g Yong Jeon
Yonsei University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by g Yong Jeon.
British Journal of Pharmacology | 2014
Ki Cheong Park; Ji Hyun Park; Jeong Yong Jeon; Sang Yong Kim; Jung Min Kim; Chang Yong Lim; Tae Hyung Lee; Hyung Kwan Kim; Hyun Gyu Lee; Sung Min Kim; Ho Jeong Kwon; Jin Suck Suh; Seung Won Kim; Seung Hoon Choi
Activation of hepatic stellate cells (HSCs) is a crucial step in the pathogenesis of hepatic fibrosis. Histone deacetylase (HDAC) is an attractive target in liver fibrosis because it plays a key role in gene expression and cell differentiation. We have developed a HDAC inhibitor, N‐hydroxy‐7‐(2‐naphthylthio)heptanomide (HNHA), and investigated the anti‐fibrotic activity of HNHA in vitro and in vivo.
BMC Cancer | 2015
Seok Mo Kim; Ki Cheong Park; Jeong Yong Jeon; Bup Woo Kim; Hyeung Kyoo Kim; Ho Jin Chang; Seung Hoon Choi; Cheong Soo Park; Hang Seok Chang
BackgroundThyroid cancer has been indicated to have a higher global proportion of DNA methylation and a decreased level of histone acetylation. Previous studies showed that histone gene reviser and epigenetic changes role significant parts in papillary and anaplastic thyroid cancer tumorigenesis. The goal of this research was to study the endoplasmic reticulum (ER) stress-mediated actions of the dominant histone deacetylase (HDAC) inhibitor, N-hydroxy-7-(2-naphthylthio) hepatonomide (HNHA), in thyroid cancer and to explore its effects on apoptotic cell death pathways.MethodsExperiments were achieved to conclude the effects of HNHA in papillary thyroid cancer (PTC) and anaplastic thyroid cancer (ATC) cell lines and xenografts, as compared with two other established HDAC inhibitors (SAHA; suberoylanilide hydroxamic acid and TSA; trichostatin A).ResultsApoptosis, which was induced by all HDAC inhibitors, was particularly significant in HNHA-treated cells, where noticeable B-cell lymphoma-2 (Bcl-2) suppression and caspase activation were observed both in vitro and in vivo. HNHA increased Ca2+ release from the ER to the cytoplasm. ER stress-dependent apoptosis was induced by HNHA, suggesting that it induced caspase-dependent apoptotic cell death in PTC and ATC. PTC and ATC xenograft studies demonstrated that the antitumor and pro-apoptotic effects of HNHA were greater than those of the established HDAC inhibitors. These HNHA activities reflected its induction of caspase-dependent and ER stress-dependent apoptosis on thyroid cancer cells.ConclusionsThe present study indicated that HNHA possibly provide a new clinical approach to thyroid cancers, including ATC.
Oncotarget | 2016
Junjeong Choi; Ji Hyun Lee; Ilkyoo Koh; Jin Kyoung Shim; Junseong Park; Jeong Yong Jeon; Mijin Yun; Se Hoon Kim; Jong In Yook; Eui Hyun Kim; Jong Hee Chang; Sun Ho Kim; Yong Min Huh; Su Jae Lee; Michael Pollak; Pilnam Kim; Seok Gu Kang; Jae Ho Cheong
Studies have investigated biguanide-derived agents for the treatment of cancers and have reported their effects against tumorspheres (TSs). The purpose of this study was determining the effects of HL156A, a newly designed biguanide with improved pharmacokinetics, on glioblastoma TSs (GMB TSs) and assess the feasibility of this drug as a new line of therapy against glioblastoma, alone or combined with a conventional therapeutic agent, temozolomide(TMZ). The effects of HL156A, alone and combined with TMZ, on the stemness and invasive properties of GBM TSs and survival of orthotopic xenograft animals were assessed. HL156A, combined with TMZ, inhibited the stemness of GBM TSs, proven by neurosphere formation assay and marker expression. Three-dimensional collagen matrix invasion assays provided evidence that combined treatment inhibited invasive properties, compared with control and TMZ-alone treatment groups. TMZ alone and combined treatment repressed the expression of epithelial-mesenchymal transition-related genes. A gene ontology comparison of TMZ and combination-treatment groups revealed altered expression of genes encoding proteins involved in cellular adhesion and migration. Combined treatment with HL156A and TMZ showed survival benefits in an orthotopic xenograft mouse model. The inhibitory effect of combination treatment on the stemness and invasive properties of GBM TSs suggest the potential usage of this regimen as a novel strategy for the treatment of GBM.
Neuro-oncology | 2016
Eui Hyun Kim; Ji Hyun Lee; Yoonjee Oh; Ilkyoo Koh; Jin Kyoung Shim; Junseong Park; Junjeong Choi; Mijin Yun; Jeong Yong Jeon; Yong Min Huh; Jong Hee Chang; Sun Ho Kim; Kyung Sup Kim; Jae Ho Cheong; Pilnam Kim; Seok Gu Kang
Background Deprivation of tumor bioenergetics by inhibition of multiple energy pathways has been suggested as an effective therapeutic approach for various human tumors. However, this idea has not been evaluated in glioblastoma (GBM). We hypothesized that dual inhibition of glycolysis and oxidative phosphorylation could effectively suppress GBM tumorspheres (TS). Methods Effects of 2-deoxyglucose (2DG) and metformin, alone and in combination, on GBM-TS were evaluated. Viability, cellular energy metabolism status, stemness, invasive properties, and GBM-TS transcriptomes were examined. In vivo efficacy was tested in a mouse orthotopic xenograft model. Results GBM-TS viability was decreased by the combination of 2DG and metformin. ATP assay and PET showed that cellular energy metabolism was also decreased by this combination. Sphere formation, expression of stemness-related proteins, and invasive capacity of GBM-TS were also significantly suppressed by combined treatment with 2DG and metformin. A transcriptome analysis showed that the expression levels of stemness- and epithelial mesenchymal transition-related genes were also significantly downregulated by combination of 2DG and metformin. Combination treatment also prolonged survival of tumor-bearing mice and decreased invasiveness of GBM-TS. Conclusion The combination of 2DG and metformin effectively decreased the stemness and invasive properties of GBM-TS and showed a potential survival benefit in a mouse orthotopic xenograft model. Our findings suggest that targeting TS-forming cells by this dual inhibition of cellular bioenergetics warrants expedited clinical evaluation for the treatment of GBM.
Biochemical and Biophysical Research Communications | 2015
Jeong Yong Jeon; Hyangkyu Lee; Jeayeo Park; Misu Lee; Sae Whan Park; Ji Sook Kim; Milim Lee; Byoungchul Cho; Kyung-Sup Kim; Augustine M. K. Choi; Chun K. Kim; Mijin Yun
The glycolytic phenotype is a dominant metabolic phenomenon in cancer and is reflected in becoming aggressive. Certain hepatocellular carcinoma lack increased glycolysis and prefer to uptake acetate than glucose for metabolism. Autophagy plays a role in preserving energies and nutrients when there is limited external nutrient supply and maintains glucose level of blood though supporting gluconeogenesis in the liver. As the role of autophagy and gluconeogenesis in HCC following the glycolic activity was not clear, we cultured HCC cells with different glycolytic levels in Hanks balanced salt solution (HBSS) to induce autophagy and conducted the activity of gluconeogenesis. Both autophagy and gluconeogenesis were induced in low glycolytic HCC cells (HepG2). In glycolytic Hep3B cells, only autophagy without gluconeogenesis was induced upon starvation. When autophagy was blocked, the level of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) was reduced in HepG2 cells and not in Hep3B. Altogether, we investigated contribution of hepatic gluconeogenesis to the metabolic phenotype of HCC cells and the role of autophagy as a potential mechanism regulating gluconeogenesis in low glycolytic HCC.
BMC Cancer | 2015
Ki Cheong Park; Jun Hyeok Heo; Jeong Yong Jeon; Hye Ji Choi; A Ra Jo; Seung Won Kim; Ho Jeong Kwon; Sung Joon Hong; Kyung Seok Han
BackgroundEpigenetic modifications play a critical role in the regulation of all DNA-based processes, such as transcription, repair, and replication. Inappropriate histone modifications can result in dysregulation of cell growth, leading to neoplastic transformation and cell death. Renal tumors have been shown to have a higher global methylation percentage and reduced histone acetylation. Preclinical models have revealed that histone gene modifiers and epigenetic alterations play important roles in renal cell carcinoma (RCC) tumorigenesis. Recently, a novel HDAC inhibitor, N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), has been introduced as an example of a new class of anti-cancer agents. The anti-cancer activity of HNHA and the underlying mechanisms of action remain to be clarified.MethodsThe MTS assay using a panel of RCC cells was used to evaluate the anti-proliferative effects of HNHA. The established HDAC inhibitors, SAHA and TSA, were used for comparison. Western blotting analysis was performed to investigate the acetylation of histone H3 and the expression of apoptotic markers in vitro and in vivo. Subcellular fractionation was performed to evaluate expression of Bax and cytochrome c in the cytosol and mitochondria, and also translocation of cytochrome c from the cytoplasm to the nucleus. A confocal microscopic evaluation was performed to confirm inhibition of cell proliferation, induction of apoptosis, and the nuclear translocation of cytochrome c in RCC cells.ResultsIn this study, we investigated the apoptosis-inducing activity of HNHA in cultured kidney cancer cells. Apoptosis in the HNHA-treated group was induced significantly, with marked caspase activation and Bcl-2 suppression in RCC cells in vitro and in vivo. HNHA treatment caused cytochrome c release from mitochondria, which was mediated by increased Bax expression and caspase activation. HNHA also induced nuclear translocation of cytochrome c, suggesting that HNHA can induce caspase-independent nuclear apoptosis in RCC cells. An in vivo study showed that HNHA had greater anti-tumor and pro-apoptotic effects on RCC xenografts than the established HDAC inhibitors.ConclusionsHNHA has more potent anti-tumor activity than established HDAC inhibitors. Its activities are mediated by caspase-dependent and cytochrome-c-mediated apoptosis in RCC cells. These results suggest that HNHA may offer a new therapeutic approach to RCC.
Neoplasia | 2017
Ki Cheong Park; Seok-Mo Kim; Jeong Yong Jeon; Bup-Woo Kim; Hyeung Kyoo Kim; Ho Jin Chang; Yong Sang Lee; Sooyoung Kim; Seung Hoon Choi; Cheong Soo Park; Hang-Seok Chang
Anaplastic thyroid carcinoma (ATC) although rare is the most deadly form of thyroid cancer. The fatality rate for ATC is high-pitched, the survival rate at 1 year after diagnosis is <20%. Control of ATC is severely hard and widespread with unpredictability. We Previous proved that histone gene reviser and epigenetic changes role significant parts in papillary and anaplastic thyroid cancer tumorigenesis. Herein, the goal of this study was to investigate the anti-tumor activities of a HDAC inhibitor, HNHA alone and in combination with sorafenib in ATC cells in vitro and in vivo and to explore its effects on apoptotic cell death pathways. Three ATC cell lines were exposed to sorafenib in the presence or absence of HNHA, and cell viability was determined by MTT assay. Effects of combined treatment on cell cycle and intracellular signaling pathways were assessed by flow cytometry and western blot analysis. The ATC cell lines xenograft model was used to examine the anti-tumor activity in vivo. Our data showed that HNHA and sorafenib synergistically decreased cell viability in ATC cells, and also significantly increased apoptotic cell death in these cells, as proved by the cleavage of caspase-3 and DNA fragmentation. HNHA and sorafenib combination was reduced anti-apoptotic factor in ATC. Thus, combination therapy with HNHA and sorafenib significantly decreased vessel density, and most significantly reduced tumor volume and increased survival in ATC xenografts. These results propose that HNHA in combination with sorafenib has significant anti-cancer activity in preclinical models, potentially suggesting a new clinical approach for patients of advanced thyroid cancer type.
Clinical Cancer Research | 2017
Ki Cheong Park; Seung Won Kim; Jeong Yong Jeon; A Ra Jo; Hye Ji Choi; Jung Min Kim; Hyun Gyu Lee; Yonjung Kim; Gordon B. Mills; Sung Hoon Noh; Min Goo Lee; Eun Sung Park; Jae Ho Cheong
Purpose: Cancer cells grow in an unfavorable metabolic milieu in the tumor microenvironment and are constantly exposed to metabolic stress such as chronic nutrient depletion. Cancer stem-like cells (CSC) are intrinsically resistant to metabolic stress, thereby surviving nutrient insufficiency and driving more malignant tumor progression. In this study, we aimed to demonstrate the potential mechanisms by which CSCs avoid Ca2+-dependent apoptosis during glucose deprivation. Experimental Design: We investigated cell viability and apoptosis under glucose deprivation, performed genome-wide transcriptional profiling of paired CSCs and parental cells, studied the effect of calcium/calmodulin-dependent protein kinase 2 alpha (CaMK2α) gene knockdown, and investigated the role of nuclear factor kappa B (NFκB) in CSCs during time-dependent Ca2+-mediated and glucose deprivation–induced apoptosis. We also observed the effect of combined treatment with 2-deoxy-d-glucose, a metabolic inhibitor that mimics glucose deprivation conditions in mouse xenograft models, and thapsigargin, a specific inhibitor of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA). Results: We demonstrated the coordinated upregulation of SERCA in CSCs. SERCA, in turn, is transcriptionally regulated by CaMK2α via NFκB activation. Combined treatment with 2-deoxy-d-glucose and thapsigargin, a specific inhibitor of SERCA, significantly reduced tumor growth compared with that in untreated control animals or those treated with the metabolic inhibitor alone. Conclusions: The current study provides compelling evidence that CaMK2α acts as a key antiapoptosis regulator in metabolic stress-resistant CSCs by activating NFκB. The latter induces expression of SERCA, allowing survival in glucose-deprived conditions. Importantly, our combination therapeutic strategy provides a novel approach for the clinical application of CSC treatment. Clin Cancer Res; 24(7); 1677–90. ©2017 AACR.
Cancer Research | 2014
Kyung Seok Han; Ki Cheong Park; Jeong Yong Jeon; Sang Yong Kim; Dami Song; Woong Kyu Han; Sung Joon Hong
Introduction: N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA) is a novel HDAC inhibitor that shows better pharmacological properties than known HDAC inhibitors. We investigated a therapeutic role and its mechanisms of HNHA in renal cell carcinoma. Methods: Renal cell carcinoma cell lines, Caki-1, A-498, 786-O and UMRC-3 cells were used for in vitro study. Crystal violet assay was used to assess cell growth. Apoptosis was evaluated by TUNEL assay and confirmed by expression of cleaved caspase-3 and caspase-7. Cytochrome c release was evaluated by mitochondria staining. For in vivo study, Caki-1 and A-498 cells were injected (1.0 x 107/mouse) into the dorsal sub-membrane of 6-week-old BALB/C nude mice. Two different drugs were injected at Lethal Dose 50: HNHA, 206 µg/mL and SAHA, 138 µg/mL. Result: HNHA suppressed tumor proliferation in all panels of renal cell carcinoma cells. TUNEL assay and Western blots showed that remarkable apoptosis was induced by HNHA in Caki-1 and A-498 cells. HNHA induced apoptotic cell death by caspase activation and Bcl 2 suppression in renal cell carcinoma in vitro and in vivo. HNHA also caused cytochrome c release from mitochondria through Bax and Bak expression. A primary caspase activation was initiated by cytochrome c release from mitochondria, which is able to induce biochemical reactions, caspase activation and subsequent cell death. Conclusion: HNHA induces more potent apoptotic cell death in renal cell carcinoma in vitro and in vivo than the previous HDAC inhibitors. HNHA-induced apoptosis is mediated by cytochrome c release from mitochondria through induction of Bax and Bak expression in renal cell carcinoma. Citation Format: Kyung Seok Han, Ki Cheong Park, Jeong Yong Jeon, Sang Yong Kim, Dami Song, Woong Kyu Han, Sung Joon Hong. A novel HDAC inhibitor, N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), induces apoptotic cell death through mitochondrial cytochrome c release in renal cell carcinoma. [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 5520. doi:10.1158/1538-7445.AM2014-5520
BMC Cancer | 2015
Jeong Yong Jeon; Seung Won Kim; Ki Cheong Park; Mijin Yun