1.Jab1 Silencing Inhibits Proliferation and Sensitizes to Cisplatin in Biliary Tract Cancer
Ah Rong NAM ; Ji Won KIM ; Ji Eun PARK ; Ju Hee BANG ; Mei Hua JIN ; Do Youn OH ; Yung Jue BANG
Cancer Research and Treatment 2019;51(3):886-900
PURPOSE: Jab1 is a coactivator of c-Jun that enhances the transcriptional function of c-Jun. Jab1 is frequently overexpressed in various cancers and is associatedwith poor prognosis of cancer patients. Thus, Jab1 could be a potential therapeutic target in cancer. However, the role of Jab1 in biliary tract cancer (BTC) has not been studied. MATERIALS AND METHODS: We performed in vitro and in vivo experiments to evaluate the therapeutic potential ofJab1 inhibition in BTC. RESULTS: Among 8 BTC cell lines, many showed higher Jab1 expression levels. In addition, Jab1 silencing by siRNA increased p27 expression levels. SNU478 and HuCCT-1 cells exhibited profound Jab1 knockdown and increased p27 expression by Jab1-specific siRNA transfection. Jab1 silencing induced anti-proliferative and anti-migratory effects and resulted in G1 cell cycle arrest in SNU478 and HuCCT-1 cells. In addition, Jab1 silencing potentiated the anti-proliferative and anti-migratory effects of cisplatin by increasing DNA damage. Interestingly,Jab1 knockdown increased PTEN protein half-life, resulting in increased PTEN expression. In the HuCCT-1 mouse xenograft model, stable knockdown of Jab1 by shRNA also showed anti-proliferative effects in vivo, with decreased Ki-67 expression and AKT phosphorylation and increased Terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling and p27 expression. CONCLUSION: Jab1 knockdown demonstrated anti-proliferative and anti-migratory effects in BTC cells by increasing DNA damage and stabilizing PTEN, resulting in G1 cell cycle arrest. In addition, Jab1 silencing potentiated the anti-proliferative effects of cisplatin. Our data suggest that Jab1 may be a potential therapeutic target in BTC that is worthy of further investigations.
Animals
;
Biliary Tract Neoplasms
;
Biliary Tract
;
Cell Line
;
Cisplatin
;
DNA Damage
;
G1 Phase Cell Cycle Checkpoints
;
Half-Life
;
Heterografts
;
Humans
;
In Vitro Techniques
;
Mice
;
Phosphorylation
;
Prognosis
;
PTEN Phosphohydrolase
;
RNA, Small Interfering
;
Transfection
2.Suppressor of Variegation 3–9 Homolog 2, a Novel Binding Protein of Translationally Controlled Tumor Protein, Regulates Cancer Cell Proliferation
A Reum KIM ; Jee Young SUNG ; Seung Bae RHO ; Yong Nyun KIM ; Kyungsil YOON
Biomolecules & Therapeutics 2019;27(2):231-239
Suppressor of Variegation 3–9 Homolog 2 (SUV39H2) methylates the lysine 9 residue of histone H3 and induces heterochromatin formation, resulting in transcriptional repression or silencing of target genes. SUV39H1 and SUV39H2 have a role in embryonic development, and SUV39H1 was shown to suppress cell cycle progression associated with Rb. However, the function of human SUV39H2 has not been extensively studied. We observed that forced expression of SUV39H2 decreased cell proliferation by inducing G1 cell cycle arrest. In addition, SUV39H2 was degraded through the ubiquitin-proteasomal pathway. Using yeast two-hybrid screening to address the degradation mechanism and function of SUV39H2, we identified translationally controlled tumor protein (TCTP) as an SUV39H2-interacting molecule. Mapping of the interacting regions indicated that the N-terminal 60 amino acids (aa) of full-length SUV39H2 and the C-terminus of TCTP (120–172 aa) were critical for binding. The interaction of SUV39H2 and TCTP was further confirmed by co-immunoprecipitation and immunofluorescence staining for colocalization. Moreover, depletion of TCTP by RNAi led to up-regulation of SUV39H2 protein, while TCTP overexpression reduced SUV39H2 protein level. The half-life of SUV39H2 protein was significantly extended upon TCTP depletion. These results clearly indicate that TCTP negatively regulates the expression of SUV39H2 post-translationally. Furthermore, SUV39H2 induced apoptotic cell death in TCTP-knockdown cells. Taken together, we identified SUV39H2, as a novel target protein of TCTP and demonstrated that SUV39H2 regulates cell proliferation of lung cancer cells.
Amino Acids
;
Apoptosis
;
Carrier Proteins
;
Cell Cycle
;
Cell Death
;
Cell Proliferation
;
Embryonic Development
;
Female
;
Fluorescent Antibody Technique
;
G1 Phase Cell Cycle Checkpoints
;
Half-Life
;
Heterochromatin
;
Histones
;
Humans
;
Immunoprecipitation
;
Lung Neoplasms
;
Lysine
;
Mass Screening
;
Pregnancy
;
Repression, Psychology
;
RNA Interference
;
Up-Regulation
;
Yeasts
3.MS-5, a Naphthalene Derivative, Induces the Apoptosis of an Ovarian Cancer Cell CAOV-3 by Interfering with the Reactive Oxygen Species Generation.
Eunsook MA ; Seon Ju JEONG ; Joon Seok CHOI ; Thi Ha NGUYEN ; Chul Ho JEONG ; Sang Hoon JOO
Biomolecules & Therapeutics 2019;27(1):48-53
Reactive oxygen species (ROS) are widely generated in biological processes such as normal metabolism and response to xenobiotic exposure. While ROS can be beneficial or harmful to cells and tissues, generation of ROS by diverse anti-cancer drugs or phytochemicals plays an important role in the induction of apoptosis. We recently identified a derivative of naphthalene, MS-5, that induces apoptosis of an ovarian cell, CAOV-3. Interestingly, MS-5 induced apoptosis by down-regulating the ROS. Cell viability was evaluated by water-soluble tetrazolium salt (WST-1) assay. Apoptosis was evaluated by flow cytometry analysis. Intracellular ROS (H₂O₂), mitochondrial superoxide, mitochondrial membrane potential (MMP) and effect on cycle were determined by flow cytometry. Protein expression was assessed by western blotting. The level of ATP was measured using ATP Colorimetric/Fluorometric Assay kit. MS-5 inhibited growth of ovarian cancer cell lines, CAOV-3, in a concentration- and time-dependent manner. MS-5 also induced G1 cell cycle arrest in CAOV-3 cells, while MS-5 decreased intracellular ROS generation. In addition, cells treated with MS-5 showed the decrease in MMP and ATP production. In this study, we found that treatment with MS-5 in CAOV-3 cells induced apoptosis but decreased ROS level. We suspect that MS-5 might interfere with the minimum requirements of ROS for survival. These perturbations appear to be concentration-dependent, suggesting that MS-5 may induce apoptosis by interfering with ROS generation. We propose that MS-5 may be a potent therapeutic agent for inducing apoptosis in ovarian cancer cell through regulation of ROS.
Adenosine Triphosphate
;
Apoptosis*
;
Biological Processes
;
Blotting, Western
;
Cell Line
;
Cell Survival
;
Flow Cytometry
;
G1 Phase Cell Cycle Checkpoints
;
Membrane Potential, Mitochondrial
;
Metabolism
;
Ovarian Neoplasms*
;
Phytochemicals
;
Reactive Oxygen Species*
;
Superoxides
4.Esculetin Inhibits the Survival of Human Prostate Cancer Cells by Inducing Apoptosis and Arresting the Cell Cycle
Kader TURKEKUL ; R Dilsu COLPAN ; Talha BAYKUL ; Mehmet D OZDEMIR ; Suat ERDOGAN
Journal of Cancer Prevention 2018;23(1):10-17
BACKGROUND: Prostate cancer (PCa) is one of the most important causes of death in men and thus new therapeutic approaches are needed. In this study, antiproliferative and anti-migration properties of a coumarin derivative esculetin were evaluated. METHODS: Human PCa cell lines PC3, DU145, and LNCaP were treated with various concentrations of esculetin for 24 to 72 hours, and cell viability was determined by the MTT test. Cell cycle and apoptosis were analyzed by using cell-based cytometer. Gene expression levels were assessed by reverse transcription and quantitative real-time PCR, cell migration was determined by the wound healing assay. The protein expression was measured by Western blotting. RESULTS: Esculetin inhibited cell proliferation in a dose- and time-dependent manner. Cell migration was inhibited by esculetin treatment. Administration of esculetin significantly reduced the cells survival, induced apoptosis and caused the G1 phase cell cycle arrest shown by image-based cytometer. The induced expression of cytochrome c, p53, p21 and p27, and down-regulated CDK2 and CDK4 may be the underlying molecular mechanisms of esculetin effect. Esculetin suppressed phosphorylation of Akt and enhanced protein expression of tumor-suppressor phosphatase and tensin homologue. CONCLUSIONS: Our findings showed that the coumarin derivative esculetin could be used in the management of PCa. However, further in vivo research is needed.
Apoptosis
;
Blotting, Western
;
Cause of Death
;
Cell Cycle Checkpoints
;
Cell Cycle
;
Cell Line
;
Cell Movement
;
Cell Proliferation
;
Cell Survival
;
Cytochromes c
;
G1 Phase
;
Gene Expression
;
Humans
;
Male
;
Passive Cutaneous Anaphylaxis
;
Phosphorylation
;
Prostate
;
Prostatic Neoplasms
;
Real-Time Polymerase Chain Reaction
;
Reverse Transcription
;
Wound Healing
5.Gomisin G Inhibits the Growth of Triple-Negative Breast Cancer Cells by Suppressing AKT Phosphorylation and Decreasing Cyclin D1.
Sony MAHARJAN ; Byoung Kwon PARK ; Su In LEE ; Yoonho LIM ; Keunwook LEE ; Hyung Joo KWON
Biomolecules & Therapeutics 2018;26(3):322-327
A type of breast cancer with a defect in three molecular markers such as the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor is called triple-negative breast cancer (TNBC). Many patients with TNBC have a lower survival rate than patients with other types due to a poor prognosis. In this study, we confirmed the anti-cancer effect of a natural compound, Gomisin G, in TNBC cancer cells. Treatment with Gomisin G suppressed the viability of two TNBC cell lines, MDA-MB-231 and MDA-MB-468 but not non-TNBC cell lines such as MCF-7, T47D, and ZR75-1. To investigate the molecular mechanism of this activity, we examined the signal transduction pathways after treatment with Gomisin G in MDA-MB-231 cells. Gomisin G did not induce apoptosis but drastically inhibited AKT phosphorylation and reduced the amount of retinoblastoma tumor suppressor protein (Rb) and phosphorylated Rb. Gomisin G induced in a proteasome-dependent manner a decrease in Cyclin D1. Consequently, Gomisin G causes cell cycle arrest in the G1 phase. In contrast, there was no significant change in T47D cells except for a mild decrease in AKT phosphorylation. These results show that Gomisin G has an anti-cancer activity by suppressing proliferation rather than inducing apoptosis in TNBC cells. Our study suggests that Gomisin G could be used as a therapeutic agent in the treatment of TNBC patients.
Apoptosis
;
Breast Neoplasms
;
Cell Cycle
;
Cell Cycle Checkpoints
;
Cell Line
;
Cell Proliferation
;
Cyclin D1*
;
Cyclins*
;
Estrogens
;
G1 Phase
;
Humans
;
Phosphorylation*
;
Prognosis
;
Receptor, Epidermal Growth Factor
;
Receptors, Progesterone
;
Retinoblastoma
;
Signal Transduction
;
Survival Rate
;
Triple Negative Breast Neoplasms*
6.Inhibitory effects of resveratrol on hepatitis B virus X protein-induced hepatocellular carcinoma.
Seungmo PARK ; Jihae LIM ; Jong Rhan KIM ; Seongbeom CHO
Journal of Veterinary Science 2017;18(4):419-429
Liver cancer occurs very frequently worldwide and hepatocellular carcinoma (HCC) accounts for more than 80% of total primary liver cancer cases. In this study, the anticarcinogenic effects of resveratrol against hepatitis B virus (HBV)-induced HCC were investigated by using HBV X-protein-overexpressing Huh7 (Huh7-HBx) human hepatoma cells. MTT assay showed that resveratrol decreased cell viability. Fluorescence-activated cell-sorter analysis showed that resveratrol induced G1 cell cycle arrest without increasing the sub-G1 phase cell population. Therefore, we evaluated its effect on regulation of cyclin D1, which is critically involved in G1/S transition. Resveratrol lowered cyclin D1 transcription. Western blot analysis of the effects of resveratrol on upstream cyclin D1 transcriptional signaling, extracellular signal-related kinase (ERK), p90(RSK), Akt, and p70(S6K) revealed inhibition of Akt but not the ERK signaling pathway. Collectively, the results indicate that resveratrol inhibits Huh7-HBx proliferation by decreasing cyclin D1 expression through blockade of Akt signaling. We investigated the anticarcinogenic effect and mechanism of resveratrol in xenograft model mice implanted with Huh7-HBx cells. Intraperitoneal resveratrol injection reduced tumor size in the mice. Expression of survivin was reduced, but cyclin D1 was not affected. The results demonstrate that resveratrol treatment may help manage HBV-induced HCC by regulating survivin.
Animals
;
Anticarcinogenic Agents
;
Blotting, Western
;
Carcinoma, Hepatocellular*
;
Cell Survival
;
Cyclin D1
;
G1 Phase Cell Cycle Checkpoints
;
Hepatitis B virus*
;
Hepatitis B*
;
Hepatitis*
;
Heterografts
;
Humans
;
Liver Neoplasms
;
Mice
;
Phosphotransferases
;
Ribosomal Protein S6 Kinases, 90-kDa
7.Solanine inhibits prostate cancer Du145 xenograft growth in nude mice by inducing cell cycle arrest in G1/S phase.
Wei-Feng ZHONG ; Si-Ping LIU ; Bin PAN ; Zhao-Feng TANG ; Jin-Guang ZHONG ; Fang-Jian ZHOU
Journal of Southern Medical University 2016;36(5):665-670
OBJECTIVETo investigate the effect of solanine on the growth of human prostate cancer cell xenograft in nude mice.
METHODSHuman prostate cancer Du145 cells were injected into the subcutaneous layers on the back of nude mice. After a week, the mice bearing subcutaneous tumor graft were randomly divided into solanine treatment group and saline control group for treatment for 3 weeks. The tumor grafts were then harvested to evaluate the inhibition rate. The mRNA and protein expressions of cell cycle-related genes in the tumors were detected by qRT-PCR and Western blotting, respectively, and tumor cell apoptosis was detected using TUNEL method.
RESULTSThe tumor growth rate in solanine-treated group was significantly slower than that in the control group (P<0.01). The mRNA and protein expressions of C-myc, cyclin D1, cyclin E1, CDK2, CDK4 and CDK6 were significantly inhibited by solanine. Solanine significantly up-regulated p21 mRNA and protein expression in the tumors and induced a higher apoptosis rate of the tumor cells than saline (P<0.01).
CONCLUSIONThe tumor-inhibition effect of solanine is probably mediated by regulating the expressions of genes related with G1/S cell cycle arrest and cell apoptosis.
Animals ; Apoptosis ; Cyclin-Dependent Kinases ; metabolism ; Cyclins ; metabolism ; G1 Phase Cell Cycle Checkpoints ; Humans ; Male ; Mice ; Mice, Nude ; Neoplasm Transplantation ; pathology ; Prostatic Neoplasms ; drug therapy ; pathology ; S Phase ; Solanine ; pharmacology
8.Dehydroglyasperin D Inhibits the Proliferation of HT-29 Human Colorectal Cancer Cells Through Direct Interaction With Phosphatidylinositol 3-kinase.
Sung Keun JUNG ; Chul Ho JEONG
Journal of Cancer Prevention 2016;21(1):26-31
BACKGROUND: Despite recent advances in therapy, colorectal cancer still has a grim prognosis. Although licorice has been used in East Asian traditional medicine, the molecular properties of its constituents including dehydroglyasperin D (DHGA-D) remain unknown. We sought to evaluate the inhibitory effect of DHGA-D on colorectal cancer cell proliferation and identify the primary signaling molecule targeted by DHGA-D. METHODS: We evaluated anchorage-dependent and -independent cell growth in HT-29 human colorectal adenocarcinoma cells. The target protein of DHGA-D was identified by Western blot analysis with a specific antibody, and direct interaction between DHGA-D and the target protein was confirmed by kinase and pull-down assays. Cell cycle analysis by flow cytometry and further Western blot analysis was performed to identify the signaling pathway involved. RESULTS: DHGA-D significantly suppressed anchorage-dependent and -independent HT-29 colorectal cancer cell proliferation. DHGA-D directly suppressed phosphatidylinositol 3-kinase (PI3K) activity and subsequent Akt phosphorylation and bound to the p110 subunit of PI3K. DHGA-D also significantly induced G1 cell cycle arrest, together with the suppression of glycogen synthase kinase 3β and retinoblastoma phosphorylation and cyclin D1 expression. CONCLUSIONS: DHGA-D has potent anticancer activity and targets PI3K in human colorectal adenocarcinoma HT-29 cells. To our knowledge, this is the first report to detail the molecular basis of DHGA-D in suppressing colorectal cancer cell growth.
Adenocarcinoma
;
Blotting, Western
;
Cell Cycle
;
Cell Proliferation
;
Colorectal Neoplasms*
;
Cyclin D1
;
Flow Cytometry
;
G1 Phase Cell Cycle Checkpoints
;
Glycogen Synthase Kinases
;
Glycyrrhiza
;
HT29 Cells
;
Humans*
;
Medicine, East Asian Traditional
;
Phosphatidylinositol 3-Kinase*
;
Phosphatidylinositols*
;
Phosphorylation
;
Phosphotransferases
;
Prognosis
;
Retinoblastoma
9.Effects of Triterpenoids from Luvunga scandens on Cytotoxic, Cell Cycle Arrest and Gene Expressions in MCF-7 Cells.
Muhammad TAHER ; Putri Nur Hidayah AL-ZIKRI ; Deny SUSANTI ; Solachuddin Jauhari Arief ICHWAN ; Mohamad Fazlin REZALI
Natural Product Sciences 2016;22(4):293-298
Plant-derived triterpenoids commonly possesses biological properties such as anti-inflammatory, anti-microbial, anti-viral and anti-cancer. Luvunga scandens is one of the plant that produced triterpenoids. The aims of the study was to analyze cell cycle profile and to determine the expression of p53 unregulated modulator of apoptosis (PUMA), caspase-8 and caspase-9 genes at mRNA level in MCF-7 cell line treated with two triterpenoids, flindissol (1) and 3-oxotirucalla-7,24-dien-21-oic-acid (2) isolated from L. scandens. The compounds were tested for cell cycle analysis using flow cytometer and mRNA expression level using quantitative RT-PCR. The number of MCF-7 cells population which distributed in Sub G1 phase after treated with compound 1 and 2 were 7.7 and 9.3% respectively. The evaluation of the expression of genes showed that both compounds exhibited high level of expression of PUMA, caspase-8 and caspase-9 as normalized to β-actin via activation of those genes. In summary, the isolated compounds of L. scandens plant showed promising anticancer properties in MCF-7 cell lines.
Apoptosis
;
Caspase 8
;
Caspase 9
;
Cell Cycle Checkpoints*
;
Cell Cycle*
;
Flow Cytometry
;
G1 Phase
;
Gene Expression*
;
MCF-7 Cells*
;
Plants
;
Puma
;
RNA, Messenger
10.Use of deferasirox, an iron chelator, to overcome imatinib resistance of chronic myeloid leukemia cells.
Dae Sik KIM ; Yoo Jin NA ; Myoung Hee KANG ; Soo Young YOON ; Chul Won CHOI
The Korean Journal of Internal Medicine 2016;31(2):357-366
BACKGROUND/AIMS: The treatment of chronic myeloid leukemia (CML) has achieved impressive success since the development of the Bcr-Abl tyrosine kinase inhibitor, imatinib mesylate. Nevertheless, resistance to imatinib has been observed, and a substantial number of patients need alternative treatment strategies. METHODS: We have evaluated the effects of deferasirox, an orally active iron chelator, and imatinib on K562 and KU812 human CML cell lines. Imatinib-resistant CML cell lines were created by exposing cells to gradually increasing concentrations of imatinib. RESULTS: Co-treatment of cells with deferasirox and imatinib induced a synergistic dose-dependent inhibition of proliferation of both CML cell lines. Cell cycle analysis showed an accumulation of cells in the subG1 phase. Western blot analysis of apoptotic proteins showed that co-treatment with deferasirox and imatinib induced an increased expression of apoptotic proteins. These tendencies were clearly identified in imatinib-resistant CML cell lines. The results also showed that co-treatment with deferasirox and imatinib reduced the expression of BcrAbl, phosphorylated Bcr-Abl, nuclear factor-kappaB (NF-kappaB) and beta-catenin. CONCLUSIONS: We observed synergistic effects of deferasirox and imatinib on both imatinib-resistant and imatinib-sensitive cell lines. These effects were due to induction of apoptosis and cell cycle arrest by down-regulated expression of NF-kappaB and beta-catenin levels. Based on these results, we suggest that a combination treatment of deferasirox and imatinib could be considered as an alternative treatment option for imatinib-resistant CML.
Antineoplastic Agents/*pharmacology
;
Apoptosis/drug effects
;
Apoptosis Regulatory Proteins/metabolism
;
Benzoates/*pharmacology
;
Cell Proliferation/drug effects
;
Dose-Response Relationship, Drug
;
Drug Resistance, Neoplasm/*drug effects
;
G1 Phase Cell Cycle Checkpoints/drug effects
;
Humans
;
Imatinib Mesylate/*pharmacology
;
Iron Chelating Agents/*pharmacology
;
K562 Cells
;
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/*drug therapy/metabolism
;
Protein Kinase Inhibitors/*pharmacology
;
Signal Transduction/drug effects
;
Triazoles/*pharmacology

Result Analysis
Print
Save
E-mail