Sie sind auf Seite 1von 14

Environmental and Molecular Mutagenesis 00:00^00 (2014)

Research Article

TiO2 Nanoparticles Induce DNA Double Strand Breaks


and Cell Cycle Arrest in Human Alveolar Cells
Krupa Kansara,1 Pal Patel,1 Darshini Shah,1 Ritesh K. Shukla,1
Sanjay Singh,1 Ashutosh Kumar,1* and Alok Dhawan1,2*
1
Institute of Life Sciences, Ahmedabad University, University Road,
Navrangpura, Ahmedabad, Gujarat, India
2
Nanomaterial Toxicology Group, CSIR-Indian Institute of Toxicology Research,
Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, India

TiO2 nanoparticles (NPs) have the second high- that TiO2 NPs can induce DNA damage and a
est global annual production (3000 tons) corresponding increase in micronucleus fre-
among the metal-containing NPs. These NPs are quency, as evident from the comet and
used as photocatalysts for bacterial disinfection, cytokinesis-block micronucleus assays. We dem-
and in various other consumer products includ- onstrate that DNA damage may be attributed to
ing sunscreen, food packaging, therapeutics, increased oxidative stress and ROS generation.
biosensors, surface cleaning agents, and others. Furthermore, genomic and proteomic analyses
Humans are exposed to these NPs during syn- showed increased expression of ATM, P53, and
thesis (laboratory), manufacture (industry), and CdC-2 and decreased expression of ATR,
use (consumer products, devices, medicines, H2AX, and Cyclin B1 in A549 cells, suggesting
etc.), as well as through environmental expo- induction of DNA double strand breaks. The
sures (disposal). Hence, there is great concern occurrence of double strand breaks was corre-
regarding the health effects caused by exposure lated with cell cycle arrest in G2/M phase.
to NPs and, in particular, to TiO2 NPs. In the Overall, the results indicate the potential for
present study, the genotoxic potential of TiO2 genotoxicity following exposure to these TiO2
NPs in A549 cells was examined, focusing on NPs, suggesting that use should be carefully
their potential to induce ROS, different types of monitored. Environ. Mol. Mutagen. 00:000
DNA damage, and cell cycle arrest. We show 000, 2014. V C 2014 Wiley Periodicals, Inc.

Key words: TiO2 NPs; genotoxicity; DNA double strand breaks; oxidative stress; micronucleus; cell cycle
arrest

INTRODUCTION Grant sponsor: UK India Education and Research Initiative (UKIERI)


standard award to Institute of Life Sciences, Ahmedabad University,
Metal oxide nanoparticles (NPs) are finding a variety India; Grant number: IND/CONT/E/1112/217.
of applications in catalysis, drug delivery, cosmetics, Grant sponsor: European Union Seventh Framework Programme (FP7/
clothing, energy conversion, water disinfection, lubrica- 20072013); Grant number: 263147.
tion, home appliances, and environmental remediation Grant sponsor: The Gujarat Institute for Chemical Technology (GICT).
[Aitken et al., 2004; Hoet et al., 2004a,b; Nohynek et al., *Correspondence to: Alok Dhawan, Institute of Life Sciences, School of Sci-
2008; Akimov and Koh, 2010; Kumar et al., 2011c; ence and Technology, Ahmedabad University, University Road, Navrangpura,
Kumar and Dhawan, 2013b]. TiO2 NPs have the second Ahmedabad 380009, Gujarat, India. E-mail: alok.dhawan@ahduni.edu.in (or)
Ashutosh Kumar, Institute of Life Sciences, School of Science and Technol-
highest annual production (estimated 3000 tons), with ogy, Ahmedabad University, University Road, Navrangpura, Ahmedabad
major applications as a photocatalyst for bacterial disin- 380009, Gujarat, India. E-mail: ashutosh.kumar@ahduni.edu.in
fection and as a component of sunscreens in the cosmet- Conflict of Interest: The authors declare that there is no conflict of
ics industry [Bondarenko et al., 2013]. TiO2 NPs are also interest.
used in products such as food packaging, therapeutics, Received 19 August 2014; provisionally accepted 19 October 2014; and
biosensors and surface cleaning agents, among others in final form 29 October 2014
[Shukla et al., 2013, 2014]. DOI 10.1002/em.21925
Worldwide use of TiO2 NPs has led to environmental Published online 00 Month 2014 in
release with reports of their presence as contaminants in Wiley Online Library (wileyonlinelibrary.com).

C 2014 Wiley Periodicals, Inc.


V
Environmental and Molecular Mutagenesis. DOI 10.1002/em
2 Kansara et al.

water, air, and soil [Klaine et al., 2008; Kumar et al. cells relies on a variety of cell cycle checkpoints and
2011b]. Humans are exposed to these NPs at various DNA repair pathways [Arenz et al., 2006; Cui et al.,
stages including: synthesis (laboratory), manufacture 2012; Prasad et al., 2013]. However, the type of DNA
(industry), use (consumer products, devices, medicines, damage and its correlation with the cell cycle arrest and
etc.) and through environmental exposure (through dis- apoptosis is not understood.
posal). The main routes exposure are inhalation, ingestion Therefore, in this study, we investigated the genotoxic
and dermal [Kumar and Dhawan, 2013a; Magdolenova potential of TiO2 NPs in A549 cells, including their
et al., 2014]. As the NPs are small in size and have high potential to induce ROS, different types of DNA damage
diffusion coefficients, they can migrate rapidly in the air and cell cycle arrest. The correlation between DNA dam-
[Aitken et al., 2008]. Therefore, inhalation is considered age and cell cycle arrest was investigated using genomics
to be the primary route of exposure by which humans are and proteomics approaches. A schematic representation of
exposed to TiO2 NPs suspended in air. In addition, expo- the study design is depicted in Figure 1.
sure of biota and humans to NPs is known to induce
adverse health effects including lung cancer, silicosis, and
autoimmune diseases [Donaldson, 2006; Fubini et al., MATERIALS AND METHODS
2011; Glista-Baker et al., 2014]. Hence, in this study, the Materials
potential toxicological effects of TiO2 NPs in the human
alveolar cell line A549 were assessed. Phosphate buffered saline (PBS; Ca21, Mg21 free), Dulbeccos
TiO2 NPs have been shown to induce cytotoxicity in Modified Eagles Medium (DMEM F12), fetal bovine serum (FBS), tita-
nium oxide 10% in water (CAS No. PL-TiO-10p-10g), low melting
organ specific cell lines. These effects have been attrib- point agarose, ethidium bromide (EtBr), 2,7-dichlorofluorescein diace-
uted to reactive oxygen species (ROS) generation, pulmo- tate, cell lysis buffer, protease inhibitor, cytochalasin B, ethyl methane-
nary inflammation, oxidative stress, and fibrosis [Borm sulfonate (EMS), sodium dodecyl sulfate, RNase from bovine pancreas,
et al., 2006; Nel, 2006; Singh et al., 2007; Jin et al., and bovine serum albumin, sodium chloride, 3-(4, 5-dimethylthiazoyl-
2008; Shukla et al., 2011b; Pfuhler et al., 2013]. Addi- 2-yl) 2,5-diphenyltetrazolium bromide (MTT), sodium citrate, normal
melting agarose, triton X-100, ethylenediaminetetraacetic acid diso-
tionally, it has been shown that the free radicals (O22 dium salt [(EDTA)Na2], trypsin-EDTA, L glutamine, tris buffer, antibi-
production) produced by TiO2 NPs interact with cellular otic/antimycotic solution (10,000 U/ml penicillin, and 10 mg/ml
components (nucleus, mitochondria, cytoplasm, etc.) and streptomycin and amphotericin-B) were purchased from Hi-Media,
induce damage by protein oxidation [Gajjar et al., 2009]. Mumbai, India. Tissue culture flasks (25 cm2, 75 cm2, and chamber
The DNA damaging potential of the TiO2 NPs and their slide), and black bottom 96 well plates were purchased from Corning,
India. Syringe filters (0.22 mm) and filter holders were purchased from
correlation with ROS and oxidative stress is also well Millipore, Mumbai, India. Syringes and needles were purchased from
documented [Shukla et al., 2011a, 2013]. Recently, Pra- Dispovan, India. End frosted conventional glass slides (75 3 25 mm2,
sad et al. [2013] demonstrated the genotoxic potential of with 19 mm frosted end) and cover slips (No. 1, 24 3 60 mm) were
TiO2 NPs in human BEAS-2B lung cells. These authors purchased from Hi-Media, Mumbai, India. Cytofunnel was purchased
observed that genotoxicity is independent of the amount from Thermo Scientific, India.
of agglomeration, cellular interaction, and cell-cycle alter-
ations [Prasad et al., 2013]. However, micronucleus for- METHODS
mation induced by TiO2 NPs was dependent on serum
concentration and the type of cell culture media used to Characterization of TiO2 NPs Using Dynamic
grow the BEAS-2B cells. Micronucleus frequency was Light Scattering (DLS)
highest in keratinocyte growth medium with 10% FBS
TiO2 NPs at a concentration of 100 mg/ml were suspended in MilliQ
concentration. This was attributed to the concentration of water and complete DMEM F-12 medium; 1 ml of TiO2 NPs suspension
FBS, as the protein present in the FBS induces the forma- was transferred into a disposable polystyrene cuvette to determine the
tion of NP protein corona and reduces the formation of size and zeta potential by DLS and phase analysis light scattering,
agglomeration. This also leads to higher internalization respectively, using a Zetasizer Nano-ZS equipped with 4.0 mW, 633nm
laser (Model ZEN3600, Malvern instruments, Malvern, UK). The stabil-
and subsequent interaction of NPs with cellular compo-
ity of NPs in the culture medium was also monitored up to 72 hr.
nents [Prasad et al., 2013, 2014]. In contrast, it has also
been reported that TiO2 NP dispersion with large agglom-
Cell Culture and Exposure to NPs
erates (3 min sonication and no serum in stock solution)
induced DNA damage in three cell lines (TK6-human The human lung alveolar carcinoma cell line (A549) was obtained
lymphoblast cells, EUE-human embryonic epithelial cells from the National Centre for Cell Sciences, Pune, India, and cultured in
and Cos-1-monkey kidney fibroblasts), while the TiO2 DMEM-F12 supplemented with 10% FBS, 0.2% sodium bicarbonate,
and 10 ml/l antibiotic and antimycotic solution at 37 C under a humidi-
NPs dispersed with agglomerates less than 200 nm (foetal fied atmosphere of 5% CO2.
serum in stock solution and sonication 15 min) had no A stock suspension of TiO2 NPs (1 mg/ml) in DMEM-F12 was
effect on genotoxicity [Magdolenova et al., 2012]. It is diluted to concentrations ranging from 1 to 200 mg/ml, respectively.
also known that the genotoxicity induced by TiO2 NPs in A549 cells were exposed at varying concentrations for each experiment.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 3

Fig. 1. Schematic showing the study design. [Color figure can be viewed in the online issue, which is available at
wileyonlinelibrary.com.]

Subsequent experiments involving: cytotoxicity assays; genotoxicity troscopy. The results were assessed by measuring the absorbance of end
assays (comet assay, micronucleus induction); oxidative stress (ROS product at their respective wavelengths using a SYNERGY-HT multi-
generation); cell cycle; gene expression (polymerase chain reaction); well plate reader (Bio-Tek, USA) using Gen5 software.
proteomics (immunoblot assay) and apoptosis markers (Annexin-V-PI)
were conducted and analyzed. For different assays, 96-, 12-, 6-well cell
culture plates and 75 cm2 cell culture flasks were used having a treat- Genotoxicity Assessment
ment volume of 0.1, 1.2, 3, and 24 ml, respectively. The volume of the
The genotoxic potential of TiO2 NPs was assessed by comet and
treatments were chosen to ensure that the concentration of NPs per cm2
cytokinesis-block micronucleus (CBMN) assays. The treatment scheme
area was equivalent in 6, 12, 96 well plates and 75 cm2 flask for all
was the same for both assays. Approximately 3 3 105 cells in 3 ml of
assays. For different plates and flasks, the concentration was equivalent
DMEM-F12 were seeded in a 6-well cell culture plate. After 24 hr, the
to 7.89 lg/cm2 for 25 lg/ml; 15.78 lg/cm2 for 50 lg/ml; 23.68 lg/cm2
cells were exposed to TiO2 NPs (25, 50, 75, and 100 mg/ml) for 6 h.
for 75 lg/ml and 31.57 lg/cm2 for 100 lg/ml.
EMS at a concentration of 2 and 4 mM was used as a positive control
for the comet and CBMN assay, respectively.
Cytotoxicity Assays
Cytotoxicity assessment of TiO2 NPs was determined by MTT and Comet assay for Detection of DNA Damage
neutral red uptake (NRU) assays. These assays were carried out accord-
ing to the method of Mosmann (1983) and Borenfreund and Puerner Briefly, cells were harvested and slides were prepared by the method
(1985), respectively, with slight modification as described in our earlier described by Singh et al. (1988) and modified by Bajpayee et al. (2005).
study [Shukla et al., 2011a, 2013]. Briefly, cells (1 3 104 cells/well) in Slides were kept overnight at 4 C in lysis solution (2.5 M NaCl,
100 ml of culture medium were seeded in 96-well plates and kept for 24 100 mM EDTA, 10 mMTris (pH10), 1% Triton X-100 added prior to
hr. Cells were then exposed to different concentrations (1 200 mg/ml) lysis step). After lysis, the slides were placed in a horizontal gel electro-
of TiO2 NPs for 6, 24 and 48 hr. phoresis tank containing fresh and chilled electrophoresis solution
NP interference with the assay reagents was also checked using a cell (1 mM EDTA and 300 mM NaOH, pH >13) for 20 min of alkaline
free system, where NPs alone were incubated with the assay reagents, unwinding of DNA, followed by electrophoresis at 0.7 V/cm and 300
including dye and buffers, and the absorption was monitored by spec- mA at 4 C for 30 min. Neutralization was done using Tris buffer
Environmental and Molecular Mutagenesis. DOI 10.1002/em
4 Kansara et al.

(400 mM, pH 7.4) and slides were stained with 20 mg/ml EtBr and tion, cells were lysed in 1ml PBS containing 0.2% triton X 100 at 4 C
stored at 4 C in a humidified slide box until scoring. for 30 min. Cells were pelleted by centrifugation and re-suspended in
Scoring was done at a final magnification of 4003 on a microscope 500 ml of PBS containing 20 ml RNAse (10 mg/ml) for 30 min at 37 C.
with a fluorescent attachment (DM 2500, Leica,Wetzlar, Germany) and Finally, cells were stained with 10 ml propidium iodide (1 mg/ml) in
using Komet 6.0 software, which was provided with the image analysis 500 ml PBS for 1015 min at 4 C and analyzed on FACS (FACS Cali-
system (Andor Technology Belfast, U.K.). The comet parameters meas- bur, BD Biosciences, CA). A histogram of count vs. intensity was made
ured were tail DNA (%) and Olive Tail Moment (OTM). Analysis of 50 to calculate ratio of cells under Go/G1 (2n), S (2n1), G2/M phase (4n),
comets (25 from replicate slides) was carried out for each experiment and under apoptosis (2n-).
according to Tice et al. (2000).
Apoptosis Assay Using Annexin V Binding Assay
CBMN Assay Cells actively undergoing apoptosis were identified by staining with
The CBMN assay was carried out according to the method described fluorescein isothiocyanate-conjugated (FITC)-Annexin V and PI accord-
by Fenech (2000). After 6 hr of exposure, the treatment of TiO2 NPs ing to manufacturers protocol (BD Biosciences, San Jose, CA). Briefly,
was aspirated, cells were washed with PBS and grown further for 16 hr 2 3 105 cells/well were seeded in a 6-well culture plate for 24 hr prior
in complete medium containing Cytochalasin-B at final concentration 3 to treatment. Cells were treated with TiO2 NPs at different concentra-
mg/ml in 5% CO2 incubator. The cells were harvested and slides were tions (0, 25, 50, 75 and 100 mg/ml) for 24 and 48 hr; 1 mM camptothe-
prepared by centrifugation using a Cytospin (Thermo Shandon, Hamp- cin was used as a positive control in this assay. After removal of
shire, UK); 300 ml of cell suspension was loaded in each block of cyto- treatment, cells were harvested and washed twice with PBS, resuspended
funnel and centrifuged at 250g for 1 min. Two slides (two dots on one in 0.1 ml binding buffer containing 5 ml of FITC-Annexin V and PI and
slide from each replicate culture) were prepared for each concentration. kept at room temperature in the dark. After 10 min of incubation, 0.4 ml
The slides were fixed in chilled methanol (90%) for 5 min, air-dried and of binding buffer was added to each sample and analyzed using flow
stored until staining. The slides were stained with 10% Giemsa dissolved cytometer.
in Sorensons buffer and examined for the presence of micronuclei in
binucleate cells using a bright field microscope (DM 2500, Leica, Wet- Genomics Studies
zlar, Germany) at 10003 magnification. Two thousand binucleate cells
from each concentration (1000 binucleate cells from each slide, 500 cells
RNA Extraction and cDNA Synthesis
per dot) were scored. Total RNA was extracted from the control and TiO2 NP-treated (25,
50, and 100 mg/ml) A549 cells using TRIzol reagent (Life Technolo-
Micronucleus Assay Using Flow Cytometry gies). cDNA was synthesized using a High-Capacity cDNA Reverse
Transcription Kit (Applied Biosystems). For RT, the 20 ml reaction mix-
The micronucleus assay using flow cytometry was carried out by the ture contained 10 ml 23 master mix [23 RT buffer (2 ml), 0.8 mM
modified method of Nusse and Kramer [1984]. After 6 hr of exposure, dNTP Mix (0.8 ml), 23 random primers (2 ml), RNase inhibitor (1 ml),
cells were harvested and centrifuged at 1200 rpm for 10 min. Cells were one liter MultiScribe reverse transcriptase (RT) and 3.2 ml nuclease-free
washed in 1 3 PBS and transferred to 1 ml of solution I containing H2O] and an equal volume of diluted RNA sample. The RT reaction
sodium chloride (584 mg/l), sodium citrate (1000 mg/l), RNase (10 mg/ was carried out in a thermal cycler consisting of one cycle at 25 C for
l), EtBr (25 mg/l), Igepal (0.3 ml/l) and incubated for 1 hr at room tem- 10 min, 50 C for 15 min, 85 C for 5 min, and 4 C on hold.
perature; 1 ml of solution II containing citric acid (15 mg/L), sucrose
(0.25 M), EtBr (40 mg/L) was then added to same tube and the samples RT-PCR Analysis
were analyzed for the presence of micronuclei in the cells using flow
cytometry (FACS Calibur, BD Biosciences, CA). RT-PCR was performed in a 25 ll volume containing 100 ng cDNA,
100 lM dNTPs, 2.5 mM MgCl2, 2.5 ll PCR buffer, 1 unit of DNA
polymerase (Thermo Scientific) and 1 lM gene specific (ATM, ATR,
Intracellular ROS Measurement P53, H2AX, CDC2, and CyclinB1) forward and reverse primers (Table
I). Thirty cycles of PCR amplification were performed and the intensity
Intracellular ROS generation was estimated by the method of Wan
of the amplified product was analyzed after electrophoresis using the
et al. [1993] modified by Shukla et al. (2011b) using 2, 7-
ImageQuantLAS 500 software (GE Healthcare Bio-Sciences AB,
dichlorofluorescein diacetate (DCFDA) dye. Briefly, cells (1 3 105cells/
Sweden).
well) were seeded in a 12-well plate. After 24 hr, the cells were exposed
to TiO2 NPs for 3, 6, 24, and 48 hr. The TiO2 NP-containing media was
aspirated and the cells were washed twice with PBS. Thereafter, culture
Immunoblot Analysis
medium containing DCFDA dye (20 mM) was added to each well. The
plate was incubated for 30 min at 37 C and the medium containing A549 cells were treated with TiO2 NPs at concentrations of 25, 50,
DCFDA was discarded; 200 ml of PBS was then added to each well and and 100 mg/ml for 24 hr. After removal of treatment, cells were har-
fluorescence intensity was measured using flow cytometer at excitation vested and lysed in lysis buffer (150 mM NaCl, 1% NP-40, 0.5%
and emission wavelengths of 485 and 528 nm, respectively. In addition, sodium deoxycholate, 0.1% SDS, 50 mMTrisHCl, pH 8) containing
qualitative analysis of ROS generation was done using a microscope protease inhibitor cocktail (Sigma). Protein concentration was esti-
with fluorescence attachment (Model DM 2500 Leica, Wetzlar, mated using Bradfords method [Bradford, 1976]. Protein (30 mg) from
Germany). control and treated groups was separated on Glycine-SDS-
polyacrylamide gel (8%) and transferred to a nitrocellulose membrane
Cell Cycle Progression Analysis by electroblotting. The membrane was incubated with primary antibod-
ies specific for Hsp70, HMOX1, and b-actin (Abcam). Secondary anti-
A549 cells (1 3 105) were seeded on 6-well plate and treated with body incubation was done and protein bands were detected using
different TiO2 NP concentrations (25100 mg/ml) for 24 hr. After the chemiluminescence and densitometry analysis was carried out using
treatment, medium was aspirated into individual tubes and cells were ImageQuantLAS 500 software (GE Healthcare Bio-Sciences AB,
harvested and fixed in 70% ethanol (at 220 C for 30 min). After fixa- Sweden).
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 5

TABLE I. Primers for Different Genes


Gene Forward primer Reverse primer
0 0
ATM 5 -TGCCAGACAGCCGTGACTTAC-3 5 -ACCTCCACCTGCTCATACACAAG-30
0

ATR 50 -TGCAGTAATGTCAATGGTTGG-30 50 -CTGGAACTTCAAAGGTTTCTCC-30


p53 50 -TCAGATCCTAGCGTCGAGCCC-30 50 -GGGTGTGGAATCAACCCACAG-30
p21 50 -GTTCCTTGTGGAGCCGGAGC-30 50 -GGTACAAGACAGTGACAGGTC-30
CdC2 50 -TGGGGTCAGCTCGTTACTCA-30 50 -CACTTCTGGCCACACTTCATTTA-30
Cyclin B1 50 -CAGTCAGACCAAAATACCTACTGGGT-30 50 -ACACCAACCAGCTGCAGCATCTTCTT-30
PCNA 50 -GCCCTCAAAGACCTCATCAA-30 50 -TCTGGGATTCCAAGTTGCTC-30
H2AX 50 -AGCAAACTCAACTCGGCAAT-30 50 -ACTCCCCAATGCCTAAGGTT-30
B actin 50 -CGGCATCGTCACCAACTG-30 50 -AACATGATCTGGGTCATCTTCTC-30
GAPDH 50 -CAGGAGGCATTGCTGATGAT-30 50 -GAAGGCTGGGGCTCATTT-30

TABLE II. Characterization of TiO2 NPs in Different to 100% of control) at 150 and 200 mg/ml of TiO2 NPs
Dispersant and Stability in Complete DMEM F12 exposure after 48 hr. However, after 6 and 24 hr expo-
Medium Using DLS sure, no significant cytotoxicity was observed (Fig. 2A).
In the NR uptake assay, a statistically significant
Hydrodynamic Zeta
Dispersant size (d.nm) potential (mV) (P < 0.05) reduction in neutral red uptake was observed
down to 67, 54, and 8% (relative to 100% of control) at
Milli Q 106.7 6 8.0 213.0 6 0.9 the three higher concentrations (100, 150, and 200 mg/ml)
DMEM-F12 (10% FBS) after 0 hr 23.28 6 2.0 210.1 6 1.0
of TiO2 NPs, respectively, after 48 hr exposure (Fig. 2B).
DMEM-F12 (10% FBS) after 24 hr 22.54 6 2.4 212.6 6 1.2
DMEM-F12 (10% FBS) after 72 hr 23.97 6 1.6 211.9 6 1.1 However, a reduction in percent cell viability was also
observed after 6 and 24 hr exposure, but it was not statis-
Values represent mean 6 standard error of three experiments. tically significant.

Statistical Analysis Genotoxic Potential of TiO2 NPs


Results from each experiment were expressed as mean 6 SEM of
DNA Damage
three individual experiments and data were analyzed using one way A significant (P < 0.05) concentration-dependent
analysis of variance (ANOVA). The post hoc comparisons of mean of
independent groups were done by Dunnetts test at statistically signifi-
increase in DNA damage was observed in A549 cells at
cant values (P < 0.05). different TiO2 NP concentrations (75 and 100 lg/ml) after
6 hr exposure, as evident by the comet assay parameters,
namely, OTM and % Tail DNA respectively (Table III).
RESULTS
Micronucleus Induction
Size and Zeta Potential Analysis of TiO2 NPs Using DLS
CBMN Assay
The average hydrodynamic size and zeta potential of
TiO2 NPs in culture medium (DMEM F12) supplemented Human alveolar cells (A549) showed a statistically sig-
with 10% FBS were 23.28 6 2.0 nm and 210.1 6 1.0 mV, nificant (P < 0.05) increase in micronucleus formation after
respectively. However, in MilliQ water the size and zeta 6 hr exposure to 75 and 100 lg/ml TiO2 NPs (12.33 and
potential was 106.7 6 8.0 nm and 213 6 0.9 mV, respec- 14.66 MN/1000 BNCs) compared with control (6 MN/
tively (Table II). The particles were also found to be stable 1000 BNCs; Table III). Using flow cytometry, we observed
up to 72 hr as measured by their size and zeta potential a significant increase (200 and 500%) in micronucleus for-
(Table II). Additionally, the primary particle size as meas- mation in A549 cells following exposure to 75 and 100
ured by transmission electron microscopy and surface area mg/ml of TiO2 NPs relative to control cells (Fig. 3).
of TiO2 NPs by BrunauerEmmettTeller method (as
described in the technical sheet by the supplier) was 4 Measurement of Intracellular ROS
8 nm 1215 m2/g, respectively. The particles were 99.7%
A significant qualitative and quantitative increase in %
pure and had an anatase crystal structure.
ROS generation was observed in A549 cells after 6 hr
exposure to TiO2 NPs. The flow cytometric analysis
Cytotoxicity
showed a concentration-dependent significant (P < 0.05)
Cytotoxicity of TiO2 NPs was assessed in human alve- increase in % ROS generation based on an increase in the
olar cells (A549) for 6, 24, and 48 hr, respectively. In the fluorescence intensity of DCFDA dye following treatment
MTT assay, the mitochondrial succinate dehydrogenase with TiO2 NPs (115 and 128% at 75 and 100 mg/ml,
enzymatic activity was reduced to 68 and 64% (relative respectively; Fig. 4).
Environmental and Molecular Mutagenesis. DOI 10.1002/em
6 Kansara et al.

Fig. 2. A significant concentration- and time-dependent increase in cytotoxicity of TiO2 NPs in A549 cells. A: % MTT
reduction. B: % Neutral red uptake. The viability of the control cells was considered as 100%. The data are expressed
as mean 6 SEM from three independent experiments. *P < 0.05, when compared with control. [Color figure can be
viewed in the online issue, which is available at wileyonlinelibrary.com.]

Apoptosis
TABLE III. Genotoxic Potential of TiO2 NPs in A549 Cells as Our data demonstrate a significant (P < 0.05)
Assessed by the Micronucleus and Comet Assay concentration-dependent increase (23, 74, 189, and 249%)
in the apoptotic A549 cells population after 24 hr of TiO2
No. of Micronucleus
in 1000 binucleated NPs exposure (25, 50, 70, and 100 mg/ml; Fig. 5). How-
Groups cells Tail DNA (%) ever, after 48 hr of treatment, cells were necrotic.
Control 6.00 6 2.80 4.48 6 0.11 Cell Cycle Analysis
TiO2 NPs (25 mg/ml) 7.33 6 1.20 5.14 6 0.12
TiO2 NPs (50 mg/ml) 9.66 6 2.84 6.06 6 0.15 Our data demonstrate that TiO2 NPs caused a signifi-
TiO2 NPs (75 mg/ml) 12.33 6 2.96a 8.25 6 0.24a cant increase in the proportion of cells that are arrested in
TiO2 NPs (100 mg/ml) 14.66 6 2.33a 9.49 6 0.25a
Positive Control- EMS(4 mM) 16.66 6 3.66a 48.2 6 1.14a
G2/M phase in a concentration-dependent manner after
24 hr exposure. More cells exposed to TiO2 NPs were
a
P < 0.05 when compared with control using one way ANOVA. arrested in G2/M phase compared to control at the 50,
Values represent mean 6 S.E. of three experiments. 75, and 100 mg/ml concentrations, respectively (Fig. 6).
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 7

Fig. 3. A: Flow cytometric analysis of A549 cells after exposure to dif- sent mean 6 S.E. of three experiments for each concentration. *P < 0.05,
ferent concentrations of TiO2 NPs showed a concentration-dependent compared with control. [Color figure can be viewed in the online issue,
increase in micronucleus formation compared to controls. B: Bar graph which is available at wileyonlinelibrary.com.]
representing the percent increase in micronucleus formation. Values repre-

Expression of DNA Damage Responsive Genes


2 gene increased by 1.24, 1.42, and 1.25 fold respectively
Expression levels of DNA damage responsive genes over control cells at the 100 mg/ml concentration. How-
were analysed in A549 cells after 24 hr exposure to dif- ever, expression levels of ATR, H2AX and Cyclin B1
ferent concentrations (25, 50, and 100 mg/ml) of TiO2 decreased by 0.67, 0.21 and 0.24 fold, respectively, com-
NPs by RT-PCR. The expression of ATM, P53, and CdC- pared with controls at 100 mg/ml (Fig. 7).
Environmental and Molecular Mutagenesis. DOI 10.1002/em
8 Kansara et al.

Fig. 4. Significant concentration-dependent increase in intracellular ROS after 6 hr exposure. Values represents mean 6 S.E. of three experiments
using DCFDA dye. A: Control cells. BF: Cells treated with TiO2 NPs (*P < 0.05). [Color figure can be viewed in the online issue, which is
(5100 lg/ml). (Magnification X-400). G: Bar graph representing the available at wileyonlinelibrary.com.]
increase in intracellular ROS generation as detected by flow cytometry

Immunoblot Analysis
cant increase (P < 0.05) in the expression of the stress
Immunoblot analysis of A549 cells exposed to TiO2 proteins HSP70 (20, 40, and 52%) and HMOX-1 (13, 24,
NPs (25, 50, and 100 mg/ml) for 24 hr showed a signifi- and 32%), as shown in Figure 8.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 9

independent test systems along with a parallel set of cell-


free systems [Sharma et al., 2009, 2012; Shukla et al,.
2011b]. A range of TiO2 NPs concentrations (1200 mg/
ml) for different time points (6, 24 and 48 hr) were used
for the cytotoxicity experiments in this study. MTT and
NR uptake assays are based on two different mechanisms.
MTT represents mitochondrial activity and NR provides
information on the lysosomal activity of the cells. It was
observed that TiO2 NPs cause no significant cytotoxicity
at concentrations ranging from 1 to 200 lg/ml after 6 and
24 hr of exposure in A549 cells. Hence, these non-
Fig. 5. Bar graph showing percent apoptotic cells. Camptothecin (1 lM) cytotoxic concentrations and time points were used for
was used as positive control. Data represent means 6 SEM of three inde-
pendent experiments.*P < 0.05, when compared with control. [Color fig-
genotoxicity and mechanistic studies (ROS, cell cycle and
ure can be viewed in the online issue, which is available at expression of DNA damage responsive genes/proteins).
wileyonlinelibrary.com.] However, mild cytotoxic responses were observed after
48 hr of exposure in MTT assay (150 and 200 lg/ml) and
DISCUSSION NR uptake assay (100, 150, and 200 lg/ml). Therefore,
these concentrations were used for annexin V binding
This study reveals the genotoxic potential of TiO2 NPs assay to demonstrate apoptosis. Our data demonstrate that
in human alveolar (A549 cells) cells and provides insight TiO2 NPs induce apoptosis in A549 cells after 48 hr of
into the possible mechanism by which TiO2 NPs exert exposure.
their genotoxic effects in these cells. Our results demon- Oxidative stress is the most prevalent mechanism pro-
strate that TiO2 NPs induce DNA damage leading to cell posed in the literature to explain the toxicity of NPs
cycle arrest in A549 cells through the ATM/ATR path- [Murray et al., 2009; Zhong et al., 2010; Srinivas et al.,
way (Fig. 9). 2012; Kumar et al., 2013]. The Hsp family of proteins is
Characterization of NPs is the most essential part of the first tier of cellular defence and induces the protein
nano-toxicological studies. The behavior and activity of folding response to minimize protein degradation and cel-
NPs is largely dependent on a number of physical and lular stress [Beere et al., 2000]. Our study indicates that
chemical properties such as, particle number, mass con- TiO2 NPs induce stress in A549 cells as evident by an
centration, surface area, charge, chemistry and reactivity, induction of Hsp 70. Additionally, ROS generation is
size distribution, aggregation, elemental composition, as hypothesized as the probable mechanism involved in the
well as structure and shape [Dhawan and Sharma, 2010]. toxicity of NPs [Gurr, 2005; Long et al., 2006; Nel, 2006;
Therefore, a complete characterization is essential for bet- Kumar et al., 2011a; Dey et al., 2012]. This has been
ter understanding and interpretation of results. In this attributed to the small size and hence large surface area
study, characterization of TiO2 NPs was done in MilliQ of NPs, which is generally thought to produce ROS and
and complete medium (DMEM-F12) using DLS. Our data oxidative stress [Borm et al., 2006]. In the present study,
demonstrate that the TiO2 NPs used in the study were in TiO2 NPs were found to generate intracellular ROS when
nano form. Also, the stability of the NPs was monitored examined by the cell permeable dye DCFH-DA using
up to 72 h, as NPs have a tendency to agglomerate in flow cytometer and fluorescence microscope. A significant
aqueous medium [Dusinska et al., 2009; Gurbani et al., induction in % ROS generation in the form of fluores-
2011]. Our data demonstrate that the TiO2 NPs were sta- cence was observed in A549 cells exposed to TiO2 NPs
ble in the DMEM-F12; however, the hydrodynamic size (25, 50, and 75 mg/ml) for 6 h using fluorescence micro-
of NPs increased in MilliQ over time. This can be attrib- scope (Figs. 4A4F). Also, a significant (P < 0.05) dose-
uted to the protein coating over the NPs, or the formation dependent increase in % ROS generation was observed
of corona, as the coating reduces the tendency of by flow cytometry, in the form of increased FITC fluores-
agglomeration [Lynch and Dawson, 2008; Elsaesser and cence intensity following treatment with TiO2 NPs (115
Howard, 2011]. Additionally, the particle-plus-corona and 128% at 75 and 100 mg/ml; Fig. 4G). This observa-
plays an important role in the biological fate of NPs as it tion is consistent with earlier studies where TiO2 was
enhances the internalization into the cells. shown to produce ROS even without photoactivation
The interference of the NPs (due to their unique physi- [Shukla et al., 2011a,b, 2013, 2014]. These free radicals
cal and chemical properties) with dyes that are commonly can oxidize and reduce macromolecules (DNA, lipid, and
being used for cytotoxicity test systems is well docu- protein) in the cellular system, resulting in significant oxi-
mented [Dhawan and Sharma, 2010; Dhawan et al., dative damage to cells.
2011]. Therefore, it has been suggested that findings from Free radicals can also damage DNA, which can lead to
cytotoxicity assays should be validated by two or more genetic instability and consequently, carcinogenesis or
Environmental and Molecular Mutagenesis. DOI 10.1002/em
10 Kansara et al.

Fig. 6. TiO2 NP-induced alterations in the cell cycle of A549 cells. A: Controls. BE: Cells treated with different con-
centration of TiO2 NPs. F: Cells treated with 4 mM EMS as positive control. [Color figure can be viewed in the online
issue, which is available at wileyonlinelibrary.com.]

cell death [Luo et al., 2011]. To elucidate the genotoxic double strand breaks. However, increase in micronucleus
potential of TiO2 NPs in A549 cells, the alkaline comet frequency indicate an abundance of DNA double strand
assay was performed. We found that TiO2 NPs induce breaks [Han et al., 2010].
DNA damage in A549 cells using the comet assay. This Earlier, Kitchin et al. [2011] demonstrated that ROS
supports ROS generation as the mediator of TiO2 NP- and radical-induced DNA damage is clustered and yields
induced DNA damage. Furthermore, a statistically signifi- DNA double strand breaks that can activate the ATM-
cant (P < 0.05) dose-dependent increase in MN frequency Chk2 signalling pathway [Kitchin et al., 2011]. ATM
was observed at TiO2 NP concentrations of 25, 50, 75, belongs to a family of kinases that have sequence homol-
and 100 lg/ml, which further confirms the genotoxicity ogy to phosphoinositide 3-kinase (PI3K). The phospho-
of the TiO2 NPs. The comet assay measures single and inositide 3-kinase related kinases (PIKKs) are the key
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 11

Fig. 7. Densitometry analysis for the expression of ATM, ATR, P53, cH2AX, CdC2, Cyclin B1, and b-actin in A549
cells, at exposure concentrations of 25, 50, and 100 mg/ml TiO2 NPs by RT-PCR. [Color figure can be viewed in the
online issue, which is available at wileyonlinelibrary.com.]

components of the checkpoint signalling network, repre-


sented by Ataxia-telangiectasia mutated (ATM), ATM
and RAD3-related, and DNA-dependent protein kinase
[Shiloh, 2003]. These protein kinases play an important
role in the cell signaling pathway involved in response to
DNA double strand break in higher eukaryotes [Shiloh,
2003]. In our study, a dose-dependent increase in the
expression of ATM was observed after 24 hr of TiO2 NPs
exposure, which suggests an increase in double strand
breaks. On the contrary, a dose-dependent decrease in the
expression of ATR indicates no single strand breaks in
DNA induced by TiO2 NPs in A549 cells. ATR is a ser-
ine/threonine-specific protein kinase that is involved in
sensing DNA damage and is activated in response to per-
Fig. 8. Immunoblot analysis of proteins (HSP70 and HMOX1) of A549
cells treated with different concentrations (25, 50, and 100 lg/ml) of sistent single-stranded DNA, which is a common interme-
TiO2 NPs. [Color figure can be viewed in the online issue, which is avail- diate formed during DNA damage detection and repair
able at wileyonlinelibrary.com.] [Horne et al., 1996; Shiloh, 2003]. Moreover, our
Environmental and Molecular Mutagenesis. DOI 10.1002/em
12 Kansara et al.

Fig. 9. A schematic showing possible mechanisms of TiO2 NP-induced double strand DNA breaks and cell cycle arrest.
[Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

observations for double strand DNA breaks were also the maturation-promoting factor, which is predominantly
confirmed by decreased expression of cH2AX, which is a expressed during the G2/M phase of the cell cycle and
sensitive biomarker of DNA double strand breaks [Pod- promotes cell division [Horne et al., 1996; Zimmermann
horecka et al., 2010]. H2AX is one of several genes cod- et al., 2012]. We observed a dose-dependent decrease in
ing for histones; it is a major component of nucleosome- the expression of Cyclin B1, which indirectly provides
formation and the structure of DNA. H2AX is phospho- support to our cell cycle finding that TiO2 NP exposure
rylated on serine 139, and then called gamma-H2AX, in arrests the cells in the G2/M phase. The correlation
the presence of DNA double strand breaks [Flores-Perez between the DNA damage and cell cycle arrest induced
et al., 2014]. Because of this modification, the DNA by TiO2 NPs in A549 cells is shown in Figure 8.
becomes less condensed, potentially allowing space for
the recruitment of proteins necessary during the repair of
double strand breaks. Our data revealed a significant CONCLUSIONS
decrease in the expression of the cH2AX gene, indicating
that the double strand breaks induced by TiO2 NPs were In the present study we investigated the effects of TiO2
not repairable at the higher concentration. However, at NPs in a human lung alveolar carcinoma cell line (A549)
lower concentrations the expression of cH2AX was on and explored the different mechanisms underlying their
higher side (relative to controls), suggesting that this cytotoxic and genotoxic potential. Comet and CBMN
damage can be repaired by the different repair enzymes. assays demonstrated that TiO2 NPs can induce DNA
These findings suggest that exposure to TiO2 NPs acti- damage and a corresponding increase in micronucleus fre-
vates the ATM/Chk2 DNA damage signalling pathway in quency. The DNA damage induced by TiO2 NPs may be
A549 cells. attributed to increased oxidative stress and ROS genera-
Finally, we observed a significant dose-dependent tion. Furthermore, genomic and proteomic changes were
increase in the expression of CdC2, with concomitant consistent with the induction of DNA double strand
decreases in the expression of Cyclin B1 [Hinds et al., breaks. The occurrence of double strand breaks was cor-
1992]. Cyclin B1 is a regulatory protein involved in mito- related with cell cycle arrest in G2/M phase. Our results
sis. This gene product binds with P34 (Cdk1) and forms provide valuable insights into the mechanism of TiO2
Environmental and Molecular Mutagenesis. DOI 10.1002/em
DNA Double Strand Breaks and Cell Cycle Arrest 13

NPs induced toxicity in human lung alveolar cells. These impairs DNA damage response and sensitizes human breast can-
observations once again reiterate concern over the safety cer cells to cisplatin therapy. Cancer Biol Ther 15:777788.
Fubini B, Fenoglio I, Tomatis M, Turci F. 2011. Effect of chemical com-
of TiO2 NPs in consumer products.
position and state of the surface on the toxic response to high
aspect ratio nanomaterials. Nanomedicine (Lond) 6:899920.
REFERENCES Gajjar P, Pettee B, Britt DW, Huang W, Johnson WP, Anderson AJ.
2009. Antimicrobial activities of commercial nanoparticles
Aitken RJ, Koopman P, Lewis SE. 2004. Seeds of concern. Nature 432: against an environmental soil microbe, Pseudomonas putida
4852. KT2440. J Biol Eng 3:9.
Aitken RJ, Hankin SM, Lang Tran C, Donaldson K, Stone V, Cumpson Glista-Baker EE, Taylor AJ, Sayers BC, Thompson EA, Bonner JC.
P, Johnstone J, Chaudhry Q, Cash S, Garrod J. 2008. A multidis- 2014. Nickel nanoparticles cause exaggerated lung and airway
ciplinary approach to the identification of reference materials for remodeling in mice lacking the T-box transcription factor,
engineered nanoparticle toxicology. Nanotoxicology 2:7178. TBX21 (T-bet). Part Fibre Toxicol 11:7.
Akimov YA, Koh WS. 2010. Resonant and nonresonant plasmonic nano-
Gurbani D, Shukla RK, Pandey AK, Dhawan A. 2011. Stable metal
particle enhancement for thin-film silicon solar cells. Nanotech-
oxide nanoparticle formulation for toxicity studies. J Biomed
nology 21:235201.
Nanotechnol 7:104105.
Arenz A, Hellweg CE, Stojicic N, Baumstark-Khan C, Grotheer HH.
Gurr JR WSSA, Chen CH, Jan KY. 2005. Ultrafine titanium dioxide par-
2006. Gene expression modulation in A549 human lung cells in
ticles in the absence of photoactivation can induce oxidative
response to combustion-generated nano-sized particles. Ann N Y
damage to human bronchial cells. Toxicology 213:6673.
Acad Sci 170183.
Han X, Corson N, Wade-Mercer P, Gelein R, Jiang J, Sahu M, Biswas
Bajpayee M, Pandey AK, Parmar D, Dhawan A. 2005. Current status of
P, Finkelstein JN, Elder A, Oberdorster G. 2010. Assessing the
short-term tests for evaluation of genotoxicity, mutagenicity, and
relevance of in vitro studies in nanotoxicology by examining cor-
carcinogenicity of environmental chemicals and NCEs. Toxicol
relations between in vitro and in vivo data. Toxicology 297:19.
Mech Methods 15:155180.
Hinds PW, Mittnacht S, Dulic V, Arnold A, Reed SI, Weinberg RA.
Beere HM, Wolf BB, Cain K, Mosser DD, Mahboubi A, Kuwana T,
1992. Regulation of retinoblastoma protein functions by ectopic
Tailor P, Morimoto RI, Cohen GM, Green DR. 2000. Heat-shock
expression of human cyclins. Cell 70:9931006.
protein 70 inhibits apoptosis by preventing recruitment of
Hoet PH, Bruske-Hohlfeld I, Salata OV. 2004a. Nanoparticles - known
procaspase-9 to the Apaf-1 apoptosome. Nat Cell Biol 2:469
and unknown health risks. J Nanobiotechnol 2:12.
475.
Bondarenko O, Juganson K, Ivask A, Kasemets K, Mortimer M, Kahru Hoet PH, Nemmar A, Nemery B. 2004b. Health impact of nanomateri-
A. 2013. Toxicity of Ag, CuO and ZnO nanoparticles to selected als? Nat Biotechnol 22:19.
environmentally relevant test organisms and mammalian cells in Horne MC, Goolsby GL, Donaldson KL, Tran D, Neubauer M, Wahl
vitro: A critical review. Arch Toxicol 87:1181200. AF. 1996. Cyclin G1 and cyclin G2 comprise a new family of
Borenfreund E, Puerner E. 1985. Toxicity determined in vitro by mor- cyclins with contrasting tissue-specific and cell cycle-regulated
phological alterations and neutral red absorption. Toxicol Lett expression. J Biol Chem 271:60506061.
24:119124. Jin C, Bai L, Wu H, Liu J, Guo Z, Chen J. 2008. Paclitaxel-loaded pol-
Borm PJ, Robbins D, Haubold S, Kuhlbusch T, Fissan H, Donaldson K, y(D,L-lactide-co-glycolide) nanoparticles for radiotherapy in
Schins R, Stone V, Kreyling W, Lademann J, et al. 2006. The hypoxic human tumor cells in vitro. Cancer Biol Therapy 7:729
potential risks of nanomaterials: a review carried out for ECE- 734.
TOC. Part Fibre Toxicol 3:11. Kitchin KT, Prasad RY, Wallace K. 2011. Oxidative stress studies of six
Cui Y, Liu H, Ze Y, Zengli Z, Hu Y, Cheng Z, Cheng J, Hu R, Gao G, TiO(2) and two CeO(2) nanomaterials: immuno-spin trapping
Wang L, et al. 2012. Gene expression in liver injury caused by results with DNA. Nanotoxicology 5:546556.
long-term exposure to titanium dioxide nanoparticles in mice. Klaine SJ, Alvarez PJ, Batley GE, Fernandes TF, Handy RD, Lyon DY,
Toxicol Sci 128:171185. Mahendra S, McLaughlin MJ, Lead JR. 2008. Nanomaterials in
Dey KK, Kumar A, Shanker R, Dhawan A, Wan M, Yadav RR, the environment: Behavior, fate, bioavailability, and effects.
Srivastava AK. 2012. Growth morphologies, phase formation, Environ Toxicol Chem 27:18251851.
optical & biological responses of nanostructures of CuO and their Kumar A, Dhawan A. 2013a. Genotoxic and carcinogenic potential of
application as cooling fluid in high energy density devices. RSC engineered nanoparticles: an update. Arch Toxicol 87:18831900.
Adv 2:13871403. Kumar A, Dhawan A. 2013b. Nano-safety, Standardization and Certifica-
Dhawan A, Sharma V. 2010. Toxicity assessment of nanomaterials: tion. In Manual on Critical Issues in Nanotechnology R&D Man-
Methods and challanges. Anal Bioanal Chemy 398:589605. agement, An Asia-Pacific Perspective 140.
Dhawan A, Shanker R, Das M, Gupta KC. 2011. Guidance for safe han- Kumar A, Pandey AK, Singh SS, Shanker R, Dhawan A. 2011a. Cellular
dling of nanomaterials. J Biomed Nanotechnol 7:218224. response to metal oxide nanoparticles in bacteria. J Biomed
Donaldson K. 2006. Resolving the nanoparticles paradox. Nanomedicine Nanotechnol 7:102103.
(Lond) 1:229234. Kumar A, Pandey AK, Singh SS, Shanker R, Dhawan A. 2011b. Cellu-
Dusinska M, Dusinska M, Fjellsbo L, Magdolenova Z, Rinna A, Runden lar uptake and mutagenic potential of metal oxide nanoparticles
Pran E, Bartonova A, Heimstad E, Harju M, Tran L, et al. 2009. in bacterial cells. Chemosphere 83:11241132.
Testing strategies for the safety of nanoparticles used in medical Kumar A, Pandey AK, Singh SS, Shanker R, Dhawan A. 2011c. Engi-
applications. Nanomedicine (Lond) 4:605607. neered ZnO and TiO(2) nanoparticles induce oxidative stress and
Elsaesser A, Howard CV. 2011. Toxicology of nanoparticles. Adv Drug DNA damage leading to reduced viability of Escherichia coli.
Delivery Rev 64:129137. Free Radic Biol Med 51:18721881.
Fenech M. 2000. The in vitro micronucleus technique. Mutat Res 455: Kumar A, Sharma V, Dhawan A. 2013. Methods for detection of oxida-
8195. tive stress and genotoxicity of engineered nanoparticles. Methods
Flores-Perez A, Rafaelli LE, Ramirez-Torres N, Arechaga-Ocampo E, Mol Biol 1028:231246.
Frias S, Sanchez S, Marchat LA, Hidalgo-Miranda A, Quintanar- Long TC, Saleh N, Tilton RD, Lowry GV, Veronesi B. 2006. Titanium
Jurado V, Rodriguez-Cuevas S, et al. 2014. RAD50 targeting dioxide (P25) produces reactive oxygen species in immortalized
Environmental and Molecular Mutagenesis. DOI 10.1002/em
14 Kansara et al.

brain microglia (BV2): Implications for nanoparticle neurotoxic- Sharma V, Shukla RK, Saxena N, Parmar D, Das M, Dhawan A. 2009.
ity. Environ Sci Technol 40:43464352. DNA damaging potential of zinc oxide nanoparticles in human
Luo C, Urgard E, Vooder T, Metspalu A. 2011. The role of COX-2 and epidermal cells. Toxicol Lett 185:211218.
Nrf2/ARE in anti-inflammation and antioxidative stress: Aging Sharma V, Anderson D, Dhawan A. 2012. Zinc oxide nanoparticles
and anti-aging. Med Hypotheses 77(2):174178. induce oxidative DNA damage and ROS-triggered mitochondria
Lynch I, Dawson KA. 2008. Protein-nanoparticle interactions. Nano mediated apoptosis in human liver cells (HepG2). Apoptosis 17:
Today 3:4047. 852870.
Magdolenova Z, Bilanicova D, Pojana G, Fjellsbo LM, Hudecova A, Shiloh Y. 2003. ATM and related protein kinases: Safeguarding genome
Hasplova K, Marcomini A, Dusinska M. 2012. Impact of integrity. Nat Rev Cancer 3:155168.
agglomeration and different dispersions of titanium dioxide nano- Shukla RK, Kumar A, Pandey AK, Singh SS, Dhawan A. 2011a. Tita-
particles on the human related in vitro cytotoxicity and genotox- nium dioxide nanoparticles induce oxidative stress-mediated apo-
icity. J Environ Monit 14:455464. ptosis in human keratinocyte cells. J Biomed Nanotechnol 7:100
Magdolenova Z, Collins A, Kumar A, Dhawan A, Stone V, Dusinska M. 101.
2014. Mechanisms of genotoxicity. A review of in vitro and in Shukla RK, Sharma V, Pandey AK, Singh S, Sultana S, Dhawan A.
vivo studies with engineered nanoparticles. Nanotoxicology 8: 2011b. ROS-mediated genotoxicity induced by titanium dioxide
233278. nanoparticles in human epidermal cells. Toxicol In Vitro 25:231
Mosmann T. 1983. Rapid colorimetric assay for cellular growth and sur- 241.
vival: application to proliferation and cytotoxicity assay. Shukla RK, Kumar A, Gurbani D, Pandey AK, Singh S, Dhawan A.
J Immunol Methods 65:5563. 2013. TiO(2) nanoparticles induce oxidative DNA damage and
Murray AR, Kisin E, Leonard SS, Young SH, Kommineni C, Kagan apoptosis in human liver cells. Nanotoxicology 7:4860.
VE, Castranova V, Shvedova AA. 2009. Oxidative stress and Shukla RK, Kumar A, Vallabani NV, Pandey AK, Dhawan A. 2014.
inflammatory response in dermal toxicity of single-walled carbon Titanium dioxide nanoparticle-induced oxidative stress triggers
nanotubes. Toxicology 257:161171. DNA damage and hepatic injury in mice. Nanomedicine (Lond)
Nel A, Xia T, Madler L, Li N. 2006. Toxic potential of materials at the 9:14231434.
nanolevel. Science 311:622627. Singh NP, McCoy MT, Tice RR, Schneider EL. 1988. A Simple Tech-
Nohynek GJ, Dufour EK, Roberts MS. 2008. Nanotechnology, cosmetics nique for quantitation of low levels of DNA damage in individual
and the skin: Is there a health risk? Skin Pharmacol Physiol 21: cells. Exp Cell Res 175:184191.
136149. Singh P, Prasuhn D, Yeh RM, Destito G, Rae CS, Osborn K, Finn MG,
Nusse M, Kramer J. 1984. Flow cytometric analysis of micronuclei Manchester M. 2007. Bio-distribution, toxicity and pathology of
found in cells after irradiation. Cytometry 5:2025. cowpea mosaic virus nanoparticles in vivo. J Control Release
Pfuhler S, Elespuru R, Aardema MJ, Doak SH, Maria Donner E, Honma 120:4150.
M, Kirsch-Volders M, Landsiedel R, Manjanatha M, Singer T, Srinivas A, Rao PJ, Selvam G, Goparaju A, Murthy PB, Reddy PN.
et al. 2013. Genotoxicity of nanomaterials: Rrefining strategies 2012. Oxidative stress and inflammatory responses of rat follow-
and tests for hazard identification. Environ Mol Mutagen 54:229 ing acute inhalation exposure to iron oxide nanoparticles. Hum
239. Exp Toxicol 31:11131131.
Podhorecka M, Skladanowski A, Bozko P. 2010. H2AX phosphoryla- Wan CP, Myung E, Lau BH. 1993. An automated micro-fluorometric
tion: Its role in DNA damage response and cancer therapy. assay for monitoring oxidative burst activity of phagocytes.
J Nucleic Acids 2010. J Immunol Methods 159:131138.
Prasad RY, Chastain PD, Nikolaishvili-Feinberg N, Smeester L, Zhong CY, Zhou YM, Smith KR, Kennedy IM, Chen CY, Aust AE,
Kaufmann WK, Fry RC. 2013. Titanium dioxide nanoparticles Pinkerton KE. 2010. Oxidative injury in the lungs of neonatal
activate the ATM-Chk2 DNA damage response in human dermal rats following short-term exposure to ultrafine iron and soot par-
fibroblasts. Nanotoxicology 7:11111119. ticles. J Toxicol Environ Health A 73:837847.
Prasad RY, Simmons SO, Killius MG, Zucker RM, Kligerman AD, Zimmermann M, Arachchige-Don AS, Donaldson MS, Dallapiazza RF,
Blackman CF, Fry RC, Demarini DM. 2014. Cellular interactions Cowan CE, Horne MC. 2012. Elevated cyclin G2 expression
and biological responses to titanium dioxide nanoparticles in intersects with DNA damage checkpoint signaling and is required
HepG2 and BEAS-2B cells: Role of cell culture media. Environ for a potent G2/M checkpoint arrest response to doxorubicin.
Mol Mutagen 55:336342. J Biol Chem 287:2283822853.

Das könnte Ihnen auch gefallen