Home Silver nanowires as prospective carriers for drug delivery in cancer treatment: an in vitro biocompatibility study on lung adenocarcinoma cells and fibroblasts
Article Publicly Available

Silver nanowires as prospective carriers for drug delivery in cancer treatment: an in vitro biocompatibility study on lung adenocarcinoma cells and fibroblasts

  • Manisha Singh

    Manisha Singh was awarded a Masters in Molecular Medicine from Trinity College Dublin (Ireland) in 2013. Her thesis focused on an in vitro nanomedicine study of nanoparticles interaction with lung epithelial cells that included a comparative investigation in 2D and 3D cell culture models. In 2010, she graduated in Dental Surgery (BDS) at Rama Dental College (India). Her primary research interest includes nanomedicine, cell cultures and dental science.

    , Dania Movia

    Dania Movia is a post-doctoral researcher at the Centre for Research on Advanced and Adaptive Nanostructures (CRANN)/School of Medicine of Trinity College Dublin (Ireland). In 2007 she was awarded a BSc in Medicinal Chemistry at University of Trieste (Italy). In 2011 she completed her PhD in Chemistry at Trinity College Dublin with a thesis entitled “Single-walled carbon nanotubes as novel NIR fluorescent probes for biomedical optical imaging”.

    , Omar K. Mahfoud

    Omar K. Mahfoud is a PhD candidate in the Nanomedicine and Molecular Imaging Group at Trinity College Dublin (Ireland), where he is involved in an EU FP7-funded project (NAMDIATREAM) aimed at developing the next generation of nanomaterial-based diagnostic systems for the advanced detection of breast, lung and prostate cancer biomarkers. In 2008, he was awarded a BSc in Neuroscience at Trinity College Dublin and he completed his MSc in Molecular Medicine at the University Medical Centre Goettingen (Germany).

    , Yuri Volkov

    Yuri Volkov received his MD from the Moscow Medical University and subsequently a PhD in biomedical sciences, Institute of Immunology, Moscow. He is a Professor at the Department of Clinical Medicine and the Director of Research of the School of Medicine at Trinity College Dublin. Prof. Volkov coordinates a large scale EU FP-7 NAMDIATREAM with 22 European academic, research, clinical and industrial partners for early diagnostics and monitoring of malignant diseases. Lead TCD partner for EU FP-7 MULTIFUN and Principal investigator in many grants. Prof. Volkov published more than 80 articles, several patents and book chapters.

    and Adriele Prina-Mello

    Adriele Prina-Mello is a CRANN Investigator, a Senior Research Fellow of the School of Medicine and a part-time lecture at Trinity College Dublin (Ireland), a Nanosafety Cluster member and the vice-chair of the Nanodiagnostic working group of the European Technology Platform of Nanomedicine. Dr Prina-Mello is involved in developing and advancing several multidisciplinary research projects between University, Research Hospital and Industry partners for future applications in medicine and nanotechnology industry. Currently he is involved in 5 EU FP7 funded projects: NAMDIATREAM, MULTIFUN, QNANO, NANoREG, and AMCARE. Dr Prina-Mello has published more than 45 articles in biomedicine, nanotechnology, nanotoxicology and nanomedicine research area.

    EMAIL logo
Published/Copyright: December 2, 2013
Become an author with De Gruyter Brill

Abstract

Lung cancer is a major and increasing global health problem. While there have been significant advances in the understanding of lung cancer biology, still no current therapy exists to reduce the inevitable and lethal progression of this disease. Silver nanowires (AgNWs) are promising candidates for a wide range of biomedical applications and the treatment of life-threatening diseases due to their unique physico-chemical and biochemical properties. However, the safety of this nanomaterial and its use as a biomedical tool are still under debate. This study evaluates the in vitro internalisation, cytotoxicity and influence on the cell cycle of AgNWs in lung adenocarcinoma (A549) cells and lung normal fibroblasts (MRC-5 cells). Our results demonstrate that AgNWs could be internalised effectively into A549 and MRC-5 cells without inducing detectable cytotoxicity, thus providing preliminary evidence on the future potential of AgNWs as biocompatible drug delivery platforms applicable in lung cancer therapies.

Introduction

In spite of considerable advancements in the medical and oncological fields, lung cancer is still a devastating disease (1) and one of the most prominent causes of cancer-related mortality in Europe and USA (2, 3). The main reason for the short survival of lung cancer patients is the poor efficacy of currently available chemotherapeutic agents (4). In order to increase the success rate of the medical treatments, the pharmaceutical industry has recently developed targeted chemotherapeutic agents that are designed to be effective against specific subtypes of lung cancer defined by precise gene mutations (5, 6). Unfortunately, these mutations define mechanisms of drug sensitivity and/or resistance that are not shared by most patients or by other lung cancer subtypes (5, 7). For example, the patients with epidermal growth factor receptor (EGFR) mutations show great response to tyrosine kinase inhibitors (e.g., gefitinib and erlotinib), but patients with mutations to the KRAS gene are resistant to such therapy (6, 8). As a consequence, novel and more efficient anti-cancer treatments are needed against lung cancer.

Nanotechnology has tremendous potential to improve the precision of lung cancer therapy, and has become a key technology in the development of more sensitive diagnostic tools and more powerful pharmaceutical treatments (frequently offered as two-in-one (theranostic) systems) against cancer (6, 9–11). Nanomedicine products can, for example, be loaded with chemotherapeutic agents and specifically targeted to the tumor site, thus decreasing the side effects and increasing the therapeutic effect of the drugs (12). In particular, silver nanomaterials can find many remarkable applications in clinical practise, since they possess optimal chemical reactivity for the functionalisation of their surfaces with biologically active moieties (such as targeting and therapeutic molecules) (13), and are characterised by localised surface plasmon modes (14) that enable the development of tunable contrast agents, thermal ablation therapies and thermal-induced drug release (15). The working hypothesis behind this study was that silver nanowires (AgNWs) can be potentially implemented as theranostic, nano-enabled drug delivery platforms for lung cancer treatment once their biocompatibility has been proven.

The use of silver for biomedical purposes is quite widespread. In colloidal form, it is used for various purposes such as preparation of ointments (e.g., silver sulfadiazine remains the main topical product used in treating burn) (16) and treatment of bacterial infections in open wounds. In nano-form (17, 18), advanced technology has enabled scientists to increasingly exploit silver nanoparticles (AgNPs) (also known as nanosilver) as an antimicrobial agent (17, 19) (e.g., in wound dressings (namely, Acticoat-7, Actisorb Silver 220, Aquacel-Ag hydrofiber and Silverlon (14)) catheters (20) and various household products), and for the treatment of retinal neurovascularisation (21) and of acquired immunodeficiency syndrome (22). Nanosilver has also been shown to improve the anti-thrombogenic surface properties of nanocomposite polymers for bypass grafts (23), providing a synergistic approach where the release of nitric oxide (24) (responsible of maintaining the haemostasis between pro-thrombotic and anti-thrombogenic states of the endothelium lining the vessels (25)) is associated with the release of Ag+ ions, thus reducing the onset of cardiovascular implant-associated infection (26). In addition, silver nanowires (AgNWs) have very high electric conductivity (27), meaning that they can transfer electricity efficiently when incorporated in elastic conductors (28), and therefore, similarly to gold nanowires (29), they might find application in the production of cardiac patches. Preclinical studies have also been carried out to evaluate the use of silver nanomaterials for cancer treatment (30–32) and/or diagnosis (33).

Despite decades of use, the toxicity of silver is still not clear. In small concentrations colloidal silver is considered non-toxic to human cells (34), even if topical and systemic side effects (such as argyria (35) and liver injury (36), respectively) have been associated with long-term exposure to silver-containing products. In parallel, results concerning the toxicity of silver nanomaterials are often inconclusive (37). Recent studies show that, in vitro, nanosilver leads to oxidative stress, lipid peroxidation, inhibition of mitochondrial activity, DNA damage and apoptotic cell death (38–48). Nanosilver has also been reported to cause hepatotoxicity (49–51), while the cytotoxicity of AgNWs is dependent on cell type, nanowire lengths, doses and incubation times (52). The proposed mechanism of action is that silver nanomaterials release Ag+ ions (53), which interact with the thiol groups of proteins in the cell cytoplasm and the inner mitochondrial membrane, interrupting and/or perturbing the normal cells activity (54, 55). However, a number of recent studies have indicated that the transformation of silver nanoparticles to (Ag2S)NPs reduces the adverse effects of these materials (56–60). Researchers have also provided evidence that in lung epithelial cells the Ag+ ions released from AgNWs precipitate in the form of insoluble metallic Ag2S, thus limiting the toxic impact of AgNWs. In addition, it is known that fibre length is an important factor in determining the cytotoxic (52) and inflammatory responses (53, 61) to AgNWs, and to HARNs more in general (62). Recent studies envisage in fact that a higher inflammatory state is associated with increasing AgNWs lengths (53, 61): this is known as the “fibre pathogenicity paradigm” (43). In vivo studies have reported frustrated macrophage uptake of AgNWs that are longer than 10 μm (61, 63); this phenomenon has been identified as an important factor in the post-exposure inflammatory response and associated asbestos-like lung pathogenicity of such nanomaterial (53, 62, 63).

In light of this diverse evidence, this study aimed to investigate the in vitro internalisation, cytotoxicity and the influence on the cell cycle of commercially-available AgNWs longer than 10 μm which were incubated with lung adenocarcinoma (A549) cells and normal lung fibroblasts (MRC-5 cells). The A549 human lung carcinoma cell line is considered one of the closest cell models mimicking alveolar epithelial type II cells (e.g., membrane-bound inclusions resemble lamellar bodies of type II cells) (64), and it has proven to be a robust cell line/alveolar model for several previous nanomaterial-based studies (52, 65–68). In addition, A549 cells are a physiologically relevant in vitro model of non-small cell lung cancer (NSCLC) (69), the most prevalent form of lung cancer originating from epithelial cells. Therefore, this cell line was chosen as an appropriate epithelial alveolar model (70) for testing AgNWs internalisation and cytotoxicity in lung cancer tissue. Conversely, the MRC-5 human fibroblast cell line was selected as a model of normal/non-cancerous lung cell type, in order to investigate the cytotoxicity of AgNWs when coming into contact with the healthy cells surrounding the malignantly transformed tissue areas in the lung.

In both cell lines tested, our data clearly demonstrated that AgNWs could penetrate and localise within the cells without causing any acute and/or detectable cytotoxicity (up to 72 h exposure) or any significant change in the cell cycle. From these observations, we conclude that AgNWs could be used as suitable candidates for subsequent functionalisation, drug delivery and diagnostic purposes (such as lung cancer treatment) with several advantages over conventional therapies.

Materials and methods

Chemicals and solvents were purchased from commercial sources [Sigma-Aldrich (Ireland), Fisher Scientific (Ireland), Invitrogen Bioscience (Ireland) and Calbiochem (USA)] and used as supplied unless differently stated in the text.

Silver nanowires (AgNWs)

AgNWs (high aspect-ratio nanostructures, HARNs) were prepared and supplied as from previous publication (53) by Seashell Technology Ltd (CA, USA). The polyol process used for the synthesis of the AgNW introduced an organic protective layer of poly(vinyl)pyrrolidone (PVP) which has been reported to be non-toxic. Prior to use, AgNWs underwent rigorous testing of their physical (morphological) characteristics. AgNWs were characterised in their geometrical size: length=17.0±3.0 μm; diameter=0.11±0.01 μm. Characterised AgNWs were suspended in sterile PBS at a concentration of 40 μg/mL (stock solution) and then diluted in cell culture media at working concentrations of 1, 5 and 10 μg/mL. The concentrations were selected as equal (5 μg/mL), higher (10 μg/mL) or lower (1 μg/mL) than the dose administered in a previously published in vivo study (53) and as within the range of the doses tested in a previous in vitro study (52).

Cell culture

Human lung adenocarcinoma (A549) and human lung fibroblast (MRC-5) cell lines (both from ATCC, Manassas, VA, USA) were cultured in F12K medium (containing L-glutamine) and EMEM medium (GIBCO®, Life Technologies™), respectively. Cell media were supplemented with 10% (v/v) foetal bovine serum (FBS) and 1% (v/v) penicillin/streptomycin. Cells were grown in Nunc™ T75 tissue culture flasks (Thermo Fisher Scientific Inc., MA, USA) in a humidified incubator at 37°C in 5% CO2 until a 70%–80% confluence was reached. At 70%–80% confluence, cells were diluted in supplemented F12K or EMEM media at concentrations appropriate for the experimental procedure:

  • For internalisation studies, A549 and MRC-5 cells were seeded in 4-well chambered Millicell EZ slides (Millipore, Invitrogen Ireland) at concentration of 106 cells/well (final volume: 500 μL/well).

  • For exposure to AgNWs for cytotoxicity and cell cycle studies, A549 and MRC-5 cells were seeded in 96-well plate (5000 cells/well).

Before exposure to AgNWs, cells were incubated at 37°C and 5% CO2 for 24 h to allow complete cell adhesion to the substrates. Cell adhesion was ascertained by microscopy inspection of the chambers or well plates.

Cellular internalisation of AgNWs

Cell cultures of A549 and MRC-5 cells were exposed to AgNWs (1, 5 and 10 μg/mL) for 24 h, then washed with PBS and fixed with 4% paraformaldehyde (PFA) for 10 min at room temperature. Cells were stained with rhodamine-phalloidin (1:40 dilution) (Molecular Probes Inc., Invitrogen, USA) to detect F-actin, and Hoechst (1:400 dilution) as nuclear counterstain. Cells were incubated for 20 min at room temperature in the dark. Specimens were imaged by a ZEISS 510 Meta confocal microscope (Carl Zeiss, Germany) equipped with Zeiss LSM 5 software. AgNWs were imaged in reflectance mode at λexc=561 nm and visualised by using pseudo-color (green). To elucidate the AgNWs internalisation, Z-stack images were recorded with a 63× oil immersion objective lens for best imaging.

Cytotoxicity of AgNWs

A549 and MRC-5 cells were exposed to AgNWs at concentrations of 1, 5 and 10 μg/mL for 24 and 72 h. Negative (untreated cells) and positive (cells treated with 70% methanol for 30 min at 37°C) controls were included in all experimental runs. The changes in cell viability were assessed by Trypan Blue exclusion assay and Live/Dead viability assay.

  • Trypan Blue exclusion assay After 24 or 72 h exposure to AgNWs, unstained (living) and stained (dead) cells were counted using a haemocytometer cell counter and the percentage of live cells was calculated as for Eq. (1) Data are reported as average±standard deviation (ntest=2).

  • Live/Dead cell viability assay After 24 or 72 h exposure to AgNWs, cells were washed twice with sterile PBS and then stained with Calcein AM (λabs=494 nm) (1:200 dilution) and Ethidium homodimer-1 (EthD-1) (λabs=528 nm) (1:500 dilution) (45 min, room temperature). Auto-fluorescence background controls were prepared for each of the dyes used. Epifluorescence microscopy was used to visualise and compare the proportion of live and dead cells in the various samples. Cell viability after exposure was quantified by absorption spectroscopy (λ=494 and 528 nm) by an Epoch microplate reader (Biotek, USA), as follows:

    1. A(494)s and A(528)s are the absorption at 494 and 528 nm in the experimental cell samples, labelled with Calcein AM and EthD-1;

    2. A(494)0 and A(528)0 are the absorption at 494 and 528 nm in the cell-free samples labelled with only one fluorescent dye.

The percentage (%) of live cells was then calculated from the absorption readings as described in Eq. (2) The data are reported as average±standard deviation (ntest=3).

The absorption spectra of AgNWs (reported in Figure S1 in the Supporting Information) were measured at the three concentrations tested (1, 5 and 10 μg/mL) by an Epoch microplate reader (Biotek, USA). The small absorbance of AgNWs at λ=494 and 528 nm confirmed that such nanonamterial could not interfere with the Live/Dead cell viability assay used in this study to quantify the cell viability after exposure to AgNWs.

Influence of AgNWs on the cell cycle

Possible cell cycle effects caused by AgNWs exposure were evaluated using the BD Cycletest™ Plus DNA Reagent Kit (BD Biosciences, Oxford, UK). After 24 or 72 h exposure to AgNWs, cells were harvested using TrypLE™ Select (GIBCO®, USA) and centrifuged for 5 min at 1600 rpm and cell cycle assay was performed according to the manufacturer’s protocol. Stained cells were analysed using BD AccuriTM C6 flow cytometer (BD Biosciences, Oxford, UK). Briefly, the stained nuclei were visualised using the SSC-H vs FSC-H scatter plot and a gate was applied (P1) to exclude debris at lower scatter intensities. Aggregate exclusion gating (P2 in P1) via doublet discrimination was then performed on the P1 population using the FL2-H vs FL2-A scatter plot. A minimum of 10,000 events was collected in the (P2 in P1) gate and visualised on the FL2-H histogram. Analysis of cell cycle stage for G0/G1, S and G2/M phase was made by manual gating on the FL2-H histogram. Data are presented as % cell population in (P2 in P1) and expressed as average±standard deviation (ntest=2).

Statistical analysis

A two-way analysis of variance (ANOVA) followed by a Bonferroni post-test analysis was carried out (Prism, Graph-Pad Software Inc., USA). p<0.05 was considered statistically significant. A complete list of p values is reported in the Supporting Information (Tables S1–S3).

Results

Cellular internalisation of AgNWs

Confocal microscopy was used to identify whether AgNWs were internalised into A549 (Figure 1A–C) and MRC-5 cells (Figure 1D–F). After 24 h exposure, it resulted evident that AgNWs were internalised into both cell culture models at all concentrations tested.

Figure 1 Representative confocal images of (A–C) A549 and (D–F) MRC-5 cells exposed to AgNWs (1, 5 and 10 μg/mL) for 24 h. Cells were stained for F-actin (in red) and nuclei (in blue). AgNWs were imaged in confocal reflectance mode and are pseudo-colored in green. Arrows highlight the internalised AgNWs. Scale bars: 10 μm (63× oil-immersion objective lens).
Figure 1

Representative confocal images of (A–C) A549 and (D–F) MRC-5 cells exposed to AgNWs (1, 5 and 10 μg/mL) for 24 h. Cells were stained for F-actin (in red) and nuclei (in blue). AgNWs were imaged in confocal reflectance mode and are pseudo-colored in green. Arrows highlight the internalised AgNWs. Scale bars: 10 μm (63× oil-immersion objective lens).

Cytotoxicity of AgNWs

Trypan Blue exclusion assay

Trypan Blue exclusion assay was used to quantify A549 and MRC-5 cell viability changes (Figure 2A and B, respectively) to three concentrations of AgNWs (1, 5 and 10 μg/mL) at two time points 24 and 72 h. No significant change in cell viability was evidenced after 24 or 72 h exposure in all cell models tested and, as the concentration of AgNWs increased, the percentage of live cells did not show any unusual decrease.

Figure 2 Percentage (%) of live cells in (A) A549 and (B) MRC-5 cells untreated (negative control), exposed to 70% methanol for 30 min (positive control) and to AgNWs (1, 5 and 10 μg/mL) for 24 or 72 h, as calculated by Trypan Blue exclusion assay. The symbol (***) indicates significant changes (p<0.001) as compared to the negative controls.
Figure 2

Percentage (%) of live cells in (A) A549 and (B) MRC-5 cells untreated (negative control), exposed to 70% methanol for 30 min (positive control) and to AgNWs (1, 5 and 10 μg/mL) for 24 or 72 h, as calculated by Trypan Blue exclusion assay. The symbol (***) indicates significant changes (p<0.001) as compared to the negative controls.

Live/Dead viability assay

Live/Dead cytotoxicity assay was performed in order to confirm the data obtained by the Trypan Blue exclusion assay. Epifluorescence microscopy (Figure 3A) and quantitative (Figure 3B–C) analysis confirmed that AgNWs did not cause any cytotoxic response in A549 cells or MRC-5 cells after 24 and 72 h exposure.

Figure 3 (A) Representative epifluorescence images of A549 cells untreated (negative control), treated with 70% methanol for 30 min (positive control), and exposed to AgNWs for 24 h. Cells were stained with Calcein AM (live cells, in green) and EthD-1 (dead cells, in red). Scale bars: 20 μm (10× objective lens). (B-C) Percentage (%) of live cells as quantified by Live/Dead cytotoxicity assay after exposing (B) A549 and (C) MRC-5 cells to AgNWs. The symbols (**) and (***) indicate significant changes (p<0.01 and p<0.001, respectively) as compared to the negative controls.
Figure 3

(A) Representative epifluorescence images of A549 cells untreated (negative control), treated with 70% methanol for 30 min (positive control), and exposed to AgNWs for 24 h. Cells were stained with Calcein AM (live cells, in green) and EthD-1 (dead cells, in red). Scale bars: 20 μm (10× objective lens). (B-C) Percentage (%) of live cells as quantified by Live/Dead cytotoxicity assay after exposing (B) A549 and (C) MRC-5 cells to AgNWs. The symbols (**) and (***) indicate significant changes (p<0.01 and p<0.001, respectively) as compared to the negative controls.

Influence of AgNWs on the cell cycle

Flow cytometry was used to evaluate the AgNWs interactions with the cell cycle in A549 and MRC-5cells exposed to such nanomaterial. Exposure of A549 cells to AgNWs for 24 h caused an increase in the cell population in G0/G1 phase at all the concentrations tested (Figure 4A). This was correlated with a decreased number of cells in G2/M phase. A similar trend was found for MRC-5 cells exposed to AgNWs for 24 h (Figure 4C). In addition, after 72 h the increase in A549 cell population in G0/G1 phase was associated with a moderate but statistically significant increased number of cells entering G2/M phase (Figure 4B). In contrast, MRC-5 cells demonstrated a rise in the G0/G1 population only after 72 h (Figure 4D).

Figure 4 Effects of AgNWs on the cell cycle (A–B) A549 and (C–D) MRC-5 cells after (A, C) 24 or (B, D) 72 h exposure. The symbols (*), (**) and (***) indicate significant changes (p<0.05, p<0.01 and p<0.001, respectively) compared to the negative controls.
Figure 4

Effects of AgNWs on the cell cycle (A–B) A549 and (C–D) MRC-5 cells after (A, C) 24 or (B, D) 72 h exposure. The symbols (*), (**) and (***) indicate significant changes (p<0.05, p<0.01 and p<0.001, respectively) compared to the negative controls.

Discussion

Analysis of the biological interactions of AgNWs with lung epithelial (A549) cells was carried out with the aim to assess (1) the capability of AgNWs as future drug delivery platforms to be internalised into lung cancer cells, and (2) to evaluate the biocompatibility of AgNWs in the lungs. Cell cultures of lung fibroblasts (MRC-5 cells) were also tested, in order to investigate the cytotoxicity of AgNWs coming into contact with non-cancerous tissue.

Confocal reflectance microscopy imaging clearly demonstrated that AgNWs internalisation was effectively achieved after 24 h exposure at all three concentrations tested (Figure 1). It has been observed that the cell type is one of the key factors that influence the mechanism of cellular entry of silver nanomaterials (37). In this study, it was not clear whether the cellular entry of AgNWs occurred through phagocytosis or some other mechanism (such as autophagy (52) in human phagocytic cells) and whether AgNWs entered into the various cell types through different mechanisms. Further studies are therefore needed to elucidate the mechanism of cellular internalisation of AgNWs in the various cell types and to identify the precise localisation of the internalised AgNWs in specific cellular compartments. Similarly to previously reported data on A549 cells’ responses to AgNWs (52), Live/Dead viability assay showed that AgNWs did not cause any detectable cytotoxicity when incubated with A549 and MRC-5 cells for up to 72 h (Figure 3). Likewise, the Trypan Blue exclusion assay (Figure 2) suggested that AgNWs did not cause any detectable cytotoxicity, thus supporting the idea that such nanomaterial could be used as a safe drug delivery platform following ad hoc engineered functionalisation. Our investigation focused on the cells response after 24 and 72 h due to the fact that AgNWs identical to those tested in this study have been reported to be subject to dissolution over longer periods (53). In particular, the authors would like to highlight that the AgNWs used in this study are reported not be bio-persistent beyond a few days (53); this partly reduces the relevance of the “fibre pathogenicity paradigm” for the nanomaterial tested. Bio-persistence is in fact one of three key factors in determining the pathogenic effects of long nanofibres (71) and HARNs (62), which are known to evoke chronic inflammatory responses and cancer development when resistant to dissolution and/or breakage in the biological environment. In addition, it has been recently reported that the length threshold for fibre pathogenic effects is a higher value in the lung than at the mesothelial surface, the cut-off length for AgNWs inducing acute pulmonary inflammation being 14 μm (63). This, together with our results, supports the concept that the AgNWs tested in this study could be translated in carriers for functionalisation and drug delivery purposes. Additional research is however needed to evaluate if, similarly to nanosilver (51), AgNWs could cause hepatotoxicity following inhalation or injection. A further issue is the formation of different silver species over time via AgNWs dissolution. The AgNWs used in this study do in fact slowly dissolve (53) and, depending on the biological environment, the resulting Ag+ ions may form soluble complexes such as AgCl2-, AgCl32-, [Ag(NH3)2]+, covalent adducts with thiol group containing compounds (e.g., glutathione, cysteine, proteins) or insoluble salts (AgCl, Ag2S) (72). Furthermore, Ag+ could be reduced to Ag0 (e.g., by glucose), thus leading to the de novo formation of Ag0 nanoparticulates. It becomes therefore crucial to fully characterise the physico-chemical properties of AgNWs and the biological effects of their products of conversion at the various timepoints as we provided in this work.

Finally, while AgNWs did not have strong influence on the cell cycle of MRC-5 cells following 72 h exposure (Figure 4D), significant changes in the percentage of cell population in the various phases were detected when A549 cells were exposed to AgNWs for 24 h and 72 h (Figure 4A–C). This indicates that AgNWs had very diverse effects on cells with different proliferation rates, with higher effect on the fast-growing, carcinogenic epithelial lineage. On the other hand, it is known that cells with damaged DNA accumulate in the G1 phase, the DNA synthesis (S) phase or the G2/M phase (73). Therefore, the increased A549 cell population in G1 and G2/M phases after 24 h or 72 h, respectively, indicated a DNA damage triggered by AgNWs exposure, which might ultimately cause genotoxicity in the chronic phase similarly to what has been demonstrated for nanosilver in various in vitro and in vivo studies (73). Further investigation is needed to better elucidate the cell line-dependent differences and understand whether the effects detected in A549 cells could translate into genotoxicity over longer exposure times.

In conclusion, our data showed that AgNWs could penetrate and localise within A549 and MRC-5 cells, without causing any detectable change in cell viability. Further investigations into the systemic side effects and bio-distribution of this nanomaterial are required, as well as to demonstrate that, by selective functionalisation with biologically active moieties, AgNWs could be used as platforms for drug delivery against lung cancer.


Corresponding author: Adriele Prina-Mello, School of Medicine, Trinity College Dublin, Ireland; and Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Ireland, E-mail:

About the authors

Manisha Singh

Manisha Singh was awarded a Masters in Molecular Medicine from Trinity College Dublin (Ireland) in 2013. Her thesis focused on an in vitro nanomedicine study of nanoparticles interaction with lung epithelial cells that included a comparative investigation in 2D and 3D cell culture models. In 2010, she graduated in Dental Surgery (BDS) at Rama Dental College (India). Her primary research interest includes nanomedicine, cell cultures and dental science.

Dania Movia

Dania Movia is a post-doctoral researcher at the Centre for Research on Advanced and Adaptive Nanostructures (CRANN)/School of Medicine of Trinity College Dublin (Ireland). In 2007 she was awarded a BSc in Medicinal Chemistry at University of Trieste (Italy). In 2011 she completed her PhD in Chemistry at Trinity College Dublin with a thesis entitled “Single-walled carbon nanotubes as novel NIR fluorescent probes for biomedical optical imaging”.

Omar K. Mahfoud

Omar K. Mahfoud is a PhD candidate in the Nanomedicine and Molecular Imaging Group at Trinity College Dublin (Ireland), where he is involved in an EU FP7-funded project (NAMDIATREAM) aimed at developing the next generation of nanomaterial-based diagnostic systems for the advanced detection of breast, lung and prostate cancer biomarkers. In 2008, he was awarded a BSc in Neuroscience at Trinity College Dublin and he completed his MSc in Molecular Medicine at the University Medical Centre Goettingen (Germany).

Yuri Volkov

Yuri Volkov received his MD from the Moscow Medical University and subsequently a PhD in biomedical sciences, Institute of Immunology, Moscow. He is a Professor at the Department of Clinical Medicine and the Director of Research of the School of Medicine at Trinity College Dublin. Prof. Volkov coordinates a large scale EU FP-7 NAMDIATREAM with 22 European academic, research, clinical and industrial partners for early diagnostics and monitoring of malignant diseases. Lead TCD partner for EU FP-7 MULTIFUN and Principal investigator in many grants. Prof. Volkov published more than 80 articles, several patents and book chapters.

Adriele Prina-Mello

Adriele Prina-Mello is a CRANN Investigator, a Senior Research Fellow of the School of Medicine and a part-time lecture at Trinity College Dublin (Ireland), a Nanosafety Cluster member and the vice-chair of the Nanodiagnostic working group of the European Technology Platform of Nanomedicine. Dr Prina-Mello is involved in developing and advancing several multidisciplinary research projects between University, Research Hospital and Industry partners for future applications in medicine and nanotechnology industry. Currently he is involved in 5 EU FP7 funded projects: NAMDIATREAM, MULTIFUN, QNANO, NANoREG, and AMCARE. Dr Prina-Mello has published more than 45 articles in biomedicine, nanotechnology, nanotoxicology and nanomedicine research area.

The authors would like to thank Kieran Crosbie-Staunton for the technical support during the flow cytometry experiment series. This work was partially supported by the EU FP7 NANoREG project (NMP.20121.3-3), EU FP7 NAMDIATREAM project (NMP-2009-LARGE-3-246479) and the MSc Molecular Medicine programme of Trinity College Dublin, Ireland.

References

1. Lung Cancer 2011. Available from: http://www.ncri.ie/pubs/pubfiles/LungCancer2011.pdf.Search in Google Scholar

2. Huber RM, Stratakis DF. Molecular oncology–perspectives in lung cancer. Lung cancer (Amsterdam, Netherlands) 2004;45(Suppl 2): S209–13.10.1016/j.lungcan.2004.07.973Search in Google Scholar

3. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA: A Cancer Journal for Clinicians 2013;63:11–30.10.3322/caac.21166Search in Google Scholar

4. Non-Small Cell Lung Cancer Collaborative Group. Chemotherapy for non-small cell lung cancer. Cochrane Database of Systematic Reviews 2000, Issue 2. Art. No.: CD002139. DOI: 10.1002/14651858.CD002139.10.1002/14651858.CD002139Search in Google Scholar

5. Pao W, Girard N. New driver mutations in non-small-cell lung cancer. Lancet Oncol 2011;12:175–80.10.1016/S1470-2045(10)70087-5Search in Google Scholar

6. Kumar M, Nagpal R, Hemalatha R, Verma V, Kumar A, Singh S, et al. Targeted cancer therapies: the future of cancer treatment. Acta Bio-medica: Atenei Parmensis 2012;83:220–33.Search in Google Scholar

7. General Information About Non-Small Cell Lung Cancer. Available from: http://www.cancer.gov/cancertopics/pdq/treatment/non-small-cell-lung/Patient/page1.Search in Google Scholar

8. Reck M, Heigener DF, Mok T, Soria JC, Rabe KF. Management of non-small-cell lung cancer: recent developments. Lancet 2013;382:709–19.10.1016/S0140-6736(13)61502-0Search in Google Scholar

9. Da Silva AL, Santos RS, Xisto DG, Alonso Sdel V, Morales MM, Rocco PR. Nanoparticle-based therapy for respiratory diseases. Anais da Academia Brasileira de Ciencias 2013;85:137–46.10.1590/S0001-37652013005000018Search in Google Scholar PubMed

10. Kurmi BD, Kayat J, Gajbhiye V, Tekade RK, Jain NK. Micro- and nanocarrier-mediated lung targeting. Expert Opin Drug Deliver 2010;7:781–94.10.1517/17425247.2010.492212Search in Google Scholar PubMed

11. Zarogoulidis P, Chatzaki E, Porpodis K, Domvri K, Hohenforst-Schmidt W, Goldberg EP, et al. Inhaled chemotherapy in lung cancer: future concept of nanomedicine. Int J Nanomedicine 2012;7:1551–72.10.2147/IJN.S29997Search in Google Scholar PubMed PubMed Central

12. Cuenca AG, Jiang H, Hochwald SN, Delano M, Cance WG, Grobmyer SR. Emerging implications of nanotechnology on cancer diagnostics and therapeutics. Cancer 2006;107:459–66.10.1002/cncr.22035Search in Google Scholar PubMed

13. Ravindran A, Chandran P, Khan SS. Biofunctionalized silver nanoparticles: advances and prospects. Colloid Surface B, Biointerfaces 2013;105:342–52.10.1016/j.colsurfb.2012.07.036Search in Google Scholar PubMed

14. Bhattacharya R, Mukherjee P. Biological properties of “naked” metal nanoparticles. Adv Drug Deliver Rev 2008;60:1289–306.10.1016/j.addr.2008.03.013Search in Google Scholar PubMed

15. Liao H, Nehl CL, Hafner JH. Biomedical applications of plasmon resonant metal nanoparticles. Nanomedicine (London, England) 2006;1:201–8.10.2217/17435889.1.2.201Search in Google Scholar PubMed

16. Atiyeh BS, Costagliola M, Hayek SN, Dibo SA. Effect of silver on burn wound infection control and healing: review of the literature. Burns: J Int Soc Burn Injuries 2007;33:139–48.10.1016/j.burns.2006.06.010Search in Google Scholar PubMed

17. Arvizo RR, Bhattacharyya S, Kudgus RA, Giri K, Bhattacharya R, Mukherjee P. Intrinsic therapeutic applications of noble metal nanoparticles: past, present and future. Chem Soc Rev 2012;41:2943–70.10.1039/c2cs15355fSearch in Google Scholar PubMed PubMed Central

18. Volker C, Oetken M, Oehlmann J. The biological effects and possible modes of action of nanosilver. Reviews of environmental contamination and toxicology 2013;223:81–106.Search in Google Scholar

19. Prabhu S, Poulose E. Silver nanoparticles: mechanism of antimicrobial action, synthesis, medical applications, and toxicity effects. Int Nano Lett 2012;2:1–10. (English).10.1186/2228-5326-2-32Search in Google Scholar

20. Samuel U, Guggenbichler JP. Prevention of catheter-related infections: the potential of a new nano-silver impregnated catheter. Int J Antimicrob Ag 2004;23(Suppl 1):S75–8.10.1016/j.ijantimicag.2003.12.004Search in Google Scholar PubMed

21. Kalishwaralal K, Barathmanikanth S, Pandian SR, Deepak V, Gurunathan S. Silver nano-a trove for retinal therapies. J Control Release: Official J Control Release Society 2010;145:76–90.10.1016/j.jconrel.2010.03.022Search in Google Scholar PubMed

22. Lara HH, Ayala-Nunez NV, Ixtepan-Turrent L, Rodriguez-Padilla C. Mode of antiviral action of silver nanoparticles against HIV-1. J Nanobiotechnology 2010;8:1.10.1186/1477-3155-8-1Search in Google Scholar PubMed PubMed Central

23. de Mel A, Chaloupka K, Malam Y, Darbyshire A, Cousins B, Seifalian AM. A silver nanocomposite biomaterial for blood-contacting implants. J Biomed Mater Res Part A 2012;100:2348–57.10.1002/jbm.a.34177Search in Google Scholar PubMed

24. Naghavi N, de Mel A, Alavijeh OS, Cousins BG, Seifalian AM. Nitric oxide donors for cardiovascular implant applications. Small (Weinheim an der Bergstrasse, Germany) 2013;9:22–35.10.1002/smll.201200458Search in Google Scholar PubMed

25. Sessa WC. Molecular control of blood flow and angiogenesis: role of nitric oxide. J Thromb Haemostasis 2009;7(Suppl 1):35–7.10.1111/j.1538-7836.2009.03424.xSearch in Google Scholar

26. Halpenny GM, Gandhi KR, Mascharak PK. Eradication of pathogenic bacteria by remote delivery of nitric oxide via light-triggering of nitrosyl-containing materials. ACS Med Chem Lett 2010;1:180–3.10.1021/ml1000646Search in Google Scholar

27. Hecht DS, Hu L, Irvin G. Emerging transparent electrodes based on thin films of carbon nanotubes, graphene, and metallic nanostructures. Adv Mater (Deerfield Beach, Fla) 2011;23: 1482–513.10.1002/adma.201003188Search in Google Scholar

28. Xu F, Zhu Y. Highly conductive and stretchable silver nanowire conductors. Adv Mater (Deerfield Beach, Fla) 2012;24:5117–22.10.1002/adma.201201886Search in Google Scholar

29. Dvir T, Timko BP, Brigham MD, Naik SR, Karajanagi SS, Levy O, et al. Nanowired three-dimensional cardiac patches. Nature Nanotechnology 2011;6:720–5.10.1038/nnano.2011.160Search in Google Scholar

30. Govender R, Phulukdaree A, Gengan RM, Anand K, Chuturgoon AA. Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. J Nanobiotechnology 2013;11:5.10.1186/1477-3155-11-5Search in Google Scholar

31. Dreaden EC, El-Sayed MA. Detecting and destroying cancer cells in more than one way with noble metals and different confinement properties on the nanoscale. Accounts Chem Res 2012;45:1854–65.10.1021/ar2003122Search in Google Scholar

32. Jeyaraj M, Sathishkumar G, Sivanandhan G, MubarakAli D, Rajesh M, Arun R, et al. Biogenic silver nanoparticles for cancer treatment: an experimental report. Colloid Surface B, Biointerfaces 2013;106:86–92.10.1016/j.colsurfb.2013.01.027Search in Google Scholar

33. Homan KA, Souza M, Truby R, Luke GP, Green C, Vreeland E, et al. Silver nanoplate contrast agents for in vivo molecular photoacoustic imaging. ACS Nano 2012;6:641–50.10.1021/nn204100nSearch in Google Scholar

34. Silver S. Bacterial silver resistance: molecular biology and uses and misuses of silver compounds. FEMS Microbiol Rev 2003;27:341–53.10.1016/S0168-6445(03)00047-0Search in Google Scholar

35. Payne CM, Bladin C, Colchester AC, Bland J, Lapworth R, Lane D. Argyria from excessive use of topical silver sulphadiazine. Lancet 1992;340:126.10.1016/0140-6736(92)90458-FSearch in Google Scholar

36. Trop M, Novak M, Rodl S, Hellbom B, Kroell W, Goessler W. Silver-coated dressing acticoat caused raised liver enzymes and argyria-like symptoms in burn patient. J Trauma 2006;60:648–52.10.1097/01.ta.0000208126.22089.b6Search in Google Scholar PubMed

37. Stensberg MC, Wei Q, McLamore ES, Porterfield DM, Wei A, Sepulveda MS. Toxicological studies on silver nanoparticles: challenges and opportunities in assessment, monitoring and imaging. Nanomedicine (London, England) 2011;6:879–98.10.2217/nnm.11.78Search in Google Scholar PubMed PubMed Central

38. Foldbjerg R, Dang DA, Autrup H. Cytotoxicity and genotoxicity of silver nanoparticles in the human lung cancer cell line, A549. Arch Toxicol 2011;85:743–50.10.1007/s00204-010-0545-5Search in Google Scholar PubMed

39. Park MV, Neigh AM, Vermeulen JP, de la Fonteyne LJ, Verharen HW, Briede JJ, et al. The effect of particle size on the cytotoxicity, inflammation, developmental toxicity and genotoxicity of silver nanoparticles. Biomaterials 2011;32:9810–7.10.1016/j.biomaterials.2011.08.085Search in Google Scholar PubMed

40. Costa CS, Ronconi JV, Daufenbach JF, Goncalves CL, Rezin GT, Streck EL, et al. In vitro effects of silver nanoparticles on the mitochondrial respiratory chain. Mol Cell Biochem 2010;342:51–6.10.1007/s11010-010-0467-9Search in Google Scholar PubMed

41. Suresh AK, Pelletier DA, Wang W, Morrell-Falvey JL, Gu B, Doktycz MJ. Cytotoxicity induced by engineered silver nanocrystallites is dependent on surface coatings and cell types. Langmuir: ACS J Surf Colloid 2012;28:2727–35.10.1021/la2042058Search in Google Scholar PubMed

42. Kim HR, Kim MJ, Lee SY, Oh SM, Chung KH. Genotoxic effects of silver nanoparticles stimulated by oxidative stress in human normal bronchial epithelial (BEAS-2B) cells. Mutat Res 2011;726:129–35.10.1016/j.mrgentox.2011.08.008Search in Google Scholar PubMed

43. Stoehr LC, Gonzalez E, Stampfl A, Casals E, Duschl A, Puntes V, et al. Shape matters: effects of silver nanospheres and wires on human alveolar epithelial cells. Part Fibre Toxicol 2011;8:36.10.1186/1743-8977-8-36Search in Google Scholar PubMed PubMed Central

44. Arora S, Jain J, Rajwade JM, Paknikar KM. Cellular responses induced by silver nanoparticles: in vitro studies. Toxicol Lett 2008;179:93–100.10.1016/j.toxlet.2008.04.009Search in Google Scholar PubMed

45. Piao MJ, Kang KA, Lee IK, Kim HS, Kim S, Choi JY, et al. Silver nanoparticles induce oxidative cell damage in human liver cells through inhibition of reduced glutathione and induction of mitochondria-involved apoptosis. Toxicol Lett 2011;201: 92–100.10.1016/j.toxlet.2010.12.010Search in Google Scholar PubMed

46. AshaRani PV, Low Kah Mun G, Hande MP, Valiyaveettil S. Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano 2009;3:279–90.10.1021/nn800596wSearch in Google Scholar PubMed

47. Perry MM, Hui CK, Whiteman M, Wood ME, Adcock I, Kirkham P, et al. Hydrogen sulfide inhibits proliferation and release of IL-8 from human airway smooth muscle cells. Am J Resp Cell Mol 2011;45:746–52.10.1165/rcmb.2010-0304OCSearch in Google Scholar PubMed PubMed Central

48. Xiu ZM, Zhang QB, Puppala HL, Colvin VL, Alvarez PJ. Negligible particle-specific antibacterial activity of silver nanoparticles. Nano Lett 2012;12:4271–5.10.1021/nl301934wSearch in Google Scholar PubMed

49. Kermanizadeh A, Gaiser BK, Hutchison GR, Stone V. An in vitro liver model–assessing oxidative stress and genotoxicity following exposure of hepatocytes to a panel of engineered nanomaterials. Part Fibre Toxicol 2012;9:28.10.1186/1743-8977-9-28Search in Google Scholar PubMed PubMed Central

50. Kermanizadeh A, Pojana G, Gaiser BK, Birkedal R, Bilanicova D, Wallin H, et al. In vitro assessment of engineered nanomaterials using a hepatocyte cell line: cytotoxicity, pro-inflammatory cytokines and functional markers. Nanotoxicology 2013;7: 301–13.10.3109/17435390.2011.653416Search in Google Scholar PubMed

51. Kermanizadeh A, Brown DM, Hutchison GR, Stone V. Engineered nanomaterial impact in the liver following exposure via an intravenous route–the role of polymorphonuclear leukocytes and gene expression in the organ. J Nanomed Nanotechol 2013;4:157.Search in Google Scholar

52. Verma NK, Conroy J, Lyons PE, Coleman J, O’Sullivan MP, Kornfeld H, et al. Autophagy induction by silver nanowires: a new aspect in the biocompatibility assessment of nanocomposite thin films. Toxicol Appl Pharmacol 2012;264:451–61.10.1016/j.taap.2012.08.023Search in Google Scholar PubMed

53. Schinwald A, Murphy FA, Prina-Mello A, Poland CA, Byrne F, Movia D, et al. The threshold length for fiber-induced acute pleural inflammation: shedding light on the early events in asbestos-induced mesothelioma. Toxicol Sci: An Off J Soc Toxicol 2012;128:461–70.10.1093/toxsci/kfs171Search in Google Scholar PubMed

54. Nowack B, Krug HF, Height M. 120 years of nanosilver history: implications for policy makers. Environ Sci Tech 2011;45: 1177–83.10.1021/es103316qSearch in Google Scholar PubMed

55. Schafer B, Brocke JV, Epp A, Gotz M, Herzberg F, Kneuer C, et al. State of the art in human risk assessment of silver compounds in consumer products: a conference report on silver and nanosilver held at the BfR in 2012. Arch Toxicol 2013: DOI: 10.1007/s00204-013-1083-8.10.1007/s00204-013-1083-8Search in Google Scholar PubMed PubMed Central

56. Kim B, Park CS, Murayama M, Hochella MF. Discovery and characterization of silver sulfide nanoparticles in final sewage sludge products. Environ Sci Tech 2010;44:7509–14.10.1021/es101565jSearch in Google Scholar

57. Nowack B. Chemistry. Nanosilver revisited downstream. Science (New York, NY) 2010;330:1054–5.10.1126/science.1198074Search in Google Scholar

58. Liu J, Pennell KG, Hurt RH. Kinetics and mechanisms of nanosilver oxysulfidation. Environ Sci Tech 2011;45:7345–53.10.1021/es201539sSearch in Google Scholar

59. Levard C, Reinsch BC, Michel FM, Oumahi C, Lowry GV, Brown GE. Sulfidation processes of PVP-coated silver nanoparticles in aqueous solution: impact on dissolution rate. Environ Sci Tech 2011;45:5260–6.10.1021/es2007758Search in Google Scholar

60. Levard C, Hotze EM, Lowry GV, Brown GE, Jr. Environmental transformations of silver nanoparticles: impact on stability and toxicity. Environ Sci Tech 2012;46:6900–14.10.1021/es2037405Search in Google Scholar

61. Schinwald A, Donaldson K. Use of back-scatter electron signals to visualise cell/nanowires interactions in vitro and in vivo; frustrated phagocytosis of long fibres in macrophages and compartmentalisation in mesothelial cells in vivo. Part Fibre Toxicol 2012;9:34.10.1186/1743-8977-9-34Search in Google Scholar

62. Donaldson K, Murphy F, Schinwald A, Duffin R, Poland CA. Identifying the pulmonary hazard of high aspect ratio nanoparticles to enable their safety-by-design. Nanomedicine (London, England) 2011;6:143–56.10.2217/nnm.10.139Search in Google Scholar

63. Schinwald A, Chernova T, Donaldson K. Use of silver nanowires to determine thresholds for fibre length-dependent pulmonary inflammation and inhibition of macrophage migration in vitro. Part Fibre Toxicol 2012;9:47.10.1186/1743-8977-9-47Search in Google Scholar

64. Shapiro DL, Nardone LL, Rooney SA, Motoyama EK, Munoz JL. Phospholipid biosynthesis and secretion by a cell line (A549) which resembles type II aleveolar epithelial cells. Biochimica et biophysica acta 1978;530:197–207.10.1016/0005-2760(78)90005-XSearch in Google Scholar

65. Mohamed BM, Movia D, Knyazev A, Langevin D, Davies AM, Prina-Mello A, et al. Citrullination as early-stage indicator of cell response to single-walled carbon nanotubes. Sci Rep 2013;3:1124.10.1038/srep01124Search in Google Scholar PubMed PubMed Central

66. Mohamed BM, Verma NK, Davies AM, McGowan A, Crosbie-Staunton K, Prina-Mello A, et al. Citrullination of proteins: a common post-translational modification pathway induced by different nanoparticles in vitro and in vivo. Nanomedicine (London, England) 2012;7:1181–95.10.2217/nnm.11.177Search in Google Scholar PubMed PubMed Central

67. Mohamed BM, Verma NK, Prina-Mello A, Williams Y, Davies AM, Bakos G, et al. Activation of stress-related signalling pathway in human cells upon SiO2 nanoparticles exposure as an early indicator of cytotoxicity. J Nanobiotechnology 2011;9:29.10.1186/1477-3155-9-29Search in Google Scholar PubMed PubMed Central

68. Verma NK, Crosbie-Staunton K, Satti A, Gallagher S, Ryan KB, Doody T, et al. Magnetic core-shell nanoparticles for drug delivery by nebulization. J Nanobiotechnology 2013;11:1.10.1186/1477-3155-11-1Search in Google Scholar PubMed PubMed Central

69. Caino MC, Lopez-Haber C, Kissil JL, Kazanietz MG. Non-small cell lung carcinoma cell motility, rac activation and metastatic dissemination are mediated by protein kinase C epsilon. PLoS One 2012;7:e31714.10.1371/journal.pone.0031714Search in Google Scholar PubMed PubMed Central

70. Rothen-Rutishauser B, Blank F, Muhlfeld C, Gehr P. In vitro models of the human epithelial airway barrier to study the toxic potential of particulate matter. Expert Opin Drug Metab Toxicol 2008;4:1075–89.10.1517/17425255.4.8.1075Search in Google Scholar PubMed

71. Donaldson K. The inhalation toxicology of p-aramid fibrils. Cr Rev Toxicol 2009;39:487–500.10.1080/10408440902911861Search in Google Scholar PubMed

72. Liu J, Sonshine DA, Shervani S, Hurt RH. Controlled release of biologically active silver from nanosilver surfaces. ACS Nano 2010;4:6903–13.10.1021/nn102272nSearch in Google Scholar PubMed PubMed Central

73. Kim S, Ryu DY. Silver nanoparticle-induced oxidative stress, genotoxicity and apoptosis in cultured cells and animal tissues. J Appl Toxicol 2013;33:78–89.10.1002/jat.2792Search in Google Scholar PubMed

Received: 2013-9-30
Accepted: 2013-10-22
Published Online: 2013-12-02
Published in Print: 2013-12-01

©2013 by Walter de Gruyter Berlin Boston

Downloaded on 22.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/ejnm-2013-0024/html
Scroll to top button