Network


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

Hotspot


Dive into the research topics where Jung Min Han is active.

Publication


Featured researches published by Jung Min Han.


Cell | 2012

Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway.

Jung Min Han; Seung Jae Jeong; Min Chul Park; G.T. Kim; Nam Hoon Kwon; Sang Hoon Ha; Sung Ho Ryu; Sunghoon Kim

Amino acids are required for activation of the mammalian target of rapamycin (mTOR) kinase, which regulates protein translation, cell size, and autophagy. However, the amino acid sensor that directly couples intracellular amino acid-mediated signaling to mTORC1 is unknown. Here we show that leucyl-tRNA synthetase (LRS) plays a critical role in amino acid-induced mTORC1 activation by sensing intracellular leucine concentration and initiating molecular events leading to mTORC1 activation. Mutation of LRS amino acid residues important for leucine binding renders the mTORC1 pathway insensitive to intracellular levels of amino acids. We show that LRS directly binds to Rag GTPase, the mediator of amino acid signaling to mTORC1, in an amino acid-dependent manner and functions as a GTPase-activating protein (GAP) for Rag GTPase to activate mTORC1. This work demonstrates that LRS is a key mediator for amino acid signaling to mTORC1.


Nature Genetics | 2003

Downregulation of FUSE-binding protein and c-myc by tRNA synthetase cofactor p38 is required for lung cell differentiation.

Min Jung Kim; Bum-Joon Park; Young-Sun Kang; Hyoung June Kim; Jae-Hyun Park; Jung Woo Kang; Sang Won Lee; Jung Min Han; Han-Woong Lee; Sunghoon Kim

p38 is associated with a macromolecular tRNA synthetase complex. It has an essential role as a scaffold for the complex, and genetic disruption of p38 in mice causes neonatal lethality. Here we investigated the molecular mechanisms underlying lethality of p38-mutant mice. p38-deficient mice showed defects in lung differentiation and respiratory distress syndrome. p38 was found to interact with FUSE-binding protein (FBP), a transcriptional activator of c-myc. Binding of p38 stimulated ubiquitination and degradation of FBP, leading to downregulation of c-myc, which is required for differentiation of functional alveolar type II cells. Transforming growth factor-β (TGF-β) induced p38 expression and promoted its translocation to nuclei for the regulation of FBP and c-myc. Thus, this work identified a new activity of p38 as a mediator of TGF-β signaling and its functional importance in the control of c-myc during lung differentiation.


FEBS Letters | 1998

Activation of phospholipase D1 by direct interaction with ADP‐ribosylation factor 1 and RalA

Jae Ho Kim; Sang Do Lee; Jung Min Han; Tae-Hoon Lee; Yong Kim; Jong Bae Park; J. David Lambeth; Pann-Ghill Suh; Sung Ho Ryu

Phospholipase D1 (PLD1) is known to be activated by ADP‐ribosylation factor 1 (ARF1). We report here that ARF1 co‐immunoprecipitates with PLD1 and that the ARF1‐dependent PLD activation is induced by the direct interaction between ARF1 and PLD1. We found that RalA, another member of the small GTP‐binding proteins, synergistically enhances the ARF1‐dependent PLD activity with an EC50 of about 30 nM. Using in vitro binding assay, we show that ARF1 and RalA directly interact with different sites of PLD1. The results suggest that the independent interactions of RalA and ARF1 with PLD1 are responsible for the synergistic activation.


Molecular Cell | 2009

LysRS serves as a key signaling molecule in the immune response by regulating gene expression.

Nurit Yannay-Cohen; Irit Carmi-Levy; Gillian Kay; Christopher Maolin Yang; Jung Min Han; D. Michael Kemeny; Sunghoon Kim; Hovav Nechushtan; Ehud Razin

Lysyl-tRNA synthetase (LysRS) was found to produce diadenosine tetraphosphate (Ap(4)A) in vitro more than two decades ago. Here, we used LysRS silencing in mast cells in combination with transfected normal and mutated LysRS to demonstrate in vivo the critical role played by LysRS in the production of Ap(4)A in response to immunological challenge. Upon such challenge, LysRS was phosphorylated on serine 207 in a MAPK-dependent manner, released from the multisynthetase complex, and translocated into the nucleus. We previously demonstrated that LysRS forms a complex with MITF and its repressor Hint-1, which is released from the complex by its binding to Ap(4)A, enabling MITF to transcribe its target genes. Here, silencing LysRS led to reduced Ap(4)A production in immunologically activated cells, which resulted in a lower level of MITF inducible genes. Our data demonstrate that specific LysRS serine 207 phosphorylation regulates Ap(4)A production in immunologically stimulated mast cells, thus implying that LysRS is a key mediator in gene regulation.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Secreted human glycyl-tRNA synthetase implicated in defense against ERK-activated tumorigenesis

Min Chul Park; Taehee Kang; Da Jin; Jung Min Han; Sang Bum Kim; Yun Jung Park; Kiwon Cho; Young Woo Park; Min Guo; Weiwei He; Xiang-Lei Yang; Paul Schimmel; Sung-Hoon Kim

Although adaptive systems of immunity against tumor initiation and destruction are well investigated, less understood is the role, if any, of endogenous factors that have conventional functions. Here we show that glycyl-tRNA synthetase (GRS), an essential component of the translation apparatus, circulates in serum and can be secreted from macrophages in response to Fas ligand that is released from tumor cells. Through cadherin (CDH)6 (K-cadherin), GRS bound to different ERK-activated tumor cells, and released phosphatase 2A (PP2A) from CDH6. The activated PP2A then suppressed ERK signaling through dephosphorylation of ERK and induced apoptosis. These activities were inhibited by blocking GRS with a soluble fragment of CDH6. With in vivo administration of GRS, growth of tumors with a high level of CDH6 and ERK activation were strongly suppressed. Our results implicate a conventional cytoplasmic enzyme in translation as an intrinsic component of the defense against ERK-activated tumor formation.


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

AIMP2/p38, the scaffold for the multi-tRNA synthetase complex, responds to genotoxic stresses via p53

Jung Min Han; Bum-Joon Park; Sang Gyu Park; Young Sun Oh; So Jung Choi; Sang-Won Lee; Soon-Kyung Hwang; Seung-Hee Chang; Myung-Haing Cho; Sunghoon Kim

AIMP2/p38 is a scaffolding protein required for the assembly of the macromolecular tRNA synthetase complex. Here, we describe a previously unknown function for AIMP2 as a positive regulator of p53 in response to genotoxic stresses. Depletion of AIMP2 increased resistance to DNA damage-induced apoptosis, and introduction of AIMP2 into AIMP2-deficient cells restored the susceptibility to apoptosis. Upon DNA damage, AIMP2 was phosphorylated, dissociated from the multi-tRNA synthetase complex, and translocated into the nuclei of cells. AIMP2 directly interacts with p53, thereby preventing MDM2-mediated ubiquitination and degradation of p53. Mutations in AIMP2, affecting its interaction with p53, hampered its ability to activate p53. Nutlin-3 recovered the level of p53 and the susceptibility to UV-induced cell death in AIMP2-deficient cells. This work demonstrates that AIMP2, a component of the translational machinery, functions as proapoptotic factor via p53 in response to DNA damage.


Journal of Biological Chemistry | 2006

Hierarchical Network between the Components of the Multi-tRNA Synthetase Complex IMPLICATIONS FOR COMPLEX FORMATION

Jung Min Han; Min Ji Lee; Sang Gyu Park; Sunhee Lee; Ehud Razin; Eung-Chil Choi; Sunghoon Kim

The macromolecular tRNA synthetase complex consists of nine different enzymes and three non-enzymatic factors. This complex was recently shown to be a novel signalosome, since many of its components are involved in signaling pathways in addition to their catalytic roles in protein synthesis. The structural organization and dynamic relationships of the components of the complex are not well understood. Here we performed a systematic depletion analysis to determine the effects of structural intimacy and the turnover of the components. The results showed that the stability of some components depended on their neighbors. Lysyl-tRNA synthetase was most independent of other components for its stability whereas it was most required for the stability of other components. Arginyl- and methionyl-tRNA synthetases had the opposite characteristics. Thus, the systematic depletion of the components revealed the functional reason for the complex formation and the assembly pattern of these multi-functional enzymes and their associated factors.


Biochemical and Biophysical Research Communications | 2003

Molecular network and functional implications of macromolecular tRNA synthetase complex

Jung Min Han; Jin Young Kim; Sunghoon Kim

Understanding the complex network and multi-functionality of proteins is one of the main objectives of post-genome research. Aminoacyl-tRNA synthetases (ARSs) are the family of enzymes that are essential for cellular protein synthesis and viability that catalyze the attachment of specific amino acids to their cognate tRNAs. However, a lot of evidence has shown that these enzymes are multi-functional proteins that are involved in diverse cellular processes, such as tRNA processing, RNA splicing and trafficking, rRNA synthesis, apoptosis, angiogenesis, and inflammation. In addition, mammalian ARSs form a macromolecular complex with three auxiliary factors or with the elongation factor complex. Although the functional meaning and physiological significance of these complexes are poorly understood, recent data on the molecular interactions among the components for the multi-ARS complex are beginning to provide insights into the structural organization and cellular functions. In this review, the molecular mechanism for the assembly and functional implications of the multi-ARS complex will be discussed.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Dual role of methionyl-tRNA synthetase in the regulation of translation and tumor suppressor activity of aminoacyl-tRNA synthetase-interacting multifunctional protein-3

Nam Hoon Kwon; Taehee Kang; Jin Young Lee; Hyo Hyun Kim; Hye Rim Kim; Jeena Hong; Young Sun Oh; Jung Min Han; Min Jeong Ku; Sang Yeol Lee; Sung-Hoon Kim

Mammalian methionyl-tRNA synthetase (MRS) plays an essential role in initiating translation by transferring Met to initiator tRNA (tRNAiMet). MRS also provides a cytosolic anchoring site for aminoacyl-tRNA synthetase-interacting multifunctional protein-3 (AIMP3)/p18, a potent tumor suppressor that is translocated to the nucleus for DNA repair upon DNA damage. However, the mechanism by which this enzyme mediates these two seemingly unrelated functions is unknown. Here we demonstrate that AIMP3 is released from MRS by UV irradiation-induced stress. Dissociation was induced by phosphorylation of MRS at Ser662 by general control nonrepressed-2 (GCN2) following UV irradiation. Substitution of Ser662 to Asp (S662D) induced a conformational change in MRS and significantly reduced its interaction with AIMP3. This mutant possessed significantly reduced MRS catalytic activity because of loss of tRNAMet binding, resulting in down-regulation of global translation. According to the Met incorporation assay using stable HeLa cells expressing MRS S662A or eukaryotic initiation factor-2 subunit-α (eIF2α) S51A, inactivation of GCN2-induced phosphorylation at eIF2α or MRS augmented the role of the other, suggesting a cross-talk between MRS and eIF2α for efficient translational inhibition. This work reveals a unique mode of regulation of global translation as mediated by aminoacyl-tRNA synthetase, specifically MRS, which we herein identified as a previously unidentified GCN2 substrate. In addition, our research suggests a dual role for MRS: (i) as a coregulator with eIF2α for GCN2-mediated translational inhibition; and (ii) as a coupler of translational inhibition and DNA repair following DNA damage by releasing bound tumor suppressor AIMP3 for its nuclear translocation.


Journal of Neurochemistry | 2002

Phospholipase D2 Activity Suppresses Hydrogen Peroxide‐Induced Apoptosis in PC12 Cells

Sang Do Lee; Byoung Dae Lee; Jung Min Han; Jae Ho Kim; Yong Kim; Pann-Ghill Suh; Sung Ho Ryu

Abstract: Phospholipase D (PLD) plays an important role as an effector in the membrane lipid‐mediated signal transduction. However, the precise physiological functions of PLD are not yet well understood. In this study, we examined the role of PLD activity in hydrogen peroxide (H2O2)‐induced apoptosis in rat pheochromocytoma (PC12) cells. Treatment of PC12 cells with H2O2 resulted in induction of apoptosis in these cells, which is accompanied by the activation of PLD. This H2O2‐induced apoptosis was enhanced remarkably when phosphatidic acid production by PLD was selectively inhibited by pretreating the PC12 cells with 1‐butanol. Expression of PLD2, but not of PLD1, correlated with increased H2O2‐induced PLD activity in a concentration‐ and time‐dependent manner. Concomitant with PLD activation, the PLD2 activity suppressed H2O2‐induced apoptosis in PC12 cells. Expression of PLD2 lipase‐inactive mutant (K758R) had no effect on either PLD activity or apoptosis. PLD2 activity also suppressed H2O2‐induced cleavage and activation of caspase‐3. Taken together, the results suggest that PLD2 activity is specifically up‐regulated by H2O2 in PC12 cells and that it plays a suppressive role in H2O2‐induced apoptosis.

Collaboration


Dive into the Jung Min Han's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sung Ho Ryu

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Yong Kim

Rockefeller University

View shared research outputs
Top Co-Authors

Avatar

Pann-Ghill Suh

Ulsan National Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Jae Ho Kim

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Nam Hoon Kwon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Sang Gyu Park

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Min Guo

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dae Gyu Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jin Young Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge