1.Helicobacter pylori inhibited cell proliferation in human periodontal ligament fibroblasts through the Cdc25C/CDK1/cyclinB1 signaling cascade
Huanying LI ; Dongsheng LIANG ; Naiming HU ; Xingzhu DAI ; Jianing HE ; Hongmin ZHUANG ; Wanghong ZHAO
Journal of Periodontal & Implant Science 2019;49(3):138-147
		                        		
		                        			
		                        			PURPOSE: Several studies have shown that the oral cavity is a secondary location for Helicobacter pylori colonization and that H. pylori is associated with the severity of periodontitis. This study investigated whether H. pylori had an effect on the periodontium. We established an invasion model of a standard strain of H. pylori in human periodontal ligament fibroblasts (hPDLFs), and evaluated the effects of H. pylori on cell proliferation and cell cycle progression. METHODS: Different concentrations of H. pylori were used to infect hPDLFs, with 6 hours of co-culture. The multiplicity of infection in the low- and high-concentration groups was 10:1 and 100:1, respectively. The Cell Counting Kit-8 method and Ki-67 immunofluorescence were used to detect cell proliferation. Flow cytometry, quantitative real-time polymerase chain reaction, and western blots were used to detect cell cycle progression. In the high-concentration group, the invasion of H. pylori was observed by transmission electron microscopy. RESULTS: It was found that H. pylori invaded the fibroblasts, with cytoplasmic localization. Analyses of cell proliferation and flow cytometry showed that H. pylori inhibited the proliferation of periodontal fibroblasts by causing G2 phase arrest. The inhibition of proliferation and G2 phase arrest were more obvious in the high-concentration group. In the low-concentration group, the G2 phase regulatory factors cyclin dependent kinase 1 (CDK1) and cell division cycle 25C (Cdc25C) were upregulated, while cyclin B1 was inhibited. However, in the high-concentration group, cyclin B1 was upregulated and CDK1 was inhibited. Furthermore, the deactivated states of tyrosine phosphorylation of CDK1 (CDK1-Y15) and serine phosphorylation of Cdc25C (Cdc25C-S216) were upregulated after H. pylori infection. CONCLUSIONS: In our model, H. pylori inhibited the proliferation of hPDLFs and exerted an invasive effect, causing G2 phase arrest via the Cdc25C/CDK1/cyclin B1 signaling cascade. Its inhibitory effect on proliferation was stronger in the high-concentration group.
		                        		
		                        		
		                        		
		                        			Blotting, Western
		                        			;
		                        		
		                        			CDC2 Protein Kinase
		                        			;
		                        		
		                        			Cell Count
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Coculture Techniques
		                        			;
		                        		
		                        			Colon
		                        			;
		                        		
		                        			Cyclin B1
		                        			;
		                        		
		                        			Cytoplasm
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			Flow Cytometry
		                        			;
		                        		
		                        			Fluorescent Antibody Technique
		                        			;
		                        		
		                        			G2 Phase
		                        			;
		                        		
		                        			Helicobacter pylori
		                        			;
		                        		
		                        			Helicobacter
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Methods
		                        			;
		                        		
		                        			Microscopy, Electron, Transmission
		                        			;
		                        		
		                        			Mouth
		                        			;
		                        		
		                        			Periodontal Ligament
		                        			;
		                        		
		                        			Periodontitis
		                        			;
		                        		
		                        			Periodontium
		                        			;
		                        		
		                        			Phosphorylation
		                        			;
		                        		
		                        			Real-Time Polymerase Chain Reaction
		                        			;
		                        		
		                        			Serine
		                        			;
		                        		
		                        			Tyrosine
		                        			
		                        		
		                        	
2.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*
		                        			
		                        		
		                        	
3.Generation of knockout mouse models of cyclin-dependent kinase inhibitors by engineered nuclease-mediated genome editing.
Bo Min PARK ; Jae il ROH ; Jaehoon LEE ; Han Woong LEE
Laboratory Animal Research 2018;34(4):264-269
		                        		
		                        			
		                        			Cell cycle dysfunction can cause severe diseases, including neurodegenerative disease and cancer. Mutations in cyclin-dependent kinase inhibitors controlling the G1 phase of the cell cycle are prevalent in various cancers. Mice lacking the tumor suppressors p16(Ink4a) (Cdkn2a, cyclin-dependent kinase inhibitor 2a), p19(Arf) (an alternative reading frame product of Cdkn2a,), and p27(Kip1) (Cdkn1b, cyclin-dependent kinase inhibitor 1b) result in malignant progression of epithelial cancers, sarcomas, and melanomas, respectively. Here, we generated knockout mouse models for each of these three cyclin-dependent kinase inhibitors using engineered nucleases. The p16(Ink4a) and p19(Arf) knockout mice were generated via transcription activator-like effector nucleases (TALENs), and p27(Kip1) knockout mice via clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9). These gene editing technologies were targeted to the first exon of each gene, to induce frameshifts producing premature termination codons. Unlike preexisting embryonic stem cell-based knockout mice, our mouse models are free from selectable markers or other external gene insertions, permitting more precise study of cell cycle-related diseases without confounding influences of foreign DNA.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			Codon, Nonsense
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase Inhibitor p16
		                        			;
		                        		
		                        			DNA
		                        			;
		                        		
		                        			Exons
		                        			;
		                        		
		                        			G1 Phase
		                        			;
		                        		
		                        			Genome*
		                        			;
		                        		
		                        			Melanoma
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Knockout*
		                        			;
		                        		
		                        			Mutagenesis, Insertional
		                        			;
		                        		
		                        			Neurodegenerative Diseases
		                        			;
		                        		
		                        			Phosphotransferases*
		                        			;
		                        		
		                        			Reading Frames
		                        			;
		                        		
		                        			Sarcoma
		                        			
		                        		
		                        	
4.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
		                        			
		                        		
		                        	
5.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
		                        			
		                        		
		                        	
6.Effect of RAD18-siRNA on proliferation and chemotherapy sensitivity of human esophageal squamous cell carcinoma ECA-109 cells.
Pengrong LOU ; Xiaonan SUN ; Jundong ZHOU ; Shitao ZOU
Journal of Zhejiang University. Medical sciences 2016;45(4):364-370
		                        		
		                        			
		                        			                    
To investigate the effect of RAD18-siRNA on cell proliferation and chemotherapy sensitivity of esophageal squamous cell carcinoma (ESCC) ECA-109 cells.RAD18-siRNA was transfected into human ECA-109 cells by Lipofectamine 3000. Quantitative PCR and Western blot were performed to detect RAD18 and CyclinD1 expression; CCK-8 assay was used to determine cell proliferation and chemotherapy drug sensitivity; flow cytometry was used to determine cell cycle. Correlation between RAD18 and CyclinD1 mRNA expression was analyzed by Pearson's correlation.Compared with non-transfected cells, the expression of RAD18 in RAD18-siRNA group was significantly decreased (<0.05). The cell proliferation was inhibited (<0.05) and the cell number of G1 phase was increased, G2/M phase cells decreased (<0.05) in RAD18-siRNA group. After treatment with different concentrations of cisplatin or 5-FU, the survival rate of the two cell groups was reduced (all<0.05), and the IC50 of RAD18-siRNA group was significantly lower than that of non-transfected group (<0.05). The mRNA expression of RAD18 was positively correlated with CyclinD1 expression in ESCC tissues(=0.478,<0.01).Down-regulated expression of RAD18 can decrease the cell proliferation and increase chemo-sensitivity of ESCC cells, and CyclinD1 may participate in the process.
		                        		
		                        		
		                        		
		                        			Adjuvants, Pharmaceutic
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cisplatin
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Cyclin D1
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			DNA-Binding Proteins
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Down-Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Drug Screening Assays, Antitumor
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Drug Synergism
		                        			;
		                        		
		                        			Esophageal Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Fluorouracil
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			G1 Phase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			G2 Phase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Metaphase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			RNA, Small Interfering
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Transfection
		                        			;
		                        		
		                        			Ubiquitin-Protein Ligases
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
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.Novel matrine derivative MD-1 attenuates hepatic fibrosis by inhibiting EGFR activation of hepatic stellate cells.
Yi FENG ; Hai-Yan YING ; Ying QU ; Xiao-Bo CAI ; Ming-Yi XU ; Lun-Gen LU
Protein & Cell 2016;7(9):662-672
		                        		
		                        			
		                        			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.
		                        		
		                        		
		                        		
		                        			Alkaloids
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Line
		                        			;
		                        		
		                        			Cyclin D1
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Dimethylnitrosamine
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Enzyme Activation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			ErbB Receptors
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			G1 Phase Cell Cycle Checkpoints
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Hepatic Stellate Cells
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Liver Cirrhosis
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			prevention & control
		                        			;
		                        		
		                        			Phosphorylation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Quinolizines
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Rats
		                        			
		                        		
		                        	
9.Total triterpenoids from Ganoderma Lucidum suppresses prostate cancer cell growth by inducing growth arrest and apoptosis.
Tao WANG ; Zi-ping XIE ; Zhan-sen HUANG ; Hao LI ; An-yang WEI ; Jin-ming DI ; Heng-jun XIAO ; Zhi-gang ZHANG ; Liu-hong CAI ; Xin TAO ; Tao QI ; Di-ling CHEN ; Jun CHEN
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):736-741
		                        		
		                        			
		                        			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.
		                        		
		                        		
		                        		
		                        			Antineoplastic Agents, Phytogenic
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Caspase 3
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Caspase 9
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			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
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Nucleosomes
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Plant Extracts
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Prostate
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Reishi
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Triterpenes
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
10.Knockdown of Bmi1 inhibits bladder cancer cell growth both in vitro and in vivo by blocking cell cycle at G1 phase and inducing apoptosis.
Hong-bo LUO ; Bin LI ; Wei-gang YUAN ; Chuan-rui XU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):730-735
		                        		
		                        			
		                        			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.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Carcinogenesis
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Carcinoma
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			therapy
		                        			;
		                        		
		                        			Caspase 3
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cyclin D1
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cyclin E
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			G1 Phase Cell Cycle Checkpoints
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Injections, Intralesional
		                        			;
		                        		
		                        			Ki-67 Antigen
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			Polycomb Repressive Complex 1
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-bcl-2
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			RNA, Small Interfering
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Tumor Burden
		                        			;
		                        		
		                        			Urinary Bladder
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Urinary Bladder Neoplasms
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			therapy
		                        			;
		                        		
		                        			Xenograft Model Antitumor Assays
		                        			;
		                        		
		                        			bcl-2-Associated X Protein
		                        			;
		                        		
		                        			agonists
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail