Network


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

Hotspot


Dive into the research topics where Ji Won Cha is active.

Publication


Featured researches published by Ji Won Cha.


International Journal of Molecular Medicine | 2014

7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways.

Min Ju Ryu; Kyoung Ah Kang; Mei Jing Piao; Ki Cheon Kim; Jian Zheng; Cheng Wen Yao; Ji Won Cha; Ha Sook Chung; Sang Cheol Kim; Eunsun Jung; Deokhoon Park; Sungwook Chae; Jin Won Hyun

This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. DHF markedly upregulated HO-1 mRNA and protein expression in human keratinocytes (HaCaT cells), resulting in increased HO-1 activity. DHF also increased the protein level of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates HO-1 expression by binding to the antioxidant response element (ARE) within the HO-1 gene promoter, in a time-dependent manner. Moreover, DHF decreased the expression of Kelch-like ECH-associated protein 1, a repressor of Nrf2 activity, and induced the translocation of Nrf2 from the cytosol into the nucleus, thereby allowing its association with the ARE site. DHF activated extracellular-regulated kinase (ERK) and protein kinase B (PKB, Akt) in keratinocytes, while the ERK and Akt inhibitors attenuated DHF-enhanced Nrf2 and HO-1 expression. DHF also protected the keratinocytes against hydrogen peroxide- and ultraviolet B-induced oxidative damage, while HO-1, ERK and Akt inhibitors markedly suppressed DHF-mediated cytoprotection. Taken together, the results suggested that DHF activates ERK- and Akt-Nrf2 signaling cascades in HaCaT cells, leading to the upregulation of HO-1 and cytoprotection against oxidative stress.


Biomolecules & Therapeutics | 2014

The Polyphenol Chlorogenic Acid Attenuates UVB-mediated Oxidative Stress in Human HaCaT Keratinocytes.

Ji Won Cha; Mei Jing Piao; Ki Cheon Kim; Cheng Wen Yao; Jian Zheng; Seong Min Kim; Chang Lim Hyun; Yong Seok Ahn; Jin Won Hyun

We investigated the protective effects of chlorogenic acid (CGA), a polyphenol compound, on oxidative damage induced by UVB exposure on human HaCaT cells. In a cell-free system, CGA scavenged 1,1-diphenyl-2-picrylhydrazyl radicals, superoxide anions, hydroxyl radicals, and intracellular reactive oxygen species (ROS) generated by hydrogen peroxide and ultraviolet B (UVB). Furthermore, CGA absorbed electromagnetic radiation in the UVB range (280–320 nm). UVB exposure resulted in damage to cellular DNA, as demonstrated in a comet assay; pre-treatment of cells with CGA prior to UVB irradiation prevented DNA damage and increased cell viability. Furthermore, CGA pre-treatment prevented or ameliorated apoptosis-related changes in UVB-exposed cells, including the formation of apoptotic bodies, disruption of mitochondrial membrane potential, and alterations in the levels of the apoptosis-related proteins Bcl-2, Bax, and caspase-3. Our findings suggest that CGA protects cells from oxidative stress induced by UVB radiation.


International Journal of Oncology | 2013

Compound K, a metabolite of ginseng saponin, inhibits colorectal cancer cell growth and induces apoptosis through inhibition of histone deacetylase activity

Kyoung Ah Kang; Mei Jing Piao; Ki Cheon Kim; Jian Zheng; Cheng Wen Yao; Ji Won Cha; Hye Sun Kim; Dong-Hyun Kim; Suk Chul Bae; Jin Won Hyun

In this study, we investigated the molecular mechanisms underlying the anti-proliferative effects of Compound K, with specific reference to histone modification. Exposure of HT-29 human colon cancer cells to Compound K resulted in time-dependent inhibition of histone deacetylase (HDAC) activity, mRNA and protein expression. Compound K treatment induced unmethylation of the RUNX3 promoter region such as TSA treatment and an accumulation of acetylated histones H3 and H4 within the total cellular chromatin, resulting in an enhanced ability of these histones to bind to the promoter sequences of the tumor suppressor gene Runt-related transcription factor 3 (RUNX3). Treatment of cells with Compound K increased the mRNA and protein expression of RUNX3, as well as p21, a downstream target of RUNX3. These alterations were consistent with cell cycle arrest at the G0/G1 phases and induction of apoptosis. Our results provide new insights into the mechanisms of Compound K action in human colorectal cancer cells and suggest that HDAC inhibition presents a novel approach to prevent or treat colorectal cancer.


In Vitro Cellular & Developmental Biology – Animal | 2014

Fisetin attenuates hydrogen peroxide-induced cell damage by scavenging reactive oxygen species and activating protective functions of cellular glutathione system

Kyoung Ah Kang; Mei Jing Piao; Ki Cheon Kim; Ji Won Cha; Jian Zheng; Cheng Wen Yao; Sungwook Chae; Jin Won Hyun

Hydrogen peroxide (H2O2) can induce cell damage by generating reactive oxygen species (ROS), resulting in DNA damage and cell death. The aim of this study is to elucidate the protective effects of fisetin (3,7,3′,4′,-tetrahydroxy flavone) against H2O2-induced cell damage. Fisetin reduced the level of superoxide anion, hydroxyl radical in cell free system, and intracellular ROS generated by H2O2. Moreover, fisetin protected against H2O2-induced membrane lipid peroxidation, cellular DNA damage, and protein carbonylation, which are the primary cellular outcomes of H2O2 treatment. Furthermore, fisetin increased the level of reduced glutathione (GSH) and expression of glutamate-cysteine ligase catalytic subunit, which is decreased by H2O2. Conversely, a GSH inhibitor abolished the cytoprotective effect of fisetin against H2O2-induced cells damage. Taken together, our results suggest that fisetin protects against H2O2-induced cell damage by inhibiting ROS generation, thereby maintaining the protective role of the cellular GSH system.


Journal of Cellular Biochemistry | 2014

Cytoprotective Effect of Eckol Against Oxidative Stress-Induced Mitochondrial Dysfunction: Involvement of the FoxO3a/AMPK Pathway

Areum Kim; Kyoung Ah Kang; Mei Jing Piao; Ki Cheon Kim; Jian Zheng; Cheng Wen Yao; Ji Won Cha; Chang Lim Hyun; Hee Kyoung Kang; Nam Ho Lee; Jin Won Hyun

This study investigated the cytoprotective effect of Ecklonia cava‐derived eckol against H2O2‐induced mitochondrial dysfunction in Chang liver cells. While H2O2 augmented levels of mitochondrial reactive oxygen species (ROS), eckol decreased it. Eckol also attenuated high intracellular Ca2+ levels stimulated by H2O2 and recovered H2O2‐diminished ATP levels and succinate dehydrogenase activity. Eckol time‐dependently increased the expression of manganese superoxide dismutase (Mn SOD), a mitochondrial antioxidant enzyme with cytoprotective effect against oxidative stress. Eckol recovered Mn SOD expression and activity that were decreased by H2O2. Finally, eckol induced Mn SOD through phosphorylated AMP‐activated protein kinase (AMPK) and forkhead box O3a (FoxO3a). Specific silencing RNAs (siRNAs) against FoxO3a and AMPK reduced eckol‐stimulated Mn SOD expression, and diethyldithiocarbamate (Mn SOD inhibitor) and siRNA against Mn SOD reduced the cytoprotective effect of eckol against H2O2‐provoked cell death. These results demonstrate that eckol protects cells from mitochondrial oxidative stress by activating AMPK/FoxO3a‐mediated induction of Mn SOD. J. Cell. Biochem. 115: 1403–1411, 2014.


Biomolecules & Therapeutics | 2015

Galangin (3,5,7-Trihydroxyflavone) Shields Human Keratinocytes from Ultraviolet B-Induced Oxidative Stress

Susara Ruwan Kumara Madduma Hewage; Mei Jing Piao; Ki Cheon Kim; Ji Won Cha; Xia Han; Yung Hyun Choi; Sungwook Chae; Jin Won Hyun

Most skin damage caused by ultraviolet B (UVB) radiation is owing to the generation of reactive oxygen species. Phytochemicals can act as antioxidants against UVB-induced oxidative stress. This study investigated the protective effects of the flavone galangin against UVB-induced oxidative damage in human keratinocytes. Galangin efficiently scavenged free radicals and reduced UVB-induced damage to cellular macromolecules, such as DNA, lipids, and proteins. Furthermore, galangin rescued cells undergoing apoptosis induced by UVB radiation via recovering mitochondrial polarization and down-regulating apoptotic proteins. These results showed that galangin protects human keratinocytes against UVB radiation-induced cellular damage and apoptosis via its antioxidant effects.


Marine Drugs | 2014

Fucoxanthin Enhances the Level of Reduced Glutathione via the Nrf2-Mediated Pathway in Human Keratinocytes

Jian Zheng; Mei Jing Piao; Ki Cheon Kim; Cheng Wen Yao; Ji Won Cha; Jin Won Hyun

Fucoxanthin, a natural carotenoid, is abundant in seaweed with antioxidant properties. This study investigated the role of fucoxanthin in the induction of antioxidant enzymes involved in the synthesis of reduced glutathione (GSH), synthesized by glutamate-cysteine ligase catalytic subunit (GCLC) and glutathione synthetase (GSS), via Akt/nuclear factor-erythroid 2-related (Nrf2) pathway in human keratinocytes (HaCaT) and elucidated the underlying mechanism. Fucoxanthin treatment increased the mRNA and protein levels of GCLC and GSS in HaCaT cells. In addition, fucoxanthin treatment promoted the nuclear translocation and phosphorylation of Nrf2, a transcription factor for the genes encoding GCLC and GSS. Chromatin immune-precipitation and luciferase reporter gene assays revealed that fucoxanthin treatment increased the binding of Nrf2 to the antioxidant response element (ARE) sequence and transcriptional activity of Nrf2. Fucoxanthin treatment increased phosphorylation of Akt (active form), an up-regulator of Nrf2 and exposure to LY294002, a phosphoinositide 3-kinase (PI3K)/Akt inhibitor, suppressed the fucoxanthin-induced activation of Akt, Nrf2, resulting in decreased GCLC and GSS expression. In accordance with the effects on GCLC and GSS expression, fucoxanthin induced the level of GSH. In addition, fucoxanthin treatment recovered the level of GSH reduced by ultraviolet B irradiation. Taken together, these findings suggest that fucoxanthin treatment augments cellular antioxidant defense by inducing Nrf2-driven expression of enzymes involved in GSH synthesis via PI3K/Akt signaling.


Biomolecules & Therapeutics | 2014

Fucodiphlorethol G Purified from Ecklonia cava Suppresses Ultraviolet B Radiation-Induced Oxidative Stress and Cellular Damage.

Ki Cheon Kim; Mei Jing Piao; Jian Zheng; Cheng Wen Yao; Ji Won Cha; Madduma Hewage Susara Ruwan Kumara; Xia Han; Hee Kyoung Kang; Nam Ho Lee; Jin Won Hyun

Fucodiphlorethol G (6’-[2,4-dihydroxy-6-(2,4,6-trihydroxyphenoxy)phenoxy]biphenyl-2,2’,4,4’,6-pentol) is a compound purified from Ecklonia cava, a brown alga that is widely distributed offshore of Jeju Island. This study investigated the protective effects of fucodiphlorethol G against oxidative damage-mediated apoptosis induced by ultraviolet B (UVB) irradiation. Fucodiphlorethol G attenuated the generation of 2, 2-diphenyl-1-picrylhydrazyl radicals and intracellular reactive oxygen species in response to UVB irradiation. Fucodiphlorethol G suppressed the inhibition of human keratinocyte growth by UVB irradiation. Additionally, the wavelength of light absorbed by fucodiphlorethol G was close to the UVB spectrum. Fucodiphlorethol G reduced UVB radiation-induced 8-isoprostane generation and DNA fragmentation in human keratinocytes. Moreover, fucodiphlorethol G reduced UVB radiation-induced loss of mitochondrial membrane potential, generation of apoptotic cells, and active caspase-9 expression. Taken together, fucodiphlorethol G protected human keratinocytes against UVB radiation-induced cell damage and apoptosis by absorbing UVB radiation and scavenging reactive oxygen species.


Journal of cancer prevention | 2013

Morin Induces Heme Oxygenase-1 via ERK-Nrf2 Signaling Pathway

Ji Young Park; Kyoung Ah Kang; Ki Cheon Kim; Ji Won Cha; Eun Hee Kim; Jin Won Hyun

Background: Oxidative stress damages to cells or tissues, however, cellular defense systems including heme oxygenase-1 (HO-1) protects them against oxidative stress. Flavonoid compounds can activate cellular defense mechanisms against oxidative stress and it can reduce cell damages. In the present study, the cytoprotective effects of morin (3,5,7,2’,4’-pentahydroxyflavone), in terms of HO-1 enzyme, against the oxidative stress and its involved mechanisms was elucidated. Methods: RT-PCR and western blot analysis were assessed to detect the mRNA and protein expression, respectively. Cell viability was measured by using MTT test. The immunocytochemistry was performed to define location of target protein. Electrophoretic mobility shift assay performed to measure transcription factor-promoter site binding activity. Results: Morin elevated mRNA and protein levels of HO-1 in human lens epithelial cells (HLE-B3). HO-1 inhibitor ZnPP attenuated the protective effect of morin against H2O2-induced cytotoxicity. Morin increased the protein level of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which up-regulates HO-1 expression by binding to the antioxidant response element (ARE) within the HO-1 gene promoter. Moreover, morin induced the translocation of Nrf2 from the cytosol into the nucleus. Morin activated extracellular-regulated kinase (ERK), while ERK inhibitor attenuated morin-enhanced Nrf2 and HO-1 expression. Conclusions: Morin activates ERK-Nrf2 signaling cascades in HLE-B3 cells, leading to the up-regulation of HO-1 and cytoprotection against oxidative stress.


International Journal of Oncology | 2014

Epigenetic alterations are involved in the overexpression of glutathione S-transferase π-1 in human colorectal cancers

Rui Zhang; Kyoung Ah Kang; Mei Jing Piao; Ki Cheon Kim; Jian Zheng; Cheng Wen Yao; Ji Won Cha; Young Hee Maeng; Weon Young Chang; Pyong-Gon Moon; Moon-Chang Baek; Jin Won Hyun

Glutathione S-transferase π-1 (GSTP-1) is a member of the glutathione S-transferase enzyme superfamily, which catalyzes the conjugation of electrophiles to glutathione during the process of detoxification. In this study, the epigenetic alterations of GSTP-1 expression in human colorectal cancers and the underlying mechanisms were investigated. In 10 colon cancer patients, proteomic analysis revealed that expression of GSTP-1 protein was higher in tumor tissues than in paired adjacent normal tissues. Likewise, in 7 of 10 colon cancer patients, GSTP-1 protein expression was more than 1.5-fold higher in tumor tissues than in adjacent normal tissues, as determined by western blotting. Immunohistochemical data confirmed that GSTP-1 protein was expressed at higher levels in colon cancer tissues compared to normal mucosa. GSTP-1 enzyme activity was closely correlated with GSTP-1 protein expression in colon cancer patients. Consistent with this, GSTP-1 mRNA, protein and activity levels were higher in the colorectal cancer cell lines Caco-2, HCT-116, HT-29, SNU-407 and SNU-1033 compared to the normal colon cell line FHC. Methylation-specific PCR results indicated that the high levels of GSTP-1 in human colorectal cancer cell lines were likely due to the lower degree of promoter methylation in colon cancer cell lines compared to the normal colon cell line, consistent with findings in colon cancer patients. Moreover, the levels of specific activator-protein complexes and histone marks were higher in human colorectal cancer cells compared to the normal human colon cell line, whereas the repressor protein complexes exhibited the opposite pattern. Furthermore, chromatin immunoprecipitation assays demonstrated that expression levels of the transcription factors AP-1 and SP-1 were correlated with the upregulation of GSTP-1 expression in colorectal cancer cells. Finally, knockdown of GSTP-1 promoted the sensitivity of SNU-407 cells to the anticancer agent 5-fluorouracil. These data indicate that GSTP-1 may serve as a clinically useful biomarker of colon cancer and a target for anti-colon cancer drugs.

Collaboration


Dive into the Ji Won Cha's collaboration.

Top Co-Authors

Avatar

Jin Won Hyun

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

Mei Jing Piao

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Cheng Wen Yao

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Jian Zheng

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Kyoung Ah Kang

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Chang Lim Hyun

Jeju National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sungwook Chae

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Eun Sook Yoo

Jeju National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge