Network


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

Hotspot


Dive into the research topics where Soonduck Lee is active.

Publication


Featured researches published by Soonduck Lee.


Cell Death & Differentiation | 2014

p34 is a novel regulator of the oncogenic behavior of NEDD4-1 and PTEN

Sujeong Hong; Moon Jh; Kim Js; Shin Js; Jung Ka; Won-Keun Lee; Jeong Sy; Hwang Jj; Soonduck Lee; Suh Ya; Ick Young Kim; Nam Ky; Han S; Kim Je; Kim Kp; Hong Ys; Lee Jl; Wang-Jae Lee; Choi Ek; Lee Js; Dong-Hoon Jin; Taehee Kim

PTEN is one of the most frequently mutated or deleted tumor suppressors in human cancers. NEDD4-1 was recently identified as the E3 ubiquitin ligase for PTEN; however, a number of important questions remain regarding the role of ubiquitination in regulating PTEN function and the mechanisms by which PTEN ubiquitination is regulated. In the present study, we demonstrated that p34, which was identified as a binding partner of NEDD4-1, controls PTEN ubiquitination by regulating NEDD4-1 protein stability. p34 interacts with the WW1 domain of NEDD4-1, an interaction that enhances NEDD4-1 stability. Expression of p34 promotes PTEN poly-ubiquitination, leading to PTEN protein degradation, whereas p34 knockdown results in PTEN mono-ubiquitination. Notably, an inverse correlation between PTEN and p34/NEDD4-1 levels was confirmed in tumor samples from colon cancer patients. Thus, p34 acts as a key regulator of the oncogenic behavior of NEDD4-1 and PTEN.


International Journal of Oncology | 2013

Oncogenic function of p34SEI-1 via NEDD4‑1‑mediated PTEN ubiquitination/degradation and activation of the PI3K/AKT pathway

Samil Jung; Chengping Li; Dongjun Jeong; Soonduck Lee; Jiyeon Ohk; Meeyoung Park; Songyi Han; Jingjing Duan; Changjin Kim; Young Yang; Keun-Il Kim; Jong-Seok Lim; Young-Sook Kang; Myeong-Sok Lee

A 34-KD protein encoded by the SEI-1 gene (p34(SEI‑1)), is a relatively recently discovered oncoprotein that has multiple important biological functions. Our data show that p34(SEI-1) enhances cancer cell survival and promotes tumorigenesis by downregulating the tumor suppressor PTEN, a negative regulator of the PI3K/AKT signaling pathway, and therefore activating the PI3K/AKT signaling pathway. In this process, p34(SEI-1) positively affects NEDD4-1 gene expression both at the transcriptional and protein levels. Furthermore, the expression levels of p34(SEI-1) and NEDD4-1 were found to be coordinated in tumor tissues obtained from patients with breast cancer. We also show that p34(SEI-1) affects the subcellular localization of PTEN.


Cancer Research | 2009

p34SEI-1 Inhibits Apoptosis through the Stabilization of the X-Linked Inhibitor of Apoptosis Protein: p34SEI-1 as a Novel Target for Anti–Breast Cancer Strategies

Seungwoo Hong; Chang-Jae Kim; Won-Sang Park; Jae-Sik Shin; Soonduck Lee; Seong-Gyu Ko; Samil Jung; In-Chul Park; Sungkwan An; Won-Keun Lee; Wang-Jae Lee; Dong-Hoon Jin; Myeong-Sok Lee

The p34(SEI-1) protein exerts oncogenic effects via regulation of the cell cycle, which occurs through a direct interaction with cyclin-dependent kinase 4. Such regulation can increase the survival of various types of tumor cells. Here, we show that the antiapoptotic function of p34(SEI-1) increases tumor cell survival by protecting the X-linked inhibitor of apoptosis protein (XIAP) from degradation. Our findings show that p34(SEI-1) inhibits apoptosis. This antiapoptotic effect was eliminated by the suppression of p34(SEI-1) expression. We also determined that direct binding of p34(SEI-1) to the BIR2 domain prevents ubiquitination of XIAP. Interestingly, p34(SEI-1) expression is absent or weak in normal tissues but is strongly expressed in tissues obtained from patients with breast cancer. Furthermore, the expression levels of p34(SEI-1) and XIAP seem to be coordinated in human breast cancer cell lines and tumor tissues. Thus, our findings reveal that p34(SEI-1) uses a novel apoptosis-inhibiting mechanism to stabilize XIAP.


Free Radical Biology and Medicine | 2012

Enhanced antitumor activity of vitamin C via p53 in Cancer cells

Jinsun Kim; Soonduck Lee; Boogi Chang; Dong-Hoon Jin; Samil Jung; Meeyoung Park; Youngsoo Han; Young Yang; Keun Il Kim; Jong-Seok Lim; Young-Sook Kang; Myeong-Sok Lee

Ascorbate is an important natural antioxidant that can selectively kill cancer cells at pharmacological concentrations. Despite its benefit, it is quite difficult to predict the antitumor effects of ascorbate, because the relative cytotoxicity of ascorbate differs between cancer cell lines. Therefore, it is essential to examine the basis for this fundamental disagreement. Because p53 is activated by DNA-damaging stress and then regulates various cellular conditions, we hypothesized that p53 can sensitize cancer cells to ascorbate. Using isogenic cancer cells, we observed that the presence of p53 can affect ascorbate cytotoxicity, and also reactivation of p53 can make cancer cells sensitive to ascorbate. p53-dependent enhancement of ascorbate cytotoxicity is caused by increased reactive oxygen species generation via a differentially regulated p53 transcriptional network. We also found that transcriptionally activated p53 was derived from MDM2 ubiquitination by ascorbate and subsequently its signaling network renders cancer cells more susceptible to oxidative stress. Similar to the p53 effect on in vitro ascorbate cytotoxicity, inhibition of tumor growth is also stronger in p53-expressing tumors than in p53-deficient ones in vivo. This is the first observation that ascorbate cytotoxicity is positively related to p53 expression, activating its transcriptional network to worsen intracellular oxidative stress and consequently enhancing its cytotoxicity. Based on our study, reactivation of p53 may help to achieve more consistent cytotoxic effects of ascorbate in cancer therapies.


Oncotarget | 2015

TRIP-Br1 oncoprotein inhibits autophagy, apoptosis, and necroptosis under nutrient/serum-deprived condition

Samil Jung; Chengping Li; Jingjing Duan; Soonduck Lee; Kyeri Kim; Yeonji Park; Young Yang; Keun-Il Kim; Jong-Seok Lim; Chung-Il Cheon; Young-Sook Kang; Myeong-Sok Lee

TRIP-Br1 oncogenic protein has been shown to have multiple biological functions in cells. In this study, we demonstrate that TRIP-Br1 functions as an oncoprotein by inhibiting autophagy, apoptosis, and necroptosis of cancer cells and eventually helping them to survive under the nutrient/serum starved condition. TRIP-Br1 expression level was significantly increased in conditions with low levels of nutrients. Nutrient depleted conditions were induced by culturing cancer cells until they were overcrowded with high cell density or in media deprived of glucose, amino acids, or serum. Among them, serum starvation significantly enhanced the expression of TRIP-Br1 only in all tested breast cancer cell lines (MCF7, MDA-MB-231, T47D, MDA-MB-435, Hs578D, BT549, and MDA-MB-435) but not in the three normal cell lines (MCF10A, HfCH8, and NIH3T3). As compared with the control cells, the introduction of TRIP-Br1 silencing siRNA into MCF7 and MDA-MB-231 cells accelerated cell death by inducing apoptosis and necroptosis. In this process, TRIP-Br1 confers resistance to serum starvation-induced cell deaths by stabilizing the XIAP protein and inhibiting cellular ROS production. Moreover, our data also show that the intracellular increase of TRIP-Br1 protein resulting from serum starvation seems to occur in part through the blockage of PI3K/AKT signaling pathway.


Molecules | 2012

Inhibitory Effect and Mechanism on Antiproliferation of Isoatriplicolide Tiglate (PCAC) from Paulownia Coreana

Samil Jung; Hyung-In Moon; Jiyeon Ohk; Soonduck Lee; Chengping Li; Soo-Ki Kim; Myeong-Sok Lee

Paulownia coreana has traditionally been used as the medicine and health food in the treatment of cancer and infectious diseases. In the present study, a new antiproliferation agent, isoatriplicolide tiglate (PCAC) was isolated from the chloroform soluble fraction of the leaves of Paulownia coreana. The antiproliferation activities of PCAC plant extract was examined in breast and cervical cancer cell lines in a time-and dose-dependent manners. Our in vitro experiments showed that PCAC suppresses the cell growth and proliferation of cancer cells at a relatively low concentration (<10 µg/mL) and induces apoptosis at a high concentration (>50 µg/mL). Western blot analysis showed that concentration higher than 50 µg/mL induces a time-dependent increase in the percentage of apoptotic cells. In this case, PCAC uses both extrinsic and intrinsic pathways for the apoptosis. PCAC treatment decreased the expression of pro-caspase 8, 9, and 3, the main regulators of apoptotic cell death, in MDA-MB-231 cells, accompanied by the activation of caspase 8, 9, and 3. More importantly, PCAC inhibited the in vitro proliferation of six other human breast and cervical cancer cell lines. In conclusion, our data strongly suggest that PCAC acts as an antiproliferation agents particularly against breast and cervical cancers by inducing cell cycle arrest in the S/G2 phase and caspase dependent apoptosis at relatively low (<10 μg/mL) and high (>50 µg/mL) concentrations, respectively.


International Journal of Oncology | 2015

SIAH1-induced p34SEI-1 polyubiquitination/degradation mediates p53 preferential vitamin C cytotoxicity

Soonduck Lee; Jinsun Kim; Samil Jung; Chengping Li; Young Yang; Keun Il Kim; Jong-Seok Lim; Yonghwan Kim; Choong-Il Cheon; Myeong-Sok Lee

Vitamin C is considered as an important anticancer therapeutic agent although this view is debatable. In this study, we introduce a physiological mechanism demonstrating how vitamin C exerts anticancer activity that induces cell cycle arrest and apoptosis. Our previous and current data reveal that p53 tumor suppressor is the prerequisite factor for stronger anticancer effects of vitamin C. In addition, vitamin C-mediated cancer cell cytotoxicity appears to be achieved at least partly through the downregulation of the p34SEI-1 oncoprotein. Our previous study showed that p34SEI-1 increases the survival of various types of cancer cells by inhibiting their apoptosis. Present data suggest that vitamin C treatment decreases the p34SEI-1 expression at the protein level and therefore alleviates its anti-apoptotic activity. Of note, SIAH1, E3 ubiquitin ligase, appears to be responsible for the p34SEI-1 polyubiquitination and its subsequent degradation, which is dependent on p53. In summary, vitamin C increases cancer cell death by inducing SIAH1-mediated polyubiquitination/degradation of the p34SEI-1 oncoprotein in a p53-dependent manner.


International Journal of Oncology | 2014

Distinct regulatory effect of the p34SEI-1 oncoprotein on cancer metastasis in HER2/neu-positive and -negative cells

Samil Jung; Jiyeon Ohk; Dongjun Jeong; Chengping Li; Soonduck Lee; Jingjing Duan; Changjin Kim; Jong-Seok Lim; Young Yang; Keun-Il Kim; Myeong-Sok Lee

The p34(SEI-1) oncoprotein is involved in a transcriptional regulation, cell cycle regulation, apoptosis, development and many other important cellular functions. Our present study suggests that p34(SEI-1) can promote metastasis by enhancing migration and invasion of cancer cells. Consistently, p34(SEI-1) expression was found to be increased as the tumor invasiveness progressed in human breast tissues. p34(SEI-1) may promote cancer metastasis by activating the PI3K/AKT signaling pathway. In this process, p34(SEI-1) activates two different serine/threonine kinases, AKT or ILK, depending on the expression status of HER2/neu oncogene. In HER2/neu suppressed cancer cells, p34(SEI-1) promoted metastasis mainly by activating AKT via phosphorylation of the 473 serine residue. In HER2/neu expressing cancer cells, p34(SEI-1) overexpression downregulates HER2/neu expression, leading to the activation of another crucial serine/threonine kinase ILK due to phosphorylation of the 178 threonine residue instead of AKT. These results suggest that p34(SEI-1) affects cancer metastasis by regulating two different signaling pathways depending on the HER2/neu expression level, in which AKT and ILK modulation can be stimulated by p34(SEI-1) overexpression.


Molecular Cancer | 2010

The anti-aging gene KLOTHO is a novel target for epigenetic silencing in human cervical carcinoma

Jaehyouk Lee; Dongjun Jeong; Jinsun Kim; Soonduck Lee; Jin-Hwa Park; Boogi Chang; Samil Jung; Lisha Yi; Youngsoo Han; Young Yang; Keun Il Kim; Jong-Seok Lim; Inchul Yang; Seob Jeon; Dong Han Bae; Chang-Jin Kim; Myeong-Sok Lee


International Journal of Molecular Medicine | 1998

Vitamin C inhibits p53-induced replicative senescence through suppression of ROS production and p38 MAPK activity.

Jee-Eun Kim; Dong-Hoon Jin; Soonduck Lee; Seungwoo Hong; Jae-Sik Shin; Seung-Koo Lee; Da-Jung Jung; Jae-Seung Kang; Wang-Jae Lee

Collaboration


Dive into the Soonduck Lee's collaboration.

Top Co-Authors

Avatar

Myeong-Sok Lee

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Samil Jung

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Chengping Li

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Jong-Seok Lim

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Young Yang

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Dongjun Jeong

Soonchunhyang University

View shared research outputs
Top Co-Authors

Avatar

Jiyeon Ohk

Sookmyung Women's University

View shared research outputs
Top Co-Authors

Avatar

Changjin Kim

Soonchunhyang University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jinsun Kim

Sookmyung Women's University

View shared research outputs
Researchain Logo
Decentralizing Knowledge