1.Antioxidant xanthones and anthraquinones isolated from a marine-derived fungus Aspergillus versicolor.
Ze-Hong WU ; Dong LIU ; Ying XU ; Jian-Liang CHEN ; Wen-Han LIN
Chinese Journal of Natural Medicines (English Ed.) 2018;16(3):219-224
		                        		
		                        			
		                        			Chemical examination of an EtOAc extract of cultured Aspergillus versicolor fungus from deep-sea sediments resulted in the isolation of four xanthones, eight anthraquinones and five alkaloids, including a new xanthone, oxisterigmatocystin D (1) and a new alkaloid, aspergillusine A (13). High resolution electron impact mass spectrometry (HR-EI-MS), FT-IR spectroscopy, and NMR techniques were used to elucidate the structures of these compounds, and the absolute configuration of compound 1 was established by its NMR features and coupling constant. Furthermore, the biosynthesis pathway of these xanthones and anthraquinones were deduced, and their antioxidant activity and cytotoxicity in human cancer cell lines (HTC-8, Bel-7420, BGC-823, A549, and A2780) were evaluated. The trolox equivalent antioxidant capacity (TEAC) assay indicated most of the xanthones and anthraquinones possessing moderate antioxidant activities. The Nrf2-dependent luciferase reporter gene assay revealed that compounds 6, 7, 9, and 12 potentially activated the expression of Nrf2-regulated gene. In addition, compounds 5 and 11 showed weak cytotoxicity on A with the IC values of 25.97 and 25.60 μmol·L, respectively.
		                        		
		                        		
		                        		
		                        			Anthraquinones
		                        			;
		                        		
		                        			Antioxidants
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Aspergillus
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Gene Expression
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Magnetic Resonance Spectroscopy
		                        			;
		                        		
		                        			Molecular Structure
		                        			;
		                        		
		                        			NF-E2-Related Factor 2
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Seawater
		                        			;
		                        		
		                        			microbiology
		                        			;
		                        		
		                        			Spectroscopy, Fourier Transform Infrared
		                        			;
		                        		
		                        			Xanthones
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
2.Saponins isolated from Schizocapsa plantaginea inhibit human hepatocellular carcinoma cell growth in vivo and in vitro via mitogen-activated protein kinase signaling.
Yue-Wen SUN ; Han-Chen QIU ; Ming-Chun OU ; Run-Li CHEN ; Gang LIANG
Chinese Journal of Natural Medicines (English Ed.) 2018;16(1):29-40
		                        		
		                        			
		                        			The underground cane of Schizocapsa plantaginea (Hance) has long been used by Chinese ethnic minority as a constituent of anti-cancer formulae. Saponins are abundant secondary metabolic products located in the underground cane of this plant. The potential therapeutic effects of total saponins isolated from Schizocapsa plantaginea (Hance) (SSPH) on human hepatocellular carcinoma (HCC) were tested in vitro in human liver cancer cell lines, SMMC-7721 and Bel-7404. Apoptosis and cell cycle arrest were determined using flow cytometry, caspase activation was determined by ELISA, and PARP, cleaved PARP, mitogen-activated protein kinase (MAPK) expression and phosphorylation were measured using Western blotting analysis. In vivo anti-HCC effects of SSPH were verified in nude mouse xenograft model. SSPH exerted markedly inhibitory effect on HCC cell proliferation in time- and concentration-dependent manner. Moreover, SSPH significantly induced apoptosis through caspase-dependent signaling and arrested cell cycle at G/M phase. These anti-proliferation effects of SSPH were associated with up-regulated phosphorylation of extracellular signal-regulated kinase-1/2 (Erk1/2) and c-jun-NH2-kinase-1/2 (JNK1/2) and reduced phosphorylation of p38MAPK. Furthermore, inhibitors of ERK, UO126, and JNK, SP600125 inhibited the anti-proliferation effects by SSPH, suggesting that Erk and JNK were the effector molecules in SSPH induced anti-proliferative action. During in vivo experiments, SSPH was found to inhibit xenograft tumor growth in nude mice, with a similar mechanism in vitro. Our study confirmed that SSPH exerted antagonistic effects on human liver cancer cells both in vitro and in vivo. Molecular mechanisms underlying SSPH action might be closely associated with MAPK signaling pathways. These results indicated that SSPH has potential therapeutic effects on HCC.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Caspases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Cycle Checkpoints
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Dioscoreaceae
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Heterografts
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			growth & development
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Inhibitory Concentration 50
		                        			;
		                        		
		                        			Liver Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			MAP Kinase Signaling System
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			Phosphorylation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Plant Tubers
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			Poly (ADP-Ribose) Polymerase-1
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Saponins
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			toxicity
		                        			
		                        		
		                        	
3.Reversal of multidrug resistance by icaritin in doxorubicin-resistant human osteosarcoma cells.
Zhen-Dong WANG ; Rui-Zhi WANG ; Yuan-Zheng XIA ; Ling-Yi KONG ; Lei YANG
Chinese Journal of Natural Medicines (English Ed.) 2018;16(1):20-28
		                        		
		                        			
		                        			Multidrug resistance (MDR) is one of the major obstacles in cancer chemotherapy. Our previous study has shown that icariin could reverse MDR in MG-63 doxorubicin-resistant (MG-63/DOX) cells. It is reported that icariin is usually metabolized to icariside II and icaritin. Herein, we investigated the effects of icariin, icariside II, and icaritin (ICT) on reversing MDR in MG-63/DOX cells. Among these compounds, ICT exhibited strongest effect and showed no obvious cytotoxicity effect on both MG-63 and MG-63/DOX cells ranging from 1 to 10 μmol·L. Furthermore, ICT increased accumulation of rhodamine 123 and 6-carboxyfluorescein diacetate and enhanced DOX-induced apoptosis in MG-63/DOX cells in a dose-dependent manner. Further studies demonstrated that ICT decreased the mRNA and protein levels of multidrug resistance protein 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1). We also verified that blockade of STAT3 phosphorylation was involved in the reversal effect of multidrug resistance in MG-63/DOX cells. Taken together, these results indicated that ICT may be a potential candidate in chemotherapy for osteosarcoma.
		                        		
		                        		
		                        		
		                        			ATP Binding Cassette Transporter, Subfamily B
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Dose-Response Relationship, Drug
		                        			;
		                        		
		                        			Doxorubicin
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Drug Resistance, Multiple
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Flavonoids
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Multidrug Resistance-Associated Proteins
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Osteosarcoma
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Phosphorylation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Rhodamine 123
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			STAT3 Transcription Factor
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Triterpenes
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
4.Effect of AG490 on JAK2/STAT3 signaling pathway in human retinoblastoma HXO-RB44 cell lines.
Bei XU ; Xiang CHEN ; Jia TAN ; Xueliang XU
Journal of Central South University(Medical Sciences) 2018;43(10):1061-1067
		                        		
		                        			
		                        			To investigate the role of Janus kinase (JAK) inhibitor AG490 in the anti-proliferation and cell cycle in human retinoblastoma HXO-RB44 cell lines in vitro, and to explore its effect on the expression of JAK2/signal transducer and activator of transcription 3 (STAT3).
 Methods: Cells were divided into an experiment group and a control group, and the experiment group was further divided into 6 sub-groups according to different AG490 concentrations (6.25, 12.50, 25.00, 50.00 or 100.00 μmol/L). Cell proliferation in the different groups was analyzed by cell vitality determination. Cell cycle distribution and apoptosis rate were examined by flow cytometry. The protein levels of STAT3, p-STAT3 and vascular endothelial growth factor (VEGF) were detected by Western blot.
 Results: After 48 h treatment with AG490, the viability of HXO-RB44 cells was reduced in a concentration-dependent manner. Compared with the control group, there was no significant difference in the experiment groups except the 6.25 μmol/L group (all P>0.05). The apoptosis rates in the experiment groups were significantly increased with increase in concentration of AG490 compared with that in the control group (all P<0.05). The cell ratio in the G1 phase in 50 or 100 μmol/L group was increased, whereas the cell ratio in the S phase was decreased. Western blot results showed that the expressions of STAT3 and p-STAT3 in the experiment groups were dramatically reduced with the increase in concentration of AG490 compared with that in the control group (all P<0.05). VEGF expression didn't obviously change in the experiment groups with AG490 concentration less than 12.5 μmol/L compared with that in the control group (both P>0.05), but there were significant differences in the other experiment groups (all P<0.05). 
 Conclusion: JAK inhibitor AG490 can inhibit proliferation and promote apoptosis of the retinoblastoma HXO-RB44 cells through down-regulation of JAK2/STAT3 signaling pathway.
		                        		
		                        		
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Enzyme Inhibitors
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Janus Kinase 2
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Retinoblastoma
		                        			;
		                        		
		                        			STAT3 Transcription Factor
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Tyrphostins
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Vascular Endothelial Growth Factor A
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
5.Effect of EGCG on oxidative stress and Nrf2/HO-1 pathway in neurons exposed to oxygen-glucose deprivation/reperfusion.
Fang HE ; Yi ZHANG ; Shang CHEN ; Bei YE ; Jianzhen CHEN ; Chang LI
Journal of Central South University(Medical Sciences) 2018;43(10):1041-1047
		                        		
		                        			
		                        			To explore the effect of epigallocatechin gallate (EGCG) on oxidative stress and Nrf2/HO-1 pathway in neurons subjected to oxygen-glucose deprivation/reperfusion (OGD/R).
 Methods: Primary cultured cerebral cortical neurons were prepared from Sprague-Dawley rats, and the OGD/R cell model was established. After pretreatment with EGCG at different concentrations (12.5, 25.0, 50.0 or 100.0 μmol/L), the neurons were subjected to OGD/R. The cell viability, reactive oxygen species (ROS) level and malondialdehyde (MDA) content were assessed after reperfusion. The superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities were measured. The expression of Nrf2 protein in nucleus, HO-1 mRNA and protein were detected.
 Results: OGD/R treatment reduced the cell viability, elevated ROS level and MDA content, decreased SOD and GSH-Px activities. The expression of Nrf2 protein in nucleus, HO-1 mRNA and protein were increased (P<0.01). Pretreatment with EGCG promoted the survival of neurons exposed to OGD/R, decreased ROS level and MDA content while increased SOD and GSH-Px activities. The levels of Nrf2 protein in nucleus, HO-1 mRNA and protein were upregulated (P<0.01).
 Conclusion: EGCG can reduce the oxidative stress of neurons subjected to OGD/R, which may be related to activation of Nrf2/HO-1 signal pathway and enhancement of the antioxidant ability of neurons.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Catechin
		                        			;
		                        		
		                        			analogs & derivatives
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Gene Expression Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Glucose
		                        			;
		                        		
		                        			Heme Oxygenase-1
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			NF-E2-Related Factor 2
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Neurons
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Neuroprotective Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Oxidative Stress
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Oxygen
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Reperfusion Injury
		                        			;
		                        		
		                        			prevention & control
		                        			
		                        		
		                        	
6.Shenmai injection protects mitochondria from oxidative injury in myocardial cells and its mechanism.
Yu ZHAO ; Feng ZHANG ; Xiaoping ZHAO ; Wei YUAN ; Jinhua ZHANG ; Yi WANG
Journal of Zhejiang University. Medical sciences 2018;47(5):507-513
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effect of Shenmai injection on myocardial cells with oxidative injury and the underlying mechanisms.
		                        		
		                        			METHODS:
		                        			Tert-butyl hydroperoxide (t-BHP) was used to induce the oxidative stress in H9c2 myocardial cells. The cell viability and ATP level were evaluated using MTT-colorimetric method and CellTiter-Glo luminescent cell viability assay. The oxygen respiration rate was examined by Clark oxygen electrode. Pyruvate and pyruvate dehydrogenase (PDH) levels were evaluated by ELISA kit. Western blot and quantitative real-time RT-PCR were employed to evaluate the expression of pyruvate dehydrogenase alpha 1(PDHA1) and pyruvate dehydrogenase kinase 1(PDK1).
		                        		
		                        			RESULTS:
		                        			Shenmai injection significantly improved viability and respiration of H9c2 myocardial cells after t-BHP injury (<0.05 or <0.01). It increased ATP contents by consuming pyruvate and increasing PDH level (<0.05 or <0.01). Furthermore, Shenmai injection had the tendency to increase protein expression of PDHA1(<0.05) and decrease mRNA expression of PDK1 (>0.05).
		                        		
		                        			CONCLUSIONS
		                        			Shenmai injection protects mitochondria from oxidative stress by increasing PDH level, which indicates that it may improve energy metabolism of myocardial cells.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Line
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Drug Combinations
		                        			;
		                        		
		                        			Drugs, Chinese Herbal
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Gene Expression Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Mitochondria
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Myocytes, Cardiac
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Oxidative Stress
		                        			;
		                        		
		                        			Protein-Serine-Threonine Kinases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Pyruvate Dehydrogenase (Lipoamide)
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Rats
		                        			
		                        		
		                        	
7.G protein-coupled receptor 17 is involved in CoCl-induced hypoxic injury in RGC-5 cells.
Kana LIN ; Meili LIN ; Yingfen GU ; Shunguo ZHANG ; Shiying HUANG
Journal of Zhejiang University. Medical sciences 2018;47(5):487-492
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effect of G protein-coupled receptor 17 (GPR17) on hypoxia injury in retinal ganglion cells .
		                        		
		                        			METHODS:
		                        			CoCl (400 μmol/L) was used to induce hypoxic injury in RGC-5 cells. The expression of GPR17 and the effect of GPR17 ligands were investigated, and the role of GPR17 in hypoxia injury was further studied by transfection of RGC-5 cells with GPR17 small interfering RNA (siRNA). The cell viability was determined by MTT and the cell apoptosis rate was detected by flow cytometry analysis. The expression of GPR17 mRNA was determined with RT-PCR.
		                        		
		                        			RESULTS:
		                        			mRNA expressions of GPR17 in RGC-5 cells with and without CoCl treatment were 0.36±0.05 and 0.26±0.08(<0.01). Compared with hypoxia without any treatment, pretreatment with GPR17 agonists (LTD, UDP, UDP-G) significantly reduced cell viability (the survival rates of cells decreased by 29.6%, 31.8% and 33.9%, all <0.01), while the effect of GPR17 antagonist (cangrelor) was the opposite (the survival rates of cells increased by 33.2%, <0.01). Transfection with GPR17 SiRNA inhibited hypoxia-induced up-expression of GPR17 mRNA (<0.01)and reduced cell apoptosis[rates of cell apoptosis were(39.73±2.06)%,(42.50±3.64)% and (24.98±2.16)% for blank control, NC siRNA and GPR17 siRNA groups, <0.01].
		                        		
		                        			CONCLUSIONS
		                        			GPR17 may mediate hypoxia injury in RGC-5 cells, while the knockdown of GPR17 can reduce the hypoxia injury.
		                        		
		                        		
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Cell Hypoxia
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Cell Line
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			Cobalt
		                        			;
		                        		
		                        			Gene Expression Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Gene Knockdown Techniques
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Hypoxia
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Receptors, G-Protein-Coupled
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Retinal Ganglion Cells
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
8.Role of PD 0332991 on the Proliferation and Apoptosis of Vascular Endothelial Cells.
Chenlong ZHAO ; Minghui LIU ; Yongwen LI ; Hongbing ZHANG ; Ying LI ; Hao GONG ; Yin YUAN ; Weiting LI ; Hongyu LIU ; Jun CHEN
Chinese Journal of Lung Cancer 2018;21(5):375-382
		                        		
		                        			BACKGROUND:
		                        			Angiogenesis is an important process in the development of tumor. PD 0332991, a cell cycle inhibitor, can specifically inhibit CD4/6 phosphorylation and cell cycle progression. In xeongraft mice models, PD 0332991 treated mice had significantly decreased angiogenesis and vascular density compared with the control group, but the mechanism remains unknown. The purpose of this study is to investigate the role and molecular mechanism of PD 0332991 on vascular endothelial cells.
		                        		
		                        			METHODS:
		                        			EA.hy926 cells, a kind of vascular endothelial cell, were used as the research model. The effects of PD 0332991 on the activity and proliferation of EA.hy926 cells were detected by the MTT, EdU assays. Wound-healing assays and transwell assays were used to determine the effects of PD 0332991 on the mobility of EA.hy926. The influence of PD 0332991 on cell cycle and apoptosis of endothelial cells was tested by flow cytometry, and the Western blot was applied to observe the expression of cell cycle related proteins in EA.hy926 cells treated by PD 0332991.
		                        		
		                        			RESULTS:
		                        			PD 0332991 significantly inhibited the proliferation and mobility of EA.hy926 cells, caused cell cycle arrest and apoptosis. At the same time, PD 0332991 inhibited the expression of CDK4/6 and phosphorylation of Rb, and thus inhibited the cell cycle progression of EA.hy926 cells.
		                        		
		                        			CONCLUSIONS
		                        			PD 0332991 can inhibit the proliferation and activity of endothelial cells and induces apoptosis.
		                        		
		                        		
		                        		
		                        			Angiogenesis Inhibitors
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Survival
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase 4
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase 6
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Endothelial Cells
		                        			;
		                        		
		                        			cytology
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Piperazines
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Pyridines
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
9.Novel nano-microspheres containing chitosan, hyaluronic acid, and chondroitin sulfate deliver growth and differentiation factor-5 plasmid for osteoarthritis gene therapy.
Zhu CHEN ; Shang DENG ; De-Chao YUAN ; Kang LIU ; Xiao-Cong XIANG ; Liang CHENG ; Dong-Qin XIAO ; Li DENG ; Gang FENG
Journal of Zhejiang University. Science. B 2018;19(12):910-923
		                        		
		                        			OBJECTIVE:
		                        			To construct a novel non-viral vector loaded with growth and differentiation factor-5 (GDF-5) plasmid using chitosan, hyaluronic acid, and chondroitin sulfate for osteoarthritis (OA) gene therapy.
		                        		
		                        			METHODS:
		                        			Nano-microspheres (NMPs) were prepared by mixing chitosan, hyaluronic acid, and chondroitin sulfate. GDF-5 plasmid was encapsulated in the NMPs through electrostatic adsorption. The basic characteristics of the NMPs were observed, and then they were co-cultured with chondrocytes to observe their effects on extracellular matrix (ECM) protein expression. Finally, NMPs loaded with GDF-5 were injected into the articular cavities of rabbits to observe their therapeutic effects on OA in vivo.
		                        		
		                        			RESULTS:
		                        			NMPs exhibited good physicochemical properties and low cytotoxicity. Their average diameter was (0.61±0.20) μm, and encapsulation efficiency was (38.19±0.36)%. According to Cell Counting Kit-8 (CCK-8) assay, relative cell viability was 75%-99% when the total weight of NMPs was less than 560 μg. Transfection efficiency was (62.0±2.1)% in a liposome group, and (60.0±1.8)% in the NMP group. There was no significant difference between the two groups (P>0.05). Immunohistochemical staining results suggested that NMPs can successfully transfect chondrocytes and stimulate ECM protein expression in vitro. Compared with the control groups, the NMP group significantly promoted the expression of chondrocyte ECM in vivo (P<0.05), as shown by analysis of the biochemical composition of chondrocyte ECM. When NMPs were injected into OA model rabbits, the expression of ECM proteins in chondrocytes was significantly promoted and the progression of OA was slowed down.
		                        		
		                        			CONCLUSIONS
		                        			Based on these data, we think that these NMPs with excellent physicochemical and biological properties could be promising non-viral vectors for OA gene therapy.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Cell Survival/drug effects*
		                        			;
		                        		
		                        			Chitosan/chemistry*
		                        			;
		                        		
		                        			Chondrocytes/cytology*
		                        			;
		                        		
		                        			Chondroitin Sulfates/chemistry*
		                        			;
		                        		
		                        			Drug Carriers
		                        			;
		                        		
		                        			Extracellular Matrix/metabolism*
		                        			;
		                        		
		                        			Genetic Therapy/methods*
		                        			;
		                        		
		                        			Growth Differentiation Factor 5/genetics*
		                        			;
		                        		
		                        			Hyaluronic Acid/chemistry*
		                        			;
		                        		
		                        			Microspheres
		                        			;
		                        		
		                        			Nanomedicine
		                        			;
		                        		
		                        			Osteoarthritis/therapy*
		                        			;
		                        		
		                        			Plasmids/metabolism*
		                        			;
		                        		
		                        			Rabbits
		                        			
		                        		
		                        	
10.Seropharmacological study on osteogenic effects of post-absorption ingredients of an osteoprotective herbal formula.
Wing-Sum SIU ; Chun-Hay KO ; Hing-Lok WONG ; Si GAO ; Wai-Ting SHUM ; Clara Bik-San LAU ; Lung-Kim HUNG ; Ping-Chung LEUNG
Chinese journal of integrative medicine 2017;23(1):25-32
OBJECTIVETo further investigate the {ptin vitro} effects of an osteoprotective herbal formula "ELP" (Herba Epimedii, Fructus Ligustri Lucidi and Fructus Psoraleae) using seropharmacological approach.
METHODSRats were fed with ELP or its individual component herbs for 2 days. The serum containing the postabsorbed ingredients of the herbal items were collected for cell culture using UMR106 cell, RAW264.7 cell and mesenchymal stem cell (MSC) isolated from the bone marrow of the rats. The effects of the herbal-containing serum on cell toxicity were detected by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay; bromodeoxyuridine assay was conducted to measure the cell proliferation of UMR106 cell and MSC; cell activity was measured using colorimetric method, and mRNA expression of runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and osteopontin (OPN) of UMR106 and MSC as well as matrix metalloproteinase 9 (MMP-9), tartrate-resistant acid phosphatase (TRAP) and cathepsin K of RAW264.7 were analyzed using real-time reverse-transcription polymerase chain reaction.
RESULTSELP and its component serum exhibited no cytotoxic effects on the cells. The ELP-containing serum increased the proliferation of UMR106 cell and MSC by 25.7% and 14.4 %, respectively and the alkaline phosphatase activity of MSC was increased by 42.6%. On the contrary, it inhibited the RAW264.7 cell differentiation by 29.2 %. ELP serum upregulated the Runx2 expression of UMR and MSC by 1.18 fold and 1.27 fold, respectively. It also upregulated ALP and OPN expression in MSC by 1.69- and 2.12-fold, respectively. On the other hand, ELP serum down-regulated MMP-9 and cathepsin K expression of RAW264.7 cell by 0.46- and 0.36-fold, respectively.
CONCLUSIONSThe serum of the animals fed with ELP contains active ingredients which are effective in promoting osteogenesis and inhibiting osteoclastogenesis.
Absorption, Physiological ; drug effects ; Animals ; Bone and Bones ; drug effects ; pathology ; Cell Differentiation ; drug effects ; Cell Proliferation ; drug effects ; Cell Survival ; drug effects ; Drugs, Chinese Herbal ; pharmacology ; Male ; Mice ; Osteoclasts ; drug effects ; metabolism ; pathology ; Osteogenesis ; drug effects ; Protective Agents ; pharmacology ; RAW 264.7 Cells ; RNA, Messenger ; genetics ; metabolism ; Rats ; Rats, Sprague-Dawley ; Real-Time Polymerase Chain Reaction ; Serum ; metabolism
            
Result Analysis
Print
Save
E-mail