Network


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

Hotspot


Dive into the research topics where Hana Jin is active.

Publication


Featured researches published by Hana Jin.


Circulation Research | 2009

Peroxisome Proliferator-Activated Receptor δ Regulates Extracellular Matrix and Apoptosis of Vascular Smooth Muscle Cells Through the Activation of Transforming Growth Factor-β1/Smad3

Hyo Jung Kim; Min Young Kim; Hana Jin; Hyun Joon Kim; Sang Soo Kang; Hye Jung Kim; Jae Heun Lee; Ki Churl Chang; Jin-Yong Hwang; Chihiro Yabe-Nishimura; Jin-Hoi Kim; Han Geuk Seo

Homeostasis of the extracellular matrix and apoptosis of vascular smooth muscle cells (VSMCs) are key components in the regulation of the stability of atherosclerotic plaques. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR)&dgr; regulates extracellular matrix synthesis and degradation through transforming growth factor-β1 and its effector, Smad3. Activation of PPAR&dgr; strongly amplified the expression of types I and III collagen, fibronectin, elastin, and TIMP-3 (tissue inhibitor of metalloproteinases 3), but not of TIMP-1, matrix metalloproteinase-2 or -9. The effect of PPAR&dgr; on the expression of type III collagen was dually regulated by the direct binding of PPAR&dgr; and Smad3 to a direct repeat-1 site and a Smad-binding element, respectively, in the type III collagen gene promoter. The activation of PPAR&dgr; attenuated apoptotic cell death in VSMCs induced by oxidized low-density lipoprotein, and similar antiapoptotic effects were observed on treatment of cells with exogenous type I and/or III collagen. Administration of a PPAR&dgr; ligand GW501516 to mice also suppressed elastase-induced cell death of aortic VSMCs. These results suggest that PPAR&dgr;-induced upregulation of extracellular matrix proteins exerts an antiapoptotic effect, thereby maintaining the stability of atherosclerotic plaques. Specific ligands of PPAR&dgr; may aid in the therapeutic intervention of atherosclerosis by improving plaque stability and patient prognosis.


Experimental Dermatology | 2014

P2Y2R activation by nucleotides promotes skin wound‐healing process

Hana Jin; Jihye Seo; So Young Eun; Young Nak Joo; Sang Won Park; Jae Heun Lee; Ki Churl Chang; Hye Jung Kim

P2Y2R has been shown to be upregulated in a variety of tissues in response to stress or injury and to mediate tissue regeneration through its ability to activate multiple signalling pathways. This study aimed to investigate the role of P2Y2R in the wound‐healing process and the mechanisms by which P2Y2R activation promotes wound healing in fibroblasts. The role of P2Y2R in skin wound healing was examined using a full‐thickness skin wound model in wildtype (WT) and P2Y2R−/− mice and an in vitro scratch wound model in control or P2Y2R siRNA‐transfected fibroblasts. WT mice showed significantly decreased wound size compared with P2Y2R−/− mice at day 14 post‐wounding, and immunohistochemical analysis showed that a proliferation marker Ki67 and extracellular matrix (ECM)‐related proteins VEGF, collagen I, fibronectin and α‐SMA were overexpressed in WT mice, which were reduced in P2Y2R−/− mice. Scratch‐wounded fibroblasts increased ATP release, which peaked at 5 min. In addition, scratch wounding increased the level of P2Y2R mRNA. Activation of P2Y2R by ATP or UTP enhanced proliferation and migration of fibroblasts in in vitro scratch wound assays and were blocked by P2Y2R siRNA. Finally, ATP or UTP also increased the levels of ECM‐related proteins through the activation of P2Y2R in fibroblasts. This study suggests that P2Y2R may be a potential therapeutic target to promote wound healing in chronic wound diseases.


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.


International Immunopharmacology | 2012

Tanshinone IIA inhibits TNF-α-mediated induction of VCAM-1 but not ICAM-1 through the regulation of GATA-6 and IRF-1.

Irina Tsoy Nizamutdinova; Young-Min Kim; Hana Jin; Kun Ho Son; Jae Heun Lee; Ki Churl Chang; Hye Jung Kim

The goal of this study was to investigate the differential effect of tanshinone IIA on the induction of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) by TNF-α and the possible molecular mechanisms by which it regulates ICAM-1 and VCAM-1 expression differentially. Stimulation of human umbilical vein endothelial cells (HUVEC) with TNF-α increased ICAM-1 and VCAM-1 expressions, and the pretreatment with tanshinone IIA concentration dependently inhibited VCAM-1 expression but not ICAM-1 expression. In previous study, PI3K/Akt, PKC and Jak/STAT-3 pathways were involved in the TNF-α-mediated induction of VCAM-1 but not ICAM-1. Thus, we examined the effect of tanshinone IIA on TNF-α-mediated activations of PI3K/Akt, PKC and Jak/STAT-3 pathways. Tanshinone IIA efficiently inhibited the phosphorylations of Akt, PKC and STAT-3 by TNF-α. Moreover, we determined the effect of tanshinone IIA on IRF-1 or GATAs induction and binding activity to VCAM-1 promoter since the upstream promoter region of VCAM-1 but not ICAM-1 contains IRF-1 and GATA binding motifs. Western blot analysis and ChIP assay showed that tanshinone IIA efficiently inhibited TNF-α-increased nuclear level of IRF-1 and GATA-6 and their binding affinity to VCAM-1 promoter region. Taken together, tanshinone IIA selectively inhibits TNF-α-mediated expression of VCAM-1 but not ICAM-1 through modulation of PI3/Akt, PKC and Jak/STAT-3 pathway as well as IRF-1 and GATA-6 binding activity.


Oncology Reports | 2013

Flavonoids from Citrus unshiu Marc. inhibit cancer cell adhesion to endothelial cells by selective inhibition of VCAM-1

Hana Jin; Won Sup Lee; Jeong Won Yun; Sang Mi Yi; Hye Jung Kim; Yung Hyun Choi; Gon-Sup Kim; Jin-Myung Jung; Chung Ho Ryu; Sung Chul Shin; Soon Chan Hong

Citrus fruits have been used as edible fruit and a component of traditional medicine for various diseases including cancer since ancient times. Herein, we investigated the anticancer activity of flavonoids of Citrus unshiu Marc. (FCM) focusing on anti-metastatic effects. We prepared FCM and performed experiments using MDA-MB-231 human breast cancer cells. FCM inhibited TNF-induced cancer cell adhesion to human umbilical vein endothelial cells (HUVECs) without showing any toxicity. FCM inhibited the expression of VCAM-1, but not of ICAM-1, on MDA-MB-231 cells as well as HUVECs. FCM inhibited protein kinase C (PKC) phosphorylation, but not Akt phosphorylation. FCM also inhibited cancer cell invasion in a dose-dependent manner, but not MMP-9 expression. In conclusion, this study suggested that FCM inhibits TNF-induced cancer cell adhesion to HUVECs by inhibiting VCAM-1 through inhibition of PKC, providing evidence that FCM have anti-metastatic activity by inhibiting adhesion molecules and invasion on human breast cancer 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.


Journal of Neuroscience Research | 2012

Activation of peroxisome proliferator-activated receptor-δ attenuates glutamate-induced neurotoxicity in HT22 mouse hippocampal cells.

Hana Jin; Sun Ah Ham; Min Young Kim; Im Sun Woo; Eun Sil Kang; Jung Seok Hwang; Ko-Woon Lee; Hye Jung Kim; Gu Seob Roh; Dae-Seog Lim; Dawon Kang; Han Geuk Seo

Glutamate‐induced neurotoxicity has been implicated in the pathogenesis of neurodegenerative disorders; however, little is known about the cellular events that underlie neurotoxicity or how to impede these events. This study demonstrates that peroxisome proliferator‐activated receptor (PPAR)‐δ regulates glutamate‐induced neurotoxicity in HT22 mouse hippocampal cells. Activation of PPARδ by GW501516, a specific ligand, significantly inhibited glutamate‐induced cell death and reactive oxygen species (ROS) production in HT22 cells. The siRNA‐mediated knockdown of PPARδ abrogated the effects of GW501516 in neuronal toxicity and ROS production induced by glutamate. In addition, ligand‐activated PPARδ reduced the glutamate‐induced level of intracellular calcium ions (Ca2+) by modulating the influx of Ca2+ from the extracellular space. Similarly, glutamate‐induced cell death and intracellular Ca2+ levels were attenuated in the presence of LY83583, an inhibitor of soluble guanylyl cyclase. Taken together, these results suggest that PPARδ plays an important role in glutamate‐induced neurotoxicity by modulating oxidative stress and Ca2+ influx.


Clinical and Experimental Pharmacology and Physiology | 2014

Dexmedetomidine-induced contraction involves c-Jun NH2-terminal kinase phosphorylation through activation of the 5-lipoxygenase pathway in the isolated endothelium-denuded rat aorta

Seong-Ho Ok; Hyo-Jin Byon; Hana Jin; Hye Jung Kim; Woochan Kim; In-Koo Nam; So Young Eun; Ju-Tae Sohn

Vasoconstriction induced by dexmedetomidine, a highly selective alpha‐2 adrenoceptor agonist, mainly involves c‐Jun NH2‐terminal kinase (JNK) phosphorylation in the isolated endothelium‐denuded aorta. We carried out an in vitro study to determine the main arachidonic acid metabolic pathway that is involved in dexmedetomidine‐induced JNK activation. Cumulative dexmedetomidine concentration‐contractile response curves were generated in the endothelium‐denuded rat aorta in the presence or absence of the following inhibitors: the JNK inhibitor SP600125, the phospholipase A2 inhibitor quinacrine dihydrochloride, the non‐specific lipoxygenase (LOX) inhibitor nordihydroguaiaretic acid, the 5‐LOX inhibitor AA‐861, the dual 5‐LOX and cyclooxygenase (COX) inhibitor phenidone, the non‐specific COX inhibitor indomethacin, the cytochrome p450 epoxygenase inhibitor fluconazole, the COX‐1 inhibitor SC‐560, and the COX‐2 inhibitor NS‐398. The effect of the alpha‐2 adrenoceptor inhibitor rauwolscine and other inhibitors, such as quinacrine dihydrochloride, nordihydroguaiaretic acid, AA‐861, phenidone, indomethacin and the protein kinase C inhibitor GF 109203X, on dexmedetomidine‐induced JNK phosphorylation was investigated in rat aortic vascular smooth muscle cells with western blotting. The effect of dexmedetomidine on 5‐LOX and COX‐2 expression was investigated in vascular smooth muscle cells. SP600125, quinacrine dihydrochloride, nordihydroguaiaretic acid, AA‐861, phenidone, rauwolscine and chelerythrine attenuated dexmedetomidine‐induced contraction. Indomethacin slightly attenuated dexmedetomidine‐induced contraction. Fluconazole and SC‐560 had no effect on dexmedetomidine‐induced contraction, whereas NS‐398 attenuated contraction. SP600125, rauwolscine, quinacrine dihydrochloride, nordihydroguaiaretic acid, AA‐861, phenidone and GF 109203X attenuated dexmedetomidine‐induced JNK phosphorylation. 5‐LOX and COX‐2 were upregulated by dexmedetomidine. Thus, dexmedetomidine‐induced alpha‐2 adrenoceptor‐mediated contraction is mediated mainly by 5‐LOX and partially by COX‐2, which leads to JNK phosphorylation.

Collaboration


Dive into the Hana Jin's collaboration.

Top Co-Authors

Avatar

Hye Jung Kim

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

Jae Heun Lee

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

So Young Eun

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Im Sun Woo

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Min Young Kim

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

Sang Won Park

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Chung Ho Ryu

Gyeongsang National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge