Network


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

Hotspot


Dive into the research topics where Hosouk Joung is active.

Publication


Featured researches published by Hosouk Joung.


Circulation | 2011

Casein Kinase-2α1 Induces Hypertrophic Response by Phosphorylation of Histone Deacetylase 2 S394 and its Activation in the Heart

Gwang Hyeon Eom; Young Kuk Cho; Jeong-Hyeon Ko; Sera Shin; Nakwon Choe; Yoojung Kim; Hosouk Joung; Hyung-Seok Kim; Kwang-Il Nam; Hae Jin Kee; Hyun Kook

Background— Cardiac hypertrophy is characterized by transcriptional reprogramming of fetal gene expression, and histone deacetylases (HDACs) are tightly linked to the regulation of those genes. We previously demonstrated that activation of HDAC2, 1 of the class I HDACs, mediates hypertrophy. Here, we show that casein kinase-2&agr;1 (CK2&agr;1)–dependent phosphorylation of HDAC2 S394 is required for the development of cardiac hypertrophy. Methods and Results— Hypertrophic stimuli phosphorylated HDAC2 S394, which was necessary for its enzymatic activation, and therefore the development of hypertrophic phenotypes in rat neonatal cardiomyocytes or in isoproterenol-administered mice hearts. Transgenic mice overexpressing HDAC2 wild type exhibited cardiac hypertrophy, whereas those expressing phosphorylation-resistant HDAC2 S394A did not. Compared with that in age-matched normal human hearts, phosphorylation of HDAC2 S394 was dramatically increased in patients with hypertrophic cardiomyopathy. Hypertrophy-induced phosphorylation of HDAC2 S394 and its enzymatic activity were completely blocked either by CK2 blockers or by CK2&agr;1 short interfering RNA. Hypertrophic stimuli led CK2&agr;1 to be activated, and its chemical inhibitors blocked hypertrophy in both phenylephrine-treated cardiomyocytes and isoproterenol-administered mice. CK2&agr;1-transgenic mice developed hypertrophy, which was attenuated by administration of trichostatin A, an HDAC inhibitor. Overexpression of CK2&agr;1 caused hypertrophy in cardiomyocytes, whereas chemical inhibitors of both CK2 and HDAC as well as HDAC2 S394A blunted it. Hypertrophy in CK2&agr;1-transgenic mice was exaggerated by crossing these mice with wild-type-HDAC2-overexpressing mice. By contrast, however, it was blocked when CK2&agr;1-transgenic mice were crossed with HDAC2 S394A-transgenic mice. Conclusions— We have demonstrated a novel mechanism in the development of cardiac hypertrophy by which CK2 activates HDAC2 via phosphorylating HDAC2 S394.


Journal of Biological Chemistry | 2009

Enhancer of polycomb1 acts on serum response factor to regulate skeletal muscle differentiation.

Ju-Ryoung Kim; Hae Jin Kee; Jiyoung Kim; Hosouk Joung; Kwang-Il Nam; Gwang Hyeon Eom; Nakwon Choe; Hyung Suk Kim; Jeong Chul Kim; Sang Beom Seo; Hyun Kook

Skeletal muscle differentiation is well regulated by a series of transcription factors. We reported previously that enhancer of polycomb1 (Epc1), a chromatin protein, can modulate skeletal muscle differentiation, although the mechanisms of this action have yet to be defined. Here we report that Epc1 recruits both serum response factor (SRF) and p300 to induce skeletal muscle differentiation. Epc1 interacted physically with SRF. Transfection of Epc1 to myoblast cells potentiated the SRF-induced expression of skeletal muscle-specific genes as well as multinucleation. Proximal CArG box in the skeletal α-actin promoter was responsible for the synergistic activation of the promoter-luciferase. Epc1 knockdown caused a decrease in the acetylation of histones associated with serum response element (SRE) of the skeletal α-actin promoter. The Epc1·SRF complex bound to the SRE, and the knockdown of Epc1 resulted in a decrease in SRF binding to the skeletal α-actin promoter. Epc1 recruited histone acetyltransferase activity, which was potentiated by cotransfection with p300 but abolished by si-p300. Epc1 directly bound to p300 in myoblast cells. Epc1+/− mice showed distortion of skeletal α-actin, and the isolated myoblasts from the mice had impaired muscle differentiation. These results suggest that Epc1 is required for skeletal muscle differentiation by recruiting both SRF and p300 to the SRE of muscle-specific gene promoters.


Nature Communications | 2016

MDM2 E3 ligase-mediated ubiquitination and degradation of HDAC1 in vascular calcification.

Duk-Hwa Kwon; Gwang Hyeon Eom; Jeong Hyeon Ko; Sera Shin; Hosouk Joung; Nakwon Choe; Yoon Seok Nam; Hyun-Ki Min; Taewon Kook; Somy Yoon; Wanseok Kang; Yong Sook Kim; Hyung-Seok Kim; Hyuck Choi; Jeong-Tae Koh; Nacksung Kim; Youngkeun Ahn; Hyun-Jai Cho; In-Kyu Lee; Dong Ho Park; Kyoungho Suk; Sang Beom Seo; Erin R. Wissing; Susan M. Mendrysa; Kwang-Il Nam; Hyun Kook

Vascular calcification (VC) is often associated with cardiovascular and metabolic diseases. However, the molecular mechanisms linking VC to these diseases have yet to be elucidated. Here we report that MDM2-induced ubiquitination of histone deacetylase 1 (HDAC1) mediates VC. Loss of HDAC1 activity via either chemical inhibitor or genetic ablation enhances VC. HDAC1 protein, but not mRNA, is reduced in cell and animal calcification models and in human calcified coronary artery. Under calcification-inducing conditions, proteasomal degradation of HDAC1 precedes VC and it is mediated by MDM2 E3 ubiquitin ligase that initiates HDAC1 K74 ubiquitination. Overexpression of MDM2 enhances VC, whereas loss of MDM2 blunts it. Decoy peptide spanning HDAC1 K74 and RG 7112, an MDM2 inhibitor, prevent VC in vivo and in vitro. These results uncover a previously unappreciated ubiquitination pathway and suggest MDM2-mediated HDAC1 ubiquitination as a new therapeutic target in VC.


Atherosclerosis | 2012

Enhancer of polycomb1 lessens neointima formation by potentiation of myocardin-induced smooth muscle differentiation.

Hosouk Joung; Jin-Sook Kwon; Ju-Ryoung Kim; Sera Shin; Wanseok Kang; Youngkeun Ahn; Hyun Kook; Hae Jin Kee

OBJECTIVE Previously, we reported that enhancer of polycomb1 (Epc1) induces skeletal muscle differentiation through the serum response factor (SRF). Considering that SRF plays a critical role in vascular smooth muscle cell (VSMC) differentiation, we expected that Epc1 also works in VSMCs. Here we examined the effect of Epc1 on neointima formation after arterial balloon injury and the underlying mechanism. METHODS Epc1 expression was examined in carotid artery injury or VSMC models. Interaction with myocardin (Myocd), a master regulator of smooth muscle differentiation, was examined by immunoprecipitation or promoter analysis with smooth muscle (SM) 22α promoter. Finally, we investigated whether local delivery of Epc1 regulated neointimal formation after injury. RESULTS Epc1 expression was down-regulated during proliferation induced by platelet-derived growth factor BB, whereas it was upregulated during differentiation in VSMCs. Forced expression of Epc1 induced VSMC differentiation. Epc1 physically interacted with Myocd to synergistically activate SM22α promoter activity. Transient transfection of Epc1 enhanced the physical interaction between Myocd and SRF, whereas that interaction was reduced when A10 cells were treated with siRNA for Epc1. Local delivery of Epc1 significantly reduced neointima formation induced by balloon injury. CONCLUSIONS Our results indicate that Epc1 induces VSMC differentiation by interacting with Myocd to induce SRF-dependent smooth muscle genes. We propose that Epc1 acts as a novel negative regulator of neointima formation after carotid injury.


Cellular Signalling | 2014

Ret finger protein mediates Pax7-induced ubiquitination of MyoD in skeletal muscle atrophy.

Hosouk Joung; Gwang Hyeon Eom; Nakwon Choe; Hye Mi Lee; Jeong-Hyeon Ko; Duk-Hwa Kwon; Yoon Seok Nam; Hyun-Ki Min; Sera Shin; Jeewon Kook; Young Kuk Cho; Jeong Chul Kim; Sang Beom Seo; Yung Hong Baik; Kwang-Il Nam; Hyun Kook

Skeletal muscle atrophy results from the net loss of muscular proteins and organelles and is caused by pathologic conditions such as nerve injury, immobilization, cancer, and other metabolic diseases. Recently, ubiquitination-mediated degradation of skeletal-muscle-specific transcription factors was shown to be involved in muscle atrophy, although the mechanisms have yet to be defined. Here we report that ret finger protein (RFP), also known as TRIM27, works as an E3 ligase in Pax7-induced degradation of MyoD. Muscle injury induced by sciatic nerve transection up-regulated RFP and RFP physically interacted with both Pax7 and MyoD. RFP and Pax7 synergistically reduced the protein amounts of MyoD but not the mRNA. RFP-induced reduction of MyoD protein was blocked by proteasome inhibitors. The Pax7-induced reduction MyoD was attenuated by RFP siRNA and by MG132, a proteasome inhibitor. RFPΔR, an RFP construct that lacks the RING domain, failed to reduce MyoD amounts. RFP ubiquitinated MyoD, but RFPΔR failed to do so. Forced expression of RFP, but not RFPΔR, enhanced Pax7-induced ubiquitination of MyoD, whereas RFP siRNA blocked the ubiquitination. Sciatic nerve injury-induced muscle atrophy as well the reduction in MyoD was attenuated in RFP knockout mice. Taken together, our results show that RFP works as a novel E3 ligase in the Pax7-mediated degradation of MyoD in response to skeletal muscle atrophy.


Circulation Research | 2014

Small Heterodimer Partner Blocks Cardiac Hypertrophy by Interfering with GATA6 Signaling

Yoon Seok Nam; Yoojung Kim; Hosouk Joung; Duk-Hwa Kwon; Nakwon Choe; Hyun-Ki Min; Y. J. Kim; Hyung-Seok Kim; Don-Kyu Kim; Young Kuk Cho; Yong Hoon Kim; Kwang-Il Nam; Hyoung Chul Choi; Dong Ho Park; Kyoungho Suk; In-Kyu Lee; Youngkeun Ahn; Chul-Ho Lee; Hueng-Sik Choi; Gwang Hyeon Eom; Hyun Kook

Rationale: Small heterodimer partner (SHP; NR0B2) is an atypical orphan nuclear receptor that lacks a conventional DNA-binding domain. Through interactions with other transcription factors, SHP regulates diverse biological events, including glucose metabolism in liver. However, the role of SHP in adult heart diseases has not yet been demonstrated. Objective: We aimed to investigate the role of SHP in adult heart in association with cardiac hypertrophy. Methods and Results: The roles of SHP in cardiac hypertrophy were tested in primary cultured cardiomyocytes and in animal models. SHP-null mice showed a hypertrophic phenotype. Hypertrophic stresses repressed the expression of SHP, whereas forced expression of SHP blocked the development of hypertrophy in cardiomyocytes. SHP reduced the protein amount of Gata6 and, by direct physical interaction with Gata6, interfered with the binding of Gata6 to GATA-binding elements in the promoter regions of natriuretic peptide precursor type A. Metformin, an antidiabetic agent, induced SHP and suppressed cardiac hypertrophy. The metformin-induced antihypertrophic effect was attenuated either by SHP small interfering RNA in cardiomyocytes or in SHP-null mice. Conclusions: These results establish SHP as a novel antihypertrophic regulator that acts by interfering with GATA6 signaling. SHP may participate in the metformin-induced antihypertrophic response.


Experimental and Molecular Medicine | 2018

Sumoylation of histone deacetylase 1 regulates MyoD signaling during myogenesis

Hosouk Joung; Sehee Kwon; Kyoung-Hoon Kim; Yun-Gyeong Lee; Sera Shin; Duk-Hwa Kwon; Yeong-Un Lee; Taewon Kook; Nakwon Choe; Jeong Chul Kim; Young-Kook Kim; Gwang Hyeon Eom; Hyun Kook

Sumoylation, the conjugation of a small ubiquitin-like modifier (SUMO) protein to a target, has diverse cellular effects. However, the functional roles of the SUMO modification during myogenesis have not been fully elucidated. Here, we report that basal sumoylation of histone deacetylase 1 (HDAC1) enhances the deacetylation of MyoD in undifferentiated myoblasts, whereas further sumoylation of HDAC1 contributes to switching its binding partners from MyoD to Rb to induce myocyte differentiation. Differentiation in C2C12 skeletal myoblasts induced new immunoblot bands above HDAC1 that were gradually enhanced during differentiation. Using SUMO inhibitors and sumoylation assays, we showed that the upper band was caused by sumoylation of HDAC1 during differentiation. Basal deacetylase activity was not altered in the SUMO modification-resistant mutant HDAC1 K444/476R (HDAC1 2R). Either differentiation or transfection of SUMO1 increased HDAC1 activity that was attenuated in HDAC1 2R. Furthermore, HDAC1 2R failed to deacetylate MyoD. Binding of HDAC1 to MyoD was attenuated by K444/476R. Binding of HDAC1 to MyoD was gradually reduced after 2 days of differentiation. Transfection of SUMO1 induced dissociation of HDAC1 from MyoD but potentiated its binding to Rb. SUMO1 transfection further attenuated HDAC1-induced inhibition of muscle creatine kinase luciferase activity that was reversed in HDAC1 2R. HDAC1 2R failed to inhibit myogenesis and muscle gene expression. In conclusion, HDAC1 sumoylation plays a dual role in MyoD signaling: enhancement of HDAC1 deacetylation of MyoD in the basally sumoylated state of undifferentiated myoblasts and dissociation of HDAC1 from MyoD during myogenesis.


Experimental and Molecular Medicine | 2018

PP2A negatively regulates the hypertrophic response by dephosphorylating HDAC2 S394 in the heart

Somy Yoon; Taewon Kook; Hyun-Ki Min; Duk-Hwa Kwon; Young Kuk Cho; Mi-Ra Kim; Sera Shin; Hosouk Joung; Seung Hoon Jeong; Sumin Lee; Gaeun Kang; Yunchul Park; Yong Sook Kim; Youngkeun Ahn; Julie R. McMullen; Ulrich Gergs; Joachim Neumann; Kyung Keun Kim; Jung-Chul Kim; Kwang-Il Nam; Young-Kook Kim; Hyun Kook; Gwang Hyeon Eom

Cardiac hypertrophy occurs in response to increased hemodynamic demand and can progress to heart failure. Identifying the key regulators of this process is clinically important. Though it is thought that the phosphorylation of histone deacetylase (HDAC) 2 plays a crucial role in the development of pathological cardiac hypertrophy, the detailed mechanism by which this occurs remains unclear. Here, we performed immunoprecipitation and peptide pull-down assays to characterize the functional complex of HDAC2. Protein phosphatase (PP) 2 A was confirmed as a binding partner of HDAC2. PPP2CA, the catalytic subunit of PP2A, bound to HDAC2 and prevented its phosphorylation. Transient overexpression of PPP2CA specifically regulated both the phosphorylation of HDAC2 S394 and hypertrophy-associated HDAC2 activation. HDAC2 S394 phosphorylation was increased in a dose-dependent manner by PP2A inhibitors. Hypertrophic stresses, such as phenylephrine in vitro or pressure overload in vivo, caused PPP2CA to dissociate from HDAC2. Forced expression of PPP2CA negatively regulated the hypertrophic response, but PP2A inhibitors provoked hypertrophy. Adenoviral delivery of a phosphomimic HDAC2 mutant, adenovirus HDAC2 S394E, successfully blocked the anti-hypertrophic effect of adenovirus-PPP2CA, implicating HDAC2 S394 phosphorylation as a critical event for the anti-hypertrophic response. PPP2CA transgenic mice were protected against isoproterenol-induced cardiac hypertrophy and subsequent cardiac fibrosis, whereas simultaneous expression of HDAC2 S394E in the heart did induce hypertrophy. Taken together, our results suggest that PP2A is a critical regulator of HDAC2 activity and pathological cardiac hypertrophy and is a promising target for future therapeutic interventions.Cardiovascular disease: A brake for heart muscle growthA regulatory mechanism that controls how cardiac muscle responds to stress could inform development of new therapies for preventing heart failure. Physiological stimuli ranging from heavy exercise to heart attack can induce hypertrophy, an increase in cardiac muscle mass that is initially beneficial but can lead to organ failure. Researchers led by Gwang Hyeon Eom and Hyun Kook at the Chonnam National University Biomedical Research Center, Hwasungun, South Korea have found that an enzyme called protein phosphatase 2A (PP2A) keeps cardiac hypertrophy in check. PP2A binds to and inhibits a second protein known as HDAC2, which would otherwise stimulate the hypertrophic response to stress. The researchers have identified the biochemical mechanism by which PP2A inactivates HDAC2, and demonstrate that this inhibition effectively protects against hypertrophic cardiac damage in mice, revealing a possible avenue for clinical intervention.


Atherosclerosis | 2012

Sulforaphane inhibits restenosis by suppressing inflammation and the proliferation of vascular smooth muscle cells

Jin-Sook Kwon; Hosouk Joung; Yong Sook Kim; Young-Sun Shim; Youngkeun Ahn; Myung Ho Jeong; Hae Jin Kee


Circulation Research | 2015

Abstract 418: Mdm2 E3 Ligase-mediated Ubiquitination of Histone Deacetylase 1 in Vascular Calcification

Hyun Kook; Duk-Hwa Kwon; Gwang Hyeon Eom; Sera Shin; Hosouk Joung; Nakwon Choe; Yoon Seok Nam; Taewon Kook; Hyung-Seok Kim; Yong Sook Kim; Jeong-Tae Koh; Nacksung Kim; Kwang-Il Nam

Collaboration


Dive into the Hosouk Joung's collaboration.

Top Co-Authors

Avatar

Hyun Kook

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Gwang Hyeon Eom

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Nakwon Choe

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Sera Shin

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Duk-Hwa Kwon

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Kwang-Il Nam

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Young Kuk Cho

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Hae Jin Kee

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Hyeon Ko

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Yoon Seok Nam

Chonnam National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge