Network


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

Hotspot


Dive into the research topics where Hee Doo Yang is active.

Publication


Featured researches published by Hee Doo Yang.


Journal of Hepatology | 2015

MicroRNA-221 governs tumor suppressor HDAC6 to potentiate malignant progression of liver cancer

Hyun Jin Bae; Kwang Hwa Jung; Jung Woo Eun; Qingyu Shen; Hyung Seok Kim; Se Jin Park; Woo Chan Shin; Hee Doo Yang; Won Sang Park; Jung Young Lee; Suk Woo Nam

BACKGROUND & AIMS Most common reason behind changes in histone deacetylase (HDAC) function is its overexpression in cancer. However, among HDACs in liver cancer, HDAC6 is uniquely endowed with a tumor suppressor, but the mechanism underlying HDAC6 inactivation has yet to be uncovered. METHODS Microarray profiling and target prediction programs were used to identify miRNAs targeting HDAC6. A series of inhibitors, activators and siRNAs was introduced to validate regulatory mechanisms for microRNA-221-3p (miR-221) governing HDAC6 in hepatocarcinogenesis. RESULTS Comprehensive miRNA profiling analysis identified seven putative endogenous miRNAs that are significantly upregulated in hepatocellular carcinoma (HCC). While miR-221 was identified as a suppressor of HDAC6 by ectopic expression of miRNA mimics in Dicer knockdown cells, targeted-disruption of miR-221 repressed cancer cell growth through derepressing HDAC6 expression. Suppression of HDAC6 via miR-221 was induced by JNK/c-Jun signaling in liver cancer cells but not in normal hepatic cells. Additionally, cytokine-induced NF-κBp65 independently regulated miR-221, thereby suppressing HDAC6 expression in HCC cells. HCC tissues derived from chemical-induced rat and H-ras12V transgenic mice liver cancer models validated that JNK/c-Jun activation and NF-κBp65 nuclear translocation are essential for the transcription of miR-221 leading to repression of HDAC6 in HCC. CONCLUSIONS Our findings suggest that the functional loss or suppression of the tumor suppressor HDAC6 is caused by induction of miR-221 through coordinated JNK/c-Jun- and NF-κB-signaling pathways during liver tumorigenesis, providing a novel target for the molecular treatment of liver malignancies.


Oncotarget | 2016

Oncogenic potential of histone-variant H2A.Z.1 and its regulatory role in cell cycle and epithelial-mesenchymal transition in liver cancer.

Hee Doo Yang; Pum-Joon Kim; Jung Woo Eun; Qingyu Shen; HyungSeok Kim; Woo Chan Shin; Young-Min Ahn; Won Park; Jung Young Lee; Suk Woo Nam

H2A.Z is a highly conserved H2A variant, and two distinct H2A.Z isoforms, H2A.Z.1 and H2A.Z.2, have been identified as products of two non-allelic genes, H2AFZ and H2AFV. H2A.Z has been reported to be overexpressed in breast, prostate and bladder cancers, but most studies did not clearly distinguish between isoforms. One recent study reported a unique role for the H2A.Z isoform H2A.Z.2 as a driver of malignant melanoma. Here we first report that H2A.Z.1 plays a pivotal role in the liver tumorigenesis by selectively regulating key molecules in cell cycle and epithelial-mesenchymal transition (EMT). H2AFZ expression was significantly overexpressed in a large cohort of hepatocellular carcinoma (HCC) patients, and high expression of H2AFZ was significantly associated with their poor prognosis. H2A.Z.1 overexpression was demonstrated in a subset of human HCC and cell lines. H2A.Z.1 knockdown suppressed HCC cell growth by transcriptional deregulation of cell cycle proteins and caused apoptotic cell death of HCC cells. We also observed that H2A.Z.1 knockdown reduced the metastatic potential of HCC cells by selectively modulating epithelial-mesenchymal transition regulatory proteins such as E-cadherin and fibronectin. In addition, H2A.Z.1 knockdown reduced the in vivo tumor growth rate in a mouse xenograft model. In conclusion, our findings suggest the oncogenic potential of H2A.Z.1 in liver tumorigenesis and that it plays established role in accelerating cell cycle transition and EMT during hepatocarcinogenesis. This makes H2A.Z.1 a promising target in liver cancer therapy.


Journal of Applied Toxicology | 2015

Characteristic molecular and proteomic signatures of drug-induced liver injury in a rat model.

Jung Woo Eun; Hyun Jin Bae; Qingyu Shen; Se Jin Park; Hyung Seok Kim; Woo Chan Shin; Hee Doo Yang; Chan Young Jin; Jueng Soo You; Hyun Joo Kang; Hoguen Kim; Young-Min Ahn; Won Sang Park; Jung Young Lee; Suk Woo Nam

Drug‐induced liver injury (DILI) is a major safety concern during drug development and remains one of the main reasons for withdrawal of drugs from the market. Although it is crucial to develop methods that will detect potential hepatotoxicity of drug candidates as early and as quickly as possible, there is still a lack of sensitive and specific biomarkers for DILI that consequently leads to a scarcity of reliable hepatotoxic data. Hence, in this study, we assessed characteristic molecular signatures in rat liver treated with drugs (pyrazinamide, ranitidine, enalapril, carbamazepine and chlorpromazine) that are known to cause DILI in humans. Unsupervised hierarchical clustering analysis of transcriptome changes induced by DILI‐causing drugs resulted in three different subclusters on dendrogram, i.e., hepatocellular, cholestatic and mixed type of DILI at early time points (2 days), and multiclassification analysis suggested 31 genes as discernible markers for each DILI pattern. Further analysis for characteristic molecular signature of each DILI pattern provided a molecular basis for different modes of DILI action. A proteomics study of the same rat livers was used to confirm the results, and the two sets of data showed 60 matching classifiers. In conclusion, the data of different DILI‐causing drug treatments from genomic analysis in a rat model suggest that DILI‐specific molecular signatures can discriminate different patterns of DILI at an early exposure time point, and that they provide useful information for mechanistic studies that may lead to a better understanding of the molecular basis of DILI. Copyright


Cancer Letters | 2014

HDAC6 sustains growth stimulation by prolonging the activation of EGF receptor through the inhibition of rabaptin-5-mediated early endosome fusion in gastric cancer

Se Jin Park; Jeong Kyu Kim; Hyun Jin Bae; Jung Woo Eun; Qingyu Shen; Hyung Seok Kim; Woo Chan Shin; Hee Doo Yang; Eun Kyung Lee; Jueng Soo You; Won Sang Park; Jung Young Lee; Suk Woo Nam

The aberrant regulation of histone deacetylase 6 (HDAC6) contributes to malignant progression in various types of cancer, but the mechanism underlying gastric carcinogenesis remains unknown. Aberrant HDAC6 overexpression was observed in a subset of human gastric cancer cells. HDAC6 knockdown caused the significant inhibition of gastric cancer cell growth without affecting the transition of cell cycles or the processing of cell death. We demonstrate that an increase in epidermal growth factor receptor (EGFR) signaling through decreased EGFR degradation was mediated by HDAC6 in gastric carcinogenesis. These results establish a molecular mechanism responsible for oncogenic HDAC6, explaining how EGFR signaling induced by the growth factor is sustained during the malignant progression of gastric cancer.


Cancer Research | 2016

Abstract 4810: Histone deacetylase 2 is a potential determinant of the sensitivity of hepatocellular carcinoma cells to sorafenib

Hyung Seok Kim; Jung Woo Eun; Qingyu Shen; Woo Chan Shin; Hee Doo Yang; Sang Yeon Kim; Suk Woo Nam

Hepatocellular carcinoma (HCC) is the fifth frequently diagnosed cancer and is the second leading cause of cancer death worldwide. Many groups have suggested possible clinical applications for liver cancer therapy but sorafenib, an orally-available kinase inhibitor, is the only standard systematic therapeutics for treatment of hepatocellular carcinoma. However, survival benefit of sorafenib is unsatisfactory due to high level heterogeneity of individual response. We previously reported that histone deacetylase 2 (HDAC2) was deregulated in HCC, and thereby contributed to liver tumorigenesis by enhancing mitotic and metastatic potential of transformed cells. Targeted-disruption of HDAC2 suppressed in vitro and in vivo tumorgigenic potential of HDAC2 in liver cancer. The goal of this study was to investigate whether both sorafenib treatment and HDAC2 inhibition elicit synergistic anti-tumor effect against HCC cells. Here, we showed that sorafenib effectively blocked ERK/MEK and AKT signaling pathway upon EGF stimulation in liver cancer cell lines. Also, we found that valproic acid (VPA), a selective inhibitor of histone deacetylase family I, and sorafenib treatment synergistically evoked liver cancer cell growth retardation. This effect was recapitulated by HDAC2 knockdown with sorafenib treatment and exhibited synergistic effect on apoptotic cell death of liver cancer cell lines, Hep3B and Huh7, compared to single treatment of sorafenib. Our data suggest that combined sorafenib treatment with HDAC2 targeting may provide more benefits toward HCC therapy providing a novel approach for future application in patients with advanced HCC. Citation Format: Hyung Seok Kim, Jung Woo Eun, Qingyu Shen, Woo Chan Shin, Hee Doo Yang, Sang Yeon Kim, Suk Woo Nam. Histone deacetylase 2 is a potential determinant of the sensitivity of hepatocellular carcinoma cells to sorafenib. [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 4810.


Cancer Research | 2015

Abstract 3111: JNK/c-Jun- and NF-κB-mediated microRNA-221 governs tumor suppressor HDAC6 to potentiate malignant progression of liver cancer

Hyung Seok Kim; Qingyu Shen; Jung Woo Eun; Woo Chan Shin; Hee Doo Yang; Won Sang Park; Joung Young Lee; Suk Woo Nam

Many clinical studies of cancer patients have established that the most common reason behind changes in HDAC function is its overexpression. However, among HDACs in liver cancer, HDAC6, a cytoplasmic tubulin deacetylase, is uniquely endowed with a tumor suppressor, but the mechanism underlying HDAC6 inactivation has yet to be uncovered. In this study we show that genetic and epigenetic dysregulation was not part of the HDAC6 inactivation mechanism. Instead, microRNA-221-3p (miR-221) directly suppressed HDAC6 in hepatocellular carcinoma (HCC) cells. A comprehensive miRNA profiling analysis identified seven putative endogenous miRNAs that are significantly upregulated in HCC. Ectopic expression of miRNA mimics in Dicer knockdown cells evidenced miR-221 to suppress HDAC6 in HCC cells. Notably, targeted-disruption of miR-221 repressed cancer cell growth and proliferation, and it recapitulated HDAC6 overexpression effects in HCC cells. Our results also demonstrate that c-Met-mediated c-Jun NH2-terminal kinase (JNK)/c-Jun signaling induced miR-221 to suppress HDAC6 and was sustained in liver cancer cells but not in normal hepatic cells. In addition, cytokine-induced NF-κBp65 independently regulated miR-221, thereby suppressing HDAC6 expression in liver cancer cells. HCC tissues derived from chemical-induced rat liver cancer models validated that JNK/c-Jun activation and NF-kBp65 nuclear translocation are essential for the transcriptional activation of oncogenic miR-221 to repress tumor suppressor HDAC6 in HCC. In conclusion, our finding suggest that the functional loss or suppression of the tumor suppressor HDAC6 is caused by miR-221 through coordinated JNK/c-Jun- and NF-κB-signaling pathways during liver tumorigenesis, providing a novel target for the molecular treatment of liver malignancies. Citation Format: Hyung Seok Kim, Qingyu Shen, Jung Woo Eun, Woo Chan Shin, Hee Doo Yang, Won Sang Park, Joung Young Lee, Suk Woo Nam. JNK/c-Jun- and NF-κB-mediated microRNA-221 governs tumor suppressor HDAC6 to potentiate malignant progression of liver cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3111. doi:10.1158/1538-7445.AM2015-3111


Experimental and Molecular Medicine | 2018

T-cell immune regulator 1 enhances metastasis in hepatocellular carcinoma

Hee Doo Yang; Jung Woo Eun; K.-W. Lee; Qingyu Shen; HyungSeok Kim; Sang Yean Kim; Dong-Wan Seo; Won Park; Jung Young Lee; Suk Woo Nam

Recurrence and metastasis are major challenges in the management of hepatocellular carcinoma (HCC) patients after resection. To identify a metastasis-associated gene signature, we performed comparative gene expression analysis with recurrent HCC tissues from HCC patients who underwent partial or total hepatectomy and from non-metastatic primary HCC tissues. From this, we were able to identify genes associated with HCC recurrence. TCIRG1 (T-Cell Immune Regulator 1) was one of the aberrantly overexpressed genes in patients with recurrent HCC who had undergone total hepatectomy. The significant overexpression of TCIRG1 was confirmed using the Liver Hepatocellular Carcinoma dataset from The Cancer Genome Atlas. High expression of TCIRG1 was significantly associated with poor 5-year disease-free and recurrence-free survival of HCC patients. TCIRG1 knockdown suppressed tumor cell growth and proliferation in HCC cell lines; caused a significant increase in the proportion of cells in the G1/S phase of cell cycle; induced cell death; suppressed the metastatic potential of HCC cells by selectively regulating the epithelial–mesenchymal transition (EMT) regulatory proteins E-cadherin, N-cadherin, Fibronectin, Snail and Slug; and significantly attenuated the metastatic potential of ras-transformed NIH-3T3 cells in vitro and in vivo. These findings suggest that TCIRG1 functions as a metastatic enhancer by modulating growth, death and EMT in HCC cells. TCIRG1 could be a therapeutic target for the treatment of liver malignancy and metastasis.


Cancer Research | 2018

Abstract 522: Histone deacetylase 6-let-7i-5p-thrombospondin-1 signaling axis suppresses CD47-dependent tumorigenic behavior of liver cancer

Hee Doo Yang; Jung Woo Eun; Qingyu Shen; Hyung Seok Kim; Sang Yean Kim; Suk Woo Nam

Histone deacetylase 6 (HDAC6) uniquely endows as tumor suppressor in liver tumorigenesis, but the underlying mechanisms leading to liver cancer are not fully understood. To identify the large-scale microRNA (miRNA) expression changes by HDAC6, we performed miRNA expression analysis in HDAC6-overexpressing Hep3B cells. From this, let-7i-5p was suggested as most highly associated miRNA with HDAC6 in liver cancer cells. We also found that let-7i-5p expression was up-regulated in a large cohort of HCC patients, and high expression of let-7i-5p was significantly associated with poor prognosis of HCC patients. Ectopic expression of antisense let-7i-5p (AS-let-7i-5p) caused cancer cell growth inhibition mediated by caspase-dependent cell death processing, and inhibited epithelial-mesenchymal transition (EMT) of liver cancer cells. Computational analysis suggested that thrombospondin-1 (TSP-1) is a specific target for let-7i-5p in liver cancer cells. Increased secretion of TSP1 was detected in AS-let-7i-5p transfected HCC cells. We found that partially purified TSP-1 from culture media (ppTSP-1) induced cellular death processing and simultaneously inhibited tumor angiogenesis and metastatic behavior of liver cancer cells. In addition, we observed that anti-TSP-1 antibody containing CD47 receptor binding motif (3F352) attenuated HDAC6, AS-let-7i-5p and TSP-1 effects on liver cancer cells. Furthermore, TSP-1 and ppTSP-1 treatments significantly enhanced macrophage phagocytosis through blocking CD47-dependent “Don9t eat me” signal in liver cancer cells. Our findings suggest that HDAC6-let-7i-5p-TSP-1 signaling axis regulates CD47-dependent neoplastic and phagocytic behaviors of hepatocytes providing a novel target for the molecular treatment of liver malignancies. Citation Format: Hee Doo Yang, Jung Woo Eun, Qingyu Shen, Hyung Seok Kim, Sang Yean Kim, Suk Woo Nam. Histone deacetylase 6-let-7i-5p-thrombospondin-1 signaling axis suppresses CD47-dependent tumorigenic behavior of liver cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 522.


Oncotarget | 2017

Identification of novel biomarkers for prediction of neurological prognosis following cardiac arrest

Jung Woo Eun; Hee Doo Yang; Soo Hyun Kim; Sungyoup Hong; Kyu Nam Park; Suk Woo Nam; Sikyoung Jeong

Background Early prognostication of neurological outcome in comatose patients after cardiac arrest (CA) is important for devising patient treatment strategies. However, there is still a lack of sensitive and specific biomarkers for easy identification of these patients. We evaluated whether molecular signatures from blood of CA patients might help to improve the prediction of neurological outcome. Methods We examined 22 comatose patients resuscitated after CA and obtained peripheral blood samples 48 hours after CA. To identify novel blood biomarkers, we aimed to measure neurological outcomes according to the Cerebral Performance Category (CPC) score at 6 months after CA and to determine blood transcriptome-based molecular signature of poor neurological outcome group. Results According to the CPC score, 10 patients exhibited a CPC score of one and 12 patients, a CPC score four to five. Blood transcriptomics revealed differently expressed profiles between the good outcome group and poor outcome group. A total of 150 genes were down-regulated and 237 genes were up-regulated in the poor neurological outcome group compared with good outcome group. From the blood transcriptome-based signatures, we identified that MAPK3, BCL2 and AKT1 were more specific and sensitive diagnostic biomarkers in poor neurological outcome with an area under the curve of 0.867 (p<0.0001), 0.800 (p=0.003), and 0.767 (p=0.016) respectively. Conclusions We identify three biomarkers as potential predictors of neurological outcome following CA. Further assessment of the prognostic value of transcriptomic analysis in larger cohorts of CA patients is needed.


Molecular & Cellular Toxicology | 2017

Identification of aberrant overexpression of long non-coding RNA MALAT1 and role as a regulatory microRNA in liver cancer

Woo Chan Shin; Jung Woo Eun; Qingyu Shen; Hyung Seok Kim; Hee Doo Yang; Sang Yean Kim; Young-Min Ahn; Won Sang Park; Jung Young Lee; Suk Woo Nam

MALAT1 is deregulated in various cancers. The underlying mechanisms of MALA T1-mediated tumorigenesis are unclear. We found that MALAT1 was highly overexpressed and its overexpression was significantly associated with poor prognosis of liver cancer patients analyzed from the TCGA Liver Hepatocellular Carcinoma and Gene Expression Omnibus databases of the National Center for Biotechnology Information. Microarray analysis to identify the miRNAs deregulated by silencing MALAT1 in two liver cancer cell lines, HepG2 and Hep3B, revealed the common deregulation of 16 miRNAs including miR-574 and miR-20b in both cell lines. The predicted targets of miR-574 and miR-20b were cancer-related pathways including the RAS, MAPK and Wnt/β-catenin pathways. Aberrant expression of MALAT1 might contribute to liver cancer tumorigenesis by deregulation of cancer-associated miRNAs.

Collaboration


Dive into the Hee Doo Yang's collaboration.

Top Co-Authors

Avatar

Jung Woo Eun

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Suk Woo Nam

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Qingyu Shen

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Hyung Seok Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Woo Chan Shin

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Jung Young Lee

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Sang Yean Kim

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Won Sang Park

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Hyun Jin Bae

Catholic University of Korea

View shared research outputs
Top Co-Authors

Avatar

Se Jin Park

Catholic University of Korea

View shared research outputs
Researchain Logo
Decentralizing Knowledge