Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
Heliyon ; 10(1): e23419, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38173472

ABSTRACT

The use of nanotechnology products with supermagnetic properties for targeted delivery of drugs has gained attention recently. Due to the anticancer features of Gingerol, the major phenolic compound from Ginger, this study aims to prepare Fe3O4@Glucose-Gingerol nanoparticles (NPs) and investigate their anticancer potential in a lung adenocarcinoma cell line. The physical and chemical features of the nanoparticles were investigated by FT-IR, XRD, zeta potential, DLS, EDS mapping, VSM, and electron microscope imaging. Cytotoxic effects of the nanoparticles for the A549 (lung adenocarcinoma) and MRC-5 (normal) cell lines was investigated by MTT assay. Furthermore, the effects of Fe3O4@Glucose-Gingerol nanoparticles on the expression of the CASP8, BAX, and BCL2 genes and the activity of Caspase 3 were characterized. The flow cytometry assay (annexin V/PI) was employed to find out the percentage of apoptotic cells. The Fe3O4@Glu-Gingerol NPs were spherical (42-67 nm), without elemental impurity, and with surface charge, DLS size, and magnetic saturation of -47.7 mV, 154 nm, and 35 emu/g, respectively. Fe3O4@Glu-Gingerol NPs showed a remarkable greater toxicity in the A549 cells than normal cell line with the 50 % inhibition concentration (IC50) of 190 and 554 µg/mL, respectively. Treatment of lung adenocarcinoma cells with the Fe3O4@Glu-Gingerol NPs led to an increase in cell apoptosis from 4.6 to 39.48 %. Also, the CASP8 and BAX genes were upregulated by 2.49 and 2.8 folds, respectively, while a downregulation by 0.75 folds was noticed for the BCL2. Moreover, apoptotic features were observed in Fe3O4@Glu-Gingerol NPs treated cells by Hoechst staining, and activation of Caspase 3 by 2.8 folds. This study revealed that the Fe3O4@Glu-Gingerol NPs have antiproliferative effects on the lung adenocarcinoma cell line by activation of intrinsic and extrinsic apoptosis that is a promising feature in cancer treatment.

2.
Biol Trace Elem Res ; 198(1): 98-108, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31983053

ABSTRACT

In recent years, researchers were attracted to nanomaterials components for their potential role in cancer treatment. This study aimed to develop a novel and biocompatible cobalt hydroxide (Co(OH)2) nano-flakes that is functionalized by glutamic acid (Glu) and conjugated to thiosemicarbazide (TSC) for anticancer activities against human breast cancer MCF-7 cells. Physico-chemical properties of the Co(OH)2@Glu-TSC nanomaterial are characterized by scanning electron microscopy (SEM), X-ray diffraction (XRD), energy dispersive X-ray (EDX) analysis, and Fourier-transform infrared (FT-IR) spectroscopy. MTT assay, flow cytometry, and caspase-3 activity analysis used for evaluating anticancer properties of the Co(OH)2@Glu-TSC nanomaterial. The MTT assay result showed cellular uptake of Co(OH)2@Glu-TSC and cell viability loss in a concentration-dependent. Results of flow cytometry and caspase-3 activity analysis indicated the stimulation of apoptosis through an increase in Caspase-3 and nucleus fragmentation. In general, our findings indicate the anticancer activities of Co(OH)2@Glu-TSC nanomaterial and so it can be considered as a new treatment for breast cancer. However, further in vivo studies are required to evaluate the accumulation of Co(OH)2@Glu-TSC nanomaterial in healthy organs, such as the liver, kidneys, brain, and testes, and potential toxic effects.


Subject(s)
Breast Neoplasms , Glutamic Acid , Breast Neoplasms/drug therapy , Cobalt , Female , Humans , Hydroxides , Semicarbazides , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL