Network


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

Hotspot


Dive into the research topics where Semi Kim is active.

Publication


Featured researches published by Semi Kim.


Journal of Virology | 2009

Proteolytic activation of the 1918 influenza virus hemagglutinin.

Chawaree Chaipan; Darwyn Kobasa; Stephanie Bertram; Ilona Glowacka; Imke Steffen; Theodros Solomon Tsegaye; Makoto Takeda; Thomas H. Bugge; Semi Kim; Young Woo Park; Andrea Marzi; Stefan Pöhlmann

ABSTRACT Proteolytic activation of the hemagglutinin (HA) protein is indispensable for influenza virus infectivity, and the tissue expression of the responsible cellular proteases impacts viral tropism and pathogenicity. The HA protein critically contributes to the exceptionally high pathogenicity of the 1918 influenza virus, but the mechanisms underlying cleavage activation of the 1918 HA have not been characterized. The neuraminidase (NA) protein of the 1918 influenza virus allows trypsin-independent growth in canine kidney cells (MDCK). However, it is at present unknown if the 1918 NA, like the NA of the closely related strain A/WSN/33, facilitates HA cleavage activation by recruiting the proprotease plasminogen. Moreover, it is not known which pulmonary proteases activate the 1918 HA. We provide evidence that NA-dependent, trypsin-independent cleavage activation of the 1918 HA is cell line dependent and most likely plasminogen independent since the 1918 NA failed to recruit plasminogen and neither exogenous plasminogen nor the presence of the A/WSN/33 NA promoted efficient cleavage of the 1918 HA. The transmembrane serine protease TMPRSS4 was found to be expressed in lung tissue and was shown to cleave the 1918 HA. Accordingly, coexpression of the 1918 HA with TMPRSS4 or the previously identified HA-processing protease TMPRSS2 allowed trypsin-independent infection by pseuodotypes bearing the 1918 HA, indicating that these proteases might support 1918 influenza virus spread in the lung. In summary, we show that the previously reported 1918 NA-dependent spread of the 1918 influenza virus is a cell line-dependent phenomenon and is not due to plasminogen recruitment by the 1918 NA. Moreover, we provide evidence that TMPRSS2 and TMPRSS4 activate the 1918 HA by cleavage and therefore may promote viral spread in lung tissue.


Carcinogenesis | 2010

TMPRSS4 induces invasion and epithelial–mesenchymal transition through upregulation of integrin α5 and its signaling pathways

Semi Kim; Hee Young Kang; Eun-Hee Nam; Myung-Sook Choi; Xue-Feng Zhao; Chang Soo Hong; Jung Weon Lee; Jae Hyuk Lee; Young-Kyu Park

TMPRSS4 is a novel type II transmembrane serine protease that is highly expressed on the cell surface in pancreatic, thyroid and other cancer tissues, although its oncogenic significance and molecular mechanisms are unknown. Previously, we have shown that TMPRSS4 promotes invasion, migration and metastasis of human tumor cells by facilitating an epithelial-mesenchymal transition (EMT). In this study, we explored the molecular basis underlying TMPRSS4-mediated effects. We show that multiple downstream signaling pathways, including focal adhesion kinase (FAK), extracellular signal-regulated kinase (ERK), Akt, Src and Rac1, are activated by TMPRSS4 expression and that FAK signaling and ERK activation are required for TMPRSS4-induced invasiveness and EMT, including cadherin switch. Inhibition of PI3K or Src reduced invasiveness and actin rearrangement mediated by TMPRSS4 without restoring E-cadherin expression. Downregulation of E-cadherin was required for TMPRSS4-mediated effects but was not sufficient to induce EMT and invasion. TMPRSS4 induced integrin alpha5 expression and its signal transduction, leading to invasiveness and EMT accompanied by downregulation of E-cadherin. Functional blocking confirmed that integrin alpha5beta1 is a critical signaling molecule that is sufficient to induce TMPRSS4-mediated effects. Immunohistochemical analysis showed that TMPRSS4 expression was significantly higher in human colorectal cancer tissues from advanced stages than in that of early stage. Furthermore, upregulation of TMPRSS4 was correlated with enhanced integrin alpha5 expression. These observations implicate integrin alpha5 upregulation as a molecular mechanism by which TMPRSS4 induces invasion and contributes to cancer progression.


Oncotarget | 2016

TMPRSS4 induces invasion and proliferation of prostate cancer cells through induction of Slug and cyclin D1

Yunhee Lee; Dongjoon Ko; Hye-Jin Min; Sol Bi Kim; Hye-Mi Ahn; Young-Hoon Lee; Semi Kim

TMPRSS4 is a novel type II transmembrane serine protease found at the cell surface that is highly expressed in pancreatic, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates tumor cell invasion, migration, and metastasis. We also found that TMPRSS4 activates the transcription factor activating protein-1 (AP-1) to induce cancer cell invasion. Here, we explored TMPRSS4-mediated cellular functions and the underlying mechanisms. TMPRSS4 induced Slug, an epithelial-mesenchymal transition (EMT)-inducing transcription factor, and cyclin D1 through activation of AP-1, composed of c-Jun and activating transcription factor (ATF)-2, which resulted in enhanced invasion and proliferation of PC3 prostate cancer cells. In PC3 cells, not only c-Jun but also Slug was required for TMPRSS4-mediated proliferation and invasion. Interestingly, Slug induced phosphorylation of c-Jun and ATF-2 to activate AP-1 through upregulation of Axl, establishing a positive feedback loop between Slug and AP-1, and thus induced cyclin D1, leading to enhanced proliferation. Using data from The Cancer Genome Atlas, we found that Slug expression positively correlated with that of c-Jun and cyclin D1 in human prostate cancers. Expression of Slug was positively correlated with that of cyclin D1 in various cancer cell lines, whereas expression of other EMT-inducing transcription factors was not. This study demonstrates that TMPRSS4 modulates both invasion and proliferation via Slug and cyclin D1, which is a previously unrecognized pathway that may regulate metastasis and cancer progression.


mAbs | 2016

Generation, characterization and preclinical studies of a human anti-L1CAM monoclonal antibody that cross-reacts with rodent L1CAM

Seulki Cho; Insoo Park; Haejung Kim; Mun Sik Jeong; Mooney Lim; Eung Suk Lee; Jin Hong Kim; Semi Kim; Hyo Jeong Hong

ABSTRACT L1 cell adhesion molecule (L1CAM) is aberrantly expressed in malignant tumors and plays important roles in tumor progression. Thus, L1CAM could serve as a therapeutic target and anti-L1CAM antibodies may have potential as anticancer agents. However, L1CAM is expressed in neural cells and the druggability of anti-L1AM antibody must be validated at the earliest stages of preclinical study. Here, we generated a human monoclonal antibody that is cross-reactive with mouse L1CAM and evaluated its pharmacokinetic properties and anti-tumor efficacy in rodent models. First, we selected an antibody (Ab4) that binds human and mouse L1CAM from the human naïve Fab library using phage display, then increased its affinity 45-fold through mutation of 3 residues in the complementarity-determining regions (CDRs) to generate Ab4M. Next, the affinity of Ab4M was increased 1.8-fold by yeast display of single-chain variable fragment containing randomly mutated light chain CDR3 to generate Ab417. The affinities (KD) of Ab417 for human and mouse L1CAM were 0.24 nM and 79.16 pM, respectively. Ab417 specifically bound the Ig5 domain of L1CAM and did not exhibit off-target activity, but bound to the peripheral nerves embedded in normal human tissues as expected in immunohistochemical analysis. In a pharmacokinetics study, the mean half-life of Ab417 was 114.49 h when a single dose (10 mg/kg) was intravenously injected into SD rats. Ab417 significantly inhibited tumor growth in a human cholangiocarcinoma xenograft nude mouse model and did not induce any adverse effect in in vivo studies. Thus, Ab417 may have potential as an anticancer agent.


Theranostics | 2017

Anti-cancer Activity of Novel TM4SF5-Targeting Antibodies through TM4SF5 Neutralization and Immune Cell-Mediated Cytotoxicity

Hye-Mi Ahn; Jihye Ryu; Jin Myeong Song; Yunhee Lee; Hye-Jin Kim; Dongjoon Ko; Inpyo Choi; Sang Jick Kim; Jung Weon Lee; Semi Kim

The transmembrane four L6 family member 5 (TM4SF5) protein is a novel molecular target for the prevention and treatment of hepatocellular carcinoma. TM4SF5 is highly expressed in liver, colon, esophageal, and pancreatic cancers and is implicated in tumor progression. Here, we screened monoclonal antibodies that specifically bound to the extracellular loop 2 (EC2) of TM4SF5 from a phage-displayed murine antibody (single-chain variable fragment; scFv) library. We constructed and characterized chimeric antibodies, Ab27 and Ab79, of scFv fused with Fc domain of human IgG1. The affinity (KD) of Ab27 and Ab79 for soluble EC2 was approximately 9.2 nM and 16.9 nM, respectively, as determined by surface plasmon resonance analysis. Ab27 and Ab79 efficiently bound to native TM4SF5 on the cell surface were internalized into the cancer cells, leading to a decrease in cell surface TM4SF5. Ab27 and Ab79 inhibited the proliferation and invasion of TM4SF5-positive liver and colon cancer cells and reduced FAK and c-Src phosphorylation. Ab27 and Ab79 also enhanced anoikis sensitivity and reduced survivin. Ab27 mediated antibody-dependent cell-mediated cytotoxicity in vitro. Ab27 and Ab79 efficiently inhibited tumor growth in a liver cancer xenograft model. These results strongly support the further development of Ab27 as a novel anti-cancer agent in the clinic.


Oncotarget | 2018

Cooperation between ZEB2 and Sp1 promotes cancer cell survival and angiogenesis during metastasis through induction of survivin and VEGF

Dongjoon Ko; Semi Kim

Epithelial-mesenchymal transition (EMT) is a process implicated in tumor invasion and metastasis. During EMT, epithelial cells undergo molecular changes to acquire mesenchymal phenotypes, which are mediated by EMT-inducing transcription factors. Previously, we showed that ZEB2 cooperates with the transcription factor Sp1 to function as a transcriptional activator of vimentin, integrin α5, and cadherin-11, which promotes cancer cell invasion. We hypothesized that ZEB2, through cooperation with Sp1, would mediate diverse cellular functions beyond EMT and invasion during metastasis. ZEB2 upregulated the expression of Sp1-regulated genes such as survivin, bcl-2, cyclin D1, and vascular endothelial growth factor in an Sp1-dependent manner, resulting in increased cancer cell survival and proliferation and endothelial cell activation in vitro, and increased circulating tumor cell survival and tumor angiogenesis in vivo. In addition, Sp1 enhanced ZEB2 stability, suggesting the presence of a positive feedback loop between ZEB2 and Sp1. Clinical data showed that ZEB2 expression was positively associated with Sp1 expression, and that the expression of both of these factors had prognostic significance for predicting survival in cancer patients. This study suggests that invasion is linked to cancer cell survival and angiogenesis by ZEB2 during cancer progression, and increases our understanding of the pathways via which EMT-inducing transcription factors regulate the complex process of metastasis.


Cancer Research | 2016

Abstract 1604: TMPRSS4 induces invasion and proliferation of prostate cancer cells through induction of Slug and cyclin D1

Yunhee Lee; Semi Kim

TMPRSS4 is a novel type II transmembrane serine protease found at the cell surface that is highly expressed in pancreatic, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates tumor cell invasion, migration, and metastasis. We also found that TMPRSS4 activates the transcription factor activating protein-1 (AP-1) to induce cancer cell invasion. Here, we explored TMPRSS4-mediated cellular functions and the underlying mechanisms. TMPRSS4 induced Slug, an epithelial-mesenchymal transition (EMT)-inducing transcription factor, and cyclin D1 through activation of AP-1, composed of c-Jun and activating transcription factor (ATF)-2, which resulted in enhanced invasion and proliferation of PC3 prostate cancer cells. In PC3 cells, not only c-Jun but also Slug was required for TMPRSS4-mediated proliferation and invasion. Interestingly, Slug induced phosphorylation of c-Jun and ATF-2 to activate AP-1, establishing a positive feedback loop between Slug and AP-1, and thus induced cyclin D1, leading to enhanced proliferation. Using data from The Cancer Genome Atlas, we found that Slug expression positively correlated with that of c-Jun and cyclin D1 in human prostate cancers. This study demonstrates that TMPRSS4 modulates both invasion and proliferation via Slug and cyclin D1, which is a previously unrecognized pathway that may regulate metastasis and cancer progression. Citation Format: Yunhee Lee, Semi Kim. TMPRSS4 induces invasion and proliferation of prostate cancer cells through induction of Slug and cyclin D1. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1604.


Cancer Research | 2015

Abstract A71: TMPRSS4 proteolytically activates pro-uPA to induce cancer cell invasion

Hye-Jin Min; Semi Kim

The invasive nature of tumor cells is critical for cancer metastasis. Urokinase-type plasminogen activator (uPA), which is usually derived from stromal cells, is a well-known serine protease involved in invasion and metastasis. uPA is produced as an inactive single-chain protein (known as pro-uPA or sc-uPA) that is processed into the active disulfide-linked two-chain form of uPA by a proteolytic event. TMPRSS4 is a novel type II transmembrane serine protease that is highly expressed on the cell surface in pancreatic, thyroid, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates colorectal tumor cell invasion, migration, and metastasis and increased TMPRSS4 expression is associated with colorectal cancer progression. We also demonstrated that TMPRSS4 upregulates uPA gene expression through c-Jun N-terminal kinase signaling activation to induce cancer cell invasion. However, it remains unknown how proteolytic activity of TMPRSS4 contributes to invasion. Here, we report that TMPRSS4 directly converted inactive pro-uPA into the active form through its proteolytic activity. Analysis of conditioned medium from cells overexpressing TMPRSS4 demonstrated that the active TMPRSS4 protease domain is released from the cells and is associated with the plasma membrane. Furthermore, TMPRSS4 could increase pro-uPA-mediated invasion in a serine proteolytic activity-dependent manner. These observations suggest that TMPRSS4 is an upstream regulator of invasion through the regulation of uPA. This study provides valuable insights into the proteolytic function of TMPRSS4 as well as mechanisms for the control of invasion. Citation Format: Hye-Jin Min, Semi Kim. TMPRSS4 proteolytically activates pro-uPA to induce cancer cell invasion. [abstract]. In: Abstracts: AACR Special Conference on Cellular Heterogeneity in the Tumor Microenvironment; 2014 Feb 26-Mar 1; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(1 Suppl):Abstract nr A71. doi:10.1158/1538-7445.CHTME14-A71


Molecular Cancer Research | 2013

Abstract A096: MT-SP2/TMPRSS4 upregulates uPA gene expression through JNK signaling activation to induce cancer cell invasion

Yunhee Lee; Hye-Jin Min; Hana Lee; Xue-Feng Zhao; Young-Kyu Park; Semi Kim

The invasive nature of tumor cells is critical for cancer metastasis. Urokinase-type plasminogen activator (uPA) is a well-known serine protease involved in normal tissue remodeling, such as wound healing, and in pathological events, such as invasion and metastasis. Increased levels of uPA correlate with invasive properties and poor prognosis in breast, lung, stomach, bladder, colon, prostate and ovarian cancers. MT-SP2/TMPRSS4 is a novel type II transmembrane serine protease that is highly expressed in pancreatic, colon, and other cancer tissues. Previously, we demonstrated that MT-SP2/TMPRSS4 mediates tumor cell invasion, migration, and metastasis. However, the mechanisms by which MT-SP2/TMPRSS4 contributes to invasion are not fully understood. Here, we demonstrated that MT-SP2/TMPRSS4 induced the transcription of the uPA gene through activating the transcription factors Sp1, Sp3, and AP-1 in mainly a JNK-dependent manner and that the induction of uPA was required for MT-SP2/TMPRSS4-mediated cancer cell invasion and signaling events. In addition, the uPA receptor was involved in MT-SP2/TMPRSS4-induced signaling activation and subsequent uPA expression probably through its association with TMPRSS4 on the cell surface. Immunohistochemical analysis showed that uPA expression was significantly correlated with MT-SP2/TMPRSS4 expression in human carcinomas. These observations suggest that MT-SP2/TMPRSS4 is an important regulator of uPA gene expression; the upregulation of uPA by TMPRSS4 contributes to invasion and may represent a novel mechanism for the control of invasion. Citation Format: Yunhee Lee, Hye-Jin Min, Hana Lee, Xue-Feng Zhao, Young-Kyu Park, Semi Kim. MT-SP2/TMPRSS4 upregulates uPA gene expression through JNK signaling activation to induce cancer cell invasion. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research: Genetics, Biology, and Clinical Applications; Oct 3-6, 2013; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2013;11(10 Suppl):Abstract nr A096.


Cancer Research | 2010

Abstract 1032: Impact of HOXC9 recapitulation on cancer biology and clinical outcome in gastric cancer

Sungmin Moon; Chang-Soo Hong; Sang-Rok Lee; Xue-Feng Zhao; Hye Won Lee; Semi Kim; Sang-Uk Han; Ik-Joo Chung; Young-Kyu Park

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC The HOX genes encode transcription factors and may be implicated in the growth and the progression of many types of tumors. There are growing lines of evidence that dysregulation of HOX gene expression plays important roles in cancer. The aim of this study is to determine the level of HOXC9 expression during gastric cancer progression and the impact of its alteration on cancer biology and clinical outcome. HOXC9 expression was evaluated in normal mucosa, malignant legions, gastric cancer tissues by RT-PCR, western blot and immunohistochemistry. We established AGS gastric cancer cell in which HOXC9 was knock-down by shRNA and conditionally knock-down the HOXC9 expression by tetracycline-regulated gene expression (Tet-Off) system. The effect of HOXC9 knock-down on cell proliferation and apoptosis in AGS gastric cancer line were analyzed by tetrazolium salt-based proliferation assay (WST assay) and FACScan. Knockdown of HOXC9 by specific shRNA (67.3%) increased apoptosis, compared to scambled (7.3%) after 48 hours treatment of doxycycline, and increased active form of cellular apoptosis-related enzyme capsae-9, caspase-3 and poly (ADP-ribose) polymerase, whereas decreased expression of Bcl-xL, a mitochondria dependent anti-apoptosis factor. There was a gain or substantial increase of HOXC9 protein expression according to gastric cancer progression. Kaplan-Meier survival curves showed that HOXC9 overexpression is correlated with inferior disease-specific survival (P<0.001). multivariate Cox repression analysis revealed HOXC9 expression was an independent poor prognostic factor (Hazard ratio, 1.74; 95% CI, 1.01 to 2.99; P=0.047). We then demonstrated that HOXC9 directly regulated Bcl-xL transcription by binding to a HOXC9-responsive element in the Bcl-xL promoter by reporter gene assay, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA). In conclusion, these results suggest that HOXC9 overexpressed in advanced gastric cancer setting and its expression should be useful as a prognostic marker in gastric cancer patients. HOXC9 dependent Bcl-xL overexpresion may be an important mechanism by which HOXC9 protects gastric cancer cells from apoptosis. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1032.

Collaboration


Dive into the Semi Kim's collaboration.

Top Co-Authors

Avatar

Young Woo Park

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Hyo Jeong Hong

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar

Young-Kyu Park

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dong Heon Lee

Kangwon National University

View shared research outputs
Top Co-Authors

Avatar

Dong Sup Lee

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hye-Jin Min

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Researchain Logo
Decentralizing Knowledge