Network


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

Hotspot


Dive into the research topics where Youn Sang Jung is active.

Publication


Featured researches published by Youn Sang Jung.


Laboratory Investigation | 2013

Elevated TRAF2/6 expression in Parkinson’s disease is caused by the loss of Parkin E3 ligase activity

Ji Yun Chung; Hee Ra Park; Su Jin Lee; Sun Hye Lee; Jin Sik Kim; Youn Sang Jung; Sang Hyun Hwang; Nam Chul Ha; Won Gi Seol; Jaewon Lee; Bum Joon Park

Parkinson’s disease (PD) is the second leading neurodegenerative disease, and is known to be induced by environmental factors or genetic mutations. Among the verified genetic mutations of PD, Parkin, isolated from the PARK2 locus, shows an autosomal recessive inheritance pattern and is known to be an E3 ligase. However, the physiological target of Parkin and the molecular mechanism of Parkin-deficiency-induced PD have not been clearly demonstrated until now. It has recently been proposed that inflammation, suggesting as a causal factor for PD, is enhanced by Parkin deficiency. Thus, we examined the relationship between inflammation-related factors and Parkin. Here, we provide the evidence that Parkin suppresses inflammation and cytokine-induced cell death by promoting the proteasomal degradation of TRAF2/6 (TNF-α receptor-associated factor 2/6). Overexpression of Parkin can reduce the half-lives of TRAF2 and TRAF6, whereas si-Parkin can extend them. However, mutant Parkins did not alter the expression of TRAF2/6. Thus, loss of Parkin enhances sensitivity to TNF-α- or IL-1β-induced JNK activation and NF-κB activation. Indeed, si-Parkin-induced apoptosis is suppressed by the knockdown of TRAF6 or TRAF2. We also observed elevated expression levels of TRAF6 and a reduction of IκB in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mouse model. Moreover, elevated expression levels or aggregation of TRAF6 were detected in approximately half of the human PD tissues (7/15 cases) and 2 cases, respectively. In addition, TRAF6 and Parkin expression levels show a reverse relationship in human PD tissues. Our results strongly suggest that the reduction of Parkin or overexpression of TRAF2/6 by chronic inflammation would be the reason for occurrence of PD.


Cell Cycle | 2012

Estrogen receptor α is a novel target of the Von Hippel-Lindau protein and is responsible for the proliferation of VHL-deficient cells under hypoxic conditions

Youn Sang Jung; Su Jin Lee; Min Ho Yoon; Nam Chul Ha; Bum Joon Park

The Von Hippel-Lindau gene (VHL) is frequently deleted or mutated in human renal cell carcinoma (RCC) at the early stage. According to the well-established theory, pVHL acts as a tumor suppressor through its E3 ligase activity, which targets hypoxia-inducing factor-1α (HIF-1α). However, the elevated expression of HIF-1α did not promote cell proliferation, indicating that there would be another target, which could promote cell proliferation at the early cancer stage of RCC. In this study, we show that estrogen receptor-α (ER-α) is a novel proteasomal degradation target of the pVHL E3 ligase. Indeed, the overexpression of VHL suppresses exo- and endogenous ER-α expression, whereas si-pVHL can increase ER-α expression. The negative regulation of pVHL on ER-α expression is achieved by its E3 ligase activity. Thus, pVHL can promote the ER-α ubiquitinylation. In addition, we revealed that ER-α and HIF-1α are competitive substrates of pVHL. Thus, under normal conditions, ER-α overexpression can increase the transcription factor activity of HIF-1α. Under the hypoxic condition, where HIF-1α is not a suitable target of pVHL, ER-α is more rapidly degraded by pVHL. However, in VHL-deficient cells, the expression of ER-α and HIF-1α is retained, so that the hypoxic condition did not suppress cell proliferation obviously compared with cells that are expressing pVHL. Thus, blocking of ER-α using its inhibitor could suppress the proliferation of VHL-deficient cells as effectively as hypoxia-induced growth suppression. Considering our results, blocking of ER-α signaling in VHL-deficient cancer cells would be beneficial for cancer suppression. Indeed, we showed the anti-proliferative effect of Faslodex in VHL-deficient cells.


Journal of Clinical Investigation | 2016

Interruption of progerin–lamin A/C binding ameliorates Hutchinson-Gilford progeria syndrome phenotype

Su Jin Lee; Youn Sang Jung; Min Ho Yoon; So Mi Kang; Ah Young Oh; Jee Hyun Lee; So Young Jun; Tae Gyun Woo; Ho Young Chun; Sang Kyum Kim; Kyu Jin Chung; Ho Young Lee; Kyeong Lee; Guanghai Jin; Min Kyun Na; Nam Chul Ha; Clea Bárcena; José M. P. Freije; Carlos López-Otín; Gyu Yong Song; Bum Joon Park

Hutchinson-Gilford progeria syndrome (HGPS) is a rare autosomal dominant genetic disease that is caused by a silent mutation of the LMNA gene encoding lamins A and C (lamin A/C). The G608G mutation generates a more accessible splicing donor site than does WT and produces an alternatively spliced product of LMNA called progerin, which is also expressed in normal aged cells. In this study, we determined that progerin binds directly to lamin A/C and induces profound nuclear aberrations. Given this observation, we performed a random screening of a chemical library and identified 3 compounds (JH1, JH4, and JH13) that efficiently block progerin-lamin A/C binding. These 3 chemicals, particularly JH4, alleviated nuclear deformation and reversed senescence markers characteristic of HGPS cells, including growth arrest and senescence-associated β-gal (SA-β-gal) activity. We then used microarray-based analysis to demonstrate that JH4 is able to rescue defects of cell-cycle progression in both HGPS and aged cells. Furthermore, administration of JH4 to LmnaG609G/G609G-mutant mice, which phenocopy human HGPS, resulted in a marked improvement of several progeria phenotypes and an extended lifespan. Together, these findings indicate that specific inhibitors with the ability to block pathological progerin-lamin A/C binding may represent a promising strategy for improving lifespan and health in both HGPS and normal aging.


Oncotarget | 2015

Wnt2 complements Wnt/β-catenin signaling in colorectal cancer.

Youn Sang Jung; Sohee Jun; Sun Hye Lee; Amrish Sharma; Jae Il Park

Wnt2 is implicated in various human cancers. However, it remains unknown how Wnt2 is upregulated in human cancer and contributes to tumorigenesis. Here we found that Wnt2 is highly expressed in colorectal cancer (CRC) cells. In addition to co-expression of Wnt2 with Wnt/β-catenin target genes in CRC, knockdown or knockout of Wnt2 significantly downregulates Wnt/β-catenin target gene expression in CRC cells. Importantly, depletion or ablation of endogenous Wnt2 inhibits CRC cell proliferation. Similarly, neutralizing secreted Wnt2 reduces Wnt target gene expression and suppresses CRC cell proliferation. Conversely, Wnt2 increases cell proliferation of intestinal epithelial cells. Intriguingly, WNT2 expression is transcriptionally silenced by EZH2-mediated H3K27me3 histone modification in non-CRC cells, However, WNT2 expression is de-repressed by the loss of PRC2s promoter occupancy in CRC cells. Our results reveal the unexpected roles of Wnt2 in complementing Wnt/β-catenin signaling for CRC cell proliferation.


Neurosignals | 2011

Direct Interaction of α-Synuclein and AKT Regulates IGF-1 Signaling: Implication of Parkinson Disease

Ji Yun Chung; Su Jin Lee; Sun Hye Lee; Youn Sang Jung; Nam Chul Ha; Wongi Seol; Bum Joon Park

Genetic mutation of α-synuclein (α-SYN) is clearly verified as the causal factor of human and mouse Parkinson’s disease. However, biological function of α-SYN has not been clearly demonstrated until now. In this investigation, we reveal that α-SYN is a co-regulator of growth factor-induced AKT activation. Elimination of SYN reduces the IGF-1-mediated AKT activation. Similarly, mutant SYN suppresses the IGF-1-induced AKT activation. Wild-type SYN can interact with AKT and enhance the solubility and plasma localization of AKT in response to IGF-1, whereas mutant α-SYNs do not interact with AKT. In addition, elevated expression of SYN blocks the AKT activation. We also find that si-RNA against α-SYN abolished the protective effect of IGF-1 against DNA damage-induced apoptosis. Our result strongly indicates that Parkinson’s disease, induced by α-SYN mutation, is evoked by deregulation of the AKT-signaling cascade.


Cell Cycle | 2013

Loss of VHL promotes progerin expression, leading to impaired p14/ARF function and suppression of p53 activity.

Youn Sang Jung; Su Jin Lee; Sun Hye Lee; Ji Yun Chung; Youn Jin Jung; Sang Hyun Hwang; Nam Chul Ha; Bum Joon Park

Renal cell carcinomas (RCCs) are frequently occurring genitourinary malignancies in the aged population. A morphological characteristic of RCCs is an irregular nuclear shape, which is used to index cancer grades. Other features of RCCs include the genetic inactivation of the von Hippel-Lindau gene, VHL, and p53 genetic-independent inactivation. An aberrant nuclear shape or p53 suppression has not yet been demonstrated. We examined the effect of progerin (an altered splicing product of the LMNA gene linked to Hutchinson Gilford progeria syndrome; HGPS) on the nuclear deformation of RCCs in comparison to that of HGPS cells. In this study, we showed that progerin was suppressed by pVHL and was responsible for nuclear irregularities as well as p53 inactivation. Thus, progerin suppression can ameliorate nuclear abnormalities and reactivate p53 in response to genotoxic addition. Furthermore, we found that progerin was a target of pVHL E3 ligase and suppressed p53 activity by p14/ARF inhibition. Our findings indicate that the elevated expression of progerin in RCCs results from the loss of pVHL and leads to p53 inactivation through p14/ARF suppression. Interestingly, we showed that progerin was expressed in human leukemia and primary cell lines, raising the possibility that the expression of this LMNA variant may be a common event in age-related cancer progression.


Cancer Research | 2016

Inhibiting DX2-p14/ARF interaction exerts antitumor effects in lung cancer and delays tumor progression

Ah Young Oh; Youn Sang Jung; Jiseon Kim; Jee Hyun Lee; Jung Hyun Cho; Ho Young Chun; Soyoung Park; Hyun-Chul Park; Si-Keun Lim; Nam Chul Ha; Jong Sook Park; Choon-Sik Park; Gyu Yong Song; Bum Joon Park

The aminoacyl tRNA synthetase complex-interacting multifunctional protein 2 (AIMP2) splice variant designated DX2 is induced by cigarette smoke carcinogens and is often detected in human lung cancer specimens. However, the function of DX2 in lung carcinogenesis is obscure. In this study, we found that DX2 expression was induced by oncogenes in human lung cancer tissues and cells. DX2 prevented oncogene-induced apoptosis and senescence and promoted drug resistance by directly binding to and inhibiting p14/ARF. Through chemical screening, we identified SLCB050, a novel compound that blocks the interaction between DX2 and p14/ARF in vitro and in vivo SLCB050 reduced the viability of human lung cancer cells, especially small cell lung cancer cells, in a p14/ARF-dependent manner. Moreover, in a mouse model of K-Ras-driven lung tumorigenesis, ectopic expression of DX2 induced small cell and non-small cell lung cancers, both of which could be suppressed by SLCB050 treatment. Taken together, our findings show how DX2 promotes lung cancer progression and how its activity may be thwarted as a strategy to treat patients with lung cancers exhibiting elevated DX2 levels. Cancer Res; 76(16); 4791-804. ©2016 AACR.


Neoplasia | 2014

Elevated estrogen receptor-α in VHL-deficient condition induces microtubule organizing center amplification via disruption of BRCA1/Rad51 interaction.

Youn Sang Jung; Ho Young Chun; Min Ho Yoon; Bum Joon Park

Since loss of VHL is frequently detected early phase genetic event in human renal cell carcinoma, pVHL is assumed to be indispensable for suppression of tumor initiation step. However, induction of HIF-1α, target of pVHL E3 ligase, is more adequate to angiogenesis step after tumor mass formation. Concerning this, it has been reported that pVHL is involved in centrosome location during metaphase and regulates ER-α signaling. Here, we provide the evidences that pVHL-mediated ER-α suppression is critical for microtubule organizing center (MTOC) maintaining and elevated ER-α promotes MTOC amplification through disruption of BRCA1-Rad51 interaction. In fact, numerous MTOC in VHL- or BRCA1-deficient cells are reduced by Fulvestrant, inhibitor of ER-α expression as well as antagonist. In addition, we reveal that activation of ER signaling can increase γ-tubulin, core factor of TuRC and render the resistance to Taxol. Thus, Fulvestrant but not Tamoxifen, antagonist against ER-α, can restore the Taxol sensitivity in VHL- or BRCA1-deficient cells. Our results suggest that pVHL-mediated ER-α suppression is important for regulation of MTOC as well as drug resistance.


Oncotarget | 2016

Prevention effect of rare ginsenosides against stress-hormone induced MTOC amplification

Jung Hyun Cho; Ho Young Chun; Jung Suk Lee; Jee Hyun Lee; Kyu Jin Cheong; Youn Sang Jung; Tae Gyun Woo; Min Ho Yoon; Ah Young Oh; So Mi Kang; Chunghui Lee; Hokeun Sun; Jihwan Hwang; Gyu Yong Song; Bum Joon Park

Stress has been suggested as one of important cause of human cancer without molecular biological evidence. Thus, we test the effect of stress-related hormones on cell viability and mitotic fidelity. Similarly to estrogen, stress hormone cortisol and its relative cortisone increase microtubule organizing center (MTOC) number through elevated expression of γ-tubulin and provide the Taxol resistance to human cancer cell lines. However, these effects are achieved by glucocorticoid hormone receptor (GR) but not by estrogen receptor (ER). Since ginsenosides possess steroid-like structure, we hypothesized that it would block the stress or estrogen-induced MTOC amplification and Taxol resistance. Among tested chemicals, rare ginsenoside, CSH1 (Rg6) shows obvious effect on inhibition of MTOC amplification, γ-tubulin induction and Taxol resistance. Comparing to Fulvestant (FST), ER-α specific inhibitor, this chemical can block the cortisol/cortisone-induced MTOC deregulation as well as ER-α signaling. Our results suggest that stress hormone induced tumorigenesis would be achieved by MTOC amplification, and CSH1 would be useful for prevention of stress-hormone or steroid hormone-induced chromosomal instability.


International Journal of Oncology | 2009

Isolation of a chemical inhibitor against K-Ras-induced p53 suppression through natural compound screening

Su Jin Lee; Youn Sang Jung; Sun Hye Lee; Hye Young Chung; Bum Joon Park

Collaboration


Dive into the Youn Sang Jung's collaboration.

Top Co-Authors

Avatar

Bum Joon Park

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Nam Chul Ha

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Su Jin Lee

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Gyu Yong Song

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Ho Young Chun

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Min Ho Yoon

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Sun Hye Lee

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Ah Young Oh

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Jee Hyun Lee

Chungnam National University

View shared research outputs
Top Co-Authors

Avatar

Ji Yun Chung

Pusan National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge