You are on page 1of 12

http://informahealthcare.

com/dct
ISSN: 0148-0545 (print), 1525-6014 (electronic)

Drug Chem Toxicol, Early Online: 1–12


! 2013 Informa Healthcare USA, Inc. DOI: 10.3109/01480545.2013.866134

Effects of sub-acute exposure to TiO2, ZnO and Al2O3 nanoparticles on


oxidative stress and histological changes in mouse liver and brain
Rupal Shrivastava*, Saimah Raza*, Abhishek Yadav, Pramod Kushwaha, and Swaran J. S. Flora
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

Division of Regulatory Toxicology, Defence Research and Development Establishment, Gwalior, India

Abstract Keywords
Nanomaterials are at the leading edge of the rapidly developing field of nanotechnology. Al2O3, morphological changes, metal oxides
However the information regarding toxicity of these nanoparticles on humans and environ- nanoparticles, nanotoxicity, oxidative
ment is still deficient. The present study investigated the toxic effects of three metal oxide stress, TiO2, ZnO
nanoparticles, TiO2, ZnO and Al2O3 on mouse erythrocytes, brain and liver. Male mice were
administered a single oral dose of 500 mg/kg of each nanoparticles for 21 consecutive days. The History
results suggest that exposure to these nano metallic particles produced a significant oxidative
stress in erythrocyte, liver and brain as evident from enhanced levels of Reactive Oxygen Received 11 March 2013
Species (ROS) and altered antioxidant enzymes activities. A significant increase in dopamine Revised 8 October 2013
and norepinephrine levels in brain cerebral cortex and increased brain oxidative stress suggest Accepted 10 November 2013
neurotoxic potential of these nanoparticles. Transmission electron microscopic (TEM) analysis Published online 17 December 2013
indicated the presence of these nanoparticles inside the cytoplasm and nucleus. These changes
were also supported by the inhibition of CuZnSOD and MnSOD, considered as important
biomarkers of oxidative stress. The toxic effects produced by these nanoparticles were more
For personal use only.

pronounced in the case of zinc oxide, followed by aluminum oxide and titanium dioxide,
respectively. The present results further suggest the involvement of oxidative stress as one of
the main mechanisms involved in nanoparticles induced toxic manifestations.

Introduction to oxidative stress. There are toxicological evidences that


ultrafine nanoparticles are capable of producing adverse
Nanoparticles (NPs) are currently having a widespread use in
effects (Englert, 2004; Oberdörster, 2000; Utell & Frampton,
modern technology, however there is insufficient data for
2000). A well defined mechanism of toxicity induced by NPs
their possible health and environmental hazards in human.
is not completely clear, however, several possible mechanisms
The rising concern for nanoparticles emerges because of their
have been proposed based on reported results. The change in
unique chemistry, extremely small size, large reactive surfaces
the physicochemical and structural properties of NPs may be
and non-biodegradability. They rapidly get dispersed through-
responsible for the interactions that could lead to toxico-
out the environment with unknown consequences (Wang
logical effects (Brown et al., 2001). These changes could lead
et al., 2009). Their adverse effects in human have raised
to specific surface groups (e.g. thiol groups, carboxyl groups,
serious concerns. It is therefore critical to understand the
carbonyl groups, SDS, DMSO, surfactants) to be reactive sites
nature and origin of the toxicity of these nanoparticles.
(Hussain et al., 2009). These reactive sites act as electron-
Metallic NPs are closely related to the generation of reactive
donors or electron-acceptors and work with oxygen to induce
oxygen species (ROS) and reactive nitrogen species (RNS)
superoxide radical (O2), which can then generate reactive
due to redox-cycling reactions. Currently, ROS and oxidative
oxygen species (ROS) by dismutation or the Fenton reaction
stress induced by NPs have been one of the best-developed
(Cabiscol et al., 2000).
paradigms for nanoparticle toxicity (Dick et al., 2003; Xia
Various metal oxide nanoparticles, such as, titanium
et al., 2006). Oxidative stress is a normal cellular process
dioxide (TiO2), aluminum oxide (Al2O3), and zinc oxide
involved in many aspects of cellular signaling, though
(ZnO) are receiving increasing attention for a large variety of
excessive oxidative stress can be harmful (Huang et al.,
applications due to their unique physical and chemical
2010). Recent studies have increased our understanding of the
properties (Huang et al., 2010; Nel et al., 2006; Schrand
nanotoxicity of metal oxide particles, particularly with respect
et al., 2010). TiO2 NPs possess inherent advantages of
physical stability, anticorrosion and nanoscale-enhanced
photocatalysis due to which they are widely used in paints,
*These authors contributed equally to the study.
printing ink, rubber, paper, cosmetics, sunscreens, car mater-
Address for correspondence: Swaran J. S. Flora, Division of Regulatory ials, cleaning air products, industrial photocatalytic processes,
Toxicology, Defence Research and Development Establishment, Jhansi
Road, Gwalior 474 002, India. Fax: +91 751 2341148; E-mail: and decomposing organic matters in wastewater (Iavicoli
sjsflora@hotmail.com or sjsflora@drde.drdo.in et al., 2012). Titanium dioxide and zinc oxide NPs are used in
2 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12

toothpaste, beauty products, sunscreens and textiles. While BDH chemicals (Mumbai, India). Triple distilled water
Aluminum oxide possess good dielectric and abrasive proper- prepared by Millipore (New Delhi, India) was used through-
ties and are widely used as an abrasive agent or insulator (Lai out the experiment to avoid contamination, and for the
et al., 2008; Prabhakar et al., 2012). TiO2 is known to get preparation of reagents and buffers used for various biochem-
accumulated in liver, kidney, and spleen with the related ical assays in the study. According to the information
toxicity (Schrand et al., 2010). There is also concern about provided by manufacturer the particles size of TiO2, ZnO
possible neurotoxicity, as it is known that TiO2 NPs may pass and Al2O3 nanoparticles are 575 nm, 5100 nm and 45 nm
through the blood brain barrier on exposure (Li et al., 2010). respectively. According to the company’s data sheet, TiO2
Several studies have shown that TiO2 NPs could be nanoparticles obtained is a mixture of rutile and anatase
translocated into the central nervous system (CNS) via the nanoparticles, in the form of suspension. Prior to dosing the
olfactory pathway in mice and accumulate in the entire brain, suspension was sonicated for 30 minutes.
mainly in the hippocampus regions (Wang et al., 2007; Wang
et al., 2008). However, to verify the generalization of this Characterization of nanoparticles by SEM
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

phenomenon, further studies are needed in other animal


The particle size and distribution were measured using
species. Titanium dioxide (TiO2) NPs injected in the abdom-
electron microscopic studies by FEI Quanta-400, SEM.
inal cavity of mice for 14 days significantly increased LPO,
Samples were coated with a thin layer of gold produced by
decreased GSH levels and altered antioxidant enzyme activity
ion sputtering in Ion Sputter JFC-1100 before observing these
in a dose-dependent manner along with histopathological
in the electron microscope at 5 kV.
changes in (Ma et al., 2010) Previous reports also indicate that
exposure to TiO2 NPs through various routes lead to enhanced
Animals and treatment
oxidative stress and also alteration of neurotransmitters in brain
(Wang et al., 2008; Shimizu et al., 2009). Adverse effects of Experiments were performed on healthy 6 weeks old Swiss
ZnO NPs can be mainly attributed to the generation of ROS albino male mice, weighing approximately 25–30 g. Animals
leading to membrane damage, directly or indirectly (Jingxia were obtained from the animal house facility of the Defence
et al., 2009). However little is known about the neurotoxic Research and Development Establishment (DRDE), Gwalior.
potential of zinc and the main mechanism involved. Prabhakar All animals received humane care in compliance with the
et al. (2012) have shown the possible involvement of oxidative guidelines of the Committee for the Purpose of Control and
For personal use only.

stress and altered antioxidant status in eliciting toxicity of Supervision of Experiments on Animals (CPCSEA). The
Al2O3 NPs after acute oral treatment. There is evidence that Animal Ethical Committee of DRDE, Gwalior, India also
exposure to aluminum may lead to an increased oxidative approved the protocols for the experiments. Prior to dosing,
stress, inflammatory events, and/or the breakdown of the they were acclimatized for 7 days to light from 06:00 to
blood-brain barrier (BBB) (Lockman et al., 2004). Due to their 18:00 h, alternating with 12 h darkness. The animals were
extremely small size, nanoparticles can translocate from these housed in stainless steel cages in an air-conditioned room
entry portals into the more sensitive circulatory and lymphatic with temperature maintained at 25  2  C. Mice were allowed
systems, and ultimately to body tissues and organs (Borm et al., standard chow diet (Amrut feeds, Pranav Agro, New Delhi,
2006). There are also reports which show their accumulation in India; metal content of diet, in ppm dry weight: Cu 10.0, Zn
different organs leading to systemic toxicity (van der Zande 45.0, Mn 55.0, Co 5.0, Fe 75.0) throughout the experiment
et al., 2012). and water ad libitum.
The study was designed to elucidate the sub-acute toxicity Thirty two mice were randomized into four groups of
of oral exposure to metal NPs TiO2, Al2O3 and ZnO 8 animals each and were treated as follows for 21 days:
nanoparticles in terms of oxidative stress and antioxidant Group I: Control (received normal water)
defense system in erythrocytes and soft tissues of mice. Most Group II: TiO2 nanoparticles (500 mg/kg b.w.) oral, once,
of the toxic chemicals are metabolized in liver due to which daily
there is a high risk of free radical attack leading to lipid Group III: ZnO nanoparticles (500 mg/kg b.w.) oral, once,
peroxidation, which in turn leads to hepatotoxicity (Patlolla daily
et al., 2011). High metabolic rate and poor antioxidant Group IV: Al2O3 nanoparticles (500 mg/kg b.w.) oral, once,
defense system renders brain highly vulnerable to oxidative daily
stress. The ability of NPs to cross the blood–brain barrier We selected oral route of exposure for mouse as these
further enhances the risk (Ma et al., 2010). Hence, liver and nanoparticles are generally being used in various products like
brain were selected for studying the oxidative stress-inducing food packaging, cosmetics and coating etc and there is
potential of TiO2, ZnO and Al2O3 NPs. possibility of gaining a direct entry into the body. They may
also enter into the gastrointestinal tract after their accidental
Materials and methods release into the environment. The doses at which humans
may get exposed to these nanoparticles at the above
Chemicals and reagents
mentioned scenario are not clear as no guidelines and
Titanium dioxide (TiO2) and Zinc oxide (ZnO) nanoparticles standard methodologies for in vivo toxicity assessment of
were procured from Sigma Aldrich (St. Louis, MO). Alumina these nanoparticles. We thus chose to work with the highest
(Al2O3) was procured from Alfa Aesar (Ward Hill, MA). All dose suggested by OECD guidelines for a new compound.
other laboratory chemicals and reagents were purchased from This dose does not necessarily reflect the actual concentration
Merck (Darmstadt, Germany), Sigma (St. Louis, MO) or of these nanoparticles in the real environment however, this
DOI: 10.3109/01480545.2013.866134 Metal oxides nanoparticles induced toxicity 3

can be used to assess the health risks from exposure to these corresponded to 44 xg RNA per ml. The purity of RNA was
nanoparticles (Sharma et al., 2012). determined by checking A260/A280 ratio, where a value in
After 21 days the exposure was stopped and animals were the range of 1.9–2.1 indicated the extraction of pure RNA.
sacrificed under light ether anesthesia, 48 h, after last dosing. Integrity of RNA isolated was checked using formamide gel
Blood was collected in heparinized vials. Liver and brain electrophoresis. Firstly, 10X MOPS buffer was prepared
were collected and stored in RNA later for gene expression (0.4 M MOPS was mixed with 0.1 M NaOAc and 10 mM
analysis (RT-PCR). For biochemical estimation, the tissues EDTA, pH of the buffer was adjusted to 7.0 with conc.
were washed with cold normal saline, blotted and all the NaOH). To prepare formamide gel (1.2%) agarose was
extraneous materials were removed. Liver and brain tissues prepared and 1X MOPS buffer and formaldehyde was further
from two animals were processed for the TEM analysis. added to it to cast the gel.
Whole brain of four animals was used for the biochemical
assays while the brain of remaining four animals was excised RT-PCR (reverse transcriptase PCR)
quickly and cerebral cortex was used for the estimation of Once the purity of RNA had been confirmed, the same was
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

neurotransmitter under cold conditions. used for the preparation of cDNA by means of reverse
transcription followed by specific amplification using Qiagen
Separation of red blood cells
One Step RT-PCR Kit. For this purpose, template RNA,
Blood was collected from orbital plexus of mice in specific primer solutions, dNTP mix, 5X Qiagen OneStep RT-
heparinized tubes. RBCs were isolated following the PCR Buffer, and RNase-free water was thawed and placed on
method of Steck and Kant (1974). Briefly, blood was ice. As per the reaction volume, a master mix was made and
centrifuged at 1500  g for 10 min at 4  C. Plasma and its appropriate volumes were dispensed into PCR tubes. To
buffy coat were removed by aspiration. The RBCs were the master mix the RNA sample was added and preceded to a
washed three times in phosphate buffer saline (0.1 M). The thermal cycler leading to both, cDNA preparation by reverse
packed cell volume (PCV) obtained was divided into two transcription, as well as amplification of the formed cDNA.
parts, one part of which was diluted with chilled distilled Reverse transcription was carried out by setting thermal
water kept for the analysis of reactive oxygen species (ROS), cycler at 45  C for 50 min followed by a 5 min heating step at
catalase, thiobarbituric acid reactive substances (TBARS) and 95  C during which the DNA polymerase gets activated,
reduced glutathione (GSH). Other part of the packed cell reverse transcriptases get inactivated and the cDNA template
For personal use only.

volume was used for the estimation of other antioxidant is denatured.


enzymes i.e. glutathione peroxidase (GPx), superoxide PCR was performed with the initial denaturation cycle at
dismutase (SOD) and glutathione-S-tranferase (GST). For 94  C for 5 min, followed by 25–40 cycles of 94  C for 1 min,
this, hemoglobin was precipitated in PCV by means of annealing at 50–68  C for 1 min and extension at 72  C for
chloroform and ethanol. After centrifugation at 3000  g for 10 min. Sequence of the primer and the size of the PCR
10 min at 4  C, the supernatant obtained was used for all product generated are described in the Table 1.
above mentioned enzymatic assays.
Biochemical assays
RNA isolation and quantification
Reactive Oxygen Species (ROS)
Total RNA was isolated from the liver and brain samples
Amount of ROS in blood was measured using 20 , 70 -
using kit protocol as provided by the manufacturers (RNeasy
dichlrofluorescein diacetate (DCF-DA) that gets converted
Protect Mini Kit and RNeasy Lipid Tissue Mini Kit respect-
into highly fluorescent DCF by cellular peroxides (including
ively, QIAGEN Sciences, Maryland). About 30 mg of liver
hydrogen peroxide). The assay was performed as described by
tissue and 100 mg brain tissues stored in RNA later were
Socci et al. (1999). For estimation of ROS in blood, 5% RBC
homogenized using a rotor-stator homogenizer until it was
hemolysate was prepared and diluted to 1.5% with ice–cold
uniformly homogenous. This was carried out in the presence
40 mM tris-HCl buffer (pH 7.4). Similarly for estimation of
of specialized highly denaturing guanidine-thiocyanate con-
ROS in tissues, 10% tissue homogenate was prepared. The
taining buffer (1 ml). The lysate was centrifuged for 3 min at
tissue was homogenized (10 mg) in 1 ml of ice–cold 40 mM
high speed. Supernatant was carefully removed and transfered
tris –HCl buffer (pH 7.4), further diluted to 0.25% with the
to a microcentrifuge tube. 1 volume of 70% ethanol was
same buffer and placed on ice. Thenafter, 40 ml of 1.25 mM
added to the cleared lysate, and mixed immediately by
DCF-DA in methanol was added for ROS estimation. All
pipetting. 700 ml of the sample was transferred to an RNeasy
samples were incubated for 15 min in a 37  C water bath.
spin column placed in a 2 ml collection tube and centrifuged
Fluorescence was determined at 488 nm excitation and
for 15 s at 10 000 rpm. The flow-through was discarded and
525 nm emission using a fluorescence plate reader (Tecan
spin column membrane was washed with 700 ml RW1 Buffer
Spectra Fluor Plus).
by centrifuging at 10 000 rpm for 15 sec. Washing of spin
column was repeated again using 500 ml RPE Buffer. The
Glutathione (GSH)
above mentioned steps were repeated twice. Finally, RNA was
eluted using 30–50 ml RNase-free water directly to the spin Analysis of blood GSH concentration was performed by the
column membrane. method described by Ellman (1959) and modified by Jollow
Concentration of isolated RNA was spectrophotometrically et al. (1974). In brief, 0.2 ml of whole blood was added to
determined at 260 nm and absorbance of 1 unit at 260 nm 1.8 ml of distilled water and incubated for 10 minutes at
4 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12

Table 1. Sequence of primer and size of the PCR product. in a boiling water bath for 15 min to get the red color of
thiobarbituric acid-malondialdehyde complex, the absorbance
Product
Genes Primer sequence (50 to 30 ) Ta size
of which was recorded at 535 nm. The amount of TBARS was
calculated using a molar extinction coefficient of 1.56  105/
b-actin F0 -GAGAGGGAAATCGTGCGTGAC 65  C 453 bp M/cm.
R0-CATCTGCTGGAAGGTGGACA
In case of blood, 0.1 ml of 5% RBC hemolysate was added
Catalase F0 -TGGCCTCCGAGATCTTTTCAATG 63  C 452 bp
R0-GCGCTGAAGCTGTTGGGGTAGTA to 0.2 ml of 8.1% SDS (w/v) and incubated for 10 min. 1.5 ml
GPx F0 -CGCTCATGACCGACCCCAAGT 65  C 221 bp
of 20% acetic acid (pH 3.5) was added followed by the
R0-GCCAGCCATCACCAAGCCAATA addition of 1.5 ml of 0.8% thiobarbituric acid (w/v) and 0.7 ml
CuZnSOD F0 -GCGGCTTCTCTCGTCTCCTTGC 65  C 201 bp distilled water. The mixture was then incubated for 1 hour in
R0-TTGATGGACATGGAACCCATGCTCG boiling water bath. 1 ml of distilled water was added to the
MnSOD F0 -ACAGCAAGCACCACGCGACC 67  C 560 bp solution after cooling and centrifuged at 6000 rpm for 15
R0-AACACCCACCACGGGCCTGA minutes. The absorbance of supernatant was read at 532 nm
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

b-actin was used as an internal control. The PCR-amplified products and the values were expressed as moles of MDA/ml.
were then subjected to electrophoresis on 1.5% agarose gels.
Glutathione peroxidase (GPX)

37  C, for complete hemolysis. After adding 3 ml of 4% Glutathione peroxidase activity was measured by the proced-
sulfosalicylic acid, tubes were centrifuged at 2500 rpm for 15 ure of Flohe and Gunzler (1984). Supernatant obtained after
minutes. To the supernatant, 0.2 ml of 10 mM solution of 5, centrifuging 5% tissue homogenate for 10 min at 1500  g,
50 -dithiobis-(2-nitrobenzoic acid), DTNB, was added in followed by 10 000  g for 30 minutes at 4  C, was used for
presence of phosphate buffer (0.1 M, pH 7.4). Absorbance GPx assay. 1 ml of reaction mixture was prepared which
recorded at 412 nm was used for calculation of GSH contained 0.3 ml of phosphate buffer (0.1 M, pH 7.4), 0.2 ml
concentration. of GSH (2 mM), 0.1 ml of sodium azide (10 mM), 0.1 ml of
H2O2 (1 mM) and 0.3 ml of tissue supernatant. After incuba-
Superoxide dismutase (SOD) tion at 37  C for 15 min, reaction was terminated by addition
of 0.5 ml of 5% TCA. Tubes were centrifuged at 1500  g for
Superoxide dismutase (SOD) activity in whole brain, liver and 5 min and supernatant was collected. 0.2 ml of phosphate
For personal use only.

blood was assayed spectrophotometrically as described by buffer (0.1 M, pH 7.4) and 0.7 ml of DTNB (0.4 mg/ml) were
Kakkar et al. (1984). Reaction mixture contained 1.2 ml added to 0.1 ml of the reaction supernatant. After mixing well,
(0.052 mM) sodium pyrophosphate buffer, 0.1 ml (186 mM) absorbance was recorded at 420 nm.
phenazine methosulphate, 0.3 ml (300 mM) nitro blue tetra-
zolium. 0.2 ml of the supernatant obtained after centrifugation
Glutathione-S-transferase (GST)
(1500  g, 10 min followed by 10 000  g, 15 min) of 5%
hemolysate/homogenate was added to the reaction mixture. GST activity was determined following the procedure of
Reaction was initiated by adding 0.2 ml NADH (780 mM) and Habig et al. (1974). Supernatant was obtained after centrifu-
stopped by adding 1 ml glacial acetic acid. Color intensity of ging 5% tissue homogenate at 1500  g for 10 min followed
the chromogen was measured at 560 nm and the activity was by 10 000  g for 30 minutes at 4  C. Reaction mixture
expressed as units/min/mg of protein. contained 0.02 ml of 1-chloro-2, 4-dinitrobenzene (1 mM),
2.9 ml of GSH (0.3 mg GSH/ml in 0.2 M phosphate buffer, pH
Catalase 7.4) and 30ml of tissue supernatant. Change in color was
Catalase activity in tissue was assayed following the proced- monitored by recording absorbance (340 nm) at 30 sec
ure of Sinha (1972), at room temperature. 0.1 ml of 5% RBC intervals for 3 min.
hemolysate/tissue homogenate was incubated with 0.5 ml of
H2O2 (0.2 M) at 37  C for 90 seconds precisely, in the Brain biogenic amines level
presence of 0.01 M phosphate buffer (pH 7.4). Reaction was The frozen brain tissue samples were weighed and homo-
stopped by adding 5% dichromate solution. Further samples genized in acidified butanol. Dopamine (DA), norepinephrine
were incubated at 100  C for 15 min in boiling water bath. (NE) and 5-hydroxytryptamine (5-HT) were estimated
Amount of H2O2 consumed was determined by recording according to the procedure of Jacobwitz and Richardson,
absorbance at 570 nm. (1978). The aliquots of butanol extracts were re-extracted
with phosphate buffer (0.1 M, pH 6.5), part of which was
Thiobarbituric acid reactive substances (TBARS)
derivatized by adding sodium EDTA and iodine solution. The
Measurement of lipid peroxidation was done by the method reaction was terminated by alkaline sulfite and neutralized
described by Ohkawa et al. (1979). Tissue lipid peroxidation with acetic acid (5 M). Fluorescence was read by excitation at
was measured in tissue homogenate [5% homogenate (w/v) 385 nm and emission at 485 nm for NE, and 320 and 385 nm
in 150 mM KCl for brain, and 10% homogenate (w/v) in respectively, for DA. To determine 5-HT, the butanol layer
150 mM KCl for liver] for 30 min at 37  C. The incubation was extracted with 0.1 M HCl in the presence of n-heptane
was interrupted by adding 0.1 ml of 10% trichloroacetic acid. and the acid layer was mixed with o-phthaldehyde. After
After centrifugation, 1 ml of the supernatant was mixed with boiling and cooling, fluorescence was read by excitation at
1 ml of 0.65% thiobarbituric acid. The mixture was then kept 360 nm and emission at 470 nm.
DOI: 10.3109/01480545.2013.866134 Metal oxides nanoparticles induced toxicity 5

Transmission electron microscopy (TEM) of biological DCF, an indicative of oxidative stress increased significantly
samples in erythrocytes on exposure to TiO2, ZnO and Al2O3
nanoparticles compared to the controls, however increased
Transmission electron microscopy was performed to assess
free radical generation was observed in case of ZnO NPs,
the uptake and subcellular localization of TiO2, ZnO and
indicating highest toxicity in blood followed by TiO2 and
Al2O3 nanoparticles in the brain and liver tissues of mice.
Al2O3. Table 2 also depicts effect of TiO2, Al2O3 and ZnO
Slices of brain and liver from the animals post sacrifice (one
nanoparticles on TBARS level in mouse erythrocyte, which is
animal per group) were randomly removed and fixed with
an important marker of lipid peroxidation. Following NP
2.5% glutaraldehyde in 0.1 M sodium cacodylate buffer for
exposure, TBARS level increased significantly in all the
4 h. The specimens were postfixed with 2% osmium tetroxide,
groups compared to the control, however the comparative
dehydrated through graded alcohol solutions and embedded in
level of TBARS among exposed groups were not found to be
Epon. Ultrathin sections mounted on copper grids (300 mesh)
significant.
were contrasted with uranyl acetate and lead citrate for TEM
TiO2 induced alterations in GSH, GPx and GST activities
analysis (ZeissbEM 10 A; Carl Zeiss, Oberkochen, Germany)
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

in mouse erythrocytes as depicted in Table 2. Erythrocyte


at an accelerating voltage of 60 kV. To obtain a global
GSH increased specifically on TiO2 and Al2O3 nanoparticles
characterization of the tissues, several grids were prepared (4–
exposure while ZnO nanoparticles had no significant effect on
5 grids per animal, each one having 3–4 sectioning cuts) from
blood GSH. Significant reduction in the activity of GPx was
the different regions.
observed in mouse erythrocyte on exposure to TiO2 and ZnO
Statistical analysis nanoparticles, However Al2O3 nanoparticles had no effect on
GPx activity compared to the control. Activity of GST on the
The results are expressed as the mean  SEM of number of other hand remained unaffected following exposure to all the
observations. Comparisons of means were carried out using three nanoparticles.
one way analysis of variance (ANOVA) followed by Turkey’s
post hoc test to compare means between the different
treatment groups. Differences were considered significant at Table 2. Effect of TiO2, Al2O3 and ZnO nanoparticles on oxidative
p50.05 unless otherwise stated in the text. stress variables/phase II drug metabolizing enzyme in mouse
erythrocyte.
For personal use only.

Results
Erythrocyte
Particle characterization
Normal TiO2 ZnO Al2O3
The SEM was done for the characterization of TiO2, ZnO and
ROS 0.73  0.02* 0.86  0.04y 0.83  0.01y 0.86  0.02y
Al2O3 NPs and chemical analysis of these nanoparticles by GSH 0.96  0.02* 1.10  0.01y 1.09  0.01* 1.18  0.04y
EDX method and the data is shown in Figure 1. The spectrum TBARS 270.8  1.29* 345.0  6.93y 351.8  4.76y 372.5  34.32y
suggested that the nanoparticle size of TiO2 nanoparticles is SOD 0.25  0.03* 0.10  0.01y 0.12  0.02y 0.09  0.04y
between 50–75 nm, ZnO nanoparticles is 80–100 nm and Catalase 0.79  0.07* 0.54  0.05y 0.63  0.04y 0.56  0.02y
GPx 1.17  0.10* 3.20  0.55y 2.22  0.38y 1.57  0.43*
Al2O3 nanoparticles is 40–50 nm (Figure 1). GST 0.06  0.01* 0.08  0.02* 0.08  0.01* 0.13  0.02y

Effects on oxidative stress variables in mouse Abbreviation used and unit: ROS – Reactive Oxygen Species as Zmoles/
erythrocyte min/ml of RBC; GSH – Reduced Glutathione as as mg/ml of RBCs;
TBARS – Thiobarbituric Acid Reactive Substances as nmole MDA
Erythrocytes were more commonly employed in the evalu- produced/ml RBC; SOD – Superoxide Dismutase as U/mg protein/min;
Catalase as m moles H2O2 degraded/min/mg protein; GPx –
ation of oxidative stress. Effects of TiO2, ZnO and Al2O3 Glutathione Peroxidase as U/mg protein/min; GST – Glutathione-S-
nanoparticles on the generation of reactive oxygen species Transferase as U/mg protein/min. Values are mean  SE; n ¼ 5.
(ROS) are depicted in Table 2. The fluorescence intensity of *,yp50.05 compared to normal control as evaluated by Student’s
‘‘t’’ test.

Figure 1. Particle size distribution of NPs, (A) SEM image of TiO2 NPs, (B) SEM image of ZnO NPs and (C) SEM image of Al2O3 NPs.
6 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12

Table 3. Effect of TiO2, Al2O3 and ZnO nanoparticles on oxidative (Table 4) on exposure to TiO2, ZnO and Al2O3 nanoparticles
stress variables in mouse liver.
compared to the control group. However in both the tissues,
Liver
levels of ROS were found to be highest in the group exposed to
ZnO NP, indicating it to be most toxic, followed by Al2O3 and
Normal TiO2 ZnO Al2O3 TiO2. In liver, TBARS level was found to be significantly
ROS 230.6  13.2* 270.5  3.1y 327.8  3.8y 313.4  6.4y elevated on exposure to TiO2 and Al2O3 (Table 3) whereas, it
TBARS 2.1  7.5* 3.0  33.5y 2.0  8.4* 2.9  5.2y remained unaltered in the group exposed to ZnO nanoparticle.
SOD 1.33  0.29* 0.45  0.08y 0.52  0.10y 0.61  0.08y
Catalase 1.76  0.31* 0.76  0.02y 0.76  0.13y 0.60  0.14y None of the nanoparticle was found to alter the level of
GPx 2.60  0.51* 1.43  0.07y 1.50  0.17y 1.29  0.04y TBARS in brain (Table 4). Activity of antioxidant enzymes
GST 1.01  0.22* 0.84  0.04* 0.72  0.04* 0.75  0.11* was also evaluated to validate the role of oxidative stress in
elucidating toxic manifestations. Activity of both SOD and
Abbreviations used and units: ROS – Reactive Oxygen Species as
Fluorescent Intensity unit (FIU); TBARS – Thiobarbituric Acid Catalase declined significantly in both liver and brain on
Reactive Substances as mg/gm of tissue weight; SOD – Superoxide exposure to all the nanoparticles compared to control.
Dismutase as units min1 mg protein1; Catalase as nmoles of H2O2
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

However highest decrease was observed in case of ZnO NPs,


consumed min1 mg protein1; GPx – glutathione peroxidase as
ngmin1 mg protein1; GST – Glutathione-S-transferase as nmole
suggesting enhanced toxicity compared to Al2O3 and TiO2
conjugate1 mg1 protein. NPs. Activity of GPx was also depleted significantly in the
Values are mean  SE; n ¼ 5. *,yDifferences between values with group exposed to ZnO NPs in both liver and brain compared to
matching symbol notations within each column are not statistically control and other exposed groups. GST activity remained
significant at 5% level of probability
unaltered in all the nanoparticle exposed groups except for a
significant depletion in ZnO exposed group in liver and brain.
Table 4. Effect of TiO2, Al2O3 and ZnO nanoparticles on oxidative
stress variables and neurotransmitter levels in mouse brain. Effects on the level of norepinephrine (NE), dopamine
(DA) 5-hydroxytryptamine (5-HT) in cortex region of
Brain
mouse brain
Normal TiO2 ZnO Al2O3
Table 4 depicts the levels of norepinephrine, dopamine and
Oxidative Stress Biomarkers
ROS 317.7  3.5* 374.7  10.2y 428.6  6.0y 407.7  8.0y
5-hydroxytryptamine in cortex region of mouse brain.
TBARS 3.3  31.4* 3.7  26.0* 2.6  11.3* 2.5  12.7* Administration of all the three nanoparticles TiO2, ZnO and
For personal use only.

SOD 0.29  0.03* 0.24  0.02y 0.26  0.02y 0.42  0.05 Al2O3 led to a significant increase in norepinephrine and
Catalase 1.79  0.11* 0.75  0.29y 1.18  0.19y 0.79  0.19y dopamine levels compared to the control group. Among the
GPx 1.77  0.20* 1.06  0.02y 1.14  0.04y 1.08  0.08y
GST 0.08  0.01* 2.09  0.03y 0.15  0.01* 0.17  0.01* exposed groups, levels of norepinephrine and dopamine were
Neurotransmitters (cerebral cortex) found to be the highest in case of ZnO NP, suggesting
NE 10.37  2.90* 25.84  3.28y 131.4  38.39y 58.72  12.37y enhanced neurotoxic potential of ZnO NP in comparison to
DA 24.90  7.12* 45.49  8.87y 86.53  16.15y 46.39  9.07y other NPs. However, the levels of 5-hydroxytryptamine
5-HT 35.69  2.10* 35.65  1.00* 36.14  1.90* 35.48  1.70*
remained unaltered in all the groups compared to control.
Abbreviations used and units: ROS – Reactive Oxygen Species as
Fluorescent Intensity unit (FIU); TBARS – Thiobarbituric Acid Effects on antioxidant mRNA expression in mouse
Reactive Substances as mg/gm of tissue weight; SOD – Superoxide liver
Dismutase as units min1 mg protein1; Catalase as nmoles of H2O2
consumed min1 mg protein1; GPx – glutathione peroxidase as Total RNA was isolated from all control and exposed samples
ngmin1 mg protein1; GST – Glutathione-S-transferase as nmole by a kit protocol as described earlier and purity and quantity
conjugate1 mg1 protein; NE – Norepinephrine as mg/g tissue weight;
DA – Dopamine as mg/g tissue weight; 5-HT – 5-hydroxytryptamin as of isolated RNA were checked in denaturing formamide
mg/g tissue weight. agarose gel. Figure 2 depicts the expression level of different
Values are mean  SE; n ¼ 4. *,y,Differences between values with antioxidant enzymes in mouse liver following exposure to
matching symbol notations within each column are not statistically
TiO2, ZnO and Al2O3 nanoparticles. Unaltered expression
significant at 5% level of probability
was observed in catalase and GPx transcript on exposure to
NPs, however a marked decrease in the expression of
CuZnSOD and MnSOD transcript was observed. A more
Effects of TiO2, ZnO and Al2O3 nanoparticles on SOD pronounced inhibition in the expression of CuZnSOD and
and catalase activities are shown in Table 2. Activity of SOD MnSOD transcript was noted in case of mice exposed to ZnO
and Catalase reduced significantly following exposure to all and Al2O3 NPs, indicating their enhanced toxicity in
the three NPs in mouse erythrocytes. No difference however comparison to TiO2. The expression of b-actin gene was
was noted in comparative activity of these enzymes among used as control (housekeeping gene) to reveal any variation in
exposed groups. antioxidant expression due to purity of RNA or error in
estimation or loading.
Effects on oxidative stress variables in mouse liver
and brain Effects of TiO2, ZnO and Al2O3 nanoparticles on
antioxidant mRNA expression in mouse brain
Effect of TiO2, ZnO and Al2O3 nanoparticles on the
generation of reactive oxygen species in liver and brain of Figure 2 shows RNA expression level of b-actin and
mice is depicted in Table 3 and Table 4. Levels of ROS, antioxidant enzymes isolated from brain after exposure to
increased significantly in both liver (Table 3) and brain nanoparticles for 21 days. An unaltered expression of catalase
DOI: 10.3109/01480545.2013.866134 Metal oxides nanoparticles induced toxicity 7
Figure 2. Differential mRNA expression of
antioxidant enzymes of control and TiO2,
ZnO and Al2O3 nanoparticles exposed
mouse. mRNA expression of b-actin (453 bp,
housekeeping gene), catalase (452 bp), GPx
(221 bp), Cu-Zn SOD (201 bp) and MnSOD
(560 bp) in nanoparticles exposed mouse
liver and brain compared with control.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13
For personal use only.

transcript was noted following exposure to TiO2, ZnO and Effects on TEM micrographs of mouse brain
Al2O3 nanoparticles, however a slight upregulation in GPx
No abnormalities were noted in normal brain tissues
expression was observed on exposure to Al2O3 nanoparticles
(Figure 4A), whereas the presence of endosomes was observed
compared to other nanoparticles. Significant inhibition in the
near the cell membrane with a large number of nanoparticles
expression of CuZnSOD and MnSOD transcript was observed
trapped inside in case of ZnO and TiO2 exposed groups
following exposure to ZnO and Al2O3 NPs compared to
(Figure 4C and D). Clumps of nanoparticles were observed
control and TiO2 NP. The expression of b-actin gene was used
inside the endosomes resembling nanoaggregates. However,
as control (housekeeping gene) to reveal any variation in
magnified images show the presence of individual nanoparti-
antioxidant expression due to purity of RNA or error in
cles inside the clump (Figure 4B and C). Few nanoparticles
estimation or loading. No alteration in the level of b-actin was
were also noted inside the cylindraxile of neurons (Figure 4C).
observed in brain.
The presence of rounded nuclei in neuron was observed in the
control group, however, the ultrastructure of neuron in mice
Effects on TEM micrographs of mouse liver sections exposed to Al2O3 NP revealed significant shrinkage of the
TEM analyses of the liver sections were performed to study nucleus (Figure 4). The phenotypic appearance bears resem-
the biodistribution of nanoparticles. No abnormality was blance to classical morphological characteristics of apoptosis.
noted in normal tissues (Figure 3A). TiO2 treated groups
showed the presence particles entrapped within compartments
Discussion
resembling endosomes and Kupffer cells (Figure 3B). The present study investigated the toxicological profile and
Numerous cytoplasmic vesicles and Golgi complexes con- general mechanism involved in TiO2, ZnO and Al2O3
taining nanoparticles trapped inside were observed in liver nanoparticles-induced sub-acute toxicity in mice. We selected
sections of ZnO and Al2O3 exposed mice (Figure 3C and D). oral route of administration as the route of exposure for mice
Ultrastructure of hepatocytes isolated from Al2O3 treated as most of these nanoparticles are being used in food
group revealed tumescent mitochondria and vacuolization. packaging and thus may gain entry into the body principally
Additionally, enhanced deformity and damaged structure near through this route. These nanoparticle are also used in other
the nucleus exhibiting signs of necrosis were also observed in consumer products (coating and dermal applications) and thus
hepatocytes (Figure 3D). there is always a risk of ingestion during use. The doses at
8 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13
For personal use only.

Figure 3. TEM micrographs of mouse liver showing no abnormalities (A); presence of particles entrapped within compartments resembling endosomes
and Kupffer cells in TiO2 exposed (B); cytoplasmic vesicles and Golgi complexes containing nanoparticles in ZnO exposed (C); cytoplasmic vesicles
and Golgi complexes containing nanoparticles and also necrosis of nucleus in Al2O3 exposed mouse.

which humans may be exposed to nanoparticles in these dosage (500 mg/kg) for an increased period of time (21 days)
scenarios are yet to be identified as there are no available at both effecter (biochemical) and expression (mRNA) levels.
guidelines for conducting in vivo toxicity assessment. We thus Oral administration with 500 mg/kg of TiO2, ZnO and Al2O3
selected a dose defined in literature for studying sub-acute oral nanoparticles for 21 consecutive days induced biochemical
toxicity. This dose does not necessarily reflect the actual alterations in blood, liver and brain in mice. Cellular oxidative
concentrations of nanoparticles found in the real environ- stress was evident by elevated ROS level, reduced glutathione
ments, however, they can provide a baseline data to assess the level, increased lipid peroxidation and impaired antioxidant
health risks from exposure to these nanoparticles. Mice were defense status. Despite the fact that there is growing concern in
chosen as the in vivo model of exposure because of their nanotoxicology studies, little is known about the interaction of
similarity with human metabolic, biochemical and physio- nanoparticles with biological system. TiO2, ZnO and Al2O3
logical pathways. Numerous reports are available suggesting NPs are three metal oxide nanoparticles widely used in
the toxicity of metal oxide NPs at different doses and industrial products, such as cosmetics and pharmaceuticals.
durations, without any reported mortality following exposure Therefore, potential widespread exposure may occur during
to these three NPs (TiO2, ZnO and Al2O3) (Sharma et al., both manufacturing and their applications. After entering the
2012; Sycheva et al., 2011). Moreover, acute toxicity studies body through different routes, they reach the target organs
have been carried out using high doses of these metal oxide through blood. Erythrocytes, being the dominant cell type in
NPs upto 5 g/kg (Klien & Godnic-Cvar, 2012). Thus an blood are vulnerable to the toxic effect by these nanoparticles
attempt was made in this study to further investigate the toxic (Li et al., 2008). Due to their characteristics small size and
effects of TiO2, ZnO and Al2O3 nanoparticles at an increased enhanced absorption properties, nanoparticles interact with
DOI: 10.3109/01480545.2013.866134 Metal oxides nanoparticles induced toxicity 9
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13
For personal use only.

Figure 4. TEM micrographs of mouse brain showing normal brain tissue with no abnormalities i.e. rounded nuclei in neurons (A); presence of
individual nanoparticles inside clump in TiO2 exposed (B); presence of nanoparticles inside the cylindraxile of neurons in ZnO exposed (C); and
significant shrinkage of nucleus, endosomes near cell membrane with large number of nanoparticles trapped inside in case of Al2O3 nanoparticles (D).

erythrocyte membrane and cause its agglutination by changing group shows the adapting response against the toxic effect of
the cell membrane structure and properties (Li et al., 2008). nanoparticles. GSH can function directly in the destruction of
We studied the effects of TiO2, ZnO and Al2O3 ROS and an increased level of GSH indicates enhanced
nanoparticles on erythrocyte oxidative stress in the present defense mechanism of the body (Cantin & Begin, 1991; Khan
study. Our results indicate an increased generation of reactive et al., 1990; Martensson & Meister, 1991). Whereas, the level
oxygen species and elevated TBARS levels in all the of GSH in erythrocytes remain unaffected upon exposure ZnO
nanoparticles intoxicated groups indicating oxidative stress. nanoparticles.
This can be attributed to the phagocytosis of nanoparticles by Organisms use a diverse array of antioxidant enzymes
erythrocytes which in turn activate a membrane bound which play crucial role in providing protection against toxic
NADPH oxidase leading to the formation of ROS (Afaq effects of free radicals. Our studies in erythrocytes have shown
et al., 1998). Elevated level of TBARS in erythrocyte a significant alteration in the activities of most of the
following exposure to TiO2, ZnO and Al2O3 nanoparticles antioxidant enzymes investigated. Decreased activities of
revealed enhanced lipid per-oxidative products. Nanoparticles SOD and catalase in TiO2, ZnO and Al2O3 nanoparticles
can get embedded in the lipid protein layer present in the exposed groups indicate increased generation of free radicals.
erythrocyte membrane and cause damage by exerting strong However, increased activities of GPx in TiO2 and ZnO exposed
oxidizing ability (Afaq et al., 1998). Several other studies also groups again depict an adaptive mechanism against nanopar-
support lipid peroxidation via free radical generation due to ticles induced oxidative stress. Moreover, increased activity of
nanoparticles induced toxicity (Li et al., 2008; Park et al., GPx can be directly related to the increased level of GSH
2008; Xia et al., 2006). Further, a significant increase in observed indicating elevated defense mechanism by these
reduced glutathione level in TiO2, and Al2O3 NPs exposed antioxidants during detoxification mechanism (Nel et al.,
10 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12

2006). However, GST activity remained unaltered on exposure of certain neurotransmitters and receptors in nerve cells,
to all the nanoparticles studied. leading to neural damage. In addition, ROS could attack the
There are few reports which indicated that nanoparticles various kinds of neuronal connections and decrease the
can cross the blood-brain barrier and enter the central nervous memory in experimental animals (Ma et al., 2010). The
system (Kreyling et al., 2002; Lockman et al., 2004; resulting oxidative stress and brain injury is due to a cascade
Oberdörster et al., 2004; Sharma & Sharma, 2007; Wang of reactions triggered by metal oxide nanoparticles, such as
et al., 2008). Rapid utilization of nano particles and their lipid peroxidation, decreased activity of antioxidant enzymes,
potential risk to central nervous system (CNS) has elicited release of nitric oxide, reduction of glutamic acid, and
much concern recently. Considering this, one of the major downregulation of acetylcholinesterase activity.
focuses of the present study was to study the potential role of Nanoparticles have been found to be accumulated in liver
metal oxide nanoparticles (TiO2, ZnO and Al2O3) in the which means that nanoparticles could be transported to the
induction of brain oxidative stress and alterations in neuro- organs after uptake by the gastrointestinal tract (Liu et al.,
transmitters level in mouse. Our data showed increased 2009; Ma et al., 2009). Moreover, some recent studies have
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

production of reactive oxygen species following exposure to unequivocally showed that exposure to nanoparticles causes
all the three metal oxide nanoparticles, indicating that mice oxidative damage and liver dysfunction in vivo (Cui et al.,
treated with these nanoparticles underwent oxidative stress. 2010; Liu et al., 2009; Ma et al., 2010; Wang et al., 2007).
Generally direct particles are translocated to various tissues in In the present study also, elevated level of ROS following
case of TiO2 and Al2O3 NPs, whereas ZnO nanoparticles are exposure to TiO2, ZnO and Al2O3 nanoparticles in liver was
mostly dissolved in the stomach to form Zn2þ ions, and are observed. Nanoparticles can generate ROS by different
then taken up in the tissues by interaction between zinc and mechanisms. The uptaken metal oxide nanoparticles can
sulfur-containing ligands in proteins (Baek et al., 2012; lose the metallic ions which directly interact with NADPH
Huang et al., 2010). The binding of zinc to L-histidine (His) oxidases from the plasma membrane or mitochondria thus
in cerebrospinal fluids seems to be involved in transferring disturbing the electron transport chain and generating a
zinc to target sites, which regulate its uptake across the brain superoxide anion (Klotz & Sies, 2009). Level of TBARS
barrier systems. Free Zn2þ modulates many membrane increased significantly in TiO2 and Al2O3 nanoparticles
receptors, transporters and channels (Takeda et al., 2002). exposed groups while ZnO nanoparticles did not produce
The role of oxidative stress in nanoparticles induced CNS any change. Moreover, activities of SOD, CAT and GPx, were
For personal use only.

toxicity has been widely reported. These studies suggest that significantly inhibited due to exposure of all the three
nanoparticles could be phagocytized by neurons and micro- nanoparticles suggesting triggered oxidative stress via
glia, resulting in the release of ROS. Overproduction of ROS increased ROS production and lipid peroxidation with a
would break down the balance of the oxidative/antioxidative subsequent reduction in the level of antioxidant defense
system in the brain, which is closely related to the reduction enzymes of the mouse liver (Cui et al., 2010). On the basis of
of the antioxidant enzymes (Long et al., 2007; Wang et al., above findings, it can be concluded that being the primary
2008). In the present study also, the activities of SOD, metabolizing organ of the body liver might be the most
Catalase, and GPx, were significantly inhibited in the groups susceptible organ to nanoparticle exposure and liver damage
exposed to the nanoparticles except a significant enhancement is a consequence of direct enzyme inhibition by these
in SOD activity in Al2O3 nanoparticles. Ma et al. (2010) has nanoparticles (Liu et al., 2009; Ma et al., 2009). This study
also reported decreased activities of antioxidant enzymes in is first in line to report the effect of three metal oxides
rat brain exposed to TiO2 nanoparticles. Whereas, GST nanoparticles (TiO2, ZnO and Al2O3) on gene expression
remained unaffected, except a significant elevation in TiO2 profile of antioxidant enzymes in mice liver and brain.
nanoparticles. Role of GST during TiO2 nanoparticles However, there are few reports which have investigated the
administration needs to be further investigated. changes in gene expression of inflammatory markers (Park
Administration of chemicals disturbs the spontaneous et al., 2008) and antioxidant enzymes in lung after inhalation
activity of the cells and influences neurotransmitter turnover. and intratracheal instillation exposure to nanoparticles
Thus the level of neurotransmitters in cerebral cortex region (Janssen et al., 1994). Inhalation of ultra-fine TiO2 nanopar-
of mice has been evaluated. Nanoparticles led to an increased ticles led to an increase in gene expression of manganese
level of norepinephrine (NE) and dopamine (DA) in cortex superoxide dismutase (MnSOD), catalase and GPx (Janssen
region of mouse brain suggesting weakened ability to et al., 1994). In contrast, the expression profile of copper-zinc
maintain an appropriate state of activation in the central superoxide dismutase (CuZnSOD) was less striking in these
nervous system due to toxicity. However, the level of 5-HT studies. On the contrary, our results show that following the
remained unaffected following exposure of these nanoparti- exposure to TiO2, ZnO and Al2O3 nanoparticles the expres-
cles. Some researchers have also reported impaired glutamate sion level of catalase and GPX remained unaffected in both
and AChE activities in TiO2 nanoparticles exposed animals mice liver and brain. However, significant inhibition in
indicating disruption in the normal functioning of central mRNA expression profile of CuZnSOD and MnSOD tran-
nervous system (Ma et al., 2010). Neurotransmitters are script was observed which is an important finding of the
known to play a key role in memory, awareness, thought, and present study (Ma et al., 2010). The downregulated expression
consciousness and allow the organism to become alert and of CuZnSOD and MnSOD transcripts suggests decreased
guards against the intensification of reflex reactions and other activity of detoxifying excess NPs and ROS scavenging which
behavior. Our results suggest that accumulation of these supports our biochemical results. Cui et al. (2010) also found
nanoparticles in brain resulted in altered synthesis and release decreased expressions of liver antioxidant enzymes after
DOI: 10.3109/01480545.2013.866134 Metal oxides nanoparticles induced toxicity 11

intragastric administration of TiO2 nanoparticles to mice. mitochondria and nucleus. The study revealed ZnO NP as the
There was a slight upregulation in GPx mRNA expression most toxic NP, followed by Al2O3 and TiO2, as evident from
level upon exposure to Al2O3 nanoparticles in brain which the observed results. Nanoparticles thus, possess potential
needs to be further investigated. toxicity and risk assessment is needed prior to their wide
In order to study the biodistribution of TiO2, ZnO and consumption to ensure safe utilization and disposal of
Al2O3, Transmission Electron Microscopy (TEM) analyses of nanoparticles.
liver and brain tissues were performed. The results obtained
with TEM data are in line with oxidative stress parameters. Acknowledgements
The nanoparticles were found to be distributed throughout the
cytoplasm and nucleus. Clumps of nanoparticles are found We thank Dr. M.P. Kaushik, Director, Defence Research and
inside endosomes and in cytoplasm. Large endosomes with Development Establishment for offering all facilities and
nanoparticles in the cytoplasm of the cells and near the cell support required for this study. Ms. Rupal Shrivastava is
and nuclear membrane were observed, which suggested that recipient of MAPCOST Junior Research Fellowship.
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

nanoparticles were entering the cells through endocytosis.


The current evidence from electron micrographs sheds light Declarartion of interest
on the endocytic pathway of nanoparticle uptake. Further, a None declared.
detailed study should be conducted to unravel the mechanism This work was supported by the grant from Ministry of
involved in metal oxide nanoparticles uptake. Moreover, Defence, Government of India.
deformity and damaged structure near the nucleus in hepato-
cytes could be observed which exhibited signs of necrosis
References
whereas, ultrastructure of neurons from the Al2O3 exposed
group presented significant shrinkage of the nucleus with Afaq F, Abidi P, Matin R, Rahman Q. (1998). Cytotoxicity, pro-oxidant
effects and antioxidant depletion in rat lung alveolar macrophages
apoptotic signal. exposed to ultrafine titanium dioxide. J Appl Toxicol 18:307–312.
As indicated at the beginning that the nanoparticles of the Baek M, Chung H, Yu J, et al. (2012). Pharmacokinetics, tissue
three chosen metals induced toxicity might be attributed due distribution, and excretion of zinc oxide nanoparticles. Int J Nanomed
to high dosing rather than the particle size and distribution. It 7:3081–3097.
Borm PJA, Robbins D, Haubold S, et al. (2006). The potential risks of
is known that metal ions released by three nanoparticles might nanomaterials: a review carried out for ECETOC. Part Fibre Toxicol
For personal use only.

be contributing to the toxicities. Although we didn’t measure 3:11–23.


concentration of these metals in blood or soft tissues, this Brown DM, Wilson MR, MacNee W, et al. (2001). Size dependent pro
might be the main lethal mechanism for the more pronounced inflammatory e ects of ultrafine polystyrene particles. A role for
surface area and oxidative stress in the enhanced activity of ultrafines.
toxicity of ZnO NPs that the other two. TiO2 NPs is known to Toxicol Appl Pharmacol 175:191–199.
enter central nervous system and may damage brain. Our Cabiscol E, Tamarit J, Ros J. (2000). Oxidative stress in bacterial and
result suggest that among the three nanoparticles the changes protein damage by reactive oxygen species. Inter Microbiol 3:3–8.
in the biogenic levels were more pronounced in ZnO Cantin AM, Begin R. (1991). Glutathione and inflammatory disorders of
the lung. Lung 169:123–138.
nanoparticle exposed mice while TiO2 and Al2O3 exposed Cui Y, Gong X, Duan Y, et al. (2010). Hepatocyte apoptosis and its
mice elicited similar effects. It has recently been reported that molecular mechanisms in mice caused by titanium dioxide nanopar-
ZnO NPs are slightly soluble after entering the organism, it ticles. J Hazar Mat 183:874–880.
can be ionized to release Zn2þ, which may access cells Deng X, Luan Q, Chen W, et al. (2009). Nanosoized zinc oxide particles
induce neural stem cell apootosis. Nanotechnology 20:115101.
through ion channels (Deng et al., 2009; Liu et al., 2013). Dick CA, Brown DM, Donaldson K, Stone V. (2003). The role of free
These authors further suggested that Zn2þ can subsequently radicals in the toxic and inflammatory effects of four different
enter the blood circulation to damage cell metabolism and ultrafine particle types. Inhal Toxicol 15:39–52.
stimulate cells to produce large amounts of ROS to cause Ellman GL. (1959). Tissue sulfhydryl groups. Arch Biochem 82:70–77.
Englert N. (2004). Fine particles and human health – a review of
oxidative stress and ultimately apoptosis (Deng et al., 2009; epidemiological studies. Toxicol Lett 149:235–242.
Liu et al., 2013). Flohe L, Gunzler WA. (1984). Assays of glutathione peroxidase. Meth
Enzymol 105:114–121.
Habig WH, Pabst MJ, Jakoby WD. (1974). Glutathione transferase, the
Conclusions first enzymatic step in mecapturic acid formation. J Biol Chem 249:
7130–7139.
In conclusion, the study confirms that interaction of absorbed Huang YW, Wu CH, Aronstam RS. (2010). Toxicity of transition metal
nanoparticles with cellular components could generate ROS oxide nanoparticles: recent insights from in vitro studies. Materials 3:
and lead to cellular toxicity if the magnitude of ROS 4842–4859.
Hussain SM, Stolle LKB, Schrand AM, et al. (2009). Toxicity evaluation
production overwhelms the antioxidant defense status of the for safe use of nanomaterials: Recent achievements and technical
cell. Accumulation of these nanoparticles into brain subse- challenges. Adv Mater 21:1549–1559.
quently disturbs the normal metabolism of neurotransmitters, Iavicoli I, Leso V, Bergamaschi A. (2012). Toxicological effects of
ultimately leading to brain damage and thereby indicating the titanium dioxide nanoparticles: a review of in vivo studies. Eur Rev
Med Pharmacol Sci 15:481–508.
neurotoxic potential of these NPs. Interestingly, inhibition in Jacobwitz DW, Richardson JS. (1978). Method for rapid determination
gene expression profile of CuZnSOD and MnSOD in liver of norepinephrine, dopamine and serotonin in the same brain region.
and brain, also validates the hypothesis, in turn establishing Pharmacol Biochem Behav 8:515–519.
oxidative stress as the major mechanism inducing toxic Janssen YMW, Marsh JP, Driscoll KE, et al. (1994). Increased
expression of manganese-containing superoxide dismutase in rat
manifestations. Electron micrographs confirmed significant lungs after inhalation of inflammatory and fibrogenic minerals.
number of nanoparticles in vital organs such as cytoplasm, Free Radic Biol Med 16:315–322.
12 R. Shrivastava et al. Drug Chem Toxicol, Early Online: 1–12

Jingxia Z, Lanju X, Tao Z, et al. (2009). Influences of nanoparticle zinc Ohkawa H, Onishi N, Yagi K. (1979). Assay for lipid peroxides in
oxide on acutely isolated rat hippocampal CA3 pyramidal neurons. animal tissues by thiobarbituric acid reaction. Anal Biochem 95:
Neurotoxicol 30:220–230. 351–358.
Jollow DJ, Mitchell JR, Zamppaglione Z, Gillette JR. (1974). Park EJ, Yi J, Chung KH, et al. (2008). Oxidative stress and apoptosis
Bromobenzene induced liver necrosis. Protective role of glutathione induced by titanium dioxide nanoparticles in cultured BEAS-2B cells.
and evidence for 3,4-bromobenzene oxide as the hepatotoxic metab- Toxicol Lett 180:222–229.
olites. Pharmacology 11:151–157. Patlolla A, McGinnis B, Tchounwou P. (2011). Biochemical and
Kakkar P, Das B, Viswanathan PN. (1984). A modified spectrophoto- histopathological evaluation of functionalized single-walled carbon
metric assay of superoxide dismutase. Ind J Biochem Biophys 21: nanotubes in Swiss–Webster mice. J Appl Toxicol 31:75–83.
130–132. Prabhakar PV, Reddy UA, Singh SP, et al. (2012). Oxidative stress
Khan SG, Ali S, Rahman Q. (1990). Protective role of ascorbic acid induced by aluminium oxide nanomaterials after acute oral treatment
against asbestos induced toxicity in rat lungs: in vitro study. Drug in Wistar rats. J Appl Toxicol 32:436–445.
Chem Toxicol 13:249–256. Schrand AM, Rahman AF, Hussain SM, et al. (2010). Metal based
Klien K, Godnic-Cvar J. (2012). Genotoxicity of metal nanoparticles. nanoparticles and their toxicity assessment. Nanomed Nanobiotechnol
Arh Hig Rada Toksikol 63:133–145. 2:544–568.
Klotz LO, Sies H. (2009). Cellular generation of oxidants: relation to Sharma HS, Sharma A. (2007). Nanoparticles aggravate heat stress
oxidative stress. In:Jacob C, Winyard PG, eds. Redox signaling and induced cognitive deficits, blood-brain barrier disruption, edema
Drug and Chemical Toxicology Downloaded from informahealthcare.com by Defence Research & Development on 12/17/13

regulation in biology and medicine. Weinheim, Germany: Wiley-VCH formation and brain pathology. Prog Brain Res 162:245–273.
Verlag, 45–61. Sharma V, Singh P, Pandey AK, Dhawan A. (2012). Induction of
Kreyling W, Semmler M, Erbe F, et al. (2002). Translocation of ultrafine oxidative stress, DNA damage and apoptosis in mouse liver after sub-
insoluble iridium particles from lung epithelium to extrapulmonary acute oral exposure to zinc oxide nanoparticles. Mutat Res 745:84–91.
organs is size dependent but very low. J Toxicol Environ Health A 65: Shimizu M, Tainaka H, Oba T, et al. (2009). Maternal exposure to
1513–1530. nanoparticulate titanium dioxide during the prenatal period alters gene
Lai JCK, Lai MB, Jandhyam S, et al. (2008). Exposure to titanium expression related to brain development in the mouse. Part Fibre
dioxide and other metallic oxide nanoparticles induces cytotoxicity on Toxicol 6:20.
Sinha AK. (1972). Colorimetric assay of catalase. Anal Biochem 47:
human neural cells and fibroblasts. Int J Nanomed 3:533–545.
389–394.
Li SQ, Zhu RR, Zhu H, et al. (2008). Nanotoxicity of TiO2 nanoparticles
Socci DJ, Bjugstad KB, Jones HC, et al (1999). Evidence that oxidative
to erythroctyte in vitro. Fd Chem Toxicol 46:3626–3631.
stress is associated with the pathophysiology of inherited hydroceph-
Li Y, Li J, Yin J, et al. (2010). Systematic influence induced by 3 nm
alus in the H-Tx rat model. Exp Neurol 155:109–117.
titanium dioxide following intratracheal instillation of mice. J Nanosci
Steck TL, Kant JA. (1974). Preparation of impermeable ghosts and
Nanotechnol 10:8544–8549. inside-out vesicles from human erythrocyte membranes. Method
Liu H, Ma L, Zhao J, et al. (2009). Biochemical toxicity of nano-anatase Enzymol 31:172–180.
TiO2 Particles in mice. Biol Trace Elem Res 129:170–180. Sycheva LP, Zhurkov VS, Iurchenko VV, et al. (2011). Investigation of
Liu H, Danga D, Yanga H, et al. (2013). Comparative study of
For personal use only.

genotoxic and cytotoxic effects of micro- and nanosized titanium


respiratory tract immune toxicity induced by three sterilisation dioxide in six organs of mice in vivo. Mutat Res 726:8–14.
nanoparticles: Silver, zinc oxide and titanium dioxide. J Hazard Takeda A, Suzuki M, Oku N. (2002). Possible involvement of plasma
Mater 248–249:478–486. histidine in differential brain permeability to zinc and cadmium.
Lockman PR, Koziara JM, Mumper RJ, Allen DD. (2004). Nanoparticle Biometals 15:371–375.
surface charges alter blood–brain barrier integrity and permeability. Utell MJ, Frampton MW. (2000). Acute health effects of ambient air
J Drug Target 12:635–641. pollution: the ultrafine particle hypothesis. J Aerosol Med 13:
Long TC, Tajuba J, Sama P, et al. (2007). Nanosize titanium dioxide 355–359.
stimulates reactive oxygen species in brain microglia and damages van der Zande M, Vandebriel RJ, Van Doren E, et al. (2012).
neurons in vitro. Environ Health Perspect 115:1631–1637. Distribution, elimination, and toxicity of silver nanoparticles and
Ma L, Liu J, Li N, et al. (2010). Oxidative stress in the brain of mice silver ions in rats after 28-day oral exposure. ACS Nano 28:
caused by translocated nanoparticulate TiO2 delivered to the abdom- 7427–7442.
inal cavity. Biomaterials 31:99–105. Wang J, Liu Y, Jiao F, et al. (2008). Time-dependent translocation and
Ma L, Zhao J, Wang J, et al. (2009). The Acute Liver Injury in Mice potential impairment on central nervous system by intranasally
Caused by Nano-Anatase TiO2. Nanoscale Res Lett 4:1275–1285. instilled TiO2 nanoparticles. Toxicol 254:82–90.
Martensson J, Meister A. (1991). Glutathione deficiency decreases tissue Wang H, Wick RL, Xing B. (2009). Toxicity of nanoparticulate and bulk
ascorbate levels in new born rats: Ascorbate spares glutathione and ZnO, Al2O3 and TiO2 to the nematode Caenorhabditis elegans.
protects. Proc Natl Acad Sci USA 88:4656–4660. Environ Pollut 157:1171–1177.
Nel A, Xia T, Madler L, Li N. (2006). Toxic potential of materials at the Wang J, Zhou G, Chen C, et al. (2007). Acute toxicity and
nano level. Science 311:622–627. biodistribution of different sized titanium dioxide particles in mice
Oberdörster G. (2000). Toxicology of ultrafine particles: in vivo studies. after oral administration. Toxicol Lett 168:176–185.
Philos Trans R Soc Lond A 358:2719–2740. Xia T, Kovochich M, Nel A. (2006). The role of reactive oxygen species
Oberdörster G, Sharp Z, Atudorei V, et al. (2004). Translocation of and oxidative stress in mediating particulate matter injury. Clin Occup
inhaled ultrafine particles to the brain. Inhal Toxicol 16:437–445. Environ Med 5:817–836.

You might also like