Network


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

Hotspot


Dive into the research topics where Im Sun Woo is active.

Publication


Featured researches published by Im Sun Woo.


Apoptosis | 2008

Ran suppresses paclitaxel-induced apoptosis in human glioblastoma cells

Im Sun Woo; Han-Su Jang; So Young Eun; Hyo Jung Kim; Sun Ah Ham; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jin-Hoi Kim; Chang Woo Han; Han Geuk Seo

Yeast-based functional screening of a human glioblastoma cDNA library identified ras-related nuclear protein (Ran) as a novel suppressor of Bcl-2-associated X protein (Bax), a pro-apoptotic member of the Bcl-2 family of proteins. Yeast cells that expressed human Ran were resistant to Bax-induced cell death. In U373MG glioblastoma cells, stable overexpression of Ran significantly attenuated apoptotic cell death induced by the chemotherapeutic agent paclitaxel. FACS analysis demonstrated that Ran is involved in paclitaxel-induced cell cycle arrest. Stable overexpression of Ran also markedly inhibited the phosphorylation of Bcl-2 by paclitaxel, and inhibited the translocation of Bax, the release of cytochrome c and activation of caspase-3. Paclitaxel-induced phosphorylation of c-JUN N-terminal kinase (JNK), but not p38, extracellular signal-regulated kinase and Akt, was markedly suppressed in U373MG cells that stably expressed Ran. These results suggest that Ran suppresses paclitaxel-induced cell death through the downregulation of JNK-mediated signal pathways.


Journal of Cellular and Molecular Medicine | 2009

PPARδ promotes wound healing by up-regulating TGF-β1-dependent or -independent expression of extracellular matrix proteins

Sun Ah Ham; Hyo Jung Kim; Hyun Joon Kim; Eun Sil Kang; So Young Eun; Gil Hyeong Kim; Myung Hyun Park; Im Sun Woo; Hye Jung Kim; Ki Churl Chang; Jae Heun Lee; Han Geuk Seo

Although the peroxisome proliferator‐activated receptor (PPAR) δ has been implicated in the wound healing process, its exact role and mechanism of action have not been fully elucidated. Our previous findings showed that PPARδ induces the expression of the transforming growth factor (TGF)‐β1, which has been implicated in the deposit of extracellular matrix proteins. Here, we demonstrate that administration of GW501516, a specific PPARδ ligand, significantly promoted wound closure in the experimental mouse and had a profound effect on the expression of collagen types I and III, alpha‐smooth muscle actin, pSmad3 and TGF‐β1, which play a pivotal role in wound healing processes. Activation of PPARδ increased migration of human epidermal keratinocytes and dermal fibroblasts in in vitro scrape‐wounding assays. Addition of a specific ALK5 receptor inhibitor SB431542 significantly suppressed GW501516‐induced migration of human keratinocytes and fibroblasts. In these cells, activated PPARδ also induced the expression of collagen types I and III and fibronectin in a TGF‐β1‐dependent or ‐independent manner. The effect of PPARδ on the expression of type III collagen was dually regulated by the direct binding of PPARδ and Smad3 to a direct repeat‐1 site and a Smad‐binding element, respectively, of the type III gene promoter. Taken together, these results demonstrated that PPARδ plays an important role in skin wound healing in vivo and that it functions by accelerating extracellular matrix‐mediated cellular interactions in a process mediated by the TGF‐β1/Smad3 signaling‐dependent or ‐ independent pathway.


Journal of Biological Chemistry | 2011

PPARδ Coordinates Angiotensin II-induced Senescence in Vascular Smooth Muscle Cells through PTEN-mediated Inhibition of Superoxide Generation

Hyo Jung Kim; Sun Ah Ham; Min Young Kim; Jung Seok Hwang; Hanna Lee; Eun Sil Kang; Taesik Yoo; Im Sun Woo; Chihiro Yabe-Nishimura; Kyung Shin Paek; Jin-Hoi Kim; Han Geuk Seo

Background: PPARδ is a ligand-activated transcriptional factor that has been implicated in the vascular homeostasis. Results: Activation of PPARδ significantly attenuated Ang II-induced senescence of VSMCs by up-regulation of PTEN and ensuing modulation of the PI3K/Akt signaling. Conclusion: PPARδ inhibits Ang II-induced senescence of VSMCs via PTEN-mediated inhibition of ROS generation. Significance: PPARδ provides a novel insight into the treatment of atherosclerotic vascular disease. Cellular senescence-associated changes in blood vessels have been implicated in aging and age-related cardiovascular disorders. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR) δ coordinates angiotensin (Ang) II-induced senescence of human vascular smooth muscle cells (VSMCs). Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly attenuated Ang II-induced generation of superoxides and suppressed senescence of VSMCs. A marked increase in the levels of p53 and p21 induced by Ang II was blunted by the treatment with GW501516. Ligand-activated PPARδ up-regulated expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and suppressed the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Knockdown of PTEN with siRNA abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt signaling, and on generation of ROS in VSMCs treated with Ang II. Finally, administration of GW501516 to apoE-deficient mice treated with Ang II significantly reduced the number of senescent cells in the aorta, where up-regulation of PTEN with reduced levels of phosphorylated Akt and ROS was demonstrated. Thus, ligand-activated PPARδ confers resistance to Ang II-induced senescence by up-regulation of PTEN and ensuing modulation of the PI3K/Akt signaling to reduce ROS generation in vascular cells.


Biochemical and Biophysical Research Communications | 2011

Transcriptional up-regulation of antioxidant genes by PPARδ inhibits angiotensin II-induced premature senescence in vascular smooth muscle cells.

Hyo Jung Kim; Sun Ah Ham; Kyung Shin Paek; Jung Seok Hwang; Si Young Jung; Min Young Kim; Hanna Jin; Eun Sil Kang; Im Sun Woo; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Chang Woo Han; Han Geuk Seo

This study evaluated peroxisome proliferator-activated receptor (PPAR) δ as a potential target for therapeutic intervention in Ang II-induced senescence in human vascular smooth muscle cells (hVSMCs). Activation of PPARδ by GW501516, a specific agonist of PPARδ, significantly inhibited the Ang II-induced premature senescence of hVSMCs. Agonist-activated PPARδ suppressed the generation of Ang II-triggered reactive oxygen species (ROS) with a concomitant reduction in DNA damage. Notably, GW501516 up-regulated the expression of antioxidant genes, such as glutathione peroxidase 1, thioredoxin 1, manganese superoxide dismutase and heme oxygenase 1. siRNA-mediated down-regulation of these antioxidant genes almost completely abolished the effects of GW501516 on ROS production and premature senescence in hVSMCs treated with Ang II. Taken together, the enhanced transcription of antioxidant genes is responsible for the PPARδ-mediated inhibition of premature senescence through sequestration of ROS in hVSMCs treated with Ang II.


Biochemical and Biophysical Research Communications | 2008

Identification of cytochrome c oxidase subunit 6A1 as a suppressor of Bax-induced cell death by yeast-based functional screening.

So Young Eun; Im Sun Woo; Han-Su Jang; Hana Jin; Min Young Kim; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jin-Hoi Kim; Han Geuk Seo

Human cytochrome c oxidase subunit VIa polypeptide 1 (COX6A1) was identified as a novel suppressor of Bcl-2-associated X protein (Bax)-mediated cell death using yeast-based functional screening of a mammalian cDNA library. The overexpression of COX6A1 significantly suppressed Bax- and N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis in yeast and human glioblastoma-derived U373MG cells, respectively. The generation of reactive oxygen species (ROS) in response to Bax or 4-HPR was inhibited in yeast and U373MG cells that expressed COX6A1, indicating that COX6A1 exerts a protective effect against ROS-induced cell damage. 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3 were markedly attenuated in U373MG cells that stably expressed COX6A1. Our results demonstrate that yeast-based functional screening of human genes for inhibitors of Bax-sensitivity in yeast identified a protein that not only suppresses the toxicity of Bax in yeast, but also has a potential role in protecting mammalian cells from 4-HPR-induced apoptosis.


Cancer Letters | 2011

TMEM14A inhibits N-(4-hydroxyphenyl)retinamide-induced apoptosis through the stabilization of mitochondrial membrane potential

Im Sun Woo; Hana Jin; Eun Sil Kang; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jae-Yong Park; Wan Sung Choi; Han Geuk Seo

Apoptosis is a highly conserved genetic process leading to death in mammalian cells. A critical step in apoptosis is mitochondrial membrane permeabilization, which results in the release of proteins critical to downstream events. Transmembrane protein 14A (TMEM14A) was identified as a novel suppressor of Bax using yeast-based functional screening. TMEM14A is a novel mitochondria-associated membrane protein containing a putative transmembrane domain. Over-expression of TMEM14A in U87MG cells inhibited N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis. TMEM14A prevented 4-HPR-induced loss of mitochondrial membrane potential (MMP), the release of cytochrome c, and the activation of caspase-3, but not the generation of reactive oxygen species, suggesting that TMEM14A regulates mitochondrial membrane potential in a ROS-independent manner. As expected, cyclosporin A, an inhibitor of membrane potential transition, inhibited 4-HPR-induced loss of MMP and apoptosis in U87MG cells, indicating that loss of MMP plays a pivotal role in 4-HPR-induced apoptosis. Suppression of TMEM14A expression using shRNA significantly increased apoptosis and MMP loss in untreated and 4-HPR-treated cells. These findings show for the first time that TMEM14A inhibits apoptosis by blocking the mitochondrial permeability transition and stabilizing mitochondrial membrane potential.


Pharmacological Research | 2008

Nrf2 regulates curcumin-induced aldose reductase expression indirectly via nuclear factor-κB

Eun Sil Kang; Gil Hyeong Kim; Hyo Jung Kim; Im Sun Woo; Sun Ah Ham; Hana Jin; Min Young Kim; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Han Geuk Seo; Jin-Yong Hwang

The osmotic response element (ORE) differs from the nuclear factor-kappaB (NF-kappaB) binding sequence by a single base pair; therefore, we investigated the involvement of NF-kappaB in the induction of aldose reductase (AR) by curcumin. Curcumin, an herb-derived polyphenolic compound, elicited an increase in the expression and promoter activity of the AR gene in a nuclear factor-erythroid 2-related factor 2 (Nrf2)-dependent manner. Small interfering RNA (siRNA) against p65 or BAY11-7082, an inhibitor of NF-kappaB, significantly suppressed the curcumin and/or Nrf2-induced increase in expression levels and promoter activity of the AR gene. BAY11-7082 or siRNA against p65 also attenuated the curcumin-induced increase in the promoter activity of the wild type AR-ORE(wt) gene, but not that of the mutated AR-ORE(mt), indicating that the ORE is essential for the response to NF-kappaB. The expression of p65, the promoter activity and DNA binding activity of NF-kappaB were enhanced in the presence of curcumin in cells that were transfected with Nrf2 compared to those treated with curcumin alone. Cells that had been preincubated with curcumin demonstrated resistance to reactive oxygen species-induced cell damage through the suppressive effects in the generation of reactive aldehydes. These effects were significantly attenuated in the presence of BAY11-7082, indicating the involvement of NF-kappaB in the cellular response of AR to oxidative stress and toxic aldehydes.


Cancer Letters | 2009

Identification of ADP-ribosylation factor 4 as a suppressor of N-(4-hydroxyphenyl)retinamide-induced cell death

Im Sun Woo; So Young Eun; Han-Su Jang; Eun Sil Kang; Gil Hyeong Kim; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jin-Hoi Kim; Chang Woo Han; Han Geuk Seo

Yeast-based functional screening for inhibitors of Bcl-2-associated X protein (Bax)-induced cell death in yeast identified ADP-ribosylation factor 4 (ARF4) as a novel anti-apoptotic gene in human glioblastoma-derived U373MG cells. Yeast or U373MG cells that overexpressed ARF4 exhibited reduced reactive oxygen species (ROS) generation in response to Bax or N-(4-hydroxyphenyl)retinamide (4-HPR), respectively, which suggests that ROS play a role in the inhibition of cell death by ARF4. The 4-HPR-mediated phosphorylation of c-JUN N-terminal kinase, p38, and extracellular signal-regulated kinase was markedly suppressed in U373MG cells that stably expressed ARF4. Stable ARF4 transfectants were also refractory to 4-HPR-induced mitochondrial translocation of Bax, release of mitochondrial cytochrome c, and activation of caspase-3. Our results suggest that ARF4 participates in the regulation of glioblastoma apoptosis through the inhibition of stress-mediated apoptotic signals.


Neuroscience Letters | 2010

Farnesyl diphosphate synthase attenuates paclitaxel-induced apoptotic cell death in human glioblastoma U87MG cells.

Im Sun Woo; So Young Eun; Hyo Jung Kim; Eun Sil Kang; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jin-Hoi Kim; Soon-Chan Hong; Han Geuk Seo

Increased expression of farnesyl diphosphate synthase (FPPS) by stable transfection appeared to attenuate paclitaxel-induced apoptotic cell death in human glioblastoma U87MG cells. The present results suggest that the apoptotic functions of p53 and c-Jun N-terminal kinase (JNK) are affected by FPPS. Farnesyl diphosphate, a catalytic product of FPPS, also attenuated mentioned paclitaxel-induced apoptotic cell death. As expected, the FPPS inhibitor, pamidronate, enhanced paclitaxel-induced apoptotic cell death. The present results suggest that FPPS plays an important role in apoptotic cell death of cancer cells by blocking the JNK signaling cascade and activating mevalonate metabolism in paclitaxel-treated glioblastoma cells.


Free Radical Research | 2008

Down-regulation of aldose reductase renders J774A.1 cells more susceptible to acrolein- or hydrogen peroxide-induced cell death

Eun Sil Kang; Gil Hyeong Kim; Im Sun Woo; Hyo Jung Kim; So Young Eun; Sun Ah Ham; Hana Jin; Min Young Kim; Myung Hyun Park; Hye Jung Kim; Ki Churl Chang; Jae Heun Lee; Jin-Hoi Kim; Chihiro Yabe-Nishimura; Han Geuk Seo

Aldose reductase (AR) is abundantly expressed in a variety of cell lineages and has been implicated in the cellular response against oxidative stress. However, the exact functional role of AR against oxidative stress remains relatively unclear. This study investigated the role of AR in acrolein- or hydrogen peroxide-induced apoptosis using the J774.A.1 macrophage cell line. Ablation of AR with a small interference RNA or inhibition of AR activity significantly enhanced the acrolein- or hydrogen peroxide-induced generation of reactive oxygen species and aldehydes, leading to increased apoptotic cell death. Blockade of AR activity in J774A.1 cells markedly augmented the acrolein- or hydrogen peroxide-induced translocation of Bax to mitochondria along with reduced Bcl-2 and increased release of cytochrome c from the mitochodria. Taken together, these findings indicate that AR plays an important role in the cellular response against oxidative stress, by sequestering the reactive molecules generated in cells exposed to toxic substances.

Collaboration


Dive into the Im Sun Woo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hye Jung Kim

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Jae Heun Lee

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Ki Churl Chang

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Eun Sil Kang

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Hyo Jung Kim

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

So Young Eun

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gil Hyeong Kim

Gyeongsang National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge