Network


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

Hotspot


Dive into the research topics where Jae-Kyo Jeong is active.

Publication


Featured researches published by Jae-Kyo Jeong.


Neuroscience Research | 2012

Autophagy induced by resveratrol prevents human prion protein-mediated neurotoxicity.

Jae-Kyo Jeong; Myung-Hee Moon; Bum-Chul Bae; You-Jin Lee; Jae-Won Seol; Hyung-Sub Kang; Jin-Shang Kim; Seog-Jin Kang; Sang-Youel Park

Our previous study revealed that resveratrol blocks prion protein peptide PrP(106-126)-induced neurotoxicity. However, the mechanism of resveratrol-mediated neuroprotection in prion diseases is not clear. Resveratrol initiates neuroprotective effects via the activation of autophagy, which protects organelles, cells, and organisms against misfolded protein-disorders, including Alzheimers disease and Parkinsons disease via regulation of mitochondrial homeostasis. Thus, we focused on elucidating the mechanisms responsible for resveratrol-mediated neuroprotection related to mitochondrial homeostasis as a result of autophagy activation. Resveratrol prevented PrP(106-126)-induced neuronal cell death by activating autophagy. Moreover, resveratrol-induced autophagy prevented the PrP(106-126)-induced reduction in mitochondrial potential and translocation of Bax to the mitochondria and cytochrome c release. Our results indicate that treatment with resveratrol appears to protect against neurotoxicity caused by prion protein peptides and the neuroprotection is induced by resveratrol-mediated autophagy signals.


Neurobiology of Aging | 2013

Autophagy induced by the class III histone deacetylase Sirt1 prevents prion peptide neurotoxicity

Jae-Kyo Jeong; Myung-Hee Moon; You-Jin Lee; Jae-Won Seol; Sang-Youel Park

Sirtuin 1 (Sirt1) is a class III histone deacetylase that mediates the protective effects of neurons in neurodegenerative disorders, including Alzheimers and prion disease. However, the mechanism directly involved in neuroprotection is still poorly understood. Recent evidence has demonstrated that activating Sirt1 induces autophagy, and that activating autophagy protects neurons against neurodegenerative disorders by regulating mitochondrial homeostasis. Thus, we focused on the mechanism of the Sirt1-mediated neuroprotective effect that was associated with regulating mitochondrial homeostasis via autophagy. Adenoviral-mediated Sirt1 overexpression prevented prion protein (PrP)(106-126)-induced neurotoxicity via autophagy processing. Moreover, Sirt1-induced autophagy protected against the PrP(106-126)-mediated decrease in the mitochondrial membrane potential value. Additionally, Sirt1 overexpression decreased PrP(106-126)-induced Bax translocation to the mitochondria and cytochrome c release into the cytosol. Sirt1 knockdown using small interfering (si) RNAs induced downregulation of Sirt1 protein expression and sensitized neuron cells to PrP(106-126)-induced cell death and mitochondrial dysfunction. Knockdown of autophagy-related 5 (ATG5) using small interfering RNA decreased autophagy-related 5 and autophagy marker microtubule-associated protein 1 light chain 3-II protein levels and blocked the effect of a Sirt1 activator against PrP(106-126)-induced mitochondrial dysfunction and neurotoxicity. Taken together, this study is the first report demonstrating that autophagy induced by Sirt1 activation plays a pivotal role protecting against prion-induced neuron cell death and also suggests that regulating autophagy including which by Sirt1 activation may be a therapeutic target for neurodegenerative disorders including the prion disease.


Osteoarthritis and Cartilage | 2013

SIRT1, a class III histone deacetylase, regulates TNF-α-induced inflammation in human chondrocytes

Myung-Hee Moon; Jae-Kyo Jeong; You-Jin Lee; Jae-Won Seol; Christopher J. Jackson; Sang-Youel Park

OBJECTIVE The present study was performed to elucidate the possible role of SIRT1 signaling in joint inflammation in human articular chondrocytes. DESIGN Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting were performed to detect gene products and proteins involved in tumor necrosis factor α (TNF-α)-induced inflammation and cartilage degradation in human primary chondrocytes. Matrix metalloproteinase (MMP)-2 and MMP-9 activity was evaluated by gelatin zymography. Overexpression and knockdown of SIRT1 were also performed to investigate whether SIRT1 is associated with the anti-inflammatory activity of resveratrol in chondrocytes. RESULTS Resveratrol dose-dependently inhibited TNF-α-induced cyclooxygenase-2 (COX-2), MMP-1, MMP-3, MMP-13 and PGE(2) production in human chondrocytes. Moreover, MMP-2 and MMP-9 activity was increased by treatment with TNF-α; however, SIRT1 activation decreased the proinflammatory effects induced by TNF-α. In addition, treatment of SIRT1 activator and overexpression of SIRT1 inhibited the expression and activation of the main proinflammatory regulator NF-κB, which was increased by TNF-α. When SIRT1 was overexpressed in chondrocytes, the anti-inflammatory action of SIRT1 was similar to that exerted by resveratrol. CONCLUSIONS SIRT1 activation deacetylates and inactivates NF-κB, and thereby, exerts an anti-inflammatory effect on chondrocytes, suggesting that SIRT1 activators could be explored as potential treatments for arthritis.


Neurobiology of Aging | 2012

SIRT1, a histone deacetylase, regulates prion protein-induced neuronal cell death

Jae-Suk Seo; Myung-Hee Moon; Jae-Kyo Jeong; Jae-Won Seol; You-Jin Lee; Byung-Hyun Park; Sang-Youel Park

Prion diseases associated with the conversion of the cellular prion protein (PrP(C)) to the misfolded isoform (PrP(Sc)), affect the central nervous system (CNS) of humans and animals. Resveratrol, an activator of class III histone deacetylase SIRT1, is important in attenuating cellular injury and oxidative stress. The present study investigated the effects of SIRT1 activation on prion protein-mediated neuronal cell death and examined its possible signals in intracellular apoptotic pathways. Resveratrol treatment significantly increased both SIRT1 protein expression and SIRT1 activity and protected neuronal cells against PrP (106-126)-induced cell death. Resveratrol-mediated SIRT1 activation decreased the acetylation of p53 and p65 induced by prion protein and SIRT1 inhibitor. SIRT1 activation also inhibited PrP (106-126)-mediated p38 mitogen-activating protein kinase (MAPK) activation and caspase-3 cleavage, and increased the expression of anti-apoptotic Bcl-xL protein. Furthermore, SIRT1 overexpression by using adenoviral vector protected neuronal cells against PrP (106-126). These results indicate that resveratrol inhibits PrP (106-126)-induced neuronal cell death by regulating SIRT1 activity and SIRT-related signaling, and suggest that prion-related disease may be attenuated by SIRT1 activation or by intake of SIRT1-activating molecules.


Journal of Pineal Research | 2012

Melatonin‐induced autophagy protects against human prion protein‐mediated neurotoxicity

Jae-Kyo Jeong; Myung-Hee Moon; You-Jin Lee; Jae-Won Seol; Sang-Youel Park

Abstract: Melatonin has neuroprotective effects in the models of neurodegenerative disease including Alzheimer’s and Parkinson’s disease. Several studies have shown that melatonin prevents neurodegeneration by regulation of mitochondrial function. However, the protective action of melatonin has not been reported in prion disease. We investigated the influence of melatonin on prion‐mediated neurotoxicity. Melatonin rescued neuronal cells from PrP(106–126)‐induced neurotoxicity by prevention of mitochondrial dysfunction. Moreover, the protective effect of melatonin against mitochondrial dysfunction was related with autophagy activation. Melatonin‐treated cells were dose‐dependently increased in LC3‐II, an autophagy marker. Melatonin‐induced autophagy prevented a PrP(106–126)‐induced reduction in mitochondrial potential and translocation of Bax to the mitochondria and cytochrome c release. On the other hand, downregulation of autophagy protein 5 with Atg5 siRNA or the autophagy blocker 3‐methyladenine prevented the melatonin‐mediated neuroprotective effects. This is the first report demonstrating that treatment with melatonin appears to protect against prion‐mediated neurotoxicity and that the neuroprotection is induced by melatonin‐mediated autophagy signals. The results of this study suggest that regulation of melatonin is a therapeutic strategy for prion peptide‐induced apoptosis.


Neurobiology of Aging | 2012

Hypoxia-inducible factor-1 alpha regulates prion protein expression to protect against neuron cell damage

Jae-Kyo Jeong; Jae-Suk Seo; Myung-Hee Moon; You-Jin Lee; Jae-Won Seol; Sang-Youel Park

The human prion protein fragment, PrP (106-126), may contain a majority of the pathological features associated with the infectious scrapie isoform of PrP, known as PrP(Sc). Based on our previous findings that hypoxia protects neuronal cells from PrP (106-126)-induced apoptosis and increases cellular prion protein (PrP(C)) expression, we hypothesized that hypoxia-related genes, including hypoxia-inducible factor-1 alpha (HIF-1α), may regulate PrP(C) expression and that these genes may be involved in prion-related neurodegenerative diseases. Hypoxic conditions are known to elicit cellular responses designed to improve cell survival through adaptive processes. Under normoxic conditions, a deferoxamine-mediated elevation of HIF-1α produced the same effect as hypoxia-inhibited neuron cell death. However, under hypoxic conditions, doxorubicin-suppressed HIF-1α attenuated the inhibitory effect on neuron cell death mediated by PrP (106-126). Knock-down of HIF-1α using lentiviral short hairpin (sh) RNA-induced downregulation of PrP(C) mRNA and protein expression under hypoxic conditions, and sensitized neuron cells to prion peptide-mediated cell death even in hypoxic conditions. In PrP(C) knockout hippocampal neuron cells, hypoxia increased the HIF-1α protein but the cells did not display the inhibitory effect of prion peptide-induced neuron cell death. Adenoviruses expressing the full length Prnp gene (Ad-Prnp) were utilized for overexpression of the Prnp gene in PrP(C) knockout hippocampal neuron cells. Adenoviral transfection of PrP(C) knockout cells with Prnp resulted in the inhibition of prion peptide-mediated cell death in these cells. This is the first report demonstrating that expression of normal PrP(C) is regulated by HIF-1α, and PrP(C) overexpression induced by hypoxia plays a pivotal role in hypoxic inhibition of prion peptide-induced neuron cell death. These results suggest that hypoxia-related genes, including HIF-1α, may be involved in the pathogenesis of prion-related diseases and as such may be a therapeutic target for prion-related neurodegenerative diseases.


Biochemical and Biophysical Research Communications | 2012

Sulforaphane induced adipolysis via hormone sensitive lipase activation, regulated by AMPK signaling pathway

Ju-Hee Lee; Myung-Hee Moon; Jae-Kyo Jeong; Yang-Gyu Park; You-Jin Lee; Jae-Won Seol; Sang-Youel Park

Sulforaphane, an aliphatic isothiocyanate derived from cruciferous vegetables, is known for its antidiabetic properties. The effects of sulforaphane on lipid metabolism in adipocytes are not clearly understood. Here, we investigated whether sulforaphane stimulates lipolysis. Mature adipocytes were incubated with sulforaphane for 24h and analyzed using a lipolysis assay which quantified glycerol released into the medium. We investigated gene expression of hormone-sensitive lipase (HSL), and levels of HSL phosphorylation and AMP-activated protein kinase on sulforaphane-mediated lipolysis in adipocytes. Sulforaphane promoted lipolysis and increased both HSL gene expression and HSL activation. Sulforaphane suppressed AMPK phosphorylation at Thr-172 in a dose-dependent manner, which was associated with a decrease in HSL phosphorylation at Ser-565, enhancing the phosphorylation of HSL Ser-563. Taken together, these results suggest that sulforaphane promotes lipolysis via hormone sensitive lipase activation mediated by decreasing AMPK signal activation in adipocytes.


Journal of Neurochemistry | 2010

Hypoxia protects neuronal cells from human prion protein fragment-induced apoptosis.

Jae-Suk Seo; Jae-Won Seol; Myung-Hee Moon; Jae-Kyo Jeong; You-Jin Lee; Sang-Youel Park

J. Neurochem. (2010) 112, 715–722.


Biochemical and Biophysical Research Communications | 2012

18β-Glycyrrhetinic acid inhibits adipogenic differentiation and stimulates lipolysis

Myung-Hee Moon; Jae-Kyo Jeong; You-Jin Lee; Jae-Won Seol; Dongchoon Ahn; In-Shik Kim; Sang-Youel Park

18β-Glycyrrhetinic acid (18β-GA) obtained from the herb liquorice has various pharmacological properties including anti-inflammatory and anti-bacterial activities. However, potential biological anti-obesity activities are unclear. In this study, novel biological activities of 18β-GA in the adipogenesis of 3T3-L1 preadipocytes and in lipolysis of differentiated adipocytes were identified. Mouse 3T3-L1 cells were used as an in vitro model of adipogenesis and lipolysis, using a mixture of insulin/dexamethasone/3-isobutyl-1-methylxanthine (IBMX) to induce differentiation. The amount of lipid droplet accumulation was determined by an AdipoRed assay. The expression of several adipogenic transcription factors and enzymes was investigated using real-time reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting. 18β-GA dose-dependently (1-40 μM) significantly decreased lipid accumulation in maturing preadipocytes. In 3T3-L1 preadipocytes, 10 μM of 18β-GA down-regulated the transcriptional levels of the peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α and adiponectin, which are markers of adipogenic differentiation via Akt phosphorylation. Also, in differentiated adipocytes, 18β-GA increased the level of glycerol release and up-regulated the mRNA of hormone-sensitive lipase, adipose TG lipase and perilipin, as well as the phosphorylation of hormone-sensitive lipase at Serine 563. The results indicate that 18β-GA alters fat mass by directly affecting adipogenesis in maturing preadipocytes and lipolysis in matured adipocytes. Thus, 18β-GA may be useful for the treatment of obesity.


International Journal of Molecular Medicine | 2012

Sphingosine-1-phosphate inhibits interleukin-1β-induced inflammation in human articular chondrocytes

Myung-Hee Moon; Jae-Kyo Jeong; You-Jin Lee; Jae-Won Seol; Sang-Youel Park

Sphingosine-1-phosphate (S1P) is a pluripotent lipid mediator that transmits signals through a family of G-protein-coupled receptors (GPCRs) to control diverse biological processes including inflammation and wound-healing. In this study, a novel biological activity of S1P in articular chondrocytes was identified. Human primary chondrocytes were cultured in a monolayer. Reverse transcription-polymerase chain reaction (RT-PCR) and western blotting were performed to detect genes and proteins involved in inflammation and cartilage degradation when human primary chondrocytes were stimulated by interleukin (IL)-1β. Matrix metalloproteinase (MMP)-2 and MMP-9 activity was evaluated by gelatin zymography. Glycosaminoglycan (GAG) degradation was evaluated using the dimethylene blue method. Prostaglandin E2 (PGE2) was measured by enzyme-linked immunosorbent assay (ELISA). By using the S1P1 receptor agonist and antagonist, we discovered the key role played by S1P1 in the S1P-dependent inhibition of IL-1β-induced inflammation in human chondrocytes. S1P dose-dependently inhibited IL-1β-induced NF-κB p65, cyclooxygenase (COX)-2, MMP-1, MMP-3, MMP-13 and MMP-14 mRNA expression in human chondrocytes and IL-1β-induced PGE2 synthesis and GAG degradation in human cartilage explants. W146, a known S1P1 receptor antagonist, inhibited the active form of NF-κB p65 and COX-2 expression induced by IL-1β. The anti-inflammatory action of the S1P1 receptor agonist SEW2871 was similar to that of S1P. This study demonstrates that S1P has anti-inflammatory effects on chondrocytes via the S1P1 receptor. Our data suggest that targeting S1P and S1P1 may be a potential therapy for arthritis.

Collaboration


Dive into the Jae-Kyo Jeong'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

Jae-Won Seol

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Myung-Hee Moon

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-Suk Seo

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Ji-Hong Moon

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Yang-Gyu Park

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Jackson

Kolling Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Seog-Jin Kang

Rural Development Administration

View shared research outputs
Researchain Logo
Decentralizing Knowledge