Network


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

Hotspot


Dive into the research topics where Hee-Jun Wee is active.

Publication


Featured researches published by Hee-Jun Wee.


Nature Communications | 2016

ARD1-mediated Hsp70 acetylation balances stress-induced protein refolding and degradation

Ji Hae Seo; Ji-Hyeon Park; Eun Lee; Tam Thuy Lu Vo; Hoon Choi; Jun Yong Kim; Jae Kyung Jang; Hee-Jun Wee; Hye Shin Lee; Se Hwan Jang; Zee Yong Park; Jaeho Jeong; Kong-Joo Lee; Seung-Hyeon Seok; Jinyoung Park; Bong-Jin Lee; Mi-Ni Lee; Goo Taeg Oh; Kyu-Won Kim

Heat shock protein (Hsp)70 is a molecular chaperone that maintains protein homoeostasis during cellular stress through two opposing mechanisms: protein refolding and degradation. However, the mechanisms by which Hsp70 balances these opposing functions under stress conditions remain unknown. Here, we demonstrate that Hsp70 preferentially facilitates protein refolding after stress, gradually switching to protein degradation via a mechanism dependent on ARD1-mediated Hsp70 acetylation. During the early stress response, Hsp70 is immediately acetylated by ARD1 at K77, and the acetylated Hsp70 binds to the co-chaperone Hop to allow protein refolding. Thereafter, Hsp70 is deacetylated and binds to the ubiquitin ligase protein CHIP to complete protein degradation during later stages. This switch is required for the maintenance of protein homoeostasis and ultimately rescues cells from stress-induced cell death in vitro and in vivo. Therefore, ARD1-mediated Hsp70 acetylation is a regulatory mechanism that temporally balances protein refolding/degradation in response to stress.


Acta Psychiatrica Scandinavica | 2008

Loss of asymmetry in D2 receptors of putamen in unaffected family members at increased genetic risk for schizophrenia

K. J. Lee; Jae Sung Lee; Sungjun Kim; Christoph U. Correll; Hee-Jun Wee; Sooyoung Yoo; Joonsoo Jeong; Dong-Youn Lee; Lee Si; Jung-Taek Kwon

Objective:  Dopamine dysregulation has been implicated in the pathophysiology of schizophrenia. The present study was performed to examine whether unaffected relatives at high genetic risk of schizophrenia have dopamine dysregulation in comparison with healthy controls.


Journal of Pineal Research | 2013

Melatonin inhibits visfatin-induced inducible nitric oxide synthase expression and nitric oxide production in macrophages

Young-Soon Kang; Yong-Gyu Kang; Hyun-Joo Park; Hee-Jun Wee; Hye-Ock Jang; Moon-Kyoung Bae; Soo-Kyung Bae

Aberrant expression of inducible nitric oxide synthase (iNOS) in macrophages, which has been reported to be suppressed by melatonin, has an important contribution in the development of pathological inflammation. Visfatin, an adipokine, regulates the expression of various inflammatory factors, leading to inflammation; however, the influence of visfatin on iNOS‐driven processes in macrophages is unclear. Here, we report the assessment of the role of visfatin in the regulation of iNOS gene expression in macrophages. Our data show that the levels of iNOS protein in peritoneal macrophages as well as nitric oxide (NO) in blood plasma were significantly lower after lipopolysaccharide treatment in visfatin+/− mice than those in the WT mice. In addition, visfatin increases iNOS mRNA and protein levels in RAW 264.7 cells, along with increasing production of NO. The enhancement of iNOS expression was prevented by treating the cells with inhibitors of the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3), nuclear factor (NF)‐κB, extracellular signal–regulated kinase 1/2, and c‐Jun N‐terminal kinase pathways. Our results also show that visfatin‐induced iNOS expression and NO production were significantly inhibited by melatonin, an effect that was closely associated with a reduction in phosphorylated JAK2/STAT3 levels and with the inhibition of p65 translocation into nucleus. In conclusion, our data show, for the first time, that melatonin suppresses visfatin‐induced iNOS upregulation in macrophages by inhibiting the STAT3 and NF‐κB pathways. Moreover, our data suggest that melatonin could be therapeutically useful for attenuating the development of visfatin–iNOS axis‐associated diseases.


Oncotarget | 2017

SAMHD1 acetylation enhances its deoxynucleotide triphosphohydrolase activity and promotes cancer cell proliferation

Eun Lee; Ji Hae Seo; Ji-Hyeon Park; Tam Thuy Lu Vo; Sunho An; Sung-Jin Bae; Hoang Le; Hye Shin Lee; Hee-Jun Wee; Danbi Lee; Young-Hwa Chung; Jeong A. Kim; Myoung-Kuk Jang; Soo Hyung Ryu; Ensil Yu; Se Hwan Jang; Zee Yong Park; Kyu-Won Kim

SAM domain and HD domain containing protein 1 (SAMHD1) is a deoxynucleotide triphosphohydrolase (dNTPase) that inhibits retroviruses by depleting intracellular deoxynucleotide triphosphates (dNTPs) in non-cycling myeloid cells. Although SAMHD1 is expressed ubiquitously throughout the human body, the molecular mechanisms regulating its enzymatic activity and function in non-immune cells are relatively unexplored. Here, we demonstrate that the dNTPase activity of SAMHD1 is regulated by acetylation, which promotes cell cycle progression in cancer cells. SAMHD1 is acetylated at residue lysine 405 (K405) in vitro and in vivo by an acetylatransferase, arrest defective protein 1 (ARD1). Acetylated SAMHD1 wildtype proteins have enhanced dNTPase activity in vitro, whereas non-acetylated arginine substituted mutants (K405R) do not. K405R mutant expressing cancer cells have reduced G1/S transition and slower proliferation compared to wildtype. SAMHD1 acetylation levels are strongest during the G1 phase, indicating a role during G1 phase. Collectively, these findings suggest that SAMHD1 acetylation enhances its dNTPase activity and promotes cancer cell proliferation. Therefore, SAMHD1 acetylation may be a potent therapeutic target for cancer treatment.


Journal of Biological Chemistry | 2014

Ninjurin1 Deficiency Attenuates Susceptibility of Experimental Autoimmune Encephalomyelitis in Mice

Bum Ju Ahn; Hoang Le; Min Wook Shin; Sung Jin Bae; Eun Lee; Hee-Jun Wee; Jong Ho Cha; Hyo Jong Lee; Hye Shin Lee; Jeong Hun Kim; Chang Yeon Kim; Ji Hae Seo; Eng H. Lo; Mi Ni Lee; Goo Taeg Oh; Guo Nan Yin; Ji Kan Ryu; Jun-Kyu Suh; Kyu-Won Kim

Background: Effect of Ninjurin1 deletion in the experimental autoimmune encephalomyelitis (EAE) mice has not been examined. Results: Ninjurin1 knock-out (KO) mice are resistance to EAE due to a defect of leukocyte recruitment into lesion sites. Conclusion: Ninjurin1 is a potent target molecule for treating inflammatory diseases such as multiple sclerosis. Significance: Our study proved contribution of Ninjurin1 in EAE pathogenesis in vivo and supports the importance of its targeting strategies. Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26–37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


PLOS ONE | 2014

AKAP12 Mediates Barrier Functions of Fibrotic Scars during CNS Repair

Jong Ho Cha; Hee-Jun Wee; Ji Hae Seo; Bum Ju Ahn; Ji Hyeon Park; Jun Mo Yang; Sae Won Lee; Eun Hee Kim; Ok Hee Lee; Ji Hoe Heo; Hyo Jong Lee; Irwin H. Gelman; Ken Arai; Eng H. Lo; Kyu-Won Kim

The repair process after CNS injury shows a well-organized cascade of three distinct stages: inflammation, new tissue formation, and remodeling. In the new tissue formation stage, various cells migrate and form the fibrotic scar surrounding the lesion site. The fibrotic scar is known as an obstacle for axonal regeneration in the remodeling stage. However, the role of the fibrotic scar in the new tissue formation stage remains largely unknown. We found that the number of A-kinase anchoring protein 12 (AKAP12)-positive cells in the fibrotic scar was increased over time, and the cells formed a structure which traps various immune cells. Furthermore, the AKAP12-positive cells strongly express junction proteins which enable the structure to function as a physical barrier. In in vivo validation, AKAP12 knock-out (KO) mice showed leakage from a lesion, resulting from an impaired structure with the loss of the junction complex. Consistently, focal brain injury in the AKAP12 KO mice led to extended inflammation and more severe tissue damage compared to the wild type (WT) mice. Accordingly, our results suggest that AKAP12-positive cells in the fibrotic scar may restrict excessive inflammation, demonstrating certain mechanisms that could underlie the beneficial actions of the fibrotic scar in the new tissue formation stage during the CNS repair process.


PLOS ONE | 2014

Nuclear translocation of hARD1 contributes to proper cell cycle progression.

Ji Hyeon Park; Ji Hae Seo; Hee-Jun Wee; Tam Thuy Lu Vo; Eun Lee; Hoon Choi; Jong Ho Cha; Bum Ju Ahn; Min Wook Shin; Sung Jin Bae; Kyu-Won Kim

Arrest defective 1 (ARD1) is an acetyltransferase that is highly conserved across organisms, from yeasts to humans. The high homology and widespread expression of ARD1 across multiple species and tissues signify that it serves a fundamental role in cells. Human ARD1 (hARD1) has been suggested to be involved in diverse biological processes, and its role in cell proliferation and cancer development has been recently drawing attention. However, the subcellular localization of ARD1 and its relevance to cellular function remain largely unknown. Here, we have demonstrated that hARD1 is imported to the nuclei of proliferating cells, especially during S phase. Nuclear localization signal (NLS)-deleted hARD1 (hARD1ΔN), which can no longer access the nucleus, resulted in cell morphology changes and cellular growth impairment. Notably, hARD1ΔN-expressing cells showed alterations in the cell cycle and the expression levels of cell cycle regulators compared to hARD1 wild-type cells. Furthermore, these effects were rescued when the nuclear import of hARD1 was restored by exogenous NLS. Our results show that hARD1 nuclear translocation mediated by NLS is required for cell cycle progression, thereby contributing to proper cell proliferation.


Nature Communications | 2014

Prompt meningeal reconstruction mediated by oxygen-sensitive AKAP12 scaffolding protein after central nervous system injury

Jong Ho Cha; Hee-Jun Wee; Ji Hae Seo; Bum Ju Ahn; Ji Hyeon Park; Jun Mo Yang; Sae Won Lee; Ok Hee Lee; Hyo Jong Lee; Irwin H. Gelman; Ken Arai; Eng H. Lo; Kyu-Won Kim

The meninges forms a critical epithelial barrier, which protects the central nervous system (CNS), and therefore its prompt reconstruction after CNS injury is essential for reducing neuronal damage. Meningeal cells migrate into the lesion site after undergoing an epithelial-mesenchymal transition (EMT) and repair the impaired meninges. However, the molecular mechanisms of meningeal EMT remain largely undefined. Here we show that TGF-β1 and retinoic acid (RA) released from the meninges, together with oxygen tension, could constitute the mechanism for rapid meningeal reconstruction. AKAP12 is an effector of this mechanism, and its expression in meningeal cells is regulated by integrated upstream signals composed of TGF-β1, RA and oxygen tension. Functionally, AKAP12 modulates meningeal EMT by regulating the TGF-β1-non-Smad-SNAI1 signalling pathway. Collectively, TGF-β1, RA and oxygen tension can modulate the dynamic change in AKAP12 expression, causing prompt meningeal reconstruction after CNS injury by regulating the transition between the epithelial and mesenchymal states of meningeal cells.


The Korean Journal of Physiology and Pharmacology | 2013

Mutant p53-Notch1 Signaling Axis Is Involved in Curcumin-Induced Apoptosis of Breast Cancer Cells.

Yun-Hee Bae; Jong Hyo Ryu; Hyun-Joo Park; Kwang Rok Kim; Hee-Jun Wee; Ok-Hee Lee; Hye-Ock Jang; Moon-Kyoung Bae; Kyu-Won Kim; Soo-Kyung Bae

Notch1 has been reported to be highly expressed in triple-negative and other subtypes of breast cancer. Mutant p53 (R280K) is overexpressed in MDA-MB-231 triple-negative human breast cancer cells. The present study aimed to determine whether the mutant p53 can be a potent transcriptional activator of the Notch1 in MDA-MB-231 cells, and explore the role of this mutant p53-Notch1 axis in curcumin-induced apoptosis. We found that curcumin treatment resulted in an induction of apoptosis in MDA-MB-231 cells, together with downregulation of Notch1 and its downstream target, Hes1. This reduction in Notch1 expression was determined to be due to the decreased activity of endogenous mutant p53. We confirmed the suppressive effect of curcumin on Notch1 transcription by performing a Notch1 promoter-driven reporter assay and identified a putative p53-binding site in the Notch1 promoter by EMSA and chromatin immunoprecipitation analysis. Overexpression of mutant p53 increased Notch1 promoter activity, whereas knockdown of mutant p53 by small interfering RNA suppressed Notch1 expression, leading to the induction of cellular apoptosis. Moreover, curcumin-induced apoptosis was further enhanced by the knockdown of Notch1 or mutant p53, but it was decreased by the overexpression of active Notch1. Taken together, our results demonstrate, for the first time, that Notch1 is a transcriptional target of mutant p53 in breast cancer cells and suggest that the targeting of mutant p53 and/or Notch1 may be combined with a chemotherapeutic strategy to improve the response of breast cancer cells to curcumin.


Biochemical and Biophysical Research Communications | 2012

The N-terminal ectodomain of Ninjurin1 liberated by MMP9 has chemotactic activity

Bum Ju Ahn; Hoang Le; Min Wook Shin; Sung Jin Bae; Eun Lee; Hee-Jun Wee; Jong Ho Cha; Ji Hyeon Park; Hye Shin Lee; Hyo Jong Lee; Hyunsook Jung; Zee Yong Park; Sang Ho Park; Byung Woo Han; Ji Hae Seo; Eng H. Lo; Kyu-Won Kim

Ninjurin1 is known as an adhesion molecule promoting leukocyte trafficking under inflammatory conditions. However, the posttranslational modifications of Ninjurin1 are poorly understood. Herein, we defined the proteolytic cleavage of Ninjurin1 and its functions. HEK293T cells overexpressing the C- or N-terminus tagging mouse Ninjurin1 plasmid produced additional cleaved forms of Ninjurin1 in the lysates or conditioned media (CM). Two custom-made anti-Ninjurin1 antibodies, Ab(1-15) or Ab(139-152), specific to the N- or C-terminal regions of Ninjurin1 revealed the presence of its shedding fragments in the mouse liver and kidney lysates. Furthermore, Matrix Metalloproteinase (MMP) 9 was responsible for Ninjurin1 cleavage between Leu(56) and Leu(57). Interestingly, the soluble N-terminal Ninjurin1 fragment has structural similarity with well-known chemokines. Indeed, the CM from HEK293T cells overexpressing the GFP-mNinj1 plasmid was able to attract Raw264.7 cells in trans-well assay. Collectively, we suggest that the N-terminal ectodomain of mouse Ninjurin1, which may act as a chemoattractant, is cleaved by MMP9.

Collaboration


Dive into the Hee-Jun Wee's collaboration.

Top Co-Authors

Avatar

Kyu-Won Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ji Hae Seo

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bum Ju Ahn

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Eun Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hye Shin Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jong Ho Cha

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Min Wook Shin

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hoang Le

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge