Network


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

Hotspot


Dive into the research topics where Ji-Hong Moon is active.

Publication


Featured researches published by Ji-Hong Moon.


Oncotarget | 2016

Activation of autophagy flux by metformin downregulates cellular FLICE–like inhibitory protein and enhances TRAIL- induced apoptosis

Uddin Md. Nazim; Ji-Hong Moon; Ju-Hee Lee; You-Jin Lee; Jae-Won Seol; Seong-Kug Eo; John-Hwa Lee; Sang-Youel Park

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. TRAIL is regarded as one of the most promising anticancer agents, because it can destruct cancer cells without showing any toxicity to normal cells. Metformin is an anti-diabetic drug with anticancer activity by inhibiting tumor cell proliferation. In this study, we demonstrated that metformin could induce TRAIL-mediated apoptotic cell death in TRAIL-resistant human lung adenocarcinoma A549 cells. Pretreatment of metformindownregulation of c-FLIP and markedly enhanced TRAIL-induced tumor cell death by dose-dependent manner. Treatment with metformin resulted in slight increase in the accumulation of microtubule-associated protein light chain LC3-II and significantly decreased the p62 protein levels by dose-dependent manner indicated that metformin induced autophagy flux activation in the lung cancer cells. Inhibition of autophagy flux using a specific inhibitor and genetically modified ATG5 siRNA blocked the metformin-mediated enhancing effect of TRAIL. These data demonstrated that downregulation of c-FLIP by metformin enhanced TRAIL-induced tumor cell death via activating autophagy flux in TRAIL-resistant lung cancer cells and also suggest that metformin may be a successful combination therapeutic strategy with TRAIL in TRAIL-resistant cancer cells including lung adenocarcinoma cells.


Oncology Reports | 2015

Quercetin-induced autophagy flux enhances TRAIL-mediated tumor cell death

Ji-Hong Moon; Seong Kug Eo; John Hwa Lee; Sang-Youel Park

Quercetin is a potent cancer therapeutic agent and dietary antioxidant present in fruit and vegetables. Quercetin prevents tumor proliferation by inducing cell cycle arrest and is a well known cancer therapeutic agent and autophagy mediator. We investigated whether quercetin enhances TRAIL-induced tumor cell death and the possible mechanism in human lung cancer cells. We identified that quercetin markedly enhanced TRAIL-mediated lung cancer cell death. Quercetin treatment dose-dependently decreased the p62 protein expression and increased GFP-LC3B. Autophagy flux inhibitor, chloroquine treatment blocked the enhancing effects of TRAIL-induced apoptosis by quercetin. Our results indicated that quercetin enhanced TRAIL-induced cell death via autophagy flux activation, and also suggest that quercetin may be a therapeutic agent against human lung cancer via combination therapy with many anticancer drugs including TRAIL.


International Journal of Molecular Sciences | 2014

Overcoming Hypoxic-Resistance of Tumor Cells to TRAIL-Induced Apoptosis through Melatonin

You-Jin Lee; Ju-Hee Lee; Ji-Hong Moon; Sang-Youel Park

A solid tumor is often exposed to hypoxic or anoxic conditions; thus, tumor cell responses to hypoxia are important for tumor progression as well as tumor therapy. Our previous studies indicated that tumor cells are resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cell apoptosis under hypoxic conditions. Melatonin inhibits cell proliferation in many cancer types and induces apoptosis in some particular cancer types. Here, we examined the effects of melatonin on hypoxic resistant cells against TRAIL-induced apoptosis and the possible mechanisms of melatonin in the hypoxic response. Melatonin treatment increased TRAIL-induced A549 cell death under hypoxic conditions, although hypoxia inhibited TRAIL-mediated cell apoptosis. In a mechanistic study, hypoxia inducible factor-1α and prolyl-hydroxylase 2 proteins, which increase following exposure to hypoxia, were dose-dependently down-regulated by melatonin treatment. Melatonin also blocked the hypoxic responses that reduced pro-apoptotic proteins and increased anti-apoptotic proteins including Bcl-2 and Bcl-xL. Furthermore, melatonin treatment reduced TRAIL resistance by regulating the mitochondrial transmembrane potential and Bax translocation. Our results first demonstrated that melatonin treatment induces apoptosis in TRAIL-resistant hypoxic tumor cells by diminishing the anti-apoptotic signals mediated by hypoxia and also suggest that melatonin could be a tumor therapeutic tool by combining with other apoptotic ligands including TRAIL, particularly in solid tumor cells exposed to hypoxia.


Journal of Pineal Research | 2014

Melatonin-mediated β-catenin activation protects neuron cells against prion protein-induced neurotoxicity

Jae-Kyo Jeong; Ju-Hee Lee; Ji-Hong Moon; You-Jin Lee; Sang-Youel Park

Activation of β‐catenin in neurons regulates mitochondrial function and protects against protein misfolding disorders, including Alzheimers disease and Huntingtons disease. Melatonin, a natural secretory product of the pineal gland, exerts neuroprotective effects through the activation of β‐catenin. In this study, melatonin increased β‐catenin protein expression and activation in human neuroblastoma cell lines SH‐SY5Y cells. Melatonin also inhibited PrP (106–126)‐induced neurotoxicity and the inhibition attenuated by treatment of β‐catenin inhibitor ICG‐001. Activation of β‐catenin blocked PrP (106–126)‐mediated downregulation of anti‐apoptotic protein survivin and Bcl‐2. Reduction of mitochondrial membrane potential, translocation of Bax, and cytochrome c release which induced by PrP (106–126) treatment were inhibited by β‐catenin activation, which contributed to prevented PrP (106–126)‐induced neuronal cell death. In conclusion, β‐catenin activation by melatonin prevented PrP (106–126)‐induced neuronal cell death through regulating anti‐apoptotic proteins and mitochondrial pathways. These results also suggest the therapeutic value of Wnt/β‐catenin signaling in prion‐related disorders as influenced by melatonin.


Oncotarget | 2016

Autophagy flux induced by ginsenoside-Rg3 attenuates human prion protein-mediated neurotoxicity and mitochondrial dysfunction

Ji-Hong Moon; Ju-Hee Lee; You-Jin Lee; Sang-Youel Park

Mitochondrial quality control is a process by which mitochondria undergo successive rounds of fusion and fission with dynamic exchange of components to segregate functional and damaged elements. Removal of mitochondrion that contains damaged components is accomplished via autophagy. In this study, we investigated whether ginsenoside Rg3, an active ingredient of the herbal medicine ginseng that is used as a tonic and restorative agent, could attenuate prion peptide, PrP (106-126)-induced neurotoxicity and mitochondrial damage. To this end, western blot and GFP-LC3B puncta assay were performed to monitor autophagy flux in neuronal cells; LC3B-II protein level was found to increase after Rg3 treatment. In addition, electron microscopy analysis showed that Rg3 enhanced autophagic vacuoles in neuronal cells. By using autophagy inhibitors wortmannin and 3-methyladenine (3MA) or autophagy protein 5 (Atg5) small interfering RNA (siRNA), we demonstrated that Rg3 could protect neurons against PrP (106-126)-induced cytotoxicity via autophagy flux. We found that Rg3 could also attenuate PrP (106-126)-induced mitochondrial damage via autophagy flux. Taken together, our results suggest that Rg3 is a possible therapeutic agent in neurodegenerative disorders, including prion diseases.


Oncology Reports | 2015

Deferoxamine inhibits TRAIL-mediated apoptosis via regulation of autophagy in human colon cancer cells

Ji-Hong Moon; Jae-Kyo Jeong; Sang-Youel Park

Deferoxamine (DFO), an iron chelator, has numerous clinical applications for patients presenting with iron overload in regards to the improvement in the quality of life and overall survival. In addition, experimental iron chelators have demonstrated potent anticancer properties. The present study investigated the effects of DFO on TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in colon cancer cells and and the mechanism involved. The experimental results showed that DFO treatment inhibited TRAIL-mediated cancer cell apoptosis by increasing Akt activation and decreasing caspase activation in human colon cancer cells. Furthermore, DFO treatment induced autophagy flux, and chloroquine, an autophagy inhibitor, blocked DFO-mediated inhibition of TRAIL-induced apoptosis. The present study demonstrated that DFO inhibited TRAIL-mediated tumor cell death via the autophagy pathway, and the results suggest that potent anticancer agent, DFO, can be an inhibitor against antitumor therapy including TRAIL protein.


International Journal of Molecular Medicine | 2014

Caffeine prevents human prion protein-mediated neurotoxicity through the induction of autophagy

Ji-Hong Moon; Ju-Hee Lee; Jin-Young Park; Sung-Wook Kim; You-Jin Lee; Seog Jin Kang; Jae-Won Seol; Dongchoon Ahn; Sang-Youel Park

The human prion protein (PrP) fragment PrP(106‑126) possesses the majority of the pathogenic properties associated with the infectious scrapie isoform of PrP, known as PrPSc. The accumulation of PrPSc in the brain of humans and animals affects the central nervous system. Recent epidemiological studies have suggested that caffeine, one of the major components of coffee, exerts protective effects against the development of neurodegeneration. However, the protective effects of caffeine against prion disease have not been reported to date. In this study, we therefore investigated the effects of caffeine on PrP-mediated neurotoxicity. The protein expression of the autophagosomal marker, LC3-II, was increased by caffeine in a dose-dependent manner, and the autophagy induced by caffeine protected the neuronal cells against PrP(106‑126)‑induced cell death. On the contrary, the downregulation of LC3-II using the autophagy inhibitors, 3-methyladenine (3-ΜΑ) and wortmannin, prevented the caffeine-mediated neuroprotective effects. To the best of our knowledge, the present study provides the first evidence that treatment with caffeine protects human neuronal cells against prion‑mediated neurotoxicity and these neuroprotective effects are mediated by caffeine-induced autophagy signals. Our data suggest that treatment with caffeine may be a novel therapeutic strategy for prion peptide‑induced apoptosis.


Oncotarget | 2016

Melatonin protects skin keratinocyte from hydrogen peroxide-mediated cell death via the SIRT1 pathway

Ju-Hee Lee; Ji-Hong Moon; Uddin Md. Nazim; You-Jin Lee; Jae-Won Seol; Seong-Kug Eo; John-Hwa Lee; Sang-Youel Park

Melatonin (N-acetyl-5-methoxytryptamine), which is primarily synthesized in and secreted from the pineal gland, plays a pivotal role in cell proliferation as well as in the regulation of cell metastasis and cell survival in a diverse range of cells. The aim of this study is to investigate protection effect of melatonin on H2O2-induced cell damage and the mechanisms of melatonin in human keratinocytes. Hydrogen peroxide dose-dependently induced cell damages in human keratinocytes and co-treatment of melatonin protected the keratinocytes against H2O2-induced cell damage. Melatonin treatment activated the autophagy flux signals, which were identified by the decreased levels of p62 protein. Inhibition of autophagy flux via an autophagy inhibitor and ATG5 siRNA technique blocked the protective effects of melatonin against H2O2-induced cell death in human keratinocytes. And we found the inhibition of sirt1 using sirtinol and sirt1 siRNA reversed the protective effects of melatonin and induces the autophagy process in H2O2-treated cells. This is the first report demonstrating that autophagy flux activated by melatonin protects human keratinocytes through sirt1 pathway against hydrogen peroxide-induced damages. And this study also suggest that melatonin could potentially be utilized as a therapeutic agent in skin disease.


Oncotarget | 2016

Hinokitiol protects primary neuron cells against prion peptide-induced toxicity via autophagy flux regulated by hypoxia inducing factor-1

Ji-Hong Moon; Ju-Hee Lee; You-Jin Lee; Sang-Youel Park

Prion diseases are fatal neurodegenerative disorders that are derived from structural changes of the native PrPc. Recent studies indicated that hinokitiol induced autophagy known to major function that keeps cells alive under stressful conditions. We investigated whether hinokitiol induces autophagy and attenuates PrP (106-126)-induced neurotoxicity. We observed increase of LC3-II protein level, GFP-LC3 puncta by hinokitiol in neuronal cells. Addition to, electron microscopy showed that hinokitiol enhanced autophagic vacuoles in neuronal cells. We demonstrated that hinokitiol protects against PrP (106-126)-induced neurotoxicity via autophagy by using autophagy inhibitor, wortmannin and 3MA, and ATG5 small interfering RNA (siRNA). We checked hinokitiol activated the hypoxia-inducible factor-1α (HIF-1α) and identified that hinokitiol-induced HIF-1α regulated autophagy. Taken together, this study is the first report demonstrating that hinokitiol protected against prion protein-induced neurotoxicity via autophagy regulated by HIF-1α. We suggest that hinokitiol is a possible therapeutic strategy in neuronal disorders including prion disease.


International Journal of Molecular Medicine | 2015

Baicalein prevents human prion protein-induced neuronal cell death by regulating JNK activation

Ji-Hong Moon; Sang-Youel Park

Prion diseases are neurodegenerative disorders characterized by the accumulation of an abnormal isoform of the protease-insensitive isoform (PrPSc) of prion protein. Human prion protein fragment 106‑126 [PrP (106‑126)] contains most of the pathological characteristics associated with PrPSc. Although a number of compounds have been identified to inhibit PrP accumulation or dissolve fibrils and aggregates in vitro, there is currenlty no treatment available for these progressive neurodegenerative diseases. Baicalein, the dried root of Scutellaria baicalensis (S. baicalensis) Georgi (known as Huang-qin in traditional Chinese medicine) has been reported to exert neuroprotective effects on neurodegenerative diseases. In the present study, we investigated the effects of baicalein on the development of prion diseases using SH-SY5Y and SK-N-SH cells in vitro. We found that baicalein protected the cells against PrP‑induced neuronal cell death by inhibiting the production of reactive oxygen species (ROS) and mitochondrial dysfunction using ROS detection assay and MTP assay. We demonstrated that baicalein treatment regulated the phosphorylation of c-Jun N-terminal kinase (JNK) by using western blot analysis and Annexin V assay. Our data suggest that baicalein has potential for use as a therapeutic drug for the treatment of various neurodegenerative diseases, including prion diseases.

Collaboration


Dive into the Ji-Hong Moon's collaboration.

Top Co-Authors

Avatar

Sang-Youel Park

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

You-Jin Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Ju-Hee Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Won Seol

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Uddin Md. Nazim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Kyo Jeong

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Sung-Wook Kim

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

John-Hwa Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Seong-Kug Eo

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Yang-Gyu Park

Chonbuk National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge