You are on page 1of 16

Author's personal copy

Int. J. Devl Neuroscience 34 (2014) 60–75

Contents lists available at ScienceDirect

International Journal of Developmental Neuroscience


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

Reversibility of changes in brain cholinergic receptors and


acetylcholinesterase activity in rats following early life arsenic
exposure
Lalit P. Chandravanshi a , Rajesh S. Yadav a,c , Rajendra K. Shukla a , Anshuman Singh a ,
Sarwat Sultana b , Aditya B. Pant a , Devendra Parmar a , Vinay K. Khanna a,∗
a
CSIR-Indian Institute of Toxicology Research, Post Box 80, MG Marg, Lucknow, 226 001, India
b
Neurotoxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi, 110 062, India
c
Department of Criminology and Forensic Science, Harisingh Gour University, Sagar,- 470003, India

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

Article history: In view of the increasing incidences of arsenic induced health effects and the vulnerability of the devel-
Received 10 October 2013 oping brain to its toxic effects, studies have been carried out to investigate the mechanism of arsenic
Received in revised form 25 January 2014 induced cholinergic alterations and understand if such changes are persistent or transient on with-
Accepted 31 January 2014
drawal of arsenic exposure. Male rats were exposed to arsenic (2 mg/kg or 4 mg/kg body weight, p.o)
from post-lactational day (PD)22 to PD59, and the effect on selected behavioral and neurochemical end
Keywords:
points associated with cholinergic functions was assessed on PD60 and PD90. Decrease in the binding
Arsenic
of muscarinic-cholinergic receptors in frontal cortex (26%, 43%) and hippocampus (21%, 34%) associated
Learning and memory
Cholinergic receptors
with reduced CHRM2 mRNA levels, acetylcholinesterase activity and expression of ChAT and PKC ␤-1
Oxidative stress was observed in arsenic exposed rats on PD60 as compared to controls. Spatial learning and memory and
Apoptosis muscle strength were affected following arsenic exposure in rats on PD60 and associated with arsenic
induced cholinergic alterations. Enhanced oxidative stress associated with increased expression of pro-
apoptotic proteins and decreased expression of anti-apoptotic proteins was distinct in both frontal cortex
and hippocampus following arsenic exposure in rats on PD60. The cholinergic alterations and other neu-
rochemical modifications were found to be linked with increased arsenic levels in frontal cortex (1.39,
3.90-fold) and hippocampus (3.23, 5.48-fold) on PD60. Although a trend of recovery was observed both in
behavioral and neurochemical endpoints on withdrawal of arsenic exposure on PD90, the results indicate
that continuous arsenic exposure may have detrimental effects.
© 2014 ISDN. Published by Elsevier Ltd. All rights reserved.

1. Introduction throughout the globe (Kris, 2009) which has resulted in increased
health problems (Chen et al., 2011; Hughes et al., 2011). Identi-
Arsenic (As), a metalloid is present in the environment in three fied as a human carcinogen, arsenic is figured number one in the
oxidation states (elemental-0, trivalent-3+ , pentavalent As-5+ ) both category of hazardous chemicals by the US Agency for Toxic Sub-
in inorganic and organic forms (Miteva et al., 2005; Kapaj et al., stances and Disease Registry (ATSDR). Risk of developing cancers
2006; Vahidnia et al., 2007). Due to natural occurrence of arsenic in of kidney, bladder, skin, lungs, breast and liver has been found to
the earth’s crust and moreover, its ubiquitous presence in rocks and increase following arsenic exposure (Heck et al., 2009; Smith et al.,
soil, high levels of arsenic are present in drinking water as a con- 2009, 2012; Yuan et al., 2010; Yorifuji et al., 2011; Du et al., 2012).
taminant in Indo-Gangetic plains, regions of South East Asia and Hyper-pigmentation and skin lesions are frequently reported in
many parts of South America (Argos et al., 2010; Lindberg et al., individuals exposed to arsenic at high levels (Ling et al., 2013). Con-
2010; Steinmaus et al., 2010; Smith et al., 2011). As per an estimate, sumption of arsenic contaminated water has been found to affect
over 140 million people are exposed to arsenic by drinking water early human development, and is therefore, a cause of concern
among the health scientists and exposed population (Hamadani
et al., 2010, 2011; Parvez et al., 2011; Roy et al., 2011; Parajuli et al.,
2013). Arsenic in drinking water has been found to be an etiological
∗ Corresponding author at: Developmental Toxicology Division, CSIR-Indian Insti-
factor for type 2 diabetes in the population of Taiwan, Bangladesh
tute of Toxicology Research, M.G. Marg, P.O. Box No. 80, Lucknow – 226 001 U.P.,
India. Tel.: +91 522 2627586; fax: +91 522 2628227.
and Serbia (Navas-Acien et al., 2006; Jovanović et al., 2012). In view
E-mail address: vkkhanna1@gmail.com (V.K. Khanna). of deleterious health effects associated with arsenic exposure, the

0736-5748/$36.00 © 2014 ISDN. Published by Elsevier Ltd. All rights reserved.


http://dx.doi.org/10.1016/j.ijdevneu.2014.01.007
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 61

maximum limit of arsenic in drinking water has been reduced to during this time. The period PD22–PD60 is pre-adolescent stage
10 ppb by the World Health Organization and US Environmental in rats during which synapse and receptor overproduction occurs
Protection Agency (WHO 2001; US EPA, 2001). and maturation of cholinergic neurotransmitter system takes place
While chronic arsenic poisoning affects the functioning of (Krinke, 2000). However, impact of arsenic exposure during this
respiratory, cardiovascular, gastro-intestinal, genito-urinary and period of brain development on the neurobehavioral outcome is
endocrine systems in humans, the nervous system is more vulner- not understood. Also, it is not known whether changes following
able to its toxic effects (Borenstein et al., 2006; Dauphine et al., arsenic exposure during this period of brain development are tran-
2011; Abhyankar et al., 2012). It has been found that arsenic may sient or persistent. The study has, therefore, been carried out to
affect both the central and peripheral nervous systems causing sub- assess the impact of arsenic exposure from post-lactational day
clinical to clinical effects (Navas-Acien et al., 2006; Kim et al., 2013). (PD)22 to PD59 on selected behavioral and neurochemical end-
Epidemiological studies have revealed an association between points and investigate the changes immediately after exposure
arsenic in drinking water and the risk of cognitive impairment on PD60 and 30 days after withdrawal of arsenic exposure on
including disturbed visual perception and visuomotor integration, PD90.
psychomotor speed, attention, speech and memory (Calderon et al.,
2. Materials and methods
2001; Rodriguez et al., 2003; Tsai et al., 2003). Decrease in the IQ
of children exposed to ground water arsenic has frequently been 2.1. Animals and treatment
reported (Wang et al., 2007). Cognitive deficits and slower nerve
conduction associated with cumulative developmental exposure Male rats (PD21) of Wistar strain obtained from the animal breeding colony
to arsenic have been reported in Taiwanese adolescents (Tsai et al., of CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India were
housed in polypropylene cages. The animals were housed in a temperature con-
2003; Tseng, 2009). Urinary arsenic levels and its metabolites have trolled room (22 ± 2 ◦ C) under standard animal house conditions in a 12-h light–dark
been found to be good indicators to assess the risk of exposure cycle and fed pellet diet and water ad libitum. Rats were divided in to three groups.
of the metal in children and exposed workers (George et al., 2013). On PD22, rats in groups I and II were administered sodium arsenite (either at 2 mg/kg
An inverse association between urinary arsenic levels and cognitive or 4 mg/kg body weight, p.o.) till PD59. Rats in the group III served as controls and
were given normal saline identically as in rats of groups I and II. Effect on behav-
functions in children has been shown by many investigators, sug-
ioral parameters – grip strength, learning and continuous alternation was studied
gesting that arsenic exposure during development can affect the 24 h after the last dose of arsenic treatment on PD60. A separate set of rats in each
functioning of the central nervous system (Calderon et al., 1999, group was sacrificed on PD90 and brain was taken out quickly and washed in ice
2001; Tsai et al., 2003; Hamadani et al., 2011). A number of exper- cold saline. Brain was dissected into specific regions – frontal cortex and hippocam-
imental studies on rats and mice have been carried out to assess pus following the standard procedure as described by Glowinski and Iversen (1966)
and kept frozen at −80 ◦ C for neurochemical assays and estimation of arsenic lev-
the impact of arsenic on neurobehavioral performance (Luo et al.,
els. To understand whether neurobehavioral changes following arsenic exposure
2009; Bardullas et al., 2009; Rodriguez et al., 2010; Yadav et al., are transient or persistent, a set of rats was left as such till PD89 and monitored
2011). It was found that arsenic exposure leads to neurochemi- daily. Effect on selected behavioral and neurochemical endpoints was studied on
cal alterations associated with impaired motor activity, learning PD90.
and memory functions (Itoh et al., 1990; Nagaraja and Desiraju,
1994; Rodriguez et al., 2001, 2003). Enhanced oxidative stress asso- 3. Behavioral studies
ciated with increased apoptosis was found to be linked to arsenic
induced neurotoxicity (Estus et al., 1994; Namgung and Xia, 2001; 3.1. Grip strength
Ahmed et al., 2011, 2012). Arsenic has been found to cross the
placental barrier and cause developmental abnormalities includ- Forelimb grip strength was measured in the control and arsenic
ing malformations, decreased growth rate, mortality and neural exposed rats using a computerized grip strength meter (TSE,
tube defects (Golub, 1994; Tabocova et al., 1996; Stump et al., Germany) following the standard procedure as described by Terry
1999). In view of increasing risk of exposure to arsenic and asso- et al. (2003). Rats were gently held by the nape of the neck and base
ciated vulnerabilities of the developing brain, studies have been of the tail. The forelimbs of the rats were placed on the tension bar.
carried out to assess the impact of arsenic exposure during pre- Followed by this, the rat was pulled back gently until it released the
natal and early post-natal periods (Anderson et al., 2000; Xi et al., bar and the reading was recorded automatically on the computer.
2010a; Lu et al., 2012; Herrera et al., 2013). In a study to assess Five successive pulls for each animal in the study group were tried
the developmental toxicity of arsenic acid, Nemec et al. (1998) by a person unaware of their treatment status. The mean of all the
found that pentavalent arsenic was equally toxic to mothers and values was taken and processed for statistical analysis.
developing offspring both in mice and rabbits. Further, it is well
reported that the risk of spontaneous abortions, still births and 3.2. Learning ability – active avoidance test
increased risk of infant mortality is enhanced in pregnant women
consuming drinking water contaminated with high arsenic lev- The learning ability of rats was measured by assessing con-
els (Vahter et al., 2006; Von Ehrenstein et al., 2006; Huyck et al., ditioned avoidance behavior using a shuttle box (Techno, India)
2007; Rahman et al., 2007, 2010). In a hospital based study on following the standard protocol as described by Yadav et al. (2009).
Nepalese, Parajuli et al. (2013) found an inverse relation with cord Briefly, rats from the control and arsenic treated groups were placed
blood arsenic levels and neurodevelopment indicators in neonates, individually in one chamber of the shuttle box and habituated for
suggesting that in utero exposure to arsenic could be detrimen- 5 min. A conditioned stimulus in the form of a buzzer for 10 s fol-
tal. Tranplacental and early life exposure to arsenic was found to lowed by a buzzer and foot shock of 0.5 mA for 10 s was given to
impair learning and memory and sensory reflexes (Xi et al., 2009). each rat. The avoidance was considered positive if the rats jumped
While studies to assess the impact of arsenic on neurobehavioral to a shock-free chamber to avoid the foot shock. All rats in the treat-
performance following prenatal and early postnatal life have been ment and control groups were subjected to 20 trials per day for 3
carried out, direct exposure to arsenic could also occur in devel- days. The inter-trial interval was 1 min. Of the 20 trials per day,
oping children during post-lactational period. Exposure to arsenic the total number of avoidances and the number of trials for the
during early postnatal life may also affect the neurobehavioral per- first avoidance were recorded for each rat. The data of the final day
formance (Martínez et al., 2011; Luo et al., 2013). Prenatal and are presented in the results. The percentage of conditioned avoid-
lactational periods in rats are critical and crucial for brain develop- ance response has been recorded as a measure of learning ability
ment as structural and functional maturation takes place primarily in treated groups and compared with controls.
Author's personal copy

62 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

3.3. Spatial memory – continuous alternation test RT-PCR (ABI, USA) using the glyceraldehydes 3-phosphated
dehydrogenase (GAPDH) for normalization. The cDNA was
Continuous alternation in a single session was measured using a amplified by RT-PCR using ABI SYBER green qPCR Kit (ABI,
computerized Y-maze (TSE, Germany), to assess short term spatial USA) on the 7900HT Fast Real-Time PCR System running at
memory (Maurice et al., 1994; Roghani et al., 2006). The equip- 40 cycles following the protocol: one cycle of denaturation
ment consisting of three arms – 40 cm long, 15 cm wide and 30 cm (95 ◦ C, 10 s), followed by 40 cycles of denaturation (95 ◦ C,
high converged in an equilateral triangular central area with 15 cm 10 s) annealing (60 ◦ C, 10 s) and extension (72 ◦ C, 45 s). A final
at its longest axis. Rat in each case was placed at the end of one extension (72 ◦ C, 5 min) was performed. The primer sequences
arm and allowed to move freely through the maze for 5 min. The used for CHRM2 and GAPDH described earlier (Borges et al.,
sequence of entry in to each arm was recorded automatically on 2001) are mentioned below, CHRM2, FP5 -GGCAAGCAAGAGTAG
the computer. Measure of spatial memory has been defined as the AATAAA-3 and CHRM2, RP 5 -GCCAACAGGATAGCCAAGATT-3
number and the sequence of entries of rat in to the arm recorded GAPDH, FP 5 -CGGGAAGCTTGTGATCAATGG-3 and GAPDH, RP 5 -
during 5 min. All the arms look alike and no reinforcer, motivation GGCAGTGATGCCATGGACT G-3 .
or punishment was used in this set of experiment. The percentage
of alternation has been calculated as the number of alterna-
tions (entries into three different arms consecutively) divided by 4.3. Assay of brain acetylcholinesterase activity
the total possible alternations (i.e. the number of arms entered
minus 2). Activity of acetylcholinesterase was assayed following the pro-
cedure as described by Ellman et al. (1961) using acetylthiocholine
iodide as a substrate and 5,5 -dithiobis-2 nitrobenzoic acid (DTNB)
4. Neurochemical studies as the coloring agent. Briefly, the reaction mixture in a final
volume of 1.0 ml contained phosphate buffer (0.1 M, pH 7.4),
4.1. Assay of brain muscarinic-cholinergic receptors post-mitochondrial fraction of frontal cortex or hippocampus (con-
taining around 15 ∼ 20 ␮g protein), acetylthiocholine iodide (ACTI),
Assay of muscarinic-cholinergic receptors in frontal cortex and and DTNB (5 mM). The degradation of acetylthiocholine iodide was
hippocampus was carried out by radioligand receptor binding fol- measured at 412 nm and the results are expressed as ␮mol ACTI
lowing the procedure as described in detail by Yadav et al. (2011). hydrolysed/min/mg protein.
Briefly, crude synaptic membrane was prepared by homogenizing
the frontal cortex and hippocampus individually in 19 volumes of
Tris–HCl buffer (5 mM, pH 7.4). The homogenate was centrifuged at 4.4. Assessment of brain oxidative stress:
40,000 × g for 15 min at 4 ◦ C and the sedimented pellet was washed
twice by resuspending in homogenization buffer. The suspended 4.4.1. Assay of lipid peroxidation, protein carbonyl and reduced
pellet in buffer was recentrifuged at the same speed for 15 min at glutathione levels
4 ◦ C and finally suspended in Tris–HCl buffer (40 mM, pH 7.4) and As a measure of lipid peroxidation, malonaldehyde (MDA) for-
stored at −20 ◦ C till binding assays. mation was estimated in brain regions following the method of
Binding incubations in triplicate were carried out in a final vol- Ohkawa et al. (1979) and the intensity of pink color formed dur-
ume of 1.0 ml. For the assay of muscarinic-cholinergic receptors, ing the reaction was read at 532 nm.The values are expressed
3 H-QNB (42 Ci/mmol, Perkin Elmer, USA) was used as a radioli- as ␮mol/hour/mg protein Protein carbonyl content in brain
gand and atropine sulfate (1 × 10−6 M, Sigma, USA) was used as regions was measured following the procedure of Levine et al.
a competitor to assess the extent of non-specific binding. The reac- (1990) using 2,4-dinitrophenylhydrazine (DNPH) as a substrate.
tion mixture containing Tris–HCl buffer (40 mM, pH 7.4) together Amount of protein carbonyl was calculated using a molar extinc-
with membrane protein (300–400 ␮g) and 3 H-QNB (1 × 10−9 M) tion coefficient (D) 22 mm−1 cm−1 for aliphatic hydrazine and
was incubated for 15 min at 37 ◦ C in the presence or absence of values are expressed as n moles/mg protein. Reduced glu-
competitor to assess the non-specific and total binding respec- tathione (GSH) levels both in frontal cortex and hippocampus
tively. At the end of incubation, contents of the binding tubes were measured spectrophotometrically using 5,5 -dithiobis (2-
were immediately filtered on glass fiber discs (25 mm diameter, nitrobenzoic acid) (DTNB) as the color reagent following the
0.3 ␮m pore size, Whatman GF/B) and washed twice rapidly with method of Hasan and Haider (1989). A range (2–10 ␮g) of
5 ml chilled Tris–HCl buffer (40 mM, pH 7.4). Filters were dried and glutathione as standard was also run in parallel to plot the stan-
transferred into vials and scintillation cocktail containing the mix- dard curve and the values are expressed as ␮g GSH/g tissue
ture (2,5-diphenyl oxazole; 1,4-bis-5, phenylozazolyl-benzene; weight.
naphthalene; toluene; methanol and 1,4 dioxane) added. The
radioactivity was counted using ␤-scintillation counter (Packard,
4.4.2. Assay of superoxide dismutase, catalase and glutathione
USA) at an efficiency of 30–40% for 3 H to determine membrane
peroxidase activity
bound radioactivity. Specific binding has been determined by
Activity of superoxide dismutase (SOD) in frontal cortex and hip-
subtracting the non-specific binding from the total binding and
pocampus was assayed following the method of Kakkar et al. (1984)
expressed as pmoles ligand bound/g protein. Scatchard analysis has
using NADH as a substrate in the post-mitochondrial fraction. The
been carried out at varying concentrations of 3 H-QNB (ranging from
SOD activity has been expressed as units/min/mg protein. Activity
0.25 nM to 4 nM) to ascertain whether change in the binding is due
of catalase in both the brain regions was assayed spectrophotomet-
to alteration in the affinity (Kd) or number of receptor binding sites
rically following the method of Aebi (1984) in post-mitochondrial
(Bmax).
fraction using hydrogen peroxide (H2 O2 ) as substrate. The activity
of the enzyme was calculated using the molar extinction coefficient
4.2. mRNA expression of brain muscarinic-cholinergic receptor (D) 43.6 M cm−1 and has been expressed in ␮mole/min/mg protein.
(CHRM2) gene Activity of glutathione peroxidase (GPx) both in frontal cortex and
hippocampus was measured following the procedure of Flohe and
CHRM2 receptor gene expression in frontal cortex and hip- Gunzler (1984). The activity of the enzyme has been expressed as
pocampus of arsenic exposed rats has been determined by nmol GSH oxidized/min/mg protein.
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 63

5. Immunoexpression of proteins by Western blotting Table 1


Early life exposure (PD22–PD59) to sodium arsenite and effect on body and brain
weights of rats on PD60 and PD90.
5.1. Expression of ChAT, PKC ˇ-1 and selected proteins associated
with apoptosis Age in postnatal day

Treatment groups PD60 PD90


Expression of choline acetyltransferase, PKC ␤-1 and proteins
Body weight (gm)
linked with apoptosis and stress (Caspase-3, Cleaved Caspase- Control 120.7 ± 6.50 138.3 ± 4.13
3, Bax, Bcl2, phospho-p38, phospho-JNK, Ap-1) in frontal cortex ARS I (2 mg/kg) 112.7 ± 4.50 131.0 ± 3.97
and hippocampus was assayed following the method of Jamal ARS II (4 mg/kg) 101.3 ± 5.9* 126.0 ± 3.62
et al. (2007). Briefly, brain regions were homogenized in RIPA Brain weight (gm)
buffer containing Tris–HCl (50 mM, pH 6.8), NaCl (150 mM), sodium Control 1.51 ± 0.01 1.55 ± 0.02
deoxycholate (0.5%), SDS (0.1%), protease inhibitor and Triton- ARS I (2 mg/kg) 1.45 ± 0.02 1.51 ± 0.02
1.47 ± 0.01 1.49 ± 0.02
100X (1%) and centrifuged at 12,000 × g (15 min, at 4 ◦ C) to remove ARS II (4 mg/kg)

the insoluble material. The pellet was discarded and the super- Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body
natant further mixed with loading buffer containing Tris–HCl weight/day, p.o.) from PD22 to PD59. Effect on body and brain weight parameters
studied on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90.
(60 mM, pH 6.8), SDS (2%), glycerol (10%), ␤-mercaptoethanol
Values are mean ± SEM of 15 animals in each group for body weight and five ani-
(5%), bromophenol blue (0.01%) and boiled for 5 – 7 min. The
mals in each group for brain weight. Data have been analyzed by one-way analysis
prepared samples (30 ␮g protein/lane) were electrophoresed on of variance followed by Newman–Keuls test.
12% SDS–PAGE, electroblotted on to nitrocellulose membranes *
Significantly differs from controls (p < 0.05).
(Millipore, USA) and blocked with blocking buffer (Western
blocker solution TM Sigma, USA). After subsequent washing, the
6. Results
blots were incubated with primary antibody (Anti-TH, Sigma,
USA, 1:2000 and Anti-ChAT, Sigma, USA, 1:2000) for 24 h at
To assess if changes in the behavioral and neurochemical param-
4 ◦ C followed by incubation with horseradish peroxidase linked
eters are transient or persistent, the effect of arsenic on body and
secondary antibody (anti-mouse IgG, 1:4000) at room temper-
brain weights and selected behavioral and neurochemical end-
ature for 60 min. After the incubation, blots were washed and
points was studied in rats following early life exposure from PD22
developed using an immobilon western chemiluminescent HRP
to PD59 on PD60. The effect was further monitored, 30 days after
substrate (Millipore, USA) following the recommended procedure.
withdrawal of arsenic exposure on PD90.
␤-actin was probed as an internal control and used to confirm
that an equal amount of protein was loaded in each lane. A
6.1. Effect of arsenic on body and brain weight of rats
digital gel image analysis system (VersaDoc, Model 1000, Bio
Rad,Quantity 1) was used for semi-quantification of immunoreac-
Early life exposure to arsenic (2.0 mg/kg or 4.0 mg/kg body
tivity.
weight) caused a decrease in the body weight (F(2,42) = 3.101, 6%,
p > 0.05; 15%, p < 0.05) of rats with significant changes in those
5.2. Estimation of arsenic levels in brain regions exposed at high dose of arsenic on PD60 as compared to con-
trols exhibiting impairment in the growth of developing rats
Arsenic levels in both brain regions were estimated using the (Table 1). However, a trend of recovery was observed in the body
hydride system 60 atomic absorption spectrophotometer (HSAAS, weight (F(2,42) = 2.511, 5%, p > 0.05; 8%, p > 0.05) of rats exposed to
ZEEnit 700) following the method of Ballentine and Burford (1957). arsenic at both the doses, 30 days after withdrawal of exposure on
Briefly, concentrated nitric acid (1 ml) and perchloric acid (1 ml) PD90.
were added in the flask containing tissue (100 mg approximately). No significant change in the brain weight was observed in rats
The sample was then digested over a sand bath until the solu- exposed to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) during
tion becomes yellow in color. If the color of the digest was early life on PD60 and 30 days after withdrawal of exposure on
brown, more nitric acid and perchloric acid were added and the PD90 as compared to respective controls (Table 1).
process of oxidation was repeated. The digest was made up to
known volume with deionized water and aliquots were used
6.2. Behavioral studies
to estimate arsenic using atomic absorption spectrophotometer.
A calibration curve was constructed by adding known amounts
6.2.1. Effect on grip strength
of arsenic standard (Sigma) to calculate arsenic levels in brain
To assess the effect on muscle weakness if any, fore limb grip
regions.
strength was monitored following early life exposure of rats from
PD22 to PD59 to arsenic (2.0 mg/kg or 4.0 mg/kg body weight). A
5.3. Protein estimation significant decrease in the forelimb grip strength (F(2,12) = 13.81,
18%, p < 0.01; 30%, p < 0.001) was observed in rats exposed to arsenic
Protein content in samples was measured following the method at both the doses (2.0 mg/kg or 4.0 mg/kg body weight) respectively
of Lowry et al. (1951) using bovine serum albumin as a reference on PD60 in comparison to controls (Fig. 1). Interestingly, a trend
standard. of recovery in the grip strength was observed (F(2,12) = 1.951, 3%,
p > 0.05; 8%, p > 0.05) in arsenic exposed rats 30 days after with-
5.4. Statistical analysis drawal of exposure on PD90 in comparison to respective controls
(Fig. 1).
Graph Pad prism software has been used to analyze data
of neurobehavioral, neurochemical and immunoblotting end- 6.2.2. Effect on learning ability – active avoidance test
points. The data have been analyzed by one-way analysis Effect of arsenic exposure on learning and memory was assessed
of variance (ANOVA) followed by Newman–Keuls test for by monitoring the active avoidance response using shuttle box and
posthoc comparisons. Values up to p < 0.05 have been considered the results are presented in Fig. 2A. Impairment in the learning and
significant. memory (F(2,12) = 31.94, 22%, p < 0.01; 50%, p < 0.001) was observed
Author's personal copy

64 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

6.2.3. Effect on spatial memory – continuous alternation test


Effect on the spatial memory was assessed by monitoring the
continuous alternation test using Y-Maze in rats exposed to arsenic
during early life and the results are presented in Fig. 2B. A signifi-
cant decrease in the spatial memory (F(2,14) = 19.23, 35%, p < 0.01;
59.6%, p < 0.001) was observed in rats exposed to arsenic dur-
ing early life from PD22 to PD59 at both the doses (2.0 mg/kg or
4.0 mg/kg body weight) on PD60 as compared to rats in the control
group (Fig. 2B). Although a trend of recovery in the spatial mem-
ory was observed (F(2,14) = 2.364, 15%, p > 0.05; 23%, p < 0.05) in
arsenic exposed rats 30 days after withdrawal of exposure on PD90,
the decrease was found to persist in rats exposed to arsenic at the
higher dose (Fig. 2B).
Fig. 1. Early life exposure (PD22–PD59) to sodium arsenite and effect on grip
strength of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 7. Neurochemical studies
4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on grip strength assessed
immediately after exposure to sodium arsenite on PD60 and 30 days after with-
Effect on muscarinic-cholinergic receptors, expression of
drawal of sodium arsenite exposure on PD90. Values are mean ± SEM of five animals
in each group. Data have been analyzed by one-way analysis of variance followed CHRM2 receptor gene and acetylcholinesterase activity associated
by Newman–Keuls test. *Significantly differs from controls (p < 0.01); **significantly with the integrity of brain cholinergic system was assessed follow-
differs from controls (p < 0.001). ing early life exposure from PD22–PD59 to arsenic (2.0 mg/kg or
4.0 mg/kg body weight) on PD60 and 30 days after withdrawal of
arsenic exposure on PD90 and the results are described below:
in rats exposed to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) dur-
ing early life as compared to controls (Fig. 2A). The decrease in the 7.1. Effect on brain muscarinic-cholinergic receptors
learning and memory was more marked in rats treated with arsenic
at the higher dose. It was interesting to observe a trend of recovery Fig. 3A illustrates effect of early life arsenic exposure of rats
in learning and memory (F(2,13) = 3.25, 6%, p > 0.05; 10%, p > 0.05) on brain muscarinic-cholinergic receptors. A significant decrease
in arsenic exposed rats 30 days after withdrawal of exposure on in the binding of 3 H-QNB to frontocortical (F(2,10) = 15.74, 26%,
PD90 as compared to respective controls (Fig. 2A). p < 0.01; 43%, p < 0.01), and hippocampal (F(2,9) = 7.792, 21%,
p < 0.05; 34%, p < 0.01) membranes, known to label muscarinic-
cholinergic receptors was observed in arsenic treated rats on PD60
as compared to controls (Fig. 3A). Scatchard analysis revealed that
decrease in the binding of 3 H-QNB to frontocortical and hippocam-
pal membranes was due to decreased number of binding sites
(Table 2). A trend of recovery was observed in the binding of
3 H-QNB to frontocortical and hippocampal membranes in arsenic

treated rats on PD90 as compared to respective controls (Fig. 3A).

7.2. Effect on mRNA expression CHRM2 receptor gene

Early life exposure of rats to arsenic (2.0 mg/kg or 4.0 mg/kg


body weight) caused a significant decrease in the expression
of CHRM2 receptor genes in frontal cortex (F(2,6) = 39.46, 29%,
p < 0.01; 48%, p < 0.001) and hippocampus (F(2,6) = 32.00, 23%,
p < 0.05; 43%, p < 0.001) on PD60 as compared to controls. These
changes exhibited a trend of recovery 30 days after withdrawal of
arsenic exposure as compared to respective controls (Fig. 3B).

7.3. Effect on brain acetylcholinesterase activity

Early life exposure of rats from PD22 to PD59 to arsenic


(2.0 mg/kg or 4.0 mg/kg body weight) caused a significant decrease
in the activity of acetylcholinesterase (AChE) in frontal cor-
tex (F(2,9) = 7.519, 24%, p < 0.05; 39%, p < 0.05) and hippocampus
(F(2,12) = 22.35, 26%, p < 0.001; 42%, p < 0.001) on PD60 as com-
pared to controls (Fig. 4). A trend of recovery in the activity of AChE
Fig. 2. Early life exposure (PD22–PD59) to sodium arsenite and effect on learning both in frontal cortex (F(2,10) = 1.040, 6%, p > 0.05; 14%, p > 0.05)
and memory assessed by active avoidance response (A) and continuous alternation and hippocampus (F(2,10) = 1.457, 10%, p > 0.05; 14%, p > 0.05) was
test (B) of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg
observed 30 days after withdrawal of exposure on PD90 in com-
body weight/day, p.o.) from PD22 to PD59. Effect on active avoidance response (A)
and continious alternation test (B) assessed immediately after exposure to sodium
parison to respective controls (Fig. 4).
arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on
PD90. Values are mean ± SEM of four to five animals in each group in the data pre- 7.4. Assessment of oxidative stress
sented for active avoidance response (A) and five to six animals in each group in
the data presented for continuous alternation test (B). Data have been analyzed by
one-way analysis of variance followed by Newman–Keuls test. *Significantly differs To assess the extent of oxidative stress following early life expo-
from controls (p < 0.01); **significantly differs from controls (p < 0.001). sure to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) in rats, effect
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 65

Fig. 3. Early life exposure (PD22–PD59) to sodium arsenite and effect on muscarinic-cholinergic receptors (A) and CHRM2 mRNA levels (B) in frontal cortex and hippocampus
of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on muscarinic-cholinergic receptors (A)
and CHRM2 mRNA levels (B) assessed immediately after exposure to sodium arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90. Values
are mean ± SEM of four to five animals in each group in the data presented for muscarinic-cholinergic receptors (A) and three animals in each group in the data presented
for CHRM2 mRNA levels (B). Data have been analyzed by one-way analysis of variance followed by Newman–Keuls test. *Significantly differs from controls (p < 0.01);
**significantly differs from controls (p < 0.001).

Fig. 4. Early life exposure (PD22–PD59) to sodium arsenite and effect on the acetylcholinestrase activity in frontal cortex and hippocampus of rats. Rats were exposed to
sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on acetylcholinestrase activity assessed immediately after exposure to
sodium arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90. Values are mean ± SEM of four to five animals in each group. Data have been
analyzed by one-way analysis of variance followed by Newman–Keuls test. *Significantly differs from controls (p < 0.05); **significantly differs from controls (p < 0.001).
Author's personal copy

66 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

Table 2
Scatchard analysis of 3 H-QNB to frontocortical and hippocampal membranes in rats exposed to sodium arsenite during early life (PD22–PD59).

Age in postnatal day

Treatment groups PD60 PD90

Kd Bmax Kd Bmax

Frontal cortex
Control 1.56 ± 0.013 737.9 ± 84 1.96 ± 0.12 805 ± 58
ARS I (2 mg/kg) 1.83 ± 0.18 592 ± 54* 2.06 ± 0.14 764 ± 46
ARS II (4 mg/kg) 2.09 ± 0.18 459 ± 48** 2.11 ± 0.21 749 ± 57

Hippocampus
Control 1.48 ± 0.09 816.6 ± 79 1.67 ± 0.10 993.6 ± 80
ARS I (2 mg/kg) 1.74 ± 0.14 646.9 ± 65* 1.75 ± 0.11 884.6 ± 91
ARS II (4 mg/kg) 1.93 ± 0.18 465.1 ± 53** 1.82 ± 0.12 841.4 ± 68

Values are mean ± SEM of three observations in each group. Kd – dissociation constant expressed in nM, Bmax – maximum number of binding sites expressed in pmoles
3
H-QNB bound/g protein. Data have been analyzed by one-way analysis of variance followed by Newman–Keuls test.
*
Significantly differs from controls (p < 0.05).
**
Significantly differs from controls (p < 0.01).

on lipid peroxidation and protein carbonyls implicated as pro- 7.4.2. Effect on superoxide dismutase, catalase and glutathione
oxidants and on the activity of superoxide dismutase, catalase and peroxidase activity
glutathione peroxidase and glutathione levels associated with anti- A significant decrease in the activity of superoxide dismu-
oxidant defense was studied immediately after arsenic exposure on tase was observed in frontal cortex (F(2,10) = 50.4039%, p < 0.001;
PD60 and 30 days after withdrawal of arsenic exposure on PD90 and 55%, p < 0.001) and hippocampus (F(2,12) = 48.09, 46%, p < 0.001;
the results are presented below: 65%, p < 0.001) following early life exposure of rats to arsenic
(2.0 mg/kg or 4.0 mg/kg body weight) in comparison to con-
trols (Fig. 6A). A trend of recovery in the activity of superoxide
7.4.1. Effect on brain lipid peroxidation, protein carbonyl levels dismutase was observed 30 days after withdrawal of exposure
and reduced glutathione levels on PD90 both in frontal cortex (F(2,12) = 0.9796, 5%, p > 0.05;
Levels of lipid peroxidation, protein carbonyl and reduced glu- 12%, p > 0.05) and hippocampus (F(2,9) = 5.670, 12%, p > 0.05; 30%,
tathione were measured both in hippocampus and frontal cortex p < 0.05) of arsenic exposed rats as compared to respective controls
to assess the extent of oxidative stress. Early life exposure of rats to (Fig. 6A).
arsenic (2.0 mg/kg or 4.0 mg/kg body weight) caused a significant Rats treated with arsenic (2.0 mg/kg or 4.0 mg/kg body weight)
increase in the TBARS levels associated with enhanced lipid peroxi- exhibited a decrease in the activity of catalase in frontal cor-
dation in frontal cortex (F(2,9) = 27.23, 40%, p < 0.05; 72%, p < 0.001) tex (F(2,11) = 13.01, 25%, p < 0.01; 43%, p < 0.01) and hippocampus
and hippocampus (F(2,9) = 14.33, 41%, p < 0.05; 72%, p < 0.01) on (F(2,11) = 19.47, 26%, p < 0.05; 57%, p < 0.01) in comparison to con-
PD60 as compared to controls (Fig. 5A). Increase in the TBARS trols (Fig. 6B). The activity of catalase remained decreased in arsenic
levels was more marked in rats exposed to arsenic at a higher treated rats even after withdrawal of exposure for 30 days although
dose (4.0 mg/kg body weight) suggesting enhanced lipid peroxi- the changes were not significant in frontal cortex (F(2,9) = 4.922,
dation in the brain. Although a trend of recovery in TBARS levels 13%, p < 0.05; 29%, p < 0.05) and hippocampus (F(2,9) = 5.260, 15%,
was observed both in frontal cortex (F(2,9) = 5.176, 28%, p > 0.05; p > 0.05; 22%, p < 0.05) in comparison to respective controls (Fig. 6B).
40%, p < 0.05) and hippocampus (F(2,9) = 4.004, 19%, p > 0.05; 41%, A significant decrease in the activity of glutathione peroxi-
p < 0.05) 30 days after withdrawal of exposure, levels of TBARS dase was observed in frontal cortex (F(2,9) = 8.45, 28%, p < 0.05;
remained significantly higher both in frontal cortex and hippocam- 40%, p < 0.01) and hippocampus (F(2,11) = 11.26, 32%, p < 0.01; 57%,
pus of rats exposed to arsenic at a higher dose as compared to p < 0.001) of rats following their exposure to arsenic (2.0 mg/kg or
respective controls (Fig. 5A). 4.0 mg/kg body weight) from PD22 to PD59 as compared to con-
Exposure to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) from trols on PD60 (Fig. 6C). The activity of glutathione peroxidase in
PD22 to PD59 significantly increased the levels of protein carbonyl arsenic treated rats exhibited a trend of recovery in frontal cor-
in frontal cortex (F(2,12) = 14.10, 35%, p < 0.01; 68%, p < 0.001) and tex (F(2,10) = 3.55, 7%, p > 0.05; 15%, p > 0.05) and hippocampus
hippocampus (F(2,12) = 15.02, 37%, p < 0.05; 78%, p < 0.01) in com- (F(2,9) = 1.78, 5%, p > 0.05; 14%, p < 0.05) 30 days after withdrawal
parison to controls (Fig. 5B). Although a trend of recovery in protein of exposure in the arsenic treated rats as compared to respective
carbonyl levels was observed both in frontal cortex (F(2,12) = 5.47, controls (Fig. 6C).
27%, p > 0.05; 48%, p < 0.05) and hippocampus (F(2,12) = 4.883, 19%,
p > 0.05; 46%, p < 0.05) 30 days after withdrawal, protein carbonyl 8. Immunoexpression of proteins by Western blotting
levels remained significantly higher in rats exposed to arsenic at the
higher dose on PD90 in comparison to respective controls (Fig. 5B). Effect on the expression of selected proteins associated with
Decrease in the levels of reduced glutathione in frontal cortex the cholinergic circuitry and those involved in pro-apoptotic, anti-
(F(2,12) = 33.62, 28%, p < 0.001; 40%, p < 0.001), and hippocampus apoptotic and stress pathways was assessed following early life
(F(2,9) = 9.24, 31%, p < 0.05; 49%, p < 0.01) was observed in rats exposure from PD22–PD59 to arsenic (2.0 mg/kg or 4.0 mg/kg body
treated with arsenic (2.0 mg/kg or 4.0 mg/kg body weight) during weight) in rats to investigate their involvement in arsenic induced
early life as compared to controls (Fig. 5C). A trend of recovery in neurotoxicity and results are presented below:
reduced glutathione was observed in frontal cortex (F(2,10) = 6.79,
9%, p > 0.05; 17%, p < 0.05) and hippocampus (F(2,12) = 3.891, 8%, 8.1. Effect on the expression of brain ChAT and PKC ˇ-1 proteins
p > 0.05; 21%, p < 0.05) of arsenic exposed rats 30 days after with-
drawal of exposure on PD90 in comparison to respective controls A decrease in the expression of ChAT protein in frontal cor-
(Fig. 5C). tex (F(2,6) = 59.79, 1.34, 1.44-fold) and hippocampus (F(2,6) = 167.8,
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 67

Fig. 5. Early life exposure (PD22–PD59) to sodium arsenite and effect on lipid peroxidation levels (A), protein carbonyl levels (B) and reduced glutathione levels (C) in frontal
cortex and hippocampus of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on these weight
parameters assessed immediately after exposure to sodium arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90. Values are mean ± SEM of
four to five animals in each group. Data have been analyzed by one-way analysis of variance followed by Newman–Keuls test. *Significantly differs from controls (p < 0.05);
**significantly differs from controls (p < 0.01); ***significantly differs from controls (p < 0.001).

1.60, 1.84-fold) was clearly evident following early life expo- 8.2. Effect on the expression of pro-apoptotic (Bax, Caspase-3,
sure of rats to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) Cleaved Caspase-3) and anti-apoptotic (Bcl-2) proteins
from PD22 to PD59 in comparison to controls on PD60 (Fig. 7A).
No significant effect on the expression of ChAT protein both in A significant increase in the Bcl-2/Bax protein ratio in frontal
frontal cortex and hippocampus was observed in rats 30 days cortex (F(2,6) = 50.18, 1.47-fold, p < 0.01; 2.19-fold, p < 0.001)
after withdrawal of exposure as compared to respective controls and hippocampus (F(2,6) = 76.42, 1.63-fold, p < 0.001; 2.61-fold,
(Fig. 7A). p < 0.001) was observed in arsenic (2.0 mg/kg or 4.0 mg/kg body
Early life exposure to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) exposed rats on PD60 as compared to controls. In spite of a
weight) from PD22 to PD59 in rats caused a significant decrease in trend of recovery observed both in frontal cortex and hippocampus,
the expression of PKC ␤-1 protein in hippocampus (F(2,6) = 58.30, ratio of Bcl-2/Bax remained increased in both the brain regions of
1.5-fold, p < 0.001; 2.10-fold, p < 0.001) on PD60 as compared to rats exposed to arsenic at the higher dose 30 days after withdrawal
controls (Fig. 7B). No significant effect on the expression of PKC of exposure on PD90 in comparison to respective controls (Fig. 8A).
␤-1 protein in hippocampal region was observed in rats 30 days A significant increase in the expression of Caspase-3 pro-
after withdrawal of arsenic exposure as compared to respective tein in frontal cortex (F(2,6) = 309.8, 1.27-fold, p < 0.001; 1.44-fold,
controls (Fig. 7B). p < 0.001) and hippocampus (F(2,6) = 167.3, 1.35-fold, p < 0.001;
Author's personal copy

68 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

Fig. 6. Early life exposure (PD22–PD59) to sodium arsenite and effect on superoxide dismutase (A), catalase (B) and glutathione peroxidase (C) activity in frontal cortex and
hippocampus of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on these parameters assessed
immediately after exposure to sodium arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90. Values are mean ± SEM of four to five animals
in each group. Data have been analyzed by one-way analysis of variance followed by Newman–Keuls test. *Significantly differs from controls (p < 0.05); **significantly differs
from controls (p < 0.01); ***significantly differs from controls (p < 0.001).

2.01-fold, p < 0.001) was observed on PD60 in arsenic (2.0 mg/kg 8.3. Effect on the expression of stress marker (phospho-p38, AP-1
or 4.0 mg/kg body weight) exposed rats in comparison to controls and phospho-JNK) proteins
(Fig. 8A). Exposure to arsenic also resulted to increase the expres-
sion of Cleaved Caspase-3 in frontal cortex (F(2,6) = 79.7, 1.52-fold, Expression of phospho-p38 protein in frontal cortex
p < 0.05; 2.84-fold, p < 0.001) and hippocampus (F(2,6) = 11.35, 2.33- (F(2,6) = 69.0, 1.41-fold, p < 0.001; 1.66-fold, p < 0.001) and hip-
fold, p < 0.01; 2.50-fold, p < 0.05) on PD60 as compared to rats in the pocampus (F(2,6) = 38.2, 1.78-fold, p < 0.01; 2.07-fold, p < 0.001)
control group (Fig. 8B). While a trend of recovery was observed in was found increased in arsenic (2.0 mg/kg or 4.0 mg/kg body
the expression of Caspase-3 and Cleaved Caspase-3, 30 days after weight) exposed rats in comparison to controls on PD60 (Fig. 9A).
withdrawal of exposure, changes were found to persist both in Change in the expression was more marked in both brain regions
frontal cortex and hippocampus of rats exposed to arsenic at higher at the higher dose of arsenic. A trend of recovery in the expres-
dose on PD90 in comparison to respective controls (Fig. 8A and B). sion of phospho-p38 both in frontal cortex and hippocampus
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 69

Fig. 7. Early life exposure (PD22–PD59) to sodium arsenite and effect on the expression of PKC ␤-1 (A) and ChAT (B). Rats were exposed to sodium arsenite (ARS I – 2.0 or
ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on the expression of these proteins studied immediately after exposure to sodium arsenite on PD60 and
30 days after withdrawal of sodium arsenite exposure on PD90. Values are mean ± SEM of three animals in each group. Data have been analyzed by one-way analysis of
variance followed by Newman–Keuls test. *Significantly differs from controls (p < 0.001).

was observed 30 days after withdrawal of exposure on PD90 in weight) during early life from PD22 to PD59 on PD60 and PD90 both
comparison to respective controls (Fig. 9A). in frontal cortex and hippocampus (Fig. 9B).
Exposure to arsenic (2.0 mg/kg or 4.0 mg/kg body weight)
resulted to increase the expression of AP-1 in frontal cortex 9. Effect on arsenic levels
(F(2,6) = 73.5, 1.26-fold, p < 0.001; 1.51-fold, p < 0.001) and hip-
pocampus (F(2,6) = 47.1, 1.45-fold, p < 0.001; 1.67-fold, p < 0.001) on A significant increase in the levels of arsenic in frontal cortex
PD60 as compared to controls (Fig. 9A). Increase in the expression (F(2,9) = 26.10, 1.39-fold, p < 0.05; 3.90-fold, p < 0.001), and hip-
of AP-1 was more marked both in frontal cortex and hippocam- pocampus (F(2,6) = 65.02, 3.23-fold, p < 0.001; 5.48-fold, p < 0.001)
pus of rats exposed to arsenic at a higher dose. Interestingly, a was observed on PD60 in rats following early life exposure to
trend of recovery in the expression of AP-1 was observed both arsenic from PD22 to PD59 as compared to controls (Table 3). A
in the frontal cortex and hippocampus 30 days after withdrawal trend of recovery in arsenic levels was observed 30 days after
of exposure in rats on PD90 in comparison to respective controls withdrawal of exposure in arsenic exposed rats in frontal cortex
(Fig. 9A). (F(2,9) = 12.77, 0.45, fold, p > 0.05; 1.00-fold, p < 0.01) and hip-
No significant change in the expression of phospho-JNK was pocampus (F(2,9) = 7.39, 0.27, fold, p > 0.05; 1.00-fold, p < 0.05) as
observed in rats exposed to arsenic (2.0 mg/kg or 4.0 mg/kg body compared to respective controls (Table 3).
Author's personal copy

70 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

Fig. 8. Early life exposure (PD22–PD59) to sodium arsenite and effect on the expression of selected pro-apoptotic (Caspase-3, Bax (A), Cleaved Caspase-3 (B)) and anti-
apoptotic (Bcl-2 (A)) proteins in frontal cortex and hippocampus of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.)
from PD22 to PD59. Effect on the expression of these proteins studied immediately after exposure to sodium arsenite on PD60 and 30 days after withdrawal of sodium
arsenite exposure on PD90. Values are mean ± SEM of three animals in each group. Data have been analyzed by one-way analysis of variance followed by Newman–Keuls
test. *Significantly differs from controls (p < 0.05); **significantly differs from controls (p < 0.01); ***significantly differs from controls (p < 0.001).

10. Discussion doses of arsenic (2 mg/kg or 4 mg/kg body weight/day) selected in


the present study are based on these reports.
Although a number of experimental studies have been carried Reduction in the body weight following arsenic exposure in
out to investigate the mechanism of arsenic induced neurotoxicity the progeny has been observed both in clinical and experimental
at different ages and doses with varying period of exposure, the studies (Holson et al., 1999; Schulz et al., 2002; Saha et al., 2008).
focus in the present study is to understand the mechanism of brain Inverse association between environmental arsenic exposure and
cholinergic alterations and associated functions during early life growth of children has been reported (Saha et al., 2012; Gardner
arsenic exposure at low doses and to assess if such changes are tran- et al., 2013). Interestingly, arsenic exposure in early childhood has
sient or persistent. Exposure to arsenic at doses 0.3–0.4 mg/kg/day a greater effect on body weight and growth than prenatal expo-
has been found to cause only morphological alterations in the sure (Saha et al., 2012). While the prevalence of decreased body
myelinated central tracts with no toxic effects. However, structural weight and growth was higher in boys, the effect was more marked
alterations in myelinated tracts in brain regions have been observed in girls (Saha et al., 2012). Adverse effect on body weight due to
following exposure to arsenic at doses 3–4 mg/kg body weight/day arsenic exposure in experimental studies was irrespective of time
(Rios et al., 2009; Zarazua et al., 2010; Martínez et al., 2011). The and dose in most of the studies (Stump et al., 1999; Ekong et al.,
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 71

Fig. 9. Early life exposure (PD22–PD59) to sodium arsenite and effect on the expression of selected stress marker proteins (Phospho-p38, AP-1 (A) and Phospho-JNK-1,2 (B))
in frontal cortex and hippocampus of rats. Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body weight/day, p.o.) from PD22 to PD59. Effect on the
expression of these proteins studied immediately after exposure to sodium arsenite on PD60 and 30 days after withdrawal of sodium arsenite exposure on PD90. Values are
mean ± SEM of three animals in each group. Data have been analyzed by one-way analysis of variance followed by Newman–Keuls test. *Significantly differs from controls
(p < 0.05); **significantly differs from controls (p < 0.01); ***significantly differs from controls (p < 0.001).

2012). Loss of body weight following early life exposure to arsenic at Bryant, 2010), there is a great need to understand the involve-
low dose as observed in the present study indicates growth impair- ment and the mechanism of cholinergic modifications. An inverse
ment and exhibit vulnerability of developing rats to the toxic effects relationship between arsenic exposure and plasma cholinesterase
of arsenic and is consistent with earlier experimental reports (Dhar activity has been observed in a population based study by Ali et al.
et al., 2005; Bashir et al., 2006). Recovery of body weight on with- (2010). It was suggested that alteration in plasma cholinesterase
drawal of arsenic exposure suggests that the effect is transient, and activity could be one of the potential biological mechanisms by
continuous arsenic exposure may have detrimental health effects. which arsenic induces neurotoxicity in humans. In brain, acetyl-
While arsenic has been found to affect different body organs, cholinesterase (AChE), an enzyme largely present in the cholinergic
the nervous system is a soft target to its deleterious effects. Increas- neurons plays an important role in the metabolism of acetylcholine,
ing incidences of neuropathy and neurological disorders associated an important neurotransmitter associated with synaptic plastic-
with acute and chronic arsenic exposure further indicate that ity (Roger et al., 2006). A number of environmental chemicals
arsenic may affect both the peripheral and the central nervous including arsenic have been found to inhibit the activity of brain
systems (Vahidnia et al., 2007; Banerjee et al., 2011). A num- acetylcholinesterase in rats (Sankhwar et al., 2012; Yadav et al.,
ber of studies have shown that arsenic exposure is closely linked 2011; Ansari et al., 2012). Arsenic exposure has also been found
with increased incidences of neuropsychological disturbances and to inhibit acetylcholinesterase activity in vitro in cultured hip-
cognitive deficits with greater vulnerability in children (Wright pocampal neurons of rats and neuroblastoma cells of mice (Repetto
et al., 2006; Rosado et al., 2007). As environmental arsenic expo- et al., 1994). Synthesis and generation of acetylcholine in brain
sure has been found to be a potential risk factor for Alzheimer’s slices has been found to be affected as a result of arsenic expo-
disease (Giasson et al., 2002; Borenstein et al., 2006; Gong and sure (Kobayashi et al., 1987). As most of these studies have been
Author's personal copy

72 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

Table 3 exposure. In an interesting study, Liu et al. (2012) found that arsenic
Early life exposure (PD22–PD59) to sodium arsenite and effect on arsenic levels in
induced effect on hippocampal cell proliferation and neurogenesis
frontal cortex and hippocampus of rats on PD60 and PD90.
could be recovered in adult mice.
Age in postnatal day Although there are evidences exhibiting that arsenic may cause
Brain region/Treatment groups PD60 PD90 oxidative damage in the brain by creating an imbalance between
pro-oxidant and anti-oxidant system (Yen et al., 2011), the impact
Frontal cortex
Control 357 ± 19.55 340 ± 39.87 of arsenic during early life exposure on the intensity of brain
ARS I (2 mg/kg) 853.5 ± 108.5* 495 ± 69.49 oxidative stress is not understood. Increased formation of malon-
ARS II (4 mg/kg) 1751 ± 212.6*** 706 ± 38.9** aldehyde and protein carbonyls in frontal cortex and hippocampus
Hippocampus of arsenic exposed rats indicate enhanced oxidative damage due
Control 336.5 ± 33.33 296 ± 42.41 to increased generation of reactive species and the finding is con-
ARS I (2 mg/kg) 1517 ± 159.49*** 377 ± 54.24 sistent with earlier reports (Bhadauria and Flora, 2007; Flora et al.,
ARS II (4 mg/kg) 2182 ± 116.6*** 591 ± 68.41*
2009; Wang et al., 2012). As arsenic has a high affinity with thiols, it
Rats were exposed to sodium arsenite (ARS I – 2.0 or ARS II – 4.0 mg/kg body may affect free thiols and catalytic activity of proteins and enzymes
weight/day, p.o.) from PD22 to PD59 and arsenic levels in frontal cortex and hip- containing thiols (Shen et al., 2012).
pocampus were analyzed by atomic absorption spectrophotometer immediately
after exposure on PD60 and 30 days after withdrawal of exposure on PD90. Values
Decrease in reduced glutathione levels both in hippocampus
are mean ± SEM of four animals in each group. Data have been analyzed by one-way and frontal cortex as observed in the present study could be due
analysis of variance followed by Newman–Keuls test. Arsenic levels expressed as to arsenic induced toxic manifestations and indicates enhanced
ng/gm tissue weight. stress. Arsenic exposure has also been found to impair the integrity
*
Significantly differs from controls (p < 0.05). of enzymes associated with the antioxidant defense (Kumar et al.,
**
Significantly differs from controls (p < 0.01).
***
Significantly differs from controls (p < 0.001).
2013; Sannadia et al., 2013). Decrease in the activity of super-
oxide dismutase following arsenic exposure in frontal cortex and
hippocampus indicates that arsenic may affect the dismutation of
carried out at high doses of arsenic and in adult animals, impact of superoxide radicals and thus enhances the vulnerability of both
low doses of arsenic on the integrity of brain acetylcholinesterase the brain regions to toxic effects of superoxide radicals. Further,
activity in developing animals is not understood. Decrease in decreased activity of catalase and glutathione peroxidase, enzymes
acetylcholinesterase activity both in frontal cortex and hippocam- associated to reduce hydrogen peroxide into water and oxygen
pus following arsenic exposure in rats during early life as observed in both the brain regions of arsenic exposed rats indicate that
in the present study is consistent with earlier reports on adult enzymatic antioxidant defense in the brain is impaired following
rats (Patlolla and Tchounwou, 2005; Rodriguez et al., 2005; Flora arsenic exposure. The fact that the brain has high utilization of
et al., 2009). Being the metabolic enzyme, inhibition in the activ- oxygen associated with impaired antioxidant defense, vulnerabil-
ity of acetylcholinesterase may be associated with pharmacological ity of the brain to arsenic induced oxidative stress is likely to be
consequences including enhanced accumulation of acetylcholine at enhanced as observed in the present study. Oxidative injury in the
the synaptic cleft and subsequently down regulation of cholinergic brain like other body organs following exposure to environmental
receptors both in frontal cortex and hippocampus. Desensitization chemicals including arsenic may cause pharmacological, physio-
of brain cholinergic receptors due to accumulation of acetylcholine logical and pathological changes including cellular apoptosis (De
has been demonstrated earlier (Zheng et al., 2000). Interestingly, Vizcaya-Ruiz et al., 2009; Khan et al., 2013; Sannadia et al., 2013).
decrease in the mRNA expression of ChRM2 receptor gene both Upregulation in the expression of Caspase-3, cleaved Caspase-3
in the frontal cortex and hippocampus following arsenic expo- and Bcl-2/Bax protein ratio both in frontal cortex and hippocampus
sure observed in the present study is consistent with decreased indicate enhanced apoptosis and could be associated with arsenic
binding of cholinergic receptors. Further, decrease in the expres- induced oxidative stress during early life. Increased expression of
sion of choline acetyltransferase (ChAT) both in the frontal cortex AP-1, a stress marker protein in arsenic exposed rats also appears
and hippocampus following arsenic exposure at low doses could to be associated with enhanced oxidative stress. While no signif-
be due to auto-feed back regulation as a result of accumulation of icant effect on the expression of phospho-JNK was observed in
acetylcholine and indicate vulnerability of cholinergic system. The the present study, increased expression of phospho-p38 both in
role of cholinergic system in modulating learning and memory is frontal cortex and hippocampus suggest that arsenic induced apo-
well established (Paul, 2003; Havekesa et al., 2011). Decrease in ptosis may be modulated by p38 MAPKs protein. The functional
learning and memory assessed by active avoidance response and role of MAPKs in modulating proliferation, differentiation, apopto-
continuous alternation test in arsenic exposed rats could be asso- sis and other important cellular processes through intracellular
ciated with impaired cholinergic system, specially decreased brain signaling is well accepted (Estus et al., 1994; Ham et al., 1995;
cholinergic-muscarinic receptors in the present study. Moreover, Mesner et al., 1995; Namgung and Zhengui, 2000). As there are
the role of PKC ␤-1 in modulating long term potentiation and dif- reports exhibiting a close link between increased MAPK signaling
ferent forms of learning is well accepted (Wu et al., 2007; Paratcha and neurodegenerative disorders associated with enhanced oxida-
et al., 2000). Decreased expression of hippocampal PKC ␤-1 there- tive stress and apoptosis (Miloso et al., 2008; Kim and Choi, 2010),
fore could also be associated with impaired learning in arsenic the increased oxidative stress associated with enhanced apoptosis
exposed rats as observed in the present study. While studying the following early life arsenic exposure in the present study is convinc-
neuroprotective efficacy of curcumin in arsenic induced choliner- ing and could be easily linked. It was noticeable that the intensity of
gic dysfunctions, decreased sensitivity of cholinergic-muscarinic oxidative stress and apoptosis was reduced and exhibited a trend
receptors associated with impaired learning was observed by us in of recovery both in frontal cortex and hippocampus on withdrawal
arsenic exposed rats. However, the dose of arsenic (sodium arsen- of arsenic exposure.
ite, 20 mg/kg body weight/day) was very high in these studies. While deciphering the mechanism of recovery in cholinergic
Cholinergic dysfunctions following early life exposure to arsenic modifications and learning, intensity of oxidative stress and apo-
at low doses as observed in the present study are striking. Another ptosis in arsenic exposed rats on withdrawal of exposure, the
interesting phenomenon observed in the present study was a trend presence of arsenic in frontal cortex and hippocampus appear to
of recovery in the cholinergic modifications, PKC ␤-1 expression be an important factor. Arsenic, like many other metals, crosses
and learning and memory in arsenic exposed rats on withdrawal of the blood brain barrier and accumulates in the brain through active
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 73

the Director, CSIR-Indian Institute of Toxicology Research (CSIR-


IITR), Lucknow for his keen interest in the present study. One
of the co-authors, Lalit P Chandravanshi is grateful to the Uni-
versity Grants Commission, New Delhi for providing the research
fellowship. The authors acknowledge the help of Mr. Ritul Kamal,
CSIR-IITR, Lucknow in analyzing the data and Mr. B.S. Pandey
for extending the technical support. The authors also thank Pro-
fessor Madhu Mehrotra, Former Head, Department of European
Languages, University of Lucknow, Lucknow and Mr. B.D. Bhat-
tacharji, CSIR-IITR, Lucknow for going through the manuscript and
making the necessary changes in the language.

References

Abhyankar, L.N., Jones, M.R., Guallar, E., Navas-Acien, A., 2012. Arsenic exposure and
hypertension: a systematic review. Environ. Heal. Perspect. 120, 494–500.
Aebi, H., 1984. Catalase in vitro. In: Packer, L. (Ed.), Methods in Enzymology, vol. 105.
Academic Press, New York, pp. 121–126.
Ahmed, S., Ahsan, K.B., Kippler, M., Mily, A., Wagatsuma, Y., Hoque, A.M., Ngom, P.T.,
El Arifeen, S., Raqib, R., Vahter, M., 2012. In utero arsenic exposure is associated
with impaired thymic function in new borns possibly via oxidative stress and
apoptosis. Toxicol. Sci. 129 (2), 305–314.
Ahmed, S., Mahabbat-e Khoda, S., Rekha, R.S., Gardner, R.M., Ameer, S.S., Moore,
S., Ekström, E.C., Vahter, M., Raqib, R., 2011. Arsenic-associated oxidative
stress,inflammation, and immune disruption in human placenta and cord blood.
Fig. 10. Schematic representation of sodium arsenite induced cholinergic deficits
Environ. Health Perspect. 119 (2), 258–264.
associated with oxidative stress and apoptosis in rat brain. Ali, N., Hoque, M.A., Haque, A., Salam, K.A., Karim, M.R., Rahman, A., Islam, K., Saud,
Z.A., Khalek, M.A., Akhand, A.A., Hossain, M., Mandal, A., Karim, M.R., Miyataka,
H., Himeno, S., Hossain, K., 2010. Association between arsenic exposure and
uptake (Xi et al., 2010b; Lu et al., 2012). It has been shown in vitro plasma cholinesterase activity: a population based study in Bangladesh. Environ.
that the transporter for arsenic uptake into astrocytes has high Km Health 9, 36.
value as there was a proportional increase in cellular content of Anderson, L.M., Diwan, B.A., Fear, N.T., Roman, E., 2000. Critical windows of exposure
for children’s health: cancer in human epidemiological studies and neoplasm in
arsenic in the system (Meyer et al., 2013; Calatayud et al., 2010; experimental animal models. Environ. Health Perspect. 108 (Suppl. 3), 573–594.
Thomas, 2007). Decreased arsenic levels both in the frontal cortex Ansari, R.W., Shukla, R.K., Yadav, R.S., Seth, K., Pant, A.B., Singh, D., Agrawal, A.K.,
and hippocampus on withdrawal of exposure is quite fascinating. Islam, F., Khanna, V.K., 2012. Cholinergic dysfunctions and enhanced oxidative
stress in the neurobehavioral toxicity of lambda-cyhalothrin in developing rats.
While it is difficult to explain the exact mechanism of low arsenic Neurotox. Res. 22 (4), 292–309.
levels both in the frontal cortex and the hippocampus on with- Argos, M., Kalra, T., Rathouz, P.J., Chen, Y., Pierce, B., Parvez, F., Islam, T., Ahmed, A.,
drawal of exposure as observed in the present study, decreased Rakibuz-Zaman, M., Hasan, R., 2010. Arsenic exposure from drinking water, and
all-cause and chronic-disease mortalities in Bangladesh (HEALS): a prospective
arsenic levels in the cerebrum of adult mice was observed on cohort study. Lancet 376 (9737), 252–258.
cessation of arsenic exposure by Liu et al. (2012). Arsenic forms Ballentine, R., Burford, D.D., 1957. Determination of metals. Methods Enzymol. 3,
conjugates with cellular GSH and possible role of these conjugates 1002–1035.
Banerjee, N., Nandy, S., Kearns, J.K., Bandyopadhyay, A.K., Das, J.K., Majumder, P.,
to eliminate/export arsenic from the cells has been suggested by a
Basu, S., Banerjee, S., Sau, T.J., States, J.C., 2011. Polymorphisms in the TNF-␣
number of investigators (Kala et al., 2000; Liu et al., 2001; Thomas, and IL10 gene promoters and risk of arsenic-induced skin lesions and other
2007; Meyer et al., 2013). nondermatological health effects. Toxicol. Sci. 121, 132–139.
Bardullas, U., Limón-Pacheco, J.H., Giordano, M., Carrizales, L., Mendoza-Trejo,
The results of the present study exhibit that early life exposure
M.S., Rodríguez, V.M., 2009. Chronic low-level arsenic exposure causes
to arsenic at low doses may cause cholinergic alterations both in gender-specific alterations in locomotor activity, dopaminergic systems, and
frontal cortex and hippocampus and impair learning and mem- thioredoxin expression in mice. Toxicol. Appl. Pharmacol. 239 (2), 169–177.
ory. Increased oxidative stress associated with enhanced apoptosis Bashir, S., Sharma, Y., Irshad, M., Nag, T.C., Tiwari, M., Kabra, M., et al., 2006. Arsenic
induced apoptosis in rat liver following repeated 60 days exposure. Toxicology
appear to be important contributors in arsenic induced cholin- 217 (1), 63–70.
ergic dysfunctions and exhibit vulnerability of developing rats Bhadauria, S., Flora, S.J.S., 2007. Response of arse-nic-induced oxidative stress,
to the toxic effects of arsenic. Based on these findings a hypo- DNA damage, and metal imbal-ance to combined administration of DMSA and
monoisoa-myl-DMSA during chronic arsenic poisoning in rats. Cell Biol. Toxicol.
thetical model of arsenic induced neurochemical alterations in 23, 91–104.
developing rats has been proposed (Fig. 10). A trend of recov- Borenstein, A.R., Copenhaver, C.I., Mortimer, J.A., 2006. Early-life risk factors for
ery in behavioral and neurochemical changes during early life Alzheimer disease. Alzheimer Dis. Assoc. Disord. 20 (1), 63–72.
Borges, M.O., Abreu, M.L., Porto, C.S., Avellar, M.C., 2001. Characterization of mus-
arsenic exposure clearly exhibits that these effects are transient carinic acetylcholine receptor in rat Sertoli cells. Endocrinology 142 (11),
but continuous arsenic exposure may have detrimental effects. 4701–4710.
Although it is difficult to comment at present, assay of blood acetyl- Calatayud, M., Gimeno, J., Vélez, D., Devesa, V., Montoro, R., 2010. Characterization
of the intestinal absorption of arsenate, monomethylarsonic acid, and dimethy-
cholinesterase activity could be further explored as a biomarker
larsinic acid using the Caco-2 cell line. Chem. Res. Toxicol. 23 (3), 547–556.
to assess the risk of arsenic induced neurotoxicity in the popu- Calderon, J., Navarro, M.E., Jimenez-Capdeville, M.E., Santos-Diaz, M.A., Golden, A.,
lation. The findings are interesting and will help the regulatory Rodriguez-Leyva, I., et al., 2001. Exposure to arsenic and lead and neuropsycho-
logical development in Mexican children. Environ. Res. 85 (2), 69–76.
agencies and scientists to plan the strategy for biomonitoring on
Calderon, R.L., Hudgens, E., Le, X.C., Schreinemachers, D., Thomas, D.J., 1999. Excre-
arsenic exposed population and specially children to re-evaluate tion of arsenic in urine as a function of exposure to arsenic in drinking water.
the Tolerable Daily Intake (TDI) of arsenic levels in food or drinking Environ. Health Perspect. 107, 663–667.
water. Chen, Y., Graziano, J.H., Parvez, F., Liu, M., Slavkovich, V., Kalra, T., Argos, M., Islam, T.,
Ahmed, A., Rakibuz-Zaman, M., 2011. Arsenic exposure from drinking water and
mortality from cardiovascular disease in Bangladesh: prospective cohort study.
Acknowledgments BMJ 342, d2431.
Dauphine, D.C., Ferreccio, C., Guntur, S., Yuan, Y., Hammond, S.K., Balmes, J., Smith,
A.H., Steinmaus, C., 2011. Lung function in adults following in utero and child-
The study is a part of INDEPTH programme of CSIR, New Delhi. hood ex-502 posure to arsenic in drinking water: preliminary findings. Int. Arch.
CSIR–IITR, Lucknow Communication no. 3076. The authors thank Occup. Environ. Health 84, 591–600.
Author's personal copy

74 L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75

De Vizcaya-Ruiz, A., Barbier, O., Ruiz-Ramos, R., Cebrian, M.E., 2009. Biomarkers of Kapaj, S., Peterson, H., Liber, K., Bhattacharya, P., 2006. Human health effects
oxidative stress and damage in human populations exposed to arsenic. Mutat. from chronic arsenic poisoning – a review. J. Environ. Sci. Health A 41,
Res. 674, 85–92. 2399–2428.
Dhar, P., Jaitley, M., Kalaivani, M., Mehra, R.D., 2005. Preliminary morphological and Khan, S., Telang, A.G., Malika, J.K., 2013. Arsenic-induced oxidative stress, apoptosis
histochemical changes in rat spinal cord neurons following arsenic ingestion. and alterations in testicular steroidogenesis and spermatogenesis in wistar rats:
Neurotoxicology 26, 309–320. ameliorative effect of curcumin. Wudpecker J. Pham. Pharmacol. 2 (3), 033–048.
Du, J., Zhou, N., Liu, H., Jiang, F., Wang, Y., Hu, C., Qi, H., Zhong, C., Wang, X., Li, Kim, E.K., Choi, E.J., 2010. Pathological roles of MAPK signaling pathways in human
Z., 2012. Arsenic induces functional re-expression of estrogen receptor ␣ by diseases. Biochim. Biophys. Acta 1802 (4), 396–405.
demethylation of DNA in estrogen receptor-negative human breast cancer. PLoS Kim, S., Takeuchi, A., Kawasumi, Y., Endo, Y., Lee, H., Kim, Y., 2013. A Guillain-Barré
One 7 (4), e35957. syndrome-like neuropathy associated with arsenic exposure. J. Occup. Health
Ekong, M.B., Ekanem, T.B., Sunday, A.O., Aquaisua, A.N., Akpanabiatu, M.I., 2012. Eval- 54 (4), 344–347.
uation of calabash chalk effect on femur bone morphometry and mineralization Kobayashi, H., Yuyama, A., Ishihara, M., Matsusaka, N., 1987. Effects of arsenic
in young wistar rats: a pilot study. Int. J. Appl. Basic Med. Res. 2 (2), 107–110. on cholinergic parameters in brain in vitro. Neuropharmacology 26 (12),
Ellman, G.L., Courtney Jr., K.D., Andresand, V., Featherstone, R.M., 1961. A new 1707–1713.
and rapid calorimetric determination of acetylcholinesterase activity. Biochem. Kris, F., 2009. Nutrient protection against arsenic toxicity: folate, cysteine support
Pharmacol. 7, 88–95. methylation in children. Environ. Health Perspect. 117 (5), A211.
Estus, S., Zaks, W.J., Freeman, R.S., Gruda, M., Bravo, R., Johnson, E.M., 1994. Altered Krinke, G., 2000. In: Bullock, G., Bunton, T.E. (Eds.), The Laboratory Rat: A Volume in
gene expression in neurons during programmed cell death: identification of Handbook of Experimental Animals, Chapter 12 Developmental Neurotoxicity.
c-jun as necessary for neuronal apoptosis. J. Cell Biol. 127, 1717–1727. Academic Press, San Diego, p. 75.
Flohe, L., Gunzler, W.A., 1984. Assays of glutathione peroxidase. Method Enzymol. Kumar, M.R., Flora, S.J.S., Reddy, G.R., 2013. Monoisoamyl 2,3-dimercaptosuccinic
105, 114–121. acid attenuates arsenic induced toxicity: behavioral and neurochemical
Flora, S.J., Bhatt, K., Mehta, A., 2009. Arsenic moiety in gallium arsenide is respon- approach. Environ. Toxicol. Pharmacol. 36 (1), 231–242.
sible for neuronal apoptosis and behavioral alterations in rats. Toxicol. Appl. Levine, R.L., Garland, D., Oliver, C.N., Amici, A., Climent, I., Lenz, A.G., Ahn, B., Shaltiel,
Pharmacol. 240 (2), 236–244. S., Stadtman, E.R., 1990. Determination of carbonyl content in oxidatively mod-
Gardner, R.M., Kippler, M., Tofail, F., Bottai, M., Hamadani, J., Grandér, M., Nermell, ified proteins. Methods Enzymol. 186, 464–478.
B., Palm, B., Rasmussen, K.M., Vahter, M., 2013. Environmental exposure to met- Lindberg, A.L., Sohel, N., Rahman, M., Persson, L.A., Vahter, M., 2010. Impact of
als and children’s growth to age 5 years: a prospective cohort study. Am. J. smoking and chewing tobacco on arsenic-induced skin lesions. Environ. Health
Epidemiol. 177 (Jun(12)), 1356–1367. Perspect. 118, 533.
George, C.M., Gamble, M., Slavkovich, V., Levy, D., Ahmed, A., Ahsan, H., Graziano, Ling, I.H., Gwo-Shing, C., Chih-Hung, L., Tse-Yen, Y., Yu-Hsin, C., Yuan-Hung, W., Yu-
J., 2013. A cross sectional study of the Impact of blood selenium on blood and Mei, H., Hung-Yi, C., Meei-Maan, W., Chien-Jen, C., 2013. Use of Arsenic-induced
urinary arsenic concentrations in Bangladesh. Environ. Health 12, 52. palmoplantar hyperkeratosis and skin cancers to predict risk of subsequent.
Giasson, B.I., Ischiropoulos, H., Lee, V.M., Trojanowski, J.Q., 2002. The relationship Intern. Malig. Am. J. Epidemiol. 177 (3), 202–212.
between oxidative/nitrative stress and pathological inclusions in Alzheimer’s Liu, J., Chen, H., Miller, D.S., Saavedra, J.E., Keefer, L.K., Johnson, D.R., Klaassen, C.D.,
and Parkinson’s diseases. Free Radic. Biol. Med. 32 (12), 1264–1275. Waalkes, M.P., 2001. Overexpression of glutathione S-transferase II and mul-
Glowinski, J., Iversen, L.L., 1966. Regional studies of catecholamines in the rat brain. I. tidrug resistance transport proteins is associated with acquired tolerance to
The disposition of [3H]norepinephrine, [3H]dopamine and [3H]dopa in various inorganic arsenic. Mol. Pharmacol. 60 (2), 302–309.
regions of the brain. J. Neurochem. 13 (8), 655–659. Liu, S., Piao, F., Sun, X., Bai, L., Peng, Y., Zhong, Y., Ma, N., Sun, W., 2012.
Golub, M.S., 1994. Maternal toxicity and the identification of inorganic arsenic as a Arsenic-induced inhibition of hippocampal neurogenesis and its reversibility.
developmental toxicant. Reprod. Toxicol. 8, 283–295. Neurotoxicology 33 (5), 1033–1039.
Gong, G.O’., Bryant, S.E., 2010. The arsenic exposure hypothesis for Alzheimer’s Lowry, O.H., Rosenbrough, N.J., Farr, A.L., Randall, R.J., 1951. Protein measurement
disease. Alzheimer Dis. Assoc. Disord. 24, 311–316. with the folin phenol reagent. J. Biol. Chem. 192, 265–275.
Ham, J., Babij, C., Whitfield, J., Pfarr, C.M., Lallemand, D., Yaniv, M., Rubin, L.L., 1995. Lu, C.W., Zhao, F.H., Sun, D., Zhong, Y., Yu, X.Y., Li, G.X., Lv, X.Q., Sun, G.F., Jin, Y.P.,
A c-jun dominant negative mutant protects sympathetic neurons against pro- 2012. Comparison of speciated arsenic levels in the liver and brain of mice-
grammed cell death. Neuron 14, 927–939. between arsenate and arsenite exposure at the early life. Environ. Toxicol.,
Hamadani, J.D., Grantham-McGregor, S.M., Tofail, F., Nermell, B., Fangstrom, B., Huda, http://dx.doi.org/10.1002/tox.21808 [Epub ahead of print].
S.N., Yesmin, S., Rahman, M., Vera-Hernandez, M., Arifeen, S.E., Vahter, M., 2010. Luo, J.H., Qiu, Z.Q., Shu, W.Q., Zhang, Y.Y., Zhang, L., Chen, J.A., 2009. Effects of arsenic
Pre- and postnatal arsenic exposure and child development at 18 months of age: exposure from drinking water on spatial memory, ultra-structures and NMDAR
a cohort study in rural Bangladesh. Int. J. Epidemiol. 39, 1206–1216. gene expression of hippocampus in rats. Toxicol. Lett. 184 (2), 121–125.
Hamadani, J.D., Tofail, F., Nermell, B., Gardner, R., Shiraji, S., Bottai, M., Arifeen, S.E., Luo, J., Qiu, Z., Chen, J., Zhang, L., Liu, W., Tan, Y., Shu, W., 2013. Maternal and early
Huda, S.N., Vahter, M., 2011. Critical windows of exposure for arsenic-associated life arsenite exposure impairs neurodevelopmentand increases the expression
impairment of cognitive function in preschool girls and boys: a population- of PSA-NCAM in hippocampus of rat offspring. TOX 51243, 1–8.
based cohort study. Int. J. Epidemiol. 40, 1593–1604. Martínez, L., Jiménez, V., García-Sepúlveda, C., Ceballos, F., Delgado, J.M., Niño-
Hasan, M., Haider, S.S., 1989. Acetyl homocysteine thiolactone protect against some Moreno, P., Doniz, L., Saavedra-Alanís, V., Castillo, C.G., Santoyo, M.E.,
neurotoxic effects of thallium. Neurotoxicology 10, 257–262. González-Amaro, R., Jiménez-Capdeville, M.E., 2011. Impact of early develop-
Havekesa, R., Abela, T., Eddy, A., Van der, Z., 2011. The cholinergic system and neos- mental arsenic exposure on promotor CpG-island methylation of genes involved
triatal memory functions. Behav. Brain Res. 221, 412–423. in neuronal plasticity. Neurochem. Int. 58 (5), 574–581.
Heck, J.E., Andrew, A.S., Onega, T., Rigas, J.R., Jackson, B.P., Karagas, M.R., Duell, E.J., Maurice, T., Su, T.-P., Parish, W., Nabeshima, T., Privat, A., 1994. A ␣ selective PCP
2009. Lung cancer in a U. S. population with low to moderate arsenic exposure. derivative, attenuates MK-801- induced impairment of learning in mice. Phar-
Environ. Health Perspect. 117, 1718–1723. macol. Biochem. Behav. 49, 859–869.
Herrera, A., Pineda, J., Antonio, M.T., 2013. Toxic effects of perinatal arsenic exposure Mesner, P.W., Epting, C.L., Hegarty, J.L., Green, S.H., 1995. A time table of events dur-
on the brain of developing rats and the beneficial role of natural antioxidants. ing programmed cell death induced by trophic factor withdrawal from neuronal
Environ. Toxicol. Pharmacol. 36 (1), 73–79. PC12 cells. J. Neurosci. 15, 7357–7366.
Holson, J.F., Stump, D.G., Ulrich, C.E., 1999. Absence of prenatal developmental tox- Meyer, N., Koehler, Y., Tulpule, K., Dringen, R., 2013. Arsenate accumulation and
icity from inhaled arsenic trioxide in rats. Toxicol. Sci. 51, 87–97. arsenate-induced glutathione export in astrocyte-rich primary cultures. Neu-
Hughes, M.F., Beck, B.D., Chen, Y., Lewis, A.S., Thomas, D.J., 2011. Arsenic exposure rochem. Int. 62 (7), 1012–1019.
and toxicology: a historical perspective. Toxicol. Sci. 123 (2), 305–332. Miloso, M., Scuteri, A., Foudah, D., Tredici, G., 2008. MAPKs as mediators of cell fate
Huyck, K.L., Kile, M.L., Mahiuddin, G., Quamruzzaman, Q., Rahman, M., Breton, determination: an approach to neurodegenerative diseases. Curr. Med. Chem.
C.V., Dobson, C.B., Frelich, J., Hoffman, E., Yousuf, J., Afroz, S., Islam, S., Chris- 15, 538–548.
tiani, D.C., 2007. Maternal arsenic exposure associated with low birth weight in Miteva, E., Hristova, D., Nenova, V., Manava, S., 2005. Arsenic as a factor affecting
Bangladesh. J. Occup. Environ. Med. 49, 1097–1104. virus infection in tomato plants: changes in plant growth, peroxidase activity
Itoh, T., Zhang, Y.F., Murai, S., Saito, H., Nagahama, H., Miyate, H., et al., 1990. The and chloroplast pigments. Sci. Hortic-Amsterdam 105, 343–358.
effect of arsenic trioxide on brain monoamine metabolism and locomotor activ- Nagaraja, T.N., Desiraju, T., 1994. Effects on operant learning and brain acetylcholine
ity of mice. Toxicol. Lett. 54, 345–353. esterase activity in rats following chronic inorganic arsenic intake. Hum. Exp.
Jamal, M., Ameno, K., Ameno, S., Morishita, J., Wang, W., Kumihashi, M., 2007. Toxicol. 13, 353–356.
Changes in cholinergic function in the frontal cortex and hippocampus of Namgung, U., Xia, Z., 2001. Arsenic induces apoptosis in rat cerebellar neurons via
rat exposed to ethanol and acetaldehyde. Neuroscience 144 (January(1)), activation of JNK3 and p38 MAP kinases. Toxicol. Appl. Pharmacol. 174, 130–138.
232–238. Namgung, U.K., Zhengui, Xia., 2000. Arsenite-induced apoptosis in cortical neurons
Jovanović, D.D., Paunović, K., Manojlović, D.D., Jakovljević, B., Rasic-Milutinović, Z., is mediated by c Jun N-terminal protein kinase 3 and p38 mitogen-activated
Dojcinović, B.P., 2012. Arsenic in drinking water and acute coronary syndrome protein kinase. J. Neurosci. 20 (17), 6442–6451.
in Zrenjanin municipality, Serbia. Environ. Res. 117, 75–82. Navas-Acien, A., Silbergeld, E.K., Streeter, R.A., Clark, J.M., Burke, T.A., Guallar, E.,
Kakkar, P., Das, B., Viswanathan, P.N., 1984. A modified spectrophotometric assay of 2006. Arsenic exposure and type 2 diabetes: a systematic review of the experi-
superoxide dismutase. Indian J. Biochem. Biophys. 21, 130–132. mental and epidemiological evidence. Environ. Health Perspect. 114, 641–648.
Kala, S.V., Neely, M.W., Kala, G., Prater, C.I., Atwood, D.W., Rice, J.S., Lieberman, Nemec, M.D., Holson, J.F., Farr, C.H., 1998. Developmental toxicity assessment of
M.W., 2000. The MRP2/cMOAT transporter and arsenic-glutathione complex arsenic acid in mice and rabbits. Reprod. Toxicol. 12 (6), 647–658.
formation are required for biliary excretion of arsenic. J. Biol. Chem. 275 (43), Ohkawa, H., Ohishi, N., Yagi, K., 1979. Assay for lipid peroxides in animal tissues by
33404–33408. thiobarbituric acid reaction. Anal. Biochem. 95, 351–358.
Author's personal copy

L.P. Chandravanshi et al. / Int. J. Devl Neuroscience 34 (2014) 60–75 75

Parajuli, R.P., Fujiwara, T., Umezaki, M., Watanabe, C., 2013. Association of cord Steinmaus, C., Yuan, Y., Kalman, D., Rey, O.A., Skibola, C.F., Dauphine, D., Basu, A.,
blood levels of lead, arsenic, and zinc with neurodevelopmental indicators in Porter, K.E., Hubbard, A., Bates, M.N., Smith, M.T., Smith, A.H., 2010. Individual
newborns: a birth cohort study in Chitwan Valley, Nepal. Environ. Res. 121, differences in arsenic metabolism and lung cancer in a case-control study in
45–51. Cordoba, Argentina. Toxicol. Appl. Pharmacol. 247 (2), 138–145.
Paratcha, G., Furman, M., Bevilaqua, L., Cammarota, M., Vianna, M., De Stein, M.L., Stump, D.G., Holson, J.F., Fleeman, T.L., Nemec, M.D., Farr, C.H., 1999. Com-
Izquierdo, I., Medina, J.H., 2000. Involvement of hippocampal PKCbeta1 isoform parative effects of single intraperitoneal or oral doses of sodium arsenate
in the early phase of memory formation of an inhibitory avoidance learning. or arsenic trioxide during in utero development. Teratology 60 (5),
Brain Res. 855, 199–205. 283–291.
Parvez, F., Wasserman, G.A., Factor-Litvak, P., Liu, X., Slavkovich, V., Siddique, A.B., Tabocova, S., Hunter 3rd, E.S., Gladen, B.C., 1996. Developmental toxicity of inorganic
Sultana, R., Sultana, R., Islam, T., Levy, D., Mey, J.L., Van Geen, A., Khan, K., Kline, arsenic in whole embryo culture: oxidation state, dose, time, and gestational age
J., Ahsan, H., Graziano, J.H., 2011. Arsenic exposure and motor function among dependence. Toxicol. Appl. Pharmacol. 138 (2), 298–307.
children in Bangladesh. Environ. Health Perspect. 119 (11), 1665–1670. Terry Jr., A.V., Stone, J.D., Buccafusco, J.J., Sickles, D.W., Sood, A., Prendergast, M.A.,
Patlolla, A.K., Tchounwou, P.B., 2005. Serum acetyl cholinesterase as a biomarker of 2003. Repeated exposure to subthreshhold doses of chlorpyrifos in rats: hip-
arsenic induced neurotoxicity in Sprague–Dawley rats. Int. J. Environ. Res. Public pocampal damage, impaired axonal transport and deficits in spatial learning. J.
Health 2, 80–83. Pharmacol. Exp. Ther. 305 (1), 375–384.
Paul, E., 2003. Gold Acetylcholine modulation of neural systems involved in learning Thomas, D.J., 2007. Molecular processes in cellular arsenic metabolism. Toxicol. Appl.
and memory. Neurobiol. Learn. Mem. 80 (3), 194–210. Pharmacol. 222 (3), 365–373.
Rahman, A., Persson, L.A., Nermell, B., El Arifeen, S., Ekstrom, E.C., Smith, A.H., Vahter, Tsai, S.Y., Chou, H.Y., The, H.W., Chen, C.M., Chen, C.J., 2003. The effects of chronic
M., 2010. Arsenic exposure and risk of spontaneous abortion, stillbirth, and arsenic exposure from drinking water on the neurobehavioral development in
infant mortality. Epidemiology 21, 797–804. adolescence. Neurotoxicology 24, 747–753.
Rahman, A., Vahter, M., Ekstrom, E.C., Rahman, M., Golam Mustafa, A.H., Wahed, M.A., Tseng, C.H., 2009. A review on environmental factors regulating arsenic methylation
Yunus, M., Persson, L.A., 2007. Association of arsenic exposure during pregnancy in humans. Toxicol. Appl. Pharmacol. 235 (3), 338–350.
with fetal loss and infant death: a cohort study in Bangladesh. Am. J. Epidemiol. US EPA, 2001. Federal Register, 66., pp. 6975–7066.
165, 1389–1396. Vahidnia, A., van der Straaten, R.J., Romijn, F., van Pelt, J., van der Voet, G.B., de Wolff,
Repetto, G., Sanz, P., Repetto, M., 1994. Comparative in vitro effects of sodium arsen- F.A., 2007. Arsenic metabolites affect expression of the neurofilament and tau
ite and sodium arsenate on neuroblastoma cells. Toxicology 92 (1–3), 143–153. genes: an in vitro study into the mechanism of arsenic neurotoxicity. Toxicol. In
Rios, R., Zarazua, S., Santoyo, M.E., Sepulveda-Saavedra, J., Romero-Diaz, V., Jimenez, Vitro 21 (6), 1104–1112.
V., Perez-Severiano, F., Vidal-Cantu, G., Delgado, J.M., Jimenez-Capdeville, M.E., Vahter, M.E., Li, L., Nermell, B., Rahman, A., El Arifeen, S., Rahman, M., Persson, L.A.,
2009. Decreased nitric oxide markers and morphological changes in the brain Ekström, E.C., 2006. Arsenic exposure in pregnancy: a population-based study
of arsenic-exposed rats. Toxicology 261 (1–2), 68–75. in Matlab, Bangladesh. J. Health Popul. Nutr. 24 (2), 236–245.
Rodriguez, V.M., Carrizales, L., Jimenez-Capdeville, M.E., Dufour, L., Giordano, M., Von Ehrenstein, O.S., Guha Mazumder, D.N., Hira-Smith, M., Ghosh, N., Yuan, Y.,
2001. The effect of sodium arsenite exposure on behavioral parameters in the Windham, G., Ghosh, A., Haque, R., Lahiri, S., Kalman, D., Das, S., Smith, A.H.,
rat. Brain Res. Bull. 55 (2), 301–308. 2006. Pregnancy outcomes, infant mortality, and arsenic in drinking water in
Rodriguez, V.M., Del Razo, L.M., Limon-Pacheco, J.H., Giordano, M., Sanchez-Pena, 684 West Bengal, India. Am. J. Epidemiol. 163 (7), 662–669.
L.C., Uribe-Querol, E., Gutierrez-Ospina, G., Gonsebatt, M.E., 2005. Glutathione Wang, S.X., Wang, Z.H., Cheng, X.T., Li, J., Sang, Z.P., Zhang, X.D., Han, L.L., Qiao, X.Y.,
reductase inhibition and methylated arsenic distribution in Cd1 mice brain and Wu, Z.M., Wang, Z.Q., 2007. Arsenic and fluoride exposure in drinking water:
liver. Toxicol. Sci. 84 (1), 157–166. children’s IQ and growth in Shanyin county, Shanxi province, China. Environ.
Rodriguez, V.M., Jimenez-Capdeville, M.E., Giordano, M., 2003. The effect of arsenic Health Perspect. 115, 643–647.
exposure on the nervous system. Toxicol. Lett. 145, 1–18. Wang, J., Zhang, W., Sun, D., Song, L., Li, Y., Xu, C., 2012. Analysis of neuroglobin
Rodriguez, V.M., Limon-Pacheco, J.H., Carrizales, L., Mendoza-Trejo, M.S., Giordano, mRNA expression in rat brain due to arsenite-induced oxidative stress. Environ.
M., 2010. Chronic exposure to low levels of inorganic arsenic causes alterations Toxicol. 27 (Sep(9)), 503–509.
in locomotoractivity and in the expression of dopaminergic and antioxidant World Health Organization, 2001. United Nations Synthesis Report on Arsenic in
systems in the albino rat. Neurotoxicol. Teratol. 32 (6), 640–647. Drinking Water, Geneva.
Roger, M.L., Steven, G.P., Albert, E., 2006. Targeting acetylcholinesterase and butyryl- Wright, R.O., Amarasiriwardena, C., Woolf, A.D., Jim, R., Bellinger, D.C., 2006. Neu-
cholinesterase in dementia. Int. J. Neuropsychopharmacol. 9, 101–124. ropsychological correlates of hair arsenic, manganese, and cadmium levels in
Roghani, M., Joghataie, M.T., Jalali, M.R., Baluchnejadmojarad, T., 2006. Time course school-age children residing near a hazardous waste site. Neurotoxicology 27
of changes in passive avoidance and Y – maze performance in male diabetic rats. (2), 210–216.
Iran. Biomed. J. 10 (2), 99–104. Wu, J., Song, T.B., Li, Y.J., He, K.S., Ge, L., Wang, L.R., 2007. Prenatal restraint
Rosado, J.L., Ronquillo, D., Kordas, K., Rojas, O., Alatorre, J., Lopez, P., Garcia-Vargas, stress impairs learning and memory and hippocampal PKCbeta1 expression and
G., Del Carmen Caamaño, M., Cebrián, M.E., Stoltzfus, R.J., 2007. Arsenic exposure translocation in offspring rats. Brain Res. 1141, 205–213.
and cognitive performance in Mexican schoolchildren. Environ. Health Perspect. Xi, S., Guo, L., Qi, R., Sun, W., Jin, Y., Sun, G., 2010a. Prenatal and early life arsenic
115 (9), 1371–1375. exposure induced oxidative damage and altered activities and mRNA expres-
Roy, A., Kordas, K., Lopez, P., Rosado, J.L., Cebrian, M.E., Vargas, G.G., Ronquillo, D., sions of neurotransmitter metabolic enzymes in offspring rat brain. J. Biochem.
Stoltzfus, R.J., 2011. Association between arsenic exposure and behavior among Mol. Toxicol. 6, 368–378.
636 first-graders from Torreon, Mexico. Environ. Res. 111, 670–676. Xi, S., Jin, Y., Lv, X., Sun, G., 2010b. Distribution and speciation of arsenic by transpla-
Saha, K.K., Engström, A., Hamadani, J.D., Tofail, F., Rasmussen, K.M., Vahter, M., 2012. cental and early life exposure to inorganic arsenic in offspring rats. Biol. Trace
Pre- and postnatal arsenic exposure and body size to 2 years of age: a cohort Elem. Res. 134 (1), 84–97.
study in rural Bangladesh. Environ. Health Perspect. 120 (8), 1208–1214. Xi, S., Sun, W., Wang, F., Jin, Y., Sun, G., 2009. Transplacental and early life exposure
Saha, K.K., Frongillo, E.A., Alam, D.S., Arifeen, S.E., Persson, L.A., Rasmussen, K.M., to inorganic arsenic aVected development and behavior in of spring rats. Arch.
2008. Appropriate infant feeding practices result in better growth of infants and Toxicol. 83, 549–556.
young children in rural Bangladesh. Am. J. Clin. Nutr. 87 (6), 1852–1859. Yadav, R.S., Chandravanshi, L.P., Shukla, R.K., Sankhwar, M.L., Ansari, R.W., Shukla,
Sankhwar, M.L., Yadav, R.S., Shukla, R.K., Pant, A.B., Singh, D., Parmar, D., Khanna, V.K., P.K., Pant, A.B., Khanna, V.K., 2011. Neuroprotective efficacy of curcumin
2012. Impaired cholinergic mechanisms following exposure to monocrotophos in arsenic induced cholinergic dysfunctions in rats. Neurotoxicology 32 (6),
in young rats. Hum. Exp. Toxicol. 31 (Jun(6)), 606–616. 760–768.
Sannadia, S., Kadeyalaa, P.K., Gottipolu, R.R., 2013. Reversal effect of monoisoamyl Yadav, R.S., Sankhwar, M.L., Shukla, R.K., Chandra, R., Pant, A.B., Islam, F., et al.,
dimercaptosuccinic acid (MiADMSA) for arsenic and lead induced perturba- 2009. Attenuation of arsenic neurotoxicity by curcumin in rats. Toxicol. Appl.
tions in apoptosis and antioxidant enzymes in developing rat brain. Int. J. Dev. Pharmacol. 240 (3), 367–376.
Neurosci. 31, 586–597, Issue. Yen, C.C., Ho, T.J., Wu, C.C., Chang, C.F., Su, C.C., Chen, Y.W., Jinn, T.R., Lu, T.H., Cheng,
Schulz, H., Nagymajtenyi, L., Institoris, L., 2002. A study on behavioral, neurotoxi- P.W., Su, Y.C., Shing, H.L., Chun, F.H., 2011. Inorganic arsenic causes cell apoptosis
cological, and immunotoxicological effects of subchronic arsenic treatment in in mouse cerebrum through an oxidative stress regulated signaling pathway.
rats. J. Toxicol. Environ. Health A 65 (16), 1181–1193. Arch. Toxicol. 85, 565–575.
Shen, S.-C., Lee, W.-R., Yang, L.-Y., Tsai, H.-H., Yang, L.-L., Chen, Y.-C., 2012. Quercetin Yorifuji, T., Tsuda, T., Doi, H., Grandjean, P., 2011. Cancer excess after arsenic expo-
enhancement of arsenic-induced apoptosis via stimulating ROS dependent p53 sure 732 from contaminated milk powder. Environ. Health Prev. Med. 16 (3),
protein ubiquitination in human HaCaT keratinocytes. Exp. Dermatol. 21 (5), 164–170.
370–375. Yuan, Y., Marshall, G., Ferreccio, C., Steinmaus, C., Liaw, J., Bates, M., Smith, A.H., 2010.
Smith, A.H., Ercumen, A., Yuan, Y., Steinmaus, C.M., 2009. Increased lung cancer risks Kidney cancer mortality: fifty-year latency patterns related to arsenic exposure.
are similar whether arsenic is ingested or inhaled. J. Exp. Sci. Environ. Epidemiol. Epidemiology 21 (1), 103–108.
19 (4), 343–348. Zarazua, S., Rios, R., Delgado, J.M., Santoyo, M.E., Ortiz-Perez, D., Jimenez-Capdeville,
Smith, A.H., Marshall, G., Liaw, J., Yuan, Y., Ferreccio, C., Steinmaus, C., 2012. Mortality M.E., 2010. Decreased arginine methylation and myelin alterations in arsenic
648 in young adults following in utero and childhood exposure to arsenic in exposed rats. Neurotoxicology 31 (1), 94–100.
drinking 649 water. Environ. Health Perspect. 120 (11), 1527–1531. Zheng, Q., Olivier, K., Wan, Y.K., Pope, C.N., 2000. Comparative cholinergic neurotox-
Smith, A.H., Marshall, G., Yuan, Y., Liaw, J., Ferreccio, C., Steinmaus, C., 2011. Evi- icity of oral chlorpygrifos exposures in preweanling and adult rats. Toxicol. Sci.
dence from Chile that arsenic in drinking water may increase mortality from 55 (1), 124–132.
pulmonary tuberculosis. Am. J. Epidemiol. 173 (4), 414–420.

You might also like