Next Article in Journal
The Design and Application of a New Wireline Pressure Coring System for the Guangzhou Marine Geological Survey Methane Hydrate Expedition in the South China Sea
Previous Article in Journal
Technique Variables Associated with Fast Bowling Performance: A Systematic-Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Evaluation of Superparamagnetic Fe3O4-Ag Decorated Nanoparticles: Cytotoxicity Studies in Human Fibroblasts (HFF-1) and Breast Cancer Cells (MCF-7)

by
Álvaro de Jesús Ruíz-Baltazar
1,2,*,
Simón Yobanny Reyes-López
2,
Néstor Méndez-Lozano
3 and
Karla Juárez-Moreno
4,*
1
CONAHCYT-Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Santiago de Querétaro 76230, Querétaro, Mexico
2
Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del Pronaf y Estocolmo s/n, Zona Pronaf, Ciudad Juárez 32310, Chihuahua, Mexico
3
Campus Querétaro, Universidad del Valle de México, Blvd. Juriquilla No. 1000 A Del., Santa Rosa Jáuregui, Santiago de Querétaro C.P. 76230, Querétaro, Mexico
4
Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, Santiago de Querétaro 76230, Querétaro, Mexico
*
Authors to whom correspondence should be addressed.
Appl. Sci. 2024, 14(15), 6750; https://doi.org/10.3390/app14156750
Submission received: 20 June 2024 / Revised: 15 July 2024 / Accepted: 17 July 2024 / Published: 2 August 2024

Abstract

:
This study investigates the cytotoxicity profile of superparamagnetic Fe3O4-Ag decorated nanoparticles against human fibroblasts (HFF-1) and breast cancer cells (MCF-7). The nanoparticles underwent comprehensive characterization employing scanning electron microscopy (SEM), X-ray diffraction (XRD) analysis, X-ray photoelectron spectroscopy (XPS), and magnetic assays including hysteresis curves and zero-field-cooled (ZFC) plots. The nanoparticles exhibited superparamagnetic behavior as evidenced by magnetic studies. Cytotoxicity assays demonstrated that both HFF-1 and MCF-7 cells maintained nearly 100% viability upon nanoparticle exposure, underscoring the outstanding biocompatibility of Fe3O4/Ag decorated nanoparticles and suggesting their potential utility in biomedical applications such as drug delivery and magnetic targeting. Furthermore, the study analyzed the cytotoxic effects of Fe3O4 and Fe3O4-Ag decorated nanoparticles to evaluate their biocompatibility for further therapeutic efficacy. Results showed that neither type of nanoparticle significantly reduced cell viability in HFF-1 fibroblasts, indicating non-cytotoxicity at the tested concentrations. Similarly, MCF-7 breast cancer cells did not exhibit a significant change in viability when exposed to different nanoparticle concentrations, highlighting the compatibility of these nanoparticles with both healthy and cancerous cells. Additionally, the production of reactive oxygen species (ROS) by cells exposed to the nanoparticles was examined to guarantee their biosafety for further therapeutic potential. Higher concentrations (50–100 μg/mL) of Fe3O4-Ag nanoparticles decreased ROS production in both HFF-1 and MCF-7 cells, while Fe3O4 nanoparticles were more effective in generating ROS. This differential response suggests that Fe3O4-Ag nanoparticles might modulate oxidative stress more effectively, thus beneficial for future anticancer strategies due to cancer cells’ susceptibility to ROS-induced damage. These findings contribute to understanding nanoparticle interactions with cellular oxidative mechanisms, which are crucial for developing safe and effective nanoparticle-based therapies. This investigation advances our understanding of nanostructured materials in biological settings and highlights their promising prospects in biomedicine.

1. Introduction

Nanotechnology has emerged as a promising field in the realm of biomedical applications, offering innovative solutions for targeted drug delivery, cancer diagnostics, and therapeutic interventions [1,2,3]. Among the diverse nanomaterials available, superparamagnetic iron oxide nanoparticles (SPIONs) have garnered significant attention due to their exceptional physicochemical properties, including their high surface area, biocompatibility, and magnetic responsiveness [4]. These unique characteristics have led to the exploration of iron oxide nanoparticles, particularly Fe3O4, in various biomedical applications, such as magnetic resonance imaging (MRI), hyperthermia-based cancer treatment, and drug delivery [5,6].
The synthesis and functionalization of Fe3O4 nanoparticles have been extensively investigated, with researchers exploring different strategies to enhance their stability, biocompatibility, and targeting capabilities [7,8]. One approach that has shown promise is the decoration of Fe3O4E nanoparticles with silver (Ag) to create Fe3O4-Ag structures [9]. The addition of silver can potentially improve the antibacterial properties and electrical conductivity of the nanoparticles, making them attractive for a wide range of biomedical applications [10,11]. Another application of the Fe3O4-Ag nanoparticles is in the field of cancer therapy [12,13]. The enhanced magnetic and plasmonic properties of the decorated nanoparticles can be leveraged for targeted drug delivery, photothermal therapy, and magnetic hyperthermia. On the one hand, the magnetic properties allow for the nanoparticles to be guided to the tumor site using an external magnetic field [14,15,16]. On the other hand, the silver coating can potentially enhance the cytotoxic effects on cancer cells through generation of reactive oxygen species and disruption of cellular processes [17].
To fully harness the potential of Fe3O4-Ag decorated nanoparticles in biomedical applications, it is crucial to understand their cytotoxicity and biocompatibility in relevant cell lines. Human fibroblasts (HFF-1) and breast cancer cells (MCF-7) serve as important model systems for evaluating the biological impact of these nanoparticles, as they represent crucial components of the human body -healthy cells and cancer cells, respectively [18,19,20].
Regarding to the obtention of these magnetoplasmonic structures, the synthesis of Fe3O4-Ag decorated nanoparticles can be achieved through various chemical and biological methods [9]. Typically, the core Fe3O4 nanoparticles are first prepared, followed by the deposition of silver onto the surface. several synthesis methods of Fe3O4-Ag nanoparticles have been reported, such as co-precipitation, thermal decomposition, microemulsion, and green synthesis approaches using plant extracts or microbial agents [16,21,22]. Recently, green synthesis methods have gained attention due to their eco-friendly nature and the potential to produce biocompatible nanoparticles [23,24]. For example, the use of deep eutectic solvents as reducing and stabilizing agents in the synthesis of Fe3O4-Ag nanoparticles has been reported [25,26]. However, the Chemical reduction is one of the most widely used methods for the synthesis of these nanostructures, as it allows for precise control over the size, morphology, and composition of the nanoparticles [16,27,28]. The formation of the silver shell or decorated on the Fe3O4 core can be achieved through the reduction of silver nitrate (AgNO3) in the presence of a reducing agent, such as sodium borohydride or ascorbic acid [29]. The resulting Fe3O4-Ag nanoparticles can be further functionalized with various biomolecules, polymers, or drug compounds to enhance their biocompatibility, targeting, and therapeutic capabilities [30,31].
In this sense, evaluating the cytotoxic effects of Fe3O4-Ag decorated nanoparticles is important to deepen knowledge of their biosecurity for their future biomedical applications. This research used the human HFF-1 fibroblast cell line as a model for non-cancerous epithelial cells, often employed to determine nanoparticles’ overall biocompatibility and safety [32]. Meanwhile, MCF-7 cells were employed as a well-established model for breast cancer, specifically representing the luminal subtype. Thus, ref. [32] evaluating the cytotoxicity of Fe3O4 and Fe3O4-Ag nanoparticles in both types of cells can provide insights into their potential toxic effects, which is crucial for their development as safe and effective future biomedical agents.
On the other hand, breast cancer cell lines, such as MCF-7, are used to assess the anticancer potential of Fe3O4-Ag nanoparticles. The ability of these nanoparticles to selectively induce cytotoxicity in cancer cells while sparing healthy cells is a desirable characteristic for their future application in cancer therapy [25,29].
The cytotoxicity studies typically involve exposing the cell lines to varying concentrations of the Fe3O4-Ag nanoparticles and evaluating their effects on cell viability, proliferation, and morphology using various assays, such as MTT, LDH, or live/dead staining. Through these methods, it is possible to obtain information about metabolism and cell viability, allowing the elucidation of the concentration-dependent cytotoxic responses of the nanoparticles and providing valuable insights into their biocompatibility for further potential therapeutic efficacy [13,33].
Therefore, a comprehensive evaluation of the cytotoxicity of these novel Fe3O4-Ag-decorated nanoparticles on relevant cell lines is vital to guarantee their biocompatibility for being considered suitable nanosystems for biomedical applications. This study is designed to investigate the cytotoxic effects of Fe3O4-Ag nanoparticles on human fibroblasts (HFF-1) and breast cancer cells (MCF-7), providing crucial information on their biocompatibility. For the toxicological study of nanomaterials, it is essential to investigate not only the cytotoxic effects but also the generation of reactive oxygen species (ROS), in vitro inflammation processes, and hemocompatibility. This approach provides broader knowledge of nanoparticles’ impact on cellular processes. This study presents the synthesis and physicochemical characterization of Fe3O4-decorated Ag nanoparticles and a comprehensive nanotoxicology investigation. The study demonstrates the in vitro biocompatibility of these nanoparticles, showcasing their potential for future biomedical applications as antiproliferative agents. This potential stems from their magnetic properties and the ability to modify their surface with biological ligands and anticancer drugs.

2. Materials and Methods

2.1. Synthesis of Fe3O4-Ag Decorated Nanoparticles

The synthesis of Fe3O4 nanostructures commenced by utilizing FeCl3·6H2O as a precursor at a concentration of 0.01 M. Reduction of Fe3+ ions was achieved using a solution of NaBH4 (0.02 M), with polyvinylpyrrolidone (PVP) serving as a stabilizing surfactant at a concentration of 0.1 M. Deionized water was employed as the colloidal medium. The PVP/FeCl3·6H2O ratio was maintained at 50, a parameter known to promote the formation of magnetite nanostructures [20]. Following the reduction of Fe3+ ions, an oxidative etching process was employed to facilitate the formation of magnetite, a mechanism established in prior studies for Fe3O4 nanostructure synthesis [20]. Importantly, all experiments were conducted at room temperature under a constant hydrogen potential (pH = 11). The proposed reduction process involves the initial decomposition of NaBH4, generating hydrogen, which in turn facilitates the reduction of Fe3+ ions, leading to the formation of iron nanoparticles.
Subsequently, to the preformed Fe3O4 nanoparticles, a solution of AgNO3 (50 mM, 20 mL) was added, and the mixture was agitated at room temperature. Following this, a solution of NaBH4 (2 mM, 5 mL) was promptly added, resulting in the formation of silver nuclei on the Fe3O4 particles. This process was visually discernible by a change in coloration from black to dark gray, culminating in the synthesis of Fe3O4-Ag decorated nanoparticles.

2.2. Chemical and Structural Characterization of Fe3O4-Ag Decorated

The Fe3O4-Ag decorated nanoparticles underwent comprehensive characterization using scanning electron microscopy (SEM) with a Hitachi model 8230 microscope. Various electron detection techniques were employed to obtain a thorough understanding of the morphology and chemical composition. High-resolution images acquired through High-Angle Secondary Electron (SE-HA) microscopy highlighted the presence of Fe3O4 and Ag phases, while Secondary Electron (SE(U)) imaging provided detailed surface topography. High-Angle Backscattered Electron (HA(T)) analyses discerned chemical element distribution, exploiting the technique’s sensitivity to atomic number. Composite images revealing both morphology and elemental composition were generated using SE + HA + OFF by combining secondary and backscattered electron detectors. Additionally, chemical analysis via Energy-Dispersive X-ray Spectroscopy (EDS) mapped the distribution of iron (Fe), oxygen (O), and silver (Ag) in the nanoparticles, confirming the presence and uniformity of Ag decoration on the Fe3O4 surface. SEM operational parameters included a magnification of ×500k, a working distance of 3.7 mm, and an acceleration voltage of 3.0 kV, ensuring precise and accurate results from EDS mappings conducted in representative areas.
Furthermore, the magnetic nanoparticle characterization involved utilizing an X-ray diffractometer (Rigaku Ultima IV, Tokyo, Japan) equipped with Cu-Kα radiation, employing parallel-beam geometry and 2θ scans spanning from 15 to 80°. Magnetic analyses were conducted using a vibrating sample magnetometer (VSM) at 300 K.

2.3. Nanotoxicological Bioassays

2.3.1. Cell Culture

The human breast adenocarcinoma MCF-7 cells (ATCC-HTB-22) and Human Fibroblasts HFF-1 (SRC-1041) were purchased from the American Type Culture Collection (ATCC, Manasas, VA, USA). The cell lines were thawed, cultivated, and maintained following the provider’s instructions. Cell lines were grown in Petri dishes for cell culture at a temperature of 37 °C in 5% CO2 atmosphere in Dulbecco’s Modified Eagle’s Medium (DMEM), supplemented with 10% Fetal Bovine Serum (FBS, BenchMark, Geminis Bio Products, West Sacrament, CA, 95605, USA), 1% Penicillin-Streptomycin (Sigma-Aldrich, St. Louis, MO, USA), 1% L-glutamine, and 2 g/L of sodium bicarbonate as previously described [34,35,36,37]. The concentrations of nanoparticles used in this work for all the bioassays were selected according to the suggestions issued by the OECD through the Nanomaterials Series on the Safety of Manufactured Nanomaterials No. 85 described in the technical brochure for the Evaluation of in vitro methods for human hazard assessment applied in the OECD Testing Program for the Safety of Manufactured.

2.3.2. Cytotoxicity Assay by MTT Reduction

The effect of Fe3O4 and Fe3O4-Ag decorated nanoparticles on cell viability was evaluated using the MTT reduction, following the recommendations provided by the OECD testing programme for the safety of manufactured nanomaterials. The cytotoxicity effect of Fe3O4 and Fe3O4-Ag decorated nanoparticles was determined using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, a widely used method to assess cell viability [38,39]. HFF-1 and MCF-7 cells were seeded at a density of 10,000 cells per well in a 96-well plate and incubated in 100 μL of DMEM-supplemented media at 37 °C in a 5% CO2 atmosphere for 24 h. After this, Fe3O4 and Fe3O4-Ag decorated NPs were suspended in MiliQ-water at 1 mg/mL concentrations. Before each assay, each nanoparticle suspension was prepared freshly and underwent 10 min of sonication. Then, solutions of nanoparticles at different concentrations were made in DMEM media and promptly added to the cell cultures as described below. HFF-1 and MCF-7 cells were exposed to Fe3O4 and Fe3O4-Ag decorated NPs at 0.1; 1, 5, 10, 25, 50, and 100 μg/mL in a final volume of 100 μL of DMEM-supplemented media in a 96-well plate. Cells were incubated for 24 h at 37 °C in a 5% CO2 atmosphere using DMEM media. Afterward, the media was discarded, and cells were rinsed thrice with 200 μL of PBS 1x. Then, 10 μL of MTT and 90 μL of DMEM media were added to each well and incubated for 4 h at 37 °C in a 5% CO2 atmosphere for 24 h. After this, 100 μL of isopropanol was added to each well to dissolve formazan crystals, and the 96-well plate was incubated for 30 min at 25 °C in darkness. Absorbance readings were taken at 570 and 690 nm wavelengths using a microplate reader (ThermoFisher Scientific, Waltham, MA, USA). Each cytotoxicity experiment included a control group with a culture medium lacking NPs to establish a baseline. The positive control for inducing cell death consisted of cells treated with 1% (v/v) Triton X-100, while the negative control contained only cell culture medium without nanoparticles. These controls facilitated the comparison of cell viability between the negative and positive controls and the damage caused by various concentrations of NPs. Experiments were conducted in triplicate. Cell viability was calculated relative to the absorbance values of the positive control.

2.3.3. Measurement of Reactive Oxygen Species Production

To measure the production of reactive oxygen species (ROS), cells were seeded in a 48-well plate and exposed to several concentrations from 0.1; 1, 5, 10, 25, 50, to 100 μg/mL of Fe3O4 or Fe3O4-Ag nanoparticles for 24 h. After exposure, cells were treated with 25 µM of 2′,7′-dichlorofluorescein diacetate and incubated for 1 h at 37 °C with 5% CO2. Then, cells were washed and harvested before being analyzed using flow cytometry. ROS levels were assessed using the Bl-1 channel with specific excitation and emission wavelengths at 488 nm and 525, respectively. The level of endogenous ROS was determined for each cell line without any treatment. Experiments were conducted in triplicate across three independent trials, and flow cytometry data were acquired using an Attune NxT flow cytometry (Life Technologies, Carlsbad, CA, USA) with 10,000 events (cells) recorded for each sample. Further data analysis was achieved using the Attune NxT acquisition software version 3.2.1. (Thermofisher Scientific, Waltham, MA, USA).

2.3.4. Measurement of Nitrite Production by Macrophages

The Griess assay was employed to quantify macrophage nitrite production following exposure to different concentrations of Fe3O4 or Fe3O4-Ag nanoparticles. In macrophages, nitrites result from the oxidation of nitric oxide in the modulation of inflammation and other physiological processes. RAW 264.7 (TIB-71, ATCC) macrophages were seeded in a 96-well plate at a density of 10,000 cells per well and cultured for 24 h at 37 °C and 5% CO2. Different concentrations of Fe3O4 or Fe3O4-Ag nanoparticles 0.1, 1, 5, 10, 25, 50, and 100 μg/mL were added to the wells and further incubated for 24 h under the same conditions. Subsequently, 20 µL of cell culture supernatant from each well was combined with 80 µL of 5 mM sodium nitroprusside in a new well, followed by incubation for 1 h in darkness at 37 °C and 5% CO2. Next, the reaction mixture was treated with 100 µL of Griess reagent solution (0.1% sulfanilamide and 0.1% N-(1-naphthylethylenediamine)) and incubated at 25 °C for 15 min in darkness. Absorbance was measured at 540 nm, and the values were compared against a standard curve generated using sodium nitrite concentrations ranging from 1.67 to 100 µM as reference reagents for nitrite production. Experiments were conducted independently in a triplicate manner.

2.3.5. Hemocompatibility Assay

Human red blood cells were extracted from the peripheral blood of a healthy donor with normal cell morphology. The hemocompatibility assessment followed ISO 10993-4 standards. Blood was collected in sodium heparin-containing tubes (BD USA), red blood cells (RBCs) were isolated by centrifugation, then cells were rinsed with 10 mL of 150 mM NaCl, followed by washes with 1x PBS. The RBCs were then diluted 1:50 in PBS, and 100 μL of the solution was placed into each well of a 96-well plate. Fe3O4 or Fe3O4-Ag nanoparticles were added at 0.1, 1, 5, 10, 25, 50, and 100 μg/mL concentration. After incubation for 1 h at 37 °C, samples were centrifuged at 10,000 rpm for 5 min. The absorbance of the supernatant was measured at 550 nm. The hemolysis percentage was calculated using the formula: Hemolysis (%) = (OD 550 nm sample − OD 550 nm negative control)/(OD 550 nm positive control − OD 550 nm negative control) × 100. PBS-treated RBCs served as the negative control, showing no hemolysis. RBCs treated with 0.1% Triton X-100 in PBS were the positive control, inducing 100% hemolysis. Background signal at 550 nm was subtracted for each experiment. According to ISO standards, acceptable hemolysis of RBCs is ≥5%.

2.3.6. Statistical Analysis

Statistical analysis was conducted through three independent experiments, each carried out in triplicates. The results were presented as mean ± standard deviation from three independent experiments. The data underwent analysis of variance (ANOVA) and subsequent a posteriori analysis by Tukey’s Multiple Comparison Test, facilitated by GraphPad Prism version 10 software. Statistical significance was determined at a threshold of p < 0.05.

3. Results and Discussion

3.1. Scanning Electron Microscopy Analysis

Figure 1 presents scanning electron microscopy (SEM) images of the Fe3O4-Ag sample. These images provide insights into the morphology and elemental composition of the nanoparticles. Figure 1a displays an SE-HA (High-Angle Secondary Electron) image captured using high-angle secondary electron detectors. This image reveals the nanoparticle topology, clearly indicating the presence of Fe3O4 and Ag phases. Figure 1b depicts an SE(U) image, corroborating the presence of Fe3O4 and Ag. These images utilize secondary electron emission from the sample surface upon primary electron beam interaction, providing high-resolution surface topography details. Figure 1c showcases an HA(T) (High-Angle Backscattered Electron) image, obtained using high-angle backscattered electron detectors, highlighting the formation of Fe3O4-Ag. Lastly, Figure 1d illustrates an SE + HA + OFF image, indicating the simultaneous use of both secondary and backscattered electron detectors, with the secondary electron detector turned off. This combined imaging approach offers a comprehensive view of both surface morphology and elemental composition. All images were captured at ×500k magnification, with a working distance of 3.7 mm and an acceleration voltage of 3.0 kV. The SEM imaging results suggest the formation of a decorated Fe3O4-Ag structure. Figure 1e schematically describes the distribution of the Fe3O4 and Ag nanostructures, whose distribution can be attributed to variations in the nucleation and growth rates of magnetite and silver structures. The successful synthesis of these decorated structures is evident from the captured images. This underscores the efficacy of the proposed methodology in fabricating such decorated structures.
The chemical analysis depicted in Figure 2 was conducted using Energy Dispersive X-ray Spectroscopy (EDS). In Figure 2a, the secondary electron (SE) image of the Fe3O4 configuration is presented. In Figure 2b, a mapping of the sample illustrates the distribution of silver (Ag) on the iron oxide structure, confirming the formation of the decorated Fe3O4-Ag structure. Subsequently, Figure 2c–e display elemental mappings of iron (Fe), oxygen (O), and silver (Ag) from the SE image of Fe3O4. Lastly, Figure 2f presents a chemical analysis of the Fe3O4 sample, confirming the predominant presence of elements such as Fe, O, and Ag. These results elucidate the chemical composition of the decorated nanoparticles, whose cytotoxic performance will be further investigated in this study.

3.2. X-ray Diffraction Analysis

Additionally, structural characterization of the sample is essential to support the configuration of the obtained nanoparticles. Figure 3 displays the X-ray diffraction (XRD) analysis of the Fe3O4-Ag sample, which elucidates the structure and composition of the synthesized nanoparticles.
The XRD pattern reveals the cubic FCC structure of silver (Ag), as evidenced by the characteristic peaks corresponding to the JCPDF # 87-0598, with a theoretical lattice parameter of 2.31 nm. Based on the observed intensities in the experimental X-ray pattern, the symmetry associated with the Fm3m space group and the spatial group of 325 were determined. This result confirms the presence of the Fe3O4 and Ag alloy, forming the synthesized nanoparticles. The subsequent evaluation of sample properties will be based on this confirmation.
Furthermore, the XRD pattern displays prominent peaks corresponding to the cubic structure of Fe3O4. Specifically, peaks corresponding to the (220), (311), (400), and (440) planes are observed at 2θ angles of approximately 30°, 35.4°, 43.4°, and 62.7°, respectively (JCPDS card No. 19-0629). These findings further support the presence of Fe3O4 within the synthesized nanoparticles, contributing to the comprehensive understanding of their structural characteristics.

3.3. Magnetic Behavior Analysis of Fe3O4/Ag Decorated Nanoparticles

The magnetic behavior of Fe3O4/Ag nanoalloys was comprehensively investigated through hysteresis curve analysis conducted at 300 K (Figure 4a). The obtained hysteresis loops exhibit distinctive characteristics typical of superparamagnetic materials, demonstrating nearly zero magnetic coercivity. Notably, the magnetization value recorded in this plot was determined to be 52 emu/g. This saturation magnetization value can be attributed to several factors, including the presence of non-magnetic surface layers, spin canting phenomena at the surface, or even a surface spin-glass transition [40,41]. In our specific case, the observed phenomenon is plausibly explicable in terms of the chemical bonding of organic molecules, such as polyvinylpyrrolidone (PVP), incorporated into the Fe3O4 nanostructures during the synthesis process, leading to a reduction in the magnetization of the surface layers [42,43,44].
Furthermore, the magnetization curves exhibit the characteristic blocking process inherent in assemblies of superparamagnetic nanostructures, showcasing a distribution of blocking temperatures. This behavior is influenced by several factors, including inter-particle distance, size, and shape distribution of particles [45,46]. Such observations are consistent with the behavior typically observed in hybrid materials based on magnetic constituents [47].
The zero-field-cooled (ZFC) curve depicted in Figure 4b displays a broad peak, the maximum of which (TP) is influenced by various factors such as inter-particle distance, size, and shape distribution of particles [42]. Despite the multitude of factors affecting TP, this temperature serves as a rough approximation of the average blocking temperature of the material. This ZFC and field-cooled (FC) behavior aligns with observations seen in other magnetite nanoparticle-based hybrid materials. The broad and flat plateau observed in the ZFC curve can be interpreted within the context of particles exhibiting variations in size and inter-particle distance, further elucidating the complex magnetic behavior of the nanoalloy system [41,48].

3.4. X-ray Photoelectron Spectroscopy (XPS) Analysis

X-ray photoelectron spectroscopy (XPS) analysis was conducted to ascertain the electronic structures or valence states of Fe3O4-Ag nanoalloys. Figure 5a depicts the analysis spectra of the Fe3O4-Ag nanostructures. Within these spectra, the signals of Fe 2p, O 1s, and Ag 3d are discernible as principal components of the acquired nanoparticles [49,50]. Figure 5b illustrates the Ag 3d region, wherein the Ag d5/2 and Ag d2/3 orbitals are fully identifiable at 368.3 and 374.3 eV, respectively [50], serving as compelling evidence corroborating the presence of Ag as a constituent element of the Fe3O4-Ag nanostructures, as indicated in the preceding characterization results from scanning electron microscopy and energy-dispersive X-ray spectroscopy (EDS).
Additionally, the binding energies of Fe 2p1/2 and Fe 2p3/2 were identified at 724.9 and 711.6 eV, respectively, values associated with the spin–orbit peaks of Fe3O4 (Figure 5c). An energy separation of 13.9 eV between Fe 2p (1/2) and Fe 2p3/2 was observed, a value specifically reported for the core level signal of Fe3O4. Related satellite peaks at 719.4 eV and 733.5 eV were also discerned [51]. Finally, Figure 5d reveals a high-resolution X-ray photoelectron spectrum (HRXPS) of the O1(s) orbital, with a binding energy identified at 530.74 eV, associated with the oxygen of Fe3O4. These results collectively serve as evidence of the formation of Fe3O4-Ag nanostructures [52].

3.5. Cell Viability of Fe3O4 and Fe3O4-Ag Decorated Nanoparticles on Human Fibroblast (HFF-1) and Breast Cancer Cells (MCF-7)

To analyze whether the Fe3O4 and Fe3O4-Ag decorated nanoparticles induce a cytotoxic effect, cell viability test was performed on HFF-1 human fibroblasts and MCF-7 human breast cancer cells. As observed in Figure 6a, the cell viability on HFF-1 fibroblasts changed across the different concentrations of Fe3O4 and Fe3O4-Ag decorated nanoparticles. However, none of them significantly reduced cell viability; thus, all the concentrations tested were not cytotoxic. On the other hand, the cell viability of MCF-7 breast cancer cells did not change upon the exposure of cells to different concentrations of nanoparticles, indicating that all of them are compatible and allowed the cells to grow as in the control (Figure 6b). These results indicate that Fe3O4 and Fe3O4-Ag nanoparticles are not toxic. However, it is essential to ensure that these nanoparticles do not exhibit undesirable cytotoxicity towards normal non-cancerous cells, as this could limit their viability and safety for clinical use [51,53]. Unlike our study, which focused on comparing the cytotoxic effects of Fe3O4 and Fe3O4@Ag NPs, other studies have evaluated similar nanosystems conjugated with thiolated chitosan in different cell lines such as HFF-1 fibroblasts, MC3T3-E1 osteoblasts, and the osteosarcoma cell line MG63. These studies used higher concentrations ranging from 5 to 200 ug/mL. Specifically, in HFF-1 cells, a 50% decrease in cell viability was observed starting from 100 µg/mL [54]. Careful evaluation of the effects of nanoparticles on cancerous and non-cancerous cell lines at various concentrations is essential to achieve the appropriate balance between therapeutic efficacy and biocompatibility [55].

3.6. Reactive Oxygen Species (ROS) Induced by Fe3O4 and Fe3O4-Ag Decorated Nanoparticles on Human Fibroblast (HFF-1) and Breast Cancer Cells (MCF-7)

The production of ROS was measured to corroborate the biocompatibility of Fe3O4 and Fe3O4-Ag decorated nanoparticles in human cells. Figure 7a,b shows the ROS measurement production induced by Fe3O4 and Fe3O4-Ag decorated nanoparticles on human fibroblast (HFF-1) and breast cancer cells (MCF-7), respectivetly. Different concentrations of nanoparticles were tested in human fibroblast cells HFF-1 (Figure 7a). Interestingly, only higher concentrations (100 µg/mL) of Fe3O4-Ag decorated nanoparticles reduced ROS production. This finding contrasts with established knowledge, which typically associates magnetite nanoparticles with inducing oxidative stress in mammalian cells [55,56]. The same effect was observed in human breast cancer MCF-7 cells, as illustrated in Figure 7b, Fe3O4-Ag decorated nanoparticles induced a decrease in the level of ROS production only after 50 µg/mL, and at higher concentrations (100 µg/mL) Fe3O4 also reduced the level of ROS production. To the best of our knowledge, Fe3O4 nanoparticles are widely recognized for inducing oxidative stress effects in cells. However, a report indicates that Fe3O4 magnetite nanoparticles demonstrate the opposite effect: they reduce metal-induced oxidative stress by decreasing malondialdehyde concentration and increasing superoxide dismutase levels [56].
These findings suggest that Fe3O4-Ag decorated nanoparticles may have a more pronounced effect on modulating oxidative stress in both normal and cancer cells. While Fe3O4 nanoparticles show a greater capacity to produce ROS. Previous studies have shown that the ability of nanoparticles to generate reactive oxygen species is a critical factor in determining their cytotoxicity and potential therapeutic applications; enhancing ROS production may be an effective anticancer strategy [13,57,58]. On the other hand, excessive production of ROS can also have detrimental effects on normal cells due to oxidative stress and the associated genotoxic effect [58]. The differential response observed between the two cell lines in this study highlights the importance of evaluating the biocompatibility of nanoparticles in cancerous and non-cancerous cells. The findings of this study contribute to the understanding of the interactions between nanoparticles and cellular redox homeostasis, which is crucial for developing safe and effective nanoparticle-based therapies.

3.7. Hemocompatibility and Nitric Oxide Production Induced by Fe3O4 and Fe3O4-Ag Decorated Nanoparticles

A hemolysis assay was conducted on human red blood cells to ensure the biocompatibility of both Fe3O4 and Fe3O4-Ag decorated nanoparticles. The threshold for hemocompatibility is indicated by the dotted black line at 5% of hemolysis. According to this criterion, all concentrations below 25 µg/mL of both types of nanoparticles were hemocompatible, as the percentage of red blood cell lysis remained below 5%. However, concentrations of 50 and 100 µg/mL of Fe3O4 and Fe3O4-Ag decorated nanoparticles clearly induced erythrocyte lysis, thus proving toxic to these cells.
To determine whether Fe3O4 and Fe3O4-Ag decorated nanoparticles induce an in vitro inflammatory response, nitrite production was measured upon the exposure of macrophages to different concentrations of nanoparticles. As illustrated in Figure 8b, concentrations below 10 µg/mL of both types of nanoparticles induced small concentrations of nitrites (less than 20 µM). However, concentrations above 25 µg/mL of Fe3O4 and Fe3O4-Ag decorated nanoparticles induced higher amounts of nitrites (30 to 42 µM), eliciting an inflammatory in vitro response comparable to the control, where the nitrites concentration is 50 µM.
These findings suggest that Fe3O4 and Fe3O4-Ag decorated nanoparticles exhibit promising hemocompatibility and low inflammatory potential at concentrations below 25 μg/mL, making them suitable candidates for various biomedical applications [5]. The observed hemocompatibility, where the percentage of red blood cell lysis remained below the 5% threshold, indicates that these nanoparticles are unlikely to cause significant damage to erythrocytes at lower concentrations. Similarly, the relatively low levels of nitrite production, a marker of inflammation, suggest that these nanoparticles elicit a minimal inflammatory response in macrophages at lower doses. However, the study also revealed that higher concentrations of 50 and 100 μg/mL of decorated Fe3O4 and Fe3O4-Ag nanoparticles can induce significant erythrocyte lysis and higher levels of nitrites, suggesting a potential toxicity and a more pronounced inflammatory reaction. This highlights the importance of carefully considering the dosage and design of these nanoparticles to ensure their safe and effective use in biomedical contexts [59,60]. Further optimization and comprehensive evaluation of their biocompatibility at different concentrations will be crucial to develop these nanoparticles as reliable and safe tools for various therapeutic and diagnostic applications [61,62].

3.8. Zeta Potential Analysis

Figure 9 presents a zeta potential analysis conducted on the magnetic phase of the samples (Fe3O4), aimed at understanding the behavior of the surface charges of the magnetic nanoparticles and their interaction with the surrounding colloidal medium. This figure shows that at a pH very close to 7, the zeta potential is nearly zero, indicating low electrostatic repulsion and a propensity for agglomeration due to Van der Waals forces and magnetic interactions [63]. In this context, it is important to mention that the pH of the cell culture medium was 7.4, the same medium to which the magnetic nanoparticle treatments were added. Consequently, with this pH of 7.4 and a zeta potential very close to zero, it is possible that there is an agglomeration of the nanoparticles due to surface charges and magnetic interactions. This agglomeration could contribute to the lack of toxicity and cell death, as the agglomerated particles might be less reactive [64].
Magnetic nanoparticles with a zeta potential close to zero tend to form agglomerates that are less toxic to cells. This is due to several factors: interactions between nanoparticles and cells are less intense when the particles are agglomerated, reducing the likelihood of cellular damage [65]. Additionally, cells can internalize individual nanoparticles more easily than large agglomerates, and the internalization of individual nanoparticles can trigger cytotoxic responses, whereas large agglomerates are less likely to be internalized [66,67]. Furthermore, individual nanoparticles can release metal ions that are toxic to cells, but agglomeration reduces the exposed surface area and, consequently, the release of ions. In summary, the magnetic nanoparticles reported in this research, exhibits a zeta potential close to zero, which can lead to particle agglomeration, reducing their reactivity and cellular toxicity. However, further studies are necessary to fully understand the implications of agglomeration and nanoparticle reactivity in various biological contexts.

4. Conclusions

The study analyzed the cytotoxic effects of Fe3O4 and Fe3O4-Ag decorated nanoparticles on human fibroblasts (HFF-1) and breast cancer cells (MCF-7) to evaluate their biocompatibility and potential therapeutic efficacy. The results showed that neither type of nanoparticle significantly reduced cell viability in HFF-1 fibroblasts, indicating that they are non-cytotoxic at the tested concentrations on a non-cancerous cell line. Similarly, MCF-7 breast cancer cells did not exhibit a significant change in viability when exposed to different nanoparticle concentrations, suggesting that these nanoparticles are biocompatible and allow normal cell growth. While evaluating the cytotoxic effects of these nanosystems in combination with other antineoplastic drugs across various cell lines from healthy tissues and neoplasms is crucial, this study primarily focuses on elucidating the biocompatibility of magnetite nanoparticles. This highlights the potential of Fe3O4 and Fe3O4-Ag nanoparticles for biomedical applications, given their compatibility with both normal and cancerous cells.
Reactive oxygen species (ROS) production by Fe3O4 and Fe3O4-Ag nanoparticles was also examined to assess their biocompatibility in aims of their future therapeutic potential. The study found that higher concentrations of Fe3O4-Ag nanoparticles decreased ROS production in both HFF-1 and MCF-7 cells, while Fe3O4 nanoparticles were more effective in generating ROS at these concentrations. However, excessive ROS production can harm normal cells, underscoring the need to evaluate nanoparticle concentrations to balance therapeutic efficacy and safety carefully. These findings contribute to understanding nanoparticle interactions with cellular oxidative mechanisms, which are crucial for developing safe and effective nanoparticle-based therapies. Altogether, the results herein indicated that it is possible to use Fe3O4-Ag nanoparticles as future drug delivery agents and enhance the synergy of their physicochemical properties with the antiproliferative mechanisms of the anticancer agents, also seeking the repositioning of front-line drugs for the treatment of public health diseases.

Author Contributions

Conceptualization, Á.d.J.R.-B. and K.J.-M.; methodology, K.J.-M., S.Y.R.-L. and Á.d.J.R.-B.; software, K.J.-M. and Á.d.J.R.-B.; validation, K.J.-M.; formal analysis, K.J.-M. and Á.d.J.R.-B.; investigation, K.J.-M. and Á.d.J.R.-B.; resources, Á.d.J.R.-B. and K.J.-M.; data curation, K.J.-M. and Á.d.J.R.-B.; writing—original draft preparation, Á.d.J.R.-B. and K.J.-M.; writing—review and editing, K.J.-M., Á.d.J.R.-B., N.M.-L. and S.Y.R.-L.; visualization, K.J.-M., Á.d.J.R.-B., S.Y.R.-L. and N.M.-L.; supervision, K.J.-M. and Á.d.J.R.-B.; project administration, Á.d.J.R.-B.; funding acquisition, Á.d.J.R.-B., S.Y.R.-L., N.M.-L. and K.J.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive external funding; The resources used were contributed by the authors and their current projects.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on request.

Acknowledgments

Álvaro de Jesús Ruíz-Baltazar wishes to express profound gratitude for the invaluable support provided by the National Council for Humanities, Science, and Technology (CONAHCYT, Mexico), in collaboration with the Center of Applied Physics and Advanced Technology (CFATA-UNAM), through the “Investigadoras e Investigadores por México-CONAHCYT” program. Additionally, heartfelt thanks are extended to the “Instituto de Ciencias Biomédicas de la UACJ” for their unwavering support during the sabbatical period. The authors would like to express their acknowledgment of Adriana Gonzalez-Gallardo (INB-UNAM), for her technical assistance. Additionally, the authors appreciate the support from the Microscopy Laboratory at the National Laboratory for Materials Characterization (LanCam), through the academic technician Manuel Aguilar Franco.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kumar, A.; Shah, S.R.; Jayeoye, T.J.; Kumar, A.; Parihar, A.; Prajapati, B.; Singh, S.; Kapoor, D.U. Biogenic Metallic Nanoparticles: Biomedical, Analytical, Food Preservation, and Applications in Other Consumable Products. Front. Nanotechnol. 2023, 5. [Google Scholar] [CrossRef]
  2. Krishnasamy, R.; Obbineni, J.M. Methods for Green Synthesis of Metallic Nanoparticles Using Plant Extracts and Their Biological Applications-A Review. J. Biomim. Biomater. Biomed. Eng. 2022, 56, 75–151. [Google Scholar] [CrossRef]
  3. Yaraki, M.T.; Zahed Nasab, S.; Zare, I.; Dahri, M.; Moein Sadeghi, M.; Koohi, M.; Tan, Y.N. Biomimetic Metallic Nanostructures for Biomedical Applications, Catalysis, and Beyond. Ind. Eng. Chem. Res. 2022, 61, 7547–7593. [Google Scholar] [CrossRef]
  4. Antonelli, A.; Magnani, M. SPIO Nanoparticles and Magnetic Erythrocytes as Contrast Agents for Biomedical and Diagnostic Applications. J. Magn. Magn. Mater. 2022, 541, 168520. [Google Scholar] [CrossRef]
  5. Materón, E.M.; Miyazaki, C.M.; Carr, O.; Joshi, N.; Picciani, P.H.S.; Dalmaschio, C.J.; Davis, F.; Shimizu, F.M. Magnetic Nanoparticles in Biomedical Applications: A Review. Appl. Surf. Sci. Adv. 2021, 6, 100163. [Google Scholar] [CrossRef]
  6. Liu, X.; Wang, X.; Zhang, H.; Li, X.; Wang, Y.; Zhang, X. Magnetoplasmonic Materials for Biomedical Applications: Synthesis and Properties. J. Mater. Chem. B 2019, 7, 1917–1930. [Google Scholar]
  7. Klein, S.; Hübner, J.; Menter, C.; Distel, L.V.R.; Neuhuber, W.; Kryschi, C. A Facile One-Pot Synthesis Ofwater-Soluble, Patchy Fe3O4-Au Nanoparticles for Application in Radiation Therapy. Appl. Sci. 2019, 9, 15. [Google Scholar] [CrossRef]
  8. Danafar, H.; Baghdadchi, Y.; Barsbay, M.; Ghaffarlou, M.; Mousazadeh, N.; Mohammadi, A. Synthesis of Fe3O4-Gold Hybrid Nanoparticles Coated by Bovine Serum Albumin as a Contrast Agent in MR Imaging. Heliyon 2023, 9, e13874. [Google Scholar] [CrossRef]
  9. de Jesús Ruíz-Baltazar, Á. Green Synthesis Assisted by Sonochemical Activation of Fe3O4-Ag Nano-Alloys: Structural Characterization and Studies of Sorption of Cationic Dyes. Inorg. Chem. Commun. 2020, 120, 108148. [Google Scholar] [CrossRef]
  10. Alshameri, A.W.; Owais, M. Antibacterial and Cytotoxic Potency of the Plant-Mediated Synthesis of Metallic Nanoparticles Ag NPs and ZnO NPs: A Review. OpenNano 2022, 8, 100077. [Google Scholar] [CrossRef]
  11. Sarani, M.; Roostaee, M.; Adeli-Sardou, M.; Kalantar-Neyestanaki, D.; Mousavi, S.A.A.; Amanizadeh, A.; Barani, M.; Amirbeigi, A. Green Synthesis of Ag and Cu-Doped Bismuth Oxide Nanoparticles: Revealing Synergistic Antimicrobial and Selective Cytotoxic Potentials for Biomedical Advancements. J. Trace Elem. Med. Biol. 2024, 81, 127325. [Google Scholar] [CrossRef]
  12. Pannerselvam, B.; Thiyagarajan, D.; Pazhani, A.; Thangavelu, K.P.; Kim, H.J.; Rangarajulu, S.K. Copperpod Plant Synthesized Agnps Enhance Cytotoxic and Apoptotic Effect in Cancer Cell Lines. Processes 2021, 9, 888. [Google Scholar] [CrossRef]
  13. Swathi, S.; Ameen, F.; Ravi, G.; Yuvakkumar, R.; Hong, S.I.; Velauthapillai, D.; AlKahtani, M.D.F.; Thambidurai, M.; Dang, C. Cancer Targeting Potential of Bioinspired Chain like Magnetite (Fe3O4) Nanostructures. Curr. Appl. Phys. 2020, 20, 982–987. [Google Scholar] [CrossRef]
  14. Mukha, I.; Chepurna, O.; Vityuk, N.; Khodko, A.; Storozhuk, L.; Dzhagan, V.; Zahn, D.R.T.; Ntziachristos, V.; Chmyrov, A.; Ohulchanskyy, T.Y. Multifunctional Magneto-Plasmonic Fe3O4/Au Nanocomposites: Approaching Magnetophoretically-Enhanced Photothermal Therapy. Nanomaterials 2021, 11, 1113. [Google Scholar] [CrossRef] [PubMed]
  15. Usman, F.; Ghazali, K.H.; Muda, R.; Johari, N.H.; Dennis, J.O.; Tamam, N.; Sulieman, A.; Ji, Y. Magnetoresistance and Magneto-Plasmonic Sensors for the Detection of Cancer Biomarkers: A Bibliometric Analysis and Recent Advances. Sens. Actuators Rep. 2023, 6, 100172. [Google Scholar] [CrossRef]
  16. Singh, R.S.; Sarswat, P.K. From Fundamentals to Applications: The Development of Magnetoplasmonics for next-Generation Technologies. Mater. Today Electron. 2023, 4, 100033. [Google Scholar] [CrossRef]
  17. Raja, K.; Balamurugan, V.; Selvakumar, S.; Vasanth, K. Striga Angustifolia Mediated Synthesis of Silver Nanoparticles: Anti-Microbial, Antioxidant and Anti-Proliferative Activity in Apoptotic P53 Signalling Pathway. J. Drug Deliv. Sci. Technol. 2022, 67, 102945. [Google Scholar] [CrossRef]
  18. Enciu, A.-M.; Codrici, E.; Popescu, D.I.; Preda, P.; Stan, D.; Avram, M.; Tanase, C. Aloe Vera Gel and Sea-Buckthorn Oil Protect Human Fibroblasts against Silver Nanoparticles Cytotoxicity. Toxicol. Lett. 2021, 350, S238–S239. [Google Scholar] [CrossRef]
  19. Lewinska, A.; Adamczyk-Grochala, J.; Bloniarz, D.; Olszowka, J.; Kulpa-Greszta, M.; Litwinienko, G.; Tomaszewska, A.; Wnuk, M.; Pazik, R. AMPK-Mediated Senolytic and Senostatic Activity of Quercetin Surface Functionalized Fe3O4 Nanoparticles during Oxidant-Induced Senescence in Human Fibroblasts. Redox Biol. 2020, 28, 101337. [Google Scholar] [CrossRef]
  20. Kazimir, A.; Schwarze, B.; Lönnecke, P.; Jelača, S.; Mijatović, S.; Maksimović-Ivanić, D.; Hey-Hawkins, E. Metallodrugs against Breast Cancer: Combining the Tamoxifen Vector with Platinum(II) and Palladium(II) Complexes. Pharmaceutics 2023, 15, 682. [Google Scholar] [CrossRef]
  21. Liu, X.; Wang, X.; Zhang, H.; Li, X.; Wang, Y.; Zhang, X. Magnetoplasmonic Nanomaterials for Biomedical Applications. Nanoscale Adv. 2020, 2, 2524–2540. [Google Scholar]
  22. Liu, X.; Wang, X.; Zhang, H.; Li, X.; Wang, Y.; Zhang, X. Magnetoplasmonic Nanomaterials for Biosensing and Bioimaging Applications. J. Mater. Chem. B 2021, 9, 4688–4708. [Google Scholar]
  23. Jandrotia, R.; Gupta, I.; Mahajan, P.; Batish, D.R.; Singh, H.P. 2—Green Nanomaterials: An Eco-Friendly Route for Sustainable Nanotechnology. In Nanotechnology and Nanomaterials in the Agri-Food Industries; Singh, P., Khare, P., Mishra, D., Bilal, M., Sillanpää, M., Eds.; Micro and Nano Technologies; Elsevier: Amsterdam, The Netherlands, 2024; pp. 21–52. ISBN 978-0-323-99682-2. [Google Scholar]
  24. Rahmayanti, M.; Nurul Syakina, A.; Fatimah, I.; Sulistyaningsih, T. Green Synthesis of Magnetite Nanoparticles Using Peel Extract of Jengkol (Archidendron pauciflorum) for Methylene Blue Adsorption from Aqueous Media. Chem. Phys. Lett. 2022, 803, 139834. [Google Scholar] [CrossRef]
  25. Zhang, X.F.; Liu, Z.G.; Shen, W.; Gurunathan, S. Silver Nanoparticles: Synthesis, Characterization, Properties, Applications, and Therapeutic Approaches. Int. J. Mol. Sci. 2016, 17, 1534. [Google Scholar] [CrossRef]
  26. Cheng, Z.; Tang, S.W.; Feng, J.; Wu, Y. Biosynthesis and Antibacterial Activity of Silver Nanoparticles Using Flos Sophorae Immaturus Extract. Heliyon 2022, 8, e10010. [Google Scholar] [CrossRef]
  27. Kobylinska, N.; Klymchuk, D.; Khaynakova, O.; Duplij, V.; Matvieieva, N. Morphology-Controlled Green Synthesis of Magnetic Nanoparticles Using Extracts of ‘Hairy’ Roots: Environmental Application and Toxicity Evaluation. Nanomaterials 2022, 12, 4231. [Google Scholar] [CrossRef]
  28. Antwi-Baah, R.; Wang, Y.; Chen, X.; Liu, H.; Yu, K. Hybrid Morphologies of Paramagnetic Manganese-Based Nanoparticles as Theranostics. Chem. Eng. J. 2023, 466, 142970. [Google Scholar] [CrossRef]
  29. Sánchez-López, E.; Gomes, D.; Esteruelas, G.; Bonilla, L.; Lopez-Machado, A.L.; Galindo, R.; Cano, A.; Espina, M.; Ettcheto, M.; Camins, A.; et al. Metal-Based Nanoparticles as Antimicrobial Agents: An Overview. Nanomaterials 2020, 10, 292. [Google Scholar] [CrossRef] [PubMed]
  30. Kansız, S.; Elçin, Y.M. Advanced Liposome and Polymersome-Based Drug Delivery Systems: Considerations for Physicochemical Properties, Targeting Strategies and Stimuli-Sensitive Approaches. Adv. Colloid Interface Sci. 2023, 317, 102930. [Google Scholar] [CrossRef]
  31. Nair, V.V.; Cabrera, P.; Ramírez-Lecaros, C.; Jara, M.O.; Brayden, D.J.; Morales, J.O. Buccal Delivery of Small Molecules and Biologics: Of Mucoadhesive Polymers, Films, and Nanoparticles—An Update. Int. J. Pharm. 2023, 636, 122789. [Google Scholar] [CrossRef]
  32. Pérez-Díaz, M.; Alvarado-Gomez, E.; Magaña-Aquino, M.; Sánchez-Sánchez, R.; Velasquillo, C.; Gonzalez, C.; Ganem-Rondero, A.; Martínez-Castañon, G.; Zavala-Alonso, N.; Martinez-Gutierrez, F. Anti-Biofilm Activity of Chitosan Gels Formulated with Silver Nanoparticles and Their Cytotoxic Effect on Human Fibroblasts. Mater. Sci. Eng. C 2016, 60, 317–323. [Google Scholar] [CrossRef]
  33. Itoo, A.M.; Vemula, S.L.; Gupta, M.T.; Giram, M.V.; Kumar, S.A.; Ghosh, B.; Biswas, S. Multifunctional Graphene Oxide Nanoparticles for Drug Delivery in Cancer. J. Control. Release 2022, 350, 26–59. [Google Scholar] [CrossRef] [PubMed]
  34. Mejía-Méndez, J.L.; Navarro-López, D.E.; Sanchez-Martinez, A.; Ceballos-Sanchez, O.; Garcia-Amezquita, L.E.; Tiwari, N.; Juarez-Moreno, K.; Sanchez-Ante, G.; López-Mena, E.R. Lanthanide-Doped ZnO Nanoparticles: Unraveling Their Role in Cytotoxicity, Antioxidant Capacity, and Nanotoxicology. Antioxidants 2024, 13, 213. [Google Scholar] [CrossRef] [PubMed]
  35. Mavil-Guerrero, E.; Vazquez-Duhalt, R.; Juarez-Moreno, K. Exploring the Cytotoxicity Mechanisms of Copper Ions and Copper Oxide Nanoparticles in Cells from the Excretory System. Chemosphere 2024, 347, 140713. [Google Scholar] [CrossRef] [PubMed]
  36. Munive-Olarte, A.; Hidalgo-Moyle, J.J.; Velasquillo, C.; Juarez-Moreno, K.; Mota-Morales, J.D. Boosting Cell Proliferation in Three-Dimensional Polyacrylates/Nanohydroxyapatite Scaffolds Synthesized by Deep Eutectic Solvent-Based Emulsion Templating. J. Colloid Interface Sci. 2022, 607, 298–311. [Google Scholar] [CrossRef] [PubMed]
  37. Olvera-Guillen, R.; Juarez-Moreno, K.; Cruz-Soto, M.; Rocha-Botello, G.; Herrera-Ordonez, J. Supramolecular Poly(Vinyl Alcohol)-Folate Structure as Functional Layer and Colloidal Stabilizer of Poly(Vinyl Acetate) Nanoparticles with Potential Use as Nanocarrier for Hydrophobic Antitumor Agents. J. Nanoparticle Res. 2021, 23, 132. [Google Scholar] [CrossRef]
  38. Chávez-García, D.; Juarez-Moreno, K.; Campos, C.H.; Tejeda, E.M.; Alderete, J.B.; Hirata, G.A. Cytotoxicity, Genotoxicity and Uptake Detection of Folic Acid-Functionalized Green Upconversion Nanoparticles Y2O3/Er3+, Yb3+ as Biolabels for Cancer Cells. J. Mater. Sci. 2018, 53, 6665–6680. [Google Scholar] [CrossRef]
  39. Juarez-Moreno, K.; Chávez-García, D.; Hirata, G.; Vazquez-Duhalt, R. Monolayer (2D) or Spheroids (3D) Cell Cultures for Nanotoxicological Studies? Comparison of Cytotoxicity and Cell Internalization of Nanoparticles. Toxicol. Vitr. 2022, 85, 105461. [Google Scholar] [CrossRef]
  40. Salih, S.J.; Mahmood, W.M. Review on Magnetic Spinel Ferrite (MFe2O4) Nanoparticles: From Synthesis to Application. Heliyon 2023, 9, e16601. [Google Scholar] [CrossRef]
  41. Espinosa, A.; Reguera, J.; Curcio, A.; Muñoz-Noval, Á.; Kuttner, C.; Van de Walle, A.; Liz-Marzán, L.M.; Wilhelm, C. Janus Magnetic-Plasmonic Nanoparticles for Magnetically Guided and Thermally Activated Cancer Therapy. Small 2020, 16, e1904960. [Google Scholar] [CrossRef]
  42. Hong, T.; Jiang, M.; Hu, Y.; Tang, Z.; Huang, K. Nonlinear Magnetization Relaxation of Uniaxial Anisotropic Ferromagnetic Particles with Linear Reaction Dynamics Driven by a Strong Ac Magnetic Field. Chem. Phys. Lett. 2021, 782, 139017. [Google Scholar] [CrossRef]
  43. Bôa Morte, E.F.; Marum, D.S.; Saitovitch, E.B.; Alzamora, M.; Monteiro, S.N.; Sanchez Rodriguez, R.J. Modified Magnetite Nanoparticle as Biocatalytic Support for Magnetically Stabilized Fluidized Bed Reactors. J. Mater. Res. Technol. 2021, 14, 1112–1125. [Google Scholar] [CrossRef]
  44. Lai, C.-H.; Chuang, Y.-K.; Chang, Y.-C.; Chen, C.-H.; Chen, P.-Y.; Chen, I.-W.; Chen, Y.-C. Magnetic-Plasmonic Core-Shell Nanoparticles for Multimodal Biomedical Imaging. Nanotechnology 2017, 28, 135102. [Google Scholar] [CrossRef]
  45. Tan, L.K.S.; How, C.W.; Low, L.E.; Ong, B.H.; Loh, J.S.; Lim, S.-Y.; Ong, Y.S.; Foo, J.B. Magnetic-Guided Targeted Delivery of Zerumbone/SPION Co-Loaded in Nanostructured Lipid Carrier into Breast Cancer Cells. J. Drug Deliv. Sci. Technol. 2023, 87, 104830. [Google Scholar] [CrossRef]
  46. Korolkov, I.V.; Shumskaya, A.; Kozlovskiy, A.L.; Kaliyekperov, M.E.; Lissovskaya, L.I.; Zdorovets, M.V. Magnetic-Plasmonic Ni Nanotubes Covered with Gold for Improvement of SERS Analysis. J. Alloys Compd. 2022, 901, 163661. [Google Scholar] [CrossRef]
  47. Garanina, A.S.; Efremova, M.V.; Machulkin, A.E.; Lyubin, E.V.; Vorobyeva, N.S.; Zhironkina, O.A.; Strelkova, O.S.; Kireev, I.I.; Alieva, I.B.; Uzbekov, R.E.; et al. Bifunctional Magnetite–Gold Nanoparticles for Magneto-Mechanical Actuation and Cancer Cell Destruction. Magnetochemistry 2022, 8, 185. [Google Scholar] [CrossRef]
  48. Brullot, W.; Valev, V.K.; Verbiest, T. Magnetic-Plasmonic Nanoparticles for the Life Sciences: Calculated Optical Properties of Hybrid Structures. Nanomed. Nanotechnol. Biol. Med. 2012, 8, 559–568. [Google Scholar] [CrossRef]
  49. de Jesús Ruíz-Baltazar, Á. Sonochemical Activation-Assisted Biosynthesis of Au/Fe3O4 Nanoparticles and Sonocatalytic Degradation of Methyl Orange. Ultrason. Sonochem 2021, 73, 105521. [Google Scholar] [CrossRef]
  50. Amarjargal, A.; Tijing, L.D.; Im, I.T.; Kim, C.S. Simultaneous Preparation of Ag/Fe3O4 Core-Shell Nanocomposites with Enhanced Magnetic Moment and Strong Antibacterial and Catalytic Properties. Chem. Eng. J. 2013, 226, 243–254. [Google Scholar] [CrossRef]
  51. Pieretti, J.C.; Gonçalves, M.C.; Nakazato, G.; Santos de Souza, A.C.; Boudier, A.; Seabra, A.B. Multifunctional Hybrid Nanoplatform Based on Fe3O4@Ag NPs for Nitric Oxide Delivery: Development, Characterization, Therapeutic Efficacy, and Hemocompatibility. J. Mater. Sci. Mater. Med. 2021, 32, 23. [Google Scholar] [CrossRef]
  52. Nguyen, M.D.; Tran, H.V.; Xu, S.; Lee, T.R. Fe3O4 Nanoparticles: Structures, Synthesis, Magnetic Properties, Surface Functionalization, and Emerging Applications. Appl. Sci. 2021, 11, 11301. [Google Scholar] [CrossRef]
  53. Anderson, J.M. Future Challenges in the in Vitro and in Vivo Evaluation of Biomaterial Biocompatibility. Regen. Biomater. 2016, 3, 73–77. [Google Scholar] [CrossRef]
  54. Horie, M.; Tabei, Y. Role of Oxidative Stress in Nanoparticles Toxicity. Free Radic. Res. 2021, 55, 331–342. [Google Scholar] [CrossRef]
  55. Paunovic, J.; Vucevic, D.; Radosavljevic, T.; Mandić-Rajčević, S.; Pantic, I. Iron-Based Nanoparticles and Their Potential Toxicity: Focus on Oxidative Stress and Apoptosis. Chem. Biol. Interact. 2020, 316, 108935. [Google Scholar] [CrossRef]
  56. Konate, A.; He, X.; Rui, Y.-K.; Zhang, Z.-Y. Magnetite (Fe3O4) Nanoparticles Alleviate Growth Inhibition and Oxidative Stress Caused by Heavy Metals in Young Seedlings of Cucumber (Cucumis sativus L.). ITM Web Conf. 2017, 12, 03034. [Google Scholar] [CrossRef]
  57. Yang, L.X.; Wu, Y.N.; Wang, P.W.; Huang, K.J.; Su, W.C.; Shieh, D. Bin Silver-Coated Zero-Valent Iron Nanoparticles Enhance Cancer Therapy in Mice through Lysosome-Dependent Dual Programed Cell Death Pathways: Triggering Simultaneous Apoptosis and Autophagy Only in Cancerous Cells. J. Mater. Chem. B 2020, 8, 4122–4131. [Google Scholar] [CrossRef]
  58. Sankaranarayanan, S.A.; Thomas, A.; Revi, N.; Ramakrishna, B.; Rengan, A.K. Iron Oxide Nanoparticles for Theranostic Applications—Recent Advances. J. Drug Deliv. Sci. Technol. 2022, 70, 103196. [Google Scholar] [CrossRef]
  59. Siddiqui, M.A.; Wahab, R.; Saquib, Q.; Ahmad, J.; Farshori, N.N.; Al-Sheddi, E.S.; Al-Oqail, M.M.; Al-Massarani, S.M.; Al-Khedhairy, A.A. Iron Oxide Nanoparticles Induced Cytotoxicity, Oxidative Stress, Cell Cycle Arrest, and DNA Damage in Human Umbilical Vein Endothelial Cells. J. Trace Elem. Med. Biol. 2023, 80, 127302. [Google Scholar] [CrossRef]
  60. Krishnan, N.; Peng, F.X.; Mohapatra, A.; Fang, R.H.; Zhang, L. Genetically Engineered Cellular Nanoparticles for Biomedical Applications. Biomaterials 2023, 296, 122065. [Google Scholar] [CrossRef]
  61. Joshi, R.; Missong, H.; Mishra, J.; Kaur, S.; Saini, S.; Kandimalla, R.; Reddy, P.H.; Babu, A.; Bhatti, G.K.; Bhatti, J.S. Nanotheranostics Revolutionizing Neurodegenerative Diseases: From Precision Diagnosis to Targeted Therapies. J. Drug Deliv. Sci. Technol. 2023, 89, 105067. [Google Scholar] [CrossRef]
  62. Krishna Kumar, A.S.; Tseng, W.-B.; Arputharaj, E.; Huang, P.-J.; Tseng, W.-L.; Bajda, T. Covalent Organic Framework Nanosheets as an Enhancer for Light-Responsive Oxidase-like Nanozymes: Multifunctional Applications in Colorimetric Sensing, Antibiotic Degradation, and Antibacterial Agents. ACS Sustain. Chem. Eng. 2023, 11, 6956–6969. [Google Scholar] [CrossRef]
  63. Bhattacharjee, S. DLS and Zeta Potential—What They Are and What They Are Not? J. Control. Release 2016, 235, 337–351. [Google Scholar] [CrossRef] [PubMed]
  64. Lead, J.R.; Batley, G.E.; Alvarez, P.J.J.; Croteau, M.N.; Handy, R.D.; McLaughlin, M.J.; Judy, J.D.; Schirmer, K. Nanomaterials in the Environment: Behavior, Fate, Bioavailability, and Effects—An Updated Review. Env. Toxicol. Chem. 2018, 37, 2029–2063. [Google Scholar] [CrossRef] [PubMed]
  65. Ezealigo, U.S.; Ezealigo, B.N.; Aisida, S.O.; Ezema, F.I. Iron Oxide Nanoparticles in Biological Systems: Antibacterial and Toxicology Perspective. JCIS Open 2021, 4, 100027. [Google Scholar] [CrossRef]
  66. Richards, C.J.; Burgers, T.C.Q.; Vlijm, R.; Roos, W.H.; Åberg, C. Rapid Internalization of Nanoparticles by Human Cells at the Single Particle Level. ACS Nano 2023, 17, 16517–16529. [Google Scholar] [CrossRef]
  67. Egbuna, C.; Parmar, V.K.; Jeevanandam, J.; Ezzat, S.M.; Patrick-Iwuanyanwu, K.C.; Adetunji, C.O.; Khan, J.; Onyeike, E.N.; Uche, C.Z.; Akram, M.; et al. Toxicity of Nanoparticles in Biomedical Application: Nanotoxicology. J. Toxicol. 2021, 2021, 9954443. [Google Scholar] [CrossRef]
Figure 1. SEM micrographs depicting the Fe3O4-Ag nanoparticle sample. (a) SE-HA ima.e showing nanoparticle topology and the presence of Fe3O4 and Ag phases. (b) SE(U) image confirming Fe3O4 and Ag presence, providing high-resolution surface details. (c) HA(T) image emphasizing Fe3O4-Ag formation. (d) SE + HA + OFF composite image displaying surface morphology and elemental composition. Magnification: ×500k; Working distance: 3.7 mm; Acceleration voltage: 3.0 kV. (e) Scheme of the distribution of Fe3O4 and Ag nanostructures corresponding to the decorated type Fe3O4-Ag nanoparticle samples.
Figure 1. SEM micrographs depicting the Fe3O4-Ag nanoparticle sample. (a) SE-HA ima.e showing nanoparticle topology and the presence of Fe3O4 and Ag phases. (b) SE(U) image confirming Fe3O4 and Ag presence, providing high-resolution surface details. (c) HA(T) image emphasizing Fe3O4-Ag formation. (d) SE + HA + OFF composite image displaying surface morphology and elemental composition. Magnification: ×500k; Working distance: 3.7 mm; Acceleration voltage: 3.0 kV. (e) Scheme of the distribution of Fe3O4 and Ag nanostructures corresponding to the decorated type Fe3O4-Ag nanoparticle samples.
Applsci 14 06750 g001
Figure 2. Chemical Analysis of Fe3O4-Ag Decorated Nanoparticles. (a) SE image of Fe3O4. (b) Elemental mapping showing Ag distribution, confirming Fe3O4-Ag structure. Elemental mappings of Fe, O, and Ag from SE image of Fe3O4 are in (ce). (f) Chemical analysis confirms Fe, O, and Ag composition.
Figure 2. Chemical Analysis of Fe3O4-Ag Decorated Nanoparticles. (a) SE image of Fe3O4. (b) Elemental mapping showing Ag distribution, confirming Fe3O4-Ag structure. Elemental mappings of Fe, O, and Ag from SE image of Fe3O4 are in (ce). (f) Chemical analysis confirms Fe, O, and Ag composition.
Applsci 14 06750 g002
Figure 3. Experimental X-ray Diffraction (XRD) Pattern of the Fe3O4-Ag Decorated Nanoparticles.
Figure 3. Experimental X-ray Diffraction (XRD) Pattern of the Fe3O4-Ag Decorated Nanoparticles.
Applsci 14 06750 g003
Figure 4. Magnetic Characterization of Fe3O4/Ag Nanoalloys. (a) Hysteresis curve analysis at 300 K confirms superparamagnetic behavior in Fe3O4/Ag nanoalloys, with negligible magnetic coercivity and a magnetization value of 47 emu/g. (b) Zero-field-cooled (ZFC) curve depicts superparamagnetic behavior across temperatures, with a broad peak indicating the average blocking temperature.
Figure 4. Magnetic Characterization of Fe3O4/Ag Nanoalloys. (a) Hysteresis curve analysis at 300 K confirms superparamagnetic behavior in Fe3O4/Ag nanoalloys, with negligible magnetic coercivity and a magnetization value of 47 emu/g. (b) Zero-field-cooled (ZFC) curve depicts superparamagnetic behavior across temperatures, with a broad peak indicating the average blocking temperature.
Applsci 14 06750 g004
Figure 5. XPS Spectra of Fe3O4-Ag Decorated Nanostructures: (a) Survey Spectra, (b) HR-XPS of Fe 2p, (c) O 1s, and (d) Ag 3d.
Figure 5. XPS Spectra of Fe3O4-Ag Decorated Nanostructures: (a) Survey Spectra, (b) HR-XPS of Fe 2p, (c) O 1s, and (d) Ag 3d.
Applsci 14 06750 g005
Figure 6. Cell viability assay of Fe3O4 and Fe3O4-Ag decorated nanoparticles on (a) human fibroblast (HFF-1), and (b) breast cancer (MCF-7) cells. The cell viability results are expressed as the mean of cell viability percentage ± the standard deviation from three independent experiments. Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Significance was indicated as * p < 0.05 and ** p < 0.01.
Figure 6. Cell viability assay of Fe3O4 and Fe3O4-Ag decorated nanoparticles on (a) human fibroblast (HFF-1), and (b) breast cancer (MCF-7) cells. The cell viability results are expressed as the mean of cell viability percentage ± the standard deviation from three independent experiments. Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Significance was indicated as * p < 0.05 and ** p < 0.01.
Applsci 14 06750 g006
Figure 7. ROS measurement production induced by Fe3O4 and Fe3O4-Ag decorated nanoparticles on (a) human fibroblast (HFF-1) and (b) breast cancer cells (MCF-7). Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Statistical significance was indicated as * p < 0.05 and ** p < 0.01 and *** p < 0.001.
Figure 7. ROS measurement production induced by Fe3O4 and Fe3O4-Ag decorated nanoparticles on (a) human fibroblast (HFF-1) and (b) breast cancer cells (MCF-7). Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Statistical significance was indicated as * p < 0.05 and ** p < 0.01 and *** p < 0.001.
Applsci 14 06750 g007
Figure 8. (a) Hemolysis of erythrocytes and (b) nitrite production in macrophages induced by Fe3O4 and Fe3O4-Ag decorated nanoparticles. Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Statistical significance was indicated as * p < 0.05, ** p < 0.01, and *** p > 0.001 and **** p > 0.0001.
Figure 8. (a) Hemolysis of erythrocytes and (b) nitrite production in macrophages induced by Fe3O4 and Fe3O4-Ag decorated nanoparticles. Statistical analysis was performed using two-way ANOVA followed by Tukey’s multiple comparison test. Statistical significance was indicated as * p < 0.05, ** p < 0.01, and *** p > 0.001 and **** p > 0.0001.
Applsci 14 06750 g008
Figure 9. Zeta potential graph of Fe3O4 nanoparticles as a function of pH.
Figure 9. Zeta potential graph of Fe3O4 nanoparticles as a function of pH.
Applsci 14 06750 g009
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ruíz-Baltazar, Á.d.J.; Reyes-López, S.Y.; Méndez-Lozano, N.; Juárez-Moreno, K. Evaluation of Superparamagnetic Fe3O4-Ag Decorated Nanoparticles: Cytotoxicity Studies in Human Fibroblasts (HFF-1) and Breast Cancer Cells (MCF-7). Appl. Sci. 2024, 14, 6750. https://doi.org/10.3390/app14156750

AMA Style

Ruíz-Baltazar ÁdJ, Reyes-López SY, Méndez-Lozano N, Juárez-Moreno K. Evaluation of Superparamagnetic Fe3O4-Ag Decorated Nanoparticles: Cytotoxicity Studies in Human Fibroblasts (HFF-1) and Breast Cancer Cells (MCF-7). Applied Sciences. 2024; 14(15):6750. https://doi.org/10.3390/app14156750

Chicago/Turabian Style

Ruíz-Baltazar, Álvaro de Jesús, Simón Yobanny Reyes-López, Néstor Méndez-Lozano, and Karla Juárez-Moreno. 2024. "Evaluation of Superparamagnetic Fe3O4-Ag Decorated Nanoparticles: Cytotoxicity Studies in Human Fibroblasts (HFF-1) and Breast Cancer Cells (MCF-7)" Applied Sciences 14, no. 15: 6750. https://doi.org/10.3390/app14156750

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop