Network


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

Hotspot


Dive into the research topics where Kyung Mi Woo is active.

Publication


Featured researches published by Kyung Mi Woo.


Bone | 2002

The phosphatidylinositol 3-Kinase, p38, and extracellular signal-regulated kinase pathways are involved in osteoclast differentiation

S.E. Lee; Kyung Mi Woo; S.Y. Kim; Hyunjong Kim; K. Kwack; Zang Hee Lee; Hong-Hee Kim

Phosphatidylinositol 3-kinase (PI 3-kinase) and mitogen-activated protein kinases (MAPKs) have been implicated in diverse cellular functions, including proliferation, migration, and survival. In this study, we examined the involvement of these kinases in osteoclast differentiation by employing specific inhibitors of the kinases. The osteoclast differentiation was assessed in three different culture systems: a coculture of mouse bone marrow cells with mouse calvarial osteoblasts, a mouse bone marrow cell culture in the presence of receptor activator of NF-kappaB ligand (RANKL) and macrophage-colony stimulating factor (M-CSF), and a culture of bone-resident osteoclast precursor cells driven by RANKL and M-CSF. LY294002, a specific inhibitor of PI 3-kinase, potently inhibited osteoclast differentiation in all culture systems when assessed by both tartrate-resistant acid phosphatase (TRAP) staining and dentine resorption assays. Inhibition of p38 MAPK by SB202190 resulted in a strong suppression in the exogenous RANKL dependent mouse bone marrow and bone resident precursor cell cultures. Another MAPK pathway inhibitor (PD98059), which blocks the activation of extracellular signal-regulated kinase (ERK) by inhibiting the upstream kinase MAPK-ERK kinase (MEK) 1, exerted an inhibitory effect on osteoclast differentiation only at the highest concentration tested (30 micromol/L) in many cases. Whether the signaling pathways involving these kinases are activated by RANKL was also examined. The RANKL-stimulated phosphorylation of Akt, a downstream target of PI 3-kinase, and that of ERK were observed. RANKL also stimulated the activity of p38. These results suggest that PI 3 kinase, p38, and ERK play roles in osteoclast differentiation, at least in part, by participating in RANKL signaling.


European Journal of Immunology | 2001

Osteoclastogenesis is enhanced by activated B cells but suppressed by activated CD8+ T cells

Youngnim Choi; Kyung Mi Woo; Seong-Hee Ko; Yoon Jung Lee; Su-Jin Park; Hyun-Man Kim; Byoung S. Kwon

Host immune response is known to contribute to the progression of periodontitis, and alveolar bone destruction in periodontitis is associated with enhanced osteoclast activity. Therefore, we evaluated the roles of activated lymphocyte subsets in osteoclastogenesis. Osteoclast precursors were co‐cultured with activated lymphocytes (B, CD4+ T, CD8+ T) in the presence of either macrophage colony‐stimulating factor (M‐CSF) alone or M‐CSF plus soluble receptor activator of NF‐κB ligand (sRANKL), and subsequent differentiation into active osteoclasts was evaluatedby a resorption assay. The activated B and CD4+ cells, but not CD8+ T cells, induced osteoclast differentiation in the presence of M‐CSF alone. In the presence of M‐CSF and sRANKL, B cells induced the formation of small but highly active osteoclasts and increased resorption, while CD8+ T cells profoundly suppressed osteoclastogenesis. Co‐culture using an insert wellor supernatant suggested that both B and CD8+ T cells acted on osteoclasts mostly via soluble proteins. Activated B cells expressed many osteoclastogenic factors including RANKL, TNF‐α, IL‐6, MIP‐1α, and MCP‐3. CD8+ T cells expressed a substantial amount of osteoprotegerin (OPG) along with RANKL. However, blocking antibody to OPG did not reverse the suppression by CD8+ T cells, suggesting that other factor(s) are involved. Taken together, activated B cells promoted osteoclastogenesis, while CD8+ T cells inhibited the osteoclast formation via direct interaction. The results imply the importance of lymphocyte subpopulations in the development of periodontitis.


Journal of Biological Chemistry | 2010

BMP2-activated Erk/MAP Kinase Stabilizes Runx2 by Increasing p300 Levels and Histone Acetyltransferase Activity

Ji Hae Jun; Won-Joon Yoon; Sang-Beom Seo; Kyung Mi Woo; Gwan-Shik Kim; Hyun-Mo Ryoo; Jeong-Hwa Baek

Runx2 is a critical transcription factor for osteoblast differentiation. Regulation of Runx2 expression levels and transcriptional activity is important for bone morphogenetic protein (BMP)-induced osteoblast differentiation. Previous studies have shown that extracellular signal-regulated kinase (Erk) activation enhances the transcriptional activity of Runx2 and that BMP-induced Runx2 acetylation increases Runx2 stability and transcriptional activity. Because BMP signaling induces Erk activation in osteoblasts, we sought to investigate whether BMP-induced Erk signaling regulates Runx2 acetylation and stability. Erk activation by overexpression of constitutively active MEK1 increased Runx2 transcriptional activity, whereas U0126, an inhibitor of MEK1/2, suppressed basal Runx2 transcriptional activity and BMP-induced Runx2 acetylation and stabilization. Overexpression of constitutively active MEK1 stabilized Runx2 protein via up-regulation of acetylation and down-regulation of ubiquitination. Erk activation increased p300 protein levels and histone acetyltransferase activity. Knockdown of p300 using siRNA diminished Erk-induced Runx2 stabilization. Overexpression of Smad5 increased Runx2 acetylation and stabilization. Erk activation further increased Smad-induced Runx2 acetylation and stabilization, whereas U0126 suppressed these functions. On the other hand, knockdown of Smad1 and Smad5 by siRNA suppressed both basal and Erk-induced Runx2 protein levels. Erk activation enhanced the association of Runx2 with p300 and Smad1. Taken together these results indicate that Erk signaling increases Runx2 stability and transcriptional activity, partly via increasing p300 protein levels and histone acetyltransferase activity and subsequently increasing Runx2 acetylation by p300. In addition to the canonical Smad pathway, a BMP-induced non-Smad Erk signaling pathway cooperatively regulates osteoblast differentiation partly via increasing the stability and transcriptional activity of Runx2.


Journal of Biological Chemistry | 2010

Molecular Consequences of the ACVR1R206H Mutation of Fibrodysplasia Ossificans Progressiva

Gin-Ah Song; Hyunjung Kim; Kyung Mi Woo; Jeong-Hwa Baek; Gwan-Shik Kim; Jin Young Choi; Hyun-Mo Ryoo

Fibrodysplasia ossificans progressiva (FOP), a rare genetic and catastrophic disorder characterized by progressive heterotopic ossification, is caused by a point mutation, c.617G>A; p.R206H, in the activin A receptor type 1 (ACVR1) gene, one of the bone morphogenetic protein type I receptors (BMPR-Is). Although altered BMP signaling has been suggested to explain the pathogenesis, the molecular consequences of this mutation are still elusive. Here we studied the impact of ACVR1 R206H mutation on BMP signaling and its downstream signaling cascades in murine myogenic C2C12 cells and HEK 293 cells. We found that ACVR1 was the most abundant of the BMPR-Is expressed in mesenchymal cells but its contribution to osteogenic BMP signal transduction was minor. The R206H mutant caused weak activation of the BMP signaling pathway, unlike the Q207D mutant, a strong and constitutively active form. The R206H mutant showed a decreased binding affinity for FKBP1A/FKBP12, a known safeguard molecule against the leakage of transforming growth factor (TGF)-β or BMP signaling. The decreased binding affinity of FKBP1A to the mutant R206H ACVR1 resulted in leaky activation of the BMP signal, and moreover, it decreased steady-state R206H ACVR1 protein levels. Interestingly, while WT ACVR1 and FKBP1A were broadly distributed in plasma membrane and cytoplasm without BMP-2 stimulation and then localized in plasma membrane on BMP-2 stimulation, R206H ACVR1 and FKBP1A were mainly distributed in plasma membrane regardless of BMP-2 stimulation. The impaired binding to FKBP1A and an altered subcellular distribution by R206H ACVR1 mutation may result in mild activation of osteogenic BMP-signaling in extraskeletal sites such as muscle, which eventually lead to delayed and progressive ectopic bone formation in FOP patients.


Journal of Biological Chemistry | 2010

FGF2-activated ERK Mitogen-activated Protein Kinase Enhances Runx2 Acetylation and Stabilization

Ok-Jin Park; Hyun Jung Kim; Kyung Mi Woo; Jeong-Hwa Baek; Hyun-Mo Ryoo

Runx2 is a key transcription factor regulating osteoblast differentiation and skeletal morphogenesis, and FGF2 is one of the most important regulators of skeletal development. The importance of the ERK mitogen-activated protein (MAP) kinase pathway in cranial suture development was demonstrated by the findings that the inhibition of FGF/FGF receptor (FGFR) signaling by a MEK blocker prevents the premature suture closure caused by an Fgfr2 mutation in mice. We previously demonstrated that ERK activation does not affect Runx2 gene expression but that it stimulates Runx2 transcriptional activity. However, the molecular mechanism underlying Runx2 activation by FGF/FGFR or ERK was still unclear. In this study, we found that FGF2 treatment increased the protein level of exogenously overexpressed Runx2 and that this increase is reversed by ERK inhibitors. In contrast, overexpression of constitutively active MEK strongly increased the Runx2 protein level, which paralleled an increase in Runx2 acetylation. As Runx2 protein phosphorylation mediated by ERK directly correlates with Runx2 protein stabilization, acetylation, and ubiquitination, we undertook to identify the ERK-dependent phosphorylation sites in Runx2. Analysis of two C-terminal Runx2 deletion constructs showed that the middle third of the protein is responsible for ERK-induced stabilization and activation. An in silico analysis of highly conserved ERK targets indicated that there are three relevant serine residues in this domain. Site-directed mutagenesis implicated Ser-301 in for ERK-mediated Runx2 stabilization and acetylation. In conclusion, the FGF2-induced ERK MAP kinase strongly increased the Runx2 protein level through an increase in acetylation and a decrease in ubiquitination, and these processes require the phosphorylation of Runx2 Ser-301 residue.


Biochemical and Biophysical Research Communications | 2010

Tumor necrosis factor-α increases alkaline phosphatase expression in vascular smooth muscle cells via MSX2 induction

Hye-Lim Lee; Kyung Mi Woo; Hyun-Mo Ryoo; Jeong-Hwa Baek

Vascular calcification is implicated in many diseases including atherosclerosis and diabetes. Tumor necrosis factor-alpha (TNF-alpha) has been shown to promote vascular calcification both in vitro and in vivo. However, the molecular mechanism of TNF-alpha-mediated vascular calcification has not yet been fully defined. Therefore, in this study, we aimed to investigate whether MSX2 acts as a crucial regulator in TNF-alpha-induced vascular calcification and to define the regulatory mechanism of MSX2 induction in human vascular smooth muscle cells (VSMCs). TNF-alpha increased the expression of osteogenic marker genes including RUNX2, osterix, alkaline phosphatase (ALP), and bone sialoprotein, and it also promoted matrix mineralization in VSMCs. In addition, TNF-alpha enhanced MSX2 expression in a dose- and time-dependent manner. MSX2 over-expression alone induced ALP expression, whereas knockdown of MSX2 with small interfering RNA completely blocked TNF-alpha-induced ALP expression. New protein synthesis was dispensable for MSX2 induction by TNF-alpha, and the inhibition of NF-kappaB by BAY-11-7082 or by dominant negative IkappaBalpha abolished the TNF-alpha-directed induction of MSX2 expression. However, inhibition of NADPH oxidase did not affect MSX2 expression. In conclusion, our study suggests that TNF-alpha directly induces MSX2 expression through the NF-kappaB pathway, which in turn induces expression of ALP, a key molecule in mineralization, in VSMCs.


Tissue Engineering Part A | 2009

Comparative Evaluation of Nanofibrous Scaffolding for Bone Regeneration in Critical-Size Calvarial Defects

Kyung Mi Woo; Victor J. Chen; Hong Moon Jung; Tae-Il Kim; Hong In Shin; Jeong Hwa Baek; Hyun-Mo Ryoo; Peter X. Ma

In a previous study we found that nanofibrous poly(l-lactic acid) (PLLA) scaffolds mimicking collagen fibers in size were superior to solid-walled scaffolds in promoting osteoblast differentiation and bone formation in vitro. In this study we used an in vivo model to confirm the biological properties of nanofibrous PLLA scaffolds and to evaluate how effectively they support bone regeneration against solid-walled scaffolds. The scaffolds were implanted in critical-size defects made on rat calvarial bones. Compared with solid-walled scaffolds, nanofibrous scaffolds supported substantially more new bone tissue formation, which was confirmed by micro-computed tomography measurement and von Kossa staining. Goldners trichrome staining showed abundant collagen deposition in nanofibrous scaffolds but not in the control solid-walled scaffolds. The cells in these scaffolds were immuno-stained strongly for Runx2 and bone sialoprotein (BSP). In contrast, solid-walled scaffolds implanted in the defects were stained weakly with trichrome, Runx2, and BSP. These in vivo results demonstrate that nanofibrous architecture enhances osteoblast differentiation and bone formation.


Journal of Cellular Physiology | 2008

Epidermal growth factor receptor regulates osteoclast differentiation and survival through cross-talking with RANK signaling

TacGhee Yi; Hye-Lim Lee; Ji-Hoon Cha; Soo-Il Ko; Hye-Jin Kim; Hong-In Shin; Kyung Mi Woo; Hyun-Mo Ryoo; Gwan-Shik Kim; Jeong-Hwa Baek

The epidermal growth factor receptor (EGFR) functions in various cellular physiological processes such as proliferation, differentiation, and motility. Although recent studies have reported that EGFR signaling is involved in osteoclast recruitment and formation, the molecular mechanism of EGFR signaling for the regulation of osteoclastogenesis remains unclear. We investigated the role of the EGFR in osteoclast differentiation and survival and show that the expression of the EGFR was highly up‐regulated by receptor activator of nuclear factor‐κB ligand (RANKL) during osteoclast differentiation. EGFR‐specific tyrosine kinase inhibitors and EGFR knockdown blocked RANKL‐dependent osteoclast formation, suggesting that EGFR signaling plays an important role in osteoclastogenesis. EGFR inhibition impaired the RANKL‐mediated activation of osteoclastogenic signaling pathways, including c‐Jun N‐terminal kinase (JNK), NF‐κB, and Akt/protein kinase B (PKB). In addition, EGFR inhibition in differentiated osteoclasts caused apoptosis through caspase activation. Inhibition of the phosphoinositide‐3 kinase (PI3K)‐Akt/PKB pathway and subsequent activation of BAD and caspases‐9 and ‐3 may be responsible for the EGFR inhibition‐induced apoptosis. The EGFR physically associated with receptor activator of nuclear factor‐κB (RANK) and Grb2‐associated binder 2 (Gab2). Moreover, RANKL transactivated EGFR. These data indicate that EGFR regulates RANKL‐activated signaling pathways by cross‐talking with RANK, suggesting that the EGFR may play a crucial role as a RANK downstream signal and/or a novel type of RANK co‐receptor in osteoclast differentiation and survival. J. Cell. Physiol. 217: 409–422, 2008.


Experimental and Molecular Medicine | 2007

Trichostatin A-mediated upregulation of p21 WAF1 contributes to osteoclast apoptosis

TacGhee Yi; Jeong-Hwa Baek; Hye-Jin Kim; Mi-Hye Choi; Sang-Beom Seo; Hyun-Mo Ryoo; Gwan-Shik Kim; Kyung Mi Woo

Histone deacetylase inhibitors (HDIs), a new class of anti-cancer agents, have been reported to suppress formation of osteoclast precursors and their fusion into multinucleated cells. However, little is known about the effect of HDIs on mature osteoclasts, which may have significance for their therapeutic use. Here, we demonstrate a novel action of HDIs on osteoclast apoptosis. Primary multinucleated mature osteoclasts were prepared from mouse bone marrow cells. Treatment of osteoclasts with the HDI trichostatin A (TSA) caused apoptosis, as confirmed by annexin V staining and caspase activation. TSA caused the upregulation of p21WAF1 in osteoclasts. To understand the role of p21WAF1 upregulation in TSA-treated osteoclasts, shRNA against p21WAF1-containing lentivirus was introduced into osteoclasts. The suppression of p21WAF1 decreased TSA-directed osteoclast apoptosis. Collectively, our results provide evidence that TSA causes osteoclast apoptosis, which involves, in part, TSA-induced upregulation of p21WAF1, and strongly supports HDIs as potential therapeutic agents for excessive bone resorption.


Experimental and Molecular Medicine | 2010

Msx2 mediates the inhibitory action of TNF-α on osteoblast differentiation

Hye-Lim Lee; TacGhee Yi; Kyung Mi Woo; Hyun-Mo Ryoo; Gwan-Shik Kim; Jeong-Hwa Baek

TNF-α, a proinflammatory cytokine, inhibits osteoblast differentiation under diverse inflammatory conditions; however, the underlying mechanisms in terms of the TNF-α signaling pathway remain unclear. In this study, we examined the role of Msx2 in TNF-α-mediated inhibition of alkaline phosphatase (ALP) expression and the signaling pathways involved. TNF-α down-regulated ALP expression induced by bone morphogenetic protein 2 (BMP2) in C2C12 and Runx2-/- calvarial cells. Over-expression of Msx2 suppressed BMP2-induced ALP expression. Furthermore, TNF-α induced Msx2 expression, and the knockdown of Msx2 by small interfering RNAs rescued ALP expression, which was inhibited by TNF-α. TNF-α activated the NF-κB and the JNK pathways. Inhibition of NF-κB or JNK activation reduced the inhibitory effect of TNF-α on ALP expression, whereas TNF-α-induced Msx2 expression was only suppressed by the inhibition of the NF-κB pathway. Taken together, these results indicate that Msx2 mediates the inhibitory action of TNF-α on BMP2-regulated osteoblast differentiation and that the TNF-α-activated NF-κB pathway is responsible for Msx2 induction.

Collaboration


Dive into the Kyung Mi Woo's collaboration.

Top Co-Authors

Avatar

Hyun-Mo Ryoo

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Jeong-Hwa Baek

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Won-Joon Yoon

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Gwan-Shik Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hye-Lim Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Young-Dan Cho

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Joung-Hwan Oh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Gene Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Rabia Islam

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Abdul S. Qadir

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge