1.Effects of interferon-α combined with homoharringtonine on K562 cell proliferation and β-catenin expression.
Yu-Ye SHI ; Wei-Ke CAO ; Xiao-Ning LIU ; Zhi-Kui DENG ; Hua GUO ; Wan-Ting FENG ; Li-Lin YE ; Jia-Bing ZHU ; Yu-Feng LI
Journal of Experimental Hematology 2012;20(1):43-47
The study was aimed to investigate the synergistically effect of interferon-α (IFN-α) and homoharringtonine (HHT) on the proliferation, apoptosis, cell cycle of K562 cells and the expression of β-catenin. The proliferation, apoptosis, cell cycle and β-catenin mRNA expression of K562 cells treated with IFN-α and/or HHT were assayed with MTT, flow cytometry or RT-PCR respectively. The results showed that HHT alone, but not IFN-α alone, displayed a proliferation inhibition, apoptosis induction, G(0)/G(1) phase block and down-regulation of β-catenin expression in K562 cells with concentration- and time-dependent manners. The expression level of β-catenin mRNA after being treated with HHT was 0.5576 ± 0.0373, which were lower than that in control group (0.9369 ± 0.0142). The down-regulation of β-catenin expression in group of IFN-α combined with HHT was higher significantly than that in HHT group (0.3737 ± 0.0529 vs 0.5576 ± 0.0373, P < 0.05). Otherwise, HHT combined with IFN-α did not demonstrate obvious toxicologic effect on bone marrow mononuclear cells. It is concluded that IFN-α combined with HHT can enhance the cytotoxic effect of HHT on K562 cells, which may be associated with down-regulation of β-catenin expression.
Cell Proliferation
;
drug effects
;
Harringtonines
;
pharmacology
;
Humans
;
Interferon-alpha
;
pharmacology
;
K562 Cells
;
beta Catenin
;
genetics
;
metabolism
2.Total Ginsenoside Extract from Panax ginseng Enhances Neural Stem Cell Proliferation and Neuronal Differentiation by Inactivating GSK-3β.
Kai-Li LIN ; Ji ZHANG ; Hau-Lam CHUNG ; Xin-Yi WU ; Bin LIU ; Bo-Xin ZHAO ; Stephen Cho-Wing SZE ; Ping-Zheng ZHOU ; Ken Kin-Lam YUNG ; Shi-Qing ZHANG
Chinese journal of integrative medicine 2022;28(3):229-235
OBJECTIVE:
To study the effects of total ginsenosides (TG) extract from Panax ginseng on neural stem cell (NSC) proliferation and differentiation and their underlying mechanisms.
METHODS:
The migration of NSCs after treatment with various concentrations of TG extract (50, 100, or 200 µ g/mL) were monitored. The proliferation of NSCs was examined by a combination of cell counting kit-8 and neurosphere assays. NSC differentiation mediated by TG extract was evaluated by Western blotting and immunofluorescence staining to monitor the expression of nestin and microtubule associated protein 2 (MAP2). The GSK-3β/β-catenin pathway in TG-treated NSCs was examined by Western blot assay. The NSCs with constitutively active GSK-3β mutant were made by adenovirus-mediated gene transfection, then the proliferation and differentiation of NSCs mediated by TG were further verified.
RESULTS:
TG treatment significantly enhanced NSC migration (P<0.01 or P<0.05) and increased the proliferation of NSCs (P<0.01 or P<0.05). TG mediation also significantly upregulated MAP2 expression but downregulated nestin expression (P<0.01 or P<0.05). TG extract also significantly induced GSK-3β phosphorylation at Ser9, leading to GSK-3β inactivation and, consequently, the activation of the GSK-3β/β-catenin pathway (P<0.01 or P<0.05). In addition, constitutive activation of GSK-3β in NSCs by the transfection of GSK-3β S9A mutant was found to significantly suppress TG-mediated NSC proliferation and differentiation (P<0.01 or P<0.05).
CONCLUSION
TG promoted NSC proliferation and neuronal differentiation by inactivating GSK-3β.
Animals
;
Cell Differentiation
;
Cell Proliferation
;
Ginsenosides/pharmacology*
;
Glycogen Synthase Kinase 3 beta/metabolism*
;
Neural Stem Cells/metabolism*
;
Panax
;
Plant Extracts/pharmacology*
;
Rats
;
beta Catenin/metabolism*
3.Amentoflavone induces apoptosis in SW480 human colorectal cancer cells via regulating β-catenin and caspase-3 expressions.
Yu YANG ; Wenjuan XU ; Kang PENG ; Xuegang SUN
Journal of Southern Medical University 2014;34(7):1035-1038
OBJECTIVETo investigate the role of β-catenin and caspase-3 in amentoflavone-induced apoptosis of human colorectal cancer SW480 cells.
METHODSMTT assay was used to detect the viability of SW480 cells exposed to amentoflavone, and flow cytometry was employed to assess the cell apoptosis. Western blotting was performed to determine the protein expressions of β-catenin and caspase-3 in the exposed cells.
RESULTSAmentoflavone dose-dependently inhibited the viability of SW480 cells, and a high concentration of amentoflavone (150 µmol/L) obviously induced apoptosis of the cells. Amentoflavone exposure caused significantly increased expression of caspase-3 and suppressed β-catenin expression in the cells.
CONCLUSIONAmentoflavone-induced apoptosis in SW480 human colorectal cancer cells is associated with altered expressions of β-catenin and caspase-3.
Apoptosis ; Biflavonoids ; pharmacology ; Caspase 3 ; metabolism ; Cell Line, Tumor ; drug effects ; Colorectal Neoplasms ; pathology ; Humans ; beta Catenin ; metabolism
4.Effects of sodium selenite on the expressions of beta-catenin and its target cyclin D1 in colorectal cancer cells HCT 116 and SW480.
Hui LUO ; Yang YANG ; Cai-Min XU
Acta Academiae Medicinae Sinicae 2011;33(6):654-658
OBJECTIVETo explore the effects of sodium selenite on the expressions of beta-catenin and cyclin D1 in colorectal cancer cells HCT 116 and SW480.
METHODSHCT 116 and SW480 cells were treated by 10 micromol/L sodium selenite at different time points. The expressions and transcription of beta-catenin and cyclin D1 were detected by Western blot analysis and reverse transcriptase polymerase chain reaction (RT-PCR), respectively. Meanwhile, the impact of MG132 (a proteasome inhibitor) pretreatment on the expressions of beta-catenin and cyclin D1 was observed through Western blot analysis. The interaction between beta-catenin and T cell factor 4 (TCF4) after selenite treatment was evaluated using co-immunoprecipitation assay.
RESULTSSodium selenite inhibited the expression of beta-catenin and transcription of its target such as cyclin D1. MG132 pretreatment prevented the inhibition of beta-catenin signaling triggered by selenite in HCT 116 and SW480 cells. Furthermore, selenite treatment disrupted the interaction between beta-catenin and TCF4 in HCT 116 and SW480 cells.
CONCLUSIONSSodium selenite can lower the expression levels of beta-catenin and its target cyclin D1, during which the proteasome-mediated degradative pathway may be involved. The decreased interaction between beta-catenin and TCF4 due to sodium selenite may be also involved in the regulation of beta-catenin signaling.
Cell Line, Tumor ; Colorectal Neoplasms ; metabolism ; Cyclin D1 ; metabolism ; HCT116 Cells ; Humans ; Sodium Selenite ; pharmacology ; beta Catenin ; metabolism
5.Sijunzi Decoction Inhibits Stemness by Suppressing β-Catenin Transcriptional Activity in Gastric Cancer Cells.
Yue-Jun LI ; Lin-Li LIAO ; Pei LIU ; Ping TANG ; Hong WANG ; Qing-Hua PENG
Chinese journal of integrative medicine 2022;28(8):702-710
OBJECTIVE:
To investigate a previously uncharacterized function of Sijunzi Decoction (SJZD) in inhibition of gastric cancer stem cells (GCSCs).
METHODS:
MKN74 and MKN45, two CD44 positive gastric cancer cell lines with stem cell properties were used. The cells were divided into 2 groups. Treatment group was treated with SJZD (1-5 mg/mL) for indicated time (48 h-14 days). The control group was treated with equal volume of phosphate buffered saline. Cell Counting Assay Kit-8 were used to measure cell viability. Spheroid colony formation and GCSCs marker expression were performed to determine GCSCs stemness. Cell fractionation and chromatin immunoprecipitation assays were used to assess the distribution and DNA-binding activity of β-catenin after SJZD treatment, respectively.
RESULTS:
SJZD treatment repressed cell growth and induced apoptosis in MKN74 and MKN45 cell lines (P<0.05). Moreover, SJZD dramatically inhibited formation of spheroid colony and expression of GCSC markers in GC cells (P<0.05). Mechanistically, SJZD reduced nuclear accumulation and DNA binding activity of β-catenin (P<0.05), the key regulator for maintaining CSC stemness.
CONCLUSION
SJZD inhibits GCSCs by attenuating the transcriptional activity of β-catenin.
Cell Line, Tumor
;
DNA/metabolism*
;
Drugs, Chinese Herbal/pharmacology*
;
Humans
;
Neoplastic Stem Cells/metabolism*
;
Stomach Neoplasms/genetics*
;
beta Catenin/metabolism*
6.Dexmedetomidine-mediated Wnt Pathway Inhibits Sevoflurane-induced Cognitive Impairment in Neonatal Rats.
Yue YANG ; Lei YANG ; Yu WU ; Jun YUAN
Acta Academiae Medicinae Sinicae 2021;43(2):235-246
Objective To explore the effect of dexmedetomidine(Dex)on sevoflurane-induced cognitive impairment in neonatal rats through Wnt signaling pathway. Methods Sixty 7-day-old SD rats were assigned into five groups:control group(without any intervention),Dex group(intraperitoneal injection of 25 μg/kg Dex),sevoflurane group(3% sevoflurane treatment for 4 hours),sevoflurane+Dex group(inhalation of 3% sevoflurane after injection of 25 μg/kg Dex for 4 hours),and sevoflurane+Dex+Wnt inhibitor group(Wnt inhibitor XAV393 and 25 μg/kg Dex were injected and 3% sevoflurane was inhaled for 4 hours).Three weeks later,Morris water maze was used to detect the cognitive function;TdT-mediated dUTP nick end labeling(TUNEL)staining was performed to detect the apoptosis of hippocampal neurons;neuronal nuclei (NeuN) staining was conducted to detect the survival of hippocampal neurons;Western blot was carried out to detect the expression of apoptosis-related proteins.The expression of the factors involved in Wnt/GSK-3β/β-catenin signaling pathway was detected by fluorescence quantitative polymerase chain reaction,and Western blot. Results Compared with the control group,there was no significant difference in the escape latency of Dex group(t=0.304,P=0.768);the escape latency in sevoflurane group(t=5.823,P=0.002),sevoflurane+Dex group(t=3.188,P=0.010),and sevoflurane+Dex+Wnt inhibitor group(t=5.784,P=0.002)was significantly prolonged.Compared with that in the sevoflurane group,the escape latency in sevoflurane+Dex group(t=3.646,P=0.005)was significantly shortened.Compared with that in sevoflurane+Dex group,the escape latency in sevoflurane+Dex+Wnt inhibitor group(t=3.296,P=0.008)was prolonged.Compared with that in the control group,the times of crossing platform in sevoflurane group(t=5.179, P=0.004),sevoflurane+Dex group(t=2.309,P=0.043),and sevoflurane+Dex+Wnt inhibitor group(t=3.871, P=0.003)decreased.Compared with that in sevoflurane group,the times of crossing platform in sevoflurane+Dex group(t=3.296,P=0.008)significantly increased.Compared with that in sevoflurane+Dex group,the times of crossing platform in sevoflurane+Dex+Wnt inhibitor group(t=2.361, P=0.041)reduced.Compared with the control group,there was no significant difference in the number of apoptotic cells in Dex group(t=1.920,P=0.127),and the number of apoptotic cells in sevoflurane group,sevoflurane+Dex group,and sevoflurane+Dex+Wnt inhibitor group increased by 16%(t=13.436,P=0.002),5%(t=7.752, P=0.001),and 11.5%(t=12.612,P=0.002),respectively.Compared with that in the sevoflurane group,the number of apoptotic cells in sevoflurane+Dex group and sevoflurane+Dex+Wnt inhibitor group decreased by 11%(t=8.521,P=0.002)and 5.5%(t=3.123,P=0.036),respectively.Compared with that in the sevoflurane+Dex group,the number of apoptotic cells in sevoflurane+Dex+Wnt inhibitor group increased by 6.5%(t=6.250,P=0.003).Compared with that in the control group,the number of positive cells in 0.15 mm
Animals
;
Animals, Newborn
;
Cognitive Dysfunction/chemically induced*
;
Dexmedetomidine/pharmacology*
;
Glycogen Synthase Kinase 3 beta
;
Rats
;
Rats, Sprague-Dawley
;
Sevoflurane/toxicity*
;
Wnt Signaling Pathway
;
beta Catenin/metabolism*
7.Low dose of triptolide ameliorates podocyte epithelial-mesenchymal transition induced by high dose of D-glucose via inhibiting Wnt3α/β-catenin signaling pathway activation.
Ge SHI ; Wei WU ; Yi-Gang WAN ; He Wei-Ming HEX ; Yue TU ; Wen-Bei HAN ; Bu-Hui LIU ; Ying-Lu LIU ; Zi-Yue WAN
China Journal of Chinese Materia Medica 2018;43(1):139-146
To explore the effects and molecular mechanisms of triptolide(TP)on improving podocyte epithelial-mesenchymal transition(EMT)induced by high dose of D-glucose(HG), the immortalized podocytes of mice were divided into the normal group(N), the high dose of D-glucose group(HG), the low dose of TP group(L-TP), the high dose of TP group(H-TP)and the mannitol group(MNT), and treated by the different measures respectively. More specifically, the podocytes in each group were separately treated by D-glucose(DG, 5 mmol·L⁻¹)or HG(25 mmol·L⁻¹)or HG(25 mmol·L⁻¹)+ TP(3 μg·L⁻¹)or HG(25 mmol·L⁻¹)+ TP(10 μg·L⁻¹)or DG(5 mmol·L⁻¹)+ MNT(24.5 mmol·L⁻¹). After the intervention for 24, 48 and 72 hours, firstly, the activation of podocyte proliferation was investigated. Secondly, the protein expression levels of the epithelial markers in podocytes such as nephrin and podocin, the mesenchymal markers such as desmin and collagen Ⅰ and the EMT-related mediators such as snail were detected respectively. Finally, the protein expression levels of Wnt3α and β-catenin as the key signaling molecules in Wnt3α/β-catenin pathway were examined severally. The results indicated that, HG could cause the low protein expression levels of nephrin and podocin and the high protein expression levels of desmin, collagen Ⅰ and snail in podocytes, and inducing podocyte EMT. On the other hand, HG could cause the high protein expression levels of Wnt3α and β-catenin in podocytes, and activating Wnt3α/β-catenin signaling pathway. In addition, L-TP had no effect on the activation of podocyte proliferation, the co-treatment of L-TP and HG could significantly recover the protein expression levels of nephrin and podocin, inhibit the protein expression levels of desmin, collagen I and snail in podocytes, thus, further improving podocyte EMT. And that, the co-treatment of L-TP and HG could obviously decrease the high protein expression levels of Wnt3α and β-catenin induced by HG in podocytes, and inhibit Wnt3α/β-catenin signaling pathway activation. On the whole, HG can induce podocyte EMT by activating Wnt3α/β-catenin signaling pathway; L-TP can ameliorate podocyte EMT through inhibiting Wnt3α/β-catenin signaling pathway activation, which may be one of the effects and molecular mechanisms .
Animals
;
Cells, Cultured
;
Diterpenes
;
pharmacology
;
Epithelial-Mesenchymal Transition
;
Epoxy Compounds
;
pharmacology
;
Glucose
;
Mice
;
Phenanthrenes
;
pharmacology
;
Podocytes
;
drug effects
;
Wnt Signaling Pathway
;
Wnt3A Protein
;
metabolism
;
beta Catenin
;
metabolism
8.Study on the relationship of beta-catenin level and sensitivity to Bortezomib of myeloma cell lines.
Li-Li ZHOU ; Wei-Jun FU ; Zhen-Gang YUAN ; Dong-Xing WANG ; Jian HOU
Chinese Journal of Hematology 2008;29(4):234-237
OBJECTIVETo explore the relationship of beta-catenin and sensitivity to Bortezomib of myeloma cell lines.
METHODSMyeloma cell lines RPMI8226, CZ-1 and NCI-H929 were treated with Bortezomib and 2ME2, alone or in combination. Typan blue dye exclusion and modified MTT were used to assess the cell viability with or without treatment. Annexin V-FITC and PI staining was performed to detect apoptosis rate. RT-PCR was used to detect beta-catenin mRNA and western blot to analyze beta-catenin protein.
RESULTSThe basic expression level of beta-catenin was different in tested myeloma cell lines: RPMI8226 was the most while NCI-H929 the least and CZ-1 the intermediate. IC50 of RPMI8226, CZ-1 and NCI-H929 were (49.8 +/- 0.6), (24.7 +/- 0.4) and (8.4 +/- 0.2) nmol/L, respectively. After the treatment of Bortezomib (at 0, 1, 5, 10 nmol/L), beta-catenin level of tested cell lines accumulated in a time and dose dependent manner for western blot, while no significant change was observed in the result of RT-PCR. The beta-catenin protein levels in the Bortezomib (5 nmol/L) and 2ME2 (1 micromol/L) treated cell group were much lower than that in Bortezomib (5 nmol/L) group, the decrease of the gray scale of beta-catenin/beta-actin was 64.03% for RPMI8226, 52.56% for CZ-1, 51.48% for NCI-H929, and the apoptosis rates were 8.00, 1.86 and 1.19 times increase compared to untreated group.
CONCLUSIONMyeloma cell lines with higher beta-catenin level are less sensitive to Bortezomib, and combination treatment of low dose 2ME2 and Bortezomib can reduce beta-catenin accumulation and enhance the sensitivity to Bortezomib.
Apoptosis ; drug effects ; Boronic Acids ; pharmacology ; Bortezomib ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; Humans ; Multiple Myeloma ; metabolism ; pathology ; Pyrazines ; pharmacology ; RNA, Messenger ; genetics ; beta Catenin ; genetics ; metabolism
9.Effect of arsenic trioxide combined with bortezomib on proliferation, apoptosis and beta-catenin level in myeloma cell lines.
Li-Li ZHOU ; Wei-Jun FU ; Zhen-Gang YUAN ; Dong-Xing WANG ; Jian HOU
Journal of Experimental Hematology 2008;16(1):84-88
The aim of this study was to investigate the effect of arsenic trioxide (As(2)O(3)) combined with bortezomib on the proliferation, apoptosis and beta-catenin level in myeloma cell lines. Myeloma cell lines RPMI8226, CZ-1 and NCI-H929 were treated with As(2)O(3) and bortezomib alone or in combination for 48 hours. Trypan blue dye exclusion and modified MTT were used to assess the cell viability. Flow cytometry with Annexin V-FITC and PI staining was used to detect the apoptosis rate. The beta-catenin level was analyzed by Western blot. The results showed that IC(50) of bortezomib to RPMI8226, CZ-1 and NCI-H929 were 46.9, 20.7 and 6.8 nmol/L, respectively. After the combination treatment with bortezomib (5 nmol/L) and As(2)O(3) (1 micromol/L), the cell viability of RPMI8226, CZ-1 and NCI-H929 decreased from 88.99%, 72.23%, 51.06% to 54.01%, 39.59%, 25.00%(p<0.05), the apoptosis rate increased from 11.1+/-0.1%, 26.8+/-1.7%, 46.8+/-5.5% to 36.1+/-2.2%, 60.4+/-3.8%, 76+/-5.6% (p<0.01) respectively. The Q value of two groups lies between enhancement and significant enhancement (1.198 - 3.75). Besides, beta-catenin levels in tested cell lines were decreased to 24.15%, 31.85%, 33.72% of their basic constitutions respectively (p<0.05). It is concluded that combination treatment of As(2)O(3) and bortezomib can enhance the proliferation inhibition and apoptosis induction of bortezomib to myeloma cell lines, reduce beta-catenin level, and increase the sensitivity of myeloma cell lines to bortezomib.
Antineoplastic Agents
;
pharmacology
;
Apoptosis
;
drug effects
;
Arsenicals
;
pharmacology
;
Boronic Acids
;
pharmacology
;
Bortezomib
;
Cell Proliferation
;
drug effects
;
Drug Synergism
;
Humans
;
Multiple Myeloma
;
metabolism
;
pathology
;
Oxides
;
pharmacology
;
Pyrazines
;
pharmacology
;
Tumor Cells, Cultured
;
beta Catenin
;
metabolism