Network


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

Hotspot


Dive into the research topics where Jee Young An is active.

Publication


Featured researches published by Jee Young An.


Journal of Cellular Biochemistry | 2009

Effects of low dose quercetin: Cancer cell-specific inhibition of cell cycle progression

Jae-Hoon Jeong; Jee Young An; Yong Tae Kwon; Juong G. Rhee; Yong J. Lee

Quercetin is a flavonoid present in many vegetables, fruits, and beverages. Due to its anti‐oxidant, anti‐tumor, and anti‐inflammatory activity, quercetin has been studied extensively as a chemoprevention agent in several cancer models. Since most of these studies used higher doses of quercetin than clinically achievable, we focused on the effectiveness of physiologically relevant doses of quercetin. A low dose of quercetin exerted cancer cell‐specific inhibition of proliferation and this inhibition resulted from cell cycle arrest at the G1 phase. Quercetin induced p21 CDK inhibitor with a concomitant decrease of phosphorylation of pRb, which inhibits the G1/S cell cycle progression by trapping E2F1. A low dose of quercetin induced mild DNA damage and Chk2 activation, which is the main regulator of p21 expression by quercetin. In addition, quercetin down‐regulated the cyclin B1 and CDK1, essential components of G2/M cell cycle progression. Inhibition of the recruitment of key transcription factor NF‐Y to cyclin B1 gene promoter by quercetin led to transcriptional inhibition. This study proved that the chemo‐preventive efficacy of a physiologically relevant dose of quercetin can be achievable through the inhibition of cell cycle progression. J. Cell. Biochem. 106: 73–82, 2009.


Molecular and Cellular Biology | 2003

Female Lethality and Apoptosis of Spermatocytes in Mice Lacking the UBR2 Ubiquitin Ligase of the N-End Rule Pathway

Yong Tae Kwon; Zanxian Xia; Jee Young An; Takafumi Tasaki; Ilia V. Davydov; Jai Wha Seo; Jun Sheng; Youming Xie; Alexander Varshavsky

ABSTRACT Substrates of the ubiquitin-dependent N-end rule pathway include proteins with destabilizing N-terminal residues. UBR1−/− mice, which lacked the pathways ubiquitin ligase E3α, were viable and retained the N-end rule pathway. The present work describes the identification and analysis of mouse UBR2, a homolog of UBR1. We demonstrate that the substrate-binding properties of UBR2 are highly similar to those of UBR1, identifying UBR2 as the second E3 of the mammalian N-end rule pathway. UBR2 −/− mouse strains were constructed, and their viability was found to be dependent on both gender and genetic background. In the strain 129 (inbred) background, the UBR2 −/− genotype was lethal to most embryos of either gender. In the 129/B6 (mixed) background, most UBR2 −/− females died as embryos, whereas UBR2 −/− males were viable but infertile, owing to the postnatal degeneration of the testes. The gross architecture of UBR2 −/− testes was normal and spermatogonia were intact as well, but UBR2 −/− spermatocytes were arrested between leptotene/zygotene and pachytene and died through apoptosis. A conspicuous defect of UBR2 −/− spermatocytes was the absence of intact synaptonemal complexes. We conclude that the UBR2 ubiquitin ligase and, hence, the N-end rule pathway are required for male meiosis and spermatogenesis and for an essential aspect of female embryonic development.


Proceedings of the National Academy of Sciences of the United States of America | 2010

UBR2 mediates transcriptional silencing during spermatogenesis via histone ubiquitination

Jee Young An; Euna Kim; Yonghua Jiang; Adriana Zakrzewska; Dong Eun Kim; Min Jae Lee; Inhee Mook-Jung; Yi Zhang; Yong Tae Kwon

Ubiquitination of histones provides an important mechanism regulating chromatin remodeling and gene expression. Recent studies have revealed ubiquitin ligases involved in histone ubiquitination, yet the responsible enzymes and the function of histone ubiquitination in spermatogenesis remain unclear. We have previously shown that mice lacking the ubiquitin ligase UBR2, one of the recognition E3 components of the N-end rule proteolytic pathway, are infertile associated with meiotic arrest at prophase I. We here show that UBR2 localizes to meiotic chromatin regions, including unsynapsed axial elements linked to chromatin inactivation, and mediates transcriptional silencing via the ubiquitination of histone H2A. UBR2 interacts with the ubiquitin conjugating enzyme HR6B and its substrate H2A and promotes the HR6B–H2A interaction and the HR6B-to-H2A transfer of ubiquitin. UBR2 and ubiquitinated H2A (uH2A) spatiotemporally mark meiotic chromatin regions subject to transcriptional silencing, and UBR2-deficient spermatocytes fail to induce the ubiquitination of H2A during meiosis. UBR2-deficient spermatocytes are profoundly impaired in chromosome-wide transcriptional silencing of genes linked to unsynapsed axes of the X and Y chromosomes. Our findings suggest that insufficiency in UBR2-dependent histone ubiquitination triggers a pachytene checkpoint system, providing a new insight into chromatin remodeling and gene expression regulation.


Journal of Biological Chemistry | 2007

Evidence for two modes of development of acquired tumor necrosis factor-related apoptosis-inducing ligand resistance. Involvement of Bcl-xL.

Jae J. Song; Jee Young An; Yong Tae Kwon; Yong J. Lee

Previous studies have shown that repeated application of TRAIL induces acquired resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Using human prostate adenocarcinoma DU-145 and human pancreatic carcinoma MiaPaCa-2 cells as a model, we now demonstrate for the first time that two states of acquired TRAIL resistance can be developed after TRAIL treatment. Data from survival assay and Western blot analysis show that acquired TRAIL resistance was developed within 1 day and gradually decayed within 6 days after TRAIL treatment in both cell lines. After TRAIL treatment, the level of Bcl-xL increased and reached a maximum within 2 days and gradually decreased in both cell lines. Bcl-xL-mediated development of acquired TRAIL resistance was suppressed by knockdown of Bcl-xL expression. Protein interaction assay revealed that during the development of TRAIL resistance, Bcl-xL dissociated from Bad and then associated with Bax. Overexpression of mutant-type Bad (S136A), which prevents this dissociation, partially suppressed the development of acquired TRAIL resistance. Thus, our results suggest that (a) dissociation of Bad from Bcl-xL and (b) an increase in the intracellular level of Bcl-xL are responsible for development of acquired TRAIL resistance.


Journal of Biological Chemistry | 2005

Pretreatment of Acetylsalicylic Acid Promotes Tumor Necrosis Factor-related Apoptosis-inducing Ligand-induced Apoptosis by Down-regulating BCL-2 Gene Expression

Ki M. Kim; Jae J. Song; Jee Young An; Yong Tae Kwon; Yong J. Lee

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to be selective in the induction of apoptosis in cancer cells with minimal toxicity to normal tissues. However, not all cancers are sensitive to TRAIL-mediated apoptosis. Thus, TRAIL-resistant cancer cells must be sensitized first to become responsive to TRAIL. In this study, we observed that pretreatment by acetylsalicylic acid (ASA) augmented TRAIL-induced apoptotic death in human prostate adenocarcinoma LNCaP and human colorectal carcinoma CX-1 cells. Western blot analysis showed that pretreatment of ASA followed by TRAIL treatment activated caspases (8, 9, and 3) and cleaved poly(ADP-ribose) polymerase, the hallmark feature of apoptosis. Most interestingly, at least 12 h of pretreatment with ASA was prerequisite for promoting TRAIL-induced apoptosis and was related to down-regulation of BCL-2. Biochemical analysis revealed that ASA inhibited NF-κB activity, which is known to regulate BCL-2 gene expression, by dephosphorylating IκB-α and inhibiting IKKβ activity but not by affecting the HER-2/neu phosphatidylinositol 3-kinase-Akt signal pathway. Overexpression of BCL-2 suppressed the promotive effect of ASA on TRAIL-induced apoptosis and changes in mitochondrial membrane potential. Taken together, our studies suggested that ASA-promoted TRAIL cytotoxicity is mediated through down-regulating BCL-2 and by decreasing mitochondrial membrane potential.


Journal of Cellular Biochemistry | 2008

Quercetin‐induced ubiquitination and down‐regulation of Her‐2/neu

Jae-Hoon Jeong; Jee Young An; Yong Tae Kwon; Lu-Yuan Li; Yong J. Lee

Her‐2/neu (ErbB2) is a transmembrane tyrosine kinase and acts as a co‐receptor for the other EGFR family members. It is well known that high expression of Her‐2/neu is associated with a poor prognosis in breast cancer. Quercetin, a flavonoid present in many vegetables and fruits, has been studied extensively as a chemoprevention agent in several cancer models. In this study, we observed that quercetin decreased the level of Her‐2/neu protein in time‐ and dose‐dependent manners and also inhibited the downstream survival PI3K‐Akt signaling pathway in Her‐2/neu‐overexpressing breast cancer SK‐Br3 cells. We also observed that quercetin induced polyubiquitination of Her‐2/neu. When the proteasome pathway was blocked by MG‐132 during quercetin treatment, accumulation of the NP‐40 insoluble form of Her‐2/neu occurred. Interestingly, data from immunocomplex studies revealed that quercetin promoted interaction between Her‐2/neu and Hsp90 which is a molecular chaperone involved in stabilization of Her‐2/neu. In this condition, inhibition of Hsp90 activity by a specific inhibitor, geldanamycin (GA), or intracellular ATP depletion caused dissociation of Hsp90 from Her‐2/neu and promoted ubiquitination and down‐regulation of Her‐2/neu protein. In addition, the carboxyl terminus of Hsc70‐interacting protein (CHIP), a chaperone‐dependent E3 ubiquitin ligase, played a crucial role in the quercetin‐induced ubiquitination of Her‐2/neu. Inhibition of tyrosine kinase activity of Her‐2/neu by quercetin could indicate an lateration in the Her‐2/neu structure which promotes CHIP recruitments and down‐regulation of Her‐2/neu. We believe that by using quercetin, new therapeutic strategies can be developed to treat Her‐2/neu overexpressing cancers. J. Cell. Biochem. 105: 585–595, 2008.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Synthetic heterovalent inhibitors targeting recognition E3 components of the N-end rule pathway

Min Jae Lee; Krishnendu Pal; Takafumi Tasaki; Sayantani Roy; Yonghua Jiang; Jee Young An; Rajkumar Banerjee; Yong Tae Kwon

Multivalent binding allows high selectivity and affinity in a ligand–protein interaction. The N-end rule pathway is a ubiquitin (Ub)-dependent proteolytic system in which specific E3s, called N-recognins, mediate ubiquitylation through the recognition of types 1 and 2, destabilizing N-terminal residues of substrates. We recently identified a set of E3 Ub ligases (named UBR1–UBR7) containing the 70-residue UBR box, and we demonstrated that UBR1, UBR2, UBR4, and UBR5 can bind to destabilizing N-terminal residues. To explore a model of heterovalent interaction to the N-recognin family, we synthesized the small-molecule compound RF-C11, which bears two heterovalent ligands designed to target N-recognins, together with control molecules with two homovalent ligands. We demonstrate that heterovalent ligands of RF-C11 selectively and cooperatively bind cognate-binding sites of multiple N-recognins and thereby inhibit both types 1 and 2 N-end rule activities. Furthermore, the efficacy of heterovalent RF-C11 was substantially higher than homovalent inhibitors, which can target either a type 1 or type 2 site, providing the molecular basis of designing multivalent inhibitors for the control of specific intracellular pathways. In addition, RF-C11 exhibited higher efficacy and stability, compared with dipeptides bearing destabilizing N-terminal residues, which are known competitive inhibitors of the pathway. We also used the heterovalent compound to study the function of N-recognins in cardiac signaling. Using mouse and rat cardiomyocytes, we demonstrate that the N-end rule pathway has a cell-autonomous function in cardiac proliferation and hypertrophy, explaining our earlier results implicating the pathway in cardiac development and proteolysis of multiple cardiovascular regulators.


Cellular Signalling | 2010

c-Cbl-mediated degradation of TRAIL receptors is responsible for the development of the early phase of TRAIL resistance.

Jae J. Song; Miroslaw J. Szczepanski; So Young Kim; Joo Hang Kim; Jee Young An; Yong Tae Kwon; Marco A. Alcala; David L. Bartlett; Yong J. Lee

We previously reported two modes of development of acquired TRAIL resistance: early phase and late phase [1]. In these studies, we observed that greater Akt activity and the expression of Bcl-xL were related mainly to the late phase of acquired TRAIL resistance. Recently we became aware of a possible mechanism of early phase TRAIL resistance development through internalization and degradation of TRAIL receptors (DR4 and DR5). Our current studies demonstrate that TRAIL receptors rapidly diminish at the membrane as well as the cytoplasm within 4h after TRAIL exposure, but recover completely after one or two days. Our studies also reveal that Cbl, a ubiquitously expressed cytoplasmic adaptor protein, is responsible for the rapid degradation of TRAIL receptors; Cbl binds to them and induces monoubiquitination of these receptors concurrent with their degeneration soon after TRAIL exposure, creating the early phase of acquired TRAIL resistance.


PLOS ONE | 2010

The ubiquitin ligase Ubr2, a recognition E3 component of the N-end rule pathway, stabilizes Tex19.1 during spermatogenesis

Fang Yang; Yong Cheng; Jee Young An; Yong Tae Kwon; Sigrid Eckardt; N. Adrian Leu; K. John McLaughlin; Peijing Jeremy Wang

Ubiquitin E3 ligases target their substrates for ubiquitination, leading to proteasome-mediated degradation or altered biochemical properties. The ubiquitin ligase Ubr2, a recognition E3 component of the N-end rule proteolytic pathway, recognizes proteins with N-terminal destabilizing residues and plays an important role in spermatogenesis. Tex19.1 (also known as Tex19) has been previously identified as a germ cell-specific protein in mouse testis. Here we report that Tex19.1 forms a stable protein complex with Ubr2 in mouse testes. The binding of Tex19.1 to Ubr2 is independent of the second position cysteine of Tex19.1, a putative target for arginylation by the N-end rule pathway R-transferase. The Tex19.1-null mouse mutant phenocopies the Ubr2-deficient mutant in three aspects: heterogeneity of spermatogenic defects, meiotic chromosomal asynapsis, and embryonic lethality preferentially affecting females. In Ubr2-deficient germ cells, Tex19.1 is transcribed, but Tex19.1 protein is absent. Our results suggest that the binding of Ubr2 to Tex19.1 metabolically stabilizes Tex19.1 during spermatogenesis, revealing a new function for Ubr2 outside the conventional N-end rule pathway.


PLOS ONE | 2012

UBR2 of the N-end rule pathway is required for chromosome stability via histone ubiquitylation in spermatocytes and somatic cells.

Jee Young An; Euna Kim; Adriana Zakrzewska; Young Dong Yoo; Jun Min Jang; Dong Hoon Han; Min Jae Lee; Jai Wha Seo; Yong Jun Lee; Tae-You Kim; Dirk G. de Rooij; Bo Yeon Kim; Yong Tae Kwon

The N-end rule pathway is a proteolytic system in which its recognition components (N-recognins) recognize destabilizing N-terminal residues of short-lived proteins as an essential element of specific degrons, called N-degrons. The RING E3 ligases UBR2 and UBR1 are major N-recognins that share size (200 kDa), conserved domains and substrate specificities to N-degrons. Despite the known function of the N-end rule pathway in degradation of cytosolic proteins, the major phenotype of UBR2-deficient male mice is infertility caused by arrest of spermatocytes at meiotic prophase I. UBR2-deficient spermatocytes are impaired in transcriptional silencing of sex chromosome-linked genes and ubiquitylation of histone H2A. In this study we show that the recruitment of UBR2 to meiotic chromosomes spatiotemporally correlates to the induction of chromatin-associated ubiquitylation, which is significantly impaired in UBR2-deficient spermatocytes. UBR2 functions as a scaffold E3 that promotes HR6B/UbcH2-dependent ubiquitylation of H2A and H2B but not H3 and H4, through a mechanism distinct from typical polyubiquitylation. The E3 activity of UBR2 in histone ubiquitylation is allosterically activated by dipeptides bearing destabilizing N-terminal residues. Insufficient monoubiquitylation and polyubiquitylation on UBR2-deficient meiotic chromosomes correlate to defects in double strand break (DSB) repair and other meiotic processes, resulting in pachytene arrest at stage IV and apoptosis. Some of these functions of UBR2 are observed in somatic cells, in which UBR2 is a chromatin-binding protein involved in chromatin-associated ubiquitylation upon DNA damage. UBR2-deficient somatic cells show an array of chromosomal abnormalities, including hyperproliferation, chromosome instability, and hypersensitivity to DNA damage-inducing reagents. UBR2-deficient mice enriched in C57 background die upon birth with defects in lung expansion and neural development. Thus, UBR2, known as the recognition component of a major cellular proteolytic system, is associated with chromatin and controls chromatin dynamics and gene expression in both germ cells and somatic cells.

Collaboration


Dive into the Jee Young An's collaboration.

Top Co-Authors

Avatar

Yong Tae Kwon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Min Jae Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Yong J. Lee

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Jai Wha Seo

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Takafumi Tasaki

Kanazawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Alexander Varshavsky

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Ilia V. Davydov

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jae J. Song

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Bo Yeon Kim

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Euna Kim

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge