Network


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

Hotspot


Dive into the research topics where Sin-Ae Lee is active.

Publication


Featured researches published by Sin-Ae Lee.


Journal of Clinical Investigation | 2008

Tetraspanin TM4SF5 mediates loss of contact inhibition through epithelial-mesenchymal transition in human hepatocarcinoma

Sin-Ae Lee; Sung-Yul Lee; Ik-Hyun Cho; Min-A Oh; Eun-Sil Kang; Yong-Bae Kim; Woo Duck Seo; Suyong Choi; Ju-Ock Nam; Mimi Tamamori-Adachi; Shigetaka Kitajima; Sang-Kyu Ye; Semi Kim; Yoon-Jin Hwang; In-San Kim; Ki Hun Park; Jung Weon Lee

The growth of normal cells is arrested when they come in contact with each other, a process known as contact inhibition. Contact inhibition is lost during tumorigenesis, resulting in uncontrolled cell growth. Here, we investigated the role of the tetraspanin transmembrane 4 superfamily member 5 (TM4SF5) in contact inhibition and tumorigenesis. We found that TM4SF5 was overexpressed in human hepatocarcinoma tissue. TM4SF5 expression in clinical samples and in human hepatocellular carcinoma cell lines correlated with enhanced p27Kip1 expression and cytosolic stabilization as well as morphological elongation mediated by RhoA inactivation. These TM4SF5-mediated effects resulted in epithelial-mesenchymal transition (EMT) via loss of E-cadherin expression. The consequence of this was aberrant cell growth, as assessed by S-phase transition in confluent conditions, anchorage-independent growth, and tumor formation in nude mice. The TM4SF5-mediated effects were abolished by suppressing the expression of either TM4SF5 or cytosolic p27Kip1, as well as by reconstituting the expression of E-cadherin. Our observations have revealed a role for TM4SF5 in causing uncontrolled growth of human hepatocarcinoma cells through EMT.


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.


Journal of Biological Chemistry | 2008

Cell Adhesion-dependent Cofilin Serine 3 Phosphorylation by the Integrin-linked Kinase·c-Src Complex

Yong-Bae Kim; Suyong Choi; Moon-Chang Choi; Min-A Oh; Sin-Ae Lee; Moonjae Cho; Kensaku Mizuno; Sunghoon Kim; Jung Weon Lee

Integrin-linked kinase (ILK) is involved in signal transduction by integrin-mediated cell adhesion that leads to dynamic actin reorganization. Actin (de)polymerization is regulated by cofilin, the Ser3 phosphorylation (pS3cofilin) of which inhibits its actin-severing activity. To determine how ILK regulates pS3cofilin, we examined the effects of ILK on pS3cofilin using normal RIE1 cells. Compared with suspended cells, fibronectin-adherent cells showed enhanced pS3cofilin, depending on ILK expression and c-Src activity. The ILK-mediated pS3cofilin in RIE1 cells did not involve Rho-associated kinase, LIM kinase, or testicular protein kinases, which are known to be upstream of cofilin. The kinase domain of ILK, including proline-rich regions, appeared to interact physically with the Src homology 3 domain of c-Src. In vitro kinase assay revealed that ILK immunoprecipitates phosphorylated the recombinant glutathione S-transferase-cofilin, which was abolished by c-Src inhibition. Interestingly, epidermal growth factor treatment abolished the ILK effects, indicating that the linkage from ILK to cofilin is biologically responsive to extracellular cues. Altogether, this study provides evidence for a new signaling connection from ILK to cofilin for dynamic actin polymerization during cell adhesion, depending on the activity of ILK-associated c-Src.


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.)


Experimental and Molecular Medicine | 2008

LYR71, a derivative of trimeric resveratrol, inhibits tumorigenesis by blocking STAT3-mediated matrix metalloproteinase 9 expression

Ja-Eun Kim; Hong Sook Kim; Yong-Jae Shin; Chang Seok Lee; Cheolhee Won; Sin-Ae Lee; Jung Weon Lee; Youngsoo Kim; Jae-Seung Kang; Sang-Kyu Ye; Myung-Hee Chung

Tumor migration/invasion is the main cause of tumor progression and STAT3 is needed to enhance tumor migration/invasion by up-regulating MMP-9. Thus, agents that inhibit STAT3 activation may be used as an anticancer drug. We present herein that 6-methyl-2-propylimino-6, 7-dihydro-5H-benzo [1, 3]-oxathiol-4-one (LYR71) , a derivative of trimeric resveratrol, has an anticancer activity through inhibition of STAT3 activation. We found that LYR71 suppressed STAT3 activation and inhibited the expression and activity of MMP-9 in RANTES-stimulated breast cancer cells. In addition, LYR71 reduced RANTES-induced MMP-9 transcripts by blocking STAT3 recruitment, dissociating p300 and deacetylating histone H3 and H4 on the MMP-9 promoter. Furthermore, LYR71 inhibited tumor migration/invasion in RANTES-treated breast cancer cells and consequently blocked tumor progression in tumor-bearing mice. Taken together, the results of this study suggest that LYR71 can be therapeutically useful due to the inhibition effect of STAT3-mediated MMP-9 expression in breast cancer cells.


Meat Science | 2003

Physical evaluation of popped cereal snacks with spent hen meat

Sin-Ae Lee; Jun-Ki Min; In-Wha Kim; M. Lee

Various blends of spent hen meat and grains (potato starch, corn starch, and rice flour) were popped using a popping machine. Lowest bulk density was observed in the snack with 1:2 ratio of meat and potato starch. Except for the popped snack with meat and rice flour, as the starch content increased, bulk density decreased gradually. Popped snacks with grains only were higher in L* value than those with meat and grains. The a* and b* values increased with increasing meat content. All popped snacks were significantly different (P<0.001) in bulk density, color, and breaking force. As the grain content of snacks increased, the size of the air cells also increased. Results of scanning electron microscopic and optical microscopic observations revealed the popping degree of snack with starch and spent hen meat was affected by the presence of meat. The optimum ratios of meat to grain for high expansion ratio were determined to be 1:2 and 1:3 of meat to corn starch and potato starch.


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.


Frontiers in Bioscience | 2011

Modulation of signaling between TM4SF5 and integrins in tumor microenvironment.

Sin-Ae Lee; Ki Hun Park; Jung Weon Lee

TM4SF5 is a transmembrane glycoprotein of the transmembrane 4 L six family, a branch of the tetraspanin family and highly expressed in many types of cancers. TM4SF5 induces epithelial-mesenchymal transition (EMT) by morphological changes resulting from inactivation of RhoA mediated by stabilized cytosolic p27kip1. TM4SF5-mediated EMT can lead to loss of contact inhibition and enhanced migration/invasion, presumably depending on cross-talks between TM4SF5 and integrins. An anti-TM4SF5 agent appears to target the second extracellular domain of TM4SF5, which is important for cross-talk with integrins, leading to a blockade of TM4SF5-mediated multilayer growth and migration/invasion. In addition, TM4SF5 engages in cross-talk with integrin alpha5 to induce and secrete VEGF, which in turn causes activation of angiogenesis in endothelial cells. Therefore, TM4SF5 plays a central regulatory role in a wide variety of physiological processes through cross-talk with integrins. This review presents current knowledge from in vitro and in vivo observations of the roles of TM4SF5-integrin cooperation in hepatocellular carcinogenesis and discusses important areas for future investigation.


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.

Collaboration


Dive into the Sin-Ae Lee's collaboration.

Top Co-Authors

Avatar

Jung Weon Lee

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

Tae Kyoung Kwak

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Suyong Choi

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hyeonjung Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hyun Jeong Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Min-A Oh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Moonjae Cho

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Eun-Sil Kang

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge