Network


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

Hotspot


Dive into the research topics where Hong Seo Choi is active.

Publication


Featured researches published by Hong Seo Choi.


Stem Cells | 2013

Heterogeneous Nuclear Ribonucleoprotein A2/B1 Regulates the Self‐Renewal and Pluripotency of Human Embryonic Stem Cells Via the Control of the G1/S Transition

Hong Seo Choi; Hyun Min Lee; Young-Joo Jang; Cheorl-Ho Kim; Chun Jeih Ryu

Self‐renewal and pluripotency of human embryonic stem cells (hESCs) are a complex biological process for maintaining hESC stemness. However, the molecular mechanisms underlying these special properties of hESCs are not fully understood. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) is a multifunctional RNA‐binding protein whose expression is related to cell proliferation and carcinogenesis. In this study, we found that hnRNP A2/B1 expression was localized to undifferentiated hESCs and decreased upon differentiation of hESCs. hnRNP A2/B1 knockdown reduced the number of alkaline phosphatase‐positive colonies in hESCs and led to a decrease in the expression of pluripotency‐associated transcription factors OCT4, NANOG, and SOX2, indicating that hnRNP A2/B1 is essential for hESC self‐renewal and pluripotency. hnRNP A2/B1 knockdown increased the expression of gene markers associated with the early development of three germ layers, and promoted the process of epithelial‐mesenchymal transition, suggesting that hnRNP A2/B1 is required for maintaining the undifferentiated and epithelial phenotypes of hESCs. hnRNP A2/B1 knockdown inhibited hESC proliferation and induced cell cycle arrest in the G0/G1 phase before differentiation via degradation of cyclin D1, cyclin E, and Cdc25A. hnRNP A2/B1 knockdown increased p27 expression and induced phosphorylation of p53 and Chk1, suggesting that hnRNP A2/B1 also regulates the G1/S transition of hESC cell cycle through the control of p27 expression and p53 and Chk1 activity. Analysis of signaling molecules further revealed that hnRNP A2/B1 regulated hESC proliferation in a PI3K/Akt‐dependent manner. These findings provide for the first time mechanistic insights into how hnRNP A2/B1 regulates hESC self‐renewal and pluripotency. STEM Cells 2013;31:2647–2658


Stem Cells and Development | 2011

Identification and characterization of adenovirus early region 1B-associated protein 5 as a surface marker on undifferentiated human embryonic stem cells.

Hong Seo Choi; Won-Tae Kim; Hana Kim; Jum-Ji Kim; Ji-Yun Ko; Sang-Wang Lee; Young Joo Jang; Sang Jick Kim; Min-Jung Lee; Han-Sung Jung; Julia Kzhyshkowska; Soo-Jong Um; Mi-Young Lee; Sang-Hun Lee; Cheorl-Ho Kim; Chun Jeih Ryu

Pluripotent human embryonic stem cells (hESCs) provide appropriate systems for developmental studies and prospective donor cell sources for regenerative medicine. Identification of surface markers specific to hESCs is a prerequisite for studying hESC biology and can be used to generate clinical-level donor cell preparations that are free from tumorigenic undifferentiated hESCs. We previously reported the generation of monoclonal antibodies that specifically recognize hESC surface antigens using a decoy immunization strategy. In this study, we show that monoclonal antibody 57-C11 recognizes a phosphorylated form of adenovirus early region 1B-associated protein 5 (E1B-AP5). E1B-AP5 is a nuclear RNA-binding protein, but we report that 57-C11-reactive E1B-AP5 is expressed on the surface of undifferentiated hESCs. In undifferentiated hESCs, 57-C11-reactive E1B-AP5 is localized to SSEA3-, SSEA4-, TRA-1-60-, TRA-1-81-, OCT4-, SOX2-, and NANOG-positive hESCs. In mixtures of undifferentiated hESCs and hESC-derived neurons, 57-C11 exclusively recognizes undifferentiated hESCs but not hESC-derived neuronal cells. Further, the expression of 57-C11-reactive E1B-AP5 decreases upon differentiation. Our results demonstrate that 57-C11-reactive E1B-AP5 is a novel surface molecule that is involved in the undifferentiated state of hESCs. As far as we know, this is the first report demonstrating that heterogeneous nuclear RNA-binding protein is expressed on the surface of undifferentiated hESCs.


Stem Cells | 2014

B‐Cell Receptor‐Associated Protein 31 Regulates Human Embryonic Stem Cell Adhesion, Stemness, and Survival via Control of Epithelial Cell Adhesion Molecule

Won-Tae Kim; Hong Seo Choi; Hyun Min Lee; Young-Joo Jang; Chun Jeih Ryu

B‐Cell receptor‐associated protein 31 (BAP31) regulates the export of secreted membrane proteins from the endoplasmic reticulum (ER) to the downstream secretory pathway. Previously, we generated a monoclonal antibody 297‐D4 against the surface molecule on undifferentiated human embryonic stem cells (hESCs). Here, we found that 297‐D4 antigen was localized to pluripotent hESCs and downregulated during early differentiation of hESCs and identified that the antigen target of 297‐D4 was BAP31 on the hESC‐surface. To investigate the functional role of BAP31 in hESCs, BAP31 expression was knocked down by small interfering RNA. BAP31 depletion impaired hESC self‐renewal and pluripotency and drove hESC differentiation into multicell lineages. BAP31 depletion hindered hESC proliferation by arresting cell cycle at G0/G1 phase and inducing caspase‐independent cell death. Interestingly, BAP31 depletion reduced hESC adhesion to extracellular matrix (ECM). Analysis of cell surface molecules showed decreased expression of epithelial cell adhesion molecule (EpCAM) in BAP31‐depleted hESCs, while ectopic expression of BAP31 elevated the expression of EpCAM. EpCAM depletion also reduced hESC adhesion to ECM, arrested cell cycle at G0/G1 phase and induced cell death, producing similar effects to those of BAP31 depletion. BAP31 and EpCAM were physically associated and colocalized at the ER and cell surface. Both BAP31 and EpCAM depletion decreased cyclin D1 and E expression and suppressed PI3K/Akt signaling, suggesting that BAP31 regulates hESC stemness and survival via control of EpCAM expression. These findings provide, for the first time, mechanistic insights into how BAP31 regulates hESC stemness and survival via control of EpCAM expression. Stem Cells 2014;32:2626–2641


Biochemical and Biophysical Research Communications | 2014

Detection of mycoplasma infection in circulating tumor cells in patients with hepatocellular carcinoma

Hong Seo Choi; Hyun Min Lee; Won-Tae Kim; Min Kyu Kim; Hee Jin Chang; Hye Ran Lee; Jae-Won Joh; Dae Shick Kim; Chun Jeih Ryu

Many studies have shown that persistent infections of bacteria promote carcinogenesis and metastasis. Infectious agents and their products can modulate cancer progression through the induction of host inflammatory and immune responses. The presence of circulating tumor cells (CTCs) is considered as an important indicator in the metastatic cascade. We unintentionally produced a monoclonal antibody (MAb) CA27 against the mycoplasmal p37 protein in mycoplasma-infected cancer cells during the searching process of novel surface markers of CTCs. Mycoplasma-infected cells were enriched by CA27-conjugated magnetic beads in the peripheral blood mononuclear cells in patients with hepatocellular carcinoma (HCC) and analyzed by confocal microscopy with anti-CD45 and CA27 antibodies. CD45-negative and CA27-positive cells were readily detected in three out of seven patients (range 12-30/8.5 ml blood), indicating that they are mycoplasma-infected circulating epithelial cells. CA27-positive cells had larger size than CD45-positive hematological lineage cells, high nuclear to cytoplasmic ratios and irregular nuclear morphology, which identified them as CTCs. The results show for the first time the existence of mycoplasma-infected CTCs in patients with HCC and suggest a possible correlation between mycoplasma infection and the development of cancer metastasis.


Scientific Reports | 2017

Cell-surface major vault protein promotes cancer progression through harboring mesenchymal and intermediate circulating tumor cells in hepatocellular carcinomas

Hyun Min Lee; Jae-Won Joh; Se-Ri Seo; Won-Tae Kim; Min Kyu Kim; Hong Seo Choi; So Young Kim; Young-Joo Jang; Dong Hyun Sinn; Gyu Seong Choi; Jong Man Kim; Choon Hyuck David Kwon; Hee Jin Chang; Dae Shick Kim; Chun Jeih Ryu

Circulating tumor cells (CTCs) play a major role in the metastasis and recurrence of hepatocellular carcinoma (HCC). Here, we found that major vault protein (MVP) is expressed on the surface of HCC cells and further induced under stressful environments. MVP knockdown reduces cell proliferation and induces apoptosis in HCC cells. Treatment of HCC cells with anti-MVP antibody (α-MVP) recognizing cell-surface MVP (csMVP) inhibits cell proliferation, migration, and invasion. csMVP-positive HCC cells have a higher clonogenic survival than csMVP-negative HCC cells, and treatment of HCC cells with α-MVP inhibits clonogenic survival, suggesting that csMVP contributes to HCC cell survival, migration, and invasion. The function of csMVP is mediated through mTOR, FAK, ERK and Akt signaling pathways. csMVP-positive CTCs are detected in HCC patients (89.7%) but not in healthy donors, and the number of csMVP-positive CTCs is further increased in patients with metastatic cancers. csMVP is exclusively detectable in CTCs with mesenchymal phenotype or intermediate phenotype with neither epithelial nor mesenchymal markers, suggesting that csMVP-associated survival and metastatic potential harbor CTCs with nonepithelial phenotypes. The results suggest that csMVP promotes cancer progression and serves as a surface marker for mesenchymal and intermediate CTCs in patients with HCC and metastatic cancers.


Monoclonal antibodies in immunodiagnosis and immunotherapy | 2013

Characterization of Monoclonal Antibodies Recognizing 130 kDa Surface Proteins on Human Embryonic Stem Cells and Cancer Cell Lines

Jum-Ji Kim; Hong Seo Choi; Mi-Young Lee; Chun Jeih Ryu

To study cell surface proteins expressed on human embryonic stem cells (hESCs), we generated a panel of monoclonal antibodies (MAbs) against undifferentiated hESCs by a decoy immunization strategy in a previous study. Two of the MAbs, 63-B6 and 246-D7, bound to human pluripotent stem cells but not to human primary cells such as human peripheral blood mononuclear cells and human lung fibroblasts. They did not bind to either mouse embryonic stem cells or mouse embryonic fibroblasts. The two MAbs had similar binding profiles for many various cancer cells, with few exceptions. Expression of antigens recognized by the two MAbs was rapidly decreased during embryoid body formation of hESCs and gradually increased after initial decrease. The MAbs recognized approximately 130 kDa proteins on the surface of hESCs. Cloning and sequence analysis of antibody genes showed that although the MAbs had exactly the same light chain sequences, they had different heavy chain sequences. Taken together, the results suggest that the two MAbs may recognize two different epitopes of the same or different 130 kDa surface proteins involved in regulating the early differentiation of human pluripotent stem cells and cancer cells.


Hybridoma | 2011

Detection and Characterization of 2-E2-Specific Surface Protein in Human Pluripotent Stem Cells

Hana Kim; Hong Seo Choi; Cheorl-Ho Kim; Chun Jeih Ryu

To investigate cell surface antigens on human embryonic stem cells (hESCs), we generated a panel of monoclonal antibodies (MAbs) against undifferentiated hESCs by a decoy immunization strategy. One of the MAbs, MAb 2-E2, specifically bound to human pluripotent stem cells but not to mouse pluripotent stem cells and mouse embryonic fibroblasts. 2-E2 also bound to human differentiated cells, peripheral blood monocytes, and dermal fibroblasts. 2-E2 antigen expression drastically decreased in retinoic acid-induced differentiated hESCs. However, it gradually increased after initial decrease during embryoid body formation of hESCs. 2-E2 recognized approximately 68 and 27 kDa proteins present on the surface of human pluripotent stem cells. The results suggest that 2-E2-specific surface protein is a novel cell surface protein that plays a role in the early differentiation of human pluripotent stem cells.


Scientific Reports | 2018

Progesterone Receptor Membrane Component 1 suppresses the p53 and Wnt/β-catenin pathways to promote human pluripotent stem cell self-renewal

Ji Yea Kim; So Young Kim; Hong Seo Choi; Min Kyu Kim; Hyun Min Lee; Young-Joo Jang; Chun Jeih Ryu

Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional heme-binding protein involved in various diseases, including cancers and Alzheimer’s disease. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 against surface molecules on human pluripotent stem cells (hPSCs). Here we show that PGRMC1 is the target antigen of both MAbs, and is predominantly expressed on hPSCs and some cancer cells. PGRMC1 is rapidly downregulated during early differentiation of hPSCs. Although PGRMC1 knockdown leads to a spread-out morphology and impaired self-renewal in hPSCs, PGRMC1 knockdown hPSCs do not show apoptosis and autophagy. Instead, PGRMC1 knockdown leads to differentiation of hPSCs into multiple lineage cells without affecting the expression of pluripotency markers. PGRMC1 knockdown increases cyclin D1 expression and decreases Plk1 expression in hPSCs. PGRMC1 knockdown also induces p53 expression and stability, suggesting that PGRMC1 maintains hPSC self-renewal through suppression of p53-dependent pathway. Analysis of signaling molecules further reveals that PGRMC1 knockdown promotes inhibitory phosphorylation of GSK-3β and increased expression of Wnt3a and β-catenin, which leads to activation of Wnt/β-catenin signaling. The results suggest that PGRMC1 suppresses the p53 and Wnt/β-catenin pathways to promote self-renewal and inhibit early differentiation in hPSCs.


PLOS ONE | 2017

Correction: Epitope mapping of antibodies suggests the novel membrane topology of B-cell receptor associated protein 31 on the cell surface of embryonic stem cells: The novel membrane topology of BAP31 (PLoS ONE (2015) 10:6 (e0130670) DOI: 10.1371/journal.pone.0130670)

Won Tae Kim; Hong Seo Choi; Hyo Jeong Hwang; Han Sung Jung; Chun Jeih Ryu

[This corrects the article DOI: 10.1371/journal.pone.0130670.].


PLOS ONE | 2017

Enhanced expression of cell-surface B-cell receptor-associated protein 31 contributes to poor survival of non-small cell lung carcinoma cells

Se-Ri Seo; Hyun Min Lee; Hong Seo Choi; Won-Tae Kim; Eun-Wie Cho; Chun Jeih Ryu

B-cell receptor-associated protein 31 (BAP31) is an endoplasmic reticulum (ER) membrane protein which plays a role as a molecular chaperone for the newly synthesized transmembrane proteins. BAP31 is also an important apoptosis regulator for extrinsic apoptosis induction in the ER membrane. Recent studies have shown that BAP31 is also expressed on the surface of embryonic stem cells. However, the function of cell surface BAP31 (csBAP31) still remains unclarified. In an attempt to search for surface markers on tumorspheres, here, we generated monoclonal antibodies (MAbs) against the sphere cells from the non-small cell lung carcinoma cell (NSCLC) line A549. SP1-B7, one of the MAbs, recognized csBAP31 whose expression was further increased on A549 sphere cells, as compared with A549 adherent cells. To investigate the role of csBAP31 in A549 cells, A549 adherent and sphere cells were stained with annexin V, propidium iodide, and SP1-B7. Interestingly, annexin V-high cells showed increased expression of csBAP31 as compared with annexin V-low cells. Caspase-3/7 activity was also increased in csBAP31-high cells as compared with csBAP31-low cells, suggesting that csBAP31-high cells are more sensitive to apoptosis. To further demonstrate the survival of csBAP31-positive A549 cells, csBAP31-positive and -negative A549 cells were sorted and subjected to the clonogenic survival assay. The colony number of csBAP31-positive A549 cells was decreased by approximately 1.7-fold, as compared that of csBAP31-negative A549 cells, suggesting that csBAP31-positve cells are sensitive to cell death indeed. The results suggest that enhanced expression of csBAP31 contributes to poor survival of NSCLC cells.

Collaboration


Dive into the Hong Seo Choi's collaboration.

Top Co-Authors

Avatar

Chun Jeih Ryu

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Hyun Min Lee

Biotechnology Institute

View shared research outputs
Top Co-Authors

Avatar

Min Kyu Kim

Biotechnology Institute

View shared research outputs
Top Co-Authors

Avatar

Won-Tae Kim

Biotechnology Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jae-Won Joh

Samsung Medical Center

View shared research outputs
Top Co-Authors

Avatar

Se-Ri Seo

Biotechnology Institute

View shared research outputs
Top Co-Authors

Avatar

So Young Kim

Biotechnology Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge