1.Effects of silencing HMGB1 combined with docetaxel chemotherapy on the proliferation and apoptosis of prostate cancer cells and its action mechanism.
National Journal of Andrology 2021;27(10):876-881
Objective:
To investigate the effect of silencing the high-mobility group box-1 protein (HMGB1) combined with docetaxel (DTX) on the proliferation and apoptosis of PCa cells and its possible action mechanism.
METHODS:
The expression of HMGB1 mRNA in different PCa cell lines and normal prostatic epithelial cells was detected by RT-qPCR. The PC-3 cells were transfected with different HMGB1 small interfering RNAs (si-HMGB1, si-HMGB1-2 and si-HMGB1-3), and the silencing effect was detected. The effects of different concentrations of DTX on the proliferation of the PC-3 cells was determined by MTT. Then the PC-3 cells were randomly divided into five groups: control (conventional culture), si-HMGB1-NC (si-HMGB1-NC transfection), si-HMGB1 (si-HMGB1-3 transfection), DTX (20 nmol/L DTX), and si-HMGB1+DTX (si-HMGB1-3+20 nmol/L DTX transfection), followed by measurement of the survival rate of the cells by MTT, their apoptosis rate by flow cytometry, and the expressions of HMGB1, B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X (Bax) proteins in different groups by Western blot.
RESULTS:
The expression of HMGB1 mRNA in the PC-3 cells was the highest and the lowest after transfection with si-HMGB1-3. DTX inhibited the proliferation of the PC-3 cells at various concentrations. Compared with the control group, the si-HMGB1 and DTX groups showed significantly decreased A values, cell survival rates and HMGB1 and Bcl-2 expressions, but increased cell apoptosis rates and Bax expressions (P < 0.05). In comparison with the si-HMGB1 and DTX groups, the si-HMGB1+DTX group exhibited a remarkably decreased A value, cell survival rate and Bcl-2 expression, but increased cell apoptosis and Bax expression. The expression of the HMGB1 protein was markedly lower in the si-HMGB1+DTX than in the DTX group (P < 0.05).
CONCLUSIONS
Silencing HMGB1 combined with DTX chemotherapy can inhibit the proliferation and promote the apoptosis of PCa cells, which may be attributed to its regulatory effect on the expressions of the Bcl-2 family-related proteins.、.
Apoptosis
;
Cell Proliferation
;
Docetaxel/pharmacology*
;
HMGB1 Protein/genetics*
;
Humans
;
Male
;
Prostatic Neoplasms/genetics*
2.Study on the Metabolic Reprogramming of Lung Cancer Cells Regulated by Docetaxel Based on Metabolomics.
Haichao SUN ; Hailong PIAO ; Huan QI ; Min YAN ; Hongxu LIU
Chinese Journal of Lung Cancer 2019;22(4):208-215
BACKGROUND:
Docetaxel is a commonly used anti-tumor drug in clinic, especially as the first-line drug for advanced non-small cell lung cancer (NSCLC). However, the molecular mechanism of docetaxel against NSCLC is still unclear. Increasing studies have shown that metabolic reprogramming of tumor cells plays an important role in tumorigenesis. The aim of this study was to investigate the effects of docetaxel on the metabolic pathway of NSCLC cells based on metabolomics analysis and biological means.
METHODS:
First, we performed CCK8 assay to analyze the effects of docetaxel on cell viability of NSCLC cells and also to screen the appropriate drug concentration. Then, the differential metabolites of docetaxel-treated and untreated NSCLC cells were analyzed by gas chromatography-mass spectrometry based metabolomics. Finally, the effects of docetaxel on the expression levels of key enzymes that regulate the relevant metabolic pathways were determined by Western blot.
RESULTS:
Docetaxel inhibited cell viability of A549 and H1299 cells in a concentration- and time-dependent manner. With the prolonged treatment time of docetaxel, the apoptotic sensitive protein poly (ADP-ribose) polymerase (PARP) was gradually activated to form a P89 fragment. Metabolomics analysis showed that eight metabolites were significantly changed in both A549 and H1299 cells following docetaxel treatment, which were mainly in the tricarboxylic acid (TCA) cycle pathway. Moreover, after docetaxel treatment, the protein expression levels of isocitrate dehydrogenases, the key regulators of the TCA cycle, were obviously decreased in both A549 and H1299 cells.
CONCLUSIONS
Our findings suggest that the effect of docetaxel-induced proliferation inhibition and apoptosis in NSCLC might be associated with down-regulation of isocitrate dehydrogenases and suppression of the TCA cycle pathway.
A549 Cells
;
Apoptosis
;
drug effects
;
Carcinoma, Non-Small-Cell Lung
;
pathology
;
Docetaxel
;
pharmacology
;
Humans
;
Lung Neoplasms
;
pathology
;
Metabolomics
3.Preparation of docetaxel-loaded nanomicelles and their anti-Lewis lung cancer effect in vitro.
Ya-Jie WANG ; Jie WANG ; Dan-Li HAO ; Qiao-Xin YUE ; Ran XIE ; Ge-Jing DE ; Hong YI ; Chen ZANG ; Qing-He ZHAO ; Yan-Jun CHEN
China Journal of Chinese Materia Medica 2019;44(11):2251-2259
Docetaxel-loaded nanomicelles were prepared in this study to improve the solubility and tumor targeting effect of docetaxel(DTX),and further evaluate their anticancer effects in vitro. PBAE-DTX nanomicelles were prepared by film-hydration method with amphiphilic block copolymer polyethyleneglycol methoxy-polylactide(PELA) and pH sensitive triblock copolymer polyethyleneglycol methoxy-polylactide-poly-β-aminoester(PBAE) were used respectively to prepare PELA-DTX nanomicelles and PBAE-DTX nanomicelles. The nanomicelles were characterized by physicochemical properties and the activity of mice Lewis lung cancer cells was studied. The results of particle size measurement showed that the blank micelles and drug-loaded micelles had similar particle sizes, ranging from 10 to 100 nm. The particle size of PBAE micelles was changed under weak acidic conditions, with good pH response. The encapsulation efficiency of the above two types of DTX-loaded nanomicelles determined by HPLC was(93.8±1.70)% and(87.2±4.10)%, and the drug loading amount was(5.3±0.10)% and(4.9±0.05)%,respectively. Furthermore,the DTX micelles also showed significant inhibitory effects on Lewis lung cancer cells by MTT assay, and pH-sensitive PBAE-DTX showed better cytotoxicity. The results of flow cytometry indicated that,the apoptosis rate of lung cancer Lewis cells was(20.72±1.47)%,(29.71±2.38)%,and(40.91±1.90)%(P<0.05) at 48 h after treatment in DTX,PELA-DTX,and PBAE-DTX groups. The results showed that different docetaxel preparations could promote the apoptosis of Lewis cells, and PBAE-DTX had stronger apoptotic-promoting effect. The pH-sensitive DTX-loaded micelles are promising candidates in developing stimuli triggered drug delivery systems in acidic tumor micro-environments with improved inhibitory effects of tumor growth on Lewis lung cancer.
Animals
;
Antineoplastic Agents
;
pharmacology
;
Cell Line, Tumor
;
Docetaxel
;
pharmacology
;
Drug Carriers
;
Lung Neoplasms
;
drug therapy
;
pathology
;
Mice
;
Micelles
;
Nanoparticles
;
Particle Size
;
Taxoids
4.Bufalin Induces Programmed Necroptosis in Triple-Negative Breast Cancer Drug-Resistant Cell Lines through RIP1/ROS-Mediated Pathway.
Xiao-Dan LIU ; Cheng-Yang SONG ; Cui-Cui KONG ; Xin TIAN
Chinese journal of integrative medicine 2022;28(10):900-908
OBJECTIVE:
To explore the effect and mechanism of action of bufalin in triple-negative breast cancer (TNBC) drug-resistant cell lines.
METHODS:
The normal human mammary epithelial cell line, TNBC cell line, TNBC adriamycin-resistant cell line, and TNBC docetaxel-resistant cell line were treated with different doses of bufalin (0-1,000 nmol/L) at different time points (0-72 h). Propidium iodide staining, AV-FITC/PI double staining, Hoechst 33342/PI double staining and transmission electron microscopy (TEM) were used to evaluate the death patterns of the cell lines.
RESULTS:
Bufalin killed the TNBC cell line and its drug-resistant cell lines in a dose/time-dependent manner (all P<0.01). After treatment with bufalin for 24 h, the adriamycin-resistant cell line showed a co-existing pattern of necroptosis and apoptosis. However, at 48 h, necroptosis was the main manifestation. After treatment with bufalin, the expressions of tumor necrosis factor α, phospho-tumor necrosis factor receptor 1, phospho-receptor interacting protein 1 and c-caspase 3 increased (all P<0.01), the killing effect of bufalin could be mostly inhibited by NEC-1, and by z-VAD-fmk (both P<0.01). Besides, the intracellular reactive oxygen species (ROS) levels increased considerably (P<0.01), the antioxidant N-acetyl cysteine or Nec-1 could inhibit the increase of ROS level and the killing effect of bufalin (all P<0.01). The adriamycin-resistant cell line exhibited necroptosis characteristic after 48 h of bufalin treatment under TEM.
CONCLUSIONS
Bufalin could induce necroptosis through RIP1/ROS-mediated pathway to kill the drug-resistant TNBC cell lines. This finding provides critical experimental data and theoretical basis for the clinical application of bufalin to overcome the difficulties in the treatment of TNBC.
Antioxidants/pharmacology*
;
Apoptosis
;
Bufanolides
;
Caspase 3/metabolism*
;
Cell Line
;
Cell Line, Tumor
;
Cysteine/pharmacology*
;
Docetaxel/pharmacology*
;
Doxorubicin/pharmacology*
;
Fluorescein-5-isothiocyanate/pharmacology*
;
Humans
;
Necroptosis
;
Propidium/pharmacology*
;
Reactive Oxygen Species/metabolism*
;
Receptors, Tumor Necrosis Factor
;
Triple Negative Breast Neoplasms/drug therapy*
;
Tumor Necrosis Factor-alpha/pharmacology*
5.Expression of AXL enhances docetaxel-resistance of prostate cancer cells.
Jian-Zhong LIN ; Jia-Geng ZHU ; Hong-Fei WU ; Jiu-Ming LI ; Wei DE ; Zeng-Jun WANG
National Journal of Andrology 2017;23(4):302-308
Objective:
To explore the effect of the AXL expression on the chemosensitivity of prostate cancer PC-3 and DU145 cells to docetaxel and possible mechanisms.
METHODS:
Using Western blot, we examined the expressions of the AXL protein, p-AXL and Gas6 in the docetaxel-resistant PC-3 (PC-3-DR) and DU145 (DU145-DR) cells stimulated with gradually increased concentrations of docetaxel. We transfected the PC-3 and DU145 cells with negative NC ShRNA and AXL-ShRNA, respectively, which were confirmed to be effective, detected the proliferation, apoptosis and cycle distribution of the cells by CCK8, MTT and flow cytometry after treated with the AXL-inhibitor MP470 and/or docetaxel, and determined the expression of the ABCB1 protein in the PC-3-DR and DU145-DR cells after intervention with the AXL-inhibitor R428 and/or docetaxel.
RESULTS:
The expression of the AXL protein in the PC-3 and DU145 cells was significantly increased after docetaxel treatment (P <0.05). The expressions AXL and p-AXL were remarkably higher (P <0.05) while that of Gas6 markedly lower (P <0.05) in the PC-3 and DU145 than in the PC-3-DR and DU145-DR cells. The inhibitory effect of docetaxel on the proliferation and its enhancing effect on the apoptosis of the PC-3 and DU145 cells were significantly decreased at 48 hours after AXL transfection (P <0.05). MP470 obviously suppressed the growth and promoted the apoptosis of the PC-3-DR and DU145-DR cells, with a higher percentage of the cells in the G2/M phase when combined with docetaxel than used alone (P <0.05). R428 markedly reduced the expression of ABCB1 in the PC-3-DR and DU145-DR cells, even more significantly in combination with docetaxel than used alone (P <0.05).
CONCLUSIONS
The elevated expression of AXL enhances the docetaxel-resistance of PC-3 and DU145 prostate cancer cells and AXL intervention improves their chemosensitivity to docetaxel, which may be associated with the increased cell apoptosis in the G2/M phase and decreased expression of ABCB1.
ATP Binding Cassette Transporter, Subfamily B, Member 1
;
metabolism
;
Antineoplastic Agents
;
pharmacology
;
Apoptosis
;
drug effects
;
Cell Count
;
Cell Cycle
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Docetaxel
;
Drug Resistance, Neoplasm
;
Humans
;
Intercellular Signaling Peptides and Proteins
;
metabolism
;
Male
;
Prostatic Neoplasms
;
drug therapy
;
metabolism
;
pathology
;
Proto-Oncogene Proteins
;
drug effects
;
genetics
;
metabolism
;
Pyrimidines
;
pharmacology
;
RNA, Small Interfering
;
Receptor Protein-Tyrosine Kinases
;
drug effects
;
genetics
;
metabolism
;
Taxoids
;
pharmacology