Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Cell Oncol ; 11(1): 2299046, 2024.
Article in English | MEDLINE | ID: mdl-38196561

ABSTRACT

In this study, we investigated the effects of an ethanolic extract of Mangifera indica L. kernel on the viability and proliferation of human lung cancer cells. We utilized MTT and BrdU cell proliferation assays, morphological assessments, cell cycle analyses, and apoptosis assays to investigate the extract's effects on lung cancer (A549 and NCI-H292) and normal lung (MRC-5) cells. The extract demonstrated a toxicity toward cancer cells compared to normal cells with dose-dependent anti-proliferative effect on lung cancer cells. The extract also caused differential effects on the cell cycle, inducing G0/G1 arrest and increasing the Sub-G1 population in both lung cancer and normal lung cells. Notably, the extract induced loss of membrane integrity, shrinkage, membrane blebbing, and apoptosis in lung cancer cells, while normal cells exhibited only early apoptosis. Furthermore, the extract exhibited higher toxicity towards NCI-H292 cells, followed by A549 and normal MRC-5 cells in decreasing order of potency. Our results suggest that the ethanolic extract of M. indica L. kernel has significant potential as a novel therapeutic agent for treating lung cancer cells, given its ability to induce apoptosis in cancer cell lines while causing minimal harm to normal cells.

2.
Heliyon ; 9(5): e15560, 2023 May.
Article in English | MEDLINE | ID: mdl-37159701

ABSTRACT

Silver-doped magnesia nanoparticles (Ag/MgO) were synthesized using the precipitation method and characterized by various techniques such as X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), thermal gravimetric analysis (TGA), Brunner-Emmett-Teller (BET) surface area measurements, and dispersive X-ray spectroscopy (EDX). The morphology of Ag/MgO nanoparticles was determined by transmission and scanning electron microscopy, which revealed cuboidal shaped nanoparticles with sizes ranging from 31 to 68 nm and an average size of 43.5 ± 10.6 nm. The anticancer effects of Ag/MgO nanoparticles were evaluated on human colorectal (HT29) and lung adenocarcinoma (A549) cell lines, and their caspase-3, -8, and -9 activities, as well as Bcl-2, Bax, p53, cytochrome C protein expressions were estimated. Ag/MgO nanoparticles showed selective toxicity towards HT29 and A549 cells while remaining relatively innocuous towards the normal human colorectal, CCD-18Co, and lung, MRC-5 cells. The IC50 values of Ag/MgO nanoparticles on the HT29 and A549 cells were found to be 90.2 ± 2.6 and 85.0 ± 3.5 µg/mL, respectively. The Ag/MgO nanoparticles upregulated caspase-3 and -9 activities, downregulated Bcl-2, upregulated Bax and p53 protein expressions in the cancer cells. The morphology of the Ag/MgO nanoparticle treated HT29 and A549 cells was typical of apoptosis, with cell detachment, shrinkage, and membrane blebbing. The results suggest that Ag/MgO nanoparticles induce apoptosis in cancer cells and exhibit potential as a promising anticancer agent.

3.
Saudi Pharm J ; 30(4): 347-358, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35527823

ABSTRACT

In this study, we formulated Thymoquinone-loaded nanocomposites (TQ-NCs) using high-pressure homogenizer without sodium tripolyphosphate. The TQ-NCs were characterized and their anti-inflammatory determined by the response of the LPS-stimulated macrophage RAW 264.7 cells in the production of nitric oxide, prostaglandin E2, tumor necrosis factor-α, interleukin-6, and interleukin-1ß. The physicochemical properties of TQ-NC were determined using different machines. TQ was fully incorporated in the highly thermal stable nanoparticles. The nanoparticles showed rapid release of TQ in the acidic medium of the gastric juice. In medium of pH 6.8, TQ-NC exhibited sustained release of TQ over a period of 100 h. The results suggest that TQ-NC nanoparticles have potential application as parenterally administered therapeutic compound. TQ-NC effectively reduce production of inflammatory cytokines by the LPS-stimulated RAW 264.7 cells, indicating that they have anti-inflammatory properties. In conclusion, TQ-NC nanoparticles have the characteristics of efficient carrier for TQ and an effective anti-inflammatory therapeutic compound.

4.
Saudi Pharm J ; 29(3): 223-235, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33981171

ABSTRACT

The long-term objective of the present study was to prepare, physicochemically characterize and determine the anticancer of clausenidin/hydroxypropyl-ß-cyclodextrin (Clu/HPßCD) inclusion complex. We used differential scanning calorimetry, X-ray diffractometer, fourier transform infrared spectroscopy, ultraviolet-visible spectrophotometer and 13C and 1H nuclear magnetic resonance followed by in vitro anticancer assays. The orientation and intermolecular interactions of Clausenidin within cyclodextrin cavity were also ascertained by molecular docking simulation accomplished by AutoDock Vina. The guest molecule was welcomed by the hydrophobic cavity of the host molecule and sustained by hydrogen bond between host/guest molecules. The constant drug release with time, and increased solubility were found after successful complexation with HPßCD as confirmed by physicochemical characterizations. Clausenidin had greater cytotoxic effect on colon cancer HT29 cells when incorporated into HPßCD cavity than dissolved in DMSO. Also, from a comparison of cell viability between normal and cancer cells, a reduced side effect was observed. The Clu/HPßCD inclusion complex triggered reactive oxygen species-mediated cytotoxicity in HT29 cells. The inclusion complex-treated HT29 cells showed cell cycle arrest and death by apoptosis associated with caspases activation. The presence of HPßCD seems to aid the anticancer activity of clausenidin.

5.
Biomed Pharmacother ; 138: 111483, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33744756

ABSTRACT

The aim of this study was to prepare, characterize, and determine the in vitro anticancer effects of platinum-doped magnesia (Pt/MgO) nanoparticles. The chemical compositions, functional groups, and size of nanoparticles were determined using X-ray diffraction, Fourier transform infrared spectroscopy, energy dispersive X-ray spectroscopy, transmission electron microscopy, and scanning electron microscopy. Pt/MgO nanoparticles were cuboid and in the nanosize range of 30-50 nm. The cytotoxicity of Pt/MgO nanoparticles was determined via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay on the human lung and colonic cancer cells (A549 and HT29 respectively) and normal human lung and colonic fibroblasts cells (MRC-5 and CCD-18Co repectively). The Pt/MgO nanoparticles were relatively innocuous to normal cells. Pt/MgO nanoparticles downregulated Bcl-2 and upregulated Bax and p53 tumor suppressor proteins in the cancer cells. Pt/MgO nanoparticles also induced production of reactive oxygen species, decreased cellular glutathione level, and increased lipid peroxidation. Thus, the anticancer effects of Pt/MgO nanoparticles were attributed to the induction of oxidative stress and apoptosis. The study showed the potential of Pt/MgO nanoparticles as an anti-cancer compound.


Subject(s)
Cytotoxins/toxicity , Magnesium Oxide/toxicity , Metal Nanoparticles/toxicity , Oxidative Stress/drug effects , Platinum/toxicity , A549 Cells , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , HT29 Cells , Humans , Inflammation Mediators/metabolism , Oxidative Stress/physiology
6.
Nanomedicine (Lond) ; 15(6): 547-561, 2020 03.
Article in English | MEDLINE | ID: mdl-32063101

ABSTRACT

Aim: To prepare, physicochemically characterize and determine the anticancer effects of palladium-doped magnesia (Pd/MgO) nanoparticles. Materials & methods: Pd/MgO nanoparticles were prepared by the co-precipitation method from the aqueous solution of Mg(NO3)2.6H2O using K2CO3 and the impregnation of MgO into palladium acetylacetonate. Results: Pd/MgO nanoparticles were between 47 and 70 nm in size, cuboid in shape, and tended to form aggregates. Nanoparticles were more antiproliferative toward cancer than the normal cells. In cancer cells, Pd/MgO nanoparticles induced apoptosis by increasing caspase activities and stimulating cytochrome C release. The anticancer effects of Pd/MgO nanoparticles were accentuated by the upregulation of Bax and p53 and downregulation of Bcl-2 protein expressions. Conclusion: Pd/MgO nanoparticles have potential to be developed as an anticancer compound.


Subject(s)
Antineoplastic Agents/pharmacology , Magnesium Oxide/pharmacology , Nanoparticles , Palladium , Apoptosis , Cell Line, Tumor , Cytochromes c , Humans
7.
Eur J Pharm Sci ; 133: 167-182, 2019 May 15.
Article in English | MEDLINE | ID: mdl-30902654

ABSTRACT

Thymoquinone is an effective phytochemical compound in the treatment of various diseases. However, its practical administration has been limited due to poor aqueous solubility and bioavailability. In this work, we developed a novel inclusion complex of thymoquinone and hydroxypropyl-ß-cyclodextrin that features improved solubility and bioactivity. The drug solubility was markedly accelerated in the increasing ratio of hydroxypropyl-ß-cyclodextrin to thymoquinone amount. The formation of the thymoquinone/hydroxypropyl-ß-cyclodextrin inclusion complex was evidenced using X-ray diffraction, differential scanning calorimetry, thermal gravimetric analysis, Fourier transform infrared, scanning electron microscopy and nuclear magnetic resonance. The release behavior of the complex, as well as of their mixtures, was examined in artificial gastric (pH 1.2) and intestinal (pH 6.8) dissolution media. The formulated complex released the drug rapidly at the initial stage, followed by a slow release. Thermodynamic parameters ΔH, ΔS and ΔG were calculated with temperatures ranging from 20 to 45 °C to evaluate the complexation process. The activity of the inclusion complex was evaluated on IgE-mediated allergic response in rat basophilic leukemia (RBL-2H3) cells by monitoring key allergic mediators. The results revealed that compared with free thymoquinone, the inclusion complex more strongly inhibited the release of histamine, tumor necrosis factor-α, and interleukin-4, and was not cytotoxic at the tested thymoquinone concentrations (0.125-4 µg/mL) indicating the inclusion complex possibly had better antiallergic effects. Our finding suggested that the inclusion complex achieved prolonged action and reduced side-effect of thymoquinone.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Anti-Allergic Agents/administration & dosage , Benzoquinones/administration & dosage , Drug Delivery Systems , Animals , Anti-Allergic Agents/chemistry , Benzoquinones/chemistry , Cell Line, Tumor , Drug Liberation , Gastric Juice/chemistry , Histamine/metabolism , Interleukin-4/metabolism , Intestinal Secretions/chemistry , Rats , Tumor Necrosis Factor-alpha/metabolism
8.
Drug Des Devel Ther ; 11: 3309-3319, 2017.
Article in English | MEDLINE | ID: mdl-29200826

ABSTRACT

INTRODUCTION: Dentatin (DEN) (5-methoxy-2, 2-dimethyl-10-(1, 1-dimethyl-2propenyl) dipyran-2-one), a natural compound present in the roots of Clausena excavata Burm f, possesses pro-apoptotic and antiproliferative effects in various cancer cells. Because of its hydrophobicity, it is believed that its complexation with hydroxy-ß-cyclodextrin (HPßCD) will make it a potent inhibitor of cancer cell growth. In the current work, the molecular mechanisms of apoptosis induced by DEN and DEN-HPßCD complex were demonstrated in human colon HT-29 cancer cells. MATERIALS AND METHODS: After the human colon HT-29 cancer cells were treated with DEN and DEN-HPßCD complex, their effects on the expression of apoptotic-regulated gene markers in mitochondria-mediated apoptotic and death receptor pathways were detected by Western blot analysis and reverse transcription polymerase chain reaction. These markers included caspases-9, 3, and 8, cytochrome c, poly (ADP-ribose) polymerase, p53, p21, cyclin A as well as the Bcl-2 family of proteins. RESULTS: At 3, 6, 12, and 24 µg/mL exposure, DEN and DEN-HPßCD complex significantly affected apoptosis in HT-29 cells through the down-regulation of Bcl-2 and cyclin A in turn, and up-regulation of Bax, p53, p21, cytochrome c at both protein and mRNA levels. DEN and DEN-HPßCD complex also decreased cleaved poly (ADP-ribose) polymerase and induced caspases-3, -8, and -9. CONCLUSION: Results of this study indicate that the apoptotic pathway caused by DEN and DEN-HPßCD complex are mediated by the regulation of caspases and Bcl-2 families in human colon HT-29 cancer cells. The results also suggest that DEN-HPßCD complex may have chemotherapeutic benefits for colon cancer patients.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HT29 Cells , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Environ Sci Pollut Res Int ; 24(24): 20104-20112, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28702910

ABSTRACT

A new commercial cationic polyelectrolyte chitosan (CM), obtained from the waste of mushroom production, was examined using models of water and wastewater namely kaolin and palm oil mill effluent (pome). As it is biocompatible, widely available, and economically feasible, chitosan mushroom has high potential to be a suitable replacement for alum. Also, it can be a promising alternative to chitosan obtained traditionally from Crustaceans due to its higher zeta potential and homogeneity based on the raw material required for its production. A wide range of coagulant dose (5-60 mg l-1) and wastewater pH (2-12) were taken into account to find the optimal conditions of coagulation. The optimal doses are 10 and 20 mg l-1 at best pH (11 and 3) when treated with kaolin and palm oil mill effluent, respectively, while 1200 mg l-1 of alum was not enough to reach the efficiency of chitosan mushroom. On the other hand, the optimum dose of chitosan mushroom (20 mg l-1) at pH 3 of pome produced (75, 73, and 98%) removal of chemical oxygen demand (COD), biological oxygen demand (BOD), and total suspended solids (TSS), respectively. The significant potential of chitosan mushroom was proved by zeta potential measurement. Indeed, it possesses the highest zeta potential (+70 mV) as compared to the traditional chitosan produced from crustaceans. In short, chitosan mushroom as a biocoagulant is eco-friendly and it enhances water quality that meets the requirements of environmental conservatives.


Subject(s)
Agaricales/chemistry , Chitosan/chemistry , Polyelectrolytes/chemistry , Wastewater/chemistry , Water Pollutants, Chemical/analysis , Water Purification/methods , Biological Oxygen Demand Analysis , Chitosan/isolation & purification , Flocculation , Polyelectrolytes/isolation & purification
10.
Int J Mol Sci ; 17(10)2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27763535

ABSTRACT

Dentatin (DEN), purified from the roots of Clausena excavata Burm f., has poor aqueous solubility that reduces its therapeutic application. The aim of this study was to assess the effects of DEN-HPßCD (hydroxypropyl-ß-cyclodextrin) complex as an anticancer agent in HT29 cancer cell line and compare with a crystal DEN in dimethyl sulfoxide (DMSO). The exposure of the cancer cells to DEN or DEN-HPßCD complex leads to cell growth inhibition as determined by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. To analyze the mechanism, in which DEN or DEN-HPßCD complex causes the death in human colon HT29 cancer cells, was evaluated by the enzyme-linked immunosorbent assay (ELIZA)-based assays for caspase-3, 8, 9, and reactive oxygen species (ROS). The findings showed that an anti-proliferative effect of DEN or DEN-HPßCD complex were via cell cycle arrest at the G2/M phase and eventually induced apoptosis through both mitochondrial and extrinsic pathways. The down-regulation of poly(ADP-ribose) polymerase (PARP) which leaded to apoptosis upon treatment, was investigated by Western-blotting. Hence, complexation between DEN and HPßCD did not diminish or eliminate the effective properties of DEN as anticancer agent. Therefore, it would be possible to resolve the conventional and current issues associated with the development and commercialization of antineoplastic agents in the future.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , M Phase Cell Cycle Checkpoints/drug effects , Reactive Oxygen Species/metabolism , 2-Hydroxypropyl-beta-cyclodextrin , Antineoplastic Agents, Phytogenic/administration & dosage , Clausena/chemistry , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Carriers/chemistry , HT29 Cells , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Signal Transduction/drug effects , beta-Cyclodextrins/chemistry
11.
Mol Med Rep ; 13(5): 3945-52, 2016 May.
Article in English | MEDLINE | ID: mdl-26987078

ABSTRACT

Patients with cancer often exhibit signs of anemia as the result of the disease. Thus, cancer chemotherapies often include erythropoietin (EPO) in the regime to improve the survival rate of these patients. The aim of the present study was to determine the effect of EPO on doxorubicin-treated breast cancer cells. The cytotoxicity of doxorubicin alone or in combination with EPO against the MCF-7 and MDA-MB­231 human breast cancer cells were determined using an MTT cell viability assay, neutral red (NR) uptake assay and lactate dehydrogenase (LDH) assay. The estimated half maximal inhibitory concentration values for doxorubicin and the combination of doxorubicin with EPO were between 0.140 and 0.260 µg/ml for all cells treated for 72 h. Treatment with doxorubicin in combination with EPO led to no notable difference in cytotoxicity, compared with treatment with doxorubicin alone. The antiproliferative effect of doxorubicin at a concentration of 1 µg/ml on the MDA­MB­231 cells was demonstrated by the decrease in viable cells from 3.6x10(5) at 24 h to 2.1x10(5) at 72 h of treatment. In order to confirm apoptosis in the doxorubicin-treated cells, the activities of caspases-3/7 and ­9 were determined using a TBE assay. The results indicated that the activities of caspases-3/7 and ­9 were significantly elevated in the doxorubicin-treated MDA-MB-231 cells by 571 and 645%, respectively, and in the MCF 7 cells by 471 and 345%, respectively, compared with the control cells. EPO did not modify the effect of doxorubicin on these cell lines. The results of the present study suggested that EPO was safe for use in combination with doxorubicin in the treatment of patients with breast cancer and concurrent anemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival/drug effects , Doxorubicin/pharmacology , Erythropoietin/pharmacology , Female , Humans , MCF-7 Cells , Recombinant Proteins/pharmacology
12.
Int J Nanomedicine ; 10: 5739-50, 2015.
Article in English | MEDLINE | ID: mdl-26425082

ABSTRACT

Iron-manganese-doped sulfated zirconia nanoparticles with both Lewis and Brønsted acidic sites were prepared by a hydrothermal impregnation method followed by calcination at 650°C for 5 hours, and their cytotoxicity properties against cancer cell lines were determined. The characterization was carried out using X-ray diffraction, thermogravimetric analysis, Fourier transform infrared spectroscopy, Brauner-Emmett-Teller (BET) surface area measurements, X-ray fluorescence, X-ray photoelectron spectroscopy, zeta size potential, and transmission electron microscopy (TEM). The cytotoxicity of iron-manganese-doped sulfated zirconia nanoparticles was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays against three human cancer cell lines (breast cancer MDA-MB231 cells, colon carcinoma HT29 cells, and hepatocellular carcinoma HepG2 cells) and two normal human cell lines (normal hepatocyte Chang cells and normal human umbilical vein endothelial cells [HUVECs]). The results suggest for the first time that iron-manganese-doped sulfated zirconia nanoparticles are cytotoxic to MDA-MB231 and HepG2 cancer cells but have less toxicity to HT29 and normal cells at concentrations from 7.8 µg/mL to 500 µg/mL. The morphology of the treated cells was also studied, and the results supported those from the cytotoxicity study in that the nanoparticle-treated HepG2 and MDA-MB231 cells had more dramatic changes in cell morphology than the HT29 cells. In this manner, this study provides the first evidence that iron-manganese-doped sulfated zirconia nanoparticles should be further studied for a wide range of cancer applications without detrimental effects on healthy cell functions.


Subject(s)
Cell Proliferation/drug effects , Iron/chemistry , Manganese/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Sulfates/chemistry , Zirconium/chemistry , Humans , Microscopy, Electron, Transmission , Neoplasms/pathology , Photoelectron Spectroscopy , Tumor Cells, Cultured , X-Ray Diffraction , Zirconium/administration & dosage
13.
BMC Complement Altern Med ; 15: 45, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25881293

ABSTRACT

BACKGROUND: In this study, the effect of mango kernel extract in the induction of apoptosis of the breast cancer (MDA-MB-231) cell line was examined. This is an attempt to discover alternatives to current therapeutic regimes in the treatment of breast cancers. METHODS: The pro-apoptotic markers, Bax, cytochrome c, caspases-. -8 and -9, and anti-apoptotic markers, Bcl-2, p53 and glutathione were determined in MDA-MB231 cells treated for 12 and 24 h with mango kernel extract. RESULTS: The results showed that the extract produced a time- and dose-dependent increases in pro-apoptotic proteins and oxidative stress markers with a corresponding decrease in anti-apoptotic markers. CONCLUSIONS: Based on the findings, mango kernel extract modulates redox balance in MDA-MB-231 breast cancer cells with a tendency for apoptotic cell death. The changes observed in this study may collectively underlie the basis for the cell death induced in MDA-MB-231 breast cancer cells by mango kernel extract. Thus, mango kernel extract has potential to be developed into an antibreast cancer mixture, and hence these results are worth studying further.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Mangifera , Oxidative Stress/drug effects , Phytotherapy , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Female , Humans , Oxidation-Reduction , Plant Extracts/therapeutic use , Receptors, Estrogen/metabolism , Seeds
14.
Int J Nanomedicine ; 10: 765-74, 2015.
Article in English | MEDLINE | ID: mdl-25632233

ABSTRACT

Nanoparticle sulphated zirconia with Brønsted acidic sites were prepared here by an impregnation reaction followed by calcination at 600°C for 3 hours. The characterization was completed using X-ray diffraction, thermal gravimetric analysis, Fourier transform infrared spectroscopy, Brunner-Emmett-Teller surface area measurements, scanning electron microscopy with energy dispersive X-ray spectroscopy, and transmission electron microscopy. Moreover, the anticancer and antimicrobial effects were investigated for the first time. This study showed for the first time that the exposure of cancer cells to sulphated zirconia nanoparticles (3.9-1,000 µg/mL for 24 hours) resulted in a dose-dependent inhibition of cell growth, as determined by (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Similar promising results were observed for reducing bacteria functions. In this manner, this study demonstrated that sulphated zirconia nanoparticles with Brønsted acidic sites should be further studied for a wide range of anticancer and antibacterial applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Proliferation/drug effects , Metal Nanoparticles/chemistry , Zirconium/chemistry , Humans , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Spectrometry, X-Ray Emission , Spectroscopy, Fourier Transform Infrared , Tumor Cells, Cultured , X-Ray Diffraction
15.
Int J Nanomedicine ; 8: 4115-29, 2013.
Article in English | MEDLINE | ID: mdl-24204141

ABSTRACT

The long-term objective of the present study was to determine the ability of NiZn ferrite nanoparticles to kill cancer cells. NiZn ferrite nanoparticle suspensions were found to have an average hydrodynamic diameter, polydispersity index, and zeta potential of 254.2 ± 29.8 nm, 0.524 ± 0.013, and -60 ± 14 mV, respectively. We showed that NiZn ferrite nanoparticles had selective toxicity towards MCF-7, HepG2, and HT29 cells, with a lesser effect on normal MCF 10A cells. The quantity of Bcl-2, Bax, p53, and cytochrome C in the cell lines mentioned above was determined by colorimetric methods in order to clarify the mechanism of action of NiZn ferrite nanoparticles in the killing of cancer cells. Our results indicate that NiZn ferrite nanoparticles promote apoptosis in cancer cells via caspase-3 and caspase-9, downregulation of Bcl-2, and upregulation of Bax and p53, with cytochrome C translocation. There was a concomitant collapse of the mitochondrial membrane potential in these cancer cells when treated with NiZn ferrite nanoparticles. This study shows that NiZn ferrite nanoparticles induce glutathione depletion in cancer cells, which results in increased production of reactive oxygen species and eventually, death of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ferric Compounds/pharmacology , Metal Nanoparticles/chemistry , Nickel/pharmacology , Zinc Compounds/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation , Cytochromes c/analysis , Cytochromes c/metabolism , Ferric Compounds/chemistry , Glutathione/analysis , Glutathione/metabolism , Humans , Malondialdehyde/analysis , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Nickel/chemistry , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , Zinc Compounds/chemistry , bcl-2-Associated X Protein/analysis , bcl-2-Associated X Protein/metabolism
16.
Int J Nanomedicine ; 8: 2497-508, 2013.
Article in English | MEDLINE | ID: mdl-23885175

ABSTRACT

In this study, in vitro cytotoxicity of nickel zinc (NiZn) ferrite nanoparticles against human colon cancer HT29, breast cancer MCF7, and liver cancer HepG2 cells was examined. The morphology, homogeneity, and elemental composition of NiZn ferrite nanoparticles were investigated by scanning electron microscopy, transmission electron microscopy, and energy dispersive X-ray spectroscopy, respectively. The exposure of cancer cells to NiZn ferrite nanoparticles (15.6-1,000 µg/mL; 72 hours) has resulted in a dose-dependent inhibition of cell growth determined by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. The quantification of caspase-3 and -9 activities and DNA fragmentation to assess the cell death pathway of the treated cells showed that both were stimulated when exposed to NiZn ferrite nanoparticles. Light microscopy examination of the cells exposed to NiZn ferrite nanoparticles demonstrated significant changes in cellular morphology. The HepG2 cells were most prone to apoptosis among the three cells lines examined, as the result of treatment with NiZn nanoparticles. In conclusion, NiZn ferrite nanoparticles are suggested to have potential cytotoxicity against cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Drug Carriers/chemistry , Ferric Compounds/pharmacology , Metal Nanoparticles/chemistry , Nickel/pharmacology , Zinc Compounds/pharmacology , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , DNA Fragmentation/drug effects , Ferric Compounds/chemistry , Humans , Nickel/chemistry , Particle Size , Spectrophotometry, Infrared , Statistics, Nonparametric , X-Ray Diffraction , Zinc Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...