Network


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

Hotspot


Dive into the research topics where Jin Hyoung Cho is active.

Publication


Featured researches published by Jin Hyoung Cho.


Journal of Cellular Biochemistry | 2016

Comparative Analysis of Human Mesenchymal Stem Cells Derived From Bone Marrow, Placenta, and Adipose Tissue as Sources of Cell Therapy.

Young-Joo Jeon; Jumi Kim; Jin Hyoung Cho; Hyung-Min Chung; Jung-Il Chae

Various source‐derived mesenchymal stem cells (MSCs) with multipotent capabilities were considered for cell therapeutics of incurable diseases. The applicability of MSCs depends on the cellular source and on their different in vivo functions, despite having similar phenotypic and cytological characteristics. We characterized MSCs from different sources, including human bone marrow (BM), placenta (PL), and adipose tissue (AT), in terms of the phenotype, surface antigen expression, differentiation ability, proteome reference map, and blood flow recovery in a hindlimb ischemic disease model. The MSCs exhibit different differentiation potentials depending on the cellular source despite having similar phenotypic and surface antigen expression. We identified approximately 90 differentially regulated proteins. Most up‐ or down‐regulated proteins show cytoskeletal or oxidative stress, peroxiredoxin, and apoptosis roles according to their functional involvement. In addition, the PL‐MSCs retained a higher therapeutic efficacy than the BM‐ and AT‐MSCs in the hindlimb ischemic disease model. In summary, we examined differentially expressed key regulatory factors for MSCs that were obtained from several cellular sources and demonstrated their differentially expressed proteome profiles. Our results indicate that primitive PL‐MSCs have biological advantages relative to those from other sources, making PL‐MSCs a useful model for clinical applications of cell therapy. J. Cell. Biochem. 117: 1112–1125, 2016.


International Journal of Oncology | 2014

Licochalcone A, a natural chalconoid isolated from Glycyrrhiza inflata root, induces apoptosis via Sp1 and Sp1 regulatory proteins in oral squamous cell carcinoma

Jung Jae Cho; Jung-Il Chae; Goo Yoon; Ka Hwi Kim; Jin Hyoung Cho; Seung-Sik Cho; Young Sik Cho; Jung-Hyun Shim

Licochalcone A (LCA), a chalconoid derived from root of Glycyrrhiza inflata, has been known to possess a wide range of biological functions such as antitumor, anti-angiogenesis, antiparasitic, anti-oxidant, antibacterial and anti-inflammatory effects. However, the anticancer effects of LCA on oral squamous cell carcinoma (OSCC) have not been reported. Our data showed that LCA inhibited OSCC cell (HN22 and HSC4) growth in a concentration- and time-dependent manner. Mechanistically, it was mediated via downregulation of specificity protein 1 (Sp1) expression and subsequent regulation of Sp1 downstream proteins such as p27, p21, cyclin D1, Mcl-1 and survivin. Here, we found that LCA caused apoptotic cell death in HSC4 and HN22 cells, as characterized by sub-G1 population, nuclear condensation, Annexin V staining, and multi-caspase activity and apoptotic regulatory proteins such as Bax, Bid, Bcl(-xl), caspase-3 and PARP. Consequently, this study strongly suggests that LCA induces apoptotic cell death of OSCC cells via downregulation of Sp1 expression, prompting its potential use for the treatment of human OSCC.


International Journal of Molecular Medicine | 2012

Role of transcription factor Sp1 in the quercetin-mediated inhibitory effect on human malignant pleural mesothelioma

Jung-Il Chae; Jin Hyoung Cho; Kyung-Ae Lee; Nag-Jin Choi; Kang Seok Seo; Sang-Bum Kim; Sang-Han Lee; Jung-Hyun Shim

Quercetin (Qu) is found in plants, including red onions and in the skins of red apples, and induces the apoptosis of certain malignant cells. However, no report has been issued on the apoptotic effect of Qu on human malignant pleural mesothelioma. In the present study, it was found that MSTO-211H mesothelioma cell viability was reduced and apoptotic cell death was increased by Qu (20-80 µM), which was found to have an IC₅₀ of 58 µM. In addition, Qu increased the sub-G₁ cell population, and was found to interact with specificity protein 1 (Sp1) and significantly suppressed its expression at the protein and mRNA levels. Furthermore, Qu modulated the levels of Sp1 regulatory genes, such as cyclin D1, myeloid cell leukemia (Mcl)-1 and survivin in MSTO-211H cells. Apoptotic signaling cascades were activated by the cleavage of Bid, caspase-3 and PARP, and by the downregulation of Bcl-xL and the upregulation of Bax in MSTO-211H cells. Our results strongly suggest that Sp1 be considered as a novel molecular target of Qu in human malignant pleural mesothelioma.


Drug Development Research | 2015

Quercetin Induces Antiproliferative Activity Against Human Hepatocellular Carcinoma (HepG2) Cells by Suppressing Specificity Protein 1 (Sp1)

Ra Ham Lee; Jin Hyoung Cho; Young-Joo Jeon; Woong Bang; Jung-Jae Cho; Nag-Jin Choi; Kang Seok Seo; Jung-Hyun Shim; Jung-Il Chae

Preclinical Research


Journal of Cellular Biochemistry | 2016

Esculetin Induces Apoptosis Through EGFR/PI3K/Akt Signaling Pathway and Nucleophosmin Relocalization

Young-Joo Jeon; Jin Hyoung Cho; Seung-Yeop Lee; Yung Hyun Choi; Hong-Ju Park; Seunggon Jung; Jung-Hyun Shim; Jung-Il Chae

Esculetin, a coumarin compound, has anti‐proliferative effects on various types of human cancer cells, but its effect on oral squamous cell carcinoma (OSCC) is unknown. In this study, we determined whether esculetin had anti‐proliferative effects on two oral squamous cell lines, HN22, and HSC2. We found that esculetin inhibited cell viability by inducing apoptosis, as evinced by apoptotic cell morphologies, nuclear fragmentation, and the multi‐caspase/MMP activity. Furthermore, proteomic analysis was used to identify the target‐specific proteins involved in esculetin treatment. Intriguingly, apoptotic cell death by esculetin was associated with significant inhibition of the EGFR/PI3K/Akt signaling pathway. We also demonstrated that the expression of nucleophosmin (NPM) markedly decreased after esculetin treatment, and relocalization of NPM from the nucleous to the cytoplasm, together with p65, potentiated apoptotic stimulation. Additionally, our data indicated that NPM expression was markedly higher in OSCC tissues than in normal tissues. Our results collectively indicated that esculetin inhibited the proliferation of OSCC through EGFR‐mediated signaling pathways and down‐regulation of NPM as well as the perturbation of NPM trafficking from the nucleolus to the cytoplasm resulted in apoptosis. J. Cell. Biochem. 117: 1210–1221, 2016.


International Journal of Oncology | 2015

Licochalcone A induces apoptosis in malignant pleural mesothelioma through downregulation of Sp1 and subsequent activation of mitochondria-related apoptotic pathway

Ka Hwi Kim; Goo Yoon; Jung Jae Cho; Jin Hyoung Cho; Young Sik Cho; Jung-Il Chae; Jung-Hyun Shim

Licochalcone A (LCA) is a natural product derived from the roots of Glycyrrhiza inflata exhibiting a wide range of bioactivities such as antitumor, anti-oxidant and anti-bacterial effects. Malignant pleural mesothelioma (MPM) is an extremely aggressive type of cancer with a poor prognosis because of its rapid progression. However, LCA has not been investigated concerning its effects on MPM. Preliminarily, we observed that LCA negatively modulated not only cell growth, but also specificity protein 1 (Sp1) expression in MSTO-211H and H28 cell lines. It was found that IC50 values of LCA for growth inhibition of MSTO-211H and H28 cells were approximately 26 and 30 µM, respectively. Consistent with downregulation of Sp1, expression of Sp1 regulatory proteins such as Cyclin D1, Mcl-1 and Survivin was substantially diminished. Mechanistically, LCA triggered the mitochondrial apoptotic pathway by affecting the ratio of mitochondrial proapoptotic Bax to anti-apoptotic Bcl-xL. Bid induced loss of mitochondrial membrane potential, eventually leading to multi-caspase activation and increased sub-G1 population. Moreover, nuclear staining with DAPI highlighted nuclear condensation and fragmentation of apoptotic features. Flow cytometry analyses after staining cells with Annexin V and propiodium iodide corroborated LCA-mediated apoptotic cell death of MPM cells. In conclusion, these results present that LCA may be a potential bioactive material to control human MPM cells by apoptosis via the downregulation of Sp1.


Scientific Reports | 2017

Antitumorigenic effect of atmospheric-pressure dielectric barrier discharge on human colorectal cancer cells via regulation of Sp1 transcription factor

Duksun Han; Jin Hyoung Cho; Ra Ham Lee; Woong Bang; Kyungho Park; Min-Seok Kim; Jung-Hyun Shim; Jung-Il Chae; Se Youn Moon

Human colorectal cancer cell lines (HT29 and HCT116) were exposed to dielectric barrier discharge (DBD) plasma at atmospheric pressure to investigate the anticancer capacity of the plasma. The dose- and time-dependent effects of DBDP on cell viability, regulation of transcription factor Sp1, cell-cycle analysis, and colony formation were investigated by means of MTS assay, DAPI staining, propidium iodide staining, annexin V–FITC staining, Western blot analysis, RT-PCR analysis, fluorescence microscopy, and anchorage-independent cell transformation assay. By increasing the duration of plasma dose times, significant reductions in the levels of both Sp1 protein and Sp1 mRNA were observed in both cell lines. Also, expression of negative regulators related to the cell cycle (such as p53, p21, and p27) was increased and of the positive regulator cyclin D1 was decreased, indicating that the plasma treatment led to apoptosis and cell-cycle arrest. In addition, the sizes and quantities of colony formation were significantly suppressed even though two cancer promoters, such as TPA and epidermal growth factor, accompanied the plasma treatment. Thus, plasma treatment inhibited cell viability and colony formation by suppressing Sp1, which induced apoptosis and cell-cycle arrest in these two human colorectal cancer cell lines.


The International Journal of Biochemistry & Cell Biology | 2015

Role of transcription factor Sp1 in the 4-O-methylhonokiol-mediated apoptotic effect on oral squamous cancer cells and xenograft

Jin Hyoung Cho; Ra Ham Lee; Young Joo Jeon; Jae Cheon Shin; Seon Min Park; Nag Jin Choi; Kang Seok Seo; Goo Yoon; Seung Sik Cho; Ka Hwi Kim; Jung Jae Cho; Young Sik Cho; Dae Hwan Kim; Jin Tae Hong; Tae-Hoon Lee; Hong Ju Park; Seunggon Jung; Jae-Min Seo; Hanyong Chen; Zigang Dong; Jung Il Chae; Jung Hyun Shim

Recently, biphenolic components derived from the Magnolia family have been studied for anti-cancer, anti-stress, and anti-inflammatory pharmacological effects. However, the pharmacological mechanism of action of 4-O-methylhonokiol (MH) is not clear in oral cancer. The aim of this study was to investigate the role of MH in apoptosis and its molecular mechanism in oral squamous cell carcinoma (OSCC) cell lines, HN22 and HSC4, as well as tumor xenografts. Here, we demonstrated that MH decreased cell growth and induced apoptosis in HN22 and HSC4 cells through the regulation of specificity protein 1 (Sp1). We employed several experimental techniques such as MTS assay, DAPI staining, PI staining, Annexin-V/7-ADD staining, RT-PCR, western blot analysis, immunocytochemistry, immunohistochemistry, TUNEL assay and in vivo xenograft model analysis. MH inhibited Sp1 protein expression and reduced Sp1 protein levels via both proteasome-dependent protein degradation and inhibition of protein synthesis in HN22 and HSC4 cells; MH did not alter Sp1 mRNA levels. We found that MH directly binds Sp1 by Sepharose 4B pull-down assay and molecular modeling. In addition, treatment with MH or knocking down Sp1 expression suppressed oral cancer cell colony formation. Moreover, MH treatment effectively inhibited tumor growth and Sp1 levels in BALB/c nude mice bearing HN22 cell xenografts. These results indicated that MH inhibited cell growth, colony formation and also induced apoptosis via Sp1 suppression in OSCC cells and xenograft tumors. Thus, MH is a potent anti-cancer drug candidate for oral cancer.


International Journal of Molecular Medicine | 2012

Phosphoinositol 3-kinase, a novel target molecule for the inhibitory effects of juglone on TPA-induced cell transformation

Jung-Il Chae; Jin Hyoung Cho; Dong Joon Kim; Kyung-Ae Lee; Moon-Kyun Cho; Hae-Seon Nam; Kee-Min Woo; Sang-Han Lee; Jung-Hyun Shim

Juglone (5-hydroxy-1,4-naphthalenedione) from black walnut trees induces apoptosis and inhibits proliferation of various malignant cells. Here, we investigated whether juglone affects 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced cell transformation through the phosphoinositol 3-kinase (PI3K) pathway. The results showed that TPA- and endothelial growth factor (EGF)-induced anchorage-independent colony formation were suppressed in a dose-dependent manner by treatment of JB6 CI41 mouse skin epidermal cells with juglone (2.5 and 5 µM). We demonstrated that juglone suppressed PI3K activity via direct binding to PI3K by sepharose 4B pull-down assay and western blot analysis. Juglone significantly suppressed TPA-induced protein kinase B (AKT) and c-Jun phosphorylation and c-fos activation, but not mitogen-activated protein-kinase kinase (MEK), extracellular signaling-regulated kinase (ERK) or 90 kDa ribosomal protein S6 kinase (RSK) phosphorylation. Juglone significantly blocked activator protein-1 (AP-1) and cyclooxygenase-2 (COX-2) activation more than the PI3K inhibitors LY294002 and wortmannin. Overall, these results showed the anticancer efficacy of juglone targeting PI3K to prevent TPA-induced tumorigenesis.


International Journal of Oncology | 2016

Licochalcone B induces apoptosis of human oral squamous cell carcinoma through the extrinsic- and intrinsic-signaling pathways.

Ha-Na Oh; Goo Yoon; Jae-Cheon Shin; Seon-Min Park; Seung-Sik Cho; Jin Hyoung Cho; Mee-Hyun Lee; Kangdong Liu; Young Sik Cho; Jung-Il Chae; Jung-Hyun Shim

Licochalcone B (Lico B), which belongs to the retrochalcone family, is isolated from the roots of Chinese licorice. Lico B has been reported to have several other useful pharmacological properties, such as anti-inflammatory, antibacterial, antioxidant, antiulcer, anticancer, and anti-metastasis activities. We elucidated the underlying mechanism by which Lico B can induce apoptosis in oral squamous cell carcinoma (OSCC). Our results showed that exposure of OSCC cells (HN22 and HSC4) to Lico B significantly inhibited cell proliferation in a time- and concentration-dependent manner. Lico B caused cell cycle arrest at G1 phase along with downregulation of cyclin D1 and upregulation of p21 and p27 proteins. Lico B also facilitated the diffusion of phospholipid phosphatidylserine (PS) from inner to outer leaflets of the plasma membrane with chromatin condensation, DNA fragmentation, accumulated sub-G1 population in a concentration-dependent manner. Moreover, Lico B promoted the generation of reactive oxygen species (ROS), which, in turn, can induce CHOP, death receptor (DR) 4 and DR5. Lico B treatment induced downregulation of anti-apoptotic proteins (Bid and Bcl-xl and Mcl-1), and up-regulation of pro-apoptotic protein (Bax). Lico B also led to the loss of mitochondrial membrane potential (MMP), resulting in cytochrome c release. As can be expected from the above results, the apoptotic protease activating factor-1 (Apaf-1) and survivin were oppositely expressed in favor of apoptotic cell death. This notion was supported by the fact that Lico B activated multi-caspases with cleavage of poly (ADP-ribose) polymerase (PARP) protein. Therefore, it is suggested that Lico B is a promising drug for the treatment of human oral cancer via the induction of apoptotic cell death.

Collaboration


Dive into the Jin Hyoung Cho's collaboration.

Top Co-Authors

Avatar

Jung-Il Chae

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Jung-Hyun Shim

Mokpo National University

View shared research outputs
Top Co-Authors

Avatar

Young-Joo Jeon

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Ra Ham Lee

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Goo Yoon

Mokpo National University

View shared research outputs
Top Co-Authors

Avatar

Jae-Cheon Shin

Mokpo National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jae-Min Seo

Chonbuk National University

View shared research outputs
Top Co-Authors

Avatar

Ka Hwi Kim

Mokpo National University

View shared research outputs
Top Co-Authors

Avatar

Kang Seok Seo

Sunchon National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge