Network


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

Hotspot


Dive into the research topics where Wongi Seol is active.

Publication


Featured researches published by Wongi Seol.


Molecular Brain | 2015

Leucine-Rich Repeat Kinase 2 (LRRK2) phosphorylates p53 and induces p21(WAF1/CIP1) expression.

Dong Hwan Ho; Hye-Jung Kim; Jisun Kim; Hyuna Sim; Hyunjun Ahn; Janghwan Kim; Hyemyung Seo; Kwang Chul Chung; Bum Joon Park; Ilhong Son; Wongi Seol

BackgroundLeucine-rich repeat kinase 2 (LRRK2) is a gene in which a mutation causes Parkinson’s disease (PD), and p53 is a prototype tumor suppressor. In addition, activation of p53 in patient with PD has been reported by several studies. Because phosphorylation of p53 is critical for regulating its activity and LRRK2 is a kinase, we tested whether p53 is phosphorylated by LRRK2.ResultsLRRK2 phosphorylates threonine (Thr) at TXR sites in an in vitro kinase assay, and the T304 and T377 were identified as putative phosphorylated residues. An increase of phospho-Thr in the p53 TXR motif was confirmed in the cells overexpressing G2019S, and human induced pluripotent stem (iPS) cells of a G2019S carrier. Interactions between LRRK2 and p53 were confirmed by co-immunoprecipitation of lysates of differentiated SH-SY5Y cells. LRRK2 mediated p53 phosphorylation translocalizes p53 predominantly to nucleus and increases p21WAF1/CIP1 expression in SH-SY5Y cells based on reverse transcription-polymerase chain reaction and Western blot assay results. The luciferase assay using the p21WAF1/CIP1 promoter-reporter also confirmed that LRRK2 kinase activity increases p21 expression. Exogenous expression of G2019S and the phosphomimetic p53 T304/377D mutants increased expression of p21WAF1/CIP1 and cleaved PARP, and cytotoxicity in the same cells. We also observed increase of p21 expression in rat primary neuron cells after transient expression of p53 T304/377D mutants and the mid-brain lysates of the G2019S transgenic mice.Conclusionp53 is a LRRK2 kinase substrate. Phosphorylation of p53 by LRRK2 induces p21WAF1/CIP1 expression and apoptosis in differentiated SH-SY5Y cells and rat primary neurons.


Human Molecular Genetics | 2016

Leucine-rich repeat kinase 2 exacerbates neuronal cytotoxicity through phosphorylation of histone deacetylase 3 and histone deacetylation

Kyung Ah Han; Woo Hyun Shin; Sungyeon Jung; Wongi Seol; Hyemyung Seo; CheMyong Ko; Kwang Chul Chung

Parkinsons disease (PD) is characterized by slow, progressive degeneration of dopaminergic neurons in the substantia nigra. The cause of neuronal death in PD is largely unknown, but several genetic loci, including leucine-rich repeat kinase 2 (LRRK2), have been identified. LRRK2 has guanosine triphosphatase (GTPase) and kinase activities, and mutations in LRRK2 are the major cause of autosomal-dominant familial PD. Histone deacetylases (HDACs) remove acetyl groups from lysine residues on histone tails, promoting transcriptional repression via condensation of chromatin. Here, we demonstrate that LRRK2 binds to and directly phosphorylates HDAC3 at Ser-424, thereby stimulating HDAC activity. Specifically, LRRK2 promoted the deacetylation of Lys-5 and Lys-12 on histone H4, causing repression of gene transcription. Moreover, LRRK2 stimulated nuclear translocation of HDAC3 via the phoshorylation of karyopherin subunit α2 and α6. HDAC3 phosphorylation and its nuclear translocation were increased in response to 6-hydroxydopamine (6-OHDA) treatment. LRRK2 also inhibited myocyte-specific enhancer factor 2D activity, which is required for neuronal survival. LRRK2 ultimately promoted 6-OHDA-induced cell death via positive modulation of HDAC3. These findings suggest that LRRK2 affects epigenetic histone modification and neuronal survival by facilitating HDAC3 activity and regulating its localization.


Experimental Neurobiology | 2018

LRRK2 Kinase Activity Induces Mitochondrial Fission in Microglia via Drp1 and Modulates Neuroinflammation

Dong Hwan Ho; A Reum Je; Haejin Lee; Ilhong Son; Hee-Seok Kweon; Hyung-Gun Kim; Wongi Seol

Leucine-rich repeat kinase 2 (LRRK2) mutations are the most common genetic cause of Parkinsons disease (PD). LRRK2 contains a functional kinase domain and G2019S, the most prevalent LRRK2 pathogenic mutation, increases its kinase activity. LRRK2 regulates mitochondria morphology and autophagy in neurons. LPS treatment increases LRRK2 protein level and mitochondrial fission in microglia, and down-regulation of LRRK2 expression or inhibition of its kinase activity attenuates microglia activation. Here, we evaluated the direct role of LRRK2 G2019S in mitochondrial dynamics in microglia. Initial observation of microglia in G2019S transgenic mice revealed a decrease in mitochondrial area and shortage of microglial processes compared with their littermates. Next, we elucidated the molecular mechanisms of these phenotypes. Treatment of BV2 cells and primary microglia with LPS enhanced mitochondrial fission and increased Drp1, a mitochondrial fission marker, as previously reported. Importantly, both phenotypes were rescued by treatment with GSK2578215A, a LRRK2 kinase inhibitor. Finally, the protein levels of CD68, an active microglia marker, Drp1 and TNF-α were significantly higher in brain lysates of G2019S transgenic mice compared with the levels in their littermates. Taken together, our data suggest that LRRK2 could promote microglial mitochondrial alteration via Drp1 in a kinase-dependent manner, resulting in stimulation of pro-inflammatory responses. This mechanism in microglia might be a potential target to develop PD therapy since neuroinflammation by active microglia is a major symptom of PD.


Frontiers in Cellular Neuroscience | 2017

Leucine-Rich Repeat Kinase 2 (LRRK2) Stimulates IL-1β-Mediated Inflammatory Signaling through Phosphorylation of RCAN1

Kyung Ah Han; Lang Yoo; Jee Y. Sung; Sun A. Chung; Ji Won Um; Hyeyoung Kim; Wongi Seol; Kwang C. Chung

Leucine-rich repeat kinase 2 (LRRK2) is a Ser/Thr kinase having mixed lineage kinase-like and GTPase domains, controlling neurite outgrowth and neuronal cell death. Evidence suggests that LRRK2 is involved in innate immune response signaling, but the underlying mechanism is yet unknown. A novel protein inhibitor of phosphatase 3B, RCAN1, is known to positively regulate inflammatory signaling through modulation of several intracellular targets of interleukins in immune cells. In the present study, we report that LRRK2 phosphorylates RCAN1 (RCAN1-1S) and is markedly up-regulated during interleukin-1β (IL-1β) treatment. During IL-1β treatment, LRRK2-mediated phosphorylation of RCAN1 promoted the formation of protein complexes, including that between Tollip and RCAN1. LRRK2 decreased binding between Tollip and IRAK1, which was accompanied by increased formation of the IRAK1-TRAF6 complex. TAK1 activity was significantly enhanced by LRRK2. Furthermore, LRRK2 enhanced transcriptional activity of NF-κB and cytokine IL-8 production. These findings suggest that LRRK2 might be important in positively modulating IL-1β-mediated signaling through selective phosphorylation of RCAN1.


Parkinson's Disease | 2018

Oxidized DJ-1 Levels in Urine Samples as a Putative Biomarker for Parkinson’s Disease

Jihoon Jang; Soyeon Jeong; Sung Ik Lee; Wongi Seol; Hyemyung Seo; Ilhong Son; Dong Hwan Ho

Parkinsons disease (PD) is the second most common neurodegenerative disease. Oxidative stress is the most critical risk factor for neurodegenerative diseases, including Alzheimers disease (AD) and Huntingtons disease (HD). Numerous reports have demonstrated that oxidative stress aggravates cytotoxicity in dopaminergic neurons and accelerates the formation of protein inclusions. In addition, oxidative stress, such as 4-hydroxynonenal (HNE), oxidized protein, and dopamine quinone, are related to PD progression. DJ-1 is a PD-causative gene, and it plays a pivotal role as a sensor and eliminator of oxidative stress. Several studies have shown that oxidized DJ-1 (OxiDJ-1) formation is induced by oxidative stress. Hence, previous studies suggest that oxidized DJ-1 could be a biomarker for PD. We previously reported higher DJ-1 levels in Korean male PD patient urine exosomes than male non-PD controls. We speculate that OxiDJ-1 levels in PD patient urine might be higher than that in non-PD controls. In this study, we established an ELISA for OxiDJ-1 using recombinant DJ-1 treated with H2O2. Using Western blot assay and ELISA, we confirmed an increase of OxiDJ-1 from HEK293T cells treated with H2O2. Using our ELISA, we observed significantly higher, 2-fold, OxiDJ-1 levels in the urine of Korean PD patients than in non-PD controls.


Animal Cells and Systems | 2018

Characterization of Parkinson’s disease-related pathogenic TMEM230 mutants

Daleum Nam; Hye-Jung Kim; Dong-Joo Choi; Yun-Hee Bae; Byoung Dae Lee; Ilhong Son; Wongi Seol

ABSTRACT Parkinson’s disease (PD) is the second most common neurodegenerative disease. Although most PD cases are sporadic, 5–10% of them are hereditary and several pathogenic mutations in related genes have been identified. Mutations in TMEM230 were recently identified as a cause of autosomal dominant PD. However, the basic properties of the mutant proteins are not yet known. We examined stability and neurotoxicity, important characteristics of PD pathogenesis-related proteins, of WT TMEM230 and two pathogenic mutants, R78L and PG5ext, in a dopaminergic neuronal cell line. Our study showed that amount of protein expressed in the same vector backbone was R78L > WT > PG5ext. The stabilities of the mutant proteins were similar to each other, but lower than that of the WT. In addition, overexpression of mutants and WT TMEM230 caused similar levels of neurotoxicity upon MPP+ treatment when compared to the cells transfected with an empty vector. Because the proteins encoded by two PD-causing genes, TMEM230 and LRRK2, function in vesicle trafficking, we tested whether they interact. LRRK2 neither interacts with, nor phosphorylates TMEM230. We also investigated the levels of several Rab proteins (Rab1A, 5, 7, 8A and 11) involved in vesicle trafficking after TMEM230 overexpression. However, there was no clear difference of any Rab proteins among cells transfected with an empty vector, TMEM230 WT and mutants-expressing cells, suggesting that TMEM230 does not directly regulate these Rab proteins. Thus, these TMEM230 PG5ext and R78L mutant proteins are not distinctly different from the WT proteins except for their stability. Abbreviations: LRRK2: Leucine-rich repeat kinase 2; PD: Parkinsons disease; AD: Alzheimers disease; RT-PCR: reverse transcription-polymerase chain reaction; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; FACS: fluorescence-activated cell sorting; PBS: phosphate buffered saline; FBS: fetal bovine serum; PI: propidium iodide.


Animal Cells and Systems | 2018

Effect of leucine-rich repeat kinase 2 (LRRK2) on protein synthesis

Hye-Jung Kim; Ilhong Son; Wongi Seol

ABSTRACT Mutations in the leucine-rich repeat kinase 2 (LRRK2) cause Parkinson’s disease (PD) in an autosomal dominant manner. Pathogenic mutations of LRRK2 such as G2019S and R1441C have been observed as common genetic causes of PD. Recently, LRRK2 has been reported to increase the reporter protein synthesis in both cap-dependent and -independent manners via phosphorylation of the ribosomal protein RPS15. In this study, we tested whether LRRK2 recombinant protein would directly increase protein synthesis using a well-defined in vitro coupled transcription/translation system. Addition of commercial full-length LRRK2 or GST-fused N-terminal-deleted LRRK2 recombinant proteins to the system showed no change of protein synthesis, as measured by luciferase reporter activity. In addition, the SUnSET assay to measure newly synthesized cellular proteins showed that G2019S overexpression had a minimal effect on the total protein amount. However, we confirmed the previous result that G2019S overexpression increased the amount of protein synthesized from an exogenous gene, Flag-VAMP2, which was transfected as a reporter, whereas there was no significant change in the amount of the Flag-VAMP2 mRNA. Inhibition of protein degradation showed that protein accumulation in the vector control was higher than that of the G2019S overexpression vector. Our results suggest that LRRK2 protein influences the amount of protein by inhibiting protein degradation rather than by directly stimulating translation.


Animal Cells and Systems | 2018

Increase in anti-apoptotic molecules, nucleolin, and heat shock protein 70, against upregulated LRRK2 kinase activity

Jihoon Jang; Hakjin Oh; Daleum Nam; Wongi Seol; Mi Kyoung Seo; Sung Woo Park; Hyung Gun Kim; Hyemyung Seo; Ilhong Son; Dong Hwan Ho

ABSTRACT Leucine-rich repeat kinase 2 (LRRK2) is involved in Parkinson’s disease (PD) pathology. A previous study showed that rotenone treatment induced apoptosis, mitochondrial damage, and nucleolar disruption via up-regulated LRRK2 kinase activity, and these effects were rescued by an LRRK2 kinase inhibitor. Heat-shock protein 70 (Hsp70) is an anti-oxidative stress chaperone, and overexpression of Hsp70 enhanced tolerance to rotenone. Nucleolin (NCL) is a component of the nucleolus; overexpression of NCL reduced cellular vulnerability to rotenone. Thus, we hypothesized that rotenone-induced LRRK2 activity would promote changes in neuronal Hsp70 and NCL expressions. Moreover, LRRK2 G2019S, the most prevalent LRRK2 pathogenic mutant with increased kinase activity, could induce changes in Hsp70 and NCL expression. Rotenone treatment of differentiated SH-SY5Y (dSY5Y) cells increased LRKK2 levels and kinase activity, including phospho-S935-LRRK2, phospho-S1292-LRRK2, and the phospho-moesin/moesin ratio, in a dose-dependent manner. Neuronal toxicity and the elevation of cleaved poly (ADP-ribose) polymerase, NCL, and Hsp70 were increased by rotenone. To validate the induction of NCL and Hsp70 expression in response to rotenone, cycloheximide (CHX), a protein synthesis blocker, was administered with rotenone. Post-rotenone increased NCL and Hsp70 expression was repressed by CHX; whereas, rotenone-induced kinase activity and apoptotic toxicity remained unchanged. Transient expression of G2019S in dSY5Y increased the NCL and Hsp70 levels, while administration of a kinase inhibitor diminished these changes. Similar results were observed in rat primary neurons after rotenone treatment or G2019S transfection. Brains from G2019S-transgenic mice also showed increased NCL and Hsp70 levels. Accordingly, LRRK2 kinase inhibition might prevent oxidative stress-mediated PD progression. Abbreviations: 6-OHDA: 6-hydroxydopamine; CHX: cycloheximide; dSY5Y: differentiated SH-SY5Y; g2019S tg: g2019S transgenic mouse; GSK/A-KI: GSK2578215A kinase inhibitor; HSP70: heat shock protein 70; LDH: lactose dehydrogenase; LRRK2: leucine rich-repeat kinase 2; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; myc-GS LRRK2: myc-tagged g2019S LRRK2; NCL: nucleolin; PARP: poly(ADP-ribose) polymerase; PD: Parkinson’s disease; PINK1: PTEN-induced putative kinase 1; pmoesin: phosphorylated moesin at t558; ROS: reactive oxygen species GRAPHICAL ABSTRACT


Molecular Brain | 2017

Correction to: Leucine-Rich Repeat Kinase 2 (LRRK2) phosphorylates p53 and induces p21 WAF1/CIP1 expression

Dong Hwan Ho; Hye-Jung Kim; Jisun Kim; Hyuna Sim; Hyunjun Ahn; Janghwan Kim; Hyemyung Seo; Kwang Chul Chung; Bum-Joon Park; Ilhong Son; Wongi Seol

Correction to: Molecular brain (2015) 8:54 DOI 10.1186/s13041-015-0145-7 In the original publication of this article [1], published on 18 September 2015, it was noticed that the information on a plasmid expressing HA-tagged human p53 gene is wrong. In this Correction the incorrect and correct information on this gene are shown (and marked bold). In the first paragraph of the Methods section, the plasmid expressing HA-tagged human p53 gene is described as followed: Plasmids expressing the HA-tagged human p53 gene (16434) and FLAG-p21 (16240) were purchased from Addgene (Cambridge, MA, USA) and shLRRK2 plasmid specifically inhibiting expression of endogenous human LRRK2 (TI202451) from ORIGENE (Rockville, MD, USA). The correct description of the plasmid expressing HAtagged human p53 gene is: Ha-p53 WT was previously described [2]. FLAG-p21 was purchased from Addgene (#16240, Cambridge, MA, USA) and shLRRK2 plasmid specifically inhibiting expression of endogenous human LRRK2 (TI202451) from ORIGENE (Rockville, MD, USA).


Biochemical and Biophysical Research Communications | 2017

Phosphorylation of p53 by LRRK2 induces microglial tumor necrosis factor α-mediated neurotoxicity

Dong Hwan Ho; Wongi Seol; Jin Hwan Eun; Ilhong Son

Collaboration


Dive into the Wongi Seol's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hyuna Sim

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Hyunjun Ahn

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Janghwan Kim

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge