Network


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

Hotspot


Dive into the research topics where Jung Lim Kim is active.

Publication


Featured researches published by Jung Lim Kim.


Experimental Cell Research | 2011

Metastatic function of BMP-2 in gastric cancer cells: The role of PI3K/AKT, MAPK, the NF-κB pathway, and MMP-9 expression

Myoung Hee Kang; Sang Cheul Oh; Hyun Joo Lee; Han Na Kang; Jung Lim Kim; Jun Suk Kim; Young Yoo

Bone morphogenetic proteins (BMPs) have been implicated in tumorigenesis and metastatic progression in various types of cancer cells, but the role and cellular mechanism in the invasive phenotype of gastric cancer cells is not known. Herein, we determined the roles of phosphoinositide 3-kinase (PI3K)/AKT, extracellular signal-regulated protein kinase (ERK), nuclear factor (NF)-κB, and matrix metalloproteinase (MMP) expression in BMP-2-mediated metastatic function in gastric cancer. We found that stimulation of BMP-2 in gastric cancer cells enhanced the phosphorylation of AKT and ERK. Accompanying activation of AKT and ERK kinase, BMP-2 also enhanced phosphorylation/degradation of IκBα and the nuclear translocation/activation of NF-κB. Interestingly, blockade of PI3K/AKT and ERK signaling using LY294002 and PD98059, respectively, significantly inhibited BMP-2-induced motility and invasiveness in association with the activation of NF-κB. Furthermore, BMP-2-induced MMP-9 expression and enzymatic activity was also significantly blocked by treatment with PI3K/AKT, ERK, or NF-κB inhibitors. Immunohistochemistry staining of 178 gastric tumor biopsies indicated that expression of BMP-2 and MMP-9 had a significant positive correlation with lymph node metastasis and a poor prognosis. These results indicate that the BMP-2 signaling pathway enhances tumor metastasis in gastric cancer by sequential activation of the PI3K/AKT or MAPK pathway followed by the induction of NF-κB and MMP-9 activity, indicating that BMP-2 has the potential to be a therapeutic molecular target to decrease metastasis.


Tumor Biology | 2015

BMP-2 induces motility and invasiveness by promoting colon cancer stemness through STAT3 activation

Bo Ram Kim; Sang Cheul Oh; Dae Hee Lee; Jung Lim Kim; Suk Young Lee; Myoung Hee Kang; Sun Il Lee; Sanghee Kang; Sung Yup Joung; Byung Wook Min

Bone morphogenetic proteins (BMPs) have been involved in metastatic progression and tumorigenesis of many cancer types. However, it remains unclear how BMP-2 contributes to the initiation and development of these cancers. Here, we investigated the role of BMP-2 in colon cancer stem cell (CSC) development from colon cancer cells. We also determined the effects of BMP-2 on CSC development and epithelial-mesenchymal transition (EMT) in human colon cancer cell lines HCT-116 and SW620. We found that BMP-2 enhanced sphere formation of colon cancer cells without serum. Also, BMP-2-induced spheres displayed up-regulation of stemness markers (CD133+ and EpCAM+) and increased drug resistance, hallmarks of CSCs. Importantly, expression of EMT activators p-Smad1/5 and Snail and N-cadherin was increased in the spheres’ cells, indicating that BMP-2 signaling might result in CSC self-renewal and EMT. Furthermore, siRNA-mediated knockdown of signal transducer and activator of transcription 3 (STAT3) in HCT-116 cells reversed BMP-2-induced EMT and stem cell formation. Taken together, our results suggest that the BMP-2 induced STAT3-mediated induction of colon cancer cell metastasis requires an EMT and/or changes in CSC markers.


The Korean Journal of Hematology | 2012

Effects of oral iron chelator deferasirox on human malignant lymphoma cells

Jong Gwon Choi; Jung Lim Kim; Joohee Park; Soonwook Lee; Seh Jong Park; Jun Suk Kim; Chul Won Choi

Background Iron is essential for cell proliferation and viability. It has been reported that iron depletion by a chelator inhibits proliferation of some cancer cells. Deferasirox is a new oral iron chelator, and a few reports have described its effects on lymphoma cells. The goal of this study was to determine the anticancer effects of deferasirox in malignant lymphoma cell lines. Methods Three human malignant lymphoma cell lines (NCI H28:N78, Ramos, and Jiyoye) were treated with deferasirox at final concentrations of 20, 50, or 100 µM. Cell proliferation was evaluated by an MTT assay, and cell cycle and apoptosis were analyzed by flow cytometry. Western blot analysis was performed to determine the relative activity of various apoptotic pathways. The role of caspase in deferasirox-induced apoptosis was investigated using a luminescent assay. Results The MTT assay showed that deferasirox had dose-dependent cytotoxic effects on all 3 cell lines. Cell cycle analysis showed that the sub-G1 portion increased in all 3 cell lines as the concentration of deferasirox increased. Early apoptosis was also confirmed in the treated cells by Annexin V and PI staining. Western blotting showed an increase in the cleavage of PARP, caspase 3/7, and caspase 9 in deferasirox-treated groups. Conclusion We demonstrated that deferasirox, a new oral iron-chelating agent, induced early apoptosis in human malignant lymphoma cells, and this apoptotic effect is dependent on the caspase-3/caspase-9 pathway.


Oncotarget | 2016

Metformin enhances TRAIL-induced apoptosis by Mcl-1 degradation via Mule in colorectal cancer cells

Seong Hye Park; Dae Hee Lee; Jung Lim Kim; Bo Ram Kim; Yoo Jin Na; Min Jee Jo; Yoon A Jeong; Suk Young Lee; Sun Il Lee; Yong Yook Lee; Sang Cheul Oh

Metformin is an anti-diabetic drug with a promising anti-cancer potential. In this study, we show that subtoxic doses of metformin effectively sensitize human colorectal cancer (CRC) cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which induces apoptosis. Metformin alone did not induce apoptosis, but significantly potentiated TRAIL-induced apoptosis in CRC cells. CRC cells treated with metformin and TRAIL showed activation of the intrinsic and extrinsic pathways of caspase activation. We attempted to elucidate the underlying mechanism, and found that metformin significantly reduced the protein levels of myeloid cell leukemia 1 (Mcl-1) in CRC cells and, the overexpression of Mcl-1 inhibited cell death induced by metformin and/or TRAIL. Further experiments revealed that metformin did not affect mRNA levels, but increased proteasomal degradation and protein stability of Mcl-1. Knockdown of Mule triggered a significant decrease of Mcl-1 polyubiquitination. Metformin caused the dissociation of Noxa from Mcl-1, which allowed the binding of the BH3-containing ubiquitin ligase Mule followed by Mcl-1ubiquitination and degradation. The metformin-induced degradation of Mcl-1 required E3 ligase Mule, which is responsible for the polyubiquitination of Mcl-1. Our study is the first report indicating that metformin enhances TRAIL-induced apoptosis through Noxa and favors the interaction between Mcl-1 and Mule, which consequently affects Mcl-1 ubiquitination.


Oncotarget | 2017

Genipin suppresses colorectal cancer cells by inhibiting the Sonic Hedgehog pathway

Bo Ram Kim; Yoon A Jeong; Yoo Jin Na; Seong Hye Park; Min Jee Jo; Jung Lim Kim; Soyeon Jeong; Suk Young Lee; Hong Jun Kim; Sang Cheul Oh; Dae Hee Lee

Genipin, a major component of Gardenia jasminoides Ellis fruit, has been shown to inhibit the growth of gastric, prostate, and breast cancers. However, the anti-proliferative activity of genipin in colorectal cancer (CRC) has not been characterized. Herein, we demonstrated that genipin inhibits the proliferation of CRC cells and that genipin suppressed the Hedgehog pathway. Further investigation showed that p53 and NOXA protein levels were increased during inhibition of Hedgehog pathway-mediated apoptosis in CRC cells. We also showed that p53 modulated the expression of NOXA during genipin-induced apoptosis, and suppression via SMO also played a role in this process. Subsequently, GLI1 was ubiquitinated by the E3 ligase PCAF. In a xenograft tumor model, genipin suppressed tumor growth, which was also associated with Hedgehog inactivation. Taken together, these results suggest that genipin induces apoptosis through the Hedgehog signaling pathway by suppressing p53. These findings reveal a novel regulatory mechanism involving Hedgehog/p53/NOXA signaling in the modulation of CRC cell apoptosis and tumor-forming defects.


The International Journal of Biochemistry & Cell Biology | 2017

Cyclopamine sensitizes TRAIL-resistant gastric cancer cells to TRAIL-induced apoptosis via endoplasmic reticulum stress-mediated increase of death receptor 5 and survivin degradation

Yoo Jin Na; Dae Hee Lee; Jung Lim Kim; Bo Ram Kim; Seong Hye Park; Min Jee Jo; Soyeon Jeong; Hong Jun Kim; Suk Young Lee; Yoon A Jeong; Sang Cheul Oh

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is one of the most effective cancer treatments owing to its ability to selectively kill cancer cells, without affecting normal cells. However, it has been reported that several gastric cancer cells show resistance to TRAIL because of a scarcity of death receptor 5 (DR5) expressed on the cell surface. In this study, we show that cyclopamine sensitizes gastric cancer cells to TRAIL-induced apoptosis by elevating the expression of DR5. Interestingly, survivin hampers the existence of DR5 protein under normal conditions and cyclopamine decreases the expression of survivin, thus acting as a TRAIL sensitizer. Mechanistically, cyclopamine induces endoplasmic reticulum (ER) stress via reactive oxygen species (ROS) and CHOP, the last protein of the ER stress pathway and it regulates the proteasome degradation of survivin. Taken together, our results indicate that cyclopamine can be used for combination therapy in TRAIL-resistant gastric cancer cells.


Tumor Biology | 2016

Iron chelator-induced apoptosis via the ER stress pathway in gastric cancer cells.

Jung Lim Kim; Dae Hee Lee; Yoo Jin Na; Bo Ram Kim; Yoon A Jeong; Sun Il Lee; Sanghee Kang; Sung Yup Joung; Suk Young Lee; Sang Cheul Oh; Byung Wook Min

Many reports have shown the anticancer effects of iron deficient on cancer cells, but the effects of iron-chelators on gastric cancer have not been clearly elucidated. Recently, we reported that iron chelators induced an antiproliferative effect in human malignant lymphoma and myeloid leukemia cells. In the present study, we investigated the antitumor activity of these two iron-chelating agents, deferoxamine (DFO) and deferasirox (DFX), with gastric cancer cell lines, and their apoptosis-inducing effects as the potential mechanism. We found that iron chelators displayed significant antiproliferative activity in human gastric cancer cell lines, which may be attributed to their induction of G1 phase arrest and apoptosis. We also found that iron chelators induced reactive oxygen species (ROS) production, resulting in the activation of both c-Jun N-terminal kinase (JNK) and endoplasmic reticulum (ER) stress apoptotic pathways in gastric cancer cells. Taken together, our data suggest that iron chelators induced apoptosis in gastric cancer, involving ROS formation ER stress and JNK activation.


Cancer Research | 2015

Abstract 3805: Effect of Imatinib mesylate in gastric cancer cell progression

Jung Lim Kim; Bo Ram Kim; Yoo Jin Na; Seong Hye Park; Yoon A Jeong; Sang Cheul Oh

Imatinib is a powerful tyrosine kinase inhibitor that specifically targets BCR-ABL, KIT, and PDGFR kinases that is used in the treatment of chronic myelogenous leukemia (CML), gastrointestinal stromal tumors (GIST), and other cancers. But, imatinib mesylate has not yet been reported about anti-cancer effect in gastric cancer. This study was undertaken to evaluate the in vitro effect of imatinib mesylate in gastric cancer cells and to define molecular mechanism underlying these effects. This study was to determine the expression of PDGFR molecules in gastric cancer cells (AGS, MKN28, MKN45, SNU638) by semi-quantitative polimerase chain reaction (PCR). The effects of imatinib mesylate on gastric cancer cells were examined in four human gastric cancer cell lines. The results showed expression of PDGFR in all four gastric cancer cells. We measured the sensitivity of gastric cancer cell line (AGS, MKN28, MKN45, SNU638) to imatinib mesylate using MTT assay. Cell viability of cell treated with imatinib mesylate was decreased in high dose. But cell viability was not change in high dose. Next, the results showed expression of PDGFR and suppression of its phosphorylation by imatinib mesylate in all four gastric cancer cells. Cell cycle analysis for sub-G1 and G2/M fraction also detected the increased cell death in the cells treated with imatinib mesylates. Results by Western blotting indicated that imatinib mesylates promotes apoptosis of gastric cancer cells by both activating caspase-3 and -9 and inducing PARP cleavage. And expression of p-AKT level was decreased. Cell migration and invasion that imatinib treated cells is decreased in low dose. Cell viability assay revealed that the combination of imatinib mesylate and chemo-reagent synergistically inhibited cell growth compared to cells treated with any of the agent alone. In conclusion, inhibition the PDGFR by imatinib mesylates lead to apoptosis of gastric cancer cell lines. And Imatinib suppressed motility and invasion of gastric cancer cells. The results suggest that imatinib mesylate may be useful in the treatment of gastric cancer. Note: This abstract was not presented at the meeting. Citation Format: Jung Lim Kim, Bo Ram Kim, Yoo Jin Na, Seong Hye Park, Yoon A Jeong, Sang Cheul Oh. Effect of Imatinib mesylate in gastric cancer cell progression. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3805. doi:10.1158/1538-7445.AM2015-3805


The International Journal of Biochemistry & Cell Biology | 2018

Upregulation of EphB3 in gastric cancer with acquired resistance to a FGFR inhibitor

Suk-young Lee; Yoo Jin Na; Yoon A Jeong; Jung Lim Kim; Sang Cheul Oh; Dae Hee Lee

Amplification of fibroblast growth factor receptor2 (FGFR2) has been regarded as a druggable target in gastric cancer (GC). Despite known potential of AZD4547, a selective inhibitor of FGFR 1-3, to suppress tumorigenic effects of activated FGFR2, resistance to the targeted agent has been an unresolved issue. This study was performed to elucidate the mechanism of AZD4547 resistance in GC cells. SNU-16 cells were used to establish an AZD4547-resistant GC cell line, SNU-16R. Elevated phosphorylation of EphB3 was confirmed using the Human Phospho-Receptor Tyrosine Kinase Array kit. A tyrosine kinase inhibitor (TKI) of EphB3 was used to investigate the effects of suppressed EphB3 activity in the SNU-16R cell line. SNU-16R cells exhibited upregulated phosphorylation of EphB3. Treatment of SNU-16R cells with the EphB3 TKI resulted in induction of apoptosis, decreased cellular viability, and cell cycle arrest at sub-G1 phase. SNU-16R cells expressed upregulated levels of N-cadherin, vimentin, Snail, matrix metalloproteinase 2 (MMP-2), and MMP-9, and reduced levels of E-cadherin, characteristic of epithelial to mesenchymal transition (EMT). Matrigel invasion assay also demonstrated the increased invasiveness of SNU-16R cells. EphB3 TKI treatment inhibited EMT of SNU-16R cells. Activation of mammalian target of rapamycin (mTOR) through the Ras-ERK1/2 pathway was suggested as the signal transduction mechanism downstream EphB3 by showing enhanced phosphorylation of Raf-1, MEK1/2, ERK1/2, mTOR and its downstream substrates in SNU-16R cells. As expected, EphB3 TKI decreased phosphorylation of these proteins. Our data suggest phosphorylation of mTOR through signaling by EphB3 is a potential mechanism of AZD4547 resistance in GC cells.


Oncotarget | 2018

Correction: Genipin suppresses colorectal cancer cells by inhibiting the Sonic Hedgehog pathway [Oncotarget. 2017; 8:101952-101964] doi 10.18632/oncotarget.21882

Bo Ram Kim; Yoon A Jeong; Yoo Jin Na; Seong Hye Park; Min Jee Jo; Jung Lim Kim; Soyeon Jeong; Suk Young Lee; Hong Jun Kim; Sang Cheul Oh; Dae Hee Lee

[This corrects the article DOI: 10.18632/oncotarget.21882.].

Collaboration


Dive into the Jung Lim Kim's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge