Network


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

Hotspot


Dive into the research topics where Hong Hee Kim is active.

Publication


Featured researches published by Hong Hee Kim.


The EMBO Journal | 2006

Sphingosine 1‐phosphate as a regulator of osteoclast differentiation and osteoclast–osteoblast coupling

Jiyoon Ryu; Hyung Joon Kim; Eun Ju Chang; Hao Huang; Yoshiko Banno; Hong Hee Kim

Sphingosine 1‐phosphate (S1P), produced by sphingosine kinase (SPHK), acts both by intracellular and extracellular modes. We evaluated the role of SPHK1 and S1P in osteoclastogenesis using bone marrow‐derived macrophage (BMM) single and BMM/osteoblast coculture systems. In BMM single cultures, the osteoclastogenic factor receptor activator of NF‐κB ligand (RANKL) upregulated SPHK1 and increased S1P production and secretion. SPHK1 siRNA enhanced and SPHK1 overexpression attenuated osteoclastogenesis via modulation of p38 and ERK activities, and NFATc1 and c‐Fos levels. Extracellular S1P had no effect in these cultures. These data suggest that intracellular S1P produced in response to RANKL forms a negative feedback loop in BMM single cultures. In contrast, S1P addition to BMM/osteoblast cocultures greatly increased osteoclastogenesis by increasing RANKL in osteoblasts via cyclooxygenase‐2 and PGE2 regulation. S1P also stimulated osteoblast migration and survival. The RANKL elevation and chemotactic effects were also observed with T cells. These results indicate that secreted S1P attracts and acts on osteoblasts and T cells to augment osteoclastogenesis. Taken together, S1P plays an important role in osteoclastogenesis regulation and in communication between osteoclasts and osteoblasts or T cells.


Cell Death & Differentiation | 2006

Osteoclast differentiation requires TAK1 and MKK6 for NFATc1 induction and NF-κB transactivation by RANKL

Hao Huang; Jiyoon Ryu; Jeongim Ha; Eun-Ju Chang; Hyun-Man Kim; Hong-Hee Kim; Toshio Kitamura; Zang Hee Lee; Hong Hee Kim

Osteoclast (Oc) differentiation is fundamentally controlled by receptor activator of nuclear factor kappaB ligand (RANKL). RANKL signalling targets include mitogen-activated protein kinases (MAPKs), nuclear factor kappaB (NF-κB), and nuclear factor of activated T cells (NFAT)c1. In this study, we found that p38 MAPK upstream components transforming growth factor-beta-activated kinase 1 (TAK1), MKK3, and MKK6 increased by RANKL in an early stage of osteoclastogenesis from primary bone marrow cells, which led to enhanced p38 activation. Retroviral transduction of dominant-negative (DN) forms of TAK1 and MKK6, but not that of MKK3, reduced Oc differentiation. Transduction of TAK1-DN and MKK6-DN and treatment with the p38 inhibitor SB203580 attenuated NFATc1 induction by RANKL. TAK1-DN, MKK6-DN, and SB203580, but not MKK3-DN, also suppressed RANKL stimulation of NF-κB transcription activity in a manner dependent on p65 phosphorylation on Ser-536. These results indicate that TAK1 and MKK6 constitute the p38 signalling pathway to participate to Oc differentiation by RANKL through p65 phosphorylation and NFATc1 induction, and that MKK6 and MKK3 have differential roles in osteoclastogenesis from bone marrow precursors.


Journal of Cell Science | 2008

The JNK-dependent CaMK pathway restrains the reversion of committed cells during osteoclast differentiation

Eun Ju Chang; Jeongim Ha; Hao Huang; Hyung Joon Kim; Jung Hoon Woo; Young-Kyun Lee; Zang Hee Lee; Ju Han Kim; Hong Hee Kim

Osteoclastogenesis involves the commitment of macrophage-lineage precursors to tartrate-resistant acid phosphatase-positive (TRAP+) mononuclear pre-osteoclasts (pOCs) and subsequent fusion of pOCs to form multinuclear mature osteoclasts. Despite many studies on osteoclast differentiation, little is known about the signaling mechanisms that specifically mediate the osteoclastic commitment. In this study, we found that inhibition of JNK at the pOC stage provoked reversion of TRAP+ cells to TRAP– cells. The conversion to TRAP– cells occurred with concomitant return to the state with higher expression of macrophage antigens, and greater activity of phagocytosis and dendritic-differentiation potential. JNK inhibition at the pOC stage reduced NFATc1 and CaMK levels, and addition of active NFATc1 partially rescued the effect of JNK inhibition. In addition, the level of NFATc1 was decreased by knockdown of CaMK by RNAi and by catalytic inhibition of CaMK, which both caused the reversion of pOCs to macrophages. These data suggest that JNK activity is specifically required for maintaining the committed status during osteoclastogenesis and that the CaMK-NFATc1 pathway is the key element in that specific role of JNK.


Journal of Cell Biology | 2012

Plasma membrane calcium ATPase regulates bone mass by fine-tuning osteoclast differentiation and survival

Hyung Joon Kim; Vikram Prasad; Seok Won Hyung; Zang Hee Lee; Sang Won Lee; Aditi Bhargava; David A. Pearce; Youngkyun Lee; Hong Hee Kim

Plasma membrane calcium ATPases PMCA1 and PMCA4 regulate osteoclast differentiation and survival by regulating NFATc1 and NO.


FEBS Letters | 2009

Lyn inhibits osteoclast differentiation by interfering with PLCγ1-mediated Ca2+ signaling

Soo Hyun Yoon; Young-Kyun Lee; Hyung Joon Kim; Zang Hee Lee; Seok Won Hyung; Sang Won Lee; Hong Hee Kim

Osteoclasts differentiate from macrophage‐lineage cells to become specialized for bone resorption function. By a proteomics approach, we found that Lyn was down‐regulated by the osteoclast differentiation factor, receptor activator of NF‐κB ligand (RANKL). The forced reduction of Lyn caused a striking increase in the RANKL‐induced PLCγ1, Ca2+, and NFATc1 responses during differentiation. These data suggest that Lyn plays a negative role in osteoclastogenesis by interfering with the PLCγ1‐mediated Ca2+ signaling that leads to NFATc1 activation. Consistent with the in vitro results, in vivo injection of Lyn specific siRNA into mice calvariae provoked a fulminant bone resorption. Our study provides the first evidence of the involvement of Lyn in the negative regulation of osteoclastogenesis by RANKL.


Journal of Molecular Medicine | 2013

Mitogen- and stress-activated protein kinase 1 activates osteoclastogenesis in vitro and affects bone destruction in vivo

Jeongim Ha; Hyung Joon Kim; Hao Huang; Zang Hee Lee; Hong Hee Kim

Mitogen- and stress-activated protein kinase (MSK) 1 is an important regulator of immune response and mitogenic signaling. In this study, we report for the first time that MSK1 was activated by the osteoclast differentiation factor receptor activator of nuclear factor kappa B ligand (RANKL) in osteoclast precursor cells. Inhibition of upstream kinases ERK1/2 and p38, but not JNK, suppressed MSK activation upon RANKL stimulation. An MSK1 inhibitor efficiently prevented the induction of c-Fos and NFATc1 and CREB phosphorylation by RANKL. Inhibition of MSK1 also successfully blocked RANKL-induced osteoclastogenesis. MSK knockdown with small interfering RNA significantly inhibited osteoclast differentiation and bone resorption. MSK1 did not affect osteoclast survival. The induction of c-Fos and NFATc1 and the phosphorylation of CREB and ATF2 were also inhibited by MSK1 knockdown. Moreover, knockdown of MSK1 significantly blocked recruitment of c-Fos to the NFATc1 promoter upon RANKL stimulation. Therefore, NFATc1-inducible osteoclast-specific genes were downregulated by MSK1 blockade. NFATc1 retrovirus transduction almost completely rescued the differentiation defect of MSK1-silenced cells. In vivo knockdown of MSK1 reduced RANKL-induced bone resorption as well as osteoclast formation. Thus, our results suggested that MSK1 is an important novel molecule involved in RANKL signaling and osteoclast differentiation.


Life Sciences | 2008

The phosphatidylinositol 3-kinase-mediated production of interferon-β is critical for the lipopolysaccharide inhibition of osteoclastogenesis

Young-Kyun Lee; Hao Huang; Hyung Joon Kim; Chul Park; Hong Hee Kim

AIMSnThis study was performed to define molecular mechanisms underlying lipopolysaccharide (LPS)-induced inhibition of osteoclastogenesis.nnnMAIN METHODSnThe LPS-dependent signaling pathways leading to the inhibition of osteoclastogenesis were examined using chemical inhibitors and neutralizing antibodies.nnnKEY FINDINGSnLipopolysaccharide (LPS) has been shown to induce massive bone loss in vivo by stimulating osteoclast differentiation and activity. However, the direct effect of LPS on osteoclastogenesis in vitro is complex. Based on the context of the differentiation state of precursors, LPS has been shown to either augment or inhibit osteoclast differentiation. When added to receptor activator of NF-kappaB ligand (RANKL)-primed precursors, LPS enhances osteoclast differentiation via the production of TNF-alpha. On the other hand, LPS inhibits osteoclastogenesis from early precursors like bone marrow macrophages by hitherto unknown mechanism. In the present study, we investigated the mechanism by which LPS inhibits osteoclastogenesis. We have identified that the phosphatidylinositol 3-kinase (PI 3-kinase) dependent production of IFN-beta and resultant inhibition of c-Fos expression upon LPS stimulation of bone marrow macrophages are responsible for the LPS-induced inhibition of osteoclastogenesis. Inhibition of PI 3-kinase, neutralization of IFN-beta, and overexpression of c-Fos respectively prevented the LPS-induced inhibition of osteoclast differentiation.nnnSIGNIFICANCEnOur results provide a molecular understanding of the differentiation stage-specific dual effect of LPS on osteoclastogenesis.


Biochemical and Biophysical Research Communications | 2006

Induction of c-Fos and NFATc1 during RANKL-stimulated osteoclast differentiation is mediated by the p38 signaling pathway.

Hao Huang; Eun Ju Chang; Jiyoon Ryu; Zang Hee Lee; Young-Kyun Lee; Hong Hee Kim


Biochemical Pharmacology | 2007

Inhibition of osteoclast differentiation and bone resorption by sauchinone.

Kyoung Youn Han; Daum Yang; Eun Ju Chang; Young-Kyun Lee; Hao Huang; Sang Hyun Sung; Zang Hee Lee; Young Choong Kim; Hong Hee Kim


Experimental Cell Research | 2004

Involvement of iNOS-dependent NO production in the stimulation of osteoclast survival by TNF-α

Seung Ku Lee; Hao Huang; Soo Woong Lee; Kyung Hee Kim; Kyung Keun Kim; Hyun Man Kim; Zang Hee Lee; Hong Hee Kim

Collaboration


Dive into the Hong Hee Kim's collaboration.

Top Co-Authors

Avatar

Zang Hee Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hao Huang

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Hyung Joon Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Young-Kyun Lee

Seoul National University Bundang Hospital

View shared research outputs
Top Co-Authors

Avatar

Eun Ju Chang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jeongim Ha

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jiyoon Ryu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Daum Yang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge