1.Defining a critical period in calvarial development for Hedgehog pathway antagonist-induced frontal bone dysplasia in mice.
Yuanjing JIANG ; Shixian ZHANG ; Chuanqing MAO ; Yongzhen LAI ; Di WU ; Hu ZHAO ; Caiyu LIAO ; Weihui CHEN
International Journal of Oral Science 2019;11(1):3-3
		                        		
		                        			
		                        			The Hedgehog (Hh) signalling pathway is essential for cellular proliferation and differentiation during embryonic development. Gain and loss of function of Hh signalling are known to result in an array of craniofacial malformations. To determine the critical period for Hh pathway antagonist-induced frontal bone hypoplasia, we examined patterns of dysmorphology caused by Hh signalling inhibition. Pregnant mice received a single oral administration of Hh signalling inhibitor GDC-0449 at 100 mg•kg or 150 mg•kg body weight at preselected time points between embryonic days (E)8.5 and 12.5. The optimal teratogenic concentration of GDC-0449 was determined to be 150 mg•kg. Exposure between E9.5 and E10.5 induced frontal bone dysplasia, micrognathia and limb defects, with administration at E10.5 producing the most pronounced effects. This model showed decreased ossification of the frontal bone with downregulation of Hh signalling. The osteoid thickness of the frontal bone was significantly reduced. The amount of neural crest-derived frontal bone primordium was reduced after GDC-0449 exposure owing to a decreased rate of cell proliferation and increased cell death.
		                        		
		                        		
		                        		
		                        			Administration, Oral
		                        			;
		                        		
		                        			Anilides
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Bone Diseases, Developmental
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Frontal Bone
		                        			;
		                        		
		                        			abnormalities
		                        			;
		                        		
		                        			Hedgehog Proteins
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			Limb Deformities, Congenital
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Micrognathism
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			Osteogenesis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Pregnancy
		                        			;
		                        		
		                        			Pyridines
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
2.GSK923295 as a potential antihepatocellular carcinoma agent causing delay on liver regeneration after partial hepatectomy.
Jia-Cheng TANG ; Ke WU ; Xing ZHENG ; Ming XU ; Yi DAI ; Sai-Sai WEI ; Xiu-Jun CAI
Chinese Medical Journal 2019;132(3):311-318
		                        		
		                        			BACKGROUND:
		                        			The clinical trials emerged centromere protein E inhibitor GSK923295 as a promising anticancer drug, but its function in hepatocellular carcinoma (HCC) remain needs to be fully elucidated, especially as chemotherapy after hepatectomy for liver tumors. We aimed to describe anti-HCC activities of GSK923295 and compare its antiproliferative effects on liver regeneration after partial hepatectomy (PH).
		                        		
		                        			METHODS:
		                        			All subjects were randomized to treatment with either vehicle or GSK923295. Antitumor activity of GSK923295 was assessed by xenograft growth assays. The C57BL/6 mice were subjected to 70% PH and the proliferation was calculated by liver coefficient, further confirmed by immunohistochemistry. The proliferation and cell cycle analysis of liver cell AML12 and HCC cells LM3, HUH7, and HepG2 were investigated using the cell counting kit-8 assay and Flow Cytometry. The chromosome misalignment and segregation in AML12 cells were visualized by immunofluorescence.
		                        		
		                        			RESULTS:
		                        			Treatment with GSK923295 induced antiproliferation in HCC cell lines. It also caused delay on HCC tumor growth instead of regression both in a HCC cell line xenograft model and patient-derived tumor xenograft model. With microarray analysis, CENtromere Protein E was gradually increased in mouse liver after PH. Exposure of liver cells to GSK923295 resulted in delay on a cell cycle in mitosis with a phenotype of misaligned chromosomes and chromosomes clustered. In 70% PH mouse model, GSK923295 treatment also remarkably reduced liver regeneration in later stage, in parallel with the mitotic marker phospho-histone H3 elevation.
		                        		
		                        			CONCLUSION
		                        			The anticancer drug GSK923295 causes a significant delay on HCC tumor growth and liver regeneration after PH in later stage.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Blotting, Western
		                        			;
		                        		
		                        			Bridged Bicyclo Compounds, Heterocyclic
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			surgery
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Chromosomal Proteins, Non-Histone
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			Electrophoresis, Polyacrylamide Gel
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Fluorescent Antibody Technique
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunohistochemistry
		                        			;
		                        		
		                        			Liver Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			surgery
		                        			;
		                        		
		                        			Liver Regeneration
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Real-Time Polymerase Chain Reaction
		                        			;
		                        		
		                        			Sarcosine
		                        			;
		                        		
		                        			analogs & derivatives
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Xenograft Model Antitumor Assays
		                        			
		                        		
		                        	
3.Anti-fibrotic Effects and Mechanism of Shengmai Injection () on Human Hepatic Stellate Cells LX-2.
Yi ZHANG ; Li-Tian MA ; Jie LI ; Yu QIAO ; Jun-Ye LIU ; Jin WANG ; Qin-You REN ; Jin-Tao HU ; Jin ZHENG
Chinese journal of integrative medicine 2019;25(3):197-202
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effects of Shengmai Injection (, SMI) on the proliferation, apoptosis and N-myc downstream-regulated gene 2 (NDRG2, a tumour suppressor gene) expression in varying densities of human hepatic stellate cells LX-2.
		                        		
		                        			METHODS:
		                        			LX-2 cells were cultured in vitro. Then, cells were plated in 96-well plates at an approximate density of 2.5×10 cells/mL and cultured for 48, 72, 96 or 120 h followed by the application of different concentrations of SMI (0.6, 1.2, 2.4, 4.8 or 6 μL/mL). Cell proliferation was measured after an additional 24 or 48 h using the 3(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of SMI on different cell growth states (cultured for 48, 72, 96, or 120 h) were observed by light microscopy at 24 h after treatment. When the cells reached 80% conflfluence, apoptosis was detected by flflow cytometry after 24 h. Lastly, LX-2 cells were treated with different concentrations of SMI and extracted with protein lysis buffer. The levels of NDRG2 were measured by Western blot.
		                        		
		                        			RESULTS:
		                        			When the LX-2 cells grew for 48, 72, 96 and 120 h, 4.8 and 6 μL/mL of SMI significantly inhibited cell proliferation at 24 and 48 h after treatment (P<0.05). And 2.4 μL/mL of SMI also inhibited cell proliferation at 24 h after treatment when cell growth for 48 h (P<0.05) and at 48 h after treatment when cell growth for 72, 96 and 120 h (P<0.05). The NDRG2 expression level in the LX-2 cell was significantly increased when treated with SMI at concentrations of 1.2, 2.4, 4.8 or 6 μL/mL (P<0.05).
		                        		
		                        			CONCLUSION
		                        			The inhibitory effects of SMI on the proliferation of LX-2 cells were related to not only concentration dependent but also cell density. In addition, SMI (2.4, 4.8 and 6 μL/mL) could accelerate apoptosis in LX-2 cells, and the mechanism might be associated with NDRG2 over-expression.
		                        		
		                        		
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Drugs, Chinese Herbal
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Hepatic Stellate Cells
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Injections
		                        			;
		                        		
		                        			Liver Cirrhosis
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			Tumor Suppressor Proteins
		                        			;
		                        		
		                        			genetics
		                        			
		                        		
		                        	
4.Effects of metformin and adiponectin on endometrial cancer cells growth.
Xiao Hui WANG ; Yan ZHANG ; Lin Zhi LIU ; Chen Guang SHANG
Journal of Peking University(Health Sciences) 2018;50(5):767-773
		                        		
		                        			OBJECTIVE:
		                        			To determine the effect of metformin and adiponectin on the proliferation of EC cells and the relationship between metformin and adiponectin.
		                        		
		                        			METHODS:
		                        			The proliferation impact of different concentrations of metformin and adiponectin on two types of EC cells ishikawa (IK) and HEC-1B was confirmed by CCK-8 method. qRT-PCR and Western blot were used to detect the effect of different concentrations of metformin on the changes of adiponectin receptors (AdipoR1 and AdipoR2) of the EC cells both in mRNA and protein level and the role of compound C, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor, on the above effects.
		                        		
		                        			RESULTS:
		                        			(1) Both metformin and adiponectin could significantly promote the proliferation of endometrial cancer (EC) cells in a time and concentration dependent manner (P<0.05).(2)Metformin and adiponectin had synergy anti-proliferative effect on EC cells and the combination index (CI) value of IK cells was 0.906 34 and of HEC-1B cells was 0.827 65. (3)qRT-PCR was used to detect the mRNA levels of AdipoR1 and AdipoR2 after 5 mmol/L and 10 mmol/L metformin, respectively, stimulating IK and HEC-1B cells for 48 hours and the mRNA expressions of AdipoR1 and AdipoR2 were significantly increased when compared with the control group (0 mmol/L)(IK: AdipoR1 of 5 mmol/L and 10 mmol/L group: P<0.001,AdipoR2 of 5 mmol/L group: P<0.001; HEC-1B: AdipoR1 of 5 mmol/L group: P<0.001, 10 mmol/L group: P=0.023, AdipoR2 of 5 mmol/L group: P<0.001, 10 mmol/L group: P=0.024). When combined with compound C, the RNA levels of AdipoR1 and AdipoR2 were not different compared with the control group (0 mmol/L, P>0.05). (4) Western blot was used to detect the protein levels of AdipoR1 and AdipoR2 after 5 mmol/L and 10 mmol/L metformin, stimulating IK and HEC-1B cells for 48 hours and the protein level was significantly increased when compared with the control group (0 mmol/L)(IK: AdipoR1 of 5 mmol/L group: P=0.04, 10 mmol/L group: P=0.033, AdipoR2 of 5 mmol/L group: P=0.044, 10 mmol/L group: P=0.046; HEC-1B: AdipoR1 of 5 mmol/L group: P=0.04, 10 mmol/L group: P=0.049, AdipoR2 of 5 mmol/L group: P=0.043, 10 mmol/L group: P=0.035). When combined with compound C,the protein levels of AdipoR1 and AdipoR2 were not different compared with the control group (0 mmol/L, P>0.05).
		                        		
		                        			CONCLUSION
		                        			We find that metformin and adiponectin have synergy anti-proliferative effect on EC cells. Besides, metformin can increase adiponectin receptors expressions of EC cells both in mRNA and protein levels and this effect is accomplished by the activation of AMPK signaling pathway.
		                        		
		                        		
		                        		
		                        			Adiponectin/physiology*
		                        			;
		                        		
		                        			Cell Proliferation/drug effects*
		                        			;
		                        		
		                        			Endometrial Neoplasms/pathology*
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Hypoglycemic Agents/pharmacology*
		                        			;
		                        		
		                        			Metformin/pharmacology*
		                        			;
		                        		
		                        			Receptors, Adiponectin
		                        			;
		                        		
		                        			Signal Transduction
		                        			
		                        		
		                        	
5.AATYK is a Novel Regulator of Oligodendrocyte Differentiation and Myelination.
Chunxia JIANG ; Wanqing YANG ; Zhihong FAN ; Peng TENG ; Ruyi MEI ; Junlin YANG ; Aifen YANG ; Mengsheng QIU ; Xiaofeng ZHAO
Neuroscience Bulletin 2018;34(3):527-533
		                        		
		                        			
		                        			Oligodendrocytes (OLs) are myelinating glial cells that form myelin sheaths around axons to ensure rapid and focal conduction of action potentials. Here, we found that an axonal outgrowth regulatory molecule, AATYK (apoptosis-associated tyrosine kinase), was up-regulated with OL differentiation and remyelination. We therefore studied its role in OL differentiation. The results showed that AATYK knockdown inhibited OL differentiation and the expression of myelin genes in vitro. Moreover, AATYK-deficiency maintained the proliferation status of OLs but did not affect their survival. Thus, AATYK is essential for the differentiation of OLs.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Animals, Newborn
		                        			;
		                        		
		                        			Apoptosis Regulatory Proteins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Cuprizone
		                        			;
		                        		
		                        			toxicity
		                        			;
		                        		
		                        			Demyelinating Diseases
		                        			;
		                        		
		                        			chemically induced
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Embryo, Mammalian
		                        			;
		                        		
		                        			Gene Expression Regulation, Developmental
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Ki-67 Antigen
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Myelin Basic Protein
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Myelin Proteolipid Protein
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Myelin Sheath
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Oligodendroglia
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Protein-Tyrosine Kinases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			RNA, Small Interfering
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			
		                        		
		                        	
6.Effects of Nerve Growth Factor on Cardiac Fibroblasts Proliferation, Cell Cycle, Migration, and Myofibroblast Transformation.
Chinese Medical Journal 2018;131(7):813-817
BackgroundRecent research indicates that nerve growth factor (NGF) promotes cardiac repair following myocardial infarction by promoting angiogenesis and cardiomyocyte survival. The purpose of this study was to investigate the effects of NGF on cardiac fibroblasts (CFs) proliferation, cell cycle, migration, and myofibroblast transformation in vitro.
MethodsCFs were obtained from ventricles of neonatal Sprague-Dawley rats and incubated with various concentrations of NGF (0, 0.01, 0.1, 1, 10, and 100 ng/ml; 0 ng/ml was designated as the control group). Cell proliferation and cell cycle of the CFs were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry (FCM), respectively. A cell scratch wound model and transwell were carried out to observe effects of NGF on migration of CFs after 24 h of culture. Real-time polymerase chain reaction (RT-PCR) and Western blotting were used to measure α-smooth muscle actin (α-SMA) at mRNA and protein levels after CFs were incubated with various concentrations of NGF.
ResultsExpression of α-SMA measured by RT-PCR and Western blotting significantly increased in the 1 and 10 ng/ml NGF groups (P < 0.05). Absorbance values of CFs showed that NGF did not influence the proliferation of CFs (The Avalues were 0.178 ± 0.038, 0.182 ± 0.011, 0.189 ± 0.005, 0.178 ± 0.010, 0.185 ± 0.025, and 0.177 ± 0.033, respectively, in the 0, 0.01, 0.1, 1, 10, and 100 ng/ml NGF groups [P = 0.800, 0.428, 0.981, 0.596, and 0.913, respectively, compared with control group]), and FCM analysis showed that the percentage of CFs in G0/G1, S, and G2/M phases was not changed (P > 0.05). The cell scratch wound model and transwell showed that CFs migration was not significantly different (P > 0.05).
ConclusionNGF induces myofibroblast transformation but does not influence proliferation, cell cycle, or migration of CFs in vitro.
Actins ; metabolism ; Animals ; Cell Cycle ; drug effects ; physiology ; Cell Movement ; drug effects ; physiology ; Cell Proliferation ; physiology ; Cells, Cultured ; Myofibroblasts ; cytology ; drug effects ; Nerve Growth Factor ; metabolism ; pharmacology ; Rats ; Rats, Sprague-Dawley
7.Oridonin inhibits proliferation of Jurkat cells via the down-regulation of Brg1.
Zhen-Zhen YE ; Fei-Long XUE ; Wen-Ping DING ; Xiang KONG ; Yi-Na SHEN
Chinese Journal of Contemporary Pediatrics 2017;19(11):1208-1212
OBJECTIVETo investigate the effect of oridonin on the human acute lymphocytic leukemia cell line Jurkat and its mechanism.
METHODSJurkat cells were cultured in vitro and treated with various concentrations (0, 1.25, 2.5, 5, and 10 μmol/L) of oridonin for different lengths of time (24, 48, and 72 hours). The proliferation of Jurkat cells was analyzed by MTT assay. The changes in nuclear morphology were evaluated by fluorescence microscopy at 12 hours after treatment with various concentrations of oridonin. The expression levels of Brg1, P53, and C-myc were determined by semi-quantitative Western blot in Jurkat cells treated with various concentrations of oridonin for 24 hours or 5 μmol/L oridonin for various lengths of time (0, 2, 6, 12, and 24 hours). The expression levels of P53 and C-myc and proliferation of Jurkat cells were evaluated after Brg1 expression was knocked down by Brg1-specific siRNA.
RESULTSCompared with the control group, the proliferation of oridonin-treated Jurkat cells was significantly inhibited in a concentration- and time-dependent manner (P<0.05). According to the florescence microscopic analysis, oridonin treatment led to nuclear pyknosis in Jurkat cells. Compared with the control group, Jurkat cells treated with 5 μmol/L oridonin had reduced expression of Brg1 and C-myc but elevated expression of P53. Brg1 knock-down led to a significant reduction in proliferation of Jurkat cells (P<0.05), up-regulated expression of P53, and down-regulated expression of C-myc.
CONCLUSIONSOridonin can inhibit the proliferation of Jurkat cells, probably via the Brg1 signaling pathway.
Antineoplastic Agents, Phytogenic ; pharmacology ; Cell Proliferation ; drug effects ; DNA Helicases ; analysis ; physiology ; Diterpenes, Kaurane ; pharmacology ; Dose-Response Relationship, Drug ; Down-Regulation ; Humans ; Jurkat Cells ; Nuclear Proteins ; analysis ; physiology ; Precursor Cell Lymphoblastic Leukemia-Lymphoma ; drug therapy ; Proto-Oncogene Proteins c-myc ; analysis ; Signal Transduction ; physiology ; Transcription Factors ; analysis ; physiology ; Tumor Suppressor Protein p53 ; analysis
8.Effect of polyunsaturated fatty acids ω-3 and ω-6 on angiogenesis formation in human gastric cancer.
Jiachi MA ; Yuntao MA ; Tiankang GUO ; Quan CHEN ; Yiping LI ; He SU ; Xiaochang CHEN ; Xiaodan ZHAO ; Qinjin GUO ; Jianbo QI
Chinese Journal of Gastrointestinal Surgery 2017;20(1):84-89
OBJECTIVETo investigate the effects of polyunsaturated fatty acids (PUFA) ω-3 and ω-6, and their middle metabolites PGE2 and PGE3 on angiogenesis formation of gastric cancer, and to explore associated mechanism.
METHODSThe effects of ω-3, ω-6, PGE2, PGE3 on the proliferation and migration of human umbilical vein endothelial cell (HUVEC) were measured by proliferation and migration assay respectively. The angiogenesis assay in vivo was used to measure the effects of ω-3, ω-6, PGE2 and PGE3 on neovascularization. In all the assays, groups without ω-3, ω-6, PGE2 and PGE3 were designed as the control.
RESULTSWith the increased concentration of ω-6 from 1 μmol/L to 10 μmol/L, the proliferation ability of HUVECs enhanced, and the number of migration cells also increased from 28.2±3.0 to 32.8±2.1, which was higher than control group (21.2±3.2) respectively (both P<0.05). With the increased concentration of ω-3 from 1 μmol/L to 10 μmol/L, the proliferation ability of HUVECs was inhibited, and the number of migration cells decreased from 15.8±2.0 to 11.0±2.1, which was lower than control group (22.1±3.0) respectively (both P<0.05). In the angiogenesis assay, compared with control group (standard number: 43 721±4 654), the angiogenesis ability of HUVECs was significantly enhanced by ω-6 in concentration-dependent manner (1 μmol/L group: 63 238±4 795, 10 μmol/L group: 78 166±6 123, all P<0.01). Meanwhile, with the increased concentration of ω-3 from 1 μmol/L to 10 μmol/L, the angiogenesis ability was significantly decreased from 30 129±3 102 to 20 012±1 541(all P<0.01). The proliferation and migration ability of HUVECs were significantly promoted by ω-6 metabolites PGE2 (P<0.05) in a concentration-dependent manner. In contrast, ω-3 metabolites PGE3 significantly inhibited the proliferation and migration ability of HUVECs in a concentration-dependent manner (all P<0.05). After rofecoxib (a COX-2 specific inhibitor) inhibited the expression of COX-2, the expression level of PGE2 was significantly decreased in a dose-dependent manner. In co-culture system, whose gastric cancer cells expressed positive COX-2, ω-6 could increase angiogenesis of gastric cancer cells(P<0.01), but ω-3 could inhibit such angiogenesis(P<0.01). In co-culture system, whose gastric cancer cells did not express COX-2, ω-3 could inhibit the angiogenesis of gastric cancer cells (P<0.05), but ω-6 had no effect on angiogenesis.
CONCLUSIONSThe PUFA ω-6 can enhance the angiogenesis via the promotion of proliferation and migration of HUVECs, and COX-2 and PGE2 may play an important role in this process, whereas, the ω-3 can inhibit the angiogenesis through its middle metabolites PGE3 to inhibit the proliferation and migration of HUVECs. Results of this experiment may provide a new approach to inhibit and prevent the spread of gastric cancer.
Alprostadil ; analogs & derivatives ; pharmacology ; Angiogenesis Inducing Agents ; metabolism ; pharmacology ; Angiogenesis Inhibitors ; pharmacology ; Cell Count ; methods ; Cell Line, Tumor ; drug effects ; physiology ; Cell Migration Assays ; Cell Movement ; drug effects ; Cell Proliferation ; drug effects ; Coculture Techniques ; Cyclooxygenase 2 ; pharmacology ; Dinoprostone ; metabolism ; pharmacology ; Fatty Acids, Omega-3 ; pharmacology ; Fatty Acids, Omega-6 ; metabolism ; pharmacology ; Fatty Acids, Unsaturated ; pharmacology ; Human Umbilical Vein Endothelial Cells ; drug effects ; physiology ; Humans ; Lactones ; pharmacology ; Neovascularization, Pathologic ; physiopathology ; Stomach Neoplasms ; physiopathology ; Sulfones ; pharmacology
9.Nr2e1 Downregulation Is Involved in Excess Retinoic Acid-induced Developmental Abnormality in the Mouse Brain.
Juan YU ; Qian GUO ; Jian Bing MU ; Ting ZHANG ; Ren Ke LI ; Jun XIE
Biomedical and Environmental Sciences 2017;30(3):185-193
OBJECTIVEThis study aimed to investigate the expression pattern and function of Nuclear receptor subfamily 2 group E member 1 (Nr2e1) in retinoic acid (RA)-induced brain abnormality.
METHODSThe mouse model of brain abnormality was established by administering 28 mg/kg RA, and neural stem cells (NSCs) were isolated from the mouse embryo and cultured in vitro. Nr2e1 expression was detected by whole mount in situ hybridization, RT-PCR, and Western blotting. Nr2e1 function was determined by transducing Nr2e1 shRNA into NSCs, and the effect on the sonic hedgehog (Shh) signaling pathway was assessed in the cells. In addition, the regulation of Nr2e1 expression by RA was also determined in vitro.
RESULTSNr2e1 expression was significantly downregulated in the brain and NSCs of RA-treated mouse embryos, and knockdown of Nr2e1 affected the proliferation of NSCs in vitro. In addition, a similar expression pattern of Nr2e1 and RA receptor (RAR) α was observed after treatment of NSCs with different concentrations of RA.
CONCLUSIONOur study demonstrated that Nr2e1 could be regulated by RA, which would aid a better understanding of the mechanism underlying RA-induced brain abnormality.
Animals ; Brain ; cytology ; embryology ; Cell Proliferation ; Down-Regulation ; Gene Expression Regulation ; Gene Expression Regulation, Developmental ; drug effects ; Mice ; Mice, Inbred C57BL ; Neural Stem Cells ; drug effects ; physiology ; Receptors, Cytoplasmic and Nuclear ; genetics ; metabolism ; Tretinoin ; pharmacology
10.Research advances in the protective effect of all-trans retinoic acid against podocyte injury.
Chinese Journal of Contemporary Pediatrics 2017;19(6):719-723
		                        		
		                        			
		                        			All-trans retinoic acid (ATRA) is a vitamin A derivative and plays an important role in the regulation of cell aggregation, differentiation, apoptosis, proliferation, and inflammatory response. In recent years, some progress has been made in the role of ATRA in renal diseases, especially its protective effect on podocytes. This article reviews the research advances in podocyte injury, characteristics of ATRA, podocyte differentiation and regeneration induced by ATRA, and the protective effect of ATRA against proliferation, deposition of fibers, and apoptosis.
		                        		
		                        		
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cytoprotection
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Podocytes
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Tretinoin
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail