Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Life Sci ; 264: 118598, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33189818

ABSTRACT

AIMS: This study aimed to explore the role of Isoliquiritigenin (ISL) in the proliferation and invasion of melanoma cells and investigate the mechanism of action of this compound. MAIN METHODS: The functional roles of ISL in melanoma cells were determined by CCK8 assay, colony formation assay, flow cytometry and wound healing assay. The antitumor activity of ISL was assessed in vivo in a mouse xenograft model using A2058 cells. Quantitative real-time PCR analysis (RT-qPCR) and western blot assays were used to evaluate the gene and protein expression in cell lines or tumor tissue samples. Bioinformatic analysis, luciferase reporter assay, and gene set enrichment analysis (GSEA) were performed to confirm the mechanism of ISL effect on cell growth and metastasis of melanoma. KEY FINDINGS: ISL suppressed proliferation and migration of melanoma cells via downregulation of miR-27a expression. The inhibitory effect of ISL on growth and metastasis of melanoma cells was reversed by ectopic expression of miR-27a. Bioinformatic analysis showed that miR-27a targets POU class 2 homeobox 3 (POU2F3); this result was verified by the luciferase reporter assay and by a decrease in the expression of POU2F3 by miR-27a intervention. GSEA demonstrated that POU2F3 is associated with the c-MYC/p53 signaling pathway and metastasis. POU2F3 knockdown reversed the inhibitory effect of ISL on the growth and metastasis of melanoma. Additionally, POU2F3 was found to be downregulated in melanoma tissue samples and was negatively correlated with miR-27a. SIGNIFICANCE: ISL inhibits proliferation and metastasis of melanoma via the miR-27a/POU2F3/c-MYC/p53 axis; these results may provide a new thought for the treatment of melanoma.


Subject(s)
Apoptosis/drug effects , Chalcones/administration & dosage , Flavonoids/administration & dosage , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Animals , Apoptosis/physiology , Cell Line, Tumor , Female , Humans , Melanoma/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays/methods
2.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(4): 401-408, 2019 Apr 30.
Article in Chinese | MEDLINE | ID: mdl-31068282

ABSTRACT

OBJECTIVE: To explore whether bortezomib and a Bcl-2 inhibitor exhibit synergistic anti-tumor effect in human acute T lymphoblastic leukemia cells. METHODS: MTT assay was used to determine the cytotoxicity of bortezomib in the absence or presence of Bcl-2 inhibitors (obatoclax, AT-101 and ABT-199) in Jurkat cells. The effects of drug treatment on the expression of Bcl-2 family proteins, LC3B, p62, ubiquitin, BiP/Grp78, p-JNK, p-p38 and CHOP proteins were examined by Western blotting. Flow cytometry was used to determine the effects of bortezomib and Bcl-2 inhibitors (obatoclax, AT-101 and ABT-199) on cell apoptosis. Quantitative real-time PCR was used to measure the mRNA expression levels of the key regulatory factors of unfolded protein reaction (UPR). A zebrafish xenograft model was used to study the anti-tumor effect of bortezomib, obatoclax and their combination in vivo. RESULTS: Bortezomib or Bcl-2 inhibitors alone inhibited the cell viability of Jurkat cells, but only obatoclax and bortezomib showed synergistic cytotoxicity and pro-apoptotic effect. Obatoclax, rather than AT-101 and ABT- 199, blocked autophagic flux in the cells evidenced by concomitant accumulation of LC3B-Ⅱ and p62. Both bortezomib and obatoclax alone caused accumulation of polyubiquinated proteins, and their combination showed a synergistic effect, which was consistent with their synergistic cytotoxicity. The dual blockade of proteasome and autophagy by the combination of bortezomib and obatoclax triggered unfolded protein response followed by cell apoptosis. Preventing UPS dysfunction by tauroursodeoxycholic acid (TUDCA) significantly attenuated the cytotoxicity and pro-apoptotic effect of bortezomib in combination with obatoclax. In zebrafish xenograft models, bortezomib combined with obatoclax significantly decreased tumor foci formation. CONCLUSIONS: Bortezomib and obatoclax for dual blockade of protein degradation pathways show synergistic anti-tumor effect in human acute T lymphoblastic leukemia cells.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antineoplastic Agents , Apoptosis , Bortezomib , Cell Line, Tumor , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Humans , Indoles , Proteolysis , Proto-Oncogene Proteins c-bcl-2 , Pyrroles
3.
Oncol Rep ; 41(2): 1415-1423, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30483745

ABSTRACT

AT­101, an orally available and well­tolerated natural pan­Bcl­2 family protein inhibitor, has been reported to be effective against a variety of cancers. However, the mechanisms whereby AT­101 exhibits anticancer activity have not been fully elucidated. In this study, we demonstrated that AT­101 reduced the cell viability of human esophageal cancer cells by inducing G1/G0 phase arrest and apoptosis. Apoptotic cell death occurred later than cell cycle arrest, as evidenced by an increase in the proportion of Annexin V­positive cells and cleaved caspase­3, ­9 and PARP protein levels. AT­101 markedly downregulated the protein levels of phospho­retinoblastoma (Ser 780) and cyclin D1, whereas it elevated protein levels of p53 and p21Waf1/Cip1, contributing to the inhibition of cell cycle progression. Moreover, AT­101 substantially reduced ß­catenin expression. XAV­939, a small molecule that inhibits the Wnt/ß­catenin signaling pathway by facilitating ß­catenin degradation, lowered ß­catenin and cyclin D1 protein expression to an extent similar to AT­101. XAV­939 alone resulted in G1/G0 phase arrest and further induced cell cycle arrest in combination with AT­101, suggesting that the ß­catenin/cyclin D1 signaling pathway mediated, at least in part, the cell cycle arrest induced by AT­101. The present study may shed new light on the anticancer activity of AT­101 in relation to cell cycle arrest as well as apoptosis in human esophageal cancer cells.

4.
Acta Pharmacol Sin ; 38(12): 1632-1641, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28713162

ABSTRACT

Both the anti- and pro-apoptotic members of the Bcl-2 family are regulated by a conserved Bcl-2 homology (BH3) domain. ABT-263 (Navitoclax), a novel BH3 mimetic and orally bioavailable Bcl-2 family inhibitor with high affinity for Bcl-xL, Bcl-2 and Bcl-w has entered clinical trials for cancer treatment. But the anticancer mechanisms of ABT-263 have not been fully elucidated. In this study we investigated the effects of ABT-263 on human esophageal cancer cells in vitro and to explore its anticancer mechanisms. Treatment with ABT-263 dose-dependently suppressed the viability of 3 human esophageal cancer cells with IC50 values of 10.7±1.4, 7.1±1.5 and 8.2±1.6 µmol/L, in EC109, HKESC-2 and CaES-17 cells, respectively. ABT-263 (5-20 µmol/L) dose-dependently induced G1/G0-phase arrest in the 3 cancer cell lines and induced apoptosis evidenced by increased the Annexin V-positive cell population and elevated levels of cleaved caspase 3, cleaved caspase 9 and PARP. We further demonstrated that ABT-263 treatment markedly increased the expression of p21Waf1/Cip1 and decreased the expression of cyclin D1 and phospho-Rb (retinoblastoma tumor suppressor protein) (Ser780) proteins that contributed to the G1/G0-phase arrest. Knockdown of p21Waf1/Cip1 attenuated ABT-263-induced G1/G0-phase arrest. Moreover, ABT-263 treatment enhanced pro-survival autophagy, shown as the increased LC3-II levels and decreased p62 levels, which counteracted its anticancer activity. Our results suggest that ABT-263 exerts cytostatic and cytotoxic effects on human esophageal cancer cells in vitro and enhances pro-survival autophagy, which counteracts its anticancer activity.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , G1 Phase Cell Cycle Checkpoints/drug effects , Resting Phase, Cell Cycle/drug effects , Sulfonamides/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
5.
J Agric Food Chem ; 65(22): 4384-4394, 2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28471170

ABSTRACT

The persistence of latent HIV reservoirs presents a significant challenge to viral eradication. Effective latency reversing agents (LRAs) based on "shock and kill" strategy are urgently needed. The natural phytoalexin resveratrol has been demonstrated to enhance HIV gene expression, although its mechanism remains unclear. In this study, we demonstrated that resveratrol was able to reactivate latent HIV without global T cell activation in vitro. Mode of action studies showed resveratrol-mediated reactivation from latency did not involve the activation of silent mating type information regulation 2 homologue 1 (SIRT1), which belonged to class-3 histone deacetylase (HDAC). However, latent HIV was reactivated by resveratrol mediated through increasing histone acetylation and activation of heat shock factor 1 (HSF1). Additionally, synergistic activation of the latent HIV reservoirs was observed under cotreatment with resveratrol and conventional LRAs. Collectively, this research reveals that resveratrol is a natural LRA and shows promise for HIV therapy.


Subject(s)
HIV Infections/drug therapy , HIV-1/physiology , Heat-Shock Proteins/genetics , Histones/metabolism , Stilbenes/pharmacology , Virus Latency/drug effects , Acetylation/drug effects , Cell Line , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/drug effects , Heat-Shock Proteins/metabolism , Humans , Resveratrol
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(4): 506-13, 2016 Apr.
Article in Chinese | MEDLINE | ID: mdl-27113178

ABSTRACT

OBJECTIVE: To explore whether MG-132 could enhance the anti-tumor activity of obatoclax against esophageal cancer cell line CaES-17. METHODS: MTT assay was used to determine the cytotoxicity of obatoclax and MG-132 in CaES-17 cells. The IC(50) of obatoclax and MG-132 were used to determine the molar ratio (1:2.4) of the two drugs for combined treatment of the cells. The concentrations of obatoclax and MG-132 ranged from 1/8 IC(50) to 4 IC(50) after serial dilution, and their combination index (CI) was calculated using CompuSyn software. The expression of ubiquitin and the cleavage of PARP, caspase-9, phospho-histone H3 and phospho-aurora A/B/C in the exposed cells were examined with Western blotting; the cell apoptosis was measured by flow cytometry with Annexin V staining, and the percentage of cells in each cell cycle phase was also determined by flow cytometry. RESULTS: The CI of obatoclax and MG-132 was 0.296 for a 50% inhibition of Caes-17 cells and was 0.104 for a 95% inhibition. The cells treated with obatoclax or MG-132 alone showed increased expression of ubiquitin and cleavage of PARP and caspase-9. Compared with the cells treated with obatoclax or MG-132 alone, the cells with a combined treatment exhibited significantly increased expression of ubiquitin, cleavage of PARP and caspase-9, and expression of phospho-Histone H3 (P<0.05). The combined treatment of the cells also resulted in significantly increased expression of phospho-Aurora A/B/C compared with obatoclax treatment alone. The cells with the combined treatment showed significantly higher percentages of apoptotic cells and cells in sub-G(1) and G(2)/M phases compared with the cells treated with either of the drugs (P<0.05). CONCLUSION: Obatoclax combined with MG-132 shows a significant synergistic anti-tumor effect against esophageal cancer CaES-17 cells by inducing apoptosis and cell cycle arrest.


Subject(s)
Esophageal Neoplasms/pathology , Leupeptins/pharmacology , Pyrroles/pharmacology , Apoptosis , Caspase 9/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor/drug effects , Histones/metabolism , Humans , Indoles , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...