Network


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

Hotspot


Dive into the research topics where Tae Kyoung Kwak is active.

Publication


Featured researches published by Tae Kyoung Kwak.


Experimental Cell Research | 2008

O-GlcNAc modulation at Akt1 Ser473 correlates with apoptosis of murine pancreatic β cells

Eun-Sil Kang; Dohyun Han; Jung-Eun Park; Tae Kyoung Kwak; Min-A Oh; Sin-Ae Lee; Suyong Choi; Zee Yong Park; Youngsoo Kim; Jung Weon Lee

O-GlcNAc transferase (OGT)-mediated modification of protein Ser/Thr residues with O-GlcNAc influences protein activity, similar to the effects of phosphorylation. The anti-apoptotic Akt1 is both activated by phosphorylation and modified with O-GlcNAc. However, the nature and significance of the Akt1 O-GlcNAc modification is unknown. The relationship of O-GlcNAc modification and phosphorylation at Akt1 Ser473 was examined with respect to apoptosis of murine beta-pancreatic cells. Glucosamine treatment induced apoptosis, which correlated with enhanced O-GlcNAc modification of Akt1 and concomitant reduction in Ser473 phosphorylation. Pharmacological inhibition of OGT or O-GlcNAcase revealed an inverse correlation between O-GlcNAc modification and Ser473 phosphorylation of Akt1. MALDI-TOF/TOF mass spectrometry analysis of Akt1 immunoprecipitates from glucosamine-treated cells, but not untreated controls, showed a peptide containing S473/T479 that was presumably modified with O-GlcNAc. Furthermore, in vitro O-GlcNAc-modification analysis of wildtype and mutant Akt1 revealed that S473 was targeted by recombinant OGT. A S473A Akt1 mutant demonstrated reduced basal and glucosamine-induced Akt1 O-GlcNAc modification compared with wildtype Akt1. Furthermore, wildtype Akt1, but not the S473A mutant, appeared to be associated with OGT following glucosamine treatment. Together, these observations suggest that Akt1 Ser473 may undergo both phosphorylation and O-GlcNAc modification, and the balance between these may regulate murine beta-pancreatic cell fate.


Blood | 2009

Cooperation between integrin α5 and tetraspan TM4SF5 regulates VEGF-mediated angiogenic activity

Suyong Choi; Sin-Ae Lee; Tae Kyoung Kwak; Hyeon Jung Kim; Mi Ji Lee; Sang-Kyu Ye; Sung-Hoon Kim; Semi Kim; Jung Weon Lee

Tetraspan TM4SF5 is highly expressed in a diverse number of tumor types. Here we explore the mechanistic roles of TM4SF5 in angiogenesis. We found that TM4SF5 overexpression correlates with vascular endothelial growth factor (VEGF) expression in SNU449 hepatocytes and with vessel formation in clinical hepatocarcinoma samples. Conditioned media from TM4SF5-expressing cells enhanced viability and tube formation of primary human umbilical vein endothelial cells, and outgrowth of endothelial cells from aorta ring segments, which was abolished by treatment with an anti-VEGF antibody. TM4SF5 retained integrin alpha(5) on the cell surface for VEGF induction, and preincubation with anti-integrin alpha(5) antibody abolished TM4SF5-mediated VEGF expression and secretion. TM4SF5-mediated effects required integrin alpha(5), c-Src, and signal transducer and activator of transcription 3 (STAT3). In addition, tumors from nude mice injected with TM4SF5-expressing cells and from clinical human hepatocarcinoma tissues showed enhanced integrin alpha(5) expression, vessel formation, and signaling activity, which were inhibited by administration of anti-integrin alpha(5) or -VEGF antibody. This study suggests that TM4SF5 facilitates angiogenesis of neighboring endothelial cells through VEGF induction, mediated by cooperation between TM4SF5 and integrin alpha(5) of epithelial cells.


Hepatology | 2009

Blockade of four‐transmembrane L6 family member 5 (TM4SF5)‐mediated tumorigenicity in hepatocytes by a synthetic chalcone derivative

Sin-Ae Lee; Hyung Won Ryu; Young Mee Kim; Suyong Choi; Mi Ji Lee; Tae Kyoung Kwak; Hyeon Jung Kim; Moonjae Cho; Ki Hun Park; Jung Weon Lee

We previously reported that the four‐transmembrane L6 family member 5 (TM4SF5) was highly expressed in hepatocarcinoma, induced morphological elongation and epithelial‐mesenchymal transition, and caused abnormal cell growth in multilayers in vitro and tumor formation in vivo. In this study, we identified a synthetic compound, 4′‐(p‐toluenesulfonylamido)‐4‐hydroxychalcone (TSAHC) that antagonized both the TM4SF5‐mediated multilayer growth and TM4SF5‐enhanced migration/invasion. TSAHC treatment induced multilayer‐growing cells to grow in monolayers, recovering contact inhibition without accompanying apoptosis, and inhibited chemotactic migration and invasion. Tumor formation in nude mice injected with TM4SF5‐expressing cells and the growth of cells expressing endogenous TM4SF5, but not of TM4SF5‐null cells, was suppressed by treatment with TSAHC, but not by treatment with its analogs. The structure‐activity relationship indicated the significance of 4′‐p‐toluenesulfonylamido and 4‐hydroxy groups for the anti‐TM4SF5 effects of TSAHC. Point mutations of the putative N‐glycosylation sites abolished the TM4SF5‐specific TSAHC responsiveness. Conclusion: These observations suggest that TM4SF5‐enhanced tumorigenic proliferation and metastatic potential can be blocked by TSAHC, likely through targeting the extracellular region of TM4SF5, which is important for protein‐protein interactions. (HEPATOLOGY 2009.)


Biochemical Journal | 2012

Cross-talk between TGFβ1 and EGFR signalling pathways induces TM4SF5 expression and epithelial-mesenchymal transition.

Minkyung Kang; Suyong Choi; Soo Jin Jeong; Sin‑Ae Lee; Tae Kyoung Kwak; Hyeonjung Kim; Oisun Jung; Mi‑Sook Lee; Youra Ko; Jihye Ryu; Yoon‑Ju Choi; Doyoung Jeong; Hyo-Jeong Lee; Sang Kyu Ye; Sung Hoon Kim; Jung Weon Lee

The EMT (epithelial-mesenchymal transition) is involved in fibrosis and cancer, and is regulated by different signalling pathways mediated through soluble factors, actin reorganization and transcription factor actions. Because the tetraspan (also called tetraspanin) TM4SF5 (transmembrane 4 L6 family member 5) is highly expressed in hepatocellular carcinoma and induces EMT, understanding how TM4SF5 expression in hepatocytes is regulated is important. We explored the mechanisms that induce TM4SF5 expression and whether impaired signalling pathways for TM4SF5 expression inhibit the acquisition of mesenchymal cell features, using human and mouse normal hepatocytes. We found that TGFβ1 (transforming growth factor β1)-mediated Smad activation caused TM4SF5 expression and EMT, and activation of the EGFR [EGF (epidermal growth factor) receptor] pathway. Inhibition of EGFR activity following TGFβ1 treatment abolished acquisition of EMT, suggesting a link from Smads to EGFR for TM4SF5 expression. Further, TGFβ1-mediated EGFR activation and TM4SF5 expression were abolished by EGFR suppression or extracellular EGF depletion. Smad overexpression mediated EGFR activation and TM4SF5 expression in the absence of serum, and EGFR kinase inactivation or EGF depletion abolished Smad-overexpression-induced TM4SF5 and mesenchymal cell marker expression. Inhibition of Smad, EGFR or TM4SF5 using Smad7 or small compounds also blocked TM4SF5 expression and/or EMT. These results indicate that TGFβ1- and growth factor-mediated signalling activities mediate TM4SF5 expression leading to acquisition of mesenchymal cell features, suggesting that TM4SF5 induction may be involved in the development of liver pathologies.


Journal of Cellular Biochemistry | 2010

Transmembrane 4 L Six Family Member 5 (TM4SF5) Enhances Migration and Invasion of Hepatocytes for Effective Metastasis

Sin-Ae Lee; Tae-You Kim; Tae Kyoung Kwak; Hyeonjung Kim; Semi Kim; Hyo-Jeong Lee; Sung-Hoon Kim; Ki Hun Park; Hyun Jeong Kim; Moonjae Cho; Jung Weon Lee

Overexpression of transmembrane 4 L six family member 5 (TM4SF5), a four‐transmembrane L6 family member, causes aberrant cell proliferation and angiogenesis, but the roles of TM4SF5 in migration, invasion, and tumor metastasis remain unknown. Using in vitro hepatocarcinoma cells that ectopically or endogenously express TM4SF5 and in vivo mouse systems, roles of TM4SF5 in metastatic potentials were examined. We found that TM4SF5 expression facilitated migration, invadopodia formation, MMP activation, invasion, and eventually lung metastasis in nude mice, but suppression of TM4SF5 with its shRNA blocked the effects. Altogether, TM4SF5‐mediated migration and invasion suggest that TM4SF5 may be therapeutically targeted to deal with TM4SF5‐mediated hepatocellular cancers. J. Cell. Biochem. 111: 59–66, 2010.


Journal of Cell Science | 2012

Tetraspan TM4SF5-dependent direct activation of FAK and metastatic potential of hepatocarcinoma cells

Oisun Jung; Suyong Choi; Sun-Bok Jang; Sin-Ae Lee; Ssang-Taek Lim; Yoon-Ju Choi; Hye-Jin Kim; Do-Hee Kim; Tae Kyoung Kwak; Hyeonjung Kim; Minkyung Kang; Mi-Sook Lee; Sook Young Park; Jihye Ryu; Doyoung Jeong; Hae-Kap Cheong; Hyun Jeong Kim; Ki Hun Park; Bong-Jin Lee; David D. Schlaepfer; Jung Weon Lee

Summary Transmembrane 4 L six family member 5 (TM4SF5) plays an important role in cell migration, and focal adhesion kinase (FAK) activity is essential for homeostatic and pathological migration of adherent cells. However, it is unclear how TM4SF5 signaling mediates the activation of cellular migration machinery, and how FAK is activated during cell adhesion. Here, we showed that direct and adhesion-dependent binding of TM4SF5 to FAK causes a structural alteration that may release the inhibitory intramolecular interaction in FAK. In turn, this may activate FAK at the cells leading edge, to promote migration/invasion and in vivo metastasis. TM4SF5-mediated FAK activation occurred during integrin-mediated cell adhesion. TM4SF5 was localized at the leading edge of the cells, together with FAK and actin-organizing molecules, indicating a signaling link between TM4SF5/FAK and actin reorganization machinery. Impaired interactions between TM4SF5 and FAK resulted in an attenuated FAK phosphorylation (the signaling link to actin organization machinery) and the metastatic potential. Our findings demonstrate that TM4SF5 directly binds to and activates FAK in an adhesion-dependent manner, to regulate cell migration and invasion, suggesting that TM4SF5 is a promising target in the treatment of metastatic cancer.


Carcinogenesis | 2009

The extracellular loop 2 of TM4SF5 inhibits integrin α2 on hepatocytes under collagen type I environment

Sin-Ae Lee; Young Mee Kim; Tae Kyoung Kwak; Hyeon Jung Kim; Semi Kim; Wonil Ko; Sung-Hoon Kim; Ki Hun Park; Hyun Jeong Kim; Moonjae Cho; Jung Weon Lee

Four-transmembrane L6 family member 5 (TM4SF5) and its homolog L6, a tumor antigen, form a four-transmembrane L6 family. TM4SF5 expression causes uncontrolled cell proliferation and angiogenesis. Although other genuine transmembrane 4 superfamily (TM4SF) members co-operate with integrins for cell migration, roles of TM4SF5 in the cellular spreading and migration are unknown. Using hepatocarcinoma cell clones that ectopically express TM4SF5, we found that cross talks via an extracellular interaction between TM4SF5 and integrin alpha2 in collagen type I environment inhibited integrin alpha2 functions such as spreading on and migration toward collagen I, which were recovered by suppression of TM4SF5 or structural disturbance of its second extracellular loop using a peptide or mutagenesis. Altogether, the observations suggest that TM4SF5 in hepatocytes negatively regulates integrin alpha2 function via an interaction between the extracellular loop 2 of TM4SF5 and integrin alpha2 during cell spreading on and migration through collagen I environment.


Biochimica et Biophysica Acta | 2013

The COOH-terminus of TM4SF5 in hepatoma cell lines regulates c-Src to form invasive protrusions via EGFR Tyr845 phosphorylation.

Oisun Jung; Yoon-Ju Choi; Tae Kyoung Kwak; Minkyung Kang; Mi-Sook Lee; Jihye Ryu; Hye-Jin Kim; Jung Weon Lee

Transmembrane 4 L six family member 5 (TM4SF5) enhances cell migration and invasion, although how TM4SF5 mechanistically mediates these effects remains unknown. In the study, during efforts to understand TM4SF5-mediated signal transduction, TM4SF5 was shown to bind c-Src and thus hepatoma cell lines expressing TM4SF5 were analyzed for the significance of the interaction in cell invasion. The C-terminus of TM4SF5 bound both inactive c-Src that might be sequestered to certain cellular areas and active c-Src that might form invasive protrusions. Wildtype (WT) TM4SF5 expression enhanced migration and invasive protrusion formation in a c-Src-dependent manner, compared with TM4SF5-null control hepatoma cell lines. However, tailless TM4SF5(ΔC) cells were more efficient than WT TM4SF5 cells, suggesting a negative regulatory role by the C-terminus. TM4SF5 WT- or TM4SF5(ΔC)-mediated formation of invasive protrusions was dependent or independent on serum or epidermal growth factor treatment, respectively, although they both were dependent on c-Src. The c-Src activity of TM4SF5 WT- or TM4SF5(ΔC)-expressing cells correlated with enhanced Tyr845 phosphorylation of epidermal growth factor receptor. Y845F EGFR mutation abolished the TM4SF5-mediated invasive protrusions, but not c-Src phosphorylation. Our findings demonstrate that TM4SF5 modulates c-Src activity during TM4SF5-mediated invasion through a TM4SF5/c-Src/EGFR signaling pathway, differentially along the leading protrusive edges of an invasive cancer cell.


Biochimica et Biophysica Acta | 2010

TM4SF5 accelerates G1/S phase progression via cytosolic p27Kip1 expression and RhoA activity

Hyeonjung Kim; Minkyung Kang; Sin-Ae Lee; Tae Kyoung Kwak; Oisun Jung; Hyo-Jeong Lee; Sung-Hoon Kim; Jung Weon Lee

Transmembrane 4 L six family member 5 (TM4SF5) causes epithelial-mesenchymal transition (EMT) for aberrant cell proliferation. However, the effects of TM4SF5 expression on cell cycle are unknown so far. In this study, using hepatocytes that either ectopically or endogenously express TM4SF5 and human hepatocarcinoma tissues, the role of TM4SF5 in G1/S phase progression was examined. We found that TM4SF5 expression accelerated G1/S phase progression with facilitated cyclin D1 and E expression and Rb phosphorylation. Furthermore, TM4SF5 enhanced trafficking of CDK4 and cyclin D1 into the nucleus and induced complex formation between them. However, TM4SF5-facilitated G1/S phase progression was blocked by silencing of p27Kip1 using siRNA or by infection of active RhoA. Pharmacological inhibition of ROCK accelerated the G1/S phase progression of control TM4SF5-unexpressing cells. Altogether, these observations suggest that TM4SF5 accelerates G1/S phase progression with facilitated CDK4/cyclin D1 entry into the nucleus, which might be supported by TM4SF5-mediated actin reorganization through cytosolic p27Kip1 expression and Rho GTPase activity.


Journal of Biological Chemistry | 2012

Cell adhesion-dependent serine 85 phosphorylation of paxillin modulates focal adhesion formation and haptotactic migration via association with the C-terminal tail domain of talin.

Tae Kyoung Kwak; Mi-Sook Lee; Jihye Ryu; Yoon-Ju Choi; Minkyung Kang; Doyoung Jeong; Jung Weon Lee

Background: Paxillin, a focal adhesion (FA) adaptor, functions in migration, although how Ser-85 phosphorylation affects migration is unknown. Results: Phosphorylation of paxillin at Ser-85 is critical for its association with talin, which regulates FA formation along the cell boundary and haptotactic migration. Conclusion: Paxillin Ser-85 phosphorylation regulates FA formation and migration. Significance: Paxillin Ser-85 phosphorylation coordinates FA dynamics for migration. Integrin-mediated adhesion to extracellular matrix proteins is dynamically regulated during morphological changes and cell migration. Upon cell adhesion, protein-protein interactions among molecules at focal adhesions (FAs) play major roles in the regulation of cell morphogenesis and migration. Although tyrosine phosphorylation of paxillin is critically involved in adhesion-mediated signaling, the significance of paxillin phosphorylation at Ser-85 and the mechanism by which it regulates cell migration remain unclear. In this study, we examined how Ser-85 phosphorylation of paxillin affects FA formation and cell migration. We found that paxillin phosphorylation at Ser-85 occurred during HeLa cell adhesion to collagen I and was concomitant with tyrosine phosphorylation of both focal adhesion kinase and talin. However, the non-phosphorylatable S85A mutant of paxillin impaired cell spreading, FA turnover, and migration toward collagen I but not toward serum. Furthermore, whereas the (presumably indirect) interaction between paxillin and the C-terminal tail of talin led to dynamic FAs at the cell boundary, S85A paxillin did not bind talin and caused stabilized FAs in the central region of cells. Together, these observations suggest that cell adhesion-dependent Ser-85 phosphorylation of paxillin is important for its interaction with talin and regulation of dynamic FAs and cell migration.

Collaboration


Dive into the Tae Kyoung Kwak's collaboration.

Top Co-Authors

Avatar

Jung Weon Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Sin-Ae Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Minkyung Kang

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hyeonjung Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Oisun Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hyun Jeong Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Jihye Ryu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Ki Hun Park

Gyeongsang National University

View shared research outputs
Top Co-Authors

Avatar

Mi-Sook Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge