured cells by inducing oxidative stress, the molecular mechanisms by which AgNPs-damaged cells are unknown. Because the endoplasmic reticulum (ER) may play an important role in the response to oxidative stress-induced damage and is quite sensitive to oxidative damage, we hypothesized that AgNPs may exert cytotoxic effects on cells by modulating ER stress. In our study, AgNPs resulted in cytotoxicity and apoptotic cell death when analyzing cell viability, DNA fragmentation and the apoptotic sub-G(1) population. Flow cytometry and confocal microscopy indicated that the cells were sensitive to AgNPs with respect to the induction of mitochondrial Ca(2+) overloading and enhancement of ER stress. AgNPs induced a number of signature ER stress markers, including phosphorylation of RNA-dependent protein kinase-like ER kinase (PERK) and its downstream eukaryotic initiation factor 2α, phosphorylation of inositol-requiring protein 1 (IRE1), splicing of ER stress-specific X-box transcription factor-1, cleavage of activating transcription factor 6 (ATF6) and up-regulation of glucose-regulated protein-78 and CCAAT/enhancer-binding protein-homologous protein (CHOP/GADD153). Down-regulation of PERK, IRE1 and ATF6 expression using siRNA significantly decreased AgNPs-induced the enhancement of ER stress. In addition, down-regulation of CHOP expression with siRNA CHOP attenuated AgNPs-induced apoptosis. Taken together, the present study supports an important role for the ER stress response in mediating AgNPs-induced apoptosis.


Journal of Agricultural and Food Chemistry | 2009

Effect of Compound K, a Metabolite of Ginseng Saponin, Combined with γ-Ray Radiation in Human Lung Cancer Cells in Vitro and in Vivo

Sungwook Chae; Kyoung Ah Kang; Weon Young Chang; Min Jung Kim; Su Jae Lee; Yun Sil Lee; Hee-Sun Kim; Dong-Hyun Kim; Jin Won Hyun

Pretreatment of NCI-H460 human lung cancer cells with compound K produced by intestinal bacteria enhances gamma-ray radiation-induced cell death. Increases in apoptosis induced by combined treatment are made apparent in the observation of nuclear fragmentation, loss of mitochondrial membrane potential (Deltapsi), and activation of caspase 3. Apoptosis induced by compound K and gamma-ray radiation is associated with reactive oxygen species (ROS) generation. Furthermore, compound K, in combination with gamma-ray radiation, has an enhanced effect in the regression of NCI-H460 tumor xenografts of nude mice. These results suggest that compound K has possible application for cancer therapy when used in combination with gamma-ray radiation.


The International Journal of Biochemistry & Cell Biology | 2010

Up-regulation of Nrf2-mediated heme oxygenase-1 expression by eckol, a phlorotannin compound, through activation of Erk and PI3K/Akt.

Ki Cheon Kim; Kyoung Ah Kang; Rui Zhang; Mei Jing Piao; Gi Young Kim; Mi Young Kang; Su Jae Lee; Nam Ho Lee; Young-Joon Surh; Jin Won Hyun

The aim of the present study was to examine the cytoprotective effect of eckol, a phlorotannin found in Ecklonia cava and to elucidate underlying mechanisms. Heme oxygenase-1 (HO-1) is an important antioxidant enzyme that plays a role in cytoprotection against oxidative stress. Eckol-induced HO-1 expression both at the level of mRNA and protein in Chinese hamster lung fibroblast (V79-4) cells, resulting in increased HO-1 activity. The transcription factor NF-E2-related factor 2 (Nrf2) is a critical regulator of HO-1, achieved by binding to the antioxidant response element (ARE). Eckol treatment resulted in the enhanced level of phosphorylated form, nuclear translocation, ARE-binding, and transcriptional activity of Nrf2. Extracellular regulated kinase (Erk) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB, Akt) contributed to ARE-driven HO-1 expression. Eckol activated both Erk and Akt, and treatments with U0126 (an Erk kinase inhibitor), LY294002 (a PI3K inhibitor), specific Erk1 siRNA, and Akt siRNA suppressed the eckol-induced activation of Nrf2, resulting in a decrease in HO-1 expression. ZnPP (a HO-1 inhibitor), HO-1 siRNA, and Nrf2 siRNA markedly abolished the cytoprotective effect of eckol against hydrogen peroxide-induced cell damage. Likewise, U0126 and LY294002 inhibited the eckol-induced cytoprotective effect against oxidative cell damage. These studies demonstrate that eckol attenuates oxidative stress by activating Nrf2-mediated HO-1 induction via Erk and PI3K/Akt signaling.


Cell Death and Disease | 2013

A ginseng metabolite, compound K, induces autophagy and apoptosis via generation of reactive oxygen species and activation of JNK in human colon cancer cells

Ah-Young Kim; K A Kang; Hyun-Taek Kim; Do Hyun Kim; Yung Hyun Choi; Sang-Jin Lee; Jin Won Hyun

Compound K (20-O-(β-D-glucopyranosyl)-20(S)-protopanaxadiol) is an active metabolite of ginsenosides and induces apoptosis in various types of cancer cells. This study investigated the role of autophagy in compound K-induced cell death of human HCT-116 colon cancer cells. Compound K activated an autophagy pathway characterized by the accumulation of vesicles, the increased positive acridine orange-stained cells, the accumulation of LC3-II, and the elevation of autophagic flux. Whereas blockade of compound K-induced autophagy by 3-methyladenein and bafilomycin A1 significantly increased cell viability. In addition, compound K augmented the time-dependent expression of the autophagy-related proteins Atg5, Atg6, and Atg7. However, knockdown of Atg5, Atg6, and Atg7 markedly inhibited the detrimental impact of compound K on LC3-II accumulation and cell vitality. Compound K-provoked autophagy was also linked to the generation of intracellular reactive oxygen species (ROS); both of these processes were mitigated by the pre-treatment of cells with the antioxidant N-acetylcysteine. Moreover, compound K activated the c-Jun NH2-terminal kinase (JNK) signaling pathway, whereas downregulation of JNK by its specific inhibitor SP600125 or by small interfering RNA against JNK attenuated autophagy-mediated cell death in response to compound K. Compound K also provoked apoptosis, as evidenced by an increased number of apoptotic bodies and sub-G1 hypodiploid cells, enhanced activation of caspase-3 and caspase-9, and modulation of Bcl-2 and Bcl-2-associated X protein expression. Notably, compound K-stimulated autophagy as well as apoptosis was induced by disrupting the interaction between Atg6 and Bcl-2. Taken together, these results indicate that the induction of autophagy and apoptosis by compound K is mediated through ROS generation and JNK activation in human colon cancer cells.

Collaboration


Dive into the Jin Won Hyun's collaboration.

Top Co-Authors

Avatar

Kyoung Ah Kang

UPRRP College of Natural Sciences

View shared research outputs
Top Co-Authors

Avatar

Mei Jing Piao

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Rui Zhang

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Ki Cheon Kim

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Sungwook Chae

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Young Sang Koh

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Ji Won Cha

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Hee-Sun Kim

Ewha Womans University

View shared research outputs
Top Co-Authors

Avatar

Jae Woo Park

Jeju National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge