You are on page 1of 14

Biological Trace Element Research

https://doi.org/10.1007/s12011-018-1286-1

Developmental Neurotoxicity of Arsenic: Involvement of Oxidative


Stress and Mitochondrial Functions
Lalit P. Chandravanshi 1,2 & Richa Gupta 2 & Rajendra K. Shukla 3

Received: 2 January 2018 / Accepted: 23 February 2018


# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Over the last decade, there has been an increased concern about the health risks from exposure to arsenic at low doses, because of
their neurotoxic effects on the developing brain. The exact mechanism underlying arsenic-induced neurotoxicity during sensitive
periods of brain development remains unclear, although enhanced oxidative stresses, leading to mitochondrial dysfunctions
might be involved. Here, we highlight the generation of reactive oxygen species (ROS) and oxidative stress which leads to
mitochondrial dysfunctions and apoptosis in arsenic-induced developmental neurotoxicity. Here, the administration of sodium
arsenite at doses of 2 or 4 mg/kg body weight in female rats from gestational to lactational (GD6-PD21) resulted to increased
ROS, led to oxidative stress, and increased the apoptosis in the frontal cortex, hippocampus, and corpus striatum of developing
rats on PD22, compared to controls. Enhanced levels of ROS were associated with decreased mitochondrial membrane potential
and the activity of mitochondrial complexes, and hampered antioxidant levels. Further, neuronal apoptosis, as measured by
changes in the expression of pro-apoptotic (Bax, Caspase-3), anti-apoptotic (Bcl2), and stress marker proteins (p-p38, pJNK) in
arsenic-exposed rats, was discussed. The severities of changes were found to more persist in the corpus striatum than in other
brain regions of arsenic-exposed rats even after the withdrawal of exposure on PD45 as compared to controls. Therefore, our
results indicate that perinatal arsenic exposure leads to abrupt changes in ROS, oxidative stress, and mitochondrial functions and
that apoptotic factor in different brain regions of rats might contribute to this arsenic-induced developmental neurotoxicity.

Keywords Arsenic . Developmental neurotoxicity . Oxidative stress . Mitochondria . Apoptosis

Introduction various diseases, including cancerous and noncancerous dis-


eases, such as cardiovascular, hypertension, and cognitive im-
Arsenic, a naturally occurring neurotoxicant, is one of the risk pairment in human [5, 41, 65]. However, little is known about
factors for cognitive and neurological dysfunctions in humans the developmental neurotoxicity of low levels of arsenic dur-
[37, 50]. Even though chronic exposure to arsenic at high ing crucial periods of brain development. In utero or early life
levels [3] has been very well recognized as causing different developments of the child represent a critical window of sus-
adverse health outcomes, including neurological diseases, ex- ceptibility to the toxicant. Arsenic insult at any step during
posure to low and moderate levels [36] of arsenic has only development may lead to developing chronic diseases [34,
been more recently correlated with an increased risk for 63]. Arsenic may be a serious threat to the development of
the fetus and infant health because of their combination of
physiological immaturity and exposure during pregnancy
* Lalit P. Chandravanshi and early life. At times, the changes may not be visible imme-
chandravanshi04@gmail.com diately but reflected at later stages of life. The adverse effects
during the developmental periods are likely to have long-
1 lasting effects and may not recover.
Department of Zoology, Institute of Science, Banaras Hindu
University, Varanasi 221005, India Arsenic may induce oxidative stress, which causes many
2 neurological dysfunctions in children, particularly at low
Developmental Toxicology Division, CSIR—Indian Institute of
Toxicology Research, Post Box No. 80, MG Marg, Lucknow 226 levels. Gong and O’Bryant [20], suggested a possible relation-
001, India ship between arsenic exposure and Alzheimer’s disease.
3
Department of Biochemistry, All India Institute of Medical Sciences, However, the neurotoxic effects and underlying mechanisms
Bhopal, India of arsenic-induced developmental neurotoxicity are mostly
Chandravanshi et al.

unknown. Emerging studies have proved that accumulation of during the perinatal period. Further, we also tried to investi-
arsenic and free radicals in the central nervous system will gate the persistent effect of arsenic 23 days after the withdraw-
induce oxidative stress and that antioxidant defenses are relat- al of arsenic exposure.
ed to the pathogenesis of neurodegenerative diseases [59].
Oxidative stress induced by arsenic can lead to mitochondrial
dysfunctions and neuronal apoptosis. Arsenic generates reac- Material and Methods
tive oxygen species (ROS) and disrupts the antioxidant de-
fense mechanism [54]. Antioxidant enzymes play a very im- Animal and Treatment
portant role in protecting the cell molecules from free radical-
generated oxidative stress. A lot of animal studies have report- Perinatal exposure of rats to sodium arsenite: Adult female
ed enhanced oxidative stress accompanied by the reduction in rats of proven fertility were kept for mating with the males
antioxidant enzyme activities in rats exposed to arsenic [46, in the ratio of 2:1. The day of appearance of the vaginal plug
52]. Neurodegeneration in pathological condition can occur as was considered as day one (GD-1) of gestation, and the preg-
a result of endogenous oxidative stress and reactive oxygen nant rats were separated into individual cages. Pregnant rats
species (ROS) generation in the central nervous system (CNS) were divided into three groups (6 pregnant rats in each group)
[12, 15]. The pathogenesis resulting from neuronal cell death and treated with sodium arsenite (SIGMA, USA) on day six of
is believed to involve in the imbalance of oxidative and anti- gestation (GD-6) until postnatal day (PD) 21 as follows:
oxidant system and increase in ROS production with a con-
comitant impairment of the mitochondrial functions [14, 25]. Group I- Pregnant rats were treated with normal saline (p.o.)
Oxidative state plays an important function in the regulation identically as in groups II and III and served as
and control of neuronal cell death in the brain through its controls.
interaction with cellular molecules and cell signaling path- Group II- Pregnant rats were treated with sodium arsenite
ways. Oxidative stress has been considered as an important dissolved in distilled water (2 mg/kg body weight,
mechanism in the neurotoxicity [6, 10]. In addition, mitochon- p.o.) once daily from GD6 to PD21.
dria have been implicated in the cellular damage and induction Group III- Pregnant rats were treated with sodium arsenite
of apoptotic cell signaling pathways during this neurotoxicity (4 mg/kg body weight, p.o.) dissolved in distilled
[47, 57], and the fact that mitochondrial dysfunctions may water once daily from GD6 to PD21.
lead to the severe pathological condition in neurodegenerative
diseases. Being the major source of energy in the cell, mito- The pregnant rats were allowed to deliver naturally and to
chondria may be an important factor contributing to the cell nurse their pups until PD21. Martínez et al. [40] reported that
death and redox signaling [27]. Generation of ROS in the 200 to 250 g rats consumed 36 ppm of arsenic, a concentration
electron transport chain may cause mitochondrial respiratory that results in ingestion of 3–4 mg/kg/day of arsenic. This
chain dysfunction. Generation of ROS in the mitochondrial parts per million range (3100 μg/L) of arsenic has also been
respiratory chain has been linked to neurotoxicity-associated reported in drinking water in the United States of America
changes in the CNS through the impairment in mitochondrial (USA) and Bangladesh [60]. Exposure to arsenic at doses
functions and neuronal homeostasis. In order to better under- 0.3–0.4 mg/kg/day (an exposure level that has already been
stand the mechanisms underlying arsenic toxicity, the present reported in human population) has not been found toxic ef-
study evaluated the redox homeostasis and energy metabolism fects. However, structural alterations in myelinated tracts in
in the frontal cortex, hippocampus, and corpus striatum of brain regions have been found following exposure to arsenic
arsenic-exposed rats. Regarding bioenergetics, we investigat- at doses 3–4 mg/kg body weight/day [40, 48, 66]. The doses
ed the activities of respiratory chain complexes and mitochon- of arsenic (2 mg/kg or 4 mg/kg body weight/day) were select-
drial membrane potential (ΔΨm). Impairment in the functions ed in the present study based on these reports.
of mitochondria plays a major role in apoptosis. All the litters were culled on PD2; 8–10 pups with an equal
Although neurotoxicity of arsenic in adults is well charac- allocation of males and females. The animals were sacrificed,
terized, information regarding the developmental neurotoxic- and the brain was taken out quickly, washed in ice-cold saline,
ity is limited. The presence of arsenic in groundwater and and dissected into specific regions—frontal cortex, corpus
extensive industrial uses of arsenic have raised concerns about striatum, and hippocampus following the standard procedure
its adverse effects on public health. Therefore, the rationale of as described by Glowinski and Iversen [19]. The dissected
the present study was to elucidate the mechanism of arsenic- brain regions were stored at −80 °C for neurochemical assays.
induced neurotoxicity in the developing brain. However, no To understand whether arsenic-induced changes are transient
report currently exhibits the effect of arsenic on the oxidative or persistent, another set of experiment was done on the same
stress, mitochondrial membrane potential, mitochondrial conditions as mentioned above. In this set of experiment, male
complexes of the electron transport chain, and the apoptosis pups were separated from mothers on PD22 in each group and
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

left as such until PD45. Effects on neurochemical endpoints Estimation of ROS Generation Amount of ROS generation
were studied on PD45. Unfortunately, we did not assess the was assessed fluorometrically by measuring the conversion
effect of arsenic on female pups. of DCFH-DA dye into highly fluorescent DCF product in
The study was approved by the institutional animal ethics the presence of H2O2 [13]. Briefly, the mitochondrial fraction
committee of CSIR-IITR, Lucknow (Ref No. ITRC/ was incubated with DCFH-DA (10 mM, 5 μl) in the water
IAEC/35/11-41/12, dated September 4, 2012), and all exper- bath (30 min, 37 °C) in a total reaction mixture of 1 ml. After
iments were carried out in accordance with the guidelines incubation, the mitochondria were pelleted by centrifugation
approved by the Committee for the Purpose of Control and (1200 g, 10 min) and re-suspended in fresh HEPES buffer.
Supervision of Experiments on Animals (CPCSEA), Ministry ROS generation was assessed in triplicate for each sample,
of Environment and Forests (Government of India), New and the fluorescence was recorded at excitation/emission—
Delhi, India. 488/525 nm. The ROS generation has been expressed as
fluorescence/μg protein [51].

Assessment of Oxidative Stress and ROS Generation Assessment of Mitochondrial Membrane Potential (ΔΨm)
MMP was assessed using JC-1 dye following the procedure
Assessment of Oxidative Stress as described by Kapoor et al. [32] with minor modifications.
Briefly, the dissected brain regions—frontal cortex, hippo-
Assay of Lipid Peroxidation, Protein Carbonyl, and Reduced campus, and corpus striatum—were kept in Hank’s balanced
Glutathione (GSH) Levels Malondialdehyde (MDA) as an end salt solution (HBSS) containing antibiotic and antimycotic.
product of lipid peroxidation levels was measured in frontal Mincing of tissues was carried out, followed by trypsinization
cortex, hippocampus, and corpus striatum by the method of at 37 °C for 5–7 min with trypsin (0.5 mg/ml) in HBSS con-
Ohkawa et al. [42]. The end products were read at 535 nm, and taining BSA (0.5%). Following incubation, trypsin was aspi-
the values are expressed as μmol MDA formed/h/mg protein. rated, and tissues were incubated (37 °C, 15 min) with DMEM
Protein carbonyl content was measured in the selected containing FBS (12%). Cells were dissociated from the tissues
brain regions by the method of Levine et al. [35] using 2,4- by passing the tissue pieces 20–30 times through 1-ml pipette
dinitrophenylhydrazine (DNPH) as the substrate. Amount of tip, and then by passing it through the 200-μl pipette tip. The
protein carbonyl was calculated using a molar extinction co- resulting cell suspension was filtered through a 50-μm diam-
efficient (€) 22 mM−1 cm−1 for aliphatic hydrazine, and values eter nylon mesh and pelleted by centrifugation (800 ×g for
are expressed as nmol/mg protein. 6 min). Cells were washed with PBS, centrifuged (800 ×g
GSH was assayed in selected brain regions by the method for 6 min), and re-suspended in PBS. The prepared cell sus-
of Hasan and Haider [22] using 5′5′-dithiobis 2-nitrobenzoic pension was then incubated with JC-1 (10 μM, 15 min,
acid (DTNB) as the color reagent. Further, GSH (ranging from 37 °C), washed with PBS, and re-suspended in PBS at a con-
2 to 10 μg) as a standard was run in parallel to plot the stan- centration of 106 cells/ml and analyzed through the flow
dard curve. The values are expressed as μg GSH/g tissue cytometer. Values are expressed as the number of depolarized
weight. cells in the single-cell suspension.

Assay of Superoxide Dismutase (SOD), Catalase (CAT), and Assay of Electron Complex Activity
Glutathione Peroxidase Activity SOD activity in brain regions
was measured following the method of Kakkar et al. [31] Isolation of Rat Brain Mitochondria Mitochondria from frontal
using NADH as a substrate in the post-mitochondrial fraction. cortex, hippocampus, and corpus striatum were isolated fol-
The activity of SOD has been expressed as unit/min/mg lowing the method of Bagh et al. [4]. Briefly, the tissues were
protein. homogenized in 10 ml of buffer (pH 7.4) containing mannitol
CAT activity was determined by the method of Aebi (225 mM), sucrose (75 mM), HEPES (5 mM), EGTA (1 mM),
[1] in the post-mitochondrial fraction of selected brain and BSA (1 mg/ml) and centrifuged (2000 ×g for 3 min) at
regions using hydrogen peroxide (H2O2) as the substrate. 4 °C. The supernatant was centrifuged (20,000 ×g for 10 min)
The decrease in optical density was measured at 240 nm at 4 °C to obtain the crude pellet of synaptosomes and mito-
for 150 s using the spectrophotometer. The activity of the chondria, which was further dissolved in homogenizing buffer
enzyme was calculated using the molar extinction coef- containing digitonin (0.02%) to lyse the synaptosomes and to
ficient 43.6 M cm −1 , and values are expressed as release the intra-synaptosomal mitochondria followed by cen-
μmol/min/mg protein. trifugation (12,000 ×g for 10 min). The pellet obtained in the
The activity of glutathione peroxidase in brain regions was process was washed twice in same buffer devoid of EGTA,
assayed by the method of Flohe and Gunzler [16]. The values BSA, and digitonin. Finally, the pellet was suspended in iso-
are expressed as nmol GSH oxidized/min/mg protein. tonic phosphate buffer (50 mM, pH 7.4).
Chandravanshi et al.

Assay of activity of mitochondrial complexes I, II–III, and Protein Estimation Protein content in the samples was mea-
IV: The activity of NADH (Complex I) was measured through sured following the procedure as described by Lowry et al.
ferricyanide as the electron acceptor [23]. Briefly, the assay [38] using bovine serum albumin as a reference standard.
was carried out at 30 °C in a reaction system containing
NADH (0.17 mM), potassium ferricyanide (0.6 mM), Statistical Analysis Results of the experiments were expressed
Triton-X 100 (0.1%) in phosphate buffer (50 mM, pH 7.4). as mean ± standard error, using a statistical software package
The reaction was initiated after the addition of mitochondrial (GraphPad Prism). The differences between the mean values
suspension (10~30 μg of protein) to the sample cuvette. The were evaluated by one-way ANOVA followed by Newman–
rate of oxidation of NADH was monitored by the decrease in Keuls test for post-hoc comparisons. p values less than 0.05
absorbance at 340 nm. The activity of complex I has been have been considered significant.
expressed as nmol NADH/oxidized/min/protein (mg).
The activity of succinate-cytochrome c reductase (com-
plexes II–III) was measured by reduction of ferricytochrome Results
c to ferrocytochrome c under influence of succinate at 550 nm.
The reaction cocktail in cuvette consisted of succinate Effect on the Extent of Oxidative Stress and ROS
(2 mM), KCN (1 mM), EDTA (0.3 mM), and cytochrome c Generation
(1.2 mg/ml) in phosphate buffer (100 mM, pH 7.4) in a total
volume (1 ml). The reaction was initiated by adding mito- Effect on Lipid Peroxidation, Protein Carbonyl, and GSH
chondrial suspension (10~30 μg protein). The activity of com- Levels in Brain Regions Arsenic-induced oxidative stress was
plexes II–III has been expressed as nmol oxidized cytochrome evident by the increased thiobarbituric acid reactive sub-
c reduced/min/protein (mg) [11]. stances (TBARS) and protein carbonyl contents measured in
The activity of cytochrome oxidase (complex IV) was mea- the frontal cortex, hippocampus, and corpus striatum of devel-
sured by oxidation of reduced cytochrome c (ferrocytochrome oping rats. Perinatal exposure to arsenic (2.0 or 4.0 mg/kg
c) at 550 nm. The reaction cocktail consisted of phosphate body weight) significantly increased the levels of TBARS in
buffer (10 mM, pH 7.4) at room temperature and ferricyanide the frontal cortex (43%, p < 0.05; 74%, p < 0.01), hippocam-
(1 mM) which was added to oxidize ferrocytochrome c. The pus (34%, p < 0.05; 66%, p < 0.05), and corpus striatum
reaction was initiated by adding mitochondrial suspension (36%, p < 0.01; 62%, p < 0.001) of arsenic-exposed rats on
(10~30 μg protein) [62]. The activity of complex IV has been PD22 as compared to controls (Fig. 1a). The TBARS level
expressed as nmol reduced cytochrome c oxidized/min/pro- remained significantly increased in the frontal cortex (20%,
tein (mg). p > 0.05; 34%, p < 0.05), hippocampus (19%, p > 0.05; 22%,
p < 0.05), and corpus striatum (18%, p > 0.05; 38%, p < 0.01)
Western Blotting Expression of proteins linked with apo- of rats exposed to arsenic at a higher dose even on PD45 as
ptosis and stress (Caspase-3, Bax, Bcl2, P-38, p-JNK, Ap- compared to respective controls; a general trend of recovery
1) in the frontal cortex, hippocampus, and corpus striatum was however seen (Fig. 1a).
was assayed following the method of Yadav et al. [64]. An increase in protein carbonyl levels in the frontal cortex
The prepared samples (30 μg protein/lane) were fractioned (30%, p < 0.05; 63%, p < 0.001), hippocampus (31%,
by 12% SDS-PAGE. Proteins were electrically transferred p < 0.05; 54%, p < 0.01), and corpus striatum (54%,
to nitrocellulose membranes (Millipore, USA) and blocked p < 0.01; 77%, p < 0.01) was observed in rats perinatally ex-
with blocking buffer (Western blocker solution, TM posed to arsenic (2.0 or 4.0 mg/kg body weight) on PD22 as
Sigma, USA). After subsequent washing, the blots were compared to controls (Fig. 1b). Protein carbonyl levels
incubated with primary antibody anti-Bax, anti-Bcl2, cas- remained significantly increased in the frontal cortex (18%,
pase-3, anti-P-38, anti-p-JNK, and anti-Ap-1 (rabbit) (Cell p > 0.05; 38%, p < 0.05) and hippocampus (17%, p > 0.05;
Signaling Technology, USA, 1:1000) for 24 h at 4 °C. 25%, p > 0.05) in rats exposed to arsenic at a higher dose
Anti-β actin was used as a loading control. Blots were (4.0 mg/kg body weight) on PD45. However, protein carbonyl
subsequently incubated with horseradish peroxidase-linked levels remained significantly higher in the corpus striatum
secondary antibody (anti-rabbit IgG 1:2000) for 60 min at (21%, p < 0.05; 34%, p < 0.01) of arsenic-exposed rats at both
room temperature. After the incubation, blots were washed doses, even on PD45 as compared to respective controls (Fig.
and developed using an Immobilon western chemilumines- 1b). In the corpus striatum, recovery of protein carbonyl con-
cent HRP substrate (Millipore, USA) following the recom- tent was seen less than other regions, after the withdrawal of
mended procedure. The densitometry for protein-specific arsenic exposure.
bands was carried out using Gel Documentation System A decrease in the GSH levels in the frontal cortex (33%,
(Alpha Innotech) with the help of Alpha Ease FC Stand p < 0.01; 55%, p < 0.001), corpus striatum (28%, p < 0.01;
Alone V.4.0 software. 44%, p < 0.001), and hippocampus (27%, p < 0.01; 39%,
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

Fig. 1 Effects of perinatal A


12 CONT As I As II
exposure (GD6 to PD21) to ***

μ MDA/hour/mg protein
arsenic (2 or 4 mg/kg body 10 **
**
weight/day) on MDA (a), protein * **
8 * *
carbonyl contents (b), and the *
levels of GSH (c) were assessed 6 *
in the frontal cortex,
4
hippocampus, and corpus
striatum of rats on PD22 and 2
23 days after withdrawal of
0
exposure on PD45. Data are PD22 PD45 PD22 PD45 PD22 PD45
reported as means ± SEM for five Frontal Cortex Hippocampus Corpus Striatum
animals in each group. B
80
Statistically significant **

Unit/min/mg protein
differences from control group, **
60
*p < 0.05, **p < 0.01, and
***p < 0.001, were determined **
*
by one-way-ANOVA with 40 *** **
* * *
Newman–Keuls test
20

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum
C
25
n mole GSH oxidized/

20
min/mg protein

*
15 ** *
* * * * ***
** *
10 **

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

p < 0.001) was evident in rats perinatally (GD6-PD21) exposed (21%, p < 0.05; 35%, p < 0.01) in arsenic-exposed rats on
to arsenic (2.0 or 4.0 mg/kg body weight) on PD22 as com- PD45 as compared to controls (Fig. 2a).
pared to controls (Fig. 1c). A trend of recovery in decreased Perinatal exposure to arsenic (2.0 or 4.0 mg/kg body
levels of GSH was observed in the frontal cortex and hippo- weight) from GD6 to PD21 resulted to a decrease in the ac-
campus of rats exposed to arsenic at a low dose as compared to tivity of CAT in the frontal cortex (29%, p < 0.01; 46%,
those exposed at a higher dose on withdrawal of exposure on p < 0.001), hippocampus (23%, p < 0.05; 46%, p < 0.01),
PD45 (Fig. 1c). While a trend of recovery was also found in the and corpus striatum (25%, p < 0.01; 51%, p < 0.001) on
decreased levels of GSH in the corpus striatum (28%, p < 0.05; PD22 as compared to controls (Fig. 2b). The CAT activity in
44%, p < 0.05) of arsenic-exposed rats, changes remained in the selected brain regions was remained significantly de-
rats exposed to arsenic at both doses of arsenic, on PD45 in creased at a higher dose of arsenic (4.0 mg/kg body weight)
comparison to respective controls (Fig. 1c). on PD45 as compared to respective controls (Fig. 2b).
Perinatal exposure to arsenic (2.0 or 4.0 mg/kg body
Effect on the Activity of SOD, CAT, and Glutathione weight) resulted in decreased activity of glutathione peroxi-
Peroxidase in Brain Regions Activity of SOD was found to dase in the frontal cortex (24%, p < 0.05; 36%, p < 0.01), hip-
be decreased significantly in the frontal cortex (35%, p < 0.01; pocampus (24%, p < 0.05; 39%, p < 0.01), and corpus stria-
58%, p < 0.001), hippocampus (24%, p < 0.05; 48%, tum (31%, p < 0.01; 42%, p < 0.001) of rats on PD22 as com-
p < 0.001), and corpus striatum (31%, p < 0.01; 53%, pared to controls (Fig. 2c). The decrease in the activity of
p < 0.001) of rats perinatally exposed to arsenic on PD22 as glutathione peroxidase was found to persist in the frontal cor-
compared to controls (Fig. 2a). The activity of SOD in the tex (19%, p < 0.05; 26%, p < 0.05) and corpus striatum (23%,
frontal cortex and hippocampus of arsenic-exposed rats exhib- p < 0.05; 33%, p < 0.05) of rats exposed to arsenic at both the
ited a trend of recovery on withdrawal of arsenic exposure on doses on withdrawal of arsenic exposure as compared to re-
PD45 as compared to controls (Fig. 2a). The decrease in the spective controls on PD45 (Fig. 2c). No significant change in
activity of SOD was found to persist in the corpus striatum the activity of glutathione peroxidase in the hippocampus
Chandravanshi et al.

Fig. 2 Effects of perinatal CONT As I As II


exposure (GD6 to PD21) to A
2.5
arsenic (2 or 4 mg/kg body

Unit/min/mg protein
weight/day) on activities of SOD 2
(a), CAT (b), and glutathione *
*
peroxidase (c) were assessed in 1.5 ** *
the frontal cortex, hippocampus, ***
1 ***
and corpus striatum of rats on **
PD22 and 23 days after ***
withdrawal of exposure on PD45. 0.5
Data are reported as means ±
0
SEM for five animals in each PD22 PD45 PD22 PD45 PD22 PD45
group. Statistically significant Frontal Cortex Hippocampus Corpus Striatum
differences from control group, B 9
*p < 0.05, **p < 0.01, and 8

μmole/min/mg protein
***p < 0.001 were determined by 7
one-way ANOVA with Newman– *
6
Keuls test
5 * ** *
**
4 ** *
***
3 ***
2
1
0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

C
350
GSH μgm/gm tissue weight

300
250
*
200 * ** * *
** **
***
150 *** ***

100
50
0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

(12%, p > 0.05; 21%, p < 0.05) was observed in rats exposed Effect on the Mitochondrial Membrane Potential (ΔΨm) in
to arsenic at a low dose while the activity of glutathione per- Brain Regions Exposure to arsenic for perinatal period
oxidase remained significantly decreased in those exposed to (GD6–PD21) had a marked effect on MMP in selected brain
arsenic at a higher dose as compared to respective controls on regions. Arsenic dose dependently increased the number of
PD45 (Fig. 2c). depolarized cells in the frontal cortex (7.9%, p < 0.05;
12.1%, p < 0.001), hippocampus (8.66%, p < 0.01; 11.53%,
Effect on the ROS Generation in Brain Regions ROS genera- p < 0.001), and corpus striatum (11.57%, p < 0.01; 16.27%,
tion was found to be markedly affected by both doses of arse- p < 0.001), indicating a decrease in the MMP on PD22 as
nic on PD22. ROS generation was increased by the arsenic compared to controls. Percentage of depolarize cells was
exposure in the frontal cortex (48%, p < 0.05; 86%, found to persist in the frontal cortex (7.2%, p < 0.05; 7.8%,
p < 0.001), hippocampus (48%, p < 0.05; 68%, p < 0.01), p < 0.01), hippocampus (5.7%, p < 0.05; 6.83%, p < 0.01),
and corpus striatum (54%, p < 0.05; 90%, p < 0.01) of devel- and corpus striatum (9.36%, p < 0.05; 11.67%, p < 0.001) of
oping rats on PD22 as compared to controls. The increase in rats exposed to arsenic as compared to the respective controls
the ROS generation was more intense in corpus striatum than after the 23 days after cessation of exposure (Fig. 3b).
in frontal cortex and hippocampus. Although a trend of recov-
ery in the ROS generation was observed on withdrawal of Effect on the Activity of Mitochondrial Complexes in Brain
arsenic exposure, the increase in the ROS generation was Regions As there was a loss of MMP following arsenic
found to persist in the frontal cortex (30%, p < 0.05; 36%, administration in rats, it was of curiosity, interest to ex-
p < 0.05), hippocampus (19%, p < 0.05; 29%, p < 0.05), and plore if this would have an impact on the electron trans-
corpus striatum (32%, p < 0.05; 43%, p < 0.05) on PD45 as port chain. Activities of mitochondrial complexes were
compared to controls (Fig. 3a). found to be altered by arsenic during the perinatal period
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

Fig. 3 Effects of perinatal A


exposure (GD6 to PD21) to 8 CONT As I As II

DCF fluorescence/μg protein


**
arsenic (2 or 4 mg/kg body 7 ***
weight/day) on ROS levels (a) *
6 **
and MMP (b) were assessed in
* *
the frontal cortex, hippocampus, 5 *
*
*
and corpus striatum of rats on * *
PD22 and 23 days after 4
withdrawal of exposure on PD45. 3
In the upper panel figure of b,
representative dot plot of an 2
independent experiment in each 1
group is presented. Depolarize
0
cells (%) in each group are PD22 PD45 PD22 PD45 PD22 PD45
calculated and presented in the Frontal Cortex Hippocampus Corpus Striatum
lower panel of b. Data are
reported as means ± SEM for five B Frontal Cortex Hippocampus Corpus Striatum
animals in each group.
Statistically significant CONT
5 2.9 4.8 3.3
differences from the control 5.2 3.2

group, *p < 0.05, **p < 0.01, and


***p < 0.001 were determined by As I 5.6 11.8 7.2
7.3 6.7 8.4
one-way ANOVA with Newman–
Keuls test
As II 12.4 7 12.8 6.6 18.5 12.2

PD22 PD45 PD22 PD45 PD22 PD45


18 ***

15
Depolarize cells

*** *** **
12 ***
*
9 **
* **
* **
6 *

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

in selected brain regions of rats at both doses. The activity p < 0.05; 33%, p < 0.01) on PD22 in comparison to con-
of NADH (complex I) was found to be decreased signif- trols (Fig. 4c). While a trend of recovery in the activity of
icantly in the frontal cortex (18%, p > 0.05; 29%, complex I and complex IV was observed both in frontal
p < 0.05), hippocampus (20%, p > 0.05; 31%, p < 0.05), cortex and hippocampus, the activity of both complexes
and corpus striatum (22%, p < 0.05; 35%, p < 0.001) of remained decreased in the corpus striatum of rats exposed
rats perinatally arsenic exposed rats to 4 mg/kg arsenic to arsenic at a higher dose on withdrawal of exposure.
on PD22 as compared to controls (Fig. 4a). A significant Decrease in the activity of complexes II–III remained in
loss in the activity of succinate-cytochrome c reductase the frontal cortex (14%, p > 0.05; 31%, p < 0.05) and hip-
(complexes II–III) in the frontal cortex (29%, p < 0.05; pocampus (16%, p > 0.05; 27%, p < 0.05) of rats exposed
45%, p < 0.01), hippocampus (27%, p < 0.05; 49%, to arsenic at a higher dose on the withdrawal of exposure
p < 0.01), and corpus striatum (34%, p < 0.05; 66%, on PD45 as compared to controls. The decrease in the
p < 0.001) was also evident in rats perinatally exposed to activity of complexes II–III in the corpus striatum (22%,
arsenic (2.0 mg/kg or 4.0 mg/kg body weight) in compar- p < 0.05; 35%, p < 0.05) was found to persist in rats ex-
ison to those in the control group (Fig. 4b). Arsenic- posed to arsenic at both doses on PD45 in comparison to
exposed rats also exhibited a decrease in the activity of controls. Maximum recovery was observed in complexes I
cytochrome oxidase (complex IV) in the frontal cortex and IV in selected brain regions of arsenic-exposed rats,
(17%, p > 0.05; 32%, p < 0.05), hippocampus (24%, while complexes II–III showed less recovery after the
p < 0.05; 33%, p < 0.05), and corpus striatum (23%, withdrawal of arsenic exposure.
Chandravanshi et al.

Fig. 4 Effects of perinatal A


exposure (GD6 to PD21) to 500 CONT As I As II

n mole NADH oxidized/


arsenic (2 or 4 mg/kg body
400

min/mg/protein
weight/day) on activity of
mitochondrial complexes *
300 *
(complex I (a), complexes II and * **
*
III (b), and complex IV (c)) were 200
assessed in the frontal cortex,
hippocampus, and corpus 100
striatum of rats on PD22 and
23 days after withdrawal of 0
PD22 PD45 PD22 PD45 PD22 PD45
exposure on PD45. Data are Frontal Cortex Hippocampus Corpus Striatum
reported as means ± SEM for five
animals in each group. B
100

Reduced/ min/mg/protein
Statistically significant

n mole oxidized Cyt C


differences from control group, 80
*p < 0.05, **p < 0.01, and *
60 *
***p < 0.001, were determined * * * *
by one-way ANOVA with ** *
40 ** ***
Newman–Keuls test
20

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum
C
450
400
350
300
*
250 * ** *
200
150
100 *
*
50
0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

Effect on the Expression of Key Proteins Associated with p < 0.001), and corpus striatum (1.48-fold, p < 0.01; 1.82-fold,
Apoptosis in Brain Regions As shown in Fig. 5a, the ratio p < 0.001) of rats perinatally exposed to arsenic (2.0 mg/kg or
of Bcl-2/Bax was markedly increased in the frontal cortex 4.0 mg/kg body weight) on PD22 in comparison to controls
(1.90-fold, p < 0.01; 2.86-fold, p < 0.001), hippocampus (Fig. 5b). While a trend of recovery was observed in the ex-
(1.52-fold, p < 0.01; 1.99-fold, p < 0.001), and corpus stri- pression of caspase-3 on withdrawal of arsenic exposure.
atum (2.20-fold, p < 0.01; 3.26-fold, p < 0.01) of rats fol- However, expression of caspase-3 remained increased in the
lowing perinatal exposure to arsenic (2.0 mg/kg or frontal cortex, hippocampus, and corpus striatum even 23 days
4.0 mg/kg body weight) compared to the control. A trend after cessation of exposure in comparison to respective con-
of recovery was, however, seen in the Bcl-2/Bax ratio after trols (Fig. 5b).
withdrawal of exposure, but a significant increase in ratio Protein expression of Phospo p-38 was found to be signifi-
in the frontal cortex, hippocampus, and corpus striatum of cantly increased in the frontal cortex (1.24-fold, p < 0.05; 1.48-
rats or animals exposed to arsenic at both doses (Fig. 5a). fold, p < 0.001), hippocampus (1.27-fold, p < 0.01; 1.32-fold,
The intensity of change in the ratio of Bcl-2/Bax was more p < 0.01), and corpus striatum (1.98-fold, p < 0.001; 2.41-fold,
pronounced in the corpus striatum than in the other re- p < 0.001) of rats on PD22 in comparison to controls (Fig. 6a).
gions, even 23 days after withdrawal of exposure. Expression of Phospo p-38 was found to recover at a low dose
The results showed that the arsenic administration signifi- of arsenic in the frontal cortex and hippocampus of rats, but the
cantly increased the expression of apoptotic hallmark protein change remained persistent in the corpus striatum in arsenic-
caspase-3 in the frontal cortex (1.45-fold, p < 0.01; 1.61-fold, exposed rats on withdrawal of exposure on PD45 as compared
p < 0.001), hippocampus (1.48-fold, p < 0.01; 1.82-fold, to respective controls (Fig. 6a).
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

CONT As I As II
A Hippocampus Corpus Striatum
Frontal Cortex
Bcl2 20 kda
Bax 26 kda
β actin 42 kda
CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II
PD22 PD45 PD22 PD45 PD22 PD45

3.6 **

3 ***

2.4
Fold change

** **
** ***
1.8 ** *
** **
1.2

0.6

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum
B
Frontal Cortex Hippocampus Corpus Striatum
Caspase-3 35kda
β actin 42 kda
CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II
PD22 PD45 PD22 PD45 PD22 PD45
2
***
*** ***
1.6 *** **
***
** **
*
Fold change

1.2 * *

0.8

0.4

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum
Fig. 5 Effects of perinatal exposure (GD6 to PD21) to arsenic (2 or densitometric analysis of bands in the control and arsenic-exposed
4 mg/kg body weight/day) on the expression of Bcl2, Bax ratio (a), and groups. Data are reported as means ± SEM for three animals in each
caspase-3 (b) were assessed in frontal cortex, hippocampus, and corpus group. Statistically significant differences from the control group,
striatum of rats on PD22 and 23 days after withdrawal of exposure on *p < 0.05, **p < 0.01, and ***p < 0.001, were determined by one-way
PD45. Each panel shows a representative Western blot (top) and ANOVA with Newman–Keuls test

When compared to control, we found that perinatal expo- and no significant variations occurred in the frontal cortex and
sure to arsenic (2.0 mg/kg or 4.0 mg/kg body weight) on PD22 hippocampus. Interestingly, overexpression of AP-1 was
led to significantly enhanced expression of AP-1 in the frontal remained significantly increased in the corpus striatum of rats
cortex (1.47-fold, p < 0.05; 1.71-fold, p < 0.01), hippocampus exposed to arsenic at both doses after the withdrawal of expo-
(1.48-fold, p < 0.05; 1.57-fold, p < 0.05), and corpus striatum sure as compared to controls (Fig. 6b).
(1.55-fold, p < 0.01; 1.61-fold, p < 0.01) (Fig. 6b). A trend of Expression of JNK1 and JNK2 was significantly
recovery was recorded 23 days after withdrawal of exposure, higher in the corpus striatum of rats exposed to arsenic
Chandravanshi et al.

A CONT As I As II

Frontal Cortex Hippocampus Corpus Striatum


p-p38 40 kda
β actin 42 kda
CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II
PD22 PD45 PD22 PD45 PD22 PD45
2.8
2.4 ***
Fold change

2 ***

1.6 ***
* ** ** ** ** **
1.2 *

0.8
0.4
0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

B
Frontal Cortex Hippocampus Corpus Striatum
Ap-1 39 kda
β actin 42 kda
CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II
PD22 PD45 PD22 PD45 PD22 PD45
2
**
1.6 * ** **
* * *
*
1.2
Fold change

0.8

0.4

0
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum

Fig. 6 Effects of perinatal exposure (GD6 to PD21) to arsenic (2 or analysis of bands in the control and arsenic-exposed groups. Data are
4 mg/kg body weight/day) on the expression of p-p38 (a) and AP-1 (b) reported as means ± SEM for three animals in each group. Statistically
were assessed in the frontal cortex, hippocampus, and corpus striatum of significant differences from the control group, *p < 0.05, **p < 0.01, and
rats on PD22 and 23 days after withdrawal of exposure on PD45. Each ***p < 0.001, were determined by one-way ANOVA with Newman–
panel shows a representative Western blot (top) and densitometric Keuls test

on PD22 as compared to controls (Fig. 7). Increase in Discussion


the expression of JNK1 and JNK2 persisted in rats ex-
posed to arsenic at a higher dose (4.0 mg/kg body Developmental neurotoxicity of arsenic in human has been
weight), while recovered in these exposed at a low dose defined as elevated risks of cognitive dysfunction, learning,
(2.0 mg/kg body weight) on withdrawal of exposure on and memory impairment in children [33]. Prenatal arsenic
PD45. No significant changes were observed in the ex- exposure has been shown to target the different brain regions
pression of JNK-1 and JNK-2 proteins in frontal cortex of rats [2, 40]. At the molecular level, arsenic is known to
and hippocampus of arsenic (2.0 mg/kg or 4.0 mg/kg cross the blood brain barrier and interfere with the neuronal
body weight)-exposed rats as compared to controls on functions in the brain [28]. Previous studies also demonstrated
PD22 and PD45 (Fig. 7). that early life exposure to arsenic impairs cholinergic
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

CONT As I As II
Frontal Cortex Hippocampus Corpus Striatum
pJNK-2 54 kda
pJNK-1 46 kda

β actin 42 kda

CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II CONT As I As II


2.5 PD22 PD45 PD22 PD45 PD22 PD45

***
2
**
**
Fold change

1.5 *
* *

0.5

0 JNK-2 JNK-1 JNK-2 JNK-1 JNK-2 JNK-1 JNK-2 JNK-1 JNK-2 JNK-1 JNK-2 JNK-1
PD22 PD45 PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus Corpus Striatum
Fig. 7 Effects of perinatal exposure (GD6 to PD21) to arsenic (2 or the control and arsenic-exposed groups. Data are reported as means ±
4 mg/kg body weight/day) on the expression of p-JNK was assessed in SEM for three animals in each group. Statistically significant
the frontal cortex, hippocampus, and corpus striatum of rats on PD22 and differences from the control group, *p < 0.05, **p < 0.01, and
23 days after withdrawal of exposure on PD45. Each panel shows a ***p < 0.001 were determined by one-way ANOVA with Newman–
representative Western blot (top) and densitometric analysis of bands in Keuls test

dopaminergic dysfunctions in the frontal cortex, hippocam- correlated to enhanced oxidative stress in developing rats after
pus, and corpus striatum of rats at low doses of arsenic [8, exposure to 2 or 4 mg/kg arsenic during the perinatal period.
9]. Therefore, in order to better understand the developmental We then evaluated different biomarkers of oxidative stress in
neurotoxicity of arsenic and to confirm the toxicity exerted by the frontal cortex, hippocampus, and corpus striatum of
a prenatal exposure to different brain regions of rats, we eval- arsenic-exposed rats. We observed that GSH levels were de-
uated the effects of arsenic on ROS generation, oxidant, and creased in the frontal cortex, hippocampus, and corpus striatum
antioxidant molecules, and mitochondrial functions in brain of rats treated with arsenic in comparison with the control
regions of arsenic-exposed rats with normal. group. In order to further evaluate the activity of antioxidant
One of the most important mechanisms involved in the neu- enzymes, we examined the activities of SOD, CAT, and gluta-
rotoxicity of arsenic is their ability to cause oxidative stress and thione peroxidase. We examined that low levels of arsenic de-
mitochondrial dysfunction [46]. Enhanced oxidative stress is creased the activities of CAT, SOD, and glutathione peroxidase
found in cognitive impairment, along with increased and de- in selected brain regions of rats. Hence, a reduced level of GSH
creased levels of oxidant and antioxidant markers, respectively combined with a decreased glutathione peroxidase activity may
[17, 18, 29]. High levels of polyunsaturated fatty acids, high lead to the accumulation of oxidatively damaged molecules and
metabolic rate, lower activity of brain antioxidant (CAT, SOD, enhances the vulnerability of the brain to oxidative insult by
and glutathione peroxidase) and reduced capacity of cellular arsenic exposure [30, 54]. A decrease in SOD enzymes may
regeneration make the brain highly susceptible target to dam- lead to the generation of superoxide radical and hydrogen per-
age by reactive oxygen species [43]. Neurodegeneration, one of oxide in the brain. Hydrogen peroxide is decomposed biolog-
the neurotoxic effects of arsenic, was induced by increase in the ically by the CAT enzyme whose activity also showed a signif-
generation of ROS in the brain [13, 39]. The present study icant fall following perinatal arsenic exposure in brain regions.
indicated that prenatal exposure to 2 or 4 mg/kg arsenic results Accumulation of superoxide and peroxide radicals leads to
in oxidative stress to induce an increment in ROS. Arsenic has peroxidation of lipids in membrane resulting in increased lipid
high affinity to glutathione; it acts as a scavenger of reactive peroxidation and protein carbonyl contents as observed in the
species, which might have implications for the maintenance of present study. Based on the observation that accumulation of
thiol-disulfide balance. Thus, reduced levels of GSH are TBARS products and protein carbonyl contents was observed
Chandravanshi et al.

in brain regions of exposed rats, it indicates the loss of mem- exposure to different doses. Increased ratio of Bcl2/Bax pro-
brane integrity and function. Increased oxidant and decreased teins and increased levels of caspase-3 are reported in various
antioxidant molecules were observed in arsenic-exposed devel- neurodegenerative conditions [49, 55].
oping rats as previously shown in other reports [24, 53]. On the MAPK signaling pathways are activated in response to
other hand, there is evidence suggesting neurotoxic effects of stress. Thus, JNK and p38 MAPK signaling pathways are
arsenic, which accumulate in the frontal cortex, hippocampus, stimulated by ROS [61]. In this study, we observed that
and corpus striatum of developing rats [8, 9]. In this regard, ROS production seems to induce the activation of JNK or
arsenic has a high affinity to glutathione that can react with and p38 MAPK pathways. JNK and p38 have classically been
induce the production of free radicals [21]. In the present study, associated with apoptosis. In the present study, exposure to
we further studied the role of mitochondrial complexes and arsenic at low doses did not affect the expression of p-JNK1
mitochondrial membrane potential in arsenic-induced oxidative and p-JNK2 both in the frontal cortex and hippocampus of
stress and its mechanisms. Accumulation of ROS may trigger developing rats. However, expression of p-JNK1 and p-JNK2
the inhibition of the activity of mitochondrial complexes by was found to increase in corpus striatum of arsenic-exposed
oxidative phosphorylation. In relation to this, we observed that rats as compared to controls. Further, we observed a marked
arsenic decreased the activities of complexes I, II–III, and IV in increased the protein expression of phosphorylated p38
the frontal cortex, hippocampus, and corpus striatum of rats. MAPK in different brain regions of arsenic-exposed develop-
Our previous study also demonstrated that arsenic impairs the ing rats. AP-1 is a nuclear transcriptional factor, which is also
activities of mitochondrial complexes in the different brain re- involved in apoptosis. Expression of AP-1 can be modulated
gions of early life exposed rats, supporting the observation that by the MAPK signaling pathway [56]. Increased expression of
affected exposed rats present increase levels of ROS [9]. AP-1 observed in different brain regions could be easily asso-
Inhibition of the electron transport complex activity leads ciated with enhanced apoptosis as a result of arsenic exposure.
to abnormal accumulation of ROS. Accumulation of ROS is Up-regulation of p38 and pJNK and AP-1 in developing brain
responsible for the lipid bi-layer damage [52] and caused mi- suggest increased neuronal apoptosis, and these observations
tochondrial swelling and mitochondrial membrane potential are consistent with our earlier report demonstrating similar
(MMP) fall down [58], which suggests that mitochondrial changes in the frontal cortex and hippocampus [8]. These
function was inhibited by the increase level of ROS. In the results suggest that the arsenic-induced activation of AP-1
present study, fluctuation in the MMP and electron transport via a p38 MAPK that includes JNK is involved in the induc-
complex activity are considerable to be a distinctive triggers, tion of apoptosis in brain regions of developing rats.
underlying the pathogenesis of arsenic-induced mitochondrial In summary, our study shows that arsenic exposure im-
dysfunctions in developing rats. paired the mitochondrial functions and enhanced oxidative
Further, ample evidence suggested that oxidative stress and stress and apoptosis in selected brain regions of perinatal ex-
mitochondrial dysfunctions may cause neurodegeneration [7, posed rats. Regarding the response of different brain region
44]. Taken together, ROS and the resulting imbalance between tissues to the neurotoxic effects of arsenic, it is noteworthy
the antioxidant and oxidant molecules can be part of the signal that the corpus striatum was the most vulnerable region to
transduction pathway during apoptosis. It is now established disruption of redox homeostasis, mitochondrial dysfunctions,
that various pro-apoptotic factors are released into the cytosol and neuronal apoptosis induced by arsenic. As regards to the
during oxidative stress and mitochondrial dysfunctions. corpus striatum, we cannot explain at present the reasons by
Therefore, it seemed likely that arsenic would induce apoptosis which the corpus striatum presents more susceptibility to ar-
through ROS generation, oxidative stress, and mitochondrial senic in developing rats. In conclusion, arsenic affects the
dysfunctions. Apoptosis has shown to play a major role in the central nervous system during the perinatal period of brain
pathogenesis of various neurological disorders [26]. The bal- development. Consequently, it may be suggested that proper
ance between anti-apoptotic and apoptotic proteins determines testing of water for arsenic is important for women of child-
the fate of the cells either to undergo apoptosis or to survive in bearing age and pregnant women to avoid arsenic exposure
pathophysiology. Our data presented here reveal that arsenic during critical periods of brain development. Further, the con-
enhanced the Bcl2/Bax ratio in different brain regions of rats tinuous exposure to arsenic during perinatal periods of brain
in a dose-dependent manner. Increased ratio of Bcl2/Bax and development may be an important risk factor for increased
oxidative stress leads to the change in MMP of brain tissues of vulnerability to neurodegenerative diseases.
rats; therefore, it could enhance apoptosis. Increased Bcl2/Bax
ratio stimulates apoptotic signaling molecules along with the Acknowledgements The authors thank the Director, CSIR—Indian
Institute of Toxicology Research (CSIR-IITR), Lucknow, for his support
variety of caspases and finally caspase-3, which is the main
and the keen interest in the present study. Financial support by the
protein of mitochondrial pathway of apoptosis [45]. We ob- University Grants Commission, New Delhi, for carrying out the study
served that expression of caspase-3 was increased in the frontal is gratefully acknowledged.
cortex, hippocampus, and corpus striatum following arsenic
Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions

Compliance with Ethical Standards 18. Flora SJ, Mittal M, Mishra D (2009) Co-exposure to arsenic and
fluoride on oxidative stress, glutathione linked enzymes, biogenic
amines and DNA damage in mouse brain. J Neurol Sci 285(1–2):
Conflict of Interest None declared.
198–205
19. Glowinski J, Iversen LL (1966) Regional studies of catecholamines
in the rat brain. I. The disposition of [3H] norepinephrine, [3H]
References dopamine and [3H] dopa in variousregions of the brain. J
Neurochem 13(8):655–659
20. Gong G, O’Bryant SE (2010) The arsenic exposure hypothesis for
1. Aebi H (1984) Catalase in vitro. Methods Enzymol 105:121–126 Alzheimer disease. Alzheimer Dis Assoc Disord 24:311–316
2. Allan AM, Hafez AK, Labrecque MT, Solomon ER, Shaikh MN, 21. Hall MN, Niedzwiecki M, Liu X, Harper KN, Alam S, Slavkovich
Zheng X, Ali A (2015) Sex-dependent effects of developmental V, Ilievski V, Levy D, Siddique AB, Parvez F, Mey JL, van Geen A,
arsenic exposure on methylation capacity and methylation regula- Graziano J, Gamble MV (2013) Chronic arsenic exposure and
tion of the glucocorticoid receptor system in the embryonic mouse blood glutathione and glutathione disulfide concentrations in
brain. Toxicol Rep 2:1376–1390 Bangladeshi adults. Environ Health Perspect 121(9):1068–1074
3. Arikan I, Namdar ND, Kahraman C, Dagci M, Ece E (2015) 22. Hasan M, Haider SS (1989) Acetyl homocystein thiolactone protect
Assessment of arsenic levels in body samples and chronic exposure against some neurotoxic effects of thallium. Neurotoxicology
in people using water with a high concentration of arsenic: a field 10(2):257–261
study in Kutahya. Asian Pac J Cancer Prev 16(8):3183–3188 23. Hatefi Y (1978) Preparation and properties of NADH: ubiquinone
4. Bagh MB, Maiti AK, Jana S, Banerjee K, Roy A, Chakrabarti S oxidoreductase (complex I), EC 1.6.5.3. Methods Enzymol 53:11–14
(2008) Quinone and oxyradical scavenging properties of N- 24. Herrera A, Pineda J, Antonio MT (2013) Toxic effects of perinatal
acetylcysteine prevent dopamine mediated inhibition of Na+, K+- arsenic exposure on the brain of developing rats and the beneficial
ATPase and mitochondrial electron transport chain activity in rat role of natural antioxidants. Environ Toxicol Pharmacol 36(1):73–79
brain: implications in the neuroprotective therapy of Parkinson’s
25. Hinerfeld D, Traini MD, Weinberger RP, Cochran B, Doctrow SR,
disease. Free Radic Res 42(6):574–581
Harry J, Melov S (2004) Endogenous mitochondrial oxidative
5. Burgos S, Tenorio M, Zapata P, Cáceres DD, Klarian J, Alvarez N, stress: neurodegeneration, proteomic analysis, specific respiratory
Oviedo R, Toro-Campos R, Claudio L, Iglesias V (2017) Cognitive chain defects and efficacious antioxidant therapy in superoxide dis-
performance among cohorts of children exposed to a waste disposal mutase 2 null mice. J Neurochem 88(3):657–667
site containing heavy metals in Chile. Int J Environ Health Res
26. Honig LS, Rosenberg RN (2000) Apoptosis and neurologic disease.
27(2):117–125
Am J Med 108(4):317–330
6. Caito S, Aschner M (2015) Neurotoxicity of metals. Handb Clin 27. Hsieh HL, Yang CM (2013) Role of redox signaling in neuroin-
Neurol 131:169–189 flammation and neurodegenerative diseases. Biomed Res Int 2013:
7. Calì T, Ottolini D, Brini M (2011) Mitochondria, calcium, and 484613
endoplasmic reticulum stress in Parkinson’s disease. Biofactors 28. Huo TG, Li WK, Zhang YH, Yuan J, Gao LY, Yuan Y, Yang HL,
37(3):228–240 Jiang H, Sun GF (2015) Excitotoxicity induced by realgar in the rat
8. Chandravanshi LP, Yadav RS, Shukla RK, Singh A, Sultana S, Pant hippocampus: the involvement of learning memory injury, dysfunc-
AB, Parmar D, Khanna VK (2014a) Reversibility of changes in tion of glutamate metabolism and NMDA receptors. Mol Neurobiol
brain cholinergic receptors and acetylcholinesterase activity in rats 51(3):980–994
following early life arsenic exposure. Int J Dev Neurosci 34:60–75 29. Jomova K, Jenisova Z, Feszterova M, Baros S, Liska J, Hudecova
9. Chandravanshi LP, Shukla RK, Sultana S, Pant AB, Khanna VK D, Rhodes CJ, Valko M (2011) Arsenic: toxicity, oxidative stress
(2014b) Early life arsenic exposure and brain dopaminergic alter- and human disease. J Appl Toxicol 31(2):95–107
ations in rats. Int J Dev Neurosci 38:91–104 30. Kadeyala PK, Sannadi S, Gottipolu RR (2013) Alterations in apo-
10. Cicero CE, Mostile G, Vasta R, Rapisarda V, Signorelli SS, Ferrante ptotic caspases and antioxidant enzymes in arsenic exposed rat
M, Zappia M, Nicoletti A (2017) Metals and neurodegenerative brain regions: reversal effect of essential metals and a chelating
diseases. A systematic review. Environ Res 159:82–94 agent. Environ Toxicol Pharmacol 36(3):1150–1166
11. Clark JB, Bates TE, Boakye P, Kuimov A, Land JM (1997) 31. Kakkar P, Das B, Viswanathan PN (1984) A modified spectropho-
Investigation of mito-chondrial defects in brain and skeletal muscle. tometric assay of superoxide dismutase. Indian J Biochem Biophys
In: Turner AJ, Bachelard HS (eds) Neurochemistry: a practical 21:130–132
approach. Oxford University Press, Oxford, pp 151–174 32. Kapoor N, Pant AB, Dhawan A, Dwievedi UN, Gupta YK, Seth
12. Coyle JT, Puttfarcken P (1993) Oxidative stress, glutamate, and PK, Parmar D (2006) Differences in sensitivity of cultured rat brain
neurodegenerative disorders. Science 262(5134):689–695 neuronal and glial cytochrome P450 2E1 to ethanol. Life Sci
13. Dwivedi N, Mehta A, Yadav A, Binukumar BK, Gill KD, Flora SJ 79(16):1514–1522
(2011) MiADMSA reverses impaired mitochondrial energy metab- 33. Karri V, Schuhmacher M, Kumar V (2016) Heavy metals (Pb, Cd,
olism and neuronalapoptotic cell death after arsenic exposure in As and MeHg) as risk factors for cognitive dysfunction: a general
rats. Toxicol Appl Pharmacol 256(3):241–248 review of metal mixture mechanism in brain. Environ Toxicol
14. Fan J, Dawson TM, Dawson VL (2017) Cell death mechanisms of Pharmacol 48:203–213
neurodegeneration. Adv Neurobiol 15:403–425 34. Laborde A, Tomasina F, Bianchi F, Bruné MN, Buka I, Comba P,
15. Federico A, Cardaioli E, Da Pozzo P, Formichi P, Gallus GN, Radi Corra L, Cori L, Duffert CM, Harari R, Iavarone I, McDiarmid MA,
E (2012) Mitochondria, oxidative stress and neurodegeneration. J Gray KA, Sly PD, Soares A, Suk WA, Landrigan PJ (2015)
Neurol Sci 322(1–2):254–262 Children’s health in Latin America: the influence of environmental
16. Flohe L, Gunzler WA (1984) Assays of glutathione peroxidase. exposures. Environ Health Perspect 123(3):201–209
Methods Enzymol 105:114–121 35. Levine RL, Garland D, Oliver CN, Amici A, Climent I, Lenz AG,
17. Flora SJ, Bhadauria S, Pant SC, Dhaked RK (2005) Arsenic in- Ahn BW, Shaltiel S, Stadtman ER (1990) Determination of carbon-
duced blood and brain oxidative stress and its response to some yl content in oxidatively modified proteins. Methods Enzymol 186:
thiol chelators in rats. Life Sci 77(18):2324–2337 464–478
Chandravanshi et al.

36. Li H, Wang M, Liang Q, Jin S, Sun X, Jiang Y, Pan X, Zhou Y, Peng arsenic exposure and attention impairment in school children.
Y, Zhang B, Zhou A, Zhang Y, Chen Z, Cao J, Zhang H, Xia W, Cortex 74:370–382
Zheng T, Cai Z, Li Y, Xu S (2017) Urinary metabolomics revealed 51. Rush JW, Quadrilatero J, Levy AS, Ford RJ (2007) Chronic resver-
arsenic exposure related to metabolic alterations in general Chinese atrol enhances endothelium-dependent relaxation but does not alter
pregnant women. J Chromatogr A 1479:145–152 eNOS levels in aorta of spontaneously hypertensive rats. Exp Biol
37. Liu J, Gao Y, Liu H, Sun J, Liu Y, Wu J, Li D, Sun D (2017) Med (Maywood) 232(6):814–822
Assessment of relationship on excess arsenic intake from drinking 52. Samuel S, Kathirvel R, Jayavelu T, Chinnakkannu P (2005) Protein
water and cognitive impairment in adults and elders in arsenicosis oxidative dam-age in arsenic induced rat brain: influence of dl-
areas. Int J Hyg Environ Health 220(2 Pt B):424–430 alpha-lipoic acid. Toxicol Lett 155(1):27–34
38. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein 53. Sannadi S, Kadeyala PK, Gottipolu RR (2013) Reversal effect of
measurement with the Folin phenol reagent. J Biol Chem 193(1): monoisoamyl dimercaptosuccinic acid (MiADMSA) for arsenic
265–275 and lead induced perturbations in apoptosis and antioxidant en-
39. Lu TH, Tseng TJ, Su CC, Tang FC, Yen CC, Liu YY, Yang CY, Wu zymes in developing rat brain. Int J Dev Neurosci 31(7):586–597
CC, Chen KL, Hung DZ, Chen YW (2014) Arsenic induces reac- 54. Sarkar S, Mukherjee S, Chattopadhyay A, Bhattacharya S (2014) Low
tive oxygen species-caused neuronal cell apoptosis through JNK/ dose of arsenic trioxide triggers oxidative stress in zebrafish brain:
ERK-mediated mitochondria-dependent and GRP 78/CHOP- expression of antioxidant genes. Ecotoxicol Environ Saf 107:1–8
regulated pathways. Toxicol Lett 224(1):130–140 55. Shacka JJ, Roth KA (2005) Regulation of neuronal cell death and
40. Martínez L, Jiménez V, García-Sepúlveda C, Ceballos F, Delgado neurodegeneration by members of the Bcl-2 family: therapeutic
JM, Niño-Moreno P, Doniz L, Saavedra-Alanís V, Castillo CG, implications. Curr Drug Targets CNS Neurol Disord 4(1):25–39
Santoyo ME, González-Amaro R, Jiménez-Capdeville ME (2011) 56. Slomiany BL, Slomiany A (2013) Involvement of p38 MAPK-
Impact of early developmental arsenic exposure on promotor CpG- dependent activator protein (AP-1) activation in modulation of gas-
island methylation of genes involved in neuronal plasticity. tric mucosal inflammatory responses to Helicobacter pylori by
Neurochem Int 58(5):574–581 ghrelin. Inflammopharmacology 21(1):67–78
41. Moon KA, Navas-Acien A, Grau-Pérez M, Francesconi KA, 57. Song B, Zhou T, Liu J, Shao L (2016) Involvement of programmed
Goessler W, Guallar E, Umans JG, Best LG, Newman JD (2017) cell death in neurotoxicity of metallic nanoparticles: recent ad-
Low-moderate urine arsenic and biomarkers of thrombosis and vances and future perspectives. Nanoscale Res Lett 11(1):484
inflammation in the Strong Heart Study. PLoS One 12(8):e0182435 58. Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya
42. Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in YK, Chauhan LK, Dwivedi HN, Pant AB, Khanna VK (2014)
animal tissues by thiobarbituric acid reaction. Anal Biochem 95(2): Unraveling the mechanism of neuroprotection of curcumin in arse-
351–358 nic induced cholinergic dysfunctions in rats. Toxicol Appl
43. Pamplona R (2008) Membrane phospholipids, lipoxidative damage Pharmacol 279(3):428–440
and molecular integrity: a causal role in aging and longevity. 59. Szabo ST, Harry GJ, Hayden KM, Szabo DT, Birnbaum L (2016)
Biochim Biophys Acta 1777(10):1249–1262 Comparison of metal levels between postmortem brain and ventric-
44. Paul R, Choudhury A, Kumar S, Giri A, Sandhir R, Borah A (2017) ular fluid in Alzheimer’s disease and nondemented elderly controls.
Cholesterol contributes to dopamine-neuronal loss in MPTP mouse Toxicol Sci 150(2):292–300
model of Parkinson’s disease: involvement of mitochondrial dys- 60. Tyler CR, Allan AM (2014) The effects of arsenic exposure on
functions and oxidative stress. PLoS One 12(2):e0171285 neurological and cognitive dysfunction in human and rodent stud-
45. Pradelli LA, Bénéteau M, Ricci JE (2010) Mitochondrial control of ies: a review. Curr Environ Health Rep 1:132–147
caspase-dependent and -independent cell death. Cell Mol Life Sci 61. Wang X, Liu JZ, Hu JX, Wu H, Li YL, Chen HL, Bai H, Hai CX
67(10):1589–1597 (2011) ROS-activated p38 MAPK/ERK-Akt cascade plays a cen-
46. Prakash C, Kumar V (2016) Arsenic-induced mitochondrial oxida- tral role in palmitic acid-stimulated hepatocyte proliferation. Free
tive damage is mediated by decreased PGC-1α expression and its Radic Biol Med 51(2):539–551
downstream targets in rat brain. Chem Biol Interact 256:228–235 62. Wharton DC, Tzagoloff A (1967) Cytochrome oxidase from beef
47. Ram Kumar M, Flora SJ, Reddy GR (2013) Monoisoamyl 2,3- heart mitochondria. Methods Enzymol 10:245–250
dimercaptosuccinic acid attenuates arsenic induced toxicity: behav- 63. WHO (2011) Guideline for drinking-water quality, fourth edn.
ioral and neurochemical approach. Environ Toxicol Pharmacol World Health Organization, Geneva
36(1):231–242 64. Yadav RS, Chandravanshi LP, Shukla RK, Sankhwar ML, Ansari
48. Rios R, Zarazúa S, Santoyo ME, Sepúlveda-Saavedra J, Romero- RW, Shukla PK, Pant AB, Khanna VK (2011) Neuroprotective
Díaz V, Jiménez V, Pérez-Severiano F, Vidal-Cantú G, Delgado JM, efficacy of curcumin in arsenic induced cholinergic dysfunctions
Jiménez-Capdeville ME (2009) Decreased nitric oxide markers and in rats. Neurotoxicology 32(6):760–768
morphological changes in the brain of arsenic-exposed rats. 65. Yu Y, Guo Y, Zhang J, Xie J, Zhu Y, Yan J, Wang B, Li Z (2017) A
Toxicology 261(1–2):68–75 perspective of chronic low exposure of arsenic on non-working
49. Robert M, Friedlander MD (2003) Apoptosis and caspases in neu- women: risk of hypertension. Sci Total Environ 580:69–73
rodegenerative diseases. N Engl J Med 348:1365–1375 66. Zarazúa S, Ríos R, Delgado JM, Santoyo ME, Ortiz-Pérez D,
50. Rodríguez-Barranco M, Gil F, Hernández AF, Alguacil J, Lorca A, Jiménez-Capdeville ME (2010) Decreased arginine methylation
Mendoza R, Gómez I, Molina-Villalba I, González-Alzaga B, and myelin alterations in arsenic exposed rats. Neurotoxicology
Aguilar-Garduño C, Rohlman DS, Lacasaña M (2016) Postnatal 31(1):94–100

You might also like