Network


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

Hotspot


Dive into the research topics where Dae-Seop Shin is active.

Publication


Featured researches published by Dae-Seop Shin.


Cancer Research | 2009

Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells.

Dae-Seop Shin; Hye-Nan Kim; Ki Deok Shin; Young Ju Yoon; Seung-Jun Kim; Dong Cho Han; Byoung-Mog Kwon

Because signal transducer and activator of transcription 3 (STAT3) is constitutively activated in most human solid tumors and is involved in the proliferation, angiogenesis, immune evasion, and antiapoptosis of cancer cells, researchers have focused on STAT3 as a target for cancer therapy. We screened for natural compounds that inhibit the activity of STAT3 using a dual-luciferase assay. Cryptotanshinone was identified as a potent STAT3 inhibitor. Cryptotanshinone rapidly inhibited STAT3 Tyr705 phosphorylation in DU145 prostate cancer cells and the growth of the cells through 96 hours of the treatment. Inhibition of STAT3 Tyr705 phosphorylation in DU145 cells decreased the expression of STAT3 downstream target proteins such as cyclin D1, survivin, and Bcl-xL. To investigate the cryptotanshinone inhibitory mechanism in DU145 cells, we analyzed proteins upstream of STAT3. Although phosphorylation of Janus-activated kinase (JAK) 2 was inhibited by 7 micromol/L cryptotanshinone at 24 hours, inhibition of STAT3 Tyr705 phosphorylation occurred within 30 minutes and the activity of the other proteins was not affected. These results suggest that inhibition of STAT3 phosphorylation is caused by a JAK2-independent mechanism, with suppression of JAK2 phosphorylation as a secondary effect of cryptotanshinone treatment. Continuing experiments revealed the possibility that cryptotanshinone might directly bind to STAT3 molecules. Cryptotanshinone was colocalized with STAT3 molecules in the cytoplasm and inhibited the formation of STAT3 dimers. Computational modeling showed that cryptotanshinone could bind to the SH2 domain of STAT3. These results suggest that cryptotanshinone is a potent anticancer agent targeting the activation STAT3 protein. It is the first report that cryptotanshinone has antitumor activity through the inhibition of STAT3.


Journal of Biological Chemistry | 2005

Blocking tumor cell migration and invasion with biphenyl isoxazole derivative KRIBB3, a synthetic molecule that inhibits Hsp27 phosphorylation.

Ki Deok Shin; Mi-Young Lee; Dae-Seop Shin; Sangku Lee; Kwang-Hee Son; Sukhoon Koh; Young-Ki Paik; Byoung-Mog Kwon; Dong Cho Han

Cell migration is a prerequisite for cancer invasion and metastasis, suggesting cell motility as a potential therapeutic target for cancer treatment. A synthetic library was screened to identify inhibitors of tumor cell migration. From this, we discovered that CAC-1098 (aurintricarboxylic acid) and CBI-0997 (5-(2,4-dimethoxy-5-ethylphenyl)-4-(4-bromophenyl) isoxazole) inhibited migration of MDA-MB-231 cells with IC50 = 5 and 50 nm, respectively. We synthesized KRIBB3 (5-(5-ethyl-2-hydroxy-4-methoxyphenyl)-4-(4-methoxyphenyl) isoxazole) by replacing the bromide group of CBI-0997 with a methoxyl group. Like CBI-0997, KRIBB3 has anti-migratory and anti-invasive activities in MDA-MB-231 cells. Because KRIBB3 has a better drug-like structure, we focused our effort on further understanding its anti-migratory mechanism. Biotinyl-KRIBB3 was synthesized as an affinity probe for identification of KRIBB3-binding proteins. Using affinity chromatography, we identified Hsp27 as a target protein of KRIBB3 in vitro. Treatment of MDA-MB-231 cells with phorbol 12-myristate 13-acetate induced protein kinase C-dependent phosphorylation of Hsp27 and tumor cell migration. In contrast, treatment of MDA-MB-231 cells with KRIBB3 blocked phorbol 12-myristate 13-acetate-induced phosphorylation of Hsp27 and tumor cell migration. Furthermore, overexpression of Hsp27 antagonized the inhibitory effect of KRIBB3 on tumor cell invasion, and knockdown of Hsp27 using small interfering RNA inhibited tumor cell migration. Overall, our results demonstrate that KRIBB3 inhibits tumor cell migration and invasion by blocking protein kinase C-dependent phosphorylation of Hsp27 through its direct binding to Hsp27.


Journal of Biological Chemistry | 2011

KRIBB11 Inhibits HSP70 Synthesis through Inhibition of Heat Shock Factor 1 Function by Impairing the Recruitment of Positive Transcription Elongation Factor b to the hsp70 Promoter

Young Ju Yoon; Joo Ae Kim; Ki Deok Shin; Dae-Seop Shin; Young Min Han; Yu Jin Lee; Jin Soo Lee; Byoung-Mog Kwon; Dong Cho Han

Heat shock factor 1 (HSF1) is the master switch for heat shock protein (HSP) expression in eukaryotes. A synthetic chemical library was screened to identify inhibitors of HSF1 using a luciferase reporter under the control of a heat shock element. A compound named KRIBB11 (N2-(1H-indazole-5-yl)-N6-methyl-3-nitropyridine-2,6-diamine) was identified for its activity in abolishing the heat shock-induced luciferase activity with an IC50 of 1.2 μmol/liter. When the cells were exposed to heat shock in the presence of KRIBB11, the induction of HSF1 downstream target proteins such as HSP27 and HSP70 was blocked. In addition, treatment of HCT-116 cells with KRIBB11 induced growth arrest and apoptosis. Markers of apoptosis, such as cleaved poly(ADP-ribose) polymerase, were detected after KRIBB11 treatment. Biotinyl-KRIBB11 was synthesized as an affinity probe for the identification of KRIBB11 target proteins. Using affinity chromatography and competition assays, KRIBB11 was shown to associate with HSF1 in vitro. Chromatin immunoprecipitation analysis showed that KRIBB11 inhibited HSF1-dependent recruitment of p-TEFb (positive transcription elongation factor b) to the hsp70 promoter. Finally, intraperitoneal treatment of nude mice with KRIBB11 at 50 mg/kg resulted in a 47.4% (p < 0.05) inhibition of tumor growth without body weight loss. Immunoblotting assays showed that the expression of HSP70 was lower in KRIBB11-treated tumor tissue than in control tissues. Because HSPs are expressed at high levels in a wide range of tumors, these results strengthen the rationale for targeting HSF1 in cancer therapy.


International Journal of Cancer | 2012

Phosphatase of regenerating liver-3 promotes migration and invasion by upregulating matrix metalloproteinases-7 in human colorectal cancer cells

Su-Kyung Lee; Young-Min Han; Jieun Yun; Chang Woo Lee; Dae-Seop Shin; Young-Ran Ha; Jina Kim; Jong Sung Koh; Su-Hyung Hong; Dong Cho Han; Byoung-Mog Kwon

Phosphatase of regenerating liver (PRL)‐3, a member of a subgroup of protein tyrosine phosphatases that can stimulate the degradation of the extracellular matrix, is over‐expressed in metastatic colorectal cancer (CRC) relative to primary tumors. To determine whether PRL‐3‐induced enhancement of migration and invasion is dependent on the expression of matrix metalloproteinases (MMPs), PRL‐3 was expressed in DLD‐1 human CRC cells. The motility, migration and invasion characteristics of the cells were examined, and metastasis to the lung was confirmed in a nude mouse using PRL‐3‐overexpressing DLD‐1 cells [DLD‐1 (PRL‐3)]. Migration and invasion of the cells were inhibited by phosphatase and farnesyltransferase inhibitors. Expression of MMPs was enhanced 3‐ to 10‐fold in comparison to control cells, and migration and invasion were partially inhibited by small interfering RNA (siRNA) knockdown of MMP‐2, ‐13 or ‐14. Importantly, siRNA knockdown of MMP‐7 completely inhibited the migration and invasion of DLD‐1 (PRL‐3) cells, whereas overexpression of MMP‐7 increased migration. The expression of MMP‐7 was also downregulated by phosphatase and farnesyltransferase inhibitors. It was found that PRL‐3 induced MMP‐7 through oncogenic pathways including PI3K/AKT and ERK and that there is a relationship between the expression of PRL‐3 and MMP‐7 in human tumor cell lines. The expression of MMP‐13 and ‐14 was very sensitive to the inhibition of farnesyltransferase; however, the migration and invasion of DLD‐1 (PRL‐3) cells did not strongly depend on the expression of MMP‐13 or ‐14. These results suggest that the migration and invasion of PRL‐3‐expressing CRC cells depends primarily on the expression of MMP‐7.


Biochemical Pharmacology | 2015

Sugiol inhibits STAT3 activity via regulation of transketolase and ROS-mediated ERK activation in DU145 prostate carcinoma cells

Seung-Nam Jung; Dae-Seop Shin; Hye-Nan Kim; Yoon Jung Jeon; Jieun Yun; Yu-Jin Lee; Jong Soon Kang; Dong Cho Han; Byoung-Mog Kwon

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in various human cancers and has been used as a therapeutic target for tumors. This study screened natural products to identify compounds that inhibit STAT3 activity using a STAT3-dependent luciferase reporter system. Sugiol was identified as a compound that decreased luciferase activity in a dose-dependent manner. Sugiol specifically inhibited STAT3 phosphorylation at Tyr-705 in DU145 prostate cells, and this inhibition was independent of the STAT3 upstream kinase. Sugiol induced cell cycle arrest and decreased the expression levels of STAT3 target genes, such as cyclin D1, cyclin A, and survivin. Notably, we observed that sugiol interacted with transketolase, an enzyme in central metabolism, and increased ROS levels leading to the activation of ERK, which inhibits STAT3 activity. The protein phosphatase MEG2 was also responsible for sugiol-induced STAT3 dephosphorylation. The inhibitory effect of sugiol on cell growth was confirmed using the DU145 mouse xenograft model. We propose that sugiol inhibits STAT3 activity through a mechanism that involves the inhibition of transketolase, which leads to increased ROS levels and MEG2 activation in DU145 cells. Therefore, sugiol is the first compound regulating STAT3 activity via modulation of cancer metabolic pathway and we suggest the use of sugiol as an inhibitor of the STAT3 pathway for the treatment of human solid tumors with activated STAT3.


Bioorganic & Medicinal Chemistry Letters | 2011

2-Hydroxycurcuminoid induces apoptosis of human tumor cells through the reactive oxygen species–mitochondria pathway

Young-Min Han; Dae-Seop Shin; Yu-Jin Lee; Ismail Ahmed Ismail; Su-Hyung Hong; Dong Cho Han; Byoung-Mog Kwon

2-Hydroxycinnamaldehyde (HCA) and curcumin have been reported to have antitumor effects against various human tumor cells in vitro and in vivo by generation of ROS. Aldehyde-free HCA analogs were synthesized based on the structure of curcumin, which we have called 2-hydroxycurcuminoids. The hydroxyl group of curcuminoids enhances the ability to generate ROS. 2-Hydroxycurcuminoid (HCC-7) strongly inhibited the growth of SW620 colon tumor cells with a GI(50) value of 7μM, while the parent compounds, HCA and curcumin, displayed GI(50) values of 12 and 30μM, respectively. HCC-7 was found to induce apoptosis through the reactive oxygen species-mitochondria pathway and cell cycle arrest at G2/M phase.


Bioorganic & Medicinal Chemistry | 2011

Cosmomycin C inhibits signal transducer and activator of transcription 3 (STAT3) pathways in MDA-MB-468 breast cancer cell.

Ji-Hoon Kim; Yu-Jin Lee; Dae-Seop Shin; Sun-Hee Jeon; Kwang-Hee Son; Dong Cho Han; Seung-Nam Jung; Tae-Kwang Oh; Byoung-Mog Kwon

The signal transducer and activator of transcription 3 (STAT3) is constitutively activated in cancer cells. Therefore, blocking the aberrant activity of STAT3 in tumor cells is a validated therapeutic strategy. To discover novel inhibitors of STAT3 activity, we screened against microbial natural products using a dual-luciferase assay. Using the microbial metabolome library, we identified cosmomycin C (CosC), which was isolated from the mycelium extract of Streptomyces sp. KCTC19769, as a STAT3 pathway inhibitor. CosC inhibited STAT3 (Tyr705) phosphorylation and subsequent nuclear translocation in MDA-MB-468 breast cancer cells. CosC-mediated inhibition of STAT3 signaling pathway was confirmed by suppressed expression of STAT3 downstream target proteins including cyclin D1, Bcl-xL, survivin, Mcl-1, and VEGF in CosC-treated MDA-MB-468 cells. Flow cytometry showed that CosC caused accumulation in the G(0)-G(1) phase of the cell cycle and induced apoptosis via PARP cleavage and caspase-3 activation. Based on these findings, CosC may be a potential candidate for modulation of STAT3 pathway.


Biochemical Pharmacology | 2007

Apoptosis induction of 2'-hydroxycinnamaldehyde as a proteasome inhibitor is associated with ER stress and mitochondrial perturbation in cancer cells.

Su Hyung Hong; Jina Kim; Jung-Min Kim; Soyoung Lee; Dae-Seop Shin; Kwang-Hee Son; Dong Cho Han; Young Kwan Sung; Byoung-Mog Kwon


Life Sciences | 2006

Dehydrotrametenolic acid selectively inhibits the growth of H-ras transformed rat2 cells and induces apoptosis through caspase-3 pathway

Hyun-Mi Kang; Su-Kyung Lee; Dae-Seop Shin; Mi-Young Lee; Dong Cho Han; Nam-In Baek; Kwang-Hee Son; Byoung-Mog Kwon


Bioorganic & Medicinal Chemistry | 2006

Synthesis and biological evaluation of dimeric cinnamaldehydes as potent antitumor agents

Dae-Seop Shin; Jong Han Kim; Su-Kyung Lee; Dong Cho Han; Kwang-Hee Son; Hwan-Mook Kim; Hyae-Gyeong Cheon; Kwang-Rok Kim; Nack-Do Sung; Seung Jae Lee; Sung Kwon Kang; Byoung-Mog Kwon

Collaboration


Dive into the Dae-Seop Shin's collaboration.

Top Co-Authors

Avatar

Byoung-Mog Kwon

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Dong Cho Han

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Kwang-Hee Son

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Hye-Nan Kim

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Ki Deok Shin

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Su-Kyung Lee

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Young-Min Han

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Chang Woo Lee

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Jieun Yun

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Young Ju Yoon

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Researchain Logo
Decentralizing Knowledge