Network


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

Hotspot


Dive into the research topics where Eun Sil Kang is active.

Publication


Featured researches published by Eun Sil Kang.


Biochemical Pharmacology | 2012

The PPARδ-mediated inhibition of angiotensin II-induced premature senescence in human endothelial cells is SIRT1-dependent.

Min Young Kim; Eun Sil Kang; Sun Ah Ham; Jung Seok Hwang; Tae Sik Yoo; Hanna Lee; Kyung Shin Paek; Chankyu Park; Hoon Taek Lee; Jin-Hoi Kim; Chang Woo Han; Han Geuk Seo

Cellular senescence has been implicated in endothelial dysfunctions affecting vascular tone and regeneration. The molecular mechanisms of vascular senescence are poorly understood. The present study demonstrates that upregulation of SIRT1 by peroxisome proliferator-activated receptor (PPAR) δ attenuates premature senescence in angiotensin (Ang) II-treated human coronary artery endothelial cells (HCAECs). Activation of PPARδ by the specific ligand GW501516 significantly inhibited Ang II-induced premature senescence and generation of reactive oxygen species (ROS) in HCAECs. A marked concentration- and time-dependent increase in the mRNA levels of SIRT1 was observed in GW501516-treated HCAECs. The effects of GW501516 were almost completely abolished in the presence of small interfering (si) RNA against PPARδ, indicating that PPARδ mediates the effects of GW501516. In addition, activation of PPARδ, but not PPARα or PPARγ, significantly enhanced SIRT1 promoter activity and protein expression. Down-regulation or inhibition of SIRT1 by siRNA or sirtinol abrogated the effects of PPARδ on Ang II-induced premature senescence and ROS generation, respectively. Furthermore, resveratrol, a well-known activator of SIRT1, mimicked the action of PPARδ on Ang II-induced premature senescence and ROS generation. Taken together, these results indicate that the anti-senescent activities of PPARδ may be achieved at least in part by fine tuning the expression of SIRT1 in the vascular endothelium.


Mediators of Inflammation | 2012

Activation of Peroxisome Proliferator-Activated Receptor γ by Rosiglitazone Inhibits Lipopolysaccharide-Induced Release of High Mobility Group Box 1

Jung Seok Hwang; Eun Sil Kang; Sun Ah Ham; Taesik Yoo; Hanna Lee; Kyung Shin Paek; Chankyu Park; Jin-Hoi Kim; Dae-Seog Lim; Han Geuk Seo

Peroxisome proliferator-activated receptors (PPARs) are shown to modulate the pathological status of sepsis by regulating the release of high mobility group box 1 (HMGB1), a well-known late proinflammatory mediator of sepsis. Ligand-activated PPARs markedly inhibited lipopolysaccharide- (LPS) induced release of HMGB1 in RAW 264.7 cells. Among the ligands of PPAR, the effect of rosiglitazone, a specific ligand for PPARγ, was superior in the inhibition of HMGB1 release induced by LPS. This effect was observed in cells that received rosiglitazone before LPS or after LPS treatment, indicating that rosiglitazone is effective in both treatment and prevention. Ablation of PPARγ with small interfering RNA or GW9662-mediated inhibition of PPARγ abolished the effect of rosiglitazone on HMGB1 release. Furthermore, the overexpression of PPARγ markedly potentiated the inhibitory effect of rosiglitazone on HMGB1 release. In addition, rosiglitazone inhibited LPS-induced expression of Toll-like receptor 4 signal molecules, suggesting a possible mechanism by which rosiglitazone modulates HMGB1 release. Notably, the administration of rosiglitazone to mice improved survival rates in an LPS-induced animal model of endotoxemia, where reduced levels of circulating HMGB1 were demonstrated. Taken together, these results suggest that PPARs play an important role in the cellular response to inflammation by inhibiting HMGB1 release.


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.


Free Radical Research | 2012

Activation of PPARδ counteracts angiotensin II-induced ROS generation by inhibiting rac1 translocation in vascular smooth muscle cells.

Hanna Lee; Sun Ah Ham; Min Young Kim; Jae-Hwan Kim; Kyung Shin Paek; Eun Sil Kang; Hyo Jung Kim; Jung Seok Hwang; Taesik Yoo; Chankyu Park; Jin-Hoi Kim; Dae-Seog Lim; Chang Woo Han; Han Geuk Seo

Abstract Angiotensin II (Ang II)-mediated modification of the redox milieu of vascular smooth muscle cells (VSMCs) has been implicated in several pathophysiological processes, including cell proliferation, migration and differentiation. In this study, we demonstrate that the peroxisome proliferator-activated receptor (PPAR) δ counteracts Ang II-induced production of reactive oxygen species (ROS) in VSMCs. Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly reduced Ang II-induced ROS generation in VSMCs. This effect was, however, reversed in the presence of small interfering (si)RNA against PPARδ. The marked increase in ROS levels induced by Ang II was also eliminated by the inhibition of phosphatidylinositol 3-kinase (PI3K) but not of protein kinase C, suggesting the involvement of the PI3K/Akt signalling pathway in this process. Accordingly, ablation of Akt with siRNA further enhanced the inhibitory effects of GW501516 in Ang II-induced superoxide production. Ligand-activated PPARδ also blocked Ang II-induced translocation of Rac1 to the cell membrane, inhibiting the activation of NADPH oxidases and consequently ROS generation. These results indicate that ligand-activated PPARδ plays an important role in the cellular response to oxidative stress by decreasing ROS generated by Ang II in vascular cells.


Biochemical Journal | 2012

Ligand-activated PPARδ inhibits UVB-induced senescence of human keratinocytes via PTEN-mediated inhibition of superoxide production

Sun Ah Ham; Jung Seok Hwang; Taesik Yoo; Hanna Lee; Eun Sil Kang; Chankyu Park; Jae-Wook Oh; Hoon Taek Lee; Gyesik Min; Jin-Hoi Kim; Han Geuk Seo

UV radiation-mediated photodamage to the skin has been implicated in premature aging and photoaging-related skin cancer and melanoma. Little is known about the cellular events that underlie premature senescence, or how to impede these events. In the present study we demonstrate that PPARδ (peroxisome-proliferator-activated receptor δ) regulates UVB-induced premature senescence of normal keratinocytes. Activation of PPARδ by GW501516, a specific ligand of PPARδ, significantly attenuated UVB-mediated generation of ROS (reactive oxygen species) and suppressed senescence of human keratinocytes. Ligand-activated PPARδ up-regulated the expression of PTEN (phosphatase and tensin homologue deleted on chromosome 10) and suppressed the PI3K (phosphatidylinositol 3-kinase)/Akt pathway. Concomitantly, translocation of Rac1 to the plasma membrane, which leads to the activation of NADPH oxidases and generation of ROS, was significantly attenuated. siRNA (small interfering RNA)-mediated knockdown of PTEN abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt/Rac1 signalling and on generation of ROS in keratinocytes exposed to UVB. Finally, when HR-1 hairless mice were treated with GW501516 before exposure to UVB, the number of senescent cells in the skin was significantly reduced. Thus ligand-activated PPARδ confers resistance to UVB-induced cellular senescence by up-regulating PTEN and thereby modulating PI3K/Akt/Rac1 signalling to reduce ROS generation in keratinocytes.


Archives of Pharmacal Research | 2005

Agastache rugosa leaf extract inhibits the iNOS expression in ROS 17/2.8 cells activated with TNF-α and IL-1β

Hwa Min Oh; Youngjin Kang; Sun Hee Kim; Young Soo Lee; Min Kyu Park; Ja Myung Heo; Jin ji Sun; Hyo Jung Kim; Eun Sil Kang; Hye Jung Kim; Han Geuk Seo; Jae Heun Lee; Hye Sook Yun-Choi; Ki Churl Chang

It has been suggested that nitric oxide (NO) derived from inducible nitric oxide synthase (iNOS) may act as a mediator of cytokine-induced effects on bone turn-over. NO is also recognized as an important factor in bone remodeling, i.e., participating in osteoblast apoptosis in an arthritic joint. The components ofAgastache rugosa are known to have many pharmacological activities. In the present study, we investigated the effects of Agastache rugosa leaf extract (ELAR) on NO production and the iNOS expression in ROS 17/2.8 cells activated by a mixture of inflammatory cytokines including TNF-α and lL-1². A preincubation with ELAR significantly and concentration-dependently reduced the expression of iNOS protein in ROS 17/2.8 cells activated with the cytokine mixture. Consequently, the NO production was also significantly reduced by ELAR with an IC50 of 0.75 mg/mL The inhibitory mechanism of iNOS induction by ELAR prevented the activation and translocation of NF-κB (p65) to the nucleus from the cytosol fraction. Furthermore, ELAR concentration-dependently reduced the cellular toxicity induced by sodium nitroprusside, an NO-donor. These results suggest that ELAR may be beneficial in NO-mediated inflammatory conditions such as osteoporosis.


Journal of Investigative Dermatology | 2013

PPARδ Inhibits UVB-Induced Secretion of MMP-1 through MKP-7-Mediated Suppression of JNK Signaling

Sun A. Ham; Eun Sil Kang; Hanna Lee; Jung S. Hwang; Taesik Yoo; Kyung Shin Paek; Chankyu Park; Jin-Hoi Kim; Dae-Seog Lim; Han G. Seo

In the present study, we investigated the role of peroxisome proliferator-activated receptor (PPAR) δ in modulating matrix-degrading metalloproteinases and other mechanisms underlying photoaging processes in the skin. In human dermal fibroblasts (HDFs), activation of PPARδ by its specific ligand GW501516 markedly attenuated UVB-induced secretion of matrix metalloproteinase (MMP)-1, concomitant with decreased generation of reactive oxygen species. These effects were significantly reduced in the presence of PPARδ small interfering RNA and GSK0660. Furthermore, c-Jun N-terminal kinase (JNK), but not p38 or extracellular signal-regulated kinase, mediated PPARδ-dependent inhibition of MMP-1 secretion in HDFs exposed to UVB. PPARδ-mediated messenger RNA stabilization of mitogen-activated protein kinase phosphatase (MKP)-7 was responsible for the GW501516-mediated inhibition of JNK signaling. Inhibition of UVB-induced secretion of MMP-1 by PPARδ was associated with the restoration of types I and III collagen to levels approaching those in cells not exposed to UVB. Finally, in HR-1 hairless mice exposed to UVB, administration of GW501516 significantly reduced wrinkle formation and skin thickness, downregulated MMP-1 and JNK phosphorylation, and restored the levels of MKP-7, types I and III collagen. These results suggest that PPARδ-mediated inhibition of MMP-1 secretion prevents some effects of photoaging and maintains the integrity of skin by inhibiting the degradation of the collagenous extracellular matrix.


Free Radical Biology and Medicine | 2011

Aldose reductase in keratinocytes attenuates cellular apoptosis and senescence induced by UV radiation.

Eun Sil Kang; Kazumi Iwata; Kanako Ikami; Sun Ah Ham; Hye Jung Kim; Ki Churl Chang; Jae Heun Lee; Jae-Hwan Kim; Soo-Bong Park; Jin-Hoi Kim; Chihiro Yabe-Nishimura; Han Geuk Seo

Although aldose reductase (AR) has been implicated in the cellular response to oxidative stress, the role of AR in ultraviolet-B (UVB)-induced cellular injury has not been investigated. Here, we show that an increased expression of AR in human keratinocytes modulates UVB-induced apoptotic cell death and senescence. Overexpression of AR in HaCaT cells significantly attenuated UVB-induced cellular damage and apoptosis, with a decreased generation of reactive oxygen species (ROS) and aldehydes. Ablation of AR with small interfering RNA or inhibition of AR activity abolished these effects. We also show that increased AR activity suppressed UVB-induced activation of the p38 and c-Jun N-terminal kinases, but did not affect the extracellular signal-regulated kinase and phosphatidylinositol 3-kinase pathways. Similarly, UVB-induced translocation of Bax and Bcl-2 to mitochondria and cytosol, respectively, was markedly attenuated in cells overexpressing AR. Knockdown or inhibition of AR activity in primary cultured keratinocytes enhanced UVB-induced cellular senescence and increased the level of a cell-cycle regulatory protein, p53. Finally, cellular apoptosis induced by UVB radiation was significantly reduced in the epidermis of transgenic mice overexpressing human AR. These findings suggest that AR plays an important role in the cellular response to oxidative stress by sequestering ROS and reactive aldehydes generated in keratinocytes.

Collaboration


Dive into the Eun Sil Kang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hye Jung Kim

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

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

Im Sun Woo

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge