1.Carfilzomib inhibits the growth of lung adenocarcinoma via upregulation of Gadd45a expression.
Fang YANG ; Wang-Wang LIU ; Hui CHEN ; Jia ZHU ; Ai-Hua HUANG ; Fei ZHOU ; Yi GAN ; Yan-Hua ZHANG ; Li MA
Journal of Zhejiang University. Science. B 2020;21(1):64-76
Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.
Adenocarcinoma of Lung/pathology*
;
Apoptosis/drug effects*
;
Cell Cycle Checkpoints/drug effects*
;
Cell Cycle Proteins/genetics*
;
Cell Line, Tumor
;
Forkhead Box Protein O3/physiology*
;
Gene Expression Regulation, Neoplastic/drug effects*
;
Humans
;
Lung Neoplasms/pathology*
;
Oligopeptides/pharmacology*
;
Proto-Oncogene Proteins c-akt/physiology*
;
Up-Regulation
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.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
4.Effects of Low-dose Triamcinolone Acetonide on Rat Retinal Progenitor Cells under Hypoxia Condition.
Yao XING ; Li-Jun CUI ; Qian-Yan KANG
Chinese Medical Journal 2016;129(13):1600-1606
BACKGROUNDRetinal degenerative diseases are the leading causes of blindness in developed world. Retinal progenitor cells (RPCs) play a key role in retina restoration. Triamcinolone acetonide (TA) is widely used for the treatment of retinal degenerative diseases. In this study, we investigated the role of TA on RPCs in hypoxia condition.
METHODSRPCs were primary cultured and identified by immunofluorescence staining. Cells were cultured under normoxia, hypoxia 6 h, and hypoxia 6 h with TA treatment conditions. For the TA treatment groups, after being cultured under hypoxia condition for 6 h, RPCs were treated with different concentrations of TA for 48-72 h. Cell viability was measured by cell counting kit-8 (CCK-8) assay. Cell cycle was detected by flow cytometry. Western blotting was employed to examine the expression of cyclin D1, Akt, p-Akt, nuclear factor (NF)-κB p65, and caspase-3.
RESULTSCCK-8 assays indicated that the viability of RPCs treated with 0.01 mg/ml TA in hypoxia group was improved after 48 h, comparing with control group (P < 0.05). After 72 h, the cell viability was enhanced in both 0.01 mg/ml and 0.02 mg/ml TA groups compared with control group (all P < 0.05). Flow cytometry revealed that there were more cells in S-phase in hypoxia 6 h group than in normoxia control group (P < 0.05). RPCs in S and G2/M phases decreased in groups given TA, comparing with other groups (all P < 0.05). There was no significant difference in the total Akt protein expression among different groups, whereas upregulation of p-Akt and NF-κB p65 protein expression and downregulation of caspase-3 and cyclin D1 protein expression were observed in 0.01 mg/ml TA group, comparing with hypoxia 6 h group and control group (all P < 0.05).
CONCLUSIONLow-dose TA has anti-apoptosis effect on RPCs while it has no stimulatory effect on cell proliferation.
Animals ; Apoptosis ; drug effects ; physiology ; Caspase 3 ; metabolism ; Cell Cycle ; drug effects ; physiology ; Cell Hypoxia ; drug effects ; physiology ; Cell Proliferation ; drug effects ; physiology ; Cell Survival ; drug effects ; physiology ; Cells, Cultured ; Cyclin D1 ; metabolism ; NF-kappa B ; metabolism ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Retina ; cytology ; Stem Cells ; cytology ; drug effects ; Triamcinolone Acetonide ; pharmacology
5.Expression of miR-let-7e-3p in cervical intraepithelial neoplasm and cervix carcinoma and its clinical significance.
Xiaojing CHEN ; Junfen XU ; Jing YE ; Xiaodong CHENG ; Xing XIE ; Weiguo LYU
Journal of Zhejiang University. Medical sciences 2016;45(4):342-348
To investigate the expression of microRNA (miRNA, miR) let-7e-3p in different cervical lesions and its clinical significance.The expression of miR-let-7e-3p in the tissues of normal cervix (=26), high-grade squamous intraepithelial lesion (HSIL) (=37), and cervix carcinoma (=101) were detected by reverse transcription and quantitative polymerase chain reaction (RT-qPCR). The correlation of miR-let-7e-3p expression with the clinicopathological parameters of patients with cervical cancer was analyzed. miR-let-7e-3p mimic was transfected into cervical carcinoma Siha cells. The cell cycle and apoptosis were determined by flow cytometry; cell proliferation was determined by CCK-8 kit; and the migration and invasion of cells were determined by Transwell assay.The relative expression levels of miR-let-7e-3p in normal cervix, HSIL, and cervical carcinoma were 1.45±0.24, 0.79±0.05 and 0.46±0.04, respectively (all<0.05). After transfection with miR-let-7e-3p mimic, the S-phase fraction and apoptosis rate of Siha cells were increased significantly compared with control group[(29.76±6.6)% vs (13.38±1.3)%,<0.05; (5.98±1.38)% vs (3.53±0.79)%,<0.05, respectively]. OD of transfected Siha cells at 48, 72 and 96 h were 0.57±0.11,0.65±0.04 and 0.84±0.14, which were significantly lower than those of untransfected Siha cells (0.74±0.05, 0.93±0.10 and 1.47±0.14, all<0.05). The migration and invasion abilities of transfected Siha cells were not significantly changed (all>0.05).The expression of miR-let-7e-3p is down-regulated in cervical neoplasms, which is associated with cell cycle arrest and proliferation inhibition of cervical cancer cells.
Apoptosis
;
drug effects
;
genetics
;
Carcinoma
;
chemistry
;
genetics
;
Cell Cycle
;
drug effects
;
genetics
;
Cell Line, Tumor
;
chemistry
;
drug effects
;
physiology
;
Cell Movement
;
drug effects
;
genetics
;
Cell Proliferation
;
drug effects
;
genetics
;
Cervical Intraepithelial Neoplasia
;
chemistry
;
genetics
;
physiopathology
;
Down-Regulation
;
physiology
;
Female
;
Humans
;
MicroRNAs
;
analysis
;
pharmacology
;
Neoplasm Invasiveness
;
genetics
;
physiopathology
;
Neoplastic Processes
;
Real-Time Polymerase Chain Reaction
;
Transfection
;
Uterine Cervical Neoplasms
;
chemistry
;
genetics
;
physiopathology
6.Effect of puerarin on hypoxia induced proliferation of PASMCs by regulating reactive oxygen.
Xiao-dan ZHANG ; Li-wei WANG ; Shu-jing WANG ; Da-ling ZHU ; Yan-nan YANG ; Jie-jing SHENG ; Sha-sha SONG
China Journal of Chinese Materia Medica 2015;40(15):3027-3033
To discuss the effect of puerarin (Pue) on the proliferation of hypoxia-induced pulmonary artery smooth muscle cells (PASMCs) and discuss whether its mechanism is achieved by regulating reactive oxygen. PASMCs of primarily cultured rats (2-5 generations) were selected in the experiment. MTT, Western blot, FCM and DCFH-DA were used to observe Pue's effect the proliferation of PASMCs. The Western blot was adopted to detect whether ROS participated in Pue's effect in inhibiting PASMC proliferation. The PASMCs were divided into five groups: the normoxia group, the hypoxia group, the hypoxia + Pue group, the hypoxia + Pue + Rotenone group and the hypoxia + Rotenone group, with Rotenone as the ROS blocker. According to the results, under the conditions of normoxia, Pue had no effect on the PASMC proliferation; But, under the conditions of hypoxia, it could inhibit the PASMC proliferation; Under the conditions of normoxia and hypoxia, Pue had no effect on the expression of the tumor necrosis factor-α (TNF-α) among PASMCs, could down-regulate the expression of hypoxia-induced cell cycle protein Cyclin A and proliferative nuclear antigen (PCNA). DCFH-DA proved Pue could reverse ROS rise caused by hypoxia. Both Rotenone and Pue could inhibit the up-regulated expressions of HIF-1α, Cyclin A, PCNA caused by anoxia, with a synergistic effect. The results suggested that Pue could inhibit the hypoxia-induced PASMC proliferation. Its mechanism may be achieved by regulating ROS.
Animals
;
Cell Cycle
;
drug effects
;
Cell Proliferation
;
drug effects
;
Cells, Cultured
;
Hypoxia
;
pathology
;
Isoflavones
;
pharmacology
;
Male
;
Myocytes, Smooth Muscle
;
drug effects
;
physiology
;
Proliferating Cell Nuclear Antigen
;
analysis
;
Pulmonary Artery
;
cytology
;
drug effects
;
Rats
;
Rats, Wistar
;
Reactive Oxygen Species
;
metabolism
7.Inhibition Mechanism of Novel Pyrazolo1,5-apyrazin-4(5H)-one Derivatives Against Proliferation of A549 and H322 Cancer Cells.
Chinese Medical Sciences Journal 2015;30(4):260-265
OBJECTIVETo explore the inhibition mechanism and safety of pyrazolo[1,5-a]pyrazin-4(5H)-one derivatives against proliferation of human lung cancer A549 cells, H322 cells, and human umbilical vein endothelial cell (HUVEC).
METHODSCells were treated with 40 Μmol/L of the ppo3a, ppo3b, ppo3i, and 0.1% DMSO (control) for 48 hours, respectively. Apoptosis was determined by Hoechst 33258 staining assay in H322 and A549 cells. Cell cycle distribution was determined by flow cytometry analysis in A549 cell. LC3-II, p53, and heat shock protein (HSP) 70 protein levels were detected by Western blotting in A549 cells treated with ppo3b for 48 hours. The morphology and viability of HUVEC were observed by inverted microscope and sulforhodamine B (SRB) assay.
RESULTSPpo3a, ppo3b, and ppo3i significantly induced apoptosis in H322 and A549 cells. A strong G1-phase arrest was concomitant with the growth inhibitory effect on A549 cells. Ppo3b effectively elevated the p53 protein level, but significantly reduced the HSP70 protein level. There were no significantly inhibitory effect on the morphology and viability of HUVEC when treated with ppo3a, ppo3b, and ppo3i.
CONCLUSIONSppo3a, ppo3b, and ppo3i could inhibit H322 proliferation through apoptosis and inhibit A549 through apoptosis and G1-phase arrest. The protein p53 and HSP70 might involve in the inhibition effects. These derivatives might be a clue to find effective and safe drug for lung cancers.
Apoptosis ; drug effects ; Cell Cycle Checkpoints ; drug effects ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; HSP70 Heat-Shock Proteins ; analysis ; physiology ; Humans ; Pyrazoles ; pharmacology ; Tumor Suppressor Protein p53 ; analysis ; physiology
8.Effects of hypoxia of different duration on movement and proliferation of human epidermal cell line HaCaT.
Tiantian YAN ; Dongxia ZHANG ; Xupin JIANG ; Qiong ZHANG ; Yuesheng HUANG
Chinese Journal of Burns 2014;30(3):231-236
OBJECTIVETo study the effects of hypoxia of different duration on movement and proliferation of human epidermal cell line HaCaT.
METHODS(1) HaCaT cells in logarithmic phase were cultured in RPMI 1640 medium containing 10% FBS (the same culture method below). Cells were divided into control group (routine culture) and hypoxia for 1, 3, 6 h groups according to the random number table (the same grouping method below), with 6 wells in each group. Cells in the 3 hypoxia groups were cultured in incubator containing 5% CO2, 2% O2, and 93% N2 (the same hypoxic condition below) for corresponding duration. Range of movement of cells in 3 hours was observed under live cell imaging workstation, and their curvilinear and rectilinear movement speeds were calculated at post observation hour (POH) 1, 2, 3. (2) HaCaT cells in logarithmic phase were divided into control group (routine culture) and hypoxia for 1, 3, 6, 9, 12, 24 h groups, with 20 wells in each group. Cells in the 6 hypoxia groups were cultured under hypoxic condition for corresponding duration. Proliferation of cells was examined with cell counting kit and microplate reader (denoted as absorbance value). (3) HaCaT cells in logarithmic phase were divided into control group (routine culture) and hypoxia for 1, 3, 6, 24 h groups, with 5 wells in each group. Cells in the 4 hypoxia groups were cultured under hypoxic condition for corresponding duration. Protein expression of proliferating cell nuclear antigen (PCNA) was determined with Western blotting. Data were processed with one-way analysis of variance and Dunnett- t test.
RESULTS(1) Compared with that of control group, the movement area of cells was obviously expanded in hypoxia for 1, 3, 6 h groups. The longer the hypoxic treatment, the greater the increase was. At POH 1, 2, 3, the curvilinear movement speeds of cells in hypoxia for 1, 3, 6 h groups were respectively (43 ± 18), (44 ± 17), (43 ± 16) µm/h; (44 ± 16), (44 ± 14), (45 ± 14) µm/h; (55 ± 19), (54 ± 17), (56 ± 18) µm/h. They were significantly higher than those of control group [(33 ± 13), (33 ± 12), (33 ± 10) µm/h, with t values from 2.840 to 9.330, P < 0.05 or P < 0.01]. The curvilinear movement speed of cells was significantly higher in hypoxia for 6 h group than in hypoxia for 1 or 3 h group (with t values from 3.474 to 4.545, P < 0.05 or P < 0.01). There was no significant difference in the curvilinear movement speed among the observation time points within each group (with F values from 0.012 to 0.195, P values above 0.05). At POH 1, the rectilinear movement speed of cells in hypoxia for 1 h group was (22 ± 11) µm/h, which was obviously higher than that of control group [(15 ± 10) µm/h, t = 2.697, P < 0.01]. At POH 1, 2, 3, rectilinear movement speeds of cells in hypoxia for 3 and 6 h groups were respectively (19 ± 14), (12 ± 8), (10 ± 6) µm/h; (32 ± 19), (21 ± 13), (17 ± 12) µm/h. They were significantly higher than those of control group [(9 ± 7) and (6 ± 5) µm/h at POH 2 and 3, with t values from 1.990 to 8.231, P < 0.05 or P < 0.01]. The rectilinear movement speed of cells in hypoxia for 6 h group was obviously higher than that of hypoxia for 1 or 3 h group (with t values from 3.394 to 6.008, P < 0.05 or P < 0.01). The rectilinear movement speed of cells in each group decreased at POH 2 or 3 in comparison with POH 1 (with t values from -8.208 to -4.232, P values below 0.01). The rectilinear movement speed of cells in control group at POH 3 was significantly different from that at POH 2 (t = -1.967, P < 0.05). (2) The proliferation levels of cells in control group and hypoxia for 1, 3, 6, 9, 12, 24 h groups were respectively 1.11 ± 0.08, 1.36 ± 0.10, 1.39 ± 0.05, 1.38 ± 0.05, 1.10 ± 0.14, 1.06 ± 0.09, 0.99 ± 0.06 (F = 39.19, P < 0.01). Compared with that of control group, the rate of proliferation of cells was obviously increased in hypoxia for 1, 3, 6 h groups (with t values respectively 6.639, 7.403, 7.195, P values below 0.01), but obviously decreased in hypoxia for 24 h group (t = -3.136, P < 0.05). The proliferation of cells decreased in hypoxia for 9, 12, 24 h groups in comparison with hypoxia for 1, 3, 6 h groups (with t values from -10.538 to -6.775, P values below 0.01). (3) The protein expressions of PCNA of cells in control group and hypoxia for 1, 3, 6, 24 h groups were respectively 0.93 ± 0.12, 0.97 ± 0.14, 1.62 ± 0.18, 0.95 ± 0.09, 0.66 ± 0.21 (F = 20.11, P < 0.01). Compared with that of control group, the expression of PCNA was obviously increased in hypoxia for 1, 3, 6 h groups (with t values respectively 2.339, 5.783, 2.235, P < 0.05 or P < 0.01), but obviously decreased in hypoxia for 24 h group (t = -1.998, P < 0.05). The protein expression of PCNA was higher in hypoxia for 3 h group than in hypoxia for 1 or 6 h group (with t values respectively 4.312 and 3.947, P values below 0.01), and it was increased in the 3 groups in comparison with that of hypoxia for 24 h group (with t values respectively 2.011, 6.193, 3.287, P < 0.05 or P < 0.01).
CONCLUSIONSShort-time hypoxia (1, 3, 6 h) treatment can promote the movement and proliferation of HaCaT cells. Hypoxia for 6 h is the best condition to promote their movement, while hypoxia for 3 or 6 h is better for their proliferation.
Carbon Dioxide ; pharmacology ; Cell Cycle ; drug effects ; Cell Line ; Cell Movement ; physiology ; Cell Proliferation ; drug effects ; physiology ; Cells, Cultured ; Epithelial Cells ; cytology ; drug effects ; Humans ; Hypoxia ; physiopathology ; Nitric Oxide ; pharmacology ; Oxygen ; pharmacology ; Phosphorylation ; Proliferating Cell Nuclear Antigen ; Signal Transduction
9.Impact of PTTG1 downregulation on cell proliferation, cell cycle and cell invasion of osteosarcoma and related molecular mechanisms.
Dapeng WU ; Yonghua XIA ; Haibin XU ; Bin ZHAO ; Qiudong LIANG ; Tan LU ; Jinling JIA ; Yuzhen DONG
Chinese Journal of Pathology 2014;43(10):695-698
OBJECTIVETo downregulate the expression of pituitary tumor transforming gene 1 (PTTG1) in osteosarcoma (OS) cells by siRNA technology and to investigate related biological impact on cell proliferation, cell cycle and cell invasion of OS.
METHODSThree OS cell lines and osteoblast hFOB1.19 cell line were used in this study. Control siRNA and PTTG1 siRNA were employed to transfect OS U2OS cells, and PTTG1 protein level was detected by Western blot after the transfection. Effects of PTTG1 siRNA on cell proliferation, cell cycle and cell invasion were investigated by CCK-8, flow cytometry and Boyden chamber, respectively. Finally, activity of Akt and its downstream target gene expression were analyzed by Western blot in U2OS cells upon various treatments.
RESULTSExpression of PTTG1 protein in 3 OS cells (MG-63, SaOS-2 and U2OS) was significantly higher than that in osteoblast hFOB1.19, among which U2OS cells displayed the highest level. PTTG1 siRNA markedly downregulated the expression of PTTG1 protein in U2OS cells, leading to obvious inhibition of cell proliferation, altered cell cycle distribution and reduced ability of invasion of U2OS cells. Moreover, downregulation of PTTG1 reduced the expression of p-Akt (S473 and T308), MMP-2 and MMP-9 proteins, along with enhanced expression of p21 and E-cadherin proteins.
CONCLUSIONSPTTG1 may be tightly linked to the development of OS and therefore may serve as a novel target for precision therapy of OS.
Bone Neoplasms ; metabolism ; pathology ; Cadherins ; metabolism ; Cell Cycle ; drug effects ; physiology ; Cell Movement ; Cell Proliferation ; drug effects ; physiology ; Down-Regulation ; Humans ; Matrix Metalloproteinase 2 ; metabolism ; Matrix Metalloproteinase 9 ; metabolism ; Neoplasm Invasiveness ; Osteosarcoma ; metabolism ; pathology ; RNA, Small Interfering ; pharmacology ; Securin ; genetics ; metabolism ; Transfection
10.Effect of uighur medicine abnormal savda munzip on human hypertrophic scar fibroblasts in vitro.
Wei-Cheng GAO ; Hu-Jun WANG ; Xing QIAO ; Juan MA ; Jin DU ; Shao-Lin MA
Chinese Journal of Plastic Surgery 2013;29(6):418-421
OBJECTIVETo evaluate in vitro effect of abnormal savda munziq (ASMq) on the proliferation and apoptosis of human hypertrophic scar fibroblasts (HSFs).
METHODSHSFs were divided into six groups to receive different treatments as group A (blank control group), group B-E (ASMq in different concentration), and group F(5-Fu). Each group contains six specimens. The HSFs were cultured in vitro. After culture for 48 hours, the CCK8 test and flow cytometry methods were used to detect the proliferation, cell cycle and apoptosis.
RESULTSThe proliferation of HSFs in the B, C, D and E groups was inhibited at G2/M period, while it was inhibited at G0/S period in group F (P < 0.05). The inhibition effect of ASMq (0.1-1.0 mg/ml) on the fibroblasts enhanced in a concentration-dependent manner. Flow cytometry analysis with annexin V-FITC and PI staining confirmed the apoptotic. When HSFs were exposed to ASMq at 1.0 mg/ml (group E) for 48 h, the percentage of apoptotic cells increased to (43.7 +/- 2.58)%, which was significantly higher than that of blank control group (2.2 +/- 0.59)%. The induced apoptosis effect was also increased in a concentration-dependent manner.
CONCLUSIONASMq has a inhibitory effect on the proliferation and an enhancement effect on the apoptosis of fibroblast. ASMq could be used as an effective drug for treatment of hypertrophic scar.
Apoptosis ; Cell Cycle ; drug effects ; physiology ; Cell Division ; Cell Proliferation ; drug effects ; Cells, Cultured ; Cicatrix, Hypertrophic ; pathology ; Fibroblasts ; cytology ; drug effects ; Flow Cytometry ; Humans ; In Vitro Techniques ; Medicine, East Asian Traditional

Result Analysis
Print
Save
E-mail