Network


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

Hotspot


Dive into the research topics where Deok-Bae Park is active.

Publication


Featured researches published by Deok-Bae Park.


Food and Chemical Toxicology | 2012

Effects of brown alga, Ecklonia cava on glucose and lipid metabolism in C57BL/KsJ-db/db mice, a model of type 2 diabetes mellitus.

Seung-Hong Lee; Kwan-Hee Min; Ji-Sook Han; Dae-Ho Lee; Deok-Bae Park; Won-Kyo Jung; Pyo-Jam Park; Byong-Tae Jeon; Se-Kwon Kim; You-Jin Jeon

Recently, there has been a growing interest in alternative therapies of marine algae for diabetes. Therefore, the anti-diabetic effects of brown alga, Ecklonia cava was investigated in type 2 diabetic animal. Male C57BL/KsJ-db/db (db/db) mice were divided into control, dieckol rich extract of E. cava (AG-dieckol), or rosiglitazone (RG) groups. The blood glucose, blood glycosylated hemoglobin levels, and plasma insulin levels were significantly lower in the AG-dieckol and RG groups than in the control db/db mice group, while glucose tolerance was significantly improved in the AG-dieckol group. AG-dieckol markedly lowered plasma and hepatic lipids concentration compared to the control db/db mice group. The antioxidant enzyme activities were significantly higher in the AG-dieckol group than in the control db/db mice group, yet its TBARS level was markedly lower compared to the RG group. With regard to hepatic glucose regulating enzyme activities, glucokinase activity was enhanced in the AG-dieckol group mice, while glucose-6-phosphatase and phosphoenolpyruvate carboxykinase activities in the AG-dieckol group mice were significantly lowered than those in the control db/db mice group. These results suggest that AG-dieckol exert an anti-diabetic effect in type 2 diabetic mice by improving the glucose and lipid metabolism and antioxidant enzymes.


Experimental and Molecular Medicine | 2003

Arsenic trioxide-induced apoptosis is independent of stress-responsive signaling pathways but sensitive to inhibition of inducible nitric oxide synthase in HepG2 cells.

Shin-Hae Kang; Jihoon Song; Hee-Kyoung Kang; Ji-Hoon Kang; Se-Jae Kim; Hyun-Wook Kang; Young-Ki Lee; Deok-Bae Park

Arsenic trioxide (As2O3) has been found to be remarkably effective in the treatment of patients with acute promyelocytic leukemia (APL). Although evidences for the proapoptotic activity of As2O3 have been suggested in leukemic and other solid cancer cells, the nature of intracellular mechanisms is far from clear. In the present study, we investigated As2O3 affect on the stress-responsive signaling pathways and pretreatment with antioxidants using HepG2 cells. When treated with micromolar concentrations of As2O3, HepG2 cells became highly apoptotic paralleled with activation of caspase-3 and members of mitogen-activated protein kinases (MAPKs) including extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) but not p38 MAP kinase. However, inhibition of each kinase activity failed to inhibit apoptosis by As2O3. Addition of n-acetyl cysteine (NAC) or diphenyleneiodonium (DPI) effectively protected cells from apoptosis and significantly lowered As2O3-induced activation of caspase-3. However, neither NAC nor DPI was able to effect ERK or JNK activation induced by As2O3. Guanidinoethyldisulfide dihydrochloride (GED) and 2-ethyl- 2-thiopseudourea (ETU), known inhibitors of the inducible nitric oxide synthase (iNOS), also suppressed the apoptotic activity of As2O3. These results suggest that As2O3 induces caspase-mediated apoptosis involving a mechanism generating oxidative stress. However, activation of some stress- responsive signaling pathways by As2O3 may not be the major determinant in the course of apoptotic processes.


Neurochemical Research | 2011

6-Hydroxydopamine-Induced PC12 Cell Death is Mediated by MEF2D Down-regulation

Min-Kyoung Kim; Sang-Cheol Kim; Jung-Il Kang; Jae-Hee Hyun; Hye-Jin Boo; Su-Yong Eun; Deok-Bae Park; Eun-Sook Yoo; Hee-Kyoung Kang; Ji-Hoon Kang

Recently, it was reported that in a 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model, neuronal cell death is associated with the cdk5-mediated hyperphosphorylation of myocyte enhancer factor 2 (MEF2), a transcription factor that is critically required for neuronal survival. In the present study, we investigated the possible involvement of cdk5-mediated MEF2D down-regulation on 6-hydroxydopamine (6-OHDA)-induced PC12 cell death. 6-OHDA was found to significantly increase nitric oxide (NO) production and to induce apoptosis in a time-dependent manner in PC12 cells. Furthermore, 6-OHDA was found to markedly reduce MEF2D levels under conditions that could induce PC12 cell apoptosis. In addition, PC12 cell death and MEF2D degradation by 6-OHDA were prevented by the cdk5 inhibitor roscovitine, but roscovitine could not restore the 6-OHDA-induced inactivation of Akt. These results suggest that the cell death and MEF2D degradation caused by 6-OHDA are dependent on cdk5 activity. On the other hand, roscovitine enhanced the 6-OHDA-induced activations of ERK1/2 and JNK, but reduced the 6-OHDA-induced activation of p38. These results suggest that PC12 cell death by 6-OHDA appears to be regulated by the down-regulation of MEF2D via some interaction between cdk5 and MAP kinase.


Biochemical and Biophysical Research Communications | 2012

p-Synephrine stimulates glucose consumption via AMPK in L6 skeletal muscle cells

Na-Young Hong; Zhi-Gang Cui; Hee-Kyoung Kang; Dae-Ho Lee; Young-Ki Lee; Deok-Bae Park

Interest in p-synephrine, the primary protoalkaloid in the extract of bitter orange and other citrus species, has increased due to its various pharmacological effects and related adverse effects. The lipolytic activity of p-synephrine has been repeatedly revealed by in vitro and in vivo studies and p-synephrine is currently marketed as a dietary supplement for weight loss. The present study investigated the effect of p-synephrine on glucose consumption and its action mechanism in L6 skeletal muscle cells. Treatment of L6 skeletal muscle cells with p-synephrine (0-100μM) did not affect cell viability and increased basal glucose consumption up to 50% over the control in a dose-dependent manner. The basal- or insulin-stimulated lactic acid production as well as glucose consumption was significantly increased by the addition of p-synephrine. p-Synephrine stimulated the phosphorylation of AMPK but not of Akt. p-Synephrine-induced glucose consumption was sensitive to the inhibition of AMPK but not to the inhibition of PI3 kinase. p-Synephrine also stimulated the translocation of Glut4 from the cytoplasm to the plasma membrane; this stimulation was suppressed by the inhibition of AMPK, but not of PI3 kinase. Taken together, p-synephrine can stimulate glucose consumption (Glut4-dependent glucose uptake) by stimulating AMPK activity, regardless of insulin-stimulated PI3 kinase-Akt activity in L6 skeletal muscle cells.


Diabetes & Metabolism Journal | 2015

Metformin Promotes Apoptosis but Suppresses Autophagy in Glucose-Deprived H4IIE Hepatocellular Carcinoma Cells

Deok-Bae Park

Background Metformin, a well-known anti-diabetic drug, has gained interest due to its association with the reduction of the prevalence of cancer in patients with type 2 diabetes and the anti-proliferative effect of metformin in several cancer cells. Here, we investigated the anti-proliferative effect of metformin with respect to apoptosis and autophagy in H4IIE hepatocellular carcinoma cells. Methods H4IIE rat cells were treated with metformin in glucose-free medium for 24 hours and were then subjected to experiments examining the onset of apoptosis and/or autophagy as well as the related signaling pathways. Results When H4IIE cells were incubated in glucose-free media for 24 hours, metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) reduced the viability of cells. Inhibition of AMP-activated protein kinase (AMPK) by compound C significantly blocked cell death induced by metformin or AICAR. Pro-apoptotic events (nuclear condensation, hydrolysis of intact poly ADP ribose polymerase and caspase-3) were stimulated by metformin and then suppressed by compound C. Interestingly, the formation of acidic intracellular vesicles, a marker of autophagy, was stimulated by compound C. Although the deprivation of amino acids in culture media also induced apoptosis, neither metformin nor compound C affected cell viability. The expression levels of all of the autophagy-related proteins examined decreased with metformin, and two proteins (light chain 3 and beclin-1) were sensitive to compound C. Among the tested inhibitors against MAP kinases and phosphatidylinositol-3-kinase/mammalian target of rapamycin, SB202190 (against p38MAP kinase) significantly interrupted the effects of metformin. Conclusion Our data suggest that metformin induces apoptosis, but suppresses autophagy, in hepatocellular carcinoma cells via signaling pathways, including AMPK and p38 mitogen-activated protein kinase.


Phytotherapy Research | 2013

Mild mitochondrial depolarization is involved in a neuroprotective mechanism of Citrus sunki peel extract.

Jinji Wu; Yanji Cui; Yoon-Sil Yang; Sung-Cherl Jung; Jin Won Hyun; Young-Hee Maeng; Deok-Bae Park; Sun-Ryung Lee; Se-Jae Kim; Su-Yong Eun

Mitochondrial membrane potential (∆Ψm) contributes to determining a driving force for calcium to enter the mitochondria. It has been demonstrated that even a small mitochondrial depolarization is sufficient to prevent mitochondrial calcium overload and the subsequent apoptosis. Therefore, mild mitochondrial depolarization has been recently evaluated as a novel mechanism of neuroprotection via inhibiting neurotoxic mitochondrial calcium overload during neuronal insults. In the present study, using both real‐time recording and flow cytometric analyses of ∆Ψm, we demonstrated that ethanolic peel extract of Citrus sunki Hort. ex Tanaka (CPE) and its active compounds are capable of inducing a mild mitochondrial depolarization. Polymethoxylated flavones such as nobiletin and tangeretin were found as the active compounds responsible for CPE effects on ∆Ψm. Neuronal viability was significantly increased in a dose‐dependent manner by CPE treatment in H2O2‐stimulated HT‐22 cells as an in vitro neuronal insult model. CPE treatment significantly inhibited H2O2‐induced apoptotic processes such as chromatin condensation, caspase 3 activation and anti‐poly (ADP‐ribose) polymerase (PARP) cleavage. CPE treatment significantly blocked mitochondrial calcium overload in H2O2‐stimulated HT‐22 neurons as indicated by rhod‐2 acetoxymethyl ester. Taken together, our findings suggest that CPE and its active compounds may be considered as promising neuroprotective agents via inducing a mild mitochondrial depolarization. Copyright


Development & Reproduction | 2017

Metformin Synergistically Potentiates the Antitumor Effects of Imatinib in Colorectal Cancer Cells

Jaeryun Lee; Deok-Bae Park; Youngki Lee

ABSTRACT Metformin is the most commonly prescribed anti-diabetic drug with relatively minor side effect. Substantial evidence has suggested that metformin is associated with decreased cancer risk and anticancer activity against diverse cancer cells. The tyrosine kinase inhibitor imatinib has shown powerful activity for treatment of chronic myeloid leukemia and also induces growth arrest and apoptosis in colorectal cancer cells. In this study, we tested the combination of imatinib and metformin against HCT15 colorectal cancer cells for effects on cell viability, cell cycle and autophagy. Our data show that metformin synergistically enhances the imatinib cytotoxicity in HCT15 cells as indicated by combination and drug reduction indices. We also demonstrate that the combination causes synergistic down-regulation of pERK, cell cycle arrest in S and G2/M phases via reduction of cyclin B1 level. Moreover, the combination resulted in autophagy induction as revealed by increased acidic vesicular organelles and cleaved form of LC3-II. Inhibition of autophagic process by chloroquine led to decreased cell viability, suggesting that induction of autophagy seems to play a cell protective role that may act against anticancer effects. In conclusion, our present data suggest that metformin in combination with imatinib might be a promising therapeutic option in colorectal cancer.


Genes & Genomics | 2016

Resveratrol suppresses inflammatory responses and improves glucose uptake in adipocytes interacted with macrophages

Hye-Sun Shin; Seong-Il Kang; Deok-Bae Park; Se-Jae Kim

Obesity is associated with inflammatory status and linked with metabolic syndrome. Interaction between adipocytes and macrophages aggravates inflammation and leads to insulin resistance in adipocytes. Resveratrol improved reportedly obesity-related inflammatory responses, but the effects of resveratrol on the production of inflammatory mediators and glucose metabolism in inflamed adipose tissue is not completely known. In this study, we investigated the effects of resveratrol on inflammatory change and insulin resistance in the coculture of hypertrophied 3T3-L1 adipocytes and RAW 264.7 macrophages. Resveratrol decreased nitric oxide production and the expression of interleukin (IL)-6, IL-1β, tumor necrosis factor-α, inducible nitric oxide synthesis, and cyclooxygenase-2 in the coculture. Resveratrol increased glucose uptake by stimulating the phosphorylation of IRS-1 and AKT in the coculture. These results support that resveratrol have beneficial effect on inflammation and insulin resistance in inflamed adipose tissue.


European Journal of Pharmacology | 2015

Effects of dihydrotestosterone on rat dermal papilla cells in vitro.

Jung-Il Kang; Sang-Cheol Kim; Min-Kyoung Kim; Hye-Jin Boo; Eun-Ji Kim; Guang-Jin Im; Young Ho Kim; Jin-Won Hyun; Ji-Hoon Kang; Young-Sang Koh; Deok-Bae Park; Eun-Sook Yoo; Hee-Kyoung Kang

Androgenetic alopecia involves the action of dihydrotestosterone (DHT) on dermal papilla cells (DPCs) that line the base of the hair follicle. However, the mechanism of DHT action is not completely understood. The effects of DHT on DPCs, regulatory cells that function in follicle growth and the hair cycle, were examined in immortalized cells derived from rat vibrissa follicles. DHT did not affect the proliferation of immortalized DPCs. However, flow cytometry analysis revealed that DHT increased cell-cycle arrest in these cells, which was accompanied by an increase in the p27(kip1) level and by decreases in cyclin E, cyclin D1, and cyclin-dependent kinase 2 levels. DHT treatment resulted in the phosphorylation and nuclear translocation of Smad2/3, a mediator of the transforming growth factor-β (TGF-β) signaling pathway, which leads to the catagen phase of the hair cycle. DHT also induced the phosphorylation and nuclear translocation of heat shock protein 27 (HSP27). Moreover, DHT decreased the levels of total and nuclear β-catenin, an important regulator of hair growth and proliferation, while lithium chloride, a glycogen synthase kinase-3β inhibitor, attenuated the DHT-induced downregulation of the β-catenin level. On the other hand, DHT increased the phosphorylation of mammalian target of rapamycin (mTOR), a regulator of proliferation, in immortalized DPCs. These results illustrate that DHT could shorten the duration of the hair growth cycle by initiating cell-cycle arrest, downregulating the β-catenin level, and upregulating the TGF-β/Smad and HSP27 level, whereas activation of mTOR by DHT could attenuate the inhibition of hair growth cycle in immortalized DPCs.


Development & Reproduction | 2016

Blockage of Autophagy Rescues the Dual PI3K/mTOR Inhibitor BEZ235- induced Growth Inhibition of Colorectal Cancer Cells

Iljoong Oh; Hyunchul Cho; Yonghoon Lee; Minseok Cheon; Deok-Bae Park; Youngki Lee

Molecular targeting for the altered signaling pathways has been proven to be effective for the treatment ofmany types of human cancer, including colorectal cancer (CRC). The dual phosphatidylinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) inhibitor BEZ235 has shown to exhibit potent antitumor activity against solid tumors. Autophagy is a cellular lysosomal catabolic process to maintain metabolic homeostasis, which has been known to be induced in response to many therapeutic agents in cancer cells. This process is negatively regulated by mTOR and often acts as prosurvival or prodeath mechanism following cancer therapeutics. The current study was designed to investigate the antiproliferation activity of BEZ235 and to evaluate the role of autophagy induced by BEZ235 using HCT15 CRC cells bearing ras oncogene mutation. We found that BEZ235 decreases cell viability, which was mostly dependent on G1 arrest of cell cycle via suppression of cyclin A expression. BEZ235 affects PI3K/Akt/mTOR signaling pathway by increasing the phosphorylation of AKT at Ser473 and RAS/RAF/MEK/ERK pathway by decreasing the phosphorylation of ERK at Tyr204. BEZ235 also stimulated autophagy induction as evidenced by the increased expression of LC3-II and abundant acidic vesicular organelles (AVOs) in the cytoplasm. In addition, the combination of BEZ235 with autophagy inhibitor chloroquine, a known antagonist of autophagy, counteracted the antiproliferation effect of BEZ235. Thus, our study indicates that autophagy induced in response to BEZ235 treatment appears to act as cell death mechanism in HCT15 CRC cells.

Collaboration


Dive into the Deok-Bae Park's collaboration.

Top Co-Authors

Avatar

Hee-Kyoung Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Se-Jae Kim

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Jihoon Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Shin-Hae Kang

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Dae-Ho Lee

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Young-Ki Lee

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Eun-Sook Yoo

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Hye-Jin Boo

Jeju National University

View shared research outputs
Top Co-Authors

Avatar

Jung-Il Kang

Jeju National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge