You are on page 1of 16

Biological Trace Element Research

https://doi.org/10.1007/s12011-018-1452-5

Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic


and Dopaminergic System in Brain of Developing Rats
Lalit P. Chandravanshi 1,2 & Richa Gupta 2 & Rajendra K. Shukla 3

Received: 10 May 2018 / Accepted: 18 July 2018


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

Abstract
Chronic exposure to arsenic via drinking water throughout the globe is assumed to cause a developmental neurotoxicity. Here, we
investigated the effect of perinatal arsenic exposure on the neurobehavioral and neurochemical changes in the corpus striatum,
frontal cortex, and hippocampus that is critically involved in motor and cognition functions. In continuation of previous studies,
this study demonstrates that perinatal exposures (GD6–PD21) to arsenic (2 or 4 mg/kg body weight, p.o.) cause hypo-activity in
arsenic-exposed rats on PD22. The hypo-activity was found to be linked with a decrease in the mRNA and protein expression of
the DA-D2 receptor. Further, a protein expression of tyrosine hydroxylase (TH), levels of dopamine, and its metabolites were also
significantly impaired in corpus striatum. The arsenic-exposed groups showed spatial learning and memory significantly below
the average in a dose-dependent manner for the controls. Here, we evaluated the declined expression of CHRM2 receptor gene
and protein expression of ChAT, PKCβ-1 in the frontal cortex and hippocampus, which are critically involved in cognition
functions including learning and memory. A trend of recovery was found in the cholinergic and dopaminergic system of the brain,
but changes remained persisted even after the withdrawal of arsenic exposure on PD45. Taken together, our results indicate that
perinatal arsenic exposure appears to be critical and vulnerable as the development of cholinergic and dopaminergic system
continues during this period.

Keywords Developmental neurotoxicity . Arsenic . DA-D2 receptor . Spatial memory . Locomotor activity etc.

Introduction Argentina, Mexico, and Colombia, which have high arsenic


contamination in the drinking water [9, 14, 16, 26, 28, 33, 40].
Arsenic is one of the environmental neurotoxicant and ubiq- Growing evidence for animal and human studies indicated
uitous in nature due to its natural existence and anthropogenic that arsenic might have deleterious effects on the central ner-
sources [49]. As per an estimate, over 200 million people have vous systems [53, 89]. Furthermore, concern over develop-
been estimated to be chronically exposed to arsenic in drink- mental neurotoxicity of arsenic has been raised in the present
ing water throughout the globe [44, 84]. Higher incidence of scenario [25, 79]. Arsenic exposure could produce develop-
cancer, arsenicosis, skin lesions, cardiovascular diseases, re- mental abnormalities, including malformations, decreased
productive problems, and psychological, neurological, and growth rate, mortality, neuronal tube defects [46, 60, 68],
mental health problems is reported in many countries like and an array of behavioral changes through effects on the
India, Bangladesh, Japan, Argentina, Bolivia, Taiwan, Chile, developing brain directly since arsenic freely crosses the pla-
centa and blood–brain barrier (BBB) in humans and animals
[27, 31, 66, 67, 85]. Neural tube defects, spontaneous abor-
* Lalit P. Chandravanshi
tion, stillbirth, and neonatal deaths were reported in arsenic-
chandravanshi04@gmail.com exposed pregnant women who are consuming drinking water
contaminated with high arsenic levels [46, 48, 58, 59]. Low
1 levels of arsenic have been detected in the breast milk of
Division of Forensic Science, School of Basic and Applied Sciences,
Galgotias University, Greater Noida 201307, India Bangladeshi women and associated with adverse health ef-
2 fects on infants [20]. These findings suggest that arsenic
Developmental Toxicology Division, CSIR–Indian Institute of
Toxicology Research, Post Box No. 80, MG Marg, Lucknow 226 may cause to impaired fetal and infant health.
001, India Slow nerve conduction velocity, and sensory and motor
3
Department of Biochemistry, All India Institute of Medical Sciences, dysfunctions have also been reported in arsenic-exposed pop-
Bhopal, India ulation [55, 76]. Many investigators suggest that arsenic
Chandravanshi et al.

exposure to maternal and early life can affect the functioning The period GD6–PD21 is the developmental period in rats
of the central nervous system [70, 74]. Arsenic can alter the during which maturation of different neurotransmitter sys-
motor and cognitive functions by the dysfunctions of multiple tems, cholinergic, dopaminergic, and serotonergic receptors,
neurobiological processes including those of dopaminergic, BBB, the setting of neurons, and synaptogenesis occurs [38].
cholinergic, glutamatergic, and monoaminergic signaling However, the impact of arsenic exposure on the neurobehav-
pathways [10, 63, 87]. Arsenic can delay and inhibit the pro- ioral functions and its associated cholinergic and dopaminer-
cess of neurodevelopment of the central nervous system dur- gic system is not well understood during this perinatal period
ing the fetal and early life of the human [61]. Several scientists of brain development. Based on the previous findings [11, 12],
have investigated that arsenic could be modulating the dopa- this article generates a hypothesis by showing the need to
minergic system and associated behaviors. Dopaminergic address the cholinergic and dopaminergic dysfunctions along
neurons are important to usual dopaminergic neurotransmis- with neurobehavioral deficits in perinatally arsenic (2 or 4 mg/
sion signaling and involved in modulating reward, motivation, kg/p.o.) exposed rats on PD22. This study also assessed the
and motor activity functions by interacting with its receptors transient or persistent effect of arsenic even after 23 days of
[18, 69]. TH is a rate-limiting enzyme in the biosynthesis of withdrawal of arsenic exposure.
dopamine. Meanwhile, DA-D2 receptors in the striatum are
functionally involved in modulating the movement and cog-
nition [22]. Arsenic may significantly alter the levels of DA Materials and Methods
and their metabolites in the brain and thus affects the neuro-
chemical and behavioral abnormalities [1, 7, 30, 35, 47, 62]. Animal and Treatment
Muscarinic–cholinergic receptors play major roles in the
memory function, and evidence suggests a role in neurological Perinatal exposure of rats to sodium arsenite: Adult female
disorders like Alzheimer’s disease and schizophrenia [32, 50]. rats of proven fertility were kept for mating with the males
Acetylcholine coordinates the neuronal networks of the cho- in the ratio of 2:1. The day of appearance of the vaginal plug
linergic modulation area of the brain. Synthesis and function was considered as day 1 (GD-1) of gestation and the pregnant
of acetylcholine are regulated by the choline acetyltransferase rats were separated into individual cages. Pregnant rats were
(ChAT) and acetylcholinesterase (AChE) enzymes [80]. divided into three groups (six pregnant rats in each group) and
Cholinergic dysfunctions could be associated with the impair- treated with sodium arsenite (SIGMA, USA) on day 6 of
ment in motor coordination and learning, and could arise from gestation (GD-6) till postnatal day (PD) 21 as follows:
cholinergic dysfunctions. Reduced AChE and ChAT along
with the degeneration of cholinergic neurons have been re- Group I—Pregnant rats were treated with normal saline
ported in arsenic-exposed rats and provide support for arsenic (p.o.) identically as in groups II and III and served as
in the etiology of Alzheimer’s disease [52]. PKC β-1 is a controls.
molecule crucially involved in long-term potentiation and dif- Group II—Pregnant rats were treated with sodium arse-
ferent forms of learning [54, 86]. PKC activation induces pro- nite (2 mg/kg body weight, p.o.) dissolved in distilled
tein synthesis required for long-term memory. It appears intu- water once daily from GD6 to PD21.
itive that in some diseases, alterations in PKC β-1 cause or Group III—Pregnant rats were treated with sodium arse-
enhance the risk of learning and memory impairments, an idea nite dissolved in distilled water (4 mg/kg body weight,
that will need testing in arsenic-induced developmental neu- p.o.) once daily from GD6 to PD21.
rotoxicity. Further, deficits in cognition and motor function
following acute and chronic exposure to high dose of arsenic All pregnant rats were allowed to deliver naturally and rear
have been reported on adult animals [63, 87]. Mechanisms their pups to weaning until PD21. All the litters were culled on
underlying the neurotoxic effects of arsenic are not well un- PD2; 8–10 pups with an equal allocation of males and females.
derstood during a developing period of the brain. Most of the Cage-side observations were made to assess the adverse effects
researches on the neurobiological consequences of arsenic of arsenic if any of the pups [39]. Development of physical
exposure have focused largely on adult animals at high doses. milestones and reflexes were assessed following the standard
Generally, humans getting exposed to low level of arsenic protocol as described by Vorhees et al. [82]. Pup viability, av-
instead of high level is close to real-life exposure scenarios erage pup weight, and sex ratios were also observed from PD1
and is of greatest concern for arsenic exposure. The nervous to PD21. In this experiment, pups were exposed to arsenic
system will respond to the neurotoxic insult depending on the through the mother from GD6 (gestational exposure) to PD21
stage of brain development. Therefore, the timing of exposure (lactational exposure). On PD22, male rats were separated and
is very critical to the developmental neurotoxicity. Perinatal effect of arsenic on other behavioral endpoints studied on
period of rats is very crucial to the brain development as struc- PD22. The animals were sacrificed and the brain was taken
tural and functional maturation takes place during this period. out quickly, washed in ice-cold saline, and dissected into
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

specific regions—frontal cortex, corpus striatum, and hippo- Spontaneous Motor Activity
campus following the standard procedure as described by
Glowinski and Iversen [23]. The dissected brain regions were Spontaneous motor activity was evaluated by using auto-
stored at − 80 °C for neurochemical assays. Unfortunately, we mated Actimot system (TSE, Germany) following the
did not evaluate the effect of arsenic on female pups. To explore standard procedure as described by Chandravanshi et al.
whether these changes produced by arsenic were transient and [12]. A square-shaped frame was equipped with a high
persistent, another set of experiment was done on the same number and density of infrared beams (32 × 32) in this
conditions as mentioned above. Male pups were separated from apparatus. The activity monitor operates on light beam
mothers on PD22 in each treatment group and maintained as principle. Briefly, rats were acclimatized to half an hour
such till PD45. Neurobehavioral and neurochemical endpoints in behavioral room. The apparatus was cleaned and dried
were then assessed on PD45. with an ethanol solution (10%) before use every time.
Following this, each rat was individually placed in the
center of the cage (45 cm × 45 cm). As soon as the ani-
Behavioral Studies mals are placed in the cage, sensors become active and
counting device starts. Activity was monitored for 5 min
Developmental Landmarks and Reflexes and parameters—total distance traveled, resting time, time
moving, and stereotypical time—were recorded simulta-
The effects on the development of physical milestones and neously. Data collected by the system software were ana-
reflexes were studied in rat pups perinatally exposed to arsenic lyzed and the effect of different parameters assessed in
from GD6 to PD21. The test battery as described by Lazarini rats in the control and arsenic-exposed rats.
et al. [39] was followed to assess the criteria of development
of physical milestones and reflexes. Grip Strength
Physical developmental milestones, viz., eye-opening, pin-
na detachment, incisor eruption, and growth of fur, were ob- Forelimb grip strength was assessed in the control and arsenic-
served each day, beginning on PD1.The following reflex pa- exposed rats by using a computerized grip strength meter
rameters were assessed in the rats of control and arsenic- (TSE Germany) following the standard procedure as de-
exposed groups (2 or 4 mg/kg) as mixed sex: surface righting, scribed by Terry Jr et al. [73]. Holding the rat by the neck
negative geotaxis, and cliff avoidance. Separate pups (ten) and the base of the tail, the forelimbs were placed on the
were used for individual reflex parameters. tension bar. The rat was pulled back gently until it released
the bar. Reading was recorded automatically on the computer.
I. Surface righting reflex: The ability of rat pups to right Five successive pulls for each animal in the study group were
itself was determined on every alternate day from PD1 tried by a person unaware of their treatment status. The mean
through PD7. Rat pups were placed on their back on of all the values was taken and processed for statistical anal-
the clean body surface and the time required to turn ysis. The values are expressed in the pound.
over to ventral surface in normal upright position re-
corded. A criterion of successful righting within 2 s Spatial Memory—Continuous Alternation Test
was used. The day all rat pups per litter reached the
criterion was recorded [71]. Continuous alternation in a single session was measured using
II. Negative geotaxis reflex: Negative geotaxis was evaluat- a computerized Y-maze (TSE, Germany) to assess short-term
ed using the procedure described by Farag et al. [21]. The spatial memory [45, 64]. The equipment consisting of three
plane was made of wood and each test rat was given one arms (40 cm long × 15 cm wide × 30 cm high) converge in an
trial on PD5, PD10, and PD15. The trial was considered equilateral triangular central area with 15 cm at its longest
to be a success when the pup turned 180° within the axis. Rat in each case was placed at the end of one arm and
allotted time of 60 s. The time required for rats to reorient allowed to move freely through the maze for 5 min. The se-
toward the top when placed in a head-down position on a quence of entry in to each arm was recorded automatically.
board inclined to 25°, and the latency to rotate 180° was Spatial memory has been defined as the number and the se-
measured with the number of successful trials. quence of entries of rat in to the arm during 5 min. All the arms
III. Cliff avoidance reflex: Cliff avoidance was evaluated on look alike and no reinforcer, motivation, or punishment was
PD4 and pups were placed on a wooden platform elevat- used in this set of experiment. Further, the sequence of arm
ed 20 cm above a table top with the forepaws and nose movements was calculated and the percentage of alternation is
over the edge. The time required to return the body calculated as the number of alternations (entries into three
1.5 cm from the Bcliff^ was recorded. A successful re- different arms consecutively) divided by the total possible
sponse to rats within 30 s per trial was recorded [43]. alternations (the number of arms entered minus 2).
Chandravanshi et al.

Passive Avoidance Response blocking using blocking buffer (5% BSA). The membrane
was incubated overnight at 4 °C with antibodies DA-D2,
The passive avoidance response was assessed in rats to mon- TH, ChAT (CST), and PKC β-1 (Abcam) followed HRP-
itor the learning and memory deficits following the procedure conjugated secondary antibodies (Sigma, USA) (1:4000) and
as described by Tota et al. [75]. Briefly, the rat was placed in detected by ECL western blotting detection kit (Pierce, USA).
the lighted compartment of a shuttle box (Techno, India). A A digital gel image analysis system (ImageQuant LAS 500)
guillotine door isolates the compartment into light and dark was used for semi-quantification of immunoreactivity and
chamber. After an acclimatization period of 30 s, the guillotine normalized by β-actin.
door was opened. Soon after the entry of the rat into the dark Assay of acetylcholinesterase activity: Activity of acetyl-
compartment, the door was shut down and a low-intensity foot cholinesterase (AChE)was measured following the procedure
shock (0.5 mA for 10 s) was given. The duration of a trial was as described by Ellman et al. [19] using acetylthiocholine iodide
270 s. The first trial was acquisition. Retention tests were as a substrate and 5,5′-dithiobis-2 nitrobenzoic acid (DTNB) as
performed at various times (24, 48, and 72 h were considered the coloring agent. Briefly, the reaction mixture of a final volume
as first, second, and third retention trials respectively) after the of 1.0 ml contained phosphate buffers (0.1 M, pH 7.4), a post-
acquisition trial. The transfer latency time refers to the transfer mitochondrial fraction of frontal cortex or hippocampus (con-
of animal from one compartment to another which has been taining 15–20 μg protein), acetylthiocholine iodide (ACTI),
recorded in seconds. The criterion for learning was taken as an and DTNB (5 mM). The degradation of acetylthiocholine iodide
increase in the transfer latency time on retention trials as com- was measured at 412 nm, and the results are expressed as micro-
pared to the acquisition (first) trial. The shock was not given in moles ACTI hydrolyzed/milligram protein.
the retention trials to avoid reacquisition. Estimation of biogenic amines and their metabolites in
brain regions of rats: To determine the levels of DA (dopa-
mine), DOPAC (3, 4-dihydroxyphenylacetic acid), and HVA
Neurochemical Studies
(homovanillic acid) in the corpus striatum of rats, reversed-
phase high-performance liquid chromatography coupled with
mRNA expression DA-D2 and M2 receptor genes in brain
an electrochemical detector (HPLC–ECD) was carried out
regions of rats: mRNA levels of M2 (frontal cortex, hippo-
following the method described by Kim et al. [34] with minor
campus), D2 (corpus striatum) receptor gene in different brain
modifications. The HPLC system (Waters, Melford, USA)
regions of rats have been determined by RT-PCR (ABI, USA)
was integrated with a high-pressure isocratic pump (515
using the glyceraldehydes 3-phosphated dehydrogenase
HPLC Pump), a sample injector valve, C-18 reverse phase
(GAPDH) for normalization. The cDNA was amplified by
column (250 × 4 mm, particle size 5 μm), and an electrochem-
RT-PCR using ABI SYBER green qPCR Kit (Thermo
ical detector (464 Pulsed electrochemical detector). The brain
Scientific, USA) on the 7900HT Fast Real-Time PCR
tissue was weighed and transferred into the tube and the pre-
System running at 40 cycles following the protocol: one cycle
chilled homogenized buffer solution (0.1 M perchloric acid
of denaturation (95 °C, 10s), followed by 40 cycles of dena-
and 3,4-dihydroxy benzylamine, an internal standard at a final
turation (95 °C, 10s), annealing (60 °C, 10 s), and extension
concentration of 25 ng/ml) was added at a volume of 1 ml/
(72 °C, 45 s). A final extension (72 °C, 5 min) was performed.
100 mg sample. Brain tissues were homogenized and kept on
The primer sequences used for M2 [11], D2 [12], and GAPDH
ice for 15 min. The homogenate was centrifuged at maximal
[12] are mentioned below:
speed (36,000×g) at 4 °C for 10 min and the supernatant was
collected and transferred to a new sample tube. The superna-
DA-D2 FP 5′-AAGCGCCG RP 5′-GGCAATGATACACT
tant was filtered through nylon filters (0.22 μm; Millipore,
AGTTACTGTCAT-3′ CATTCTGGT-3′
CHRM2 FP 5′-GGCAAGCAAGA RP GCCAACAGGATAGCCA USA). Then 20-μl volumes of the sample were injected into
GTAGAATAAA-3′ AGATT-3′ a column with a mobile phase (pH 4.2) containing sodium
GAPDH FP 5′-CGGGAAGCTTGT RP 5′-CCAGCATCACCCC dihydrogen phosphate (0.15 M), ethylenediaminetetraacetic
GATCAATGG-3′ ATTTGA-3′ acid (0.25 mM), sodium octyl sulfate (1.75 mM), and 4%
methanol at a flow rate of 1.5 ml/min. The concentrations of
Immunoexpression of proteins by western blotting: As de- DA and its metabolites in corpus striatum of rats were deter-
scribed previously [13], frontal cortex hippocampus and cor- mined by using an electrochemical detector coupled with an
pus striatum tissues were homogenized in RIPA buffer and integrator. Chromatograms were analyzed with the help of
centrifuged at 12,000×g (15 min 4 °C) to remove the insoluble Empower2 software (Waters, Melford, USA) and the results
material. The supernatant was taken out carefully and the pel- were expressed in nanograms/gram tissue weight.
let discarded. The prepared samples contained 30 μg protein/ Estimation of arsenic in frontal cortex, hippocampus, and
lane and separated on 10% SDS-PAGE. The proteins were corpus striatum of rats: Arsenic levels were estimated in the
electroblotted onto nitrocellulose membranes followed by frontal cortex, hippocampus, and corpus striatum of rats
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

by using the hydride system 60 atomic absorption spec- Neurobehavioral Studies


trophotometer (HSAAS, ZEEnit 700) following the meth-
od of Ballentine and Burford [6]. Briefly, 0.1 g of tissue Effect on Development of Physical Milestones and Reflexes
was weighed and digested in 2-ml mixtures of concentrat-
ed nitric acid (1 ml) and perchloric acid (1 ml) in the Rat pups were observed once daily to examine their general
Kjeldahl flask. The sample was digested over a sand bath appearance, the physical condition, including moving activi-
until it becomes yellow in color. If the color of the digest ties and appearance of hair, nose, eyes, and limbs. There was
was brown, more nitric acid and the perchloric acid mix- no mortality in rat pups in any of the treatment groups.
tures were added and the process of oxidation was repeat- Further, there was no delay in the development of eye-open-
ed. The residue was made up to the known volume of HCl ing, pinna detachment, incisor eruption, and fur growth in
(3%) in deionized water and aliquots were used to esti- arsenic-exposed rat pups as compared to controls (data not
mate the arsenic levels using an atomic absorption spec- shown). Reflex parameters were tested to assess the effects
trophotometer. A calibration curve was constructed by of arsenic exposure on pups.
adding known amounts of arsenic standard (Sigma) to Surface righting: Following a neurotoxic dose of arsenic
calculate arsenic levels in brain regions, and data were (2 or 4 mg/kg p.o.) in rat pups, there was a delay in the
presented in nanograms/gram tissue weight. development of offspring righting reflex compared to con-
trol animals at PD7. Arsenic, however, did not affect the
righting ability of pups at a lower dose. The development
Protein Estimation of righting ability was significantly impaired by arsenic at
higher dose at PD7. The present study shows that surface
Protein content in the samples was measured following the righting remained increased on PD1, 3, 5, and 7 in the
procedure as described by Lowry et al. [41] using bovine arsenic-exposed group than controls (Fig. 2a).
serum albumin as a reference standard. Negative geotaxis effects: The results showed that arsenic
did not hamper the ability of pups to show negative geotaxis
Statistical Analysis reflex at PD5, 10, and 15 (Fig. 2b).
Cliff avoidance: The cliff avoidance response (F(2,27) =
Statistical analysis of the data has been carried out using 7.38, 23%, p < 0.05; 54%, p < 0.01) was found to be delayed
Graph Pad Prism software. The data have been analyzed using in arsenic-exposed rat pups as compared to control rat pups at
one-way analysis of variance (ANOVA) followed by both doses (Fig. 2c).
Newman–Keuls test to compare all pairs of columns. Values
are expressed as mean ± SEM and values up to p < 0.05 have Effect on Spontaneous Motor Activity
been considered significant.
Perinatal exposure to arsenic impaired the different parameters
of motor activity that was studied on PD22 and PD45, and
results are summarized in Table 1. A significant decrease in
Results the distance traveled (F(2,15) = 44.69, 37%, p < 0.001; 49%,
p < 0.001), time moving (F(2,15) = 41.98, 23%, p < 0.001;
Effect on Body and Brain Weight 42%, p < 0.001), and an increase in resting time (F(2,15) =
41.98, 36%, p < 0.001; 66%, p < 0.001) were observed in rats
Perinatal (GD6–PD21) exposure to arsenic (2.0 or 4.0 mg/ exposed to arsenic (2.0 or 4.0 mg/kg body weight) on PD22 as
kg body weight) caused a significant decrease in the body compared to controls (Table 1). At perinatal arsenic exposure,
weight (F(2,57) = 7.78, 21%, p < 0.05; 32%, p < 0.05) of we found increased stereotypic counts (F(2,15) = 15.17, 42%,
rats that was observed on PD22 as compared to controls p < 0.01; 72%, p < 0.001) in arsenic-exposed groups with re-
(Fig. 1a). The body weight remained decreased spect to the control group. Distance traveled (F(2,15) = 12.03,
(F(2,57) = 4.36, 8%, p > 0.05; 16%, p < 0.05) in arsenic- 17%, p < 0.05; 25%, p < 0.01), time moving (F(2,15) = 7.18,
exposed rats on withdrawal of arsenic exposure on PD45 17%, p < 0.05; 24%, p < 0.05), resting time (F(2,15) = 7.18,
as compared to respective controls (Fig. 1a). However, 18%, p < 0.05; 25%, p < 0.05), and stereotypic counts
changes in the body weight was not significant in rats (F(2,15) = 2.55, 21%, p > 0.05; 44%, p > 0.05) exhibit a trend
exposed to arsenic at a low dose (2.0 mg/kg body weight). of recovery, but the changes remained persistent on withdraw-
No significant change in the brain weight was observed in al of arsenic exposure in developing rats on PD45 as com-
rats perinatally (GD6–PD21) exposed to arsenic at any of pared to respective controls (Table 1). The locomotor activity
the doses (2.0 or 4.0 mg/kg body weight) on PD22 and results show that arsenic-exposed rats are characterized by a
PD45 as compared to respective controls (Fig. 1b). hypo-activity compared with control rats.
Chandravanshi et al.

Fig. 1 Body weight (a) and brain A – Body Weight


weight (b) of arsenic-exposed rats CONT As I As II
on PD22 or after the withdrawal
of arsenic exposure on PD45.
60
Values are the mean ± SEM of
five animals in each group for
brain weight and 20 animals for 50
body weight. Different from *

Body weight (g)


control group, *p < 0.05 and 40
**p < 0.01 were determined by
one-way ANOVA with Newman– 30
Keuls test
**
20 **

10

0
PD22 PD45

B – Brain Weight

1.6

1.2
Brain weight (g)

0.8

0.4

0
PD22 PD45

Effect on Grip Strength at both doses (2.0 or 4.0 mg/kg body weight) on PD22 as
compared to rats in the control group (Fig. 3b). Although
Effect of arsenic on the muscular strength of forelimb muscles a trend of recovery from the spatial memory was observed
was evaluated by grip holding the capacity of rats. A decrease (F(2,15) = 7.68, 14%, p < 0.05; 33%, p < 0.01) in arsenic-
in the forelimb grip strength (F(2,15) = 77.07, 30%, p < 0.01; exposed rats on withdrawal of arsenic exposure, a de-
40%, p < 0.01) was observed on PD22 in rats perinatally crease in spatial memory was found to persist in arsenic-
(GD6–PD21) exposed to arsenic (2.0 or 4.0 mg/kg body exposed rats on PD45.
weight) as compared to controls (Fig. 3a), while a trend of
recovery of the grip strength was observed in arsenic-
exposed rats on withdrawal of exposure. Decreased grip Effect on Learning and Memory—Passive Avoidance Test
strength was more prominent in rats exposed to arsenic at
higher dose (F(2,15) = 6.35, 17%, p < 0.05) on PD45 as com- Perinatally arsenic-exposed rats demonstrated impairment
pared to respective controls (Fig. 3a). in memory as evidenced by a reduction of transfer latency
time for the dark chamber. Perinatal arsenic exposure to
Effect on Spatial Memory—Continuous Alternation Test mothers at both doses was caused a significant impair-
ment in memory of pups on PD22. The decrease in the
Concerning spatial memory like behavior, arsenic admin- retention of memory was more pronounced in rats treated
istration reduced the percentage of alternation in arsenic- with arsenic at a higher dose. However, impairment in the
exposed groups as compared to control. A significant de- retention of memory remained persistent in rats exposed
crease in the spatial memory (F(2,15) = 24.24, 35%, p < to arsenic at a higher dose (4.0 mg/kg body weight) even
0.001; 54%, p < 0.001) tested by a Y-maze was observed on withdrawal of arsenic exposure on PD45 in compari-
in rats exposed to arsenic perinatally from GD6 to PD21 son to respective controls (Fig. 3c).
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

Fig. 2 Results of A - Surface Righting B - Negative Geotaxis


neurodevelopmental reflexes in
CONT As I As II
offspring. Surface righting reflex 12 CONT As I As II
60
(a) and negative geotaxis (b) were 10
** 50
monitored beginning on PD1 and

Time (s)
8 ***

Time (s)
PD5, respectively, cliff avoidance **
40
*
6 30
(c) was monitored beginning on
PND4. Values are the mean ± 4 20
SEM of ten animals in each 2 10
group. Different from control 0 0
group, *p < 0.05, **p < 0.01, and PD-1 PD-3 PD-5 PD-7 PD5 PD10 PD15
***p < 0.001 were determined by
one-way ANOVA with Newman–
Keuls test CONT As I As II
C - Cliff Avoidance
10 **
9
8 *

7
6

Time (s)
5
4
3
2
1
0
CONT As I As II
PD4

Neurochemical Studies gene were assessed in different brain regions. Exposure


to arsenic during the perinatal period caused a signifi-
Effects of Chronic Arsenic Exposure in DA-D2 and CHRM2 cant decrease in the mRNA expression of DA-D2 recep-
mRNA Expression tor gene (F(2,6) = 45.62, 44%, p < 0.001; 68%, p <
0.001) on PD22 as compared to controls (Fig. 4a).
In order to evaluate the effects of arsenic exposure on Further, the decrease in the mRNA expression of
receptor genes, mRNA levels DA-D2 and of CHRM2 DAR-D2 receptor gene (F(2,6) = 11.35, 29%, p < 0.05;

Table 1 Perinatal exposure


(GD6–PD21) to arsenic and effect Control Treatment groups
on different parameters of
spontaneous motor activity of rats As I As II
on PD22 and PD45 (2 mg/kg) (4 mg/kg)

Postnatal day 22
Total distance traveled (cm) 2370 ± 145.3 1487 ± 54.93*** 1207 ± 24.72***
Resting time (s) 116.8 ± 5.09 197.8 ± 8.57*** 194.8 ± 6.74***
Stereotypic count (s) 27.17 ± 1.72 38.83 ± 2.99** 46.83 ± 2.72***
Time moving (s) 183.2 ± 5.09 140.2 ± 6.13*** 105.2 ± 6.74***
Postnatal day 45
Total distance (cm) 2123 ± 93.83 1737 ± 88.86** 1512 ± 86.73***
Resting time (s) 44.7 ± 13.38 175.3 ± 5.50* 192.5 ± 5.99**
Stereotypic count (s) 31.5 ± 2.24 37.83 ± 2.34 41.33 ± 4.21
Time moving (s) 155.3 ± 13.38 124.7 ± 5.50* 107.5 ± 5.99**

Different parameters of spontaneous motor activity studied of arsenic-exposed rats on PD22 or after the with-
drawal of arsenic exposure on PD45. Values are the mean ± SEM of six animals in each group. Different from
control group, *p < 0.05, **p < 0.01, and ***p < 0.001 were determined by one-way ANOVA with Newman–
Keuls test
Chandravanshi et al.

Fig. 3 Neurobehavioral CONT As I As II


dysfunctions observed at PD22
(after the exposure) and PD45 A - Grip Strength B - Y maze - Continuous Alternation
(withdrawal of exposure) of age 400
of rats. a The forelimb grip 60
350
strength measured by grip 50
300 *

% of Alternation
strength meter. Percentage of
continuous alternation was 250 40 **
***

Pound (w)
assessed by Y-maze system (b).
200
Passive avoidance response in the 30
** ***
retention tests were performed at 150 ** 20
various times (24, 48, and 72 h, 100
respectively) after the acquisition 10
50
trial in arsenic-exposed groups
(c). Values are the mean ± SEM of 0 0
CONT As I As II CONT As I As II CONT As I As II CONT As I As II
five to six animals in each group.
Different from control group, PD22 PD45 PD22 PD45
*p < 0.05, **p < 0.01, and
***p < 0.001 were determined by C - Passive Avoidance Response
one-way ANOVA with Newman–
Keuls test Acquisition Retention 1 Retention 2 Retention 3
300 *
300
*
250 * * * *
250
*
Transfer Latency Time

Transfer Latency Time


200 200 * *

150 150

100 100

50 50

0 0
CONT As I As II CONT As I As II
PD22 PD45

44%, p < 0.01) was found to persist in arsenic-exposed Effect on the Protein Expression of TH, DA-D2 Receptor
rats even on withdrawal of arsenic exposure on PD45. (Corpus Striatum), ChAT, AChE, and PKCβ-1 (Frontal Cortex
However, the decrease in the mRNA expression was not and Hippocampus)
restored in arsenic-exposed rats.
To further explore the mechanism by which arsenic impairs
the cholinergic and dopaminergic system, we evaluated the
Effect on Muscarinic–Cholinergic Receptors and CHRM2 protein expression of the DA-D2 receptor, TH, ChAT, and
Receptor Gene in Frontal Cortex and Hippocampus PKCβ-1 by western blotting.
DA-D2 receptor and TH protein expression levels in the
Arsenic (2.0 or 4.0 mg/kg body weight) exposure during corpus striatum were analyzed. As observed in Fig. 5, expres-
perinatal from GD6 to PD21 caused a significant de- sion of DA-D2 receptor (F(2,6) = 11.87, 1.33-fold, p < 0.05;
crease in the expression of CHRM2 receptor genes in 1.63-fold, p < 0.01) and TH (F(2,6) = 32.98, 1.43-fold, p <
the frontal cortex (F(2,6) = 10.62, 38%, p < 0.01; 55%, 0.01; 2.03-fold, p < 0.001) proteins in the corpus striatum in
p < 0.001) and hippocampus (F(2,6) = 22.97, 39%, p < both arsenic-exposed groups were significantly lower than in
0.001; 59%, p < 0.001) of rats on PD22 as compared the control group on PD22. Moreover, there was no significant
to controls (Fig. 4b). While a trend of recovery was change observed in the expression of DA-D2 receptor protein
found in the mRNA expression of CHRM2 receptor at the lower dose of arsenic after the withdrawal of exposure as
genes both in frontal cortex and hippocampus, the compared to controls (Fig. 5a). Down-regulation of the DA-
changes remained significantly decreased in rats exposed D2 receptor was significantly continued at a higher dose of
to arsenic at a higher dose PD45 in comparison to con- arsenic (F(2,6) = 27.23, 1.78-fold, p < 0.01) on PD45. The
trols (Fig. 4b). expression of TH (F(2,6) = 26.79, 1.30-fold, p < 0.01; 1.51-
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

Fig. 4 Relative mRNA levels of A


DA-D2 receptor gene in the 1.2 CONT As I As II
corpus striatum (a) and CHRM2
receptor gene in frontal cortex and 1
hippocampus (b) of arsenic-

DA-D2 mRNA Levels


Relative to Controls
exposed rats on PD22 or after the 0.8 *
withdrawal of arsenic exposure
on PD45. Values are the mean ± 0.6 *** **
SEM of three animals in each
group. Different from control 0.4 ***
group, *p < 0.05, **p < 0.01, and
***p < 0.001 were determined by 0.2
one-way ANOVA with Newman–
Keuls test 0
PD22 PD45
Corpus Striatum
B
1.2
CHRM2 mRNA Levels Relative to Controls
1

0.8 **
**
** *** **
0.6 ***
***
0.4

0.2

0 PD22 PD45 PD22 PD45


Frontal Cortex Hippocampus

fold, p < 0.001) in corpus striatum remained significantly de- Densitometric analysis revealed that perinatal exposure to ar-
creased on withdrawal of arsenic exposure on PD45 as com- senic was decreased the expression of ChAT, an enzyme in-
pared to controls (Fig. 5b). volved in the synthesis of acetylcholine, a neurotransmitter in
Compared with the control, expression of PKC β-1 was frontal cortex (F(2,6) = 14.51, 1.26-fold, p < 0.05; 1.50-fold,
reduced by F(2,6) = 32.84, 1.24-fold, 1.60-fold, in the hippo- p < 0.01) and hippocampus (F(2,6) = 16.01, 1.22-fold, p <
campus of rats in groups I and II, respectively, on PD22. 0.05; 1.56-fold, p < 0.01) on PD22 in comparison to controls
However, rats exposed to arsenic at a low dose exhibited a (Fig. 6b). In arsenic-exposed rats at the lower dose, expression
trend of recovery in the expression of PKC β-1 in hippocam- of ChAT was found to recover while the expression remained
pus on PD45 as compared to respective controls (Fig. 6a). significantly decreased in rats exposed to arsenic at a higher

Fig. 5 Relative protein A B


expression levels of DA-D2 51 kda TH 40 kda
DA-D2
receptor (a) and tyrosine
β-actin 42 kda β-acn 42 kda
hydroxylase (b) in the corpus
striatum of arsenic-exposed rats
on PD22 or after the withdrawal CONT As I As II
of arsenic exposure on PD45.
Each panel shows a representative 1.2
Western blot (top) and 1.2
1
densitometric analysis of bands in
the control and As-exposed *
0.8
Fold Change

groups. Values are the mean ± **


Fold Change

** 0.8 **
SEM of three animals in each ** ***
0.6
group. Different from control
***
group, *p < 0.05, **p < 0.01, and
0.4
***p < 0.001 were determined by 0.4
one-way ANOVA with Newman–
0.2
Keuls test
0 0
PD22 PD45 PD22 PD45
Corpus Striatum Corpus Striatum
Chandravanshi et al.

Fig. 6 Relative protein


expression levels of PKC β-1 in A PKC β-1 77 kda
the hippocampus (a) and ChAT in β-acn 42 kda
the frontal cortex and
hippocampus (b) of arsenic- 1.2 CONT As I As II
exposed rats on PD22 or after the
1
withdrawal of arsenic exposure
on PD45. Each panel shows a ** *
0.8
representative western blot (top)

Fold Change
***
and densitometric analysis of 0.6
bands in the control and As-
exposed groups. Values are the 0.4

mean ± SEM of three animals in


0.2
each group. Different from
control group, *p < 0.05, **p < 0
0.01, and ***p < 0.001 were PD22 PD45
determined by one-way ANOVA Hippocampus
with Newman–Keuls test B
ChAT 68 kda
β-actin 42 kda
1.2

*
*
0.8 * **
**
Fold Change

**
0.6

0.4

0.2

0
PD22 PD45 PD22 PD45
Frontal Cortex Hippocampus

dose (4.0 mg/kg body weight) as compared to respective con- the acetylcholinesterase (AChE) activity was more
trols on PD45 (Fig. 6b). marked in rats exposed to arsenic at a higher dose
(4.0 mg/kg body weight).

Effect on Brain Acetylcholinesterase Activity in Frontal Cortex


and Hippocampus Effect on Dopamine and their Metabolites in Corpus Striatum

Acetylcholinesterase, an enzyme involved in the hydroly- The levels of DA and their metabolites showed dose-
sis of acetylcholine, a neurotransmitter in the cholinergic dependent change in the corpus striatum of arsenic-
neurons that modulate the behavior, was studied in frontal exposed rats as compared to controls. A significant decrease
cortex and hippocampus of perinatally arsenic-exposed in the levels of striatal DA (F(2,9) = 32.79, 35%, p < 0.01;
rats. A significant decrease in the activity of acetylcholin- 60%, p < 0.001) and an increase in the levels of DOPAC
esterase (AChE) was observed both in the frontal cortex (F(2,9) = 43.78, 39%, p < 0.001; 53%, p < 0.001) and HVA
(F(2,12) = 10.03, 31%, p < 0.01; 48%, p < 0.001) and hip- (F(2,9) = 18.76, 31%, p < 0.05; 64%, p < 0.001) were ob-
pocampus (F(2,12) = 8.97, 34%, p < 0.001; 45%, p < served in the corpus striatum of perinatally arsenic-exposed
0.001) of rats perinatally exposed to arsenic (2.0 or rats as compared to controls on PD22. Both doses of arsenic
4.0 mg/kg body weight) on PD22 as compared to controls had persistent effects as a significant decrease in the levels of
(Fig. 7). The decrease in the activity of acetylcholinester- DA (F(2,11) = 6.33, 28%, p < 0.05; 41%, p < 0.05) was ob-
ase (AChE) was found to persist both in the frontal cortex served even after withdrawal of arsenic exposure. Higher
(F(2,12) = 4.20, 19%, p > 0.05; 25%, p < 0.01) and hippo- levels of DOPAC (F(2,11) = 8.84, 25%, p < 0.05; 36%, p <
campus (F(2,12) = 4.52, 13%, p > 0.05; 21%, p < 0.01) 0.01) and HVA (F(2,11) = 8.22, 16%, p > 0.05; 32%, p <
even on withdrawal of arsenic exposure on PD45 as com- 0.01) were found to persist on PD45 in comparison to respec-
pared to respective controls. The intensity of changes in tive controls (Fig. 8).
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

Fig. 7 Acetylcholinesterase CONT As I As II


10
activity in the frontal cortex and

μ mole Hydrolyzed / min / mg Protein


hippocampus of arsenic-exposed
rats on PD22 or after the with- 8
drawal of arsenic exposure on
*** **
PD45. Values are the mean ±
*** **
SEM of five animals in each 6
**
group. Different from control
group, **p < 0.01 and ***p < ***
0.001 were determined by one-
4
way ANOVA with Newman–
Keuls test 2

0 PD22 PD45 PD22 PD45


Frontal Cortex Hippocampus

Effects on Arsenic Levels in Brain Regions and varying period of exposure [5, 37, 89]. Still, there is
no study on the effect of perinatal arsenic exposure to cho-
The total arsenic levels in frontal cortex, corpus striatum, linergic and dopaminergic dysfunctions along with
and hippocampus of rats were measured on PD22 and neurodevelopmental consequences of developing brain of
PD45, and the results are presented in Table 2. A signif- rats. The previous studies have demonstrated that arsenic
icant increase in the levels of arsenic in the frontal cor- exposure might lead to cholinergic and dopaminergic dys-
tex, corpus striatum, and hippocampus was observed in functions of early life exposed rats at the same doses of the
perinatally arsenic-exposed rats on PD22 as compared to present study. The present study has succeeded in
controls (Table 2). However, the decrease in arsenic unveiling a mechanism of developmental neurotoxicity
levels was observed on withdrawal of arsenic exposure linking the arsenic levels in different brain regions of rats
in the frontal cortex, corpus striatum, and hippocampus and also assessing such changes are transient or persistent.
on PD45 as compared to rats exposed to arsenic imme- The reduction of body weight following perinatal exposure
diately on PD22. However, the increase in the arsenic to arsenic reported in the present study indicated that im-
levels remained persistent in selected brain regions at pairment of growth and general health status of rats is con-
both the doses of arsenic in comparison to respective sistent with earlier experimental reports [8, 15]. Decreased
controls on PD45 (Table 2). pattern of body weight remained to persist with a higher
dose of arsenic even after withdrawal of arsenic exposure,
suggesting that the effect is persisting and perinatal arsenic
Discussion exposure may have detrimental health effects.
It is found that arsenic exposure might cause to modu-
Many laboratories have reported that exposure to arsenic late the cognitive and motor functions even at low doses,
during the developing period of the brain in experimental and the changes might be associated with cholinergic and
animals induces brain changes and promotes neurotoxicity. dopaminergic adverse outcomes. With the result of the
These reports have explained the mechanism of arsenic- present study, we found the noticeable link between de-
induced neurotoxicity in animals at different ages, dose, velopmental arsenic exposure and motor or cognitive

Fig. 8 Relative dopamine and CONT As I As II


their metabolites (HVA, DOPAC) 7000
content in the corpus striatum of
arsenic-exposed rats on PD22 or 6000
ng/gm Tissue Weight

after the withdrawal of arsenic 5000


exposure on PD45. Values are the ** *
mean ± SEM of four to five 4000 ***
* **
animals in each group. Different 3000
from control group, *p < 0.05, *** *
***
**p < 0.01, and ***p < 0.001 2000
were determined by one-way 1000 * ***
ANOVA with Newman–Keuls **
test 0
PD22 PD45 PD22 PD45 PD22 PD45
DA HVA DOPAC
Corpus Striatum
Chandravanshi et al.

Table 2 Perinatal exposure


(GD6–PD21) to arsenic and effect Brain regions/treatment groups Postnatal day 22 Postnatal day 45
on arsenic levels in different brain
regions of rats Frontal cortex
Control 2.90 ± 0.54 2.07 ± 0.81
As I (2 mg/kg) 91.02 ± 9.41* 42.97 ± 17
As II (4 mg/kg) 334.20 ± 59.50*** 206 ± 29.38***
Hippocampus
Control 1.12 ± 0.24 0.82 ± 0.21
As I (2 mg/kg) 94.50 ± 22.23** 36.27 ± 8.23*
As II (4 mg/kg) 172.80 ± 24.92*** 109.60 ± 16.64***
Corpus striatum
Control 4.82 ± 0.46 8.65 ± 0.72
As I (2 mg/kg) 416.50 ± 22.95*** 284.10 ± 40.77***
As II (4 mg/kg) 683.30 ± 12.80*** 442 ± 45.77***

Arsenic levels expressed as ng/g tissue weight. Values are the mean ± SEM of five animals in each group.
Different from control group, *p < 0.05, **p < 0.01, and ***p < 0.001 were determined by one-way ANOVA
with Newman–Keuls test

deficits in rats. Adverse health effects of arsenic on the dopamine [7]. Motor and cognitive functions and different
developing fetus and children are of particular attention to type receptor signaling are affected in rodents by the arsenic
the society and regulatory agencies. The effects of perina- exposure [35, 42, 72]. Therefore, we find out whether arsenic
tal arsenic exposure on neurodevelopment consequence, exposure alters expression of TH protein in the corpus stria-
motor and cognitive function, including learning and tum. Expression TH protein in the corpus striatum was signif-
memory, in rats were evaluated. The timing of exposure icantly decreased in both arsenic exposure groups. In addition,
has no importance to the nature and the degree of the DA-D2 receptors in the striatum have a distinct role in the
observed neurotoxicity in an adult. In contrast, the devel- regulation of motor function including locomotor activity.
oping brain will respond to a similar neurotoxic insult to a To better understand the mechanism of underlying arsenic-
different susceptibility stage of brain development. The induced developmental neurotoxicity, we further observed
high sensitivity of developing rats compared with the the declined protein expression of DA-D2 receptors as well
adults to the neurotoxicity of arsenic is most probably as mRNA level. Decrease striatal DA level was found in
due to the immaturity of detoxifying enzymes. Arsenic arsenic-exposed rats in a dose-dependent manner, while in-
may also interfere the maturation of BBB and receptors creased DOPAC and HVA levels were found in the corpus
of the brain during development, causing permanent dys- striatum of rats treated with arsenic suggesting the dopaminer-
functions [29, 57] that result in behavioral changes. gic system as a target of arsenic exposure. DOPAC and HVA
Growth delays and neural tube defects by the arsenic ex- are the main neuronal metabolites of dopamine; higher levels
posure in human have been reported [2, 78], and the neuro- of DOPAC and HVA contents are consistent with greater loss
logical and cognitive deficits in children were also observed of DA. These findings indicate that decreased dopamine level
[36, 65, 81]. Negative geotaxis can assess the sensory input, may be related to the declined expression of TH in arsenic-
while cliff avoidance and surface righting assess motor func- exposed rats at these low concentrations of arsenic during the
tion of rats. In the present study, developmental exposure to developmental period of the brain. These changes in the se-
arsenic impaired the negative geotaxis, cliff avoidance, and lected endpoints of the dopaminergic system due to the accu-
surface righting ability of neonates. Corpus striatum, frontal mulation of arsenic in the corpus striatum might be related to
cortex, and hippocampus are believed to be involved in the the behavioral or functional consequences. These findings
neurobehavioral reflexes [4, 83]. Further, neurochemical agree with the previously published reports, where transmis-
changes in the brain can be predicted by impairment of these sion of dopaminergic neurons and its associated behaviors
reflexes and provide the basic mechanisms of brain damage were vulnerable to arsenic, which induced neurobehavioral
underlying the arsenic exposure. toxicity [88]. Hyperactivity and hypo-activity have also been
It is well known that dopamine regulates the motor activity reported in arsenic-exposed rats, and these behaviors seemed
and cognitive functions of rats and closely associated with to be dependent on dose, route of exposure, and the period of
neurotransmission process. Levels of striatal dopamine are exposure to arsenic [12, 51].
necessary for the physiological function of the dopaminergic Arsenic exposure has also been found to inhibit AChE
system. TH is a rate-limiting enzyme for the synthesis of activity and ChAT expression in the brain of rats [17, 87],
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

and it has also been reported that there is an inverse link could be associated with the cholinergic and dopaminer-
between arsenic exposure and plasma cholinesterase activity gic dysfunctions. We investigated a mechanism involved
in a human [3]. As most of these studies have been designed at in arsenic-induced developmental neurotoxicity. These in-
high doses of arsenic and in adult animals, the impact of low clude modification in dopaminergic and cholinergic sig-
doses of arsenic on the integrity of brain AChE activity during naling, and behavioral deficits, including locomotion
the perinatal period of brain development is unexplored. learning and memory. This rodent model that may even-
Further, declined frontocortical and hippocampal AChE activ- tually be useful for understanding arsenic-induced neuro-
ity, ChAT protein expression, as well as CHRM2 gene expres- logical deficits in children during the duration of perinatal
sion were observed in a dose-dependent manner. Inhibition in exposure. Dysfunction of both dopaminergic and cholin-
the activity of acetylcholinesterase is a result of arsenic depo- ergic systems has been alarmed in a different type of
sition in the brain and is also associated with enhanced accu- neurological disorder characterized by motor and cogni-
mulation of acetylcholine in the synaptic cleft and subsequent- tive deficits including learning and memory. In the present
ly downregulation of cholinergic receptors both in the frontal study, functional deficits in spatial learning, memory, and
cortex and hippocampus. Accumulation of acetylcholine at motor activity following arsenic exposure at high and low
the synaptic cleft and arsenic in the frontal cortex and hippo- doses made it possible to significantly cause dopaminer-
campus of rats indicated vulnerability of the cholinergic sys- gic and cholinergic dysfunctions in perinatally exposed
tem to perinatal arsenic exposure. PKCβ-1 also plays a sig- rats. A trend of recovery was observed in selected behav-
nificant role in the long-term potentiation and different pat- ioral and neurochemical endpoints after 23 days of with-
terns of learning and memory [54, 86]. It can also be hypoth- drawal of arsenic exposure; however, most of these
esized that decreased expression of PKCβ-1 in the hippocam- changes remained persistent. It is further suggested that
pus was associated with impaired learning and memory in the intensity of these changes could be more harmful in
arsenic-exposed rats in comparison to controls as observed case exposure to arsenic occurs during the developmental
in the present study. period of the brain for the reason that development of
With these experiments, we also sought to define the cho- dopaminergic and cholinergic receptors in the brain con-
linergic dysfunction patterns of the frontal cortex and hippo- tinues from perinatal to early life periods of rats.
campus of developing rats as well as effects on learning and In conclusion, our findings indicate that perinatal arsenic
memory induced by arsenic at low doses. Arsenic may affect exposure leads to delay and disturbance of neurodevelopment.
the integrity of muscarinic–cholinergic receptors and inhibit Taking into account the possibility that arsenic might inhibit
intracellular signaling [56, 77]. They also defined the cogni- the normal development of dopaminergic and cholinergic neu-
tion and behavioral dysfunctions may occur due to the distur- rons in the developing brain when exposed during the perina-
bances of the cholinergic system of the central nervous sys- tal period, developmental neurotoxicity cannot be excluded.
tem. The decline in learning and memory caused by the rela- Further, developmental exposure to arsenic may be a risk fac-
tively low doses of arsenic used here was accompanied by tor for increased vulnerability to neurological disorders, and
distinct patterns of selected endpoints of the cholinergic sys- studies are needed in order to progress the urgently needed
tem. In our study, decrease in learning and memory is possible risk assessment. In this light, our findings are relevant: they
due to the significant severity of cholinergic dysfunctions. suggest that dopaminergic and cholinergic dysfunctions with
Furthermore, subjects with a decreased grip strength were neurobehavioral deficits represent key issues that may be as-
more likely to have a muscle weakness. This could be related sociated with accumulation of arsenic in different brain re-
to the cholinergic dysfunctions in the frontal cortex and hip- gions. These findings put forth a new perspective on how
pocampus of rats. Impairment in learning and memory clearly environmental arsenic exposure affects the pathogenesis of
suggested that even low doses of arsenic will injure the frontal neurodevelopmental disorders in children. The underlying
cortex and hippocampus of developing rats. Cognitive impair- mechanisms of these shared changes may provide an avenue
ments in humans assessed after arsenic exposures have been for novel treatments and address preventive strategies for
associated with Alzheimer’s disease and established the link arsenic-induced neurodevelopment disorders in children.
between arsenic and Alzheimer’s disease [24]. However, peri-
natal arsenic exposure is thought to inhibit the cholinergic Acknowledgements: The authors thank the Director of the CSIR–Indian
system in the frontal cortex and hippocampus, along with Institute of Toxicology Research (CSIR-IITR), Lucknow for his support
and keen interest in the present study. Financial support by the University
cognitive deficits of developing rats at these low doses. It is Grants Commission, New Delhi for carrying out the study is gratefully
well established that arsenic crosses the placenta and blood– acknowledged.
brain barrier; therefore, exposure may occur from the begin-
ning of life and accumulate in the different brain regions. An Compliance with Ethical Standards
increase of arsenic levels found in the corpus striatum, frontal
cortex, and hippocampus of perinatal arsenic-exposed rats Conflict of Interest The authors state no conflict of interest.
Chandravanshi et al.

References 18. Eisenegger C, Naef M, Linssen A, Clark L, Gandamaneni PK,


Müller U, Robbins TW (2014) Role of dopamine D2 receptors in
human reinforcement learning. Neuropsychopharmacology 39:
1. Agrawal S, Bhatnagar P, Flora SJ (2015) Changes in tissue oxida-
2366–2375
tive stress, brain biogenic amines and acetylcholinesterase follow-
19. Ellman GL, Courtney KD, Andres VJ, Feather-Stone RM (1961) A
ing co-exposure to lead, arsenic and mercury in rats. Food Chem
new and rapid calorimetric determination of acetylcholinesterase
Toxicol 86:208–216
activity. Biochem Pharmacol 7:88–95
2. Ahmed S, Mahabbat-e Khoda S, Rekha RS, Gardner RM, Ameer 20. Fängström B, Moore S, Nermell B, Kuenstl L, Goessler W, Grandér
SS, Moore S, Ekström EC, Vahter M, Raqib R (2011) Arsenic- M, Kabir I, Palm B, Arifeen SE, Vahter M (2008) Breast-feeding
associated oxidative stress, inflammation, and immune disruption protects against arsenic exposure in Bangladeshi infants. Environ
in human placenta and cord blood. Environ Health Perspect 119: Health Perspect 116:963–969
258–264 21. Farag AT, Goda NF, Shaaban NA, Mansee AH (2007) Effects of
3. Ali N, Hoque MA, Haque A, Salam KA, Karim MR, Rahman A, oral exposure of synthetic pyrethroid, cypermethrin on the behavior
Islam K, Saud ZA, Khalek MA, Akhand AA, Hossain M, Mandal of F1-progeny in mice. Reprod Toxicol 23:560–567
A, Karim MR, Miyataka H, Himeno S, Hossain K (2010) 22. Girault JA, Greengard P (2004) The neurobiology of dopamine
Association between arsenic exposure and plasma cholinesterase signaling. Arch Neurol 61:641–644
activity: a population based study in Bangladesh. Environ Health 23. Glowinski J, Iversen LL (1966) Regional studies of catecholamines
10:9–36 in the rat brain. I. The disposition of [3H]norepinephrine,
4. Altman J, Sudarshan K (1975) Postnatal development of locomo- [3H]dopamine and [3H]dopa in various regions of the brain. J
tion in the laboratory rat. Anim Behav 23:896–920 Neurochem 13:655–659
5. Aung KH, Kyi-Tha-Thu C, Sano K, Nakamura K, Tanoue A, 24. Gong G, O'Bryant SE (2010) The arsenic exposure hypothesis for
Nohara K, Kakeyama M, Tohyama C, Tsukahara S, Maekawa F Alzheimer disease. Alzheimer Dis Assoc Disord 24:311–316
(2016) Prenatal exposure to arsenic impairs behavioral flexibility 25. Gonzalez-Cortes T, Recio-Vega R, Lantz RC, Chau BT (2017)
and cortical structure in mice. Front Neurosci 10:137 DNA methylation of extracellular matrix remodeling genes in chil-
6. Ballentine R, Burford DD (1957) Determination of metals. dren exposed to arsenic. Toxicol Appl Pharmacol 329:140–147
Methods Enzymol 3:1002–1035 26. González-Horta C, Ballinas-Casarrubias L, Sánchez-Ramírez B,
7. Bardullas U, Limón-Pacheco JH, Giordano M, Carrizales L, Ishida MC, Barrera-Hernández A, Gutiérrez-Torres D, Zacarias
Mendoza-Trejo MS, Rodríguez VM (2009) Chronic low-level ar- OL, Saunders RJ, Drobná Z, Mendez MA, García-Vargas G,
senic exposure causes gender-specific alterations in locomotor ac- Loomis D, Stýblo M, Del Razo LM (2015) A concurrent exposure
tivity, dopaminergic systems, and thioredoxin expression in mice. to arsenic and fluoride from drinking water in Chihuahua, Mexico.
Toxicol Appl Pharmacol 239:169–177 Int J Environ Res Public Health 12:4587–4601
8. Bashir S, Sharma Y, Irshad M, Nag TC, Tiwari M, Kabra M, Dogra 27. Hirner AV, Rettenmeier AW (2010) Methylated metal(loid) species
TD (2006) Arsenic induced apoptosis in rat liver following repeated in humans. Met Ions Life Sci 7:465-521
60 days exposure. Toxicology 217:63–70 28. Hughes MF, Beck BD, Chen Y, Lewis AS, Thomas DJ (2011)
9. Brinkel J, Khan MH, Kraemer A (2009) Systematic review of ar- Arsenic exposure and toxicology: a historical perspective. Toxicol
senic exposure and its social and mental health effects with special Sci 123:305–332
reference to Bangladesh. Int J Environ Res Public Health 6:1609– 29. Ishii K, Itoh Y, Iwasaki N, Shibata Y, Tamaoka A (2014) Detection
1619 of diphenylarsinic acid and its derivatives in human serum and
10. Caldwell KE, Labrecque MT, Solomon BR, Ali A, Allan AM cerebrospinal fluid. Clin Chim Acta 431:227–231
(2015) Prenatal arsenic exposure alters the programming of the 30. Itoh T, Zhang YF, Murai S, Saito H, Nagahama H, Miyate H,
glucocorticoid signaling system during embryonic development. Saito Y, Abe E (1990) The effect of arsenic trioxide on brain
Neurotoxicol Teratol 47:66–79 monoamine metabolism and locomotor activity of mice.
Toxicol Lett 54:345–353
11. Chandravanshi LP, Yadav RS, Shukla RK, Singh A, Sultana S, Pant
31. Jin Y, Xi S, Li X, Lu C, Li G, Xu Y, Qu C, Niu Y, Sun G (2006)
AB, Parmar D, Khanna VK (2014a) Reversibility of changes in
Arsenic speciation transported through the placenta from mother
brain cholinergic receptors and acetylcholinesterase activity in rats
mice to their newborn pups. Environ Res 101:349–355
following early life arsenic exposure. Int J Dev Neurosci 34:60–75
32. Kamkwalala AR, Newhouse PA (2017) Beyond acetylcholinester-
12. Chandravanshi LP, Shukla RK, Sultana S, Pant AB, Khanna VK
ase inhibitors: novel cholinergic treatments for Alzheimer's disease.
(2014b) Early life arsenic exposure and brain dopaminergic alter-
Curr Alzheimer Res 14:377–392
ations in rats. Int J Dev Neurosci 38:91–104
33. Kapaj S, Peterson H, Liber K, Bhattacharya P (2006) Human health
13. Chandravanshi LP, Gupta R, Shukla RK (2018 Mar 3) effects from chronic arsenic poisoning—a review. J Environ Sci
Developmental neurotoxicity of arsenic: involvement of oxidative Health A Tox Hazard Subst Environ Eng 41:2399–2428
stress and mitochondrial functions. Biol Trace Elem Res. https:// 34. Kim C, Speisky MB, Kharouba SN (1987) Rapid and sensitive meth-
doi.org/10.1007/s12011-018-1286-1 od for measuring norepinephrine, dopamine, 5-hydroxytryptamine
14. Chen Y, Graziano JH, Parvez F, Liu M, Slavkovich V, Kalra T, and their major metabolites in rat brain by high-performance liquid
Argos M, Islam T, Ahmed A, Rakibuz-Zaman M (2011) Arsenic chromatography. Differential effect of probenecid, haloperidol and
exposure from drinking water andmortality from cardiovascular yohimbine on the concentrations of biogenic amines and metabolites
disease in Bangladesh: prospective cohort study. BMJ 342:d2431 in various regions of rat brain. J Chromatogr 386:25–35
15. Dhar P, Jaitley M, Kalaivani M, Mehra RD (2005) Preliminary 35. Kim M, Seo S, Sung K, Kim K (2014) Arsenic exposure in drinking
morphological and histochemical changes in rat spinal cord neurons water alters the dopamine system in the brains of C57BL/6 mice.
following arsenic ingestion. Neurotoxicology 26:309–320 Biol Trace Elem Res 162(1–3):175–180
16. Duker AA, Carranza EJ, Hale M (2005) Arsenic geochemistry and 36. Kordas K, Ardoino G, Coffman DL, Queirolo EI, Ciccariello
health. Environ Int 31:631–641 D, Mañay N, Ettinger AS (2015) Patterns of exposure to mul-
17. Dwivedi N, Flora SJ (2011) Concomitant exposure to arsenic and tiple metals and associations with neurodevelopment of pre-
organophosphates on tissue oxidative stress in rats. Food Chem school children from Montevideo, Uruguay. J Environ Public
Toxicol 49:1152–1159 Health 2015:493471
Arsenic-Induced Neurotoxicity by Dysfunctioning Cholinergic and Dopaminergic System in Brain of Developing...

37. Kozul-Horvath CD, Zandbergen F, Jackson BP, Enelow RI, Geen A, Khan K, Kline J, Ahsan H, Graziano JH (2011) Arsenic
Hamilton JW (2012) Effects of low-dose drinking water arsenic exposure and motor function among children in Bangladesh.
on mouse fetal and postnatal growth and development. PLoS One Environ Health Perspect 119:1665–1670
7:e38249 56. Patlolla AK, Tchounwou PB (2005) Serum acetyl cholinesterase as
38. Krinke G (2000) In: Bullock G, Bunton TE (eds) The laboratory rat: a biomarker of arsenic induced neurotoxicity in Sprague-Dawley
a volume in handbook of experimental animals, chapter 12 devel- rats. Int J Environ Res Public Health 2:80–83
opmental neurotoxicity. Academic Press, San Diego, p 75 57. Prakash C, Soni M, Kumar V (2016) Mitochondrial oxidative stress
39. Lazarini CA, Lima RY, Guedes AP, Bernardi MM (2004) Prenatal and dysfunction in arsenic neurotoxicity: a review. J Appl Toxicol
exposure to dichlorvos: physical and behavioral effects on rat off- 36(2):179–188
spring. Neurotoxicol Teratol 26:607–614 58. Rahman A, Persson LÅ, Nermell B, El Arifeen S, Ekström EC, Smith
40. Liu J, Waalkes MP (2018) Liver is a target of arsenic carcinogene- AH, Vahter M (2010) Arsenic exposure and risk of spontaneous abor-
sis. Toxicol Sci 105:24–32 tion, stillbirth, and infant mortality. Epidemiology 21:797–804
41. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) 59. Rahman A, Kumarathasan P, Gomes J (2016) Infant and moth-
Protein measurement with the Folin phenol reagent. J Biol er related outcomes from exposure to metals with endocrine
Chem 193:265–275 disrupting properties during pregnancy. Sci Total Environ 569-
42. Luo JH, Qiu ZQ, Shu WQ, Zhang YY, Zhang L, Chen JA (2009) 570:1022–1031
Effects of arsenic exposure from drinking water on spatial memory, 60. Rahman A, Granberg C, Persson LA (2017) Early life arsenic ex-
ultra-structures and NMDAR gene expression of hippocampus in posure, infant and child growth, and morbidity: a systematic review.
rats. Toxicol Lett 184:121–125 Arch Toxicol 91:3459–3467
43. Luo JH, Qiu ZQ, Zhang L, Shu WQ (2012) Arsenite exposure 61. Rodrigues EG, Bellinger DC, Valeri L, Hasan MO, Quamruzzaman
altered the expression of NMDA receptor and postsynaptic signal- Q, Golam M, Kile ML, Christiani DC, Wright RO, Mazumdar M
ing proteins in rat hippocampus. Toxicol Lett 211:39–44 (2016) Neurodevelopmental outcomes among 2- to 3-year-old chil-
44. Martinez-Finley EJ, Ali AM, Allan AM (2009) Learning deficits in dren in Bangladesh with elevated blood lead and exposure to arse-
C57BL/6J mice following perinatal arsenic exposure: consequence nic and manganese in drinking water. Environ Health 15:44
of lower corticosterone receptor levels? Pharmacol Biochem Behav 62. Rodríguez VM, Dufour L, Carrizales L, Díaz-Barriga F, Jiménez-
94:271–277 Capdeville ME (1998) Effects of oral exposure to mining waste on
45. Maurice T, Su T-P, Parish W, Nabeshima T, Privat A (1994) PRE- in vivo dopamine release from rat striatum. Environ Health Perspect
084, a sigma selective PCP derivative, attenuates MK-801-induced 106:487–491
impairment of learning in mice. Pharmacol Biochem Behav 49: 63. Rodríguez VM, Limón-Pacheco JH, Carrizales L, Mendoza-Trejo
859–869 MS, Giordano M (2010) Chronic exposure to low levels of inor-
46. Mazumdar M (2017) Does arsenic increase the risk of neural tube ganic arsenic causes alterations in locomotor activity and in the
defects among a highly exposed population? A new case–control expression of dopaminergic and antioxidant systems in the albino
study in Bangladesh. Birth Defects Res 109:92–98 rat. Neurotoxicol Teratol 32:640–647
47. Mejía JJ, Díaz-Barriga F, Calderón J, Ríos C, Jiménez-Capdeville 64. Roghani M, Joghataie MT, Jalali MR, Baluchnejadmojarad T
ME (1997) Effects of lead-arsenic combined exposure on central (2006) Time course of changes in passive avoidance and Y-maze
monoaminergic systems. Neurotoxicol Teratol 19:489–497 performance in male diabetic rats. Iran Biomed J J10:99–104
48. Milton AH, Hussain S, Akter S, Rahman M, Mouly TA, Mitchell K 65. Rosado JL, Ronquillo D, Kordas K, Rojas O, Alatorre J, Lopez P,
(2017) A review of the effects of chronic arsenic exposure on ad- Garcia-Vargas G, Del Carmen CM, Cebrian ME, Stoltzfus RJ
verse pregnancy outcomes. Int J Environ Res Public Health 6 (2007) Arsenic exposure and cognitive performance in Mexican
49. Minichilli F, Bianchi F, Ronchi AM, Gorini F, Bustaffa E schoolchildren. Environ Health Perspect 115:1371–1375
(2018) Urinary arsenic in human samples from areas charac- 66. Rudge CV, Röllin HB, Nogueira CM, Thomassen Y, Rudge MC,
terized by natural or anthropogenic pollution in Italy. Int J Odland JØ (2009) The placenta as a barrier for toxic and essential
Environ Res Public Health 15 elements in paired maternal and cord blood samples of South
50. Miyauchi M, Kishida I, Suda A, Shiraishi Y, Hattori S, Fujibayashi African delivering women. J Environ Monit 11:1322–1330
M, Taguri M, Ishii C, Ishii N, Moritani T, Hirayasu Y (2016) 67. Sanders AP, Desrosiers TA, Warren JL, Herring AH, Enright D,
Association of the cholinergic muscarinic M2 receptor with auto- Olshan AF, Meyer RE, Fry RC (2014) Association between arsenic,
nomic nervous system activity in patients with schizophrenia on cadmium, manganese, and lead levels in private wells and birth
high-dose antipsychotics. Neuropsychobiology 74:60–67 defects prevalence in North Carolina: a semi-ecologic study.
51. Moreno Ávila CL, Limón-Pacheco JH, Giordano M, BMC Public Health 14:955
Rodríguez VM (2016) Chronic exposure to arsenic in drinking 68. Shih YH, Islam T, Hore SK, Sarwar G, Shahriar MH, Yunus M,
water causes alterations in locomotor activity and decreases Graziano JH, Harjes J, Baron JA, Parvez F, Ahsan H, Argos M
striatal mRNA for the D2 dopamine receptor in CD1 male (2017) Associations between prenatal arsenic exposure with adverse
mice. J Toxicol 2016:4763434 pregnancy outcome and child mortality. Environ Res 158:456–461
52. Mufson EJ, Counts SE, Perez SE, Ginsberg SD (2008) Cholinergic 69. Shohamy D, Adcock RA (2010) Dopamine and adaptive memory.
system during the progression of Alzheimer's disease: therapeutic Trends Cogn Sci 14:464–472
implications. Expert Rev Neurother 8:1703–1718 70. Smeester L, Fry RC (2018) Long-term health effects and underly-
53. Mukherjee B, Bindhani B, Saha H, Sinha D, Ray MR (2014) ing biological mechanisms of developmental exposure to arsenic.
Platelet hyperactivity, neurobehavioral symptoms and depression Curr Environ Health Rep 1:134–144
among Indian women chronically exposed to low level of arsenic. 71. Soni I, Syed F, Bhatnagar P, Mathur R (2011) Perinatal toxicity of
Neurotoxicology 45:159–167 cyfluthrin in mice: developmental and behavioral effects. Hum Exp
54. Paratcha G, Furman M, Bevilaqua L, Cammarota M, Vianna M, de Toxicol 30:1096–1105
Stein ML, Izquierdo I, Medina JH (2000) Involvement of hippo- 72. Srivastava P, Yadav RS, Chandravanshi LP, Shukla RK, Dhuriya
campal PKCbeta1 isoform in the early phase of memory formation YK, Chauhan LK, Dwivedi HN, Pant AB, Khanna VK (2014)
of an inhibitory avoidance learning. Brain Res 855:199–205 Unraveling the mechanism of neuroprotection of curcumin in arse-
55. Parvez F, Wasserman GA, Factor-Litvak P, Liu X, Slavkovich V, nic induced cholinergic dysfunctions in rats. Toxicol Appl
Siddique AB, Sultana R, Sultana R, Islam T, Levy D, Mey JL, Van Pharmacol 279:428–440
Chandravanshi et al.

73. Terry AV Jr, Stone JD, Buccafusco JJ, Sickles DW, Sood A, 81. von Ehrenstein OS, Poddar S, Yuan Y, Mazumder DG, Eskenazi B,
Prendergast MA (2003) Repeated exposure to subthreshhold doses Basu A, Hira-Smith M, Ghosh N, Lahiri S, Haque R, Ghosh A,
of chlorpyrifos in rats: hippocampal damage, impaired axonal transport Kalman D, Das S, Smith AH (2007) Children's intellectual function
and deficits in spatial learning. J Pharmacol Exp Ther 305:375–384 in relation to arsenic exposure. Epidemiology 1:44–51
74. Tolins M, Ruchirawat M, Landrigan P (2014) The developmental 82. Vorhees CV, Brunner RL, Butcher RE (1979) Psychotropic drugs as
neurotoxicity of arsenic: cognitive and behavioral consequences of behavioral teratogens. Science 205:1220–1225
early life exposure. Ann Glob Health 80:303–314 83. Wang X, Li W, Li S, Yan J, Wilson JX, Huang G (2017) Maternal
75. Tota S, Kamat PK, Awasthi H, Singh N, Raghubir R, Nath C, Hanif folic acid supplementation during pregnancy improves neurobehav-
K (2009) Candesartan improves memory decline in mice: involve- ioral development in rat offspring. Mol Neurobiol 55:2676–2684.
ment of AT1 receptors in memory deficit induced by intracerebral https://doi.org/10.1007/s12035-017-0534-2
streptozotocin. Behav Brain Res 199:235–240 84. WHO (2008) BGuidelines for drinking-water quality, recommenda-
76. Tseng HP, Wang YH, Wu MM, The HW, Chiou HY, Chen CJ tions,^ in Incorporating 1st and 2nd Addenda. Vol. 1, 3rd Edn
(2006) Association between chronic exposure to arsenic and slow (Geneva: World Health Organization), 306–308b
nerve conduction velocity among adolescents in Taiwan. J Health 85. Willhite CC, Ferm VH (1984) Prenatal and developmental toxicol-
Popul Nutr 24:182–189 ogy of arsenicals. Adv Exp Med Biol 177:205–228
77. Tyler CR, Allan AM (2013) Adult hippocampal neurogenesis and 86. Wu J, Song TB, Li YJ, He KS, Ge L, Wang LR (2007) Prenatal
mRNA expression are altered by perinatal arsenic exposure in mice restraint stress impairs learning and memory and hippocampal
and restored by brief exposure to enrichment. PLoS One 8:e73720 PKCbeta1 expression and translocation in offspring rats. Brain
78. Vahter M (2009) Effects of arsenic on maternal and fetal health. Res 1141:205–213
Annu Rev Nutr 29:381–399 87. Yadav RS, Chandravanshi LP, Shukla RK, Sankhwar ML, Ansari
79. Valeri L, Mazumdar MM, Bobb JF, Claus Henn B, Rodrigues E, RW, Shukla PK, Pant AB, Khanna VK (2011) Neuroprotective
Sharif OIA, Kile ML, Quamruzzaman Q, Afroz S, Golam M, efficacy of curcumin in arsenic induced cholinergic dysfunctions
Amarasiriwardena C, Bellinger DC, Christiani DC, Coull BA, in rats. Neurotoxicology 32:760–768
Wright RO (2017) The joint effect of prenatal exposure to metal mix- 88. Zhang J, Liu X, Zhao L, Hu S, Li S, Piao F (2013) Subchronic
tures on neurodevelopmental outcomes at 20–40 months of age: evi- exposure to arsenic disturbed the biogenic amine neurotransmitter
dence from rural Bangladesh. Environ Health Perspect 125:067015 level and the mRNA expression of synthetase in mice brains.
80. Vathana T, Nijhuis TH, Friedrich PF, Bishop AT, Shin AY (2014) Neuroscience 241:52–58
An experimental study to determine and correlate choline acetyl- 89. Zhao F, Liao Y, Tang H, Piao J, Wang G, Jin Y (2017) Effects of
transferase assay with functional muscle testing after nerve injury. J developmental arsenite exposure on hippocampal synapses in
Neurosurg 120:1125–1130 mouse offspring. Metallomics 9:1394–1412

You might also like