Vous êtes sur la page 1sur 11

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/236086543

Cytotoxicity and genotoxicity induced by zinc


oxide nanoparticles

Article in Environment international March 2013


DOI: 10.1016/j.envint.2013.02.013 Source: PubMed

CITATIONS READS

43 448

7 authors, including:

Vanessa Valdiglesias Eduardo Psaro


University of A Corua University of A Corua
58 PUBLICATIONS 579 CITATIONS 115 PUBLICATIONS 1,650 CITATIONS

SEE PROFILE SEE PROFILE

Blanca Laffon Joo Paulo Teixeira


University of A Corua National Institute of Health Dr. Ricardo Jorge
120 PUBLICATIONS 1,860 CITATIONS 180 PUBLICATIONS 1,247 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Cost Action CA 15132 hCOMET-The comet assay as a human biomonitoring tool View
project

FUTURAR - Air quality in Portugal in 2030 - a policy support View project

All in-text references underlined in blue are linked to publications on ResearchGate, Available from: Blanca Laffon
letting you access and read them immediately. Retrieved on: 01 October 2016
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elseviers archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/authorsrights
Author's personal copy

Environment International 55 (2013) 92100

Contents lists available at SciVerse ScienceDirect

Environment International
journal homepage: www.elsevier.com/locate/envint

Neuronal cytotoxicity and genotoxicity induced by zinc oxide nanoparticles


Vanessa Valdiglesias a, b, 1, Carla Costa c,, 1, Gzde Kili a, d, Solange Costa c, Eduardo Psaro a,
Blanca Laffon a, Joo Paulo Teixeira c
a
Toxicology Unit, Department of Psychobiology, University of A Corua, Edicio de Servicios Centrales de Investigacin, Campus Elvia s/n, 15071, A Corua, Spain
b
Clinical and Molecular Epidemiology, IRCCS San Raffaele Pisana, Via di Val Cannuta, 247, 00166, Roma, Italy
c
Department of Environmental Health, Portuguese National Institute of Health, Rua Alexandre Herculano, 321, 4000-055, Porto, Portugal
d
Department of Cell and Molecular Biology, University of A Corua, Facultad de Ciencias, Campus A Zapateira s/n, 15071, A Corua, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Zinc oxide nanoparticles (ZnO NPs) are one of the most abundantly used nanomaterials in consumer products and
Received 24 December 2012 biomedical applications. As a result, human exposure to these NPs is highly frequent and they have become an
Accepted 28 February 2013 issue of concern to public health. Although toxicity of ZnO NPs has been extensively studied and they have been
Available online xxxx
shown to affect many different cell types and animal systems, there is a signicant lack of toxicological data for
ZnO NPs on the nervous system, especially for human neuronal cells and tissues. In this study, the cytotoxic and
Keywords:
Zinc oxide nanoparticles
genotoxic effects of ZnO NPs on human SHSY5Y neuronal cells were investigated under different exposure condi-
Cytotoxicity tions. Results obtained by ow cytometry showed that ZnO NPs do not enter the neuronal cells, but their presence
Genotoxicity in the medium induced cytotoxicity, including viability decrease, apoptosis and cell cycle alterations, and
Oxidative damage genotoxicity, including micronuclei production, H2AX phosphorylation and DNA damage, both primary and oxida-
SHSY5Y cells tive, on human neuronal cells in a dose- and time-dependent manner. Free Zn2+ ions released from the ZnO NPs
Zn+ 2 ions were not responsible for the viability decrease, but their role on other types of cell damage cannot be ruled out. The
results obtained in this work contribute to increase the knowledge on the genotoxic and cytotoxic potential of ZnO
NPs in general, and specically on human neuronal cells, but further investigations are required to understand the
action mechanism underlying the cytotoxic and genotoxic effects observed.
2013 Elsevier Ltd. All rights reserved.

1. Introduction (Vandebriel and De Jong, 2012), so they can potentially reach any organ
or tissue and involve a risk for human health. Toxicity of ZnO NPs has
Zinc oxide nanoparticles (ZnO NPs) are one of the most abundantly been extensively studied and they have been shown to affect many differ-
used nanomaterials in consumer products and biomedical applications ent cell types and animal systems (Chiang et al., 2012; De Berardis et al.,
due to their specic properties, e.g. transparency, high isoelectric 2010; Osman et al., 2010; Sharma et al., 2012a, 2012b; Wahab et al.,
point, biocompatibility, and photocatalytic efciency. They are widely 2011). Commonly, the toxicity of NPs is associated with their small size
employed in a variety of devices including cosmetics, toothpaste, sun- and high specic surface area and therefore, nanoforms are theoretically
screens, llings in medical materials, textiles, wall paints, and other expected to be more toxic than their bulk counterparts (Xiong et al.,
building materials, and they can be also utilized in environmental remedi- 2011).
ation for elimination or degradation of pollutants in water or air (Qiang, In recent years, there have been an increasing number of works
2001). Furthermore, ZnO NPs have promising applications in the medi- reporting that different NPs can reach the brain and cause neurological
cine eld since they have been proposed as a possible treatment for can- injuries, being associated even with neurodegenerative diseases (Block
cer and/or autoimmune diseases after being found to be selectively toxic et al., 2004; Hu and Gao, 2010; Peters et al., 2006). This translocation
towards potential disease-causing cells (Hanley et al., 2008; Premanathan can happen both directly, through axonal transport from olfactory
et al., 2011; Akhtar et al., 2012), and they are being considered to be used epithelium, or indirectly by passing to the bloodstream and crossing
in fabrication of nerve guidance channels for treatment of nerve injury the blood brain barrier (Oberdrster et al., 2004). Similarly to other
(Seil and Webster, 2008). As a result of all these uses, human exposure metal oxide NPs, it has been recently found that ZnO NPs reach the
to these NPs is highly frequent. They can enter the organism through brain of experimental animals after oral (Lee et al., 2012) and inhalatory
different pathways (respiratory tract, digestive system and parenteral (Kao et al., 2012a) administration. Still, there is a signicant lack of toxico-
routes) and have shown a systemic distribution in in vivo studies logical data for ZnO NPs on nervous system, especially for human neuro-
nal cells and tissues.
Corresponding author. Tel.: +351 223401147; fax: +351 223401149.
In vitro data have shown that ZnO NPs induce cytotoxicity in mouse
E-mail address: cstcosta@gmail.com (C. Costa). neuroblastoma Neuro-2A cells and neural stem cells (Deng et al., 2009;
1
These authors contributed equally to this work. Jeng and Swanson, 2006), in rat RSC96 Schwann cells and primary

0160-4120/$ see front matter 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.envint.2013.02.013
Author's personal copy

V. Valdiglesias et al. / Environment International 55 (2013) 92100 93

neuronal cells (Chiang et al., 2012; Yin et al., 2012), and in human glioma (1%) were employed as positive controls in the cellular uptake and the
cells (Ostrovsky et al., 2009). Besides, they were found to enhance the lactate dehydrogenase (LDH) assays, respectively.
excitability of rat neurons by altering the ion channels (Zhao et al.,
2009) and to decrease the adhesion of rat astroglial cells (Seil and 2.4. Cellular viability
Webster, 2008), although in this last case the cells were exposed to
composite materials with ZnO NPs. MTT assay (according to Mosmann, 1983) and NRU assay (according
The only in vivo study describing neurological effects after ZnO NP to Borenfreund and Puerner, 1985) were used to test the potential effects
exposure reported attenuation in spatial learning and memory ability of ZnO NPs on viability of neuronal SHSY5Y cells. In each assay, 8 differ-
by alteration of synaptic plasticity in rats after intraperitoneal adminis- ent NP concentrations (080 g/ml) and 4 exposure times (3, 6, 24, and
tration (Han et al., 2011). Besides, morphological and histochemical 48 h) were evaluated. Absorbance was measured at 595 nm (MTT) or
changes in brains of rats fed with bulk ZnO were also described (Kozik 540 nm (NRU) using a Cambrex ELx808 microplate reader (Biotek,
et al., 1980). KC4). A parallel set of experiments conducted without cells was carried
Given the wide and frequent use of ZnO NPs in many elds closely out to exclude the potential interaction of the NPs with the dyes used
related to human beings, their promising benecial applications in in MTT and NRU assays. Data obtained demonstrated no interaction
medicine, and the scarce knowledge on their potential neurotoxic effects, between the ZnO NPs tested and the dyes used for cytotoxicity assess-
the main objective of this work was to investigate the cytotoxic and ment. From the results obtained in the viability experiments, two expo-
genotoxic effects of ZnO NPs on human SHSY5Y neuronal cells and to sure times (3 and 6 h) and three different concentrations of ZnO NPs (20,
explore the underlying mechanisms involved in these effects. 30 and 40 g/ml) were selected to perform the subsequent experiments.

2. Materials and methods 2.5. Cellular uptake

2.1. Chemicals A ow cytometry methodology (FACSCalibur Flow Cytometer, Becton


Dickinson, Madrid, Spain) was employed to assess the potential of the
ZnO NPs (CAS No. 1314-13-2), mytomycin C (MMC) (CAS No. ZnO NPs to enter the cells. The analysis was carried out on the basis of
50-07-7), bleomycin (BLM) (CAS No. 9041-93-4), camptothecin the size and complexity of the cells by measuring the forward scatter
(Campt) (CAS No. 7689-03-4), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl (FSC) and the side scatter (SSC) following the protocol described by
tetrazolium bromide (MTT) (CAS No. 298-93-1), 5,5,6,6-tetrachloro- Suzuki et al. (2007).
1,1,3,3-tetraethyl-imidacarbocyanine iodide (JC-1) (CAS No. 3520-43-
2), neutral red dye (CAS No. 553-24-2), and propidium iodide (PI) were 2.6. Cell cycle
purchased from Sigma-Aldrich Co. (Madrid, Spain). MMC, BLM, and
Campt were dissolved in sterile distilled water prior use. The cell distribution along the different phases of the cell cycle was
examined in cells treated with ZnO NPs or the controls by evaluating
the relative cellular DNA content with a ow cytometry technique
2.2. Nanoparticle suspension: preparation and characterization (FACSCalibur Flow Cytometer, Becton Dickinson, Madrid, Spain) follow-
ing Valdiglesias et al. (2011a). DNA content was assessed from the PI
A stock suspension of ZnO NPs (nal concentration 80 g/ml) was signal detected by the FL2 detector in a minimum of 104 events. Cell
prepared in either deionized water or complete cell culture medium Quest Pro software (Becton Dickinson, Madrid, Spain) was used to
(with fetal bovine serum, FBS). Prior to each treatment, this suspension analyze cell cycle histograms, to calculate the percentage of occupancy
was ultrasonicated (Branson Sonier, USA) at 30 W for 5 min (1.5 min of G0/G1, S and G2/M regions.
on and 1 min off twice, and 2 min on), and diluted to prepare the differ-
ent NP concentrations tested. Average hydrodynamic size, size distribu- 2.7. Membrane integrity
tion and zeta potential of particles in suspension were determined by
Dynamic Light Scattering (DLS) using a Zetasizer Nano-ZS equipped Lactate dehydrogenase (LDH) activity in cell culture medium was
with 4.0 mW, 633 nm laser (Model ZEN 3600, Malvern Instruments measured by using a commercial kit (Roche Diagnostics Corp) according
Ltd., Malvern, UK). to the manufacturer's instructions. After exposure, half the amount of the
cell culture medium was collected for LDH measurement. Absorption
2.3. Cell culture and treatments was measured at 490 nm with a reference wavelength of 655 nm
using a Cambrex ELx808 microplate reader (Biotek, KC4). Positive con-
Human neuroblastoma SHSY5Y cell line was obtained from the trol experiments were performed with 0.1% Triton X-100 and set as
European Collection of Cell Cultures and cultured in nutrient mixture 100% cytotoxicity. LDH release was calculated by the following equation:
EMEM/F12 (1:1) medium with 1% non-essential amino acids, 1% antibi-
otic and antimycotic solution, and supplemented with 10% heat- Asample Amedium
LDH%  100
inactivated fetal bovine serum (FBS), all from Invitrogen (Barcelona, Apositivecontrol Amedium
Spain). The cells were incubated in a humidied atmosphere with 5%
CO2 at 37 C. To carry out the experiments, cells were seeded in where [A]sample, [A]medium, and [A]positive control denote the absorbance of
96-well plates (at bottom) and allowed to adhere for 24 h at 37 C. the sample, medium negative control and Triton X-100 positive control,
Cell densities were approximately in the range of 25 105 cells/well respectively.
at the beginning of cell culture. For cell treatment, these were incubated
at 37 C for different periods in the presence of a variable number of NP 2.8. Apoptosis
concentrations, depending on the assay performed, or the control solu-
tions. Complete medium was used as negative control in all experiments. 2.8.1. Analysis of apoptosis by PIannexin V staining
The following chemicals were used as positive controls: Campt (10 M) Apoptosis rate was determined by means of annexin V/PI double
in apoptosis assessment, cell cycle and H2AX analysis; and MMC staining, using the BD Pharmingen annexin VFITC apoptosis detection
(1.5 M), BLM (1 g/ml) and H2O2 (2 M for 3 h treatments, and 1 M kit I (Becton Dickinson, Madrid, Spain), following the manufacturer's rec-
for 6 h treatments) in MN test, comet assay, and oxidative DNA damage ommendations. At least 104 events were acquired with a FACSCalibur
evaluation, respectively. Besides, TiO2 NPs (120 g/ml) and triton X-100 Flow Cytometry (Becton Dickinson, Madrid, Spain). Data from annexin
Author's personal copy

94 V. Valdiglesias et al. / Environment International 55 (2013) 92100

Vuorescein isothiocyanate (FITC) (FL1) and PI (FL2) were analyzed were set according to the results obtained with FAAS. Cell viability was
using Cell Quest Pro software (Becton Dickinson, Madrid, Spain). evaluated using the MTT assay (procedure described above).

2.8.2. Mitochondrial membrane potential analysis 2.12. Statistical analysis


Changes in mitochondrial membrane potential (MMP) after ZnO
NP treatments were evaluated by ow cytometry (FACSCalibur, Statistical analyses were performed using SPSS for Windows statisti-
Becton Dickinson, Madrid, Spain) as previously described (Sharma cal package (version 20.0). Distribution of the response variables depart-
et al., 2012b). Data obtained from at least 10 4 events were analyzed ed signicantly from normality (KolmogorovSmirnov goodness of t
by Cell Quest Pro software (Becton Dickinson, Madrid, Spain). test) and therefore non-parametric tests were considered adequate for
the statistical analysis of these data. Differences among groups were
2.9. Genotoxicity tested with KruskalWallis test and MannWhitney U-test. The associa-
tions between two variables were analyzed by Spearman's correlation. A
2.9.1. Micronuclei evaluation by ow cytometry minimum of three independent experiments were performed for each
After incubation of the cells in the presence or absence of ZnO NPs, experimental condition tested. Experimental data were expressed
they were cultured in fresh medium for an additional period of 24 h. as mean standard error and a P-value of b 0.05 was considered
Then a suspension of nuclei and micronuclei (MN) was prepared signicant.
according to the procedure described by Nsse et al. (1994) and
Roman et al. (1998), with some modications (Valdiglesias et al.,
3. Results
2011b). The nal suspension was analyzed with a FACSCalibur Flow
Cytometer (Becton Dickinson, Madrid, Spain) as previously reported
3.1. Nanoparticle characterization
(Avlasevich et al., 2006).
Table 1 summarizes the description and characterization data of the
2.9.2. H2AX assay ZnO NPs employed in this study. The average hydrodynamic size, size
The general protocol proposed by Tanaka et al. (2009) with minor distribution and zeta potential of ZnO NPs in water and in complete
changes (Valdiglesias et al., 2011b) was followed for the evaluation of cell culture medium suspensions were assessed by Dynamic Light
H2AX histone phosphorylation after ZnO NP treatments. Data from Scattering (DLS). Slightly higher values of the mean hydrodynamic diam-
Alexa Fluor 488 (FL1) and PI (FL2) intensities from a minimum of eter and the zeta potential (negative) were observed in culture medium
10 4 events were acquired with a FACSCalibur Flow Cytometer (Becton supplemented with FBS (distributions shown in Fig. 1(a) and (b), respec-
Dickinson, Madrid, Spain). Data analyses were performed using Cell tively) than in water.
Quest Pro software (Becton Dickinson, Madrid, Spain).

3.2. Cellular viability


2.9.3. Comet assay
After treatments with ZnO NPs, the alkaline comet assay was
Results obtained from both cytotoxicity tests, MTT and NRU, are
performed following Singh et al. (1988), with minor changes (Costa et
represented in Fig. 2. ZnO NPs induced dose-dependent decreases in
al., 2008). Comet IV Software (Perceptive Instruments, Haverhill, UK)
viability percentages of SHSY5Y cells, particularly pronounced at 24
was used for image capture and analysis. In all cases 100 cells were
and 48 h treatments in MTT assay. For MTT assay signicance was
scored (50 from each replicate slide), and percentage of DNA in the
obtained from 10 g/ml on treatment periods longer than 6 h, and for
Comet tail (%tDNA) was used as DNA damage parameter.
both cytotoxicity tests from 25 g/ml on all the treatment times.

2.9.4. Oxidative DNA damage


A modied version of the comet assay incorporating incubation with 3.3. Uptake
8-oxoguanine DNA glycosylase (OGG1) repair enzyme was employed to
evaluate the oxidative DNA damage, specically the presence of Fig. 3 shows the results obtained from the evaluation of the cellular
8-oxoguanine, following the protocol described by Smith et al. (2006). uptake by ow cytometry. No uptake of ZnO NPs by the SHSY5Y cells
Treatments following the same protocol with just buffer and no OGG1 was observed at any concentration or exposure time tested.
enzyme served as reference. Similar to the standard comet assay, 100
cells per condition were scored to determine %tDNA. 3.4. Cell cycle

2.10. Dissolved zinc concentrations in the cell culture medium Cell percentages in the different phases of the cell cycle (G0/G1, S,
G2/M) were assessed by ow cytometry after ZnO NP treatment. Signif-
To quantify the Zn+2 ion concentrations released from the ZnO NPs, icant alterations of cell cycle were observed after both 3 and 6 h
suspensions (2, 4, 6, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50 and 55 g/ml) (Table 2). Specically, ZnO NPs induced decreases in the percentage of
were incubated in complete cell medium for 3, 6 and 24 h at 37 C in a cells in G0/G1 and G2/M phases, and an increase in the percentage of
humidied 5% CO2 environment. Then centrifugation at 14,000 rpm for cells in S-phase. Besides, signicant doseresponse relationships were
30 min removed solid phase ZnO from the liquid medium. The Zn
content in the supernatant was analyzed by ame atomic absorption Table 1
spectroscopy (FAAS) (Thermoelemental Solaar S4 v.10.02). As negative Characterization of ZnO NPs.

control, cell culture medium without NPs but subjected to the same con- Particle size Specic surface area Hydrodynamic Zeta potential
ditions was used. (nm)a (m2/g)a diameter (mV)(DLS)
(nm)(DLS)

2.11. Cytotoxic effects of Zn +2 ions Water Medium Water Medium

100(BET) 1525 243.7 273.4 8.23 11.7


In order to test whether cytotoxicity of ZnO nanoparticles was due to BET: BrunauerEmmettTeller.
Zn2+ ions released from ZnO NPs, cells were treated for 3 h with DLS: Dynamic Light Scattering.
a
ZnSO4.7H2O at 0.05, 0.1, 0.2, 0.4, 0.6, and 0.8 mM. These concentrations Provided by the commercial supplier.
Author's personal copy

V. Valdiglesias et al. / Environment International 55 (2013) 92100 95

Fig. 1. Characterization of ZnO NPs in complete medium. Hydrodynamic diameter (a) and zeta potential (b).

found in phase G2/M (r = 0.631, P b 0.01) at 3 h, and in phases S 3.8. Oxidative damage
(r = 0.759, P b 0.01) and G2/M (r = 0.743, P b 0.01) at 6 h.
The NP potential to induce oxidative DNA damage was evaluated by
3.5. Membrane integrity means of comet assay modied by adding an incubation step with the
enzyme OGG1. Increases in DNA damage (%tDNA) in cells incubated
LDH activity in extracellular medium was evaluated as indicator of with OGG1 regarding those incubated only with buffer (indicative of
membrane integrity, since LDH is released when the membrane is oxidative DNA damage) were observed after ZnO NP treatment at all
damaged. Results obtained in this test are collected in Fig. 4. No sig- the concentrations and exposure times tested (Fig. 5).
nicant alteration in the percentage of LDH released was observed
in the neuronal cells after ZnO NP exposure at any treatment time. 3.9. Zn +2 ions: release from NPs and effects

After incubation of ZnO NPs (2 to 55 g/ml) in complete cell media


3.6. Apoptosis for 3, 6 and 24 h, Zn+2 ions were quantied by FAAS in the liquid super-
natant fraction. Results obtained were very similar at all the treatment
Table 3 shows the results obtained from apoptosis assessment by times tested (Fig. 6). A dose-dependent linear increase in Zn+2 ions
annexin V/PI analysis and MMP assay. Increases in the percentage of was observed in cell culture medium up to approximately 30 g/ml
apoptotic cells were found after both 3 h and 6 h ZnO NP treatments ZnO NPs; then a plateau is reached.
(annexin V results). However, only slight increases in mitochondrial To investigate the potential cytotoxicity of the released Zn+2 ions, the
membrane potential, with just a statistically signicant result at cells were treated with a soluble Zn salt (ZnSO4) to obtain Zn+2 ion
20 g/ml after 3 h of exposure, were observed. concentrations comparable to those existing in the initial ZnO NP suspen-
sions (FAAS results: 0.05 to 0.8 mM). Signicant decreases in the cell
3.7. Genotoxicity viability were found only at the highest concentrations tested (0.6 and
0.8 mM) (Fig. 7).
Results obtained from the MN test, comet assay, and analysis of
H2AX are collected in Table 4. ZnO NPs did not induce MN production 4. Discussion
after the 3 h exposure. However, an important dose-dependent MN
induction was observed after the 6 h treatment (r = 0.635, P b 0.01). The increasing use of the ZnO NPs in a number of worldwide indus-
Furthermore, increases in the levels of H2AX phosphorylation and DNA tries and, specially, their potential benets in the medicine eld have
damage were observed in all cases. These increases were statistically brought attentions to their potential toxicity and health risks. It is well
signicant in all the conditions tested in the comet assay, except for the established that, under specic conditions, ZnO NPs may result toxic to
highest concentration after the 6 h exposure. However, statistically a variety of mammalian and human cells (De Berardis et al., 2010; Kim
signicant values of phosphorylated H2AX were obtained only at the et al., 2010; Osman et al., 2010; Sharma et al., 2009, 2012b; Wahab et
30 g/ml treatments. al., 2011) and to animals after intratracheal instillation (Fukui et al.,
Author's personal copy

96 V. Valdiglesias et al. / Environment International 55 (2013) 92100

a) Table 2
Results of cell cycle analysis in neuronal cells treated with ZnO NPs (%cells).
120
3H 6H 24H 48H Exposure time Concentration G0/G1 S-phase G2/M
100
3h Control 64.55 0.95 11.79 0.61 23.66 0.39
80 20 g/ml 54.98 1.09* 21.07 0.44* 23.96 1.44
% Viability

30 g/ml 60.19 1.22* 20.47 0.86* 19.33 2.07*


60 40 g/ml 61.25 0.51* 18.19 0.88* 20.56 1.29*
PC 69.77 0.67* 13.55 0.73 16.68 0.20*
40 6h Control 62.50 0.57 9.75 0.85 27.75 1.12
20 g/ml 50.63 0.61** 20.78 0.77** 28.59 0.77
20 30 g/ml 57.71 0.23** 32.50 3.39** 9.79 3.41**
40 g/ml 61.18 0.44 25.72 2.13** 13.10 2.33**
0 PC 65.10 1.43 15.92 0.27** 18.98 1.19**
0 10 20 30 40 50 60 70 80 90
-20 PC: positive control. *P b 0.05, **P b 0.01, signicant differences regarding the control.
ZnO NP concentration (g/ml)

b) lineage suggesting that ZnO NP toxicity is related to the proliferative po-


120 3H 6H 24H 48h
tential of the cell (Hanley et al., 2008). Our results also support that ZnO
100 NPs present an antiproliferative effect; this capacity led different authors
to indicate ZnO NPs as a promising antibacterial and anticancer agent
% Viability

80
(Premanathan et al., 2011).
60 The potential of the NPs to be taken up by the cells was evaluated by
40 means of ow cytometry. Surprisingly, no uptake of ZnO NPs was
observed in any case. These negative results in cellular uptake disagree
20 with a few previous studies in other different cell types which observed
0 internalization of these NPs either by scanning electron microscopy or by
0 10 20 30 40 50 60 70 80 90 ow cytometry (Sharma et al., 2011, 2012b). However, it should be
ZnO NP concentration (g/ml) highlighted that a number of studies on in vitro cytotoxic and/or
genotoxic effects of ZnO NPs did not report their uptake by the cells
Fig. 2. Viability of neuronal cells treated with ZnO NPs. Results from MTT assay (a) and NRU (Akhtar et al., 2012; Hsiao and Huang, 2011; Lin et al., 2009). It was
assay (b). Values inside the rectangle are statistically different from the corresponding previously shown that NPs suspended in supplemented medium can
control.
be covered by the proteins of the medium reducing their ability to be
taken up by the cells (Lesniak et al., 2010). Moreover other parameters,
2012), or after oral (Pasupuleti et al., 2012; Sharma et al., 2012b) or including the cell cycle phase or the NP size and shape, may inuence
inhalatory (Wang et al., 2010) administration. However, there is a lack the internalization rate of NPs in cells (Kim et al., 2011). Thus, it seems
of information on the effects of these NPs on neurons and on the nervous that the specic conditions employed in our experiments, especially
system, particularly those from humans. regarding the use of medium supplemented with FBS, are limiting in
In this study, the potential neurotoxic effects of ZnO NPs were someway the entry of ZnO NPs into the neuronal cells.
assessed in vitro in SHSY5Y cells. Several concentrations and exposure The potential cell cycle alterations and apoptosis induced by ZnO NPs
times were tested and a battery of cytotoxicity and genotoxicity method- were assessed by different ow cytometry methodologies. Results
ologies were employed with the aim of characterizing in depth the obtained from the cell cycle analysis show that the ZnO NPs induce
effects of ZnO NPs on human neuronal cells as well as attempting to important cell cycle alterations, including mitotic arrest, in SHSY5Y
understand the underlying action mechanisms. cells after both 3 h and 6 h treatments. Similar to these results, ZnO
Results obtained from MTT and NRU assays showed that ZnO NPs NPs were also recently found to induce cell cycle alterations and mitotic
diminished the viability of the neuronal cells in a dose-dependent man- arrest in neuronal RSC96 Schwann cells treated for 12 h (880 g/ml)
ner at all the treatment times. According to our results, a concentration- (Yin et al., 2012).
dependent decrease in cellular viability after ZnO NP exposure was pre- Likewise, results obtained from the annexin V/PI analysis showed
viously described for several cell types, including astrocytoma U87 (Lai et increases in the percentage of apoptosis induction following both 3 h
al., 2008), hepatocarcinoma HepG2 (Sharma et al., 2012b), lung carcino- and 6 h ZnO NP treatments. Under experimental conditions similar to
ma A549 (Fukui et al., 2012), and epidermal A431 (Sharma et al., 2009) the ones employed in this study, ZnO NPs were previously reported to
cells. There is also evidence that ZnO NPs are particularly cytotoxic to induce cell death in a variety of cell lines (Akhtar et al., 2012; Sharma
rapidly dividing cancer cells relative to quiescent cells of the same et al., 2012b), including cells from the nervous system such as neural
stem cells and glioma cells (Deng et al., 2009; Wahab et al., 2011; Yin
et al., 2012). On the basis of our data, this apoptosis was scarcely associ-
30
3h 6h ated with losses of the mitochondrial membrane potential, as shown by
*
% Cells with NPs

25 the slight effects observed in the MMP assessment. Therefore, our results
20 in apoptosis evaluation suggest that ZnO NPs induce death cell mainly by
a mitochondria-independent pathway. Some previous studies reported
15
mitochondrial alterations after ZnO NP treatments in other different ex-
*
10 perimental systems (De Berardis et al., 2010; Li et al., 2012; Sharma et al.,
5 2012b). Nonetheless, in the study by Sharma et al. (2012b) the NPs were
efciently taken up by the HepG2 cells, penetration of the NPs in the
0
LoVo cells was not evaluated in the work by De Berardis et al. (2010),
Control 20 30 40 PC
and Li et al. (2012) exposed rat mitochondrion directly to the ZnO
ZnO NP concentration (g/ml)
NPs. Thus there are substantial differences between these studies and
Fig. 3. Uptake of ZnO NPs by SHSY5Y cells as analyzed by ow cytometry. PC: positive the current one, so further investigation is required to conrm our
control. *P b 0.01, signicant difference with regard to the corresponding control. observation.
Author's personal copy

V. Valdiglesias et al. / Environment International 55 (2013) 92100 97

100 3h 6h
90

80

70

% LDH activity
60

50

40

30

20

10

0
PC
-10 0 5 10 15 20 25 30 35 40 45

ZnO NPs concentration (g/ml)

Fig. 4. Results of membrane integrity assessment (LDH assay) in SHSY5Y cells exposed to ZnO NPs.

Different kinds of ZnO NP-induced genotoxic damage were evaluated explain these effects. On one hand the ZnO NPs can potentially interact
by means of several approaches, i.e., MN test, H2AX analysis and comet with the cell membrane to cause the observed toxicity. In this study,
assay. Results obtained from all these biomarkers showed DNA damage the potential alterations in the membrane integrity caused by ZnO NP
(comet assay) induction by the ZnO NPs since the 3 h exposure, but exposure were assessed by LDH assay, but negative results were
only after the 6 h exposure an important dose-dependent MN induction obtained after both 3 and 6 h of treatment. However, we cannot rule
could be observed, which was accompanied by an increase in phosphor- out the possible interaction of NPs with some component of the cellular
ylation of the histone H2AX. It should be taken into account that the membrane without altering its integrity. Zhao et al. (2009) studied the
highest concentration of ZnO NPs used (40 g/ml) induced around 40% effect of ZnO NPs on ion channels and excitability of rat neurons. On
of cytotoxicity at 3 and 6 h, so this fact can be inuencing the non- the basis of their results, they suggested that ZnO NPs can enhance the
signicant results obtained with this concentration for histone phos- excitability of neurons by increasing the opening number of sodium
phorylation (at both treatment times) and comet assay (at 6 h). ZnO channels and delaying rectier potassium channels. These alterations
NPs were previously reported to induce genotoxicity in other different in ion channels may disturb the ionic homeostasis and consequently
cell types including primary keratinocytes (Sharma et al., 2011), bro- the physiological functions of neurons.
blasts (Chiang et al., 2012), epidermal A431 cells (Sharma et al., 2009), On the other hand, several previous studies concluded that ZnO NPs
lung A549 cells (Fukui et al., 2012), cervix HEp-2 carcinoma cells may induce cytotoxicity and genotoxicity by releasing Zn2+ ions. Deng
(Osman et al., 2010), and HepG2 cells (Sharma et al., 2012b). However, et al. (2009), after evaluating the cytotoxicity of ZnO NPs in mouse neural
studies evaluating the effects of these NPs on genetic material of neuro- stem cells at different levels (cell viability, apoptosis and necrosis),
nal cells are highly scarce. Particularly, Chiang et al. (2012) observed reported that ZnO NP toxicity mainly came from the Zn2+ ions dissolved
DNA damage after exposing rat primary neuronal cells to ZnO NPs, and in the culture medium or inside cells. Fukui et al. (2012) found cytotox-
Wahab et al. (2011) found results very parallel to ours in MN induction icity due to the increased levels of Zn+2 ions in vitro, in ZnO NP-treated
on human U87 glioma cells treated with different Zn nanostructures, at human lung carcinoma cells, and in vivo, in rat lung cells after
similar concentrations to those employed in the current study. intratracheal instillation of ZnO NPs. And also cytotoxic effects linked to
The NP potential to induce oxidative DNA damage was also evaluated the presence of high levels of Zn+2 ions in both cytosol and mitochondria
by means of comet assay modied with the enzyme OGG1. Results of broncho-alveolar lavage (BAL) cells and white blood cells were found
obtained show that ZnO NPs induced oxidative damage at both 3 and in rats exposed to ZnO NPs (Kao et al., 2012b). Furthermore, increases in
6 h treatments. These data agree with previous studies reporting in- intracellular levels of Zn+2 ions released from ZnO NPs were correlated
creased values of oxidative stress and ROS production after ZnO NP expo- with high levels of ROS and, consequently, with cell death (Fukui et al.,
sure in a variety of cell types (Akhtar et al., 2012; De Berardis et al., 2010;
Sharma et al., 2009) and in animal studies (Fukui et al., 2012; Hao and
Chen, 2012; Sharma et al., 2012b).
Table 4
As expounded so far, although no cellular uptake of ZnO NPs was Genotoxicity evaluation of ZnO NPs. Results of MN assay (a), comet assay (b) and H2AX
observed, we obtained induction of cytotoxic and genotoxic effects by analysis (c).
these NPs on neuronal cells. Two main reasons may be suggested to
3h 6h

(a) Increase in %MN 20 g/ml 0.72 0.78 8.85 2.71**


Table 3 30 g/ml 0.66 0.33 12.55 2.31**
Apoptotic cell rates (%) after ZnO NP exposure. 40 g/ml 0.30 0.59 12.06 1.11**
PC 1.92 0.73* 13.34 1.20**
3h 6h (b) Increase in %tDNA 20 g/ml 4.88 1.93** 3.23 0.79*
30 g/ml 5.37 0.57** 11.34 0.28**
Annexin-V MMP Annexin-V MMP
40 g/ml 4.29 0.53** 5.01 3.70
Control 4.71 0.27 4.93 0.33 6.94 0.34 5.03 0.52 PC 9.37 3.44** 7.71 1.29**
ZnO NP 20 g/ml 6.63 0.15** 7.57 0.24** 9.80 0.98** 3.52 0.48 (c) Increase in %H2AX 20 g/ml 1.81 0.53 1.19 0.58
30 g/ml 6.38 0.31** 6.91 0.72 8.17 0.37 7.68 1.74 30 g/ml 2.20 0.30* 3.87 0.82**
40 g/ml 6.33 0.21** 5.62 0.49 8.98 0.74 5.62 0.69 40 g/ml 0.07 0.42 1.39 1.29
PC 8.28 0.14** 6.69 1.09 10.27 0.41** 9.57 0.70** PC 38.23 4.20** 45.41 1.92**

PC: positive control. **P b 0.01, signicant differences regarding the control. PC: positive control. *P b 0.05, **P b 0.01, signicant differences regarding the control.
Author's personal copy

98 V. Valdiglesias et al. / Environment International 55 (2013) 92100

120
a) *
30 100
buffer OGG1
**

% Viability
25 80
20 ** 60
%tDNA

* **
15 * 40
**
10 20

5 0
0 0.05 0.1 0.2 0.4 0.6 0.8 PC
0 Zn+2 concentration (mM)
Control 20 30 40 PC
+2
ZnO NP concentration (g/ml) Fig. 7. Cytotoxicity of Zn ions: MTT assay in SHSY5Y cells treated with ZnSO4.
**P b 0.01, signicant difference with regard to the control.
b)
35
buffer OGG1 *
30
25
* means of FAAS. For all the times assayed (3, 6 and 24 h) linear increases
%tDNA

20 ** in ion concentrations were observed until 30 g/ml of NPs, and then a


15 plateau was reached at around 0.3 mM Zn2+. Other studies reporting
10 experiments of Zn2+ ion release from ZnO NP showed similar Zn2+ con-
5 centrations in Dulbecco's modied Eagle's medium (DMEM) (Reed et al.,
2012), but lower in seawater (Wong et al., 2010) or in complete minimal
0
Control 20 30 40 PC essential medium (CMEM) (Sharma et al., 2012b). But Reed et al. (2012)
demonstrated using nanopure water and different cell culture media
ZnO NP concentration (g/ml)
that solubility of ZnO NPs may be highly variable and dependent on
Fig. 5. Results of OGG1-modied comet assay in neuronal cells treated with ZnO NPs the matrix in which they are suspended.
for 3 h (a) or 6 h (b). PC: positive control. *P b 0.05, **P b 0.01, signicant difference When neuronal cells were treated with Zn2+ ions (from ZnSO4 salt) at
with regard to the corresponding buffer. a range of concentrations including the ones obtained as released from
ZnO NPs (0.050.8 mM), decrease in cell viability was only observed at
2012), agreeing with the positive results obtained in our study from doses higher than 0.4 mM. Therefore, as the NP concentrations used in
apoptosis and oxidative DNA damage assessments. Contrarily, Lin et al. this study released Zn2+ ions at levels up to 0.3 mM, the decrease in
(2009) concluded that neither free Zn2+ nor metal impurity in the ZnO neuronal cell viability observed after ZnO NP treatments cannot be attrib-
NP samples was the cause of the observed cytotoxicity and oxidative uted to the Zn2+ ions released from the NPs, but to the NPs themselves.
stress in A549 cells treated with these NPs. Regarding the brain, zinc is Other authors also suggest a size dependent toxicity, distinct from
an essential nutrient for normal neurological functions and may play adverse effects associated with the presence of dissolved ions, also re-
an important role in the processing of memory formation (Daniels et leased from bulk forms (Nair et al., 2009; Wong et al., 2010). Contrarily,
al., 2004). To understand the possible effects of this ion in brain cells is Franklin et al. (2007) found that for ZnO, there are no size related effects;
particularly relevant as transition metals (such as zinc) may contribute rather, the most likely impact of ZnO (nanoparticulate or bulk) results
to the etiology of neurodegenerative diseases, such as Alzheimer's from dissolution presumably as Zn2+.
disease, Parkinson's disease, amyotrophic lateral sclerosis, stroke or Still, as Zn 2+ ions were released from the ZnO NPs even at the
epilepsy (Pavlica et al., 2009). lowest concentrations, the possibility that these ions are involved in
In order to check the possibility that Zn+2 ions could be released from the genotoxic and cytotoxic effects observed in this study cannot be
the ZnO NPs to the cell culture medium, and so they could be involved in excluded since experiments aimed at testing the potential of Zn 2+
the cytotoxic and genotoxic effects observed, we tested the presence of ions to induce genotoxic damage, alterations in the cell cycle or apo-
free Zn2+ ions released from the NPs in our cell culture medium by ptosis on SHSY5Y cells were not performed.

0,40
3h 6h 24h
0,35
Zn2+ concentration (mM)

0,30

0,25

0,20

0,15

0,10

0,05

0,00
0 2 4 6 8 10 15 20 25 30 35 40 45 50 55
ZnO NP concentration (g/ml)

Fig. 6. Analysis of Zn+2 ions in cell culture medium released from ZnO NPs.
Author's personal copy

V. Valdiglesias et al. / Environment International 55 (2013) 92100 99

5. Conclusions Kim JA, berg C, Salvati A, Dawson KA. Role of cell cycle on the cellular uptake and
dilution of nanoparticles in a cell population. Nat Nanotechnol 2011;7:628.
Kozik MB, Maziarz L, Godlewski A. Morphological and histochemical changes occurring
Despite the fact that no cellular uptake was found, ZnO NPs induced in the brain of rats fed large doses of zinc oxide. Folia Histochem Cytochem
considerable cytotoxicity, including viability decrease, apoptosis and (Krakow) 1980;18:2016.
Lai JC, Lai MB, Jandhyam S, Dukhande VV, Bhushan A, Daniels CK, et al. Exposure to titanium
cell cycle alterations, and different kinds of genetic damage, including dioxide and other metallic oxide nanoparticles induces cytotoxicity on human neural
oxidative DNA damage, on human neuronal cells in a dose- and time- cells and broblasts. Int J Nanomedicine 2008;3(4):53345.
dependent manner. Free Zn 2+ ions released from the ZnO NPs were Lee CM, Jeong HJ, Kim DW, Sohn MH, Lim ST. The effect of uorination of zinc oxide
nanoparticles on evaluation of their biodistribution after oral administration.
not responsible for the viability decrease, but their role on other types
Nanotechnology 2012;23:205102.
of cell damage cannot be ruled out. The results obtained in this work Lesniak A, Campbell A, Monopoli MP, Lynch I, Salvati A, Dawson KA. Serum heat inac-
contribute to increase the knowledge on the genotoxic and cytotoxic tivation affects protein corona composition and nanoparticle uptake. Biomaterials
2010;31:95118.
potential of ZnO NPs in general, and specically on human neuronal
Li J, Liu X, Zhang Y, Tian F, Zhao G, Yu Q, et al. Toxicity of nano zinc oxide to mitochondria.
cells. Since neurotoxic effects of these NPs at different levels were dem- Toxicol Res 2012;1:137.
onstrated in this study, further investigations are needed to develop the Lin W, Xu Y, Huang C-C, Ma Y, Shannon KB, Chen D-R, et al. Toxicity of nano- and micro-sized
necessary safety precautions and exposure limits for people working ZnO particles in human lung epithelial cells. J Nanopart Res 2009;11:2539.
Mosmann T. Rapid colorimetric assay for cellular growth and survival, application to
with materials containing ZnO NPs and those who might be exposed proliferation and cytotoxicity assays. J Immunol Methods 1983;65:5563.
to them when they are implemented in commercial, industrial, and Nair S, Sasidharan A, Rani VVD, Menon D, Nair S, Manzoor K, et al. Role of size scale of
medical applications. ZnO nanoparticles and microparticles on toxicity toward bacteria and osteoblast
cancer cells. J Mater Sci Mater Med 2009;20:23541.
Nsse M, Beisker W, Kramer J, Miller BM, Schreiber GA, Viaggi S, et al. Measurement of
Acknowledgments micronuclei by ow cytometry. Methods Cell Biol 1994;42:14958.
Oberdrster G, Sharp Z, Atudorei V, Elder A, Gelein R, Kreyling W, et al. Inhal Toxicol
2004;16:437.
This work was funded by the European Commission (ERA NET New Osman IF, Baumgartner A, Cemeli E, Fletcher JN, Anderson D. Genotoxicity and cyto-
INDIGO Program, NanoLINEN 045-036-073, PIM2010ENI-00632) and toxicity of zinc oxide and titanium dioxide in HEp-2 cells. Nanomedicine (Lond)
by Xunta de Galicia (EM 2012/079). G. Kili was supported by a fellowship 2010;5:1193203.
Ostrovsky S, Kazimirsky G, Gedanken A, Brodie C. Selective cytotoxic effect of ZnO
from the University of A Corua. nanoparticles on glioma cells. Nano Res 2009;2:88290.
Pasupuleti S, Alapati S, Ganapathy S, Anumolu G, Pully NR, Prakhya BM. Toxicity of zinc
References oxide nanoparticles through oral route. Toxicol Ind Health 2012;28:67586.
Pavlica S, Gaunitz F, Gebhardt R. Comparative in vitro toxicity of seven zinc-salts to-
Akhtar MJ, Ahamed M, Kumar S, Khan MM, Ahmad J, Alrokayan SA. Zinc oxide wards neuronal PC12 cells. Toxicol in Vitro 2009;23:6539.
nanoparticles selectively induce apoptosis in human cancer cells through reactive Peters A, Veronesi B, Calderon-GarciduenasL Gehr P, Chen LC, Geiser M, Reed W, et al.
oxygen species. Int J Nanomedicine 2012;7:84557. Translocation and potential neurological effects of ne and ultrane particles a
Avlasevich SL, Bryce SM, Cairns SE, Dertinger SD. In vitro micronucleus scoring by ow critical update. Part Fibre Toxicol 2006;3:13.
cytometry: differential staining of micronuclei versus apoptotic and necrotic chro- Premanathan M, Karthikeyan K, Jeyasubramanian K, Manivannan G. Selective toxicity
matin enhances assay reliability. Environ Mol Mutagen 2006;47:5666. of ZnO nanoparticles toward Gram positive bacteria and cancer cells by apoptosis
Block ML, Wu X, Pei Z, Li G, Wang T, Qin L, et al. Nanometer size diesel exhaust particles through lipid peroxidation. Nanomedicine: NBM 2011;7:18492.
are selectively toxic to dopaminergic neurons: the role of microglia, phagocytosis, Qiang JL. The surface properties and photocatalytic activities of ZnO ultrane particles.
and NADPH oxidase. FASEB J 2004;18:1618. Appl Surf Sci 2001;180:30814.
Borenfreund E, Puerner JA. Toxicity determined in vitro by morphological alterations Reed RB, Ladner DA, Higgins CP, Westerhoff P, Ranville JF. Solubility of nano-zinc oxide
and neutral red absorption. Toxicol Lett 1985;24:11924. in environmentally and biologically important matrices. Environ Toxicol Chem
Chiang HM, Xia Q, Zou X, Wang C, Wang S, Miller BJ, et al. Nanoscale ZnO induces 2012;31:939.
cytotoxicity and DNA damage in human cell lines and rat primary neuronal cells. Roman D, Locher F, Suter W, Cordier A, Bobadilla M. Evaluation of a new procedure for
J Nanosci Nanotechnol 2012;12:212635. the ow cytometric analysis of in vitro, chemically induced micronuclei in V79
Costa S, Coelho P, Costa C, Silva S, Mayan O, Santos LS, et al. Genotoxic damage in pathology cells. Environ Mol Mutagen 1998;32:38796.
anatomy laboratory workers exposed to formaldehyde. Toxicology 2008;252:408. Seil JT, Webster TJ. Decreased astroglial cell adhesion and proliferation on zinc oxide
Daniels W, Hendricks J, Salie R, Rensburg S. A mechanism for zinc toxicity in neuroblas- nanoparticle polyurethane composites. Int J Nanomedicine 2008;3:52331.
toma cells. Metab Brain Dis 2004;19:7988. Sharma V, Shukla RK, Saxena N, Parmar D, Das M, Dhawan A. DNA damaging poten-
De Berardis B, Civitelli G, Condello M, Lista P, Pozzi R, Arancia G, et al. Exposure to ZnO tial of zinc oxide nanoparticles in human epidermal cells. Toxicol Lett 2009;185:2118.
nanoparticles induces oxidative stress and cytotoxicity in human colon carcinoma Sharma V, Singh SK, Anderson D, Tobin DJ, Dhawan A. Zinc oxide nanoparticle in-
cells. Toxicol Appl Pharmacol 2010;246:11627. duced genotoxicity in primary human epidermal keratinocytes. J Nanosci Nanotechnol
Deng X, Luan Q, Chen W, Wang Y, Wu M, Zhang H, et al. Nanosized zinc oxide particles 2011;11:37828.
induce neural stem cell apoptosis. Nanotechnology 2009;20:115101. Sharma V, Singh P, Pandey AK, Dhawan A. Induction of oxidative stress, DNA damage
Franklin N, Rogers N, Apte S, Batley G, Gadd G, Casey P. Comparative toxicity of and apoptosis in mouse liver after sub-acute oral exposure to zinc oxide
nanoparticulate ZnO, bulk ZnO, and ZnCl2 to a freshwater microalga (Pseudokirchneriella nanoparticles. Mutat Res 2012a;745:8491.
subcapitata): the importance of particle solubility. Environ Sci Technol 2007;41:848490. Sharma V, Anderson D, Dhawan A. Zinc oxide nanoparticles induce oxidative DNA
Fukui H, Horie M, Endoh S, Kato H, Fujita K, Nishio K, et al. Association of zinc ion release damage and ROS-triggered mitochondria mediated apoptosis in human liver cells
and oxidative stress induced by intratracheal instillation of ZnO nanoparticles to rat (HepG2). Apoptosis 2012b;17:85270.
lung. Chem Biol Interact 2012;198:2937. Singh NP, McCoy MT, Tice RR, Schneider EL. A simple technique for quantitation of low
Han D, Tian Y, Zhang T, Ren G, Yang Z. Nano-zinc oxide damages spatial cognition capability levels of DNA damage in individual cells. Exp Cell Res 1988;175:18491.
via over-enhanced long-term potentiation in hippocampus of Wistar rats. Int J Smith CC, O'Donovan MR, Martin EA. hOGG1 recognizes oxidative damage using the
Nanomedicine 2011;6:145361. comet assay with greater specicity than FPG or ENDOIII. Mutagenesis 2006;21:
Hanley C, Layne J, Punnoose A, Reddy KM, Coombs I, Coombs A, et al. Preferential killing 18590.
of cancer cells and activated human T cells using ZnO nanoparticles. Nanotechnol- Suzuki H, Toyooka T, Ibuki Y. Simple and easy method to evaluate uptake potential of
ogy 2008;19:295103. nanoparticles in mammalian cells using a ow cytometric light scatter analysis. En-
Hao L, Chen L. Oxidative stress responses in different organs of carp (Cyprinus carpio) viron Sci Technol 2007;41:301824.
with exposure to ZnO nanoparticles. Ecotoxicol Environ Saf 2012;80:10310. Tanaka T, Halicka D, Traganos F, Darzynkiewicz Z. Cytometric analysis of DNA damage,
Hsiao IL, Huang YJ. Effects of various physicochemical characteristics on the toxicities of phosphorylation of histone H2AX as a marker of DNA double-strand breaks (DSBs).
ZnO and TiO nanoparticles toward human lung epithelial cells. Sci Total Environ Methods Mol Biol 2009;523:1618.
2011;409(7):121928. Valdiglesias V, Laffon B, Psaro E, Mndez J. Okadaic acid induces morphological
Hu YL, Gao JQ. Potential neurotoxicity of nanoparticles. Int J Pharm 2010;394:11521. changes, apoptosis and cell cycle alterations in different human cell types. J Envi-
Jeng HA, Swanson J. Toxicity of metal oxide nanoparticles in mammalian cells. J Environ Sci ron Monit 2011a;13(6):183140.
Health A 2006;41:2699711. Valdiglesias V, Laffon B, Psaro E, Mndez J. Okadaic acid-induced genotoxicity in
Kao YY, Cheng TJ, Yang DM, Wang CT, Chiung YM, Liu PS. Demonstration of an olfactory human cells evaluated by micronucleus test and H2AX analysis. J Toxicol Environ
bulbbrain translocation pathway for ZnO nanoparticles in rodent cells in vitro and Health 2011b;74:98092.
in vivo. J Mol Neurosci 2012a;48:46471. Vandebriel RJ, De Jong WH. A review of mammalian toxicity of ZnO nanoparticles.
Kao YY, Chen YC, Cheng TJ, Chiung YM, Liu PS. Zinc oxide nanoparticles interfere with Nanotechnol Sci Appl 2012;5:6171.
zinc ion homeostasis to cause cytotoxicity. Toxicol Sci 2012b;125(2):46272. Wahab R, Kaushik NK, Verma AK, Mishra A, Hwang IH, Yang YB, et al. Fabrication and
Kim YH, Fazlollahi F, Kennedy IM, Yacobi NR, Hamm-Alvarez SF, Borok Z, et al. Alveolar growth mechanism of ZnO nanostructures and their cytotoxic effect on human
epithelial cell injury due to zinc oxide nanoparticle exposure. Am J Respir Crit Care brain tumor U87, cervical cancer HeLa, and normal HEK cells. J Biol Inorg Chem
Med 2010;182:1398409. 2011;16:43142.
Author's personal copy

100 V. Valdiglesias et al. / Environment International 55 (2013) 92100

Wang L, Wang L, Ding W, Zhang F. Acute toxicity of ferric oxide and zinc oxide Yin Y, Lin Q, Sun H, Chen D, Wu Q, Chen X, et al. Cytotoxic effects of ZnO hierarchical
nanoparticles in rats. J Nanosci Nanotechnol 2010;10:861724. architectures on RSC96 Schwann cells. Nanoscale Res Lett 2012;7:439.
Wong SW, Leung PT, Djurisi AB, Leung KM. Toxicities of nano zinc oxide to ve marine Zhao J, Xu L, Zhang T, Ren G, Yang Z. Inuences of nanoparticle zinc oxide on acutely
organisms: inuences of aggregate size and ion solubility. Anal Bioanal Chem isolated rat hippocampal CA3 pyramidal neurons. Neurotoxicology 2009;30:22030.
2010;396:60918.
Xiong D, Fang T, Yu L, Sima X, Zhu W. Effects of nano-scale TiO2, ZnO and their bulk
counterparts on zebrash: acute toxicity, oxidative stress and oxidative damage.
Sci Total Environ 2011;409:144452.

Vous aimerez peut-être aussi