Network


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

Hotspot


Dive into the research topics where Ji-Hee Lee is active.

Publication


Featured researches published by Ji-Hee Lee.


Cancer Research | 2017

Abstract P4-12-11: Up-regulation of SPARC is associated with breast tumor progression and epithelial SPARC expression is correlated with poor survival and MMP-2 expression in patients with breast carcinoma

Ji-Hee Lee; G-E Kim; MinHo Park; JungHan Yoon

Background: Secreted protein acidic and rich in cysteine (SPARC) plays a crucial role in the process of tumor invasion and metastasis in many cancers. Matrix metalloproteinases (MMPs) degrade the extracellular matrix and participate in several key processes of invasion and metastasis. The aims of this study were to evaluate the potential involvement of SPARC in the progression of breast tumor and to determine its association with outcome variables and MMPs expression in patients with breast carcinoma (BC). Materials and Methods: SPARC expression was examined in 8 pairs of BC tissues and surrounding normal tissues at mRNA and protein levels by quantitative real-time PCR (qRT-PCR), RNAscope in situ hybridization (ISH), Western blotting, and immunohistochemistry techniques. Immunohistochemical staining of SPARC on tissue microarray was done in 26 normal breasts, 76 ductal carcinoma in situ (DCIS), and 198 BC samples. In addition, we performed immunohistochemical staining for MMP-2 and MMP-9 in BC. Results: SPARC expression at mRNA and protein levels by qRT-PCR and Western blotting was significantly increased in BC tissues compared to the surrounding normal tissues (p Conclusion: Our results suggest that up-regulation of SPARC contributes to breast tumor progression. SPARC expression may be a useful biomarker for the prognostic prediction in patients with BC. SPARC can control extracellular matrix degradation through up-regulation of MMP-2. Citation Format: Lee JS, Kim G-E, Park MH, Yoon JH. Up-regulation of SPARC is associated with breast tumor progression and epithelial SPARC expression is correlated with poor survival and MMP-2 expression in patients with breast carcinoma [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P4-12-11.


Cancer Letters | 2018

TAp73 inhibits cell invasion and migration by directly activating KAI1 expression in colorectal carcinoma

Woo-Kyun Bae; Chang-Soo Hong; Mi-Ra Park; Eun‐Gene Sun; Ji-Hee Lee; Keunsoo Kang; Kyung-Hyun Ryu; Hyun-Jeong Shim; Jun-Eul Hwang; Sang-Hee Cho; Ik-Joo Chung

p73 is a member of the p53 family of transcription factors and, like p53, plays a role as a tumor suppressor. p73 is involved in development, proliferation, apoptosis and metastasis. However, the precise molecular mechanisms underlying its function in inhibiting metastasis remain largely unknown. Here, we show that induction of TAp73 decreased invasion and migration activity of colorectal cancer cells, whereas knockdown of TAp73 led to increased invasion and migration activity. KAI1 was identified as a transcriptional target of TAp73 and its expression is indispensable for TAp73-mediated inhibition of cell invasion and migration. Furthermore, induction of TAp73 in colorectal cancer cells elevated KAI1 expression and decreased the frequency of hepatic metastasis inxa0vivo. Whereas, the decreased invasion and migration activities caused by TAp73 induction were abrogated by knockdown of KAI1. Interestingly, TAp73 and KAI1 are overexpressed in primary colorectal cancers and a significant correlation between TAp73 and KAI1 expression was detected, but their expressions were significantly down-regulated in metastatic cancers. Taken together, our results support a novel role for TAp73 in controlling colorectal cancer cell invasion, migration and metastasis by regulating transcription of KAI1.


Cancer Research | 2016

Abstract 2834: Antitumor effect of AGM 130 (5’-OH-5-nitro-Indirubin oxime), a cyclin-dependent kinase inhibitor in colorectal cancer cells

Hyun-Jeong Shim; Jun-Eul Hwang; Woo-Kyun Bae; Myung-Sook Park; Hyo-Jeong Yun; Ji-Hee Lee; Sang-Hee Cho; Ik-Joo Chung

The AGM 130 (5’-OH-5-nitro-Indirubin oxime) compound is derived from Indirubin, which is an ingredient of Danggui Longhui Wan and used in traditional Chinese medicine. AGM 130 can inhibit the proliferation of a variety cells by arresting the cell in the G2/M phase of the cell cycle, it is a cyclin-dependent kinase (CDK) inhibitor that has anti-proliferative activity and apoptotic effects on cancer cells. AGM 130 has improved water solubility compared to Indirubin, however still displays a low solubility in biologic fluids and a poor bioavailability. This poor water solubility limits the clinical application of AGM 130 to treat cancer. In this study, we decided to develop and evaluate a SNEDDS (self-nanoemulsifying drug delivery systems) formulation containing AGM130 for improving its solubility, dissolution rate, and bioavailability. To check the anti-proliferative effect of the AGM 130 compound on colon cancer cell line, we performed the MTT assay with the CT-26 cancer cell. We also performed cell cycle analysis, apoptotic assays and western blotting. The CT-26 cell bearing mice were treated intraperitoneally with AGM 130 loaded PPG-cRGD, 5-fluorouracil (5-FU) or AGM 130 loaded PPG-cRGD combined with 5-FU. We measured tumor volume and tumor progression of each groups. In vitro, AGM 130 showed significant anti-proliferative activity in CT-26 cell (IC 50, 1.9 μM). Cells treated with AGM 130 showed G2/M cell cycle arrest and also induced apoptosis compared to untreated cells. The western blotting analysis showed that AGM 130 inhibited CKD1/cyclin B1 and also CKD2/cyclin E. The greatest tumor regression (percentage tumor growth inhibition) was observed in the group treated with AGM 130 loaded PPG-cRGD combined with 5-FU. When 5-FU was administrated alone, the anti-tumor effect was inferior to the group treated with AGM 130 loaded PPG-cRGD alone. These results suggest that intraperitoneal administration of AGM 130 loaded PPG-cRGD inhibits tumor progression in a mouse colon cancer. Thus, AGM 130, a cyclin-dependent kinase inhibitors may provide a new therapeutic approach in the treatment of colon cancer. Citation Format: Hyun-Jeong Shim, Jun-Eul Hwang, Woo-Kyun Bae, Myung-Sook Park, Hyo-Jeong Yun, Ji-Hee Lee, Sang-Hee Cho, Ik-Joo Chung. Antitumor effect of AGM 130 (5’-OH-5-nitro-Indirubin oxime), a cyclin-dependent kinase inhibitor in colorectal cancer cells. [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 2834.


Cancer Research | 2016

Abstract P2-07-06: Periostin is associated with breast tumor progression and epithelial periostin expression is correlated with poor survival in patients with invasive breast carcinoma

Ji-Hee Lee; G-E Kim; MinHo Park; JungHan Yoon

Background: Invasion and metastasis are the direct causes of mortality in patients with breast cancer and require reciprocal interactions between cancer cells and the extracellular matrix (ECM). Periostin, a fasciclin-containing adhesive ECM glycoprotein, is frequently overexpressed in various types of human cancer, and its overexpression in cancer-associated stroma and/or cancer cells is usually associated with poor clinical outcomes. However, the expression of periostin in the successive steps of breast tumorigenesis and its association with outcome variables have not been well established in breast carcinoma. The purposes of the present study were to assess the role of periostin alteration in breast tumorigenesis and evaluate the putative prognostic value of periostin as a function of its compartmentalization. Materials and Methods: Immunohistochemical staining with anti-periostin antibody was performed in a total of 300 patients (26 patients with normal breasts, 76 patients with ductal carcinoma in situ [DCIS], and 198 patients with invasive breast carcinoma [IBC]) using tissue microarray. Periostin immunoreactivity was assessed in both epithelial tissue and the surrounding stromal compartment. In addition, the mRNA and protein expression of periostin was analyzed in 10 paired normal/invasive cancer frozen specimens by quantitative real time-polymerase chain reaction and western blot analysis, respectively. Results: Periostin mRNA and protein expression in cancer tissues was increased compared with that in adjacent normal tissues. Both epithelial and stromal periostin staining scores were significantly increased with disease progression in a stepwise manner to DCIS and IBC compared with those in normal breast tissues (P = 0.000 and 0.000, respectively). High epithelial and stromal periostin expression was observed in 109 of 189 (57.7%) and 158 of 189 (83.6%) cases of IBC, respectively. High epithelial periostin expression was more frequently observed in the distant metastatic relapse-positive group than in the distant metastatic relapse-negative group (41 [80.4%] of 51 cases versus 68 [49.3%] of 138 cases [P = 0.000]). Furthermore, high epithelial periostin expression was associated with reduced disease-free and overall survival on univariate and multivariate analyses. Conclusion: Periostin might play an important role in the progression of breast tumor, and epithelial periostin expression may serve as a new parameter for prognostic prediction in patients with IBC. Further study of periostin expression and its potential as a target of therapy for IBC appears warranted. Citation Format: Lee JS, Kim G-E, Park MH, Yoon JH. Periostin is associated with breast tumor progression and epithelial periostin expression is correlated with poor survival in patients with invasive breast carcinoma. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-07-06.


Cancer Research | 2014

Abstract 3658: 90K attenuates the development of colitis-associated colorectal tumors through negative regulation of intestinal epithelial Toll-like receptor 4

Ik-Joo Chung; Jun-Eul Hwang; Ji-Hee Lee

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CAnnBackground:nnInflammation is a risk factor for colon cancer. TLR4 expression is upregulated in almost all colitis-associated cancer and dysplasia. The molecular mechanisms linking inflammation and colon carcinogenesis are incompletely understood. We found that tumor associated antigen 90K antagonized TLR4/NF-kB signaling pathway in colon cancer cells. We hypothesized that 90K is involved in TLR4 expression in colitis-induced colon carcinogenesis.nnMethods:nnThe effect of 90K against TLR4 was analysed by cell cultures, NF-kB reporter assay, and immunoprecipitation. C57BL/6 mice were treated with 3% dextran sodium sulfate (DSS) for acute colitis model. Colitis was assessed by disease activity index (DAI) including weight loss, stool consistency and rectal bleeding, and histopathology.nnResults:nn90K interacts with TLR4, suppresses the NF-kB signal in the MYD88-dependent pathway that is major adaptor molecules in TLRs/NF-kB signal. Adenoviral 90K administration attenuated the severity of acute DSS-induced colitis as assessed by DAI and histopathologic scoring compared with the control group. 90K restored body weight loss and the severity of acute DSS colitis through the suppression of TLR4/MYD88/NF-kB signal.nnConclusions:nnTumor associated antigen 90K is involved in negative regulation of inflammatory signaling triggered by TLR4-induced NF-kB activation. The data suggest that 90K might thereby suppress proliferation and progression of colon cancers.nnCitation Format: Ik-Joo Chung, Jun-Eul Hwang, Ji-Hee Lee. 90K attenuates the development of colitis-associated colorectal tumors through negative regulation of intestinal epithelial Toll-like receptor 4. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3658. doi:10.1158/1538-7445.AM2014-3658


Cancer Research | 2013

Abstract P2-10-03: Metastasis-associated in colon cancer 1 predicts poor outcomes in patients with breast cancer

Ji-Hee Lee; JinSeong Cho; MinHo Park; JungHan Yoon

Background: Metastasis-associated in colon cancer 1 (MACC1) plays a role in cancer invasion and metastasis through the upregulation of hepatocyte growth factor/mesenchymal epithelial transition-pathway, which is a known critical regulator of the mitogen-activated protein kinase (MAPK) cascades. However, the potential involvement of MACC1 in breast cancer has not been assessed. Materials and Methods: To investigate whether MACC1 expression is a prognostic marker in breast cancer, we performed immunohistochemical staining for MACC1 expression in tissue microarrays consisting of 198 invasive breast carcinomas. To demonstrate the potential correlation between MACC1 and MAPK cascades, phospho-p44/42 MAPK (ERK1/2) expression was also analyzed. Results: Expression of MACC1 was detected in 109 (55.1%) of 198 invasive breast carcinomas. MACC1 expression was significantly associated with several clinicopathologic parameters, including ER negativity (P Conclusion: Our results suggest that MACC1 may serve as a new parameter for the prognostic prediction in patients with invasive breast carcinoma. MACC1 is likely to be involved in the regulation of MAPK cascades in invasive breast carcinoma. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P2-10-03.


Cancer Research | 2011

Abstract 2221: DNA methylation of microRNA genes in gastric carcinoma and its clinicopathological association

Jin-Sook Kim; Ga-Eon Kim; Ji-Hee Lee; Jun-Eul Hwang; Woo-Kyun Bae; Jae-Hyuk Lee

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FLnnBackground: Epigenetic silencing of tumor-related genes, due to CpG island methylation, is considered to be an important mechanism for the development of many tumors, including gastric carcinoma (GC). DNA methylation of multiple CpG sites in promoter region of several tumor-related genes, such as hMLH1, p14, p16, and APC has been analyzed in GC so far. MicroRNAs (miRNA) are small, non-coding RNAs that can contribute to cancer development and progression by acting as oncogenes or tumor suppressor genes. Recent studies indicate that epigenetic silencing of miRNAs with tumor suppressor activity by CpG island methylation plays an important role in gastric carcinogenesis. To explore the role of epigenetic mechanisms in the down-regulation of miRNA genes (has-miR-9, has-miR-129, has-miR-137), we examined the presence of DNA methylation-associated silencing of miRNAs in GC and observed that aberrant methylation of these miRNAs is associated with expression of target gene products. Materials and Methods: The extent of promoter methylation of 4 miRNA genes (has-miR-9-1, has-miR-9-3, has-miR-129-2, has-miR-137) was assessed using methylation-specific polymerase chain reaction (MSP) in 100 GC tissues (50 early gastric cancers and 50 advanced gastric cancers) and corresponding non-tumor tissues. Twenty patients with benign gastric pathology were also included as a control group. The potential target gene products of miRNAs were studied by immunohistochemistry and the relationship between methylation profile of miRNAs promoter and clinicopathological parameters was analyzed. Results: Methylation of the has-miR-9-3 and has-miR-137 CpG island was frequently observed in tumor tissues (89% and 86%, respectively) and non-tumor tissues in 100 GC patients (70% and 78%), but not in normal gastric tissues (55% and 55%). Methylation of the has-miR-9-1 is slightly higher in tumor tissues (65%) than normal gastric tissue (45%). However, methylation level of the has-miR-129-2 was not shown significant difference in tumor tissues (97%) compared with non-tumor tissues (90%) and normal gastric tissues (90%). Expression of NF-κB and SOX4 protein, which are has-miR-9 and has-miR-129-2 potential target respectively, were inversely correlated with methylation level of miRNAs. The presence of H. pylori infection was significantly associated with aberrant methylation of miRNA genes (p<0.05). But, there was no significant correlation between aberrant methylation of miRNAs and other clinicopathological factors. Conclusion: These results suggest that specific miRNAs methylation in GC could be an important molecular mechanism causing loss of control on its target, and it may be correlated with the high transcriptional activity of target gene. Epigenetic silencing of some miRNAs may involve in the early stage of gastric carcinogenesis and could be used as an efficient diagnostic biomarker.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2221. doi:10.1158/1538-7445.AM2011-2221


Cancer Research | 2011

Abstract 4252: The addition of bevacizumab to paclitaxel overcomes chemoresistance to paclitaxel in a class III beta tubulin expressing gastric cancer cells

Jun-Eul Hwang; Woo-Kyun Bae; Jae-Hyuk Lee; Ji-Hee Lee; Ju-Young Yoon; Hyun-Jeong Shim; Sang-Hee Cho; Ik-Joo Chung

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FLnnBackground: Class III β tubulin (TUBB3) reduces microtubule stability and confers resistance to microtubule stabilizing taxanes, including paclitaxel and docetaxel. Expression of vascular endothelial growth factor (VEGF) is highly regulated by hypoxia and the regulation of VEGF expression by hypoxia is mediated by a hypoxia inducible factor-1a(HIF-1a), which increase transcription of the VEGF gene. Recent report showed hypoxia induces expression of TUBB3 by HIF-1a. Inhibition of VEGF system may decrease the expression of HIF-1a and TUBB3. This study was undertaken to investigate the synergistic antitumor effects of bevacizumab and palitaxel on TUBB3 expressing gastric cancer cells.nnMethods: We evaluated the expressions of TUBB3 and HIF-1a under normoxic and hypoxic conditions in human gastric cancer cells. The chemoresistance of TUBB3 to paclitaxel was checked through knockdown of TUBB3. Expressions of TUBB3 and HIF-1a were measured under blocking of VEGFR1/2 with anti-FLT1 and anti-KDR antibodies. The effects of bevacizumab and paclitaxel on expressions of TUBB3 and HIF1a were monitored by western blot and MTT assay.nnResults: The chemosensitivities to paclitaxel were increased by siTUBB3. Elevated expressions of TUBB3 and HIF-1a in gastric cancer cells under hypoxic conditions were reduced in the prescence of anti-KDR and anti-FLT1 antibodies. When we added the bevacizumab to paclitaxel, the protein expressions of TUBB3 and HIF-1a in gastric cancer cells under hypoxic conditions were significantly decreased as compared with paclitaxel alone. Apoptosis of gastric cancer cells were more increased in a combined treatment with bevacizumab and paclitaxel than paclitaxel alone. Tyrosine phosphorylation of Erk and Akt was also measured in gastric cancer cells under hypoxic conditions in the absence or prescence of anti-KDR, anti FLT-1 and paclitaxels. Hypoxia induced phosphorylation of Erk and Akt in gastric cancer cells was more decreased by addition of anti FLT-1 and anti KDR antibodies to paclitaxel as compared with paclitaxel alone.nnConclusion: Our experiments show that a VEGF/VEGFR/pERK and, or pAKT pathway is activated by hypoxic gastric cancer cells to induce HIF-1a and TUBB3 expressions. Combined treatment with paclitaxel and bevacizumab, a recombinant humanized monoclonal antibody that binds to VEGF, synergistically decreased the expressions of HIF-1a and TUBB3 in gastric cancer cells in hypoxic conditions. With increasing use of taxanes in the treatment of gastric cancer, this study showed the potential synergistic antitumor effects of bevacizumab and paclitaxel in a TUBB3, a marker of resistance to paclitaxel, overexpressing gastric cancer.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4252. doi:10.1158/1538-7445.AM2011-4252


Cancer Research | 2011

Abstract 4810: Utility of promoter hypermethylation for differentiating malignant and benign effusions in liquid-based cytology specimens

Ga-Eon Kim; Ji-Shin Lee; Jin-Sook Kim; Woo-Kyun Bae; Ji-Hee Lee; Jun-Eul Hwang; Jae-Hyuk Lee

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FLnnBackground : The cytologic differentiation between benign and malignant effusions can be difficult. Because promoter hypermethylation of tumor suppressor genes is a frequent epigenetic event in many human cancers, it could serve as a marker for the diagnosis of cancer. The aim of this study was to investigate the feasibility of promoter hypermethylation as a diagnostic tool for the differentiation between malignant and benign effusions in liquid-based cytology samples. Methods : A multiplex, nested, methylation-specific polymerase chain reaction analysis was used to examine promoter methylation of 4 genes (RAR-β, APC, Twist and HIN-1) in malignant (n=85) and benign (n=31) liquid-based cytology samples. Results : The frequencies of hypermethylation of RAR-β, APC, Twist and HIN-1 were significantly higher in malignant effusions than in benign effusions (p<0.001 for each). In receiver-operating characteristic analysis, the area under the curve (AUC) for APC was the highest. The AUC for the best two-gene combination (APC/HIN-1) was not statistically different from the best individual tumor suppressor gene (APC). Conclusions : This study suggests that promoter methylation analysis on residual liquid-based effusion samples may be a feasible approach to detect malignant effusions, and that APC is the best marker for differentiating malignant and benign effusions.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4810. doi:10.1158/1538-7445.AM2011-4810


Cancer Research | 2011

P2-11-03: High Expression of CX3CL1 by Tumor Cells Correlates with a Good Prognosis and Increased Infiltrating CD8+ T Cells, Natural Killer Cells, and Dendritic Cells in Breast Carcinoma.

Ji-Hee Lee; Min Ho Park; JungHan Yoon

CX3CL1 is the only CX3C chemokine that can chemoattract CD8+ T cells, natural killer (NK) cells, and dendritic cells (DCs). Although studies have reported that CX3CL1 regulates the host immune response, the roles of the CX3CL1 in breast carcinoma remain unknown. We evaluated the expression CX3CL1 in 198 surgical specimens of breast carcinoma and analyzed any association with the clinicopathological factors, including CD8+ T cells, CD56+ NK cells, and CD1a+ DCs, while also evaluating the impact on the prognosis. High CX3CL1 expression was found in 136 of 198 (68.7%) breast carcinomas. The numbers of stromal CD8+ T cells, intratumoral CD1a+ DCs, and stromal CD56+ NK cells were significantly increased in the high CX3CL1 expression group (19.3 ± 15.2, 0.3 ± 0.9 and 1.4 ± 1.7 per field, respectively) compared with the low expression group (10.2 ± 13.1, 0.0 ± 0.3 and 0.7 ± 0.8 per field, respectively). Patients with high CX3CL1 expression had a significantly better prognosis for disease-free and overall survival than those with low expression ( P = 0.002 and P = 0.000, respectively). In a multivariate analysis, the CX3CL1 expression was identified as one of the independent prognostic factors for overall survival (odds ratio, 2.29; P = 0.002). These data suggest that CX3CL1 expressed in the tumor cells appears to enhance the recruitment of CD8+ T cells, NK cells, and CD1a+ DCs, thereby bringing about a better prognosis in breast carcinoma. CX3CL1 is a new prognostic biomarker and be a novel candidate for development of a more effective therapeutic strategy for breast carcinoma. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P2-11-03.

Collaboration


Dive into the Ji-Hee Lee's collaboration.

Top Co-Authors

Avatar

Jun-Eul Hwang

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Ik-Joo Chung

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Woo-Kyun Bae

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

JungHan Yoon

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Hyun-Jeong Shim

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Hyuk Lee

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

MinHo Park

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Sang-Hee Cho

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

G-E Kim

Chonnam National University

View shared research outputs
Top Co-Authors

Avatar

Ga-Eon Kim

Chonnam National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge