Network


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

Hotspot


Dive into the research topics where Seung Min Shin is active.

Publication


Featured researches published by Seung Min Shin.


BMC Cancer | 2006

The bone morphogenetic protein antagonist gremlin 1 is overexpressed in human cancers and interacts with YWHAH protein

Hong Namkoong; Seung Min Shin; Hyun Kee Kim; Seon-Ah Ha; Goang Won Cho; Soo Young Hur; Tae Eung Kim; Jin Woo Kim

BackgroundBasic studies of oncogenesis have demonstrated that either the elevated production of particular oncogene proteins or the occurrence of qualitative abnormalities in oncogenes can contribute to neoplastic cellular transformation. The purpose of our study was to identify an unique gene that shows cancer-associated expression, and characterizes its function related to human carcinogenesis.MethodsWe used the differential display (DD) RT-PCR method using normal cervical, cervical cancer, metastatic cervical tissues, and cervical cancer cell lines to identify genes overexpressed in cervical cancers and identified gremlin 1 which was overexpressed in cervical cancers. We determined expression levels of gremlin 1 using Northern blot analysis and immunohistochemical study in various types of human normal and cancer tissues. To understand the tumorigenesis pathway of identified gremlin 1 protein, we performed a yeast two-hybrid screen, GST pull down assay, and immunoprecipitation to identify gremlin 1 interacting proteins.ResultsDDRT-PCR analysis revealed that gremlin 1 was overexpressed in uterine cervical cancer. We also identified a human gremlin 1 that was overexpressed in various human tumors including carcinomas of the lung, ovary, kidney, breast, colon, pancreas, and sarcoma. PIG-2-transfected HEK 293 cells exhibited growth stimulation and increased telomerase activity. Gremlin 1 interacted with homo sapiens tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, eta polypeptide (14-3-3 eta; YWHAH). YWHAH protein binding site for gremlin 1 was located between residues 61–80 and gremlin 1 binding site for YWHAH was found to be located between residues 1 to 67.ConclusionGremlin 1 may play an oncogenic role especially in carcinomas of the uterine cervix, lung, ovary, kidney, breast, colon, pancreas, and sarcoma. Over-expressed gremlin 1 functions by interaction with YWHAH. Therefore, Gremlin 1 and its binding protein YWHAH could be good targets for developing diagnostic and therapeutic strategies against human cancers.


Clinical Cancer Research | 2004

Cancer-Associated Expression of Minichromosome Maintenance 3 Gene in Several Human Cancers and Its Involvement in Tumorigenesis

Seon-Ah Ha; Seung Min Shin; Hong Namkoong; Heejeong Lee; Goang Won Cho; Soo Young Hur; Tae Eung Kim; Jin Woo Kim

Purpose: The purpose of our study was to identify an unique gene that shows cancer-associated expression, evaluates its potential usefulness in cancer diagnosis, and characterizes its function related to human carcinogenesis. Experimental Design: We used the differential display reverse transcription-PCR method with normal cervical, cervical cancer and metastatic tissues, and cervical cancer cell line to identify genes overexpressed in cancers. Results: We identified a minichromosome maintenance 3 (MCM3) gene that was overexpressed in various human cancers, including leukemia, lymphoma, and carcinomas of the uterine cervix, colon, lung, stomach, kidney and breast, and malignant melanoma. Western blot and immunohistochemical analyses also revealed that MCM3 protein was elevated in most of human cancer tissues tested. We compared the MCM3 protein expression levels in human cancers with conventional proliferation markers, Ki-67 and proliferating cell nuclear antigen. MCM3 antibody was the most specific for multiple human cancers, whereas proliferating cell nuclear antigen was relatively less effective in specificity, and Ki-67 failed to detect several human cancers. The down-regulation of MCM3 protein level was examined under serum starvation in both normal and cancer cells. Interestingly, MCM3 protein was stable in MCF-7 breast cancer cells even up to 96 hours after serum starvation, whereas it was gradually degraded in normal BJ fibroblast cells. Nude mice who received injections of HEK 293 cells stably transfected with MCM3 formed tumors in 6 weeks. Conclusions: Our study indicates that determination of MCM3 expression level will facilitate the assessment of many different human malignancies in tumor diagnosis, and MCM3 is involved in multiple types of human carcino-genesis.


Oncogene | 2003

Identification and differential expression of novel human cervical cancer oncogene HCCR-2 in human cancers and its involvement in p53 stabilization.

Jesang Ko; Young Han Lee; Seung Yong Hwang; Youn Soo Lee; Seung Min Shin; Jae Hoon Hwang; Jin Kim; Yong Wook Kim; Sung Wuk Jang; Zae Young Ryoo; In Kyung Kim; Sung Eun Namkoong; Jin Woo Kim

Basic studies of oncogenesis have demonstrated that either the elevated production of particular oncogene proteins or the occurrence of qualitative abnormalities in oncogenes can contribute to neoplastic cellular transformation. The purpose of this study was to identify unique oncogenes that are differentially expressed in human cancers and characterize their functions in tumorigenesis. To discover new putative oncogenes, the differential display RT–PCR method was applied using normal cervical tissues, cervical cancer cell lines, cervical cancer tissues, and metastatic tissues. We identified a new human cervical cancer oncogene HCCR-2 that was overexpressed in various human tumors including leukemia, lymphoma, and carcinomas of the breast, kidney, ovary, stomach, colon, and uterine cervix. Ectopic expression of HCCR-2 resulted in direct tumorigenic conversions of NIH/3T3 and Rat1 fibroblasts. Nude mice injected with NIH/3T3 cells stably transfected with HCCR-2 formed tumors in 4 weeks. The resultant tumors display characteristics of an epithelial carcinoma. In HCCR-2 transfected NCI-H460 cells and RKO cells, stabilization of the p53 tumor suppressor occurred without genetic mutation and correlated with functional impairment, as indicated by the defective induction of p53-induced p21WAF1, MDM2, and bax. These results indicate that HCCR-2 probably represents a new oncogene that is related to tumorigenesis, functioning as a negative regulator of the p53 tumor suppressor.


Oncogene | 2004

Transgenic mouse model for breast cancer: Induction of breast cancer in novel oncogene HCCR-2 transgenic mice

Jesang Ko; Seung Min Shin; Young Mi Oh; Youn Soo Lee; Zae Yoong Ryoo; Young Han Lee; Doe Sun Na; Jin Woo Kim

Transgenic mice containing novel oncogene HCCR-2 were generated to analyse the phenotype and to characterize the role of HCCR-2 in cellular events. Mice transgenic for HCCR-2 developed breast cancers and metastasis. The level of p53 in HCCR-2 transgenic mice was elevated in most tissues including breast, brain, heart, lung, liver, stomach, kidney, spleen, and lymph node. We examined whether stabilized p53 is functional in HCCR-2 transgenic mice. Defective induction of p53 responsive genes including p21WAF1, MDM2, and bax indicates that stabilized p53 in HCCR-2 transgenic mice exists in an inactive form. These results suggest that HCCR-2 represents an oncoprotein that is related to breast cancer development and regulation of the p53 tumor suppressor.


Experimental and Molecular Pathology | 2010

Minichromosome maintenance protein 3 is a candidate proliferation marker in papillary thyroid carcinoma.

Youn Soo Lee; Seon-Ah Ha; Hae Joo Kim; Seung Min Shin; Hyun Kee Kim; Sanghee Kim; Chang Suk Kang; Kyo Young Lee; Oak Kee Hong; Seung Hwan Lee; Hyuk-Sang Kwon; Bong-Yun Cha; Jin Woo Kim

The proliferative capacity of tumor cells is a characteristic feature in the whole growing tumors. Many pathologists and clinicians have used the estimation of cell proliferation for prognostic information. Minichromosome maintenance protein 3 (MCM3) is known to have a role on the initiation and regulation of DNA replication during cell cycle. The aim of this study was to evaluate the potential applicability of one of the MCM proteins, MCM3, as a proliferation marker in papillary thyroid carcinoma (PTC) with correlation to clinicopathological parameters. We performed the immunohistochemical analysis for MCM3 and Ki-67 in 60 cases of PTC and Western blot analysis for MCM3 expression in 6 PTCs and normal thyroid tissues. The comparison of MCM3 labeling index (LI) to tumor size (P=0.031) and extrathyroidal extension (P=0.037) was statistically significant while that of Ki-67 LI to them was not. Moreover, a significant association was not observed between MCM3 and Ki-67, but the MCM3 LI was considerably higher. Western blot analyses revealed that the MCM3 protein expression levels were overexpressed in all PTCs. On the contrary, the levels of MCM3 were very low or absent in all normal thyroid tissues. Our results indicate that MCM3 may be a more reliable proliferation marker than Ki-67 in accessing the growth of tumor and evaluating tumor aggressiveness of PTC.


Clinical Cancer Research | 2005

The HCCR Oncoprotein as a Biomarker for Human Breast Cancer

Sang Seol Jung; Hyung Soon Park; Insong James Lee; Hong Namkoong; Seung Min Shin; Goang Won Cho; Seon Ah Ha; Yong Gyu Park; Youn Soo Lee; Jesang Ko; Jin Woo Kim

Purpose: HCCR oncoprotein is reported to be related to tumorigenesis, including breast cancer, functioning as a negative regulator of p53. Mice transgenic for HCCR developed breast cancers. The objective of this study was to validate the HCCR oncoprotein as a candidate biomarker for breast cancer. Experimental Design: HCCR expression in breast cancer cells was analyzed by quantitative PCR, ELISA, immunohistochemistry, Western blotting, fluorescence-activated cell sorting, and confocal microscopy. Epitope areas were determined using mass spectrometry through the analysis of time-dependent tryptic fragment patterns of HCCR. HCCR expression profiles in breast cancer patient sera were analyzed, and correlations with clinicopathologic data and carbohydrate antigen 15-3 (CA15-3) levels were determined. Results: HCCR was up-regulated in breast cancer cells and tissues. The epitope regions of HCCR recognized by monoclonal antibody (BCS-1) were HFWTPK and QQTDFLDIYHAFR. According to fluorescence-activated cell sorting and confocal microscopic analysis, BCS-1 was bound to HCCR antigen on the cell surface. Serum HCCR concentrations were measured using ELISA from 299 subjects, including 129 patients with breast cancer, 24 patients with benign breast disease, and 158 normal volunteers, and comparisons were made to CA15-3. Serologic studies revealed an 86.8% sensitivity for HCCR in breast cancer, which was higher than 21.0% for CA15-3. Eighty-six of 98 (87.8%) patients with breast cancers that were negative for CA15-3 were positive for HCCR-1. A positive response rate of 83.3% was identified even at early stages for pathologic factors in breast cancer. Conclusions: The HCCR assay has an advantage over CA15-3 in diagnosing breast cancer and detecting early stages of the disease.


BMC Cell Biology | 2010

Transdifferentiation-inducing HCCR-1 oncogene

Seon-Ah Ha; Hyun Kee Kim; Jinah Yoo; Sanghee Kim; Seung Min Shin; Youn Soo Lee; Soo Young Hur; Yong Wook Kim; Tae E. Kim; Yeun J Chung; Shin Soo Jeun; Dong Wook Kim; Yong G. Park; Jin Kim; Soon Young Shin; Young Han Lee; Jin W. Kim

BackgroundCell transdifferentiation is characterized by loss of some phenotypes along with acquisition of new phenotypes in differentiated cells. The differentiated state of a given cell is not irreversible. It depends on the up- and downregulation exerted by specific molecules.ResultsWe report here that HCCR-1, previously shown to play an oncogenic role in human cancers, induces epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET) in human and mouse, respectively. The stem cell factor receptor CD117/c-Kit was induced in this transdifferentiated (EMT) sarcoma tissues. This MET occurring in HCCR-1 transfected cells is reminiscent of the transdifferentiation process during nephrogenesis. Indeed, expression of HCCR-1 was observed during the embryonic development of the kidney. This suggests that HCCR-1 might be involved in the transdifferentiation process of cancer stem cell.ConclusionsTherefore, we propose that HCCR-1 may be a regulatory factor that stimulates morphogenesis of epithelia or mesenchyme during neoplastic transformation.


BMC Cell Biology | 2007

HCCR-1, a novel oncogene, encodes a mitochondrial outer membrane protein and suppresses the UVC-induced apoptosis.

Goang-Won Cho; Seung Min Shin; Hyun Kee Kim; Seon-Ah Ha; Sanghee Kim; Joo-Hee Yoon; Soo Young Hur; Tae Eung Kim; Jin Woo Kim

BackgroundThe Human cervical cancer oncogene (HCCR-1) has been isolated as a human oncoprotein, and has shown strong tumorigenic features. Its potential role in tumorigenesis may result from a negative regulation of the p53 tumor suppressor gene.ResultsTo investigate the biological function of HCCR-1 in the cell, we predicted biological features using bioinformatic tools, and have identified a LETM1 homologous domain at position 75 to 346 of HCCR-1. This domain contains proteins identified from diverse species predicted to be mitochondrial proteins. Fluorescence microscopy and fractionation experiments showed that HCCR-1 is located in mitochondria in the COS-7, MCF-7 and HEK/293 cell lines, and subcompartamentally at the outer membrane in the HEK/293 cell line. The topological structure was revealed as the NH2-terminus of HCCR-1 oriented toward the cytoplasm. We also observed that the D1-2 region, at position 1 to 110 of HCCR-1, was required and sufficient for posttranslational mitochondrial import. The function of HCCR-1 on mitochondrial membrane is to retard the intrinsic apoptosis induced by UVC and staurosporine, respectively.ConclusionOur experiments show the biological features of HCCR-1 in the cell, and suggest that uncontrolled expression of HCCR-1 may cause mitochondrial dysfunction that can result in resisting the UVC or staurosporine-induced apoptosis and progressing in the tumor formation.


BMC Cancer | 2009

Oncoprotein HCCR-1 expression in breast cancer is well correlated with known breast cancer prognostic factors including the HER2 overexpression, p53 mutation, and ER/PR status.

Seon-Ah Ha; Youn Soo Lee; Seung Min Shin; Hyun Kee Kim; Sanghee Kim; Hong Namkoong; Hae Joo Kim; Sang Min Jung; Yu Sun Lee; Yeun Jun Chung; Sang Seol Jung; Jin Woo Kim

BackgroundOncoprotein HCCR-1 functions as a negative regulator of the p53 and contributes breast tumorigenesis. The serum HCCR-1 assay is useful in diagnosing breast cancer and mice transgenic for HCCR developed breast cancers. But it is unknown how HCCR-1 contributes to human breast tumorigenesis.MethodsOncogene HCCR-1 expression levels were determined in normal breast tissues, breast cancer tissues and cancer cell lines. We examined whether HCCR-1 protein expression in breast cancer is related to different biological characteristics, including ER, PR, p53 genotype, and HER2 status in 104 primary breast cancer tissues using immunohistochemical analyses.ResultsHCCR-1 was upregulated in breast cancer cells and tissues compared with normal breast tissues. In this study, overexpression of HCCR-1 was well correlated with known breast cancer prognostic markers including the presence of steroid receptors (ER and PR), p53 mutation and high HER2 overexpression. HCCR-1 was not detected in the ER-negative, PR-negative, p53 negative and low HER2 breast cancer tissues. These data indicate that the level of HCCR-1 in breast cancer tissues is relatively well correlated with known breast cancer factors, including the HER2 overexpression, p53 mutation, and ER/PR status.ConclusionDetermination of HCCR-1 levels as options for HER2 testing is promising although it needs further evaluation.


International Journal of Cancer | 2002

Candidate tumor suppressor, HCCS‐1, is downregulated in human cancers and induces apoptosis in cervical cancer

Tae E. Kim; Yong Wook Kim; Seung Yong Hwang; Seung Min Shin; Jin W. Shin; Young Hwan Lee; Sun Y. Shin; Ku T. Han; Joon Mo Lee; Sung Eun Namkoong; Jin W. Kim

To identify the genes involved in cervical carcinogenesis, we applied the mRNA differential display method and identified a candidate tumor suppressor gene, HCCS‐1, which was present in normal cervical tissue but absent in cervical cancer, metastatic lymph node and CUMC‐6 cervical cancer cell line. HCCS‐1 transcripts were expressed in many normal tissues including leukocyte, lung, spleen, liver, heart and uterine cervix. Its expression was absent in 8 human cancer cell lines. HCCS‐1‐transfected HeLa cells exhibited growth inhibition by about 50%. This inhibitory effect of HCCS‐1 on cervical cancer cells was associated with apoptotic process including DNA fragmentation. HCCS‐1‐transfected HeLa cells were shown to release cytochrome c from mitochondria, which activates caspase‐9 and ‐3 and finally results in cleavage of poly(ADP‐ribose) polymerase. Apoptosis formation was detected by propidium‐iodide/annexin V. HCCS‐1‐transfected HeLa cells were more sensitive to adriamycin or UVC ray triggered apoptosis. These results suggest that HCCS‐1 is downregulated in multiple human tumor types and may serve as a candidate tumor suppressor gene through apoptotic pathway against human cervical cancer.

Collaboration


Dive into the Seung Min Shin's collaboration.

Top Co-Authors

Avatar

Jin Woo Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Hyun Kee Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Seon-Ah Ha

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Youn Soo Lee

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Hong Namkoong

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Sanghee Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Soo Young Hur

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Tae Eung Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Goang Won Cho

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge