Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Biomolecules & Therapeutics ; : 48-53, 2019.
Article in English | WPRIM | ID: wpr-719642

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate , Apoptosis , Biological Phenomena , Blotting, Western , Cell Line , Cell Survival , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , Membrane Potential, Mitochondrial , Metabolism , Ovarian Neoplasms , Phytochemicals , Reactive Oxygen Species , Superoxides
2.
Biomolecules & Therapeutics ; : 231-239, 2019.
Article in English | WPRIM | ID: wpr-739653

ABSTRACT

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.


Subject(s)
Female , Humans , Pregnancy , Amino Acids , Apoptosis , Carrier Proteins , Cell Cycle , Cell Death , Cell Proliferation , Embryonic Development , Fluorescent Antibody Technique , G1 Phase Cell Cycle Checkpoints , Half-Life , Heterochromatin , Histones , Immunoprecipitation , Lung Neoplasms , Lysine , Mass Screening , Repression, Psychology , RNA Interference , Up-Regulation , Yeasts
3.
Cancer Research and Treatment ; : 886-900, 2019.
Article in English | WPRIM | ID: wpr-763190

ABSTRACT

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.


Subject(s)
Animals , Humans , Mice , Biliary Tract Neoplasms , Biliary Tract , Cell Line , Cisplatin , DNA Damage , G1 Phase Cell Cycle Checkpoints , Half-Life , Heterografts , In Vitro Techniques , Phosphorylation , Prognosis , PTEN Phosphohydrolase , RNA, Small Interfering , Transfection
4.
Journal of Veterinary Science ; : 419-429, 2017.
Article in English | WPRIM | ID: wpr-16843

ABSTRACT

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.


Subject(s)
Animals , Humans , Mice , Anticarcinogenic Agents , Blotting, Western , Carcinoma, Hepatocellular , Cell Survival , Cyclin D1 , G1 Phase Cell Cycle Checkpoints , Hepatitis B virus , Hepatitis B , Hepatitis , Heterografts , Liver Neoplasms , Phosphotransferases , Ribosomal Protein S6 Kinases, 90-kDa
5.
Journal of Southern Medical University ; (12): 665-670, 2016.
Article in Chinese | WPRIM | ID: wpr-263984

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of solanine on the growth of human prostate cancer cell xenograft in nude mice.</p><p><b>METHODS</b>Human 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.</p><p><b>RESULTS</b>The 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).</p><p><b>CONCLUSION</b>The tumor-inhibition effect of solanine is probably mediated by regulating the expressions of genes related with G1/S cell cycle arrest and cell apoptosis.</p>


Subject(s)
Animals , Humans , Male , Mice , Apoptosis , Cyclin-Dependent Kinases , Metabolism , Cyclins , Metabolism , G1 Phase Cell Cycle Checkpoints , Mice, Nude , Neoplasm Transplantation , Pathology , Prostatic Neoplasms , Drug Therapy , Pathology , S Phase , Solanine , Pharmacology
6.
Protein & Cell ; (12): 662-672, 2016.
Article in English | WPRIM | ID: wpr-757410

ABSTRACT

Matrine (MT), the effective component of Sophora flavescens Ait, has been shown to have anti-inflammation, immune-suppressive, anti-tumor, and anti-hepatic fibrosis activities. However, the pharmacological effects of MT still need to be strengthened due to its relatively low efficacy and short half-life. In the present study, we report a more effective thio derivative of MT, MD-1, and its inhibitory effects on the activation of hepatic stellate cells (HSCs) in both cell culture and animal models. Cytological experiments showed that MD-1 can inhibit the proliferation of HSC-T6 cells with a half-maximal inhibitory concentration (IC50) of 62 μmol/L. In addition, MD-1 more strongly inhibits the migration of HSC-T6 cells compared to MT and can more effectively induce G0/G1 arrest and apoptosis. Investigating the biological mechanisms underlying anti-hepatic fibrosis in the presence of MD-1, we found that MD-1 can bind the epidermal growth factor receptor (EGFR) on the surface of HSC-T6 cells, which can further inhibit the phosphorylation of EGFR and its downstream protein kinase B (Akt), resulting in decreased expression of cyclin D1 and eventual inhibition of the activation of HSC-T6 cells. Furthermore, in rats with dimethylnitrosamine (DMN)-induced hepatic fibrosis, MD-1 slowed the development and progression of hepatic fibrosis, protecting hepatic parenchymal cells and improving hepatic functions. Therefore, MD-1 is a potential drug for anti-hepatic fibrosis.


Subject(s)
Animals , Rats , Alkaloids , Pharmacology , Cell Line , Cyclin D1 , Metabolism , Dimethylnitrosamine , Toxicity , Enzyme Activation , ErbB Receptors , Metabolism , G1 Phase Cell Cycle Checkpoints , Hepatic Stellate Cells , Metabolism , Pathology , Liver Cirrhosis , Metabolism , Pathology , Phosphorylation , Proto-Oncogene Proteins c-akt , Metabolism , Quinolizines , Pharmacology
7.
Journal of Cancer Prevention ; : 26-31, 2016.
Article in English | WPRIM | ID: wpr-89894

ABSTRACT

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.


Subject(s)
Humans , Adenocarcinoma , Blotting, Western , Cell Cycle , Cell Proliferation , Colorectal Neoplasms , Cyclin D1 , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , Glycogen Synthase Kinases , Glycyrrhiza , HT29 Cells , Medicine, East Asian Traditional , Phosphatidylinositol 3-Kinase , Phosphatidylinositols , Phosphorylation , Phosphotransferases , Prognosis , Retinoblastoma
8.
The Korean Journal of Internal Medicine ; : 357-366, 2016.
Article in English | WPRIM | ID: wpr-109562

ABSTRACT

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.


Subject(s)
Humans , 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 , Imatinib Mesylate/pharmacology , Iron Chelating Agents/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Triazoles/pharmacology
9.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 428-437, 2015.
Article in English | WPRIM | ID: wpr-812525

ABSTRACT

Marsdenia tenacissima extract (MTE, trade name: Xiao-Ai-Ping injection) is an extract of a single Chinese plant medicine. It has been used for the treatment of cancer in China for decades, especially for esophageal cancer and other cancers in the digestive tract. In the present study, the potential mechanism for MTE's activity in esophageal cancer was explored. The effects of MTE on the proliferation of human esophageal cancer cells (KYSE150 and Eca-109) were investigated by the MTT assay, the BrdU (bromodeoxyuridine) incorporation immunofluorescence assay, and flow cytometric analysis. MTE inhibited cell proliferation through inducing G0/G1 cell cycle arrest in KYSE150 and Eca-109. Western blot analysis was employed to determine protein levels in the MTE treated cells. Compared with the control cells, the expression levels of the cell cycle regulatory proteins cyclin D1/D2/D3, cyclin E1, CDK2/4/6 (CDK: cyclin dependent kinase), and p-Rb were decreased significantly in the cells treated with MTE at 40 mg·mL(-1). In addition, MTE had an inhibitory effect on the MAPK (mitogen-activated protein kinase) signal transduction pathway, including ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), and p38MAPK. Moreover, MTE showed little additional effects on the regulation of cyclin D1/D3, CDK4/6, and p-Rb when the ERK pathway was already inhibited by the specific ERK inhibitor U0126. In conclusion, these data suggest that MTE inhibits human esophageal cancer cell proliferation through regulation of cell cycle regulatory proteins and the MAPK signaling pathways, which is probably mediated by the inhibition of ERK activation.


Subject(s)
Humans , Apoptosis , Carcinoma , Drug Therapy , Cell Line, Tumor , Cell Proliferation , Drugs, Chinese Herbal , Pharmacology , Esophageal Neoplasms , Drug Therapy , Extracellular Signal-Regulated MAP Kinases , Metabolism , G1 Phase Cell Cycle Checkpoints , MAP Kinase Signaling System , Marsdenia , Chemistry
10.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 672-678, 2015.
Article in English | WPRIM | ID: wpr-250360

ABSTRACT

Liver fibrosis is an important health problem that can further progress into cirrhosis or liver cancer, and result in significant morbidity and mortality. Inhibiting proliferation and inducing apoptosis of hepatic stellate cells (HSCs) may be the key point to reverse liver fibrosis. At present, anti-fibrosis drugs are rare. Kinetin is a type of plant-derived cytokinin which has been reported to control differentiation and induce apoptosis of human cells. In this study, the HSCs were incubated with different concentrations of kinetin. The proliferation of rat HSCs was measured by MTT assay, cell cycle and apoptosis were analyzed by flow cytometry, and the apoptosis was examined by TUNEL method. The expression of Bcl-2 and Bax proteins was detected by immunocytochemistry staining. It was found that kinetin could markedly inhibit proliferation of HSCs. In a concentration range of 2 to 8 μg/mL, the inhibitory effects of kinetin on proliferation of HSCs were increased with the increased concentration and the extension of time (P < 0.01). Flow cytometry indicated that kinetin could inhibit the DNA synthesis from G0/G1 to S phase in a dose-dependent manner (P < 0.01). The apoptosis rates of the HSCs treated with 8, 4 and 2 μg/mL kinetin (25.62% ± 2.21%, 15.31% ± 1.9% and 6.18% ± 1.23%, respectively) were increased significantly compared with the control group (3.81% ± 0.93%) (P < 0.01). All the DNA frequency histogram in kinetin-treated groups showed obvious hypodiploid peak (sub-G1 peak), and with the increase of kinetin concentrations, the apoptosis rate of HSCs also showed a trend of increase. It was also found that kinetin could down-regulate the expression of Bcl-2, and up-regulate the expression of Bax, leading to the decreased ratio of Bcl-2/Bax significantly. The kinetin-induced apoptosis of HSCs was positively correlated with the expression of Bax, and negatively with the expression of Bcl-2. It was concluded that kinetin can inhibit activation and proliferation of HSCs by interrupting the cell cycle at G1/S restriction point and inducing apoptosis of HSCs via reducing the ratio of Bcl-2/Bax.


Subject(s)
Animals , Rats , Apoptosis , Cell Line, Transformed , Cell Proliferation , Dose-Response Relationship, Drug , G1 Phase Cell Cycle Checkpoints , Genetics , Gene Expression Regulation , Growth Inhibitors , Pharmacology , Hepatic Stellate Cells , Cell Biology , Metabolism , Kinetin , Pharmacology , Proto-Oncogene Proteins c-bcl-2 , Genetics , Metabolism , Signal Transduction , bcl-2-Associated X Protein , Genetics , Metabolism
11.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 700-706, 2015.
Article in English | WPRIM | ID: wpr-250355

ABSTRACT

Various kinds of schiff base metal complexes have been proven to induce apoptosis of tumor cells. However, it remains largely unknown whether schiff base zinc complexes induce apoptosis in human cancer cells. Here, we synthesized a novel schiff base zinc coordination compound (SBZCC) and investigated its effects on the growth, proliferation and apoptosis of human osteosarcoma MG-63 cells. A novel SBZCC was synthesized by chemical processes and used to treat MG-63 cells. The cell viability was determined by CCK-8 assay. The cell cycle progression, mitochondrial membrane potential and apoptotic cells were analyzed by flow cytometry. The apoptosis-related proteins levels were determined by immunoblotting. Treatment of MG-63 cells with SBZCC resulted in inhibition of cell proliferation and cell cycle arrest at G1 phase. Moreover, SBZCC significantly reduced the mitochondrial membrane potential and induced apoptosis, accompanied with increased Bax/Bcl-2 and FlasL/Fas expression as well as caspase-3/8/9 cleavage. Our results demonstrated that the synthesized novel SBZCC could inhibit the proliferation and induce apoptosis of MG-63 cells via activating both the mitochondrial and cell death receptor apoptosis pathways, suggesting that SBZCC is a promising agent for the development as anticancer drugs.


Subject(s)
Humans , Antineoplastic Agents , Pharmacology , Apoptosis , Caspase 3 , Genetics , Metabolism , Caspase 8 , Genetics , Metabolism , Caspase 9 , Genetics , Metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Coordination Complexes , Pharmacology , Fas Ligand Protein , Genetics , Metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Membrane Potential, Mitochondrial , Mitochondria , Metabolism , Pathology , Osteoblasts , Metabolism , Pathology , Proto-Oncogene Proteins c-bcl-2 , Genetics , Metabolism , Schiff Bases , Chemistry , Signal Transduction , Zinc , Chemistry , bcl-2-Associated X Protein , Genetics , Metabolism , fas Receptor , Genetics , Metabolism
12.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 730-735, 2015.
Article in English | WPRIM | ID: wpr-250350

ABSTRACT

Bmi1 is a member of the polycomb group family of proteins, and it drives the carcinogenesis of various cancers and governs the self-renewal of multiple types of stem cells. However, its role in the initiation and progression of bladder cancer is not clearly known. The present study aimed to investigate the function of Bmi1 in the development of bladder cancer. Bmi1 expression was detected in human bladder cancer tissues and their adjacent normal tissues (n=10) by immunohistochemistry, qRT-PCR and Western blotting, respectively. Bmi1 small interference RNA (siRNA) was synthesized and transfected into human bladder carcinoma cells (EJ) by lipofectamine 2000. The Bmil expression at mRNA and protein levels was measured in EJ cells transfected with Bmil siRNA (0, 80, 160 nmol/L) by qRT-PCR and Western blotting, respectively. Cell viability and Ki67 expression (a marker of cell proliferation) were determined in Bmi1 siRNA-transfected cells by CCK-8 assay and qRT-PCR, respectively. Cell cycle of transfected cells was flow-cytometrically determined. Immunofluorescence and Western blotting were used to detect the expression levels of cell cycle-associated proteins cyclin D1 and cyclin E in the cells. Pro-apoptotic proteins Bax and caspase 3 and anti-apoptotic protein Bcl-2 were detected by Western blotting as well. Additionally, xenograft tumor models were established by inoculation of EJ cells (infected with Bmil shRNA/pLKO.1 lentivirus or not) into nude mice. The tumor volumes were measured every other day for 14 days. The results showed that the Bmil expression was significantly increased in bladder tumor tissues when compared with that in normal tissues (P<0.05). Perturbation of Bmi1 expression by using siRNA could significantly inhibit the proliferation of EJ cells (P<0.05). Bmi1 siRNA-transfected EJ cells were accumulated in G1 phase and the expression levels of cyclin D1 and cyclin E were down-regulated. Bax and caspase-3 expression levels were significantly increased and Bcl-2 levels decreased after Bmi1 knockdown. Tumor volume was conspicuously reduced in mice injected with EJ cells with Bmi1 knockdown. Our findings indicate that Bmi1 is a potential driver oncogene of bladder cancer and it may become a potential treatment target for human bladder cancer.


Subject(s)
Animals , Humans , Mice , Apoptosis , Genetics , Carcinogenesis , Genetics , Metabolism , Pathology , Carcinoma , Genetics , Metabolism , Pathology , Therapeutics , Caspase 3 , Genetics , Metabolism , Cell Line, Tumor , Cyclin D1 , Genetics , Metabolism , Cyclin E , Genetics , Metabolism , G1 Phase Cell Cycle Checkpoints , Genetics , Gene Expression Regulation, Neoplastic , Injections, Intralesional , Ki-67 Antigen , Genetics , Metabolism , Mice, Nude , Polycomb Repressive Complex 1 , Genetics , Metabolism , Proto-Oncogene Proteins c-bcl-2 , Genetics , Metabolism , RNA, Small Interfering , Genetics , Metabolism , Signal Transduction , Tumor Burden , Urinary Bladder , Metabolism , Pathology , Urinary Bladder Neoplasms , Genetics , Metabolism , Pathology , Therapeutics , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein , Genetics , Metabolism
13.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 736-741, 2015.
Article in English | WPRIM | ID: wpr-250349

ABSTRACT

In this study, one immortalized human normal prostatic epithelial cell line (BPH) and four human prostate cancer cell lines (LNCaP, 22Rv1, PC-3, and DU-145) were treated with Ganoderma Lucidum triterpenoids (GLT) at different doses and for different time periods. Cell viability, apoptosis, and cell cycle were analyzed using flow cytometry and chemical assays. Gene expression and binding to DNA were assessed using real-time PCR and Western blotting. It was found that GLT dose-dependently inhibited prostate cancer cell growth through induction of apoptosis and cell cycle arrest at G1 phase. GLT-induced apoptosis was due to activation of Caspases-9 and -3 and turning on the downstream apoptotic events. GLT-induced cell cycle arrest (mainly G1 arrest) was due to up-regulation of p21 expression at the early time and down-regulation of cyclin-dependent kinase 4 (CDK4) and E2F1 expression at the late time. These findings demonstrate that GLT suppresses prostate cancer cell growth by inducing growth arrest and apoptosis, which might suggest that GLT or Ganoderma Lucidum could be used as a potential therapeutic drug for prostate cancer.


Subject(s)
Humans , Male , Antineoplastic Agents, Phytogenic , Pharmacology , Apoptosis , Caspase 3 , Genetics , Metabolism , Caspase 9 , Genetics , Metabolism , Cell Line, Tumor , Cell Survival , Cyclin D1 , Genetics , Metabolism , Cyclin-Dependent Kinase 4 , Genetics , Metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Genetics , Metabolism , Dose-Response Relationship, Drug , E2F1 Transcription Factor , Genetics , Metabolism , G1 Phase Cell Cycle Checkpoints , Genetics , Gene Expression Regulation, Neoplastic , Nucleosomes , Metabolism , Pathology , Plant Extracts , Chemistry , Prostate , Metabolism , Pathology , Reishi , Chemistry , Signal Transduction , Triterpenes , Pharmacology
14.
Nutrition Research and Practice ; : 111-116, 2015.
Article in English | WPRIM | ID: wpr-204524

ABSTRACT

BACKGROUND/OBJECTIVES: Inonotus obliquus (I. obliquus, Chaga mushroom) has long been used as a folk medicine to treat cancer. In the present study, we examined whether or not ethanol extract of I. obliquus (EEIO) inhibits cell cycle progression in HT-29 human colon cancer cells, in addition to its mechanism of action. MATERIALS/METHODS: To examine the effects of Inonotus obliquus on the cell cycle progression and the molecular mechanism in colon cancer cells, HT-29 human colon cancer cells were cultured in the presence of 2.5 - 10 microg/mL of EEIO, and analyzed the cell cycle arrest by flow cytometry and the cell cycle controlling protein expression by Western blotting. RESULTS: Treatment cells with 2.5 - 10 microg/mL of EEIO reduced viable HT-29 cell numbers and DNA synthesis, increased the percentage of cells in G1 phase, decreased protein expression of CDK2, CDK4, and cyclin D1, increased expression of p21, p27, and p53, and inhibited phosphorylation of Rb and E2F1 expression. Among I. obliquus fractions, fraction 2 (fractionated by dichloromethane from EEIO) showed the same effect as EEIO treatment on cell proliferation and cell cycle-related protein levels. CONCLUSIONS: These results demonstrate that fraction 2 is the major fraction that induces G1 arrest and inhibits cell proliferation, suggesting I. obliquus could be used as a natural anti-cancer ingredient in the food and/or pharmaceutical industry.


Subject(s)
Humans , Blotting, Western , Cell Cycle , Cell Cycle Checkpoints , Cell Proliferation , Colonic Neoplasms , Cyclin D1 , DNA , Drug Industry , Ethanol , Flow Cytometry , G1 Phase , G1 Phase Cell Cycle Checkpoints , HT29 Cells , Medicine, Traditional , Methylene Chloride , Phosphorylation
15.
Clinical and Molecular Hepatology ; : 257-267, 2015.
Article in English | WPRIM | ID: wpr-157202

ABSTRACT

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, and it has a poor prognosis and few therapeutic options. Radiotherapy is one of the most effective forms of cancer treatment, and P53 protein is one of the key molecules determining how a cell responds to radiotherapy. The aim of this study was to determine the therapeutic efficacy of iodine-131 in three human HCC cell lines. METHODS: Western blotting was used to measure P53 expression. The effects of radiotherapy with iodine-131 were assessed by using the clonogenic assay to evaluate cell survival. Flow cytometry was carried out to examine the effects of iodine-131 on cell death, oxidative stress, reduced intracellular glutathione expression, the mitochondrial membrane potential, and the cell cycle. RESULTS: The P53 protein was not expressed in Hep3B2.1-7 cells, was expressed at normal levels in HepG2 cells, and was overexpressed in HuH7 cells. P53 expression in the HuH7 and HepG2 cell lines increased after internal and external irradiation with iodine-131. Irradiation induced a decrease in cell survival and led to a decrease in cell viability in all of the cell lines studied, accompanied by cell death via late apoptosis/necrosis and necrosis. Irradiation with 131-iodine induced mostly cell-cycle arrest in the G0/G1 phase. CONCLUSIONS: These results suggest that P53 plays a key role in the radiotherapy response of HCC.


Subject(s)
Humans , Apoptosis/radiation effects , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Gamma Rays , Glutathione/metabolism , Hep G2 Cells , Iodine Radioisotopes/chemistry , Liver Neoplasms/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Journal of Southern Medical University ; (12): 338-342, 2015.
Article in Chinese | WPRIM | ID: wpr-239181

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the expression of miR-223 in clear cell renal cell carcinoma (ccRcc) and its clinical implications.</p><p><b>METHODS</b>Quantitative real-time PCR was employed to detect the levels of miR- 223 expression in ccRcc, pair-matched adjacent normal tissues and different renal cancer cell lines. Transwell migration essay and wound healing essay were used to evaluate the invasion and migration of renal cancer 786-O cells transfected with miR-223 mimics. MTT essay was used to measure the cell proliferation, and the cell cycle changes following the transfection were analyzed with flow cytometry.</p><p><b>RESULTS</b>Compared with the normal tissues, the cancer samples showed up-regulated miR-223 expression, which was associated with tumor size. In 786-O cell cultures, transfection with miR-223 mimics significantly enhanced cell migration (P<0.0001) and growth (P=0.006) and induced G1 cell cycle arrest.</p><p><b>CONCLUSION</b>miR-223 promotes renal cancer cell migration and proliferation and may serve as a potential therapeutic target for ccRcc.</p>


Subject(s)
Humans , Carcinoma, Renal Cell , Metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , MicroRNAs , Metabolism , Real-Time Polymerase Chain Reaction , Transfection , Up-Regulation
17.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 370-375, 2014.
Article in English | WPRIM | ID: wpr-351069

ABSTRACT

Although previous reports showed drug-eluting stent (DES) could effectively inhibit neointima formation, in-stent restenosis (ISR) remains an important obstacle. The purpose of this study was to investigate different effects of paclitaxel on proliferation and cell cycle regulators between vascular smooth muscle cells (VSMCs) and vascular endothelial cells (VECs) of rats in vitro. The cultured VSMCs and VECs of rats from the same tissues were examined by using immunohistochemistry, flow cytometry and Western blotting in control and paclitaxel-treated groups. The results showed paclitaxel could effectively inhibit proliferation of VSMCs and VECs. However, as compared with VECs, proliferation of VSMCs in paclitaxel-treated group decreased less rapidly. The percentage of cells in G0-G1 and G2-M phases was reduced, and that in S phase increased after treatment for 72 h. The expression of cyclin D1 and B1, p27 and PCNA in VSMCs of paclitaxel-treated group was up-regulated, but that of p21 down-regulated as compared with VECs. It is concluded that there are significant differences in the expression of cell cycle regulators and proliferation rate between paclitaxel-treated VSMCs and paclitaxel-treated VECs, suggesting that the G1-S checkpoint regulated by paclitaxel may play a critical role in the development of complications of DES, which provides new strategies for treatments of ISR.


Subject(s)
Animals , Rats , Blotting, Western , Cell Cycle , Cell Cycle Proteins , Metabolism , Cell Proliferation , Cells, Cultured , Cyclin B1 , Metabolism , Cyclin D1 , Metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Metabolism , Endothelial Cells , Metabolism , Flow Cytometry , G1 Phase Cell Cycle Checkpoints , Immunohistochemistry , Microscopy, Fluorescence , Muscle, Smooth, Vascular , Cell Biology , Myocytes, Smooth Muscle , Metabolism , Paclitaxel , Pharmacology , Proliferating Cell Nuclear Antigen , Metabolism , Tubulin Modulators , Pharmacology
18.
Biol. Res ; 47: 1-13, 2014. ilus, graf, tab
Article in English | LILACS | ID: biblio-950750

ABSTRACT

BACKGROUND: Zanthoxylum heitzii is a spice used to prepare several dishes and to treat tumors, syphilis, malaria, cardiac palpitations, urogenital infections in the west region of Cameroon, but the antitumor mechanisms and chemical composition are not yet investigated. This study was aimed to determine the antiproliferative effects of four extracts from the fruits and barks of Zanthoxyllum heitzii (Rutaceae) on apoptosis in human promyelocytic cells, their mechanisms and the chemical composition. The 3-(4, 5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to determine the fifty percent inhibition (IC50) concentration of the cell lines after treatment. The effect on morphology was observed using a light or fluorescence microscopy. The rate of apoptosis and the cell cycle were measured using flow cytometry (FCM). The phytochemical analysis of the extract was carried with HPLC/MS methods. RESULTS: The phytochemical analysis of the extracts indicated the presence of four known polyphenols (Syringic acid, Juglon, Luteolin and Myricetin) in both fruits and barks of Z. heitzii but in different quantities. Syringic acid and Myricetin concentrations were between 17-21 fold higher in the fruits than the stem bark. Rhamnetin (393.35 µg/mL) and Oleuropein (63.10 µg/mL) were identified only in the stem barks of Z. heitzii. Among the four extracts tested for cytotoxicity properties, only the methanol extract of fruits and barks significantly inhibited cell proliferation of HL-60 cells with IC50 value of 20 µg/mL and 12 µg/mL respectively. HL-60 cells treated with Z. heitzii extracts significantly produced reactive oxygen species (ROS) with concurrent loss of mitochondrial membrane potential (MMP). Modifications in the DNA distribution and enhanced of G1/G0 phase cell cycle arrest were observed in a concentration dependent manner. CONCLUSIONS: Polyphenols from Z. heitzii plant exert inhibitory effect on HL-60 cells through the reactive oxygen species (ROS) generation, loss of mitochondrial membrane potential and cell cycle destabilization.


Subject(s)
Humans , Apoptosis/drug effects , Plant Bark/chemistry , Zanthoxylum/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , Fruit/chemistry , Mitochondria/physiology , Mass Spectrometry , Tetrazolium Salts , Thiazoles , Cameroon , Plant Extracts/isolation & purification , Plant Extracts/chemistry , Chromatography, High Pressure Liquid , Spices/analysis , Reactive Oxygen Species/analysis , HL-60 Cells , Inhibitory Concentration 50 , Cell Proliferation/drug effects , Membrane Potential, Mitochondrial/drug effects , Polyphenols/analysis , Flow Cytometry , Microscopy, Fluorescence
19.
Braz. j. med. biol. res ; 46(8): 643-649, ago. 2013. graf
Article in English | LILACS | ID: lil-684525

ABSTRACT

MP [4-(3′,3′-dimethylallyloxy)-5-methyl-6-methoxyphthalide] was obtained from liquid culture of Pestalotiopsis photiniae isolated from the Chinese Podocarpaceae plant Podocarpus macrophyllus. MP significantly inhibited the proliferation of HeLa tumor cell lines. After treatment with MP, characteristic apoptotic features such as DNA fragmentation and chromatin condensation were observed in DAPI-stained HeLa cells. Flow cytometry showed that MP induced G1 cell cycle arrest and apoptosis in a dose-dependent manner. Western blotting and real-time reverse transcription-polymerase chain reaction were used to investigate protein and mRNA expression. MP caused significant cell cycle arrest by upregulating the cyclin-dependent kinase inhibitor p27KIP1 protein and p21CIP1 mRNA levels in HeLa cells. The expression of p73 protein was increased after treatment with various MP concentrations. mRNA expression of the cell cycle-related genes, p21CIP1 , p16INK4a and Gadd45α, was significantly upregulated and mRNA levels demonstrated significantly increased translation of p73, JunB, FKHR, and Bim. The results indicate that MP may be a potential treatment for cervical cancer.


Subject(s)
Humans , Apoptosis/drug effects , Benzofurans/administration & dosage , Endophytes/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , Xylariales/chemistry , Apoptosis Regulatory Proteins/genetics , Benzofurans/isolation & purification , Cell Cycle Proteins/drug effects , Cell Proliferation/drug effects , /drug effects , /drug effects , DNA-Binding Proteins/drug effects , Flow Cytometry , Forkhead Transcription Factors/drug effects , Cycadopsida , /drug effects , HeLa Cells , Nuclear Proteins/drug effects , Real-Time Polymerase Chain Reaction , Transcription, Genetic , Transcription Factors/drug effects , Tumor Suppressor Proteins/drug effects
20.
Braz. j. med. biol. res ; 46(6): 546-554, 02/jul. 2013. tab, graf
Article in English | LILACS | ID: lil-679208

ABSTRACT

Multidrug resistance (MDR) poses a serious impediment to the success of chemotherapy for laryngeal cancer. To identify microRNAs and mRNAs associated with MDR of human laryngeal cancer Hep-2 cells, we developed a multidrug-resistant human laryngeal cancer subline, designated Hep-2/v, by exposing Hep-2 cells to stepwise increasing concentrations of vincristine (0.02-0.96'µM). Microarray assays were performed to compare the microRNA and mRNA expression profiles of Hep-2 and Hep-2/v cells. Compared to Hep-2 cells, Hep-2/v cells were more resistant to chemotherapy drugs (∼45-fold more resistant to vincristine, 5.1-fold more resistant to cisplatin, and 5.6-fold more resistant to 5-fluorouracil) and had a longer doubling time (42.33±1.76 vs 28.75±1.12'h, P<0.05), higher percentage of cells in G0/G1 phase (80.98±0.52 vs 69.14±0.89, P<0.05), increased efflux of rhodamine 123 (95.97±0.56 vs 12.40±0.44%, P<0.01), and up-regulated MDR1 expression. A total of 7 microRNAs and 605 mRNAs were differentially expressed between the two cell types. Of the differentially expressed mRNAs identified, regulator of G-protein signaling 10, high-temperature requirement protein A1, and nuclear protein 1 were found to be the putative targets of the differentially expressed microRNAs identified. These findings may open a new avenue for clarifying the mechanisms responsible for MDR in laryngeal cancer.


Subject(s)
Humans , Drug Resistance, Neoplasm/genetics , Laryngeal Neoplasms/genetics , MicroRNAs/isolation & purification , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , RNA, Messenger/isolation & purification , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Flow Cytometry , Fluorouracil/pharmacology , G1 Phase Cell Cycle Checkpoints , Genes, MDR , Laryngeal Neoplasms/drug therapy , Neoplasm Proteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , RGS Proteins/genetics , /pharmacokinetics , Serine Endopeptidases/genetics , Tissue Array Analysis , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL