You are on page 1of 8

Ecotoxicology and Environmental Safety 198 (2020) 110674

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


journal homepage: www.elsevier.com/locate/ecoenv

Silver nanoparticles induce size-dependent and particle-specific T


neurotoxicity to primary cultures of rat cerebral cortical neurons
Bingjie Zhanga,b, Na Liua,c, Qian S. Liua, Jianqing Zhangd,∗∗, Qunfang Zhoua,b,e,∗, Guibin Jianga,b
a
State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085,
China
b
College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
c
School of Life Science, Shanxi University, Taiyuan, 030006, China
d
Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
e
School of Environment, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China

A R T I C LE I N FO A B S T R A C T

Keywords: Silver nanoparticles (AgNPs) are widely applied in many fields because of their excellent antibacterial activities.
Silver nanoparticles Toxicological studies have showed that AgNPs can cross the blood-brain barrier and exhibit high retention in the
Size-dependent effect brain. Therefore, the potential neurotoxicity of AgNPs is raising serious concerns. This study investigated the
Particle-specific effect neurotoxicological effects of AgNPs with two different sizes (20 nm and 70 nm, AgNPs-20 and AgNPs-70) using
Neurotoxicity
primary cultures of rat cerebral cortical neurons in mature and developing stages. The contribution of silver ion
Primary cultures of cerebral cortical neurons
release was investigated by testing the effects of ionic silver in parallel. The results showed that both AgNPs-20
and AgNPs-70 significantly decreased neuronal cell viability, and AgNPs-20 had stronger toxicity compared with
AgNPs-70. AgNP applications caused the granulated skeleton structure of the mature neurons with some broken
synapses after a 24-h exposure, and inhibited neuronal growth during a 7-day exposure. Intracellular silver
accumulation at non-cytotoxic exposure levels inhibited dopamine efflux, which was particle-specific and free of
released silver ions. The findings herein can aid in guiding the proper applications of AgNPs in different areas,
especially in medical use.

1. Introduction that the exposure of AgNPs inhibited locomotive activities, and caused
cerebellar ataxia-like symptoms in Sprague Dawley (SD) rats (Zhang
Silver nanoparticles (AgNPs) are a kind of metallic silver with at et al., 2013; Yin et al., 2015). Histopathological studies showed that
least one dimension at nano scale (< 100 nm) in three-dimensional AgNPs caused obvious distortions in both Purkinje and granular layers
space. These are widely used as antimicrobial agents in various com- of rat brains, and the granular layer was degenerated with loosened and
modities, such as wound dressings and antibacterial clothes. AgNPs can separated structure (Yin et al., 2015). AgNPs were also reported to in-
be released into the environment during the processes of manufacture, duce astrocytic proliferation, neuronal degeneration, and demyelina-
use, and disposal (McShan et al., 2014). They may migrate and trans- tion (Ahmed and Hussein, 2017).
form in the atmosphere, soil, surface water and ground water (Pachapur Although there are a large quantity of existing neurotoxicological
et al., 2016). The increase of AgNP release in the environment poses a studies of AgNPs based on in vivo experiments and in vitro immortalized
threat to wildlife and human health. A large number of studies have cell tests (Zhang et al., 2018), investigations into how AgNPs affect
shown that AgNPs can cause damage to organs, including the kidneys, primary cortical neurons remain limited. A previous study demon-
heart, and lungs (Kaewamatawong et al., 2014), and induce adverse strated a direct effect of AgNP upon neurotoxicity by showing that
effects on immune and reproductive systems (Ema et al., 2017). AgNPs caused the loss of cytoskeleton components and neurite out-
Recent studies have indicated that AgNPs can penetrate the blood- growth inhibition in premature cortical neurons (Xu et al., 2013).
brain barrier (Chen et al., 2016; Dan et al., 2018) and exert high re- Nevertheless, more investigations are warranted regarding the neuro-
tention in the brain (Wen et al., 2016). Neurobehavioral studies showed logical alterations of primary cultures of mature and developing


Corresponding author. State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy
of Sciences, Beijing, 100085, China.
∗∗
Corresponding author.
E-mail addresses: jianqingzh@szcdc.net (J. Zhang), zhouqf@rcees.ac.cn (Q. Zhou).

https://doi.org/10.1016/j.ecoenv.2020.110674
Received 19 July 2019; Received in revised form 20 April 2020; Accepted 21 April 2020
Available online 05 May 2020
0147-6513/ © 2020 Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

neuronal cells. 2.3. Primary cultures of cortical neurons


In addition to studies regarding the neurotoxic effects of AgNPs, it
remains unclear whether the mechanism occurs via particulate or ionic All experimental procedures involving animals were performed in
properties. Some studies reported that the toxicological effect of silver accordance with the protocols approved by the Committee of Scientific
ions (Ag+) on cell apoptosis was stronger than that of AgNPs (Hadrup Research in the Research Center for Eco-Environmental Sciences,
et al., 2012). Other studies showed that AgNPs induced cytotoxicity by Chinese Academy of Sciences. Pure cortical neurons were obtained from
a Trojan-horse type mechanism (Eun-Jung et al., 2010), which meant SD rat embryos on embryonic day 18 as previously reported (Hossain,
that AgNPs would further release silver ions to cause toxicity after 2016). Briefly, the embryonic cortices dissected from the pregnant fe-
entering cells. Nevertheless, several recent studies have indicated that male SD rats (Beijing Vital River Laboratory Animal Technology Co.,
AgNPs may have special particulate effects (Sun et al., 2016). The Ltd., China) were cut into small pieces and enzymatically digested.
characteristics of AgNPs, such as particle size, are also considered to be After dissociation and filtration, the single cell suspension was seeded
important factors in the regulation of their biological activities (Gliga in poly-L-lysine-coated plates and cultured in plating medium at 37°C
et al., 2014). Therefore, it would be important to demonstrate how with 5% CO2 for 24 h. Then, the cells were incubated in neurobasal
AgNPs of different sizes induce neurotoxic effects on primary neuronal medium supplemented with 0.25% GlutaMAX and 2% B27, and 2.5 μM
cells. cytosine arabinoside (Sha et al., 2019), a mitotic inhibitor, was used on
In the present study, AgNPs with different particle sizes (20 nm and the second day to eliminate glial cells. The primary cultures of cortical
70 nm, AgNPs-20 and AgNPs-70) and silver ion were applied to cultures neurons were maintained with half of the culture medium renewed
of primary rat cerebral cortical neurons at early development and every 4 days afterward.
mature stages in order to evaluate cell viability, neuron-specific skeletal
protein expression, intracellular silver accumulation, and neuro-
transmitter secretion. The findings of the present study based on pri- 2.4. Cell morphology and purity evaluation
mary cultures of mature and developing neurons may provide im-
portant insight into the neurotoxicity of AgNPs of different sizes and The neuron cells plated in 6-well plates at the density of
their particle-specific effects. 4.5 × 105 cells/well were cultured for 7 days, and then fixed with 4%
paraformaldehyde for 30 min. After washing three times with phos-
phate-buffered saline (PBS), the cells were incubated in PBS solution
2. Materials and methods supplemented with 10% FBS and 1% Triton X-100 at 37°C for 2 h, and
then immunoreacted with anti-neuron-specific β-III tubulin (TuJ1) an-
2.1. Reagents tibody (R&D, USA, 1:100, monoclonal mouse IgG2A) for 3 h at room
temperature in the dark. After rinsing with PBS, the cells were subse-
Two kinds of polyvinylpyrrolidone (PVP)-coated AgNP suspensions, quently stained with 4,6-diamino-2-phenylindole (DAPI, Solarbio, USA)
including AgNPs-20 (20 nm, 1 mg/mL as silver) and AgNPs-70 (70 nm, for 1 h, and observed using the inverted fluorescence microscope
1 mg/mL as silver) were commercially purchased from Shanghai (Olympus, IX73, Japan).
Huzheng Nano Technology Co., Ltd. (Shanghai, China) and After a 7-day culture, the cells were fixed and washed in 6-well
nanoComposix, Inc. (USA), respectively. Silver nitrate (AgNO3, > 99%) plates, then permeabilized by 0.2% Triton X-100, and washed with PBS.
was obtained from Sigma-Aldrich (USA), and its stock solution (1 mg/ Then, the cells were incubated with anti-NeuN antibody (Abcam, USA,
mL as silver) in distilled water was used as the control of ionic silver. 1:300) at 4°C overnight, followed by incubation with goat anti-rabbit
The working solutions of three silver species were prepared by suitable IgG (H + L) (Thermo Fisher Scientific, USA, 1:500) at 37°C for 1 h and
dilution with neurobasal medium (Gibco, Thermo Fisher Scientific, final DAPI staining. The images were taken by the inverted fluorescence
USA) under sonication for cell exposure experiments. microscope based on the random selection of the visual fields and
analyzed by Image J software (Ver 1.4.3, National Institutes of Health).

2.2. AgNP characterization


2.5. Cell viability assay
The physicochemical properties of AgNPs-20 and AgNPs-70 used in
this study were characterized after ultrasonication in aqueous solution To study the neurotoxicity of AgNPs, mature neuronal cells after a 7-
for 5 min. The morphology of AgNPs in both water and cell culture day culture were exposed to chemicals for 24 h (method 1). To study
medium were observed by transmission electron microscopy (TEM, the neurodevelopmental toxicity of AgNPs, neuronal cells were cultured
JEOL, 2100F, Japan). Briefly, several droplets of AgNP suspensions for 24 h, exposed to chemicals for 7 days (method 2), and the neurite
were dispersed on carbon membrane-coated copper nets and air dried. outgrowth of the cells was carefully observed. Half of the exposure
The representative images were then photographed, and the sizes of medium (the culture medium containing different concentrations of
AgNPs were analyzed based on counting 200 particles in four views chemicals) was renewed every 4 days afterward.
using Nano Measurer software (Version 1.2, China). The hydrodynamic The cells plated in 24-well plates at the density of 3.0 × 105 cells/
particle sizes as well as ζ-potentials were determined by a dynamic light well were exposed to different concentrations of AgNPs and AgNO3. In
scattering (DLS) technique using Malvern Zetasizer Nano ZS (Malvern, method 1, the exposure concentrations were 0.01, 0.1, 0.5, 1, 2, 5, 10,
UK) at room temperature, and the measurements were repeated for 20, 30, and 40 μg/mL, whereas in method 2, the concentrations were
three times. Total silver levels in two kinds of AgNPs were measured by 0.01, 0.1, 0.5, 1, 2, and 5 μg/mL. When the exposure was terminated,
inductively coupled plasma mass spectrometry (ICP-MS, Agilent 8800, half of the exposure medium (400 μL) was aspirated, and 40 μL of
USA) after digestion with the mixture of HNO3:H2O2 (3:1) at 95 °C for 100 μM alarmarBlue (AB) reagent (Sigma-Aldrich, USA) was added to
2 h. Silver ion release was evaluated by setting different concentrations each well. The plates were incubated at 37°C with 5% CO2 for 2 h in the
of AgNP suspensions in neurobasal medium, which were incubated at dark. The cell viability was evaluated by measuring fluorescence in-
37°C with 5% CO2 for 24 h and 7 days, respectively, to mimic the fol- tensities at 530 nm/590 nm (λexcitation/λemission) using the multi-
lowing exposure systems. The levels of Ag+ released from AgNP sus- function microplate reader (Thermo Scientific, VARIOSKAN FLASH,
pensions were analyzed by ICP-MS after ultracentrifugation (Millipore, USA). The EC50 values of the test chemicals were calculated by
Amicon 10 kDa, 16,000 g, 30 min). Nonlinear Curve Fit (Logistic) using Origin 2017 (Version 94E).

2
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

Fig. 1. AgNP characterization in medium. (A, B) TEM images. (C, D) The particle size distribution. (E, F) The hydrodynamic particle sizes.

2.6. Immunocytochemical staining 1, and 0.5 μg/mL AgNPs and AgNO3 for 24 h, 72 h, and 168 h, re-
spectively in method 2. After exposure, the cells were washed for three
The cells plated in 6-well plates at the density of 9.0 × 105 cells/ times with cold PBS, and lysed for 30 min. The cell lysates were col-
well were stimulated by different concentrations of AgNPs and AgNO3 lected after centrifugation (9,600 g, 5 min) to remove cell debris,
(1, 2, and 5 μg/mL for method 1, and 0.1, 0.5, and 2 μg/mL for method avoiding non-specific attachment of substantial amounts of chemicals
2). After exposure, the cells were fixed with 4% paraformaldehyde and on cell surface. Total silver concentrations in cell lysates were measured
washed three times with PBS. They were incubated in PBS solution by ICP-MS (Agilent 8800, USA) after digestion in the mixture of
supplemented with 10% FBS and 1% Triton X-100 for 2 h at 37°C, and HNO3:H2O2 (3:1) at 95°C for 2 h. Untreated cell lysates were analyzed
immunoreacted with TuJ1 antibody (R&D, USA) at the dilution ratio of as negative control, and the undetectable silver content confirmed no
1:100 for 3 h at room temperature in the dark. The cytoskeleton mor- cross contamination during exposure experiments. The protein con-
phology was observed using the inverted fluorescence microscope, and centrations of cell lysates measured by Pierce BCA kit (Thermo, USA)
photographed at 493 nm/514 nm (λexcitation/λemission), based on were used to calibrate final results of intracellular silver contents.
tracking the fluorescence of fluorochrome NL493 conjugated directly
on the purified TuJ1 antibody. The total fluorescence densities of dif-
ferent 55,000 μm2 were analyzed by ImageJ software. 2.8. Measurement of dopamine efflux

The cells plated in 6-well plates at the density of 9.0 × 105 cells/
2.7. Analysis of intracellular silver well were exposed to different concentrations of AgNPs (0.01, 0.1, 0.25,
and 0.5 μg/mL) and 0.05 μg/mL AgNO3 for 24 h (method 1) and 7 days
The cells plated in 100 mm dishes at the density of 4.5 × 106 cells/ (method 2), respectively. When the exposure was terminated, the
dish were exposed to 0.5 μg/mL AgNPs and AgNO3 for 24 h in method medium in each well was collected for the evaluation of dopamine

3
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

efflux. The cells were washed three times with cold PBS, and submitted performed for AgNPs dispersed in cell culture medium at different
to subsequent analysis of protein contents. The concentration of dopa- concentrations. The results in Table S1 show that the silver ion release
mine was analyzed by a rat dopamine enzyme-linked immunosorbent ratios of AgNPs-20 and AgNPs-70 (1.0–5.0 μg/mL) were 0.9%–5.3%
assay (ELISA) kit (Shanghai Yanjin Biological, China). The final values and 2.4%–7.9% during 24 h, respectively. Regarding 7 days of ex-
were calibrated by cell protein concentrations in different conditions, posure, silver ion release ratios of AgNPs-20 and AgNPs-70 (0.1–1.0 μg/
and the results relative to the control were expressed to adjust the mL) were 2.8%–9.9% and 2.3%–11.4%, respectively. The two sizes of
variations from different batches of the experiments. AgNPs had similar silver ion release ratios. Comparatively, Ag+ release
ratios were relatively higher in lower concentrations of AgNP disper-
sions.
2.9. Statistical analysis

All results were expressed as the mean values ± standard devia- 3.2. Cell morphology and purity
tions (SDs) of at least three independent experiments for each assay.
The statistical analysis was conducted for different groups by one-way The freshly isolated cortical neuronal cells were counted and eval-
analysis of variance (ANOVA) with the Bonferroni multi-group com- uated by the cell counting instrument (Life Technologies, AMQAX1000,
parison test. The significant differences were considered when the p- USA), and the batches with living cell proportions higher than 90%
value was less than 0.05 (*) or 0.01 (**). were submitted to further culture. The cell growth observation by the
inverted microscope indicated that neurons were well spread and ad-
hered in dishes 4 h after plating. After the 7-day culture, cells differ-
3. Results
entiated into mature neurons, and the tight neuronal network structure
was well developed (Figs. S2A–S2C), which could also be evidently
3.1. AgNP characterization
characterized by the structural protein, TuJ1, expressed in neuronal cell
bodies and axons (Fig. S3A). Mature neuronal cells could well survive
It has been shown that the particle size (Wang et al., 2018), shape
for 1 more week (Figs. S2D–S2F), thus providing an attractive experi-
(Jasinski et al., 2018), and coating (Zheng et al., 2018) can affect the
mental model of primary cultures with an approximately 2-week testing
biological effects of nanomaterials. To correlate the toxicity data with
window for neurotoxicity studies.
the specific properties of nanoparticles, two kinds of AgNPs were
To characterize the purity of primary cultures, the NeuN-stained
comprehensively characterized in this study. TEM results showed that
neuronal cells (red fluorescence) and total cells indicated by a DAPI-
the test AgNPs were uniform in size and well dispersed in both water
counterstained nucleus (blue fluorescence) were evaluated after the 7-
(Fig. S1A, Fig. S1B) and cell culture medium (Fig. 1A, Fig. 1B). No
day culture, and the results are shown in Figs. S3D–S3F. The consistent
obvious agglomeration was observed in any of the image fields. By
overlay of NeuN and DAPI-stained neuronal nuclei in the merged
fitting AgNP sizes with normal distribution function (Fig. S1C, Fig. 1C),
images shows the high purity of neuronal cultures. Quantitative ana-
the particle sizes of AgNPs-20 were 27.67 ± 3.39 nm and
lysis based on counting the numbers of both NeuN- and DAPI-stained
15.56 ± 1.53 nm in water and culture medium, respectively. Those of
cells in three independent cell culture batches (four fields per batch)
AgNPs-70 were 69.86 ± 6.97 nm and 84.39 ± 10.92 nm in water and
showed that the average purity of neuronal cells was greater than
culture medium, respectively (Fig. S1D, Fig. 1D). These results were
90.7% (Table S2), confirming that the lab-prepared cell models were
consistent with the product information provided by the manufacturers.
free of glial cell contamination (Lavezzi et al., 2013).
DLS measurement indicated that the hydrodynamic particle sizes of
AgNPs-20 were 38.47 ± 5.68 nm with a polydispersity index (PDI) of
0.239 in water, and 49.35 ± 3.74 nm with a PDI of 0.264 in the 3.3. Cell viability
culture medium (Fig. S1E, Fig. 1E). As for AgNPs-70, the hydrodynamic
sizes were 87.12 ± 1.61 nm with a PDI of 0.294 in water and To investigate the potential neurotoxicity of AgNPs and silver ions,
91.71 ± 1.58 nm with a PDI of 0.342 in the culture medium (Fig. S1F, the viability of neuronal cells exposed to different concentrations of the
Fig. 1F). The zeta potentials of AgNPs-20 and AgNPs-70 in water were test agents were evaluated using AB assay. The results in Fig. 2A
−15.30 ± 0.08 mV and −14.33 ± 1.48 mV, respectively. No ob- showed that all test silver species caused exposure concentration-de-
vious aggregation was observed during any of the following exposure pendent inhibition in cell proliferation after 24-h exposure. Based on
experiments. the calculation of logistic fitting curves for their dose responses, the 24-
Silver ion release would definitely influence the biological effects of h EC50 values were obtained for AgNPs-20 (6.61 ± 0.28 μg/mL),
AgNPs, and this process could be affected by many factors, such as AgNPs-70 (38.4 ± 1.61 μg/mL), and AgNO3 (2.86 ± 0.12 μg/mL),
AgNP concentration, storage condition and pH of the dispersion (Choi respectively. The cytotoxicity order of the test silver species of
et al., 2018). The determination of silver ion release ratios was thus AgNO3 > AgNPs-20 > AgNPs-70 was thus concluded. It was

Fig. 2. Effects of AgNPs and AgNO3 on neuronal cell viability (n = 3). (A) Mature cells on day 7 treated for 24 h. (B) Developing cells treated for 7 days.

4
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

apparent that ionic silver was more toxic than particulate silver, and networks, whereas the obvious granulated skeleton structure with
AgNPs with smaller size exerted higher neurotoxicity than did those broken synapses was observed when the exposure concentration was
with larger size, potentially due to the fact that small particulate silver increased to 5 μg/mL (indicated by white arrows, Fig. 3B, Figs. S5A and
was easier for cells to take up (Chen et al., 2015) and possessed higher S5B). As for AgNPs-70, they caused few deleterious effects on neuronal
bioactivity (Riaz Ahmed et al., 2017). The no-observed-adverse-effect morphology in any of the exposure groups (Fig. 3C, Figs. S5C and S5D).
level (NOAEL) for the test silver species was approximately 1 μg/mL. In view of AgNO3 treatments, the observable damages to neuronal
Regarding the strategy of 7-day exposure, the results in Fig. 2B networks occurred in the 2 μg/mL exposure group, and more severe
showed typical exposure concentration-related decreases in cell viabi- granulation in neuronal cells with ruptured structures appeared when
lity for all three test silver species, wherein, the logistic fitting curves the exposure concentration was increased to 5 μg/mL (Fig. 3D, Figs.
for silver ion and AgNPs-20 were very similar. The calculated 7-day S5E and S5F). Quantitative analysis of fluorescence intensities in dif-
EC50 values were 1.08 ± 0.4 μg/mL, 2.02 ± 0.07 μg/mL and ferent exposure groups showed that 2 μg/mL AgNPs-20 and 2–5 μg/mL
1.22 ± 0.04 μg/mL for AgNPs-20, AgNPs-70, and AgNO3, respectively. AgNO3 significantly compromised the expressions of TuJ1 (p < 0.01),
The toxicity order based on 7-day exposure experiments was AgNPs-20, whereas the fluorescence intensities of the other groups showed no
AgNO3 > AgNPs-70, which was different from that observed in the 24- difference from that of the negative control (Fig. 4A).
h experiments. The NOAEL of the test silver species was approximately Compared with the results from the 24-h exposure experiments, 7-
0.5 μg/mL, and it was apparent that their cytotoxicity was obviously day treatments of AgNPs and AgNO3 caused more severe degeneration
elevated when the exposure period was extended to 7 days. Obvious in the cytoskeleton structures. More specifically, the granular and
decreases in cell viability were observed even when the exposure con- fractured cytoskeleton structures were observed in different treatments.
centrations were 1 μg/mL or less. In addition, the cytotoxic effect of The delayed neuronal development was observed due to the prolonged
AgNPs-70 increased sharply in the 7-day exposure group when com- chemical treatments, resulting in sparse filaments in both bright fields
pared with that in the 24-h treatment, and its EC50 value decreased (Figs. S4F–S4H) and fluorescent views (Fig. 3F–H, S6). The im-
from 38.4 ± 1.61 μg/mL to 2.02 ± 0.07 μg/mL, which suggested the munostaining was apparently decreased, showing that the neurotoxi-
importance of studying the long-term and low-dose effects of chemicals, city could be induced by all test silver species at both cytotoxic and non-
although the low toxicity of AgNPs with large sizes has been commonly cytotoxic exposure levels. The quantitative analysis for the im-
reported (Gottschalk et al., 2009). munostaining of TuJ1 expression showed significant decreases in all
test groups (p < 0.01), which were exhibited in exposure concentra-
tion-related manners (Fig. 4B).
3.4. Cytoskeleton changes

To characterize cellular morphological changes induced by the ex- 3.5. Intracellular silver contents
posures of three silver species at cytotoxic and non-cytotoxic levels,
neuronal cells with different silver treatments (1–5 μg/mL for 24 h, or Recent studies have indicated that NPs enter into cells mainly
0.1–2 μg/mL for 7 days) were submitted to inverted microscopy ob- through endocytosis (Greulich et al., 2011), and this process can be
servation under both bright and fluorescent modes based on TuJ1 im- affected by the characteristics of NPs, such as surface charge (Bannunah
munostaining. et al., 2014) and particle size (Iversen et al., 2011). The intracellular
According to the images under bright field (Fig. S4A), the well-de- AgNPs can be expelled out of cells through slow exocytosis (AshaRani
veloped neuronal networks were formed in negative controls. The 24-h et al., 2009), thus forming a dynamic equilibrium process. The eva-
treatments of AgNPs-20 and AgNO3 induced breakages in neuronal fi- luation of intracellular silver levels could aid in understanding cyto-
laments (indicated by black arrows) to different extents, whereas toxicity induced by AgNPs. The concentration of 0.5 μg/mL was se-
AgNPs-70 did not (Figs. S4B–S4D). According to TuJ1 im- lected in follow-up experiments to avoid excessive damage to cells.
munocytochemical staining images, the neuronal cells were evenly Investigations on silver contents in mature neuronal cells after a 24-
distributed with big and intact cell bodies located in filament networks, h exposure of AgNPs showed that treatments of three silver species at
and the synapses of different neurons interlaced and joined into the the concentration of 0.5 μg/mL all increased intracellular silver levels
reticular structure in negative controls (Fig. 3A). Comparatively, ranging from 152 μg/g to 197 μg/g protein (Fig. 5A), and the accu-
AgNPs-20 at 1 μg/mL or 2 μg/mL caused few changes in the neuronal mulation followed the order of AgNPs-20 > AgNPs-70 > AgNO3,

Fig. 3. The influences of three silver species on cytoskeleton structures of neuronal cells. (A–D) Fluorescent images for TuJ1 expression in mature neuronal cells
treated with 2 μg/mL silver species for 24 h. (E–H) Fluorescent images for TuJ1 expression in developing neuronal cells treated with 2 μg/mL silver species for 7 days.
The white arrows indicate the fragmentation of neuronal networks.

5
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

Fig. 4. Quantification of neuron beta-tubulin III by fluorescent TuJ1 expression in response to increasing doses of AgNPs-20, AgNPs-70, and AgNO3 exposures for (A)
24 h, and (B) 7 days. n = 3, **p < 0.01 versus the control.

showing the relatively higher bioavailability of particulate silver com- result (Fig. 6A) shows that 24-h exposure of AgNPs-20 and AgNPs-70 to
pared with ionic silver. The time course shown in Fig. 5B demonstrates mature neurons both caused significant concentration-dependent de-
that the particulate-derived intracellular silver concentrations increased creases in extracellular dopamine levels (p < 0.01). Comparatively,
over time compared with ionic-derived silver. In regard to the accu- AgNPs-20 exerted stronger inhibition effect on dopamine efflux than
mulation capacity of different silver species, the exposure of particulate did AgNPs-70. Considering the potential contribution of silver ions re-
silver caused relatively higher intracellular silver levels than did ionic leased from AgNPs to the disturbed dopamine efflux, an equivalent
silver, which was in accord with the result in Fig. 5A. In addition, the concentration of AgNO3 (0.05 μg/mL) comparable to 0.5 μg/mL AgNPs
intracellular silver levels in the AgNPs-20 exposure groups were gen- with an Ag+ release ratio less than 10%, was evaluated for its effect on
erally higher than those in AgNPs-70 treatments, suggesting that the dopamine level. The result (Fig. 6A) shows no significant effect on
penetration of AgNPs through the cell membrane could be particle size- neurons with AgNO3 when compared to control (p > 0.05), indicating
related. Moreover, intracellular silver levels in developing neurons ex- that AgNP-induced inhibition in dopamine efflux is particle-specific
posed to AgNPs for 7 days did not increase in contrast to increased rather than ionic.
levels observed in mature neurons exposed for 24 h. This indicates that For developing neuronal cells after a 7-day exposure to AgNPs, the
prolonged exposure time does not contribute to intracellular silver ac- data in Fig. 6B shows that inhibition of dopamine levels exhibits a U-
cumulation, suggesting that the dynamic equilibrium of silver between shaped curve with increasing AgNP levels (0.01–0.5 μg/mL). The dif-
endocytosis and exocytosis may exist to maintain metal homeostasis in ferent dopamine release responses in developing neuronal cells
neurons. (Fig. 6B) compared with those in mature ones (Fig. 6A) represented the
complicated regulation of neuronal function in different primary cul-
3.6. Dopamine efflux tures. When compared with AgNPs-70, AgNPs-20 exerted relatively
stronger effects on altering dopamine efflux, showing a similar particle
size-related effect to that observed in Fig. 6A. The test for 7-day ex-
Dopamine is one of the most important neurotransmitters, and it is
critical to exercise regulation, learning, memory, and addictive beha- posure of 0.05 μg/mL AgNO3 also showed no changes in dopamine
efflux (Fig. 6B), further confirming the particle-specific effect of AgNPs
vior (Langdon et al., 2018). In vitro studies have shown that excessive
dopamine secretion can cause cytotoxic effects, whereas insufficient on developing neurons. Altogether, AgNP-caused inhibition in dopa-
mine efflux in both mature and developing neurons suggested their
dopamine release may result in disease risks, such as Parkinson's dis-
ease (Hisahara and Shimohama, 2011). AgNP exposure has been re- potential disturbances in neuronal function.
ported to alter dopamine levels in Wistar rat brains (Hadrup et al.,
2012). The changes in dopamine release from the primary cultures of 4. Discussion
neuronal cells upon AgNP exposure were studied herein to find the
potential influences of the test silver species on neuronal functions. The The biosafety of AgNPs is of great importance, considering their

Fig. 5. Intracellular silver accumulation in mature neuronal cells after 24-h exposure (A), and developing neuronal cells during 7-day exposure (B). The exposure
concentration of each chemical was 0.5 μg/mL. n = 3, *p < 0.05, and **p < 0.01.

6
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

Fig. 6. AgNPs disturbed dopamine release from primary cultures of cerebral cortical neurons (n = 3). (A) Mature cells at day 7 treated for 24 h. (B) Developing cells
treated for 7 days. *p < 0.05, and **p < 0.01.

wide application in diverse fields. And tuning the characteristics of zebrafish could be affected by both AgNPs and dissolved Ag+, but the
AgNPs would influence their bioactivities and toxicities, including regulation patterns by these two silver species were opposite (Kang
neurotoxicity. In the present study, two kinds of AgNPs with different et al., 2016). AgNPs could accumulate in the nucleus of NIH3T3 cells
sizes, together with AgNO3, were evaluated for their potential size-re- and induce DNA damage, compared with Ag+ (Jiravova et al., 2016).
lated and particle-specific effects on primary cultures of cortical neu- Different neurotoxic mechanisms were found for AgNPs and AgNO3 in a
ronal cells. The findings provided new insights for toxicity-directed recent study on human embryonic stem cell-derived neuron and as-
AgNP synthesis strategy and risk assessment on commerically available trocyte network (Repar et al., 2018). Regarding intracellular silver ac-
AgNPs. cumulation and dopamine release, the findings in the present study also
With regard to AgNP-induced neurotoxicity in primary cultures of showed a series of distinct effects induced by AgNPs and AgNO3 on
neuronal cells, the cell viability assay showed the toxicity order of the primary neuron cells, suggesting that particle-specific effects induced
three silver species (Fig. 2), which was visually demonstrated by cell by AgNPs could be more than what the released Ag+ could explain.
morphological alterations (Fig. S4). The cytoskeleton characterized by When it comes to biological effects of AgNPs, particle size is usually
TuJ1 protein indicated the deleterious effect caused by the test silver recognized as a crucial factor. For instance, smaller AgNPs more easily
species (Figs. 3 and 4), which was in accordance with previous findings entered cells (Chen et al., 2015), and had relatively higher biological
of degeneration of neuronal structures and dissolution of β-tubulin activity due to higher specific surface areas. According to the results
proteins in AgNP-exposed corticle neurons (Xu et al., 2013). In- obtained herein, AgNPs-20 exposure caused relatively stronger effects
tracellular silver existence (Fig. 5) caused the internal exposure of on primary neuronal cultures than did AgNPs-70, in view of cytotoxi-
neuronal cells, which would be responsible for the incidence of neu- city, cell morphology, and dopamine efflux. Nevertheless, neurotoxic
rotoxicological effects. The decreased dopamine efflux in AgNP ex- effects induced by AgNPs-70 were obviously increased when exposure
posure groups (Fig. 6) showed the potential purturbation of neuronal time was prolonged. In addition, the vulnerability of mature and de-
function, which would be clinically related to neurodegenerative dis- veloping neurons to AgNP exposure could also be different, indicating
eases, such as Parkinson's disease (Hisahara and Shimohama, 2011). the potential neurodevelopmental toxicity of AgNPs, which was in ac-
Concerning the neurotoxicity of AgNPs, the role of silver ion release, cordance with previous findings (Ema et al., 2017; Yin et al., 2018).
and particulate- and size-related effect required clarification. Collectively, the neurotoxicity of two kinds of AgNPs (AgNPs-20 and
It is well known that Ag+ release is an important mechanism of AgNPs-70) was evaluated using primary cultures of mature and devel-
AgNPs' bactericidal effect (van Aerle et al., 2013). As a slow and con- oping neuronal cell models. AgNP exposures caused particle size-re-
tinuous release source of Ag+, AgNPs have more lasting bactericidal lated cytotoxicity, cellular silver accumulation, and dopamine release.
effects (Zhou et al., 2015; Wang et al., 2013). Most studies reported that The developing neurons upon 7-day exposure responded more mark-
Ag+ exerted higher toxicity than did AgNPs. For example, at 1.5 μg/mL edly to cytotoxic and non-cytotoxic exposure levels of three silver
total silver, A549 cells exposed to an AgNP suspension containing 39% species than did mature neurons based on cell viability assay and TuJ1-
Ag+ caused a cell viability of 92%, whereas cells exposed to an AgNP probed cytoskeleton structure observation. The disturbances in dopa-
suspension containing 69% Ag+ caused a cell viability of 54% (Beer mine efflux induced by AgNP exposure was particle-specific, although
et al., 2012). The viability of planktonic N. europaea cells was inhibited silver ions exerted higher toxicity to neuronal cells. These findings
by 95% upon 0.5 μg/mL Ag+ exposure and by 87% upon 20 μg/mL provide substantial data on neurotoxicological effects of two different
AgNP exposure (Barker et al., 2018). The 24-h EC50 values were ob- sizes of AgNPs, and this information will be very helpful in their bio-
tained for AgNPs-20 (6.61 ± 0.28 μg/mL), AgNPs-70 safety assessment.
(38.4 ± 1.61 μg/mL), and AgNO3 (2.86 ± 0.12 μg/mL), respectively,
in this study, which were consistent with previous findings that Ag+
CRediT authorship contribution statement
was more toxic than AgNPs. Moreover, 7-day EC50 values were
1.08 ± 0.4 μg/mL, 2.02 ± 0.07 μg/mL and 1.22 ± 0.04 μg/mL for
Bingjie Zhang: Methodology, Investigation, Writing - original draft.
AgNPs-20, AgNPs-70, and AgNO3 respectively, which indicated that the
Na Liu: Software, Validation. Qian S. Liu: Formal analysis, Data
ability of AgNPs to degrade cell viability was increased after chronic
curation. Jianqing Zhang: Resources. Qunfang Zhou:
exposure. Similarly, long-term continuous exposure to AgNPs lowered
Conceptualization, Writing - review & editing. Guibin Jiang:
the effective inhibitory concentration by 1–2 orders of magnitude when
Supervision.
compared with those observed in short-term exposures (Barker et al.,
2018).
Further, the unique particle-specific effects of AgNPs have also been Declaration of competing interest
extensively reported, which could be different from Ag+-induced ef-
fects. For instance, it was reported that the cell cycle pathway in larval The authors declare that they have no conflicts of interest to this

7
B. Zhang, et al. Ecotoxicology and Environmental Safety 198 (2020) 110674

work. Jasinski, D.L., Li, H., Guo, P., 2018. The effect of size and shape of RNA nanoparticles on
biodistribution. Mol. Ther. 26, 784–792. https://doi.org/10.1016/j.ymthe.2017.12.
018.
Acknowledgments Jiravova, J., Tomankova, K.B., Harvanova, M., Malina, L., Malohlava, J., Luhova, L.,
Panacek, A., Manisova, B., Kolarova, H., 2016. The effect of silver nanoparticles and
We thank Dr. Qunhui Xie for the technique support on primary silver ions on mammalian and plant cells in vitro. Food Chem. Toxicol. 96, 50–61.
https://doi.org/10.1016/j.fct.2016.07.015.
cultures of rat cerebral cortical neurons. This study was financially Kaewamatawong, T., Banlunara, W., Maneewattanapinyo, P., Thammachareon, C.,
supported by the National Key Research and Development Program of Ekgasit, S., 2014. Acute and subacute pulmonary toxicity caused by a single in-
China (2018YFA0901101), Chinese Academy of Sciences (QYZDJ-SSW- tratracheal instillation of colloidal silver nanoparticles in mice: pathobiological
changes and metallothionein responses. J. Environ. Pathol. Toxicol. Oncol. 33,
DQC017), National Natural Science Foundation of China (21876195, 59–68. https://doi.org/10.1615/JEnvironPatholToxicolOncol.2014010179.
21806178, 21527901), and Sanming Project of Medicine in Shenzhen Kang, J.S., Bong, J., Choi, J.-S., Henry, T.B., Park, J.-W., 2016. Differentially transcrip-
(SZSM201811070). tional regulation on cell cycle pathway by silver nanoparticles from ionic silver in
larval zebrafish (Danio rerio). Biochem. Biophys. Res. Commun. 479, 753–758.
https://doi.org/10.1016/j.bbrc.2016.09.139.
Appendix A. Supplementary data Langdon, A.J., Sharpe, M.J., Schoenbaum, G., Niv, Y., 2018. Model-based predictions for
dopamine. Curr. Opin. Neurobiol. 49, 1–7. https://doi.org/10.1016/j.conb.2017.10.
Supplementary data to this article can be found online at https:// 006.
Lavezzi, A.M., Corna, M.F., Matturri, L., 2013. Neuronal nuclear antigen (NeuN): a useful
doi.org/10.1016/j.ecoenv.2020.110674. marker of neuronal immaturity in sudden unexplained perinatal death. J. Neurol. Sci.
329, 45–50. https://doi.org/10.1016/j.jns.2013.03.012.
References McShan, D., Ray, P.C., Yu, H., 2014. Molecular toxicity mechanism of nanosilver. J. Food
Drug Anal. 22, 116–127. https://doi.org/10.1016/j.jfda.2014.01.010.
Pachapur, V.L., Dalila Larios, A., Cledón, M., Brar, S.K., Verma, M., Surampalli, R.Y.,
Ahmed, M.M., Hussein, M.M.A., 2017. Neurotoxic effects of silver nanoparticles and the 2016. Behavior and characterization of titanium dioxide and silver nanoparticles in
protective role of rutin. Biomed. Pharmacother. 90, 731–739. https://doi.org/10. soils. Sci. Total Environ. 563–564, 933–943. https://doi.org/10.1016/j.scitotenv.
1016/j.biopha.2017.04.026. 2015.11.090.
AshaRani, P.V., Hande, M.P., Valiyaveettil, S., 2009. Anti-proliferative activity of silver Repar, N., Li, H., Aguilar, J.S., Li, Q.Q., Damjana, D., Hong, Y., 2018. Silver nanoparticles
nanoparticles. BMC Cell Biol. 10, 65. https://doi.org/10.1186/1471-2121-10-65. induce neurotoxicity in a human embryonic stem cell-derived neuron and astrocyte
Bannunah, A.M., Vllasaliu, D., Lord, J., Stolnik, S., 2014. Mechanisms of nanoparticle network. Nanotoxicol. 12, 104–116. https://doi.org/10.1080/17435390.2018.
internalization and transport ccross an intestinal epithelial cell model: effect of size 1425497.
and surface charge. Mol. Pharm. 11, 4363–4373. https://doi.org/10.1021/ Riaz Ahmed, K.B., Nagy, A.M., Brown, R.P., Zhang, Q., Malghan, S.G., Goering, P.L.,
mp500439c. 2017. Silver nanoparticles: significance of physicochemical properties and assay in-
Barker, L.K., Giska, J.R., Radniecki, T.S., Semprini, L., 2018. Effects of short- and long- terference on the interpretation of in vitro cytotoxicity studies. Toxicol. In Vitro 38,
term exposure of silver nanoparticles and silver ions to nitrosomonas europaea bio- 179–192. https://doi.org/10.1016/j.tiv.2016.10.012.
films and planktonic cells. Chemosphere 206, 606–614. https://doi.org/10.1016/j. Sha, R., Chen, Y., Luo, L., Liu, Y., Xu, L., Xie, H.Q., Zhao, B., 2019. Effects of astrocyte
chemosphere.2018.05.017. conditioned medium on neuronal AChE expression upon 2,3,7,8-tetrachlorodibenzo-
Beer, C., Foldbjerg, R., Hayashi, Y., Sutherland, D.S., Autrup, H., 2012. Toxicity of silver p-dioxin exposure. Chem. Biol. Interact. 309, 108686. https://doi.org/10.1016/j.cbi.
nanoparticles-nanoparticle or silver ion? Toxicol. Lett. 208, 286–292. https://doi. 2019.05.052.
org/10.1016/j.toxlet.2011.11.002. Sun, C., Yin, N., Wen, R., Liu, W., Jia, Y., Hu, L., Zhou, Q., Jiang, G., 2016. Silver na-
Chen, I.C., Hsiao, I.L., Lin, H.C., Wu, C.H., Chuang, C.Y., Huang, Y.J., 2016. Influence of noparticles induced neurotoxicity through oxidative stress in rat cerebral astrocytes is
silver and titanium dioxide nanoparticles on in vitro blood-brain barrier perme- distinct from the effects of silver ions. Neurotoxicol. 52, 210–221. https://doi.org/10.
ability. Environ. Toxicol. Pharmacol. 47, 108–118. https://doi.org/10.1016/j.etap. 1016/j.neuro.2015.09.007.
2016.09.009. van Aerle, R., Lange, A., Moorhouse, A., Paszkiewicz, K., Ball, K., Johnston, B.D., de-
Chen, L., Fang, L., Ling, J., Ding, C., Kang, B., Huang, C., 2015. Nanotoxicity of silver Bastos, E., Booth, T., Tyler, C.R., Santos, E.M., 2013. Molecular mechanisms of
nanoparticles to red blood cells: size dependent adsorption, uptake, and hemolytic toxicity of silver nanoparticles in zebrafish embryos. Environ. Sci. Technol. 47,
activity. Chem. Res. Toxicol. 28, 501–509. https://doi.org/10.1021/tx500479m. 8005–8014. https://doi.org/10.1021/es401758d.
Choi, Y., Kim, H.-A., Kim, K.-W., Lee, B.-T., 2018. Comparative toxicity of silver nano- Wang, W., Zeng, C., Feng, Y., Zhou, F., Liao, F., Liu, Y., Feng, S., Wang, X., 2018. The size-
particles and silver ions to Escherichia coli. J. Environ. Sci. 66, 50–60. https://doi. dependent effects of silica nanoparticles on endothelial cell apoptosis through acti-
org/10.1016/j.jes.2017.04.028. vating the p53-caspase pathway. Environ. Pollut. 233, 218–225. https://doi.org/10.
Dan, M., Wen, H., Shao, A., Xu, L., 2018. Silver nanoparticle exposure induces neuro- 1016/j.envpol.2017.10.053.
toxicity in the rat hippocampus without increasing the blood-brain barrier perme- Wang, Z., Liu, S., Ma, J., Qu, G., Wang, X., Yu, S., He, J., Liu, J., Xia, T., Jiang, G.-B., 2013.
ability. J. Biomed. Nanotechnol. 14, 1330–1338. https://doi.org/10.1166/jbn.2018. Silver nanoparticles induced RNA polymerase-silver binding and RNA transcription
2563. inhibition in erythroid progenitor cells. ACS Nano 7, 4171–4186. https://doi.org/10.
Ema, M., Okuda, H., Gamo, M., Honda, K., 2017. A review of reproductive and devel- 1021/nn400594s.
opmental toxicity of silver nanoparticles in laboratory animals. Reprod. Toxicol. 67, Wen, R., Yang, X., Hu, L., Sun, C., Zhou, Q., Jiang, G., 2016. Brain-targeted distribution
149–164. https://doi.org/10.1016/j.reprotox.2017.01.005. and high retention of silver by chronic intranasal instillation of silver nanoparticles
Eun-Jung, P., Jongheop, Y., Younghun, K., Kyunghee, C., Kwangsik, P., 2010. Silver na- and ions in Sprague-Dawley rats. J. Appl. Toxicol. 36, 445–453. https://doi.org/10.
noparticles induce cytotoxicity by a Trojan-horse type mechanism. Toxicol. In Vitro 1002/jat.3260.
24, 872–878. https://doi.org/10.1016/j.tiv.2009.12.001. Yin, N., Zhang, Y., Yun, Z., Liu, Q., Qu, G., Zhou, Q., Hu, L., Jiang, G., 2015. Silver
Gliga, A.R., Skoglund, S., Odnevall Wallinder, I., Fadeel, B., Karlsson, H.L., 2014. Size- nanoparticle exposure induces rat motor dysfunction through decrease in expression
dependent cytotoxicity of silver nanoparticles in human lung cells: the role of cellular of calcium channel protein in cerebellum. Toxicol. Lett. 237, 112–120. https://doi.
uptake, agglomeration and Ag release. Part. Fibre Toxicol. 11, 11. https://doi.org/10. org/10.1016/j.toxlet.2015.06.007.
1186/1743-8977-11-11. Xu, F., Piett, C., Farkas, S., Qazzaz, M., Syed, N.I., 2013. Silver nanoparticles (AgNPs)
Gottschalk, F., Sonderer, T., Scholz, R.W., Nowack, B., 2009. Modeled environmental cause degeneration of cytoskeleton and disrupt synaptic machinery of cultured cor-
concentrations of engineered nanomaterials (TiO2, ZnO, Ag, CNT, Fullerenes) for tical neurons. Mol. Brain 6, 29. https://doi.org/10.1186/1756-6606-6-29.
different regions. Environ. Sci. Technol. 43, 9216–9222. https://doi.org/10.1021/ Yin, N., Hu, B., Yang, R., Liang, X., Liang, S., Faiola, F., 2018. Assessment of the devel-
es9015553. opmental neurotoxicity of silver nanoparticles and silver ions with mouse embryonic
Greulich, C., Diendorf, J., Simon, T., Eggeler, G., Epple, M., Köller, M., 2011. Uptake and stem cells in vitro. J. Interdiscip. Nanomed. 3, 133–145. https://doi.org/10.1002/
intracellular distribution of silver nanoparticles in human mesenchymal stem cells. jin2.49.
Acta Biomater. 7, 347–354. https://doi.org/10.1016/j.actbio.2010.08.003. Zhang, B., Liu, Q.S., Zhou, Q., Zhang, J., Jiang, G., 2018. Neurotoxicological effects of
Hadrup, N., Loeschner, K., Mortensen, A., Sharma, A.K., Qvortrup, K., Larsen, E.H., Lam, nanosilver. Prog. Chem. 30, 1392–1402. https://doi.org/10.7536/PC171228.
H.R., 2012. The similar neurotoxic effects of nanoparticulate and ionic silver in vivo Zhang, Y.B., Ferguson, S.A., Watanabe, F., Jones, Y., Xu, Y., Biris, A.S., Hussain, S., Ali,
and in vitro. Neurotoxicol. 33, 416–423. https://doi.org/10.1016/j.neuro.2012.04. S.F., 2013. Silver nanoparticles decrease body weight and locomotor activity in adult
008. male rats. Small 9, 1715–1720. https://doi.org/10.1002/smll.201201548.
Hisahara, S., Shimohama, S., 2011. Dopamine receptors and Parkinson’s disease. Int. J. Zheng, M., Lu, J., Zhao, D., 2018. Effects of starch-coating of magnetite nanoparticles on
Med. Chem. 2011, 403039. https://doi.org/10.1155/2011/403039. cellular uptake, toxicity and gene expression profiles in adult zebrafish. Sci. Total
Hossain, S.M.Z., 2016. Enzyme-luminescence method: tool for real-time monitoring of Environ. 622–623, 930–941. https://doi.org/10.1016/j.scitotenv.2017.12.018.
natural neurotoxins in vitro and l-glutamate release from primary cortical neurons. Zhou, Q., Sun, C., Liu, W., Jiang, G., 2015. Evaluation of nanosilver-induced toxicity in
Biotechnol. Rep. 9, 57–65. https://doi.org/10.1016/j.btre.2015.12.002. aquatic environment using the Chinese rare minnow. Chin. Sci. Bull. 60, 645–653.
Iversen, T.-G., Skotland, T., Sandvig, K., 2011. Endocytosis and intracellular transport of https://doi.org/10.1360/n972014-01183.
nanoparticles: present knowledge and need for future studies. Nano Today 6,
176–185. https://doi.org/10.1016/j.nantod.2011.02.003.

You might also like