1.The Changes of Cell Cycle Phase Fractions and Expression of p53 by the Treatment of Staurosporine in MCF-7 Cell Line.
Jung NAM ; Kyung A YEA ; Hea Nam LEE ; Hyun Hee JO ; Ki Sung RYU ; Young Oak LEW ; Jong Gu RHA ; Ku Taek HAN
Korean Journal of Obstetrics and Gynecology 2001;44(3):501-505
OBJECTIVE: We investigated the effects on the cell cycle and p53 expression by the treatment of various concentrations of staurosporine to elucidate the molecular mechanism of staurosporine induced cell cycle arrest in MCF-7 cell line. METHODS: Various concentrations of staurosporine were treated in MCF-7 cells cultured with RPMI 1640 media. The harvested cells were fixed and permeabilized with 1% paraformaldehyde and absolute methanol. Then the cells were stained indirectly with anti-p53 primary antibody and FITC conjugated goat anti-mouse(GAM)-IgG secondary antibody. Sequentially DNA were stained with 0.1% RNase and PI solution. These stained cells were analyzed by the standard FACScan flow cytometer. The obtained results were analyzed further with WinList 3.0, and ModiFit LT software program. RESULTS: MCF-7 cells were arrested mostly in G1 phase of cell cycle at 5-10 nM of staurosporine, however, the cells were arrested in G2 phase at 20-100 nM of staurosporine. The expressions of p53 protein were higher in the MCF-7 cells treated with both concentrations of 10 nM and 100 nM of staurosporine compaired with the control cells. This suggests that the p53 may be involved in the mechanism of G1 and G2M arrest of cell cycle in MCF-7 cell. CONCLUSIONS: The points of arrest in cell cycle differred depending on the concentrations of staurosporine and these cell cycle arrests at G0G1 and G2M pahse were related with p53 protein expression. It suggested that these results could be extended to study for staurosporine to be usefull as a potential anti-tumor agent.
Cell Cycle Checkpoints
;
Cell Cycle*
;
DNA
;
Fluorescein-5-isothiocyanate
;
G1 Phase
;
G2 Phase
;
Goats
;
MCF-7 Cells*
;
Methanol
;
Ribonucleases
;
Staurosporine*
2.Regulatory Mechanism of Radiation-induced Cancer Cell Death by the Change of Cell Cycle.
Soo Jin JEONG ; Min Ho JEONG ; Ji Yeon JANG ; Wol Soon JO ; Byung Hyouk NAM ; Min Za JEONG ; Young Jin LIM ; Byung Gon JANG ; Seon Min YOUN ; Hyung Sik LEE ; Won Joo HUR ; Kwang Mo YANG
The Journal of the Korean Society for Therapeutic Radiology and Oncology 2003;21(4):306-314
PURPOSE: In our previous study, we have shown the main cell death pattern induced by irradiation or protein tyrosine kinase (PTK) inhibitors in K562 human myelogenous leukemic cell line. Death of the cells treated with irradiation alone was characterized by mitotic catastrophe and typical radiation-induced apoptosis was accelerated by herbimycin A (HMA). Both types of cell death were inhibited by genistein. In this study, we investigated the effects of HMA and genistein on cell cycle regulation and its correlation with the alterations of radiation-induced cell death. MATERIALS AND METHODS: K562 cells in exponential growth phase were used for this study. The cells were irradiated with 10 Gy using 6 MeV Linac (200-300 cGy/min). Immediately after irradiation, cells were treated with 250 nM of HMA or 25 microM of genistein. The distributions of cell cycle, the expressions of cell cycle-related protein, the activities of cyclin-dependent kinase, and the yield of senescence and differentiation were analyzed. RESULTS: X-irradiated cells were arrested in the G2 phase of the cell cycle but unlike the p53-positive cells, they were not able to sustain the cell cycle arrest. An accumulation of cells in G2 phase of first cell-cycle post-treatment and an increase of cyclin B1 were correlated with spontaneous, premature, chromosome condensation and mitotic catastrophe. HMA induced rapid G2 checkpoint abrogation and concomitant p53-independent G1 accumulation. HMA-induced cell cycle modifications correlated with the increase of cdc2 kinase activity, the decrease of the expressions of cyclins E and A and of CDK2 kinase activity, and the enhancement of radiation-induced apoptosis. Genistein maintained cells that were arrested in the G2-phase, decreased the expressions of cyclin B1 and cdc25C and cdc2 kinase activity, increased the expression of p16, and sustained senescence and megakaryocytic differentiation. CONCLUSION: The effects of HMA and genistein on the radiation-induced cell death of K562 cells were closely related to the cell cycle regulatory activities. In this study, we present a unique and reproducible model in which for investigating the mechanisms of various, radiation-induced, cancer cell death patterns. Further evaluation by using this model will provide a potent target for a new strategy of radiotherapy.
Aging
;
Apoptosis
;
Cell Cycle Checkpoints
;
Cell Cycle*
;
Cell Death*
;
Cell Line
;
Cyclin B1
;
Cyclins
;
G2 Phase
;
Genistein
;
Humans
;
K562 Cells
;
Neoplasms, Radiation-Induced*
;
Phosphotransferases
;
Protein-Tyrosine Kinases
;
Radiotherapy
3.Downregulation of MCL-1 by Diallyl Disulfide Induces G/M Arrest in Human Leukemia K562 Cells and Its Mechanism.
Xiao-Xia JI ; Fang LIU ; Hong XIA ; Jie HE ; Hui TAN ; Lan YI ; Qi SU
Journal of Experimental Hematology 2018;26(3):750-755
OBJECTIVETo investigate the inducing effect of down-regulation of MCL-1 by diallyl disulfide(DADS) on the G/M arrest of human leukemia K562 cells and its mechanisms.
METHODSCCK-8 was used to detect the effect of DADS on proliferation of K562 cells, flow cytometry was employed to observe the effect of cycle arrest by DADS and RNAi silencing MCL-1 gene in K562 cells. The expressions of MCL-1, PCNA and CDK1 in K562 cells treated with DADS were detected by Western blot. The amphigamy of MCL-1 with PCNA and CDK1 was detected by Coimmunoprecipitation.
RESULTSCCK-8 detection showed that the inhibition rates of K562 cells treated with 15, 30, 60, 120, 240 µmol/L DADS were 32.48%, 59.34%, 66.42%, 77.06%, 81.05% respectively (P<0.05). Flow cytometry analysis revealed that the perecentages of G/M cells were increased to 18.6% and 34.4%, 17.5% and 28.5%, respectively at 24 and 48 h after treating K562 cells with 60 and 120 µmol/L DADS (P<0.05). And the perecentage of G/M cells of silencing MCL-1 was significantly increased (P<0.05). Silencing effects of MCL-1+DADS on the cells were enhanced more significantly as compared with DADS or MCL-1 alone (P<0.05). Western blot exhibited that DADS could markedly downregulate the expression of MCL-1, PCNA and CDK1(P<0.05). Coimmunoprecipitation revealed that MCL-1 bound with PCNA and CDK1, then forming heterodimers, which were downregulated respectively more significantly than that in the control group after treating K562 cells with DADS for 8 h (P<0.05).
CONCLUSIONDADS can inhibit the K562 cell proliferation and induce them arrest G/M through downregulation of MCL-1, then decreasing the expression of PCNA and CDK1 in hunan leukemia K562 cells. Moreover, silencing MCL-1 can enhance the effect of DADS.
Allyl Compounds ; Apoptosis ; Cell Line, Tumor ; Disulfides ; Down-Regulation ; G2 Phase Cell Cycle Checkpoints ; Humans ; K562 Cells ; Leukemia ; M Phase Cell Cycle Checkpoints ; Myeloid Cell Leukemia Sequence 1 Protein
4.ATM Signaling Pathway Is Implicated in the SMYD3-mediated Proliferation and Migration of Gastric Cancer Cells.
Lei WANG ; Qiu Tong WANG ; Yu Peng LIU ; Qing Qing DONG ; Hai Jie HU ; Zhi MIAO ; Shuang LI ; Yong LIU ; Hao ZHOU ; Tong Cun ZHANG ; Wen Jian MA ; Xue Gang LUO
Journal of Gastric Cancer 2017;17(4):295-305
PURPOSE: We previously found that the histone methyltransferase suppressor of variegation, enhancer of zeste, trithorax and myeloid-nervy-deformed epidermal autoregulatory factor-1 domain-containing protein 3 (SMYD3) is a potential independent predictive factor or prognostic factor for overall survival in gastric cancer patients, but its roles seem to differ from those in other cancers. Therefore, in this study, the detailed functions of SMYD3 in cell proliferation and migration in gastric cancer were examined. MATERIALS AND METHODS: SMYD3 was overexpressed or suppressed by transfection with an expression plasmid or siRNA, and a wound healing migration assay and Transwell assay were performed to detect the migration and invasion ability of gastric cancer cells. Additionally, an MTT assay and clonogenic assay were performed to evaluate cell proliferation, and a cell cycle analysis was performed by propidium iodide staining. Furthermore, the expression of genes implicated in the ataxia telangiectasia mutated (ATM) pathway and proteins involved in cell cycle regulation were detected by polymerase chain reaction and western blot analyses. RESULTS: Compared with control cells, gastric cancer cells transfected with si-SMYD3 showed lower migration and invasion abilities (P<0.05), and the absence of SMYD3 halted cells in G2/M phase and activated the ATM pathway. Furthermore, the opposite patterns were observed when SMYD3 was elevated in normal gastric cells. CONCLUSIONS: To the best of our knowledge, this study provides the first evidence that the absence of SMYD3 could inhibit the migration, invasion, and proliferation of gastric cancer cells and halt cells in G2/M phase via the ATM-CHK2/p53-Cdc25C pathway. These findings indicated that SMYD3 plays crucial roles in the proliferation, migration, and invasion of gastric cancer cells and may be a useful therapeutic target in human gastric carcinomas.
Ataxia Telangiectasia
;
Blotting, Western
;
Cell Cycle
;
Cell Proliferation
;
G2 Phase Cell Cycle Checkpoints
;
Histones
;
Humans
;
Plasmids
;
Polymerase Chain Reaction
;
Propidium
;
RNA, Small Interfering
;
Stomach Neoplasms*
;
Transfection
;
Wound Healing
5.Oleanolic acid induces G₂/M phase arrest and apoptosis in human hepatocellular carcinoma Bel-7402 cells.
Ling LIU ; Jian-long ZHAO ; Jian-gang WANG
China Journal of Chinese Materia Medica 2015;40(24):4897-4902
This study was to examine the mechanism of oleanolic acid (OA) induces G2/M phase arrest and apoptosis in human hepatocellular carcinoma Bel-7402 cells. MTT and trypan blue exclusion test assay were adopted to detect the proliferate status of cells treated with OA. We assayed the cell cycle by flow cytometry using PI staining. Apoptosis was determined by Annexin V-FITC staining and PI labeling. The expressions of cycle related proteins and apoptotic related proteins were determined by Western blot analysis. OA strongly inhibited human hepatoma cells proliferation. When Bel-7402 cells were pretreated with OA for 24 h, OA induced apoptosis and G₂/M phase cell cycle arrest in a concentration-dependent manner. Analysis of the cell cycle regulatory proteins demonstrated that OA decreased the protein levels of cyclin B1, but increased the protein levels of p-Cdk1 (Tyr15) and p-Cdc25C (Ser 216). Moreover, OA modulated the phosphorylation of protein kinases Chk1 and p2l. Western blotting assay also showed significant decrease of Bcl-2 protein expression and increase of Bax protein expression, the cytosol Cyt c level, cleaved-caspase-9 and cleaved-caspase-3 activity. These data suggest that OA produces anti-tumor effect via induction of G₂/M cell cycle arrest and apoptosis.
Apoptosis
;
drug effects
;
Carcinoma, Hepatocellular
;
drug therapy
;
pathology
;
Cell Line, Tumor
;
G2 Phase Cell Cycle Checkpoints
;
drug effects
;
Humans
;
Liver Neoplasms
;
drug therapy
;
pathology
;
M Phase Cell Cycle Checkpoints
;
drug effects
;
Oleanolic Acid
;
pharmacology
6.Ophiopogonin D inhibits cell proliferation, causes cell cycle arrest at G2/M, and induces apoptosis in human breast carcinoma MCF-7 cells.
Qing-qing ZANG ; Lu ZHANG ; Ning GAO ; Cheng HUANG
Journal of Integrative Medicine 2016;14(1):51-59
OBJECTIVETo investigate the effects of ophiopogonin D on human breast cancer MCF-7 cells.
METHODSCell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and colony formation experiments. Cell cycle was measured with cell cycle flow cytometry and a living cell assay. Apoptosis and terminal deoxynucleoitidyl transferase-mediated dUTP nick end labeling assays were performed to detect the apoptosis of MCF-7 cells induced by ophiopogonin D. Finally, Western blotting was used to explore the mechanism.
RESULTSExposure of cells to ophiopogonin D resulted in marked decreases in viable cells and colony formation with a dose-dependent manner. Treatment of these cells with ophiopogonin D also resulted in cell cycle arrest at the G(2)/M phase, and increased apoptosis. Mechanistically, ophiopogonin D-induced G(2)/M cell cycle arrest was associated with down-regulation of cyclin B1. Furthermore, activation of caspase-8 and caspase-9 was involved in ophiopogonin D-induced apoptosis.
CONCLUSIONThe data suggested that ophiopogonin D inhibits MCF-7 cell growth via the induction of cell cycle arrest at the G(2)/M phase.
Apoptosis ; drug effects ; Cell Proliferation ; drug effects ; G2 Phase Cell Cycle Checkpoints ; drug effects ; Humans ; M Phase Cell Cycle Checkpoints ; drug effects ; MCF-7 Cells ; Saponins ; pharmacology ; Spirostans ; pharmacology
7.miR-18a enhances the radiosensitivity of nasopharyngeal carcinoma cells through inducing autophagy.
Li Hong CHANG ; Zhou Zhou YAO ; Hong Wei BAO ; Yue LI ; Xiao Hong CHEN ; Xiao Ping LAI ; Zi Zhen HUANG ; Ge Hua ZHANG
Chinese Journal of Otorhinolaryngology Head and Neck Surgery 2021;56(7):736-745
Objective: To explore the impacts of miR-18a overexpression or depression on the radiosensitivities of nasopharyngeal carcinoma cell line CNE1 and CNE2 and underlying mechanisms. Methods: CNE1 and CNE2 were transfected with miR-18a mimics, inhibitor and the corresponding control vectors. qRT-PCR and western blot were used to determine the ataxia telangiectasia mutated (ATM) expressions in CNE1 and CNE2. CNE1 and CNE2 with stably expressing miR-18a and miR-18a siRNA were constructed. Methyl thiazolyl tetrazolium (MTT) assay was used to detect the impacts of the miR-18a overexpression or depression combined with irradiation on the cell growth. Flow cytometry was used to detect the cell apoptosis and cell cycle. Colony formation assay was used to evaluate the raodiosensitivities of cells. Acridine orange (AO) staining and western blot were used respectively to test the autophagy and the expressions of related proteins. Independent samples t test was used to compare the mean value between groups by using SPSS 16.0. Results: ATM mRNA was decreased significantly in CNE1 and CNE2 cells transfected with 100 or 200 nmol/L miR-18a mimics for 48 hours (CNE1: RQ=0.174±0.139 and 0.003±0.001, t=9.939 and 19 470.783;CNE2: RQ=0.024±0.008 and 0.019±0.012, t=270.230 and 137.746, respectively, all P<0.001). ATM proteins were also decreased after transfected with 100 or 200 nmol/L miR-18a mimics for 72 hours. While in the cells transfected with 100 and 200 nmol/L miR-18a inhibitor for 48 hours, the expressions of ATM mRNA were upregulated significantly (CNE1: RQ=9.419±2.495 and 2.500±1.063, t=-4.427 and -41.241; CNE2: RQ=7.210±0.171 and 115.875±15.805, t=-62.789 and -12.589, all P<0.05), and the expressions of ATM proteins increased after transfected for 72 hours. The growth of cells with miR-18a overexpression plus 4 Gy irradiation were obviously inhibited compared to that of cells with the 4Gy irradiation alone; while the growth of miR-18a-inhibited cells increased compared to that of cells with 4 Gy irradiation alone (all P<0.05). CNE1 transfected with 100 nmol/L miR-18a mimics plus 4 Gy irradiation showed the higher apoptosis rate than the cells with 4 Gy irradiation alone ((22.9±2.1)% vs. (16.3±1.0)%, t=-4.870, P<0.01). Compared to the cells with 4 Gy irradiation alone, miR-18a-overexpressed cells plus 4 Gy irradiation decreased their percentages in G1 phases ((20.2±3.0)% vs. (29.8±4.4)%, t=3.119) and G2/M phases ((21.5±0.9)% vs. (33.4±3.1)%, t=6.410, P<0.05 for both), and increased their percentages in S phases ((56.7±4.9)% vs. (36.8±6.4)%, t=-4.246, P<0.05), and these cells possessed less colony number after exposure to different doses of irradiation, more autophagy-lysosome number, and more expressions of LC3 proteins (all P<0.05). There were no significant differences in the expressions of p62 expressions between different groups of cells. Conclusion: Overexpression of miR-18a can enhance the radiosensitivities of NPC cells by targeting ATM to abrogate G1/S, G2/M arrest and to induce autophagy and apoptosis.
Apoptosis
;
Autophagy
;
Cell Line, Tumor
;
Cell Proliferation
;
G2 Phase Cell Cycle Checkpoints
;
Humans
;
MicroRNAs/genetics*
;
Nasopharyngeal Carcinoma/genetics*
;
Nasopharyngeal Neoplasms/genetics*
;
Radiation Tolerance
8.Effects of long non-coding RNA-HOTAIR on the cell cycle and invasiveness of prostate cancer.
Yi ZHU ; Ri-kao YU ; A-fin JI ; Xiao-lin YAO ; Jia-jie FANG ; Xiao-dong JIN
National Journal of Andrology 2015;21(9):792-796
OBJECTIVETo investigate the expression of long non-coding RNA-HOTAIR in prostate cancer cells and its effects on the growth and metastasis of the cells.
METHODSUsing quantitative reverse-transcription PCR (qRT-PCR), we determined the relative expression of HOTAIR in the normal human prostate epithelial cell line RWPE-I and prostate cancer cell lines PC-3 and DU145. We detected the effects of HOTAIR on the cell cycle and invasiveness of prostate cancer cells by RNA interference, flow cytometry, and Transwell mitration assay.
RESULTSThe expressions of HOTAIR in the PC3 and DU145 cells were increased 3.2 and 5.7 times, respectively, as compared with that in the normal RWPE-1 cells. After si-HOTAIR interference, the prostate cancer cells were arrested in the G2 phase and downregulated in the G1 phase. The invasive ability of the prostate cancer cells was evidently inhibited, with the inhibition rates of 32% and 44% of the PC3 cells and 43% and 34% of the DU145 cells for si-HOTAIR1 and si-HOTAIR2, respectively.
CONCLUSIONIncRNA HOTAIR is highly expressed in prostate cancer, which is associated with the growth and invasiveness of prostate cancer cells. HOTAIR is potentially a novel marker for the diagnosis and prognosis of prostate cancer.
Cell Cycle ; Cell Cycle Checkpoints ; Cell Division ; Cell Line, Tumor ; Cell Proliferation ; Down-Regulation ; G1 Phase ; G2 Phase ; Humans ; Male ; Neoplasm Invasiveness ; Prognosis ; Prostatic Neoplasms ; metabolism ; pathology ; RNA Interference ; RNA, Long Noncoding ; metabolism ; RNA, Untranslated ; metabolism
9.Human Papilloma Virus Type 16 E7 Oncoprotein Stabilizes p53 Protein but not Induced p53-mediated Apoptosis in HepG2 Cells after gamma-irradiation under Hypoxia.
Hye Jin HWANG ; Eun Jung CHOI ; Yoon Jung CHOI ; Won Taek LEE ; Kyung Ah PARK ; Jong Eun LEE
Korean Journal of Anatomy 2007;40(2):95-106
Human papilloma virus 16 E6 and E7 oncoproteins are well known to change cell functions, especially through p53 and pRb expression, so we studied their effects on molecular mechanisms and on the cell death associated with hypoxia and ionizing radiation. These treatments both caused cell death and increased p53 protein expression in HepG2 cells. This increased p53 expression by gamma-irradiation under hypoxia induced G1 cell cycle arrest and led to apoptosis even though HepG2 cells have a relatively reduced ability to induce p21 and pRb expression levels. Ablation of p53 expression by the HPV 16 E6 gene induced E2F-1 expression, which plays a role in cellular survival, especially under hypoxia or gamma-irradiation. The steady-state level of p53 action produced by HPV 16 E7 did not induce apoptotic cell death or the production of the apoptotic regulators, the bcl-2 family and caspase-3, so it did not appear to participate in apoptotic signaling in response to hypoxia and ionizing radiation. Thus, the HPV 16 E7 oncoprotein did not increase the rate of cell death induced by p53, although p53 might play a role in apoptosis in HepG2 cells.
Anoxia*
;
Apoptosis*
;
Caspase 3
;
Cell Death
;
G1 Phase Cell Cycle Checkpoints
;
Hep G2 Cells*
;
Human papillomavirus 16
;
Humans*
;
Oncogene Proteins
;
Papilloma*
;
Radiation, Ionizing
10.p53 Prevents Immature Escaping from Cell Cycle G2 Checkpoint Arrest through Inhibiting cdk2-dependent NF-Y Phosphorylation.
Un Jung YUN ; Heui Dong PARK ; Deug Y SHIN
Cancer Research and Treatment 2006;38(4):224-228
PURPOSE: Recent studies have suggested that p53 regulates the G2 checkpoint in the cell cycle and this function is required for the maintenance of genomic integrity. In this study, we addressed a role of p53 in escaping from cell cycle G2 arrest following DNA damage. MATERIALS AND METHODS: Cell cycle checkpoint arrest in the human colon cancer cell line HCT116 and its derivatives carry p53 or p21 deletions, were examined by FACS analysis, immunoprecipitation, Western blot and IP-kinase assay. RESULTS: While the cells with functional p53 were arrested at both the G1 and G2 checkpoints, the p53-deficient cells failed to arrest at G1, but they were arrested at G2. However, the p53-deficient cells failed to sustain G2 checkpoint arrest and they entered mitosis earlier than did the p53-positive cells and so this resulted in extensive cell death. Cdc2 kinase becomes reactivated in p53-deficient cells in association with entry into mitosis, but not in the p53-positive cells. Upon DNA damage, the p21-deficient cells, like the p53-negative cells, not only failed to repress cdk2- dependent NF-Y phosphorylation, but they also failed to repress the expression of such cell cycle G2-regulatory genes as cdc2, cyclin B, RNR-R2 and cdc25C, which have all been previously reported as targets of NF-Y transcription factor. CONCLUSION: p53 is essential to prevent immature escaping from cell cycle G2 checkpoint arrest through p21-mediated cdk2 inactivation, and this leads to inhibition of cdk2-dependent NF-Y phosphorylation and NF-Y dependent transcription of the cell cycle G2-rgulatory genes, including cdc2 and cyclin B.
Blotting, Western
;
CCAAT-Binding Factor
;
CDC2 Protein Kinase
;
Cell Cycle Checkpoints
;
Cell Cycle*
;
Cell Death
;
Cell Line
;
Colonic Neoplasms
;
Cyclin B
;
DNA Damage
;
G2 Phase
;
Humans
;
Immunoprecipitation
;
Mitosis
;
Phosphorylation*
;
Phosphotransferases
;
Transcription Factors
;
Tumor Suppressor Protein p53
;
United Nations*