11 research outputs found

    Anticancer Effects of ZnO/CNT@Fe3O4 in AML-Derived KG1 Cells: Shedding Light on Promising Potential of Metal Nanoparticles in Acute Leukemia

    Get PDF
    Background: Therapeutic approaches for acute myeloid leukemia (AML) have remained largely unchanged for over 40 years and cytarabine and an anthracycline (e.g., daunorubicin) backbone is the main induction therapy for these patients. Resistance to chemotherapy is the major clinical challenge and contributes to short-term survival with a high rate of disease recurrence. Given the established efficacy of nanoparticles in cancer treatment, this study was designed to evaluate the anticancer property of our novel nanocomposite in the AML-derived KG1 cells.Materials and Methods: To assess the anti-leukemic effects of our nanocomposite on AML cells, we used MTT and trypan blue assays. Flow cytometric analysis and q-RT-PCR were also applied to evaluate the impact of nanocomposite on cell cycle and apoptosis.Results: Our results outlined that ZnO/CNT@Fe3O4 decreased viability and metabolic activity of KG1 cells through induction of G1 arrest by increasing the expression of p21 and p27 cyclin-dependent kinase inhibitors and decreasing c-Myc transcription. Moreover, ZnO/CNT@Fe3O4 markedly elevated the percentage of apoptotic cells which was coupled with a significant alteration of Bax and Bcl-2 expressions. Synergistic experiments showed that ZnO/CNT@Fe3O4 enhances the cytotoxic effects of Vincristine on KG1 cells.Conclusion: In conclusion, this study sheds light on the potent anti-leukemic effects of ZnO/CNT@Fe3O4 and provides evidence for the application of this agent in the treatment of acute myeloid leukemia.</p

    ZnO/CNT@Fe3O4 induces ROS-mediated apoptosis in chronic myeloid leukemia (CML) cells: an emerging prospective for nanoparticles in leukemia treatment

    No full text
    AbstractThe advent of nanoparticles revolutionised the drug delivery systems in human diseases; however, their prominent role was highlighted in the cancer-based therapies, where this technology could specifically target cancer cells. Herein, we decided to combine two nanoparticles Fe3O4 and ZnO to fabricate a new anti-cancer nanocomposite. Noteworthy, hydroxylated carbon nanotube (CNT) was used to increase the water-solubility of the compound, improving its uptake by malignant cells. This study was designed to evaluate the anticancer property as well as the molecular mechanisms of ZnO/CNT@Fe3O4 nanocomposite cytotoxicity in CML-derived K562 cells. Our results outlined that ZnO/CNT@Fe3O4 decreased the proliferative capacity of K562 cells through induction of G1 arrest and induced apoptosis probably via ROS-dependent upregulation of FOXO3a and SIRT1. The results of qRT-PCR analysis also demonstrated that while ZnO/CNT@Fe3O4 significantly increased the expression of pro-apoptotic genes in K562 cells, it had no significant inhibitory effect on the expression levels of anti-apoptotic target genes of NF-κB; proposing an attenuating role of NF-κB signalling pathway in K562 cell response to ZnO/CNT@Fe3O4. Synergistic experiment showed that ZnO/CNT@Fe3O4 could enhance the cytotoxic effects of imatinib on K562 cells. Overall, it seems that pharmaceutical application of nanocomposites possesses novel promising potential for leukaemia treatment strategies

    Novel application of PEG/SDS interaction as a wettability modifier of hydrophobic carbonate surfaces

    No full text
    Wettability alteration of carbonate reservoirs from oil-wet to water-wet is an important method to increase the efficiency of oil recovery. Interaction between surfactants and polymers can enhance the effectiveness of surfactants in EOR applications. In this study, the interaction of polyethylene glycol (PEG) with an ionic surfactant, sodium dodecyl sulphate (SDS), is evaluated on an oil-wet carbonate rock surface by using contact angle measurements. The results reveal that wettability alteration of carbonate rocks is achieved through PEG/SDS interaction on the rock surface above a critical aggregation concentration (CAC). The behaviour of PEG/SDS aqueous solutions is evaluated using surface and interfacial tension measurements. Furthermore, the effect of PEG and SDS concentrations and impact of electrolyte addition on PEG/SDS interaction are investigated. It is shown that electrolyte (NaCl) can effectively decrease the CAC values and accordingly initiate the wettability alteration of rocks. Moreover, in a constant SDS concentration, the addition of NaCl leads to a reduction in the contact angle, which can also be obtained by increasing the aging time, temperature and pre-adsorption of PEG on the rock surface.Applied Geophysics and Petrophysic

    ZnO Q-dots as a potent therapeutic nanomedicine for <i>in vitro</i> cytotoxicity evaluation of mouth KB44, breast MCF7, colon HT29 and HeLa cancer cell lines, mouse ear swelling tests <i>in vivo</i> and its side effects using the animal model

    No full text
    <p>Nanoformulations derived from fine porous ZnO quantum dot nanoparticles (QD NPs) can offer strong potential medical applications; especially in cancer therapy. ZnO QD NPs was synthesized by sol–gel hydrothermal process, fast cold quenching and further smart surface functionalization methods to obtain ultrasmall size (1–4 nm) NPs. ZnO nanopolymer, a wetting agent, PEG co-solvent and water<b>/</b>oil emulsion stabilizer were considered in our nanofluid formulation. The resulting nanofluid was characterized by SEM, FTIR, photoluminescence, band gap energy, zeta potential and UV–Vis spectroscopy. The cytotoxic effects on the growth of four cancer cell lines were evaluated by MTT assay. The IC<sub>50</sub> (µg/ml) values of 30, 41, 40 and 35 for KB44, MCF-7, HT29 and HeLa cells, respectively, after 48 h of nanoformulation treatment suggested the cytotoxic effect of this nanoformulation on these cell lines in a concentration-dependent manner (<i>p</i> < .05). ZnO nanofluid destroyed cancer cell lines more efficiently than the normal HFF-2 (IC<sub>50 </sub>= 105 µg/ml). The reduction in cell viability in response to ZnO nanofluid treatment induced apoptosis in the cultured cells. Skin sensitization test plus antibacterial activity were also measured. Side effect tests on 70 white mice <i>in vivo</i> resulted in only 3–4 abnormal situations in hepatic tissue section possibly due to the idiosyncratic drug reactions.</p
    corecore