ArticlePDF Available

Standardized Bacopa monnieri Extract Ameliorates Learning and Memory Impairments through Synaptic Protein, Neurogranin, Pro-and Mature BDNF Signaling, and HPA Axis in Prenatally Stressed Rat O�spring

Authors:

Abstract and Figures

Prenatal stress (PNS) influences offspring neurodevelopment, inducing anxiety-like behavior and memory deficits. We investigated whether pretreatment of Bacopa monnieri extract (CDRI-08/BME) ameliorates PNS-induced changes in signaling molecules, and changes in the behavior of Wistar rat offspring. Pregnant rats were randomly assigned into control (CON)/prenatal stress (PNS)/PNS and exposed to BME treatment (PNS + BME). Dams were exposed to stress by placing them in a social defeat cage, where they observed social defeat from gestational day (GD)-16–18. Pregnant rats in the PNS + BME group were given BME treatment from GD-10 to their offspring’s postnatal day (PND)-23, and to their offspring from PND-15 to -30. PNS led to anxiety-like behavior; impaired memory; increased the level of corticosterone (CORT), adrenocorticotropic hormone, glucocorticoid receptor, pro-apoptotic Casepase-3, and 5-HT2C receptor; decreased anti-apoptotic Bcl-2, synaptic proteins (synaptophysin, synaptotagmin-1), 5-HT1A, receptor, phosphorylation of calmodulin-dependent protein kinase II/neurogranin, N-methyl-D-aspartate receptors (2A,2B), postsynaptic density protein 95; and conversion of pro and mature brain derived neurotropic factor in their offspring. The antioxidant property of BME possibly inhibiting the PNS-induced changes in observed molecules, anxiety-like behavior, and memory deficits. The observed results suggest that pretreatment of BME could be an effective coping strategy to prevent PNS-induced behavioral impairments in their offspring.
Content may be subject to copyright.
antioxidants
Article
Standardized Bacopa monnieri Extract Ameliorates
Learning and Memory Impairments through
Synaptic Protein, Neurogranin, Pro-and Mature
BDNF Signaling, and HPA Axis in Prenatally
Stressed Rat Ospring
Karunanithi Sivasangari and Koilmani Emmanuvel Rajan *
Department of Animal Science, Behavioural Neuroscience Laboratory, Bharathidasan University,
Tiruchirappalli 620024, India; sangarik@bdu.ac.in
*Correspondence: emmanuvel1972@yahoo.com; Tel.: +91-9791-736-009
Received: 4 November 2020; Accepted: 24 November 2020; Published: 4 December 2020


Abstract:
Prenatal stress (PNS) influences ospring neurodevelopment, inducing anxiety-like behavior
and memory deficits. We investigated whether pretreatment of Bacopa monnieri extract (CDRI-08/BME)
ameliorates PNS-induced changes in signaling molecules, and changes in the behavior of Wistar rat
ospring. Pregnant rats were randomly assigned into control (CON)/prenatal stress (PNS)/PNS and
exposed to BME treatment (PNS +BME). Dams were exposed to stress by placing them in a social
defeat cage, where they observed social defeat from gestational day (GD)-16–18. Pregnant rats in the
PNS +BME group were given BME treatment from GD-10 to their ospring’s postnatal day (PND)-23,
and to their ospring from PND-15 to -30. PNS led to anxiety-like behavior; impaired memory;
increased the level of corticosterone (CORT), adrenocorticotropic hormone, glucocorticoid receptor,
pro-apoptotic Casepase-3, and 5-HT
2C
receptor; decreased anti-apoptotic Bcl-2, synaptic proteins
(synaptophysin, synaptotagmin-1), 5-HT
1A,
receptor, phosphorylation of calmodulin-dependent
protein kinase II/neurogranin, N-methyl-D-aspartate receptors (2A,2B), postsynaptic density protein 95;
and conversion of pro and mature brain derived neurotropic factor in their ospring. The antioxidant
property of BME possibly inhibiting the PNS-induced changes in observed molecules, anxiety-like
behavior, and memory deficits. The observed results suggest that pretreatment of BME could be an
eective coping strategy to prevent PNS-induced behavioral impairments in their ospring.
Keywords:
prenatal stress; Bacopa monnieri; caspase-3; synaptophysin; N-methyl-D-aspartate
receptor (NMDAR); brain-derived neurotropic factor (BDNF)
1. Introduction
Animal model and clinical studies have demonstrated the eect of prenatal stress (PNS) and
exposure to antidepressants from the perspective of the ospring [
1
4
]. Parallel studies have
documented that PNS-induced behavioral changes were associated with changes in adrenocorticotropic
hormone (ACTH), corticosterone (CORT) and its receptor (glucocorticoid receptor; GR), and the
monoaminergic system [
5
7
]. The hyperactivity of the hypothalamic–pituitary–adrenal (HPA) axis
led to the accumulation of corticosterone, which triggered the release of excitatory neurotransmitter
(glutamate, Glu) [
8
]. The level of Glu increased in the synaptic cleft by stress, which caused learning
and memory impairment through neuronal toxicity and degeneration [
9
] and could be linked with the
regulation of pro-apoptotic caspase-3 and anti-apoptotic Bcl-2 [
10
,
11
]. Interestingly, earlier studies have
reported that the interaction of corticotrophin-releasing hormone (CRH) with serotonergic neurons
Antioxidants 2020,9, 1229; doi:10.3390/antiox9121229 www.mdpi.com/journal/antioxidants
Antioxidants 2020,9, 1229 2 of 24
possibly interacted and regulated the release of serotonin (5-hydroxytryptamine, 5-HT) and in turn,
5-HT signals regulated the HPA axis function by regulating the level of CRH [12,13].
Dysregulation of the HPA axis has been known to alter the serotonergic system, thus, synaptic proteins
(synaptophysin (SYP), synaptotagmin-1 (SYT-1)) and 5-HT receptors were also altered [
14
,
15
]. Furthermore,
the synaptic transmission, finely tuned byinteraction between the presynaptic and postsynaptic proteins and
among the presynaptic proteins [
16
], regulated behavior [
17
]. In addition, the 5-HT
1A
/5-HT
2C
receptor has
been known to criticallyregulate anxiety/anxiolytic behavior [
18
20
]. Activation/inhibition of 5-HT receptor
critically regulate the signaling pathway mediated by phosphorylation of calmodulin-dependent
protein-kinase II (CaMKII) and N-methyl-D-asparate (NMDA) receptors [
21
,
22
]. Earlier studies have
shown that activation of CaMKII led to formation of a complex with NMDA receptors (NR2A, NR2B),
which was critical for stable long-term potentiation (LTP) [
23
,
24
]. Furthermore, this complex regulates
the localization of postsynaptic density protein 95 (PSD-95) in the spine, and its interactions with
brain-derived neurotrophic factor (BDNF) [
24
,
25
]. Precursor and mature BDNF, and its downstream
signaling molecules are known to influence synaptic plasticity and have been linked with behavioral
disorders [26,27].
In addition, in the Indian traditional system of Ayurvedic medicine, Bacopa monnieri (Linn.)
has been used as a nootropic agent in Ayurvedic herbal formulations and has been classified as
a medhyarasayana, a drug used to cure mental disorders. The characterization and structural
elucidation of B. monnieri extract reveals the presence of bacosides [bacoside A (bacogenins A1, A2, A3,
and A4), bacoside B, bacopacoside II, bacopasaponin X, and bacopasaponin C], which possibly hold the
property to improve cognitive function [
28
30
]. Studies in animal models of depression and anxiety
have shown that B. monnieri extract treatment reduced anxiety-like behaviors [
31
,
32
]. In addition,
B. monnieri extract (CDRI-08) supplementation reduced anxiety and depression in elderly people [
33
,
34
].
Homeostatic adaptation of hormones and neurotransmitters mediate stress adaptation, in which a
balancing act of oxidants and antioxidants can play a crucial role [
35
]. Dietary supplement with
antioxidants has been shown to be as eective for protecting against stress-induced abnormality [36].
In this study, we hypothesized that, in our PNS model, exposure to Bacopa monnieri extract
(CDRI-08) may have a resilience eect on PNS outcome in ospring. To test this, we examined
behaviors in an experimental group of adolescent rats and further estimated the level of CORT,
ACTH, expression of GR, caspase-3, synaptic proteins (SYP, SYT-1), 5-HT receptors (5-HT
1A/2C
),
CaMKII/p-CaMKII, NR2A/B receptors, PSD-95, as well as pro-and mature BDNF, in their ospring.
2. Materials and Methods
2.1. Bacopa monnieri Extract
The extract was prepared from the whole plant of Bacopa monnieri (CDRI-08, referred to as BME in
this articles) using ethanol (70%), and then the solvent was removed. Then, the soluble was concentrated
with 95% ethanol, dried in vacuo, and macerated with acetone, obtaining the free flowing powder.
The bacoside (bacopaside A3, bacopacoside II, bacopasaponin X, and bacopasaponin C) enriched
extract was obtained from Dr. Hemant Singh as a generous gift, Lumen marketing company, Chennai,
India (batch# C15030294). The high performance liquid chromatography (HPLC) chromatogram of the
extract is provided as Supplementary Data-1 [37].
2.2. Animal Design
The experimental timeline is depicted in Supplementary Figure S1 in Supplementary Materials.
Female Wistar rats (Rattus norvegicus, 180–250 g) were selected by continuously monitored their
estrous cycle and the distribution of dierent stages during the estrous cycle [
38
]. Selected Dams
were individually housed with one sexually experienced male for mating. Gestational day (GD)-0
was designated by the presence of sperms in vaginal lavage sample and pregnant rats were housed
individually in standard laboratory cage under a controlled environment (24
±
2
C, 12 h light-dark
Antioxidants 2020,9, 1229 3 of 24
cycle) with ad libitum water and food. Pregnant rats were randomly assigned into the following three
groups: (i) control (CON); (ii) prenatal stress (PNS, received 0.5% gum acacia treatment (per-orally,
p.o.)); and (iii) prenatal stress +exposed to BME (PNS +BME 80 mg/kg +0.5% gum acacia (p.o.))
treatment. In order to select the optimum dose of BME, dierent concentrations (40, 60, 80, 100 mg/kg)
of BME were provided to the individual dams, and then they were tested using a behavioral task [
36
].
Freshly prepared BME aqueous suspension (BME +gum acacia in double distilled water) or gum
acacia aqueous suspension (gum acacia in double distilled water) were treated orally to the dams
each day (10.00 to 11.00 h) from GD-10 to their pup’s postnatal day (PND)-23 except on the day of
parturition (
8 h after parturition), and to the pups from PND-15 to -30. The day of parturition was
noted as PND-0 and the male pups were separated on PND-24 from their dam and housed (2–3/cage)
in dierent cages [
39
]. Two or three male pups were randomly selected from each dam (three dam for
each group) for dierent experimental groups (i.e., control (n=7), PNS (n=6), and PNS +BME (n=6)),
and selected pups were subjected to the behavioral test on PND-31/-32. Throughout the study, care was
taken to minimize the handling stress by partially replacing the bedding material periodically to ensure
the home cage odor, and minimally disrupt the nests. All the experimental group rats were handled
minimally to avoid handling eects. The project was recommended by the Institutional Animal Ethics
Committees of Bharathidasan University, Tiruchirappalli, India (approval no. BDU/IAEC/P22/2018
dated 7 August 2018) and approved by the Committee for the Purpose of Control and Supervision of
Experiments on Animals (CPCSEA) (reg. no. 418/GO/Re/S/01/CPCSEA dated 27 April 2018). All the
experiments were conducted in compliance with the guidelines laid down by CPCSEA, Government
of India, India.
2.3. Chemicals
All chemicals used in this study were of analytical grade.
2.4. Prenatal Stress
To induce prenatal stress (PNS), the pregnant rats were allowed to observe social defeat (SDO) from
GD-16 to GD-18, in a specially designed social defeat cage (Supplementary Figure S2 in Supplementary
Materials) for 10 min/day, following the procedure modified from Lee et al. [
40
]. The social defeat cage
consisted of three chambers (observer chamber (OC), intruder chamber (IC), resident chamber (RC))
with equal size (30
×
30
×
30 cm). The RC was connected with a standard laboratory cage through a
transparent plastic pipe (60 cm in length and 10.5 cm in diameter), the OC was permanently partitioned
with wire mesh, and the RC and IC were partitioned with sliding wire mesh doors, which facilitated
the aggressive interaction of the resident male and intruder male. Ten days before the experiment,
the aggressor (senescence male rat) was housed in the standard laboratory cage which was connected
to the RC and allowed the aggressor free access to the RC. First, the intruder (six-month-old male
rat) was introduced into the IC, and after five minutes, the sliding wire mesh door was opened and
the intruder was allowed to interact (10 min/day) with the aggressor in the RC. After the aggressive
interaction the intruder was transferred to the home cage. The control group pregnant rats were
allowed to explore the OC in the absence of aggressor and intruder from GD-16 to GD-18. The PNS
and PNS +BME group were allowed to observe the social defeat (GD-16 to GD-18) from the OC.
2.5. Behavioral Test
2.5.1. Elevated Plus Maze (EPM)
Experimental group pups were subjected to the elevated plus maze (EPM) test from PND-31 to
PND-32. The EPM test was used to identify the anxiety-like behavior in rats; the two closed arm
(30
×
5
×
15 cm) and two open arms (30
×
5
×
0.25 cm) were connected to the central platform and were
50 cm above the floor level [
41
]. Three hours before the behavioral test, animals were shifted to the
behavioral testing room and kept undisturbed. Individually, test animals were placed in the central
Antioxidants 2020,9, 1229 4 of 24
square of the EPM, facing towards an open arm and they were allowed to explore for five minutes.
Individuals’ behavior was video recorded for analysis. The entire behavior test was conducted under
bright illumination (30 W). The apparatus was cleaned with 75% ethanol after every trial to remove
olfactory cues.
2.5.2. Y-Maze Test
Individual rat’s spontaneous alternation behavior was tested using a Y-maze apparatus.
The Y-shaped apparatus was constructed with three horizontal arms to connect symmetrically at
a 120
angle (40
×
5
×
15 cm) [
42
]. The behavioral test was conducted in a behavioral room which was
illuminated with a 100 W bulb. On PND-30, the experimental animal was placed in the start arm end
and allowed to explore the maze for 15 min. The next day, the B arm was closed, and the reward was
placed in the C arm to ensure that the C arm was their first choice for 10 min. On PND-32, the testing
was conducted for 10 min by allowing the rat to investigate all three arms, in which the closed arm
was presented as a novel arm and the reward was placed in the C arm. An entry was calculated when
a rat placed all four paws in an arm. Pure chocolate was provided as the reward without nuts or any
other compounds. Training and testing were conducted as 2 trials/day with four-hour interval. On the
day of testing, individuals’ behavioral profiles, such as total number of arm entries and spontaneous
alternation (number of traits), were recorded and analyzed. As triplet sets (i.e., ABC/BCAo/CAB but
not ABA/BCB/CAC) the entries were designated to calculate spontaneous alternation. The alternations
(% alternations) were calculated as spontaneous alternations/(total number of arm entries 2) ×100.
2.6. Hormone Assay
To estimate the level of corticosterone (CORT) and adrenocorticotropic hormone (ACTH), blood
samples were collected from rats representing each group (control, PNS, PNS +BME, n=6) into
a tube with anticoagulant (sodium citrate, 0.25 mL of 3.8% sodium per 2.0 mL of blood). Plasma was
separated by centrifuging at 1800
×
gfor 10 min [
43
] and stored at
20
C. The CORT and ACTH levels
in plasma were estimated by using an ELISA kit (ALPCO Diagnostics, Salem, NH, USA).
2.7. Total RNA Isolation
Animals (n=6) representing each group were sacrificed after the behavioral test. The whole brain
was dissected out and placed on an ice-cold petri dish. The amygdala tissue was carefully dissected
from the brain slice, as reported earlier [
44
]. One portion of the dissected amygdale was used to
isolate RNA and the other side for isolation of protein. Total RNA was isolated by using TRI Reagent
(cat. # FATRR 001; Favogen Biotech Corp, Pingtung, Taiwan) and stored at
80
C. Total RNA (2
µ
g)
was used to synthesis cDNA (cat.# 170-8891; Iscript
TM
cDNA synthesis kit, Bio-Rad laboratories Inc.,
Hercules, CA, USA).
2.8. Protein Isolation
Amygdala tissue obtained from the experimental groups were homogenized in ice-cold lysis buer
(150 mM NaCl, 50 mM Tris-Hcl pH 7.5, 5 mM EDTA, 0.1% v/vNP-40, 1.0 mM DTT, 0.2 mM sodium
orthovanadate, 0.23 mM PMSF) containing protease inhibitor (4
µ
L/mL) (Sigma-Aldrich, Saint Louis,
MO, USA). The homogenate was incubated in ice for 30 min, followed by centrifuged at 4
C (10,000
×
g)
for 30 min. The supernatants were collected in anew tube and again centrifuged at 4
C (12,000
×
g)
for 15 min, after being extracted, the samples as aliquots were stored at
80
C [
36
,
45
47
]. Protein
was quantified at 595 nm by Bradford method [
48
] (cat. #5000006; Bio-Rad Protein Assay kit, Bio-Rad
laboratories Inc., Hercules, CA, USA) using a Biophotometer Plus (Eppendorf Inc., Hamburg, Germany).
Antioxidants 2020,9, 1229 5 of 24
2.9. Quantitative Real-Time PCR (qRT-PCR)
Total reaction volume (10
µ
L) contains an aliquot of real-time mixture (SYBR green super
mix, Bio-Rad laboratories Inc.) with cDNA (0.2
µ
g) and specific primers (100 pmoles) [
36
].
Specific primers were used to estimate the expression of 5-HT
1A
(accession number NM_012585.1;
forward 5
0
-GACTACGTGAACAAGAGGAC-3
0
and reverse 5
0
-TATAGAAAGCGCCGAAAGTG-3
0
),
5-HT
2C
and (accession number NM_012765.3; forward 5
0
-AAACTGCACAATGCTACCAA-3
0
and
reverse 5
0
-TGATGGACGCAGTTGAAAAT-3
0
) normalized with internal control and glyceroldehydes
-3-phosphate dehydragenase (GAPDH) (accession number NM_017008.4; forward 5
0
-AACATCAT
CCCTGCATCCAC-3
0
and reverse 5
0
-AGGAACACGGAAGGCCATGC-3
0
). The reaction starts with
92
C initial denaturation (3 min), and then 40 cycles of denaturation at 92
C (5 s), annealing (5 s)
(5-HT
1A
(52.4
C), 5-HT
2C
(59.7
C), GAPDH (56
C)), extension at 72
C (5 s), and final extension at 72
C
(10 min). Single specific PCR product amplification was confirmed by observing the dissociation curve
followed by melting curve analysis (CFX-96 Touch Real-Time PCR detection system; CFX manager
version 2 software; Bio-Rad laboratories Inc., Hercules, CA, USA) [
49
,
50
]. The data obtained from
triplicate were normalized with internal control and presented as the mean fold change relative to the
control group.
2.10. Western Blot Analysis
An equal concentration of total protein (80
µ
g) separated on polyacrylamide gel (PAGE 10%) was
mixed with a buer (2% mercaptoethanol, 100% glycerol, 4% SDS, 125 mM Tris-HCL pH 6.8, 0.006%
bromophenol blue) and boiled (2 min). Semi-dry Western blot apparatus (SD-20; Cleaver Scientific Ltd.,
Rugby, UK) was used to transfer the separated protein to the polyvinylidine difluride membrane
(PVDF) (cat. #IPVH00010; Millipore, Burlington, MA, USA). The membranes were preblocked in
Tris-buered saline (TBS, 10 mM Tris-base pH 7.5 and 150 mM NaCl) containing Tween-20 (0.1%)
and non-fat milk (5% for 2 h) at room temperature. Membranes were incubated with any one of the
following primary antibodies for 12–15 h at 4
C: rabbit-anti-GR (SC-1004, 1:1000); rabbit-anti-caspase-3
(SC-7148, 1:1000); rabbit-anti-Bcl2 (ab59348, 1:1000); rabbit-anti-SYP (M-04-1019, 1:10,000) (Millipore,
Burlington, MA, USA); mouse anti-SYT1 (BD-610433, 1:4000)(BD Biosciences, San Jose, CA, USA); mouse
anti-total-
α
CaMKII a (t-
α
CaMKII, SC-32288, 1:200); rabbit anti-phosphorylated
α
CaMKII (p-
α
CaMKII,
Thr
286
SC-12886-R, 1:200); rabbit anti-Ng (ab23570, 1:1000); rabbit anti-pNgantibody (ABN 426, 1:1000);
rabbit polyclonal NR2A antibody (BT-AP02388, 1:2000); rabbit NR2B (BT-AP02389, 1:2000); rabbit
anti-PSD-95 (SC-28941, 1:200); rabbit anti-pro-BDNF (SC-20981 (H-117), 1:500); rabbit anti-BDNF
(SC-546 (N-20), 1:500).For the internal control rabbit polyclonal anti-rabbit-
β
-actin (SC-130656) was
used. The membrane was washed with TBS-T (TBS containing 1% Tween-20), and then incubated
(for 4 h) in either alkaline phosphatase (ALP) conjugated goat anti-rabbit (MERK, cat.# 62110080011730,
1:2000) or anti-mouse (MERK cat.# 621100480011730, 1:2000) antibody to detect the membrane bounded
antibodies. Subsequently, ALP activity was detected with the substrates 5 bromo-4-chloro-3-indolyl
phosphate di-sodium salt (BCIP) and nitro blue tetrazolium chloride (NBT) (cat.# S 3771; Promega
Biotech Ltd., Madison, WI, USA), following the manufacturer’s instruction. Individuals’ blot images
were obtained using Molecular Imager and each band trace quantity was measured (Chemi Doc XRS
system, Image Lab 2 software (2.0) Bio-Rad laboratories Inc., Hercules, CA, USA). Dierences from
the control group were presented in relative fold [
36
,
45
47
]. All uncropped Western blot images are
provided in Supplementary Materials.
2.11. Statistical Analysis
KyPlot (ver 1.0) was used to plot the values (mean
±
standard error of the mean (SEM)) as a graphical
representation. The significant dierence among the experimental groups (CON, PNS, PNS +BME)
were tested with one-way analysis of variance (ANOVA) followed by post hoc (Bonferroni test) analysis
Antioxidants 2020,9, 1229 6 of 24
(Sigma Stat version 3.1). Significant dierence among groups (* p<0.05, ** p<0.01, *** p<0.001)
and NS, not significantly dierent were indicated wherever required.
3. Results
3.1. Behavioral Results
3.1.1. Elevated Plus Maze
Behavioral profile in EPM showed that exposure to the BME treatment reduced the PNS-induced
anxiety-like behavior in their ospring. The observed behavioral data showed that there was a
significant dierence in the time spent in the open arm across the experimental groups (F
(2,18)
=6.79,
p<0.01). Bonferroni post hoc comparative analysis suggests that, in the open arm, the PNS group
spent less time than the control (p<0.05) and the PNS +BME group (p<0.05), but there was no
significant dierence between the control and the PNS +BME group (p=1.00) (Figure 1a). In addition,
for individual time spent in the closed arm, there was a significant dierence across the study groups
(F
(2,18)
=7.300, p<0.01). The post hoc analysis demonstrates that the PNS group spent more time
than the control group (p<0.05) and the PNS +BME group (p<0.05). Whereas no dierence was
observed between the control group and the PNS +BME group (p=0.984) (Figure 1b). Similarly,
the PNS group resulted in a significant dierence in total number of entries (F
(2,37)
=3.76, p<0.05).
The post hoc analysis demonstrated that, unlike the control animals, the PNS group (p<0.05) exhibited
fewer entries and there was no significant dierence between the PNS group and the PNS +BME
group (p=0.212) and the control group and the PNS +BME group (p=1.00) (Figure 2). When the
percentage of open entries was calculated, a significant dierence was noted across the experimental
group (F
(2,18)
=9.53, p<0.01). The post hoc analysis revealed that the PNS group was significantly
dierent from the control group (p<0.05) and the PNS +BME group (p<0.05), but the control group
was not significantly dierent from the PNS +BME group (p=0.759). Similarly, a significant dierence
was observed across the groups regarding closed arm entries (F
(2,18)
=9.22; p<0.01). Subsequently,
the post hoc comparison showed a significant dierence between the control group and the PNS group
(p<0.01) and the PNS group and the PNS+BME group (p<0.05), where a significant dierence was
not observed between the control group and the PNS +BME group (p=1.00).
3.1.2. Spatial Memory
In addition, the Y-maze analysis showed a significant eect of the variable group (F
(2,37)
=14.28,
p<0.001), and the post hoc comparison analysis indicated that, with reference to the control group,
the PNS group spent significantly less arm entries (p<0.001), as well as the PNS +BME group
(p<0.01). However, the PNS group made significantly less entries than the PNS +BME group (p<0.05)
(Figure 3a). When we estimated the percentage of alternation during training, we found a significant
dierence across the groups (F
(2,37)
=17.42, p<0.001). The post hoc analysis revealed that the PNS
group exhibited a lower percentage of alternation than the control group (p<0.001) and the PNS +BME
group (p<0.001), but a significant dierence was not observed between the control group and the
PNS +BME group (p=0.421). Similarly, during testing, the recorded alternation between groups
was significantly dierent (F
(2,37)
=21.468, p<0.001). The post hoc analysis showed that the PNS
individuals displayed a lower percentage of alternation than the control group (p<0.001) and the
PNS +BME group (p<0.001). In fact, we did not find a significant dierence between the control and
the PNS +BME groups (p =0.11) (Figure 3b). The observed behavioral data in the EPM and the Y-maze
tests demonstrated that the exposure to the BME treatment protected against prenatal stress-induced
anxiety-like behavior and memory impairment in their ospring.
Antioxidants 2020,9, 1229 7 of 24
1
Figure 1.
Exposure to BME treatment and resilience eect on the prenatal stress (PNS)-induced
anxiety-like behaviors in the ospring. Behavioral profile in the elevated plus maze (EPM). (
a
) Total
time spent in the closed arm; (
b
) Total time spent in the open arm. The anxiety induced by the
gestational stress was significantly reduced by the maternal exposure to the BME treatment. Data are
expressed as mean
±
SEM (control n=7, PNS n=6, PNS +BME n=6). One-way ANOVA followed by
Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is indicated by
*p<0.05; NS—Not significant).
Antioxidants 2020, 9, x FOR PEER REVIEW 7 of 26
Figure 1. Exposure to BME treatment and resilience effect on the prenatal stress (PNS)-induced
anxiety-like behaviors in the offspring. Behavioral profile in the elevated plus maze (EPM). (a) Total
time spent in the closed arm; (b) Total time spent in the open arm. The anxiety induced by the
gestational stress was significantly reduced by the maternal exposure to the BME treatment. Data are
expressed as mean ± SEM (control n = 7, PNS n = 6, PNS + BME n = 6). One-way ANOVA followed by
Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is indicated by
* p < 0.05; NS—Not significant).
Figure 2.
Eect of exposure to BME treatment on PNS-induced changes in ospring. Behavioral profile
of EPM showed as the percentage of entries in the open arm and closed arm, maternal exposure to
BME treatment and resilience eect on the anxiety-like behavior in the ospring. Data are expressed
in percentage (mean
±
SEM) calculated against the total number of entries (control n=7, PNS n=6,
PNS +BME n=6). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple
comparisons, statistical significance is indicated by * p<0.05 and ** p<0.01; NS—Not significant).
Antioxidants 2020,9, 1229 8 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 9 of 26
Figure 3. Exposure to BME treatment protects against PNS-induced memory impairment in offspring.
The behavioral profile in the Y-maze. (a) The BME exposed group (a) made more arm entries than the
PNS group (b) showed a higher percentage of alternations. Data are expressed as mean ± SEM (control
n = 7, PNS n = 6, PNS + BME n = 6). One-way ANOVA followed by Bonferroni post hoc test (for all
pairwise multiple comparisons, statistical significance is indicated by * p < 0.05, ** p < 0.01 and *** p <
0.001; NS—Not significant)
3.2. Hormonal Results
3.2.1. Effect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Corticosterone
and Adrenocorticotropic Hormone
Subsequently, we estimated the level of corticosterone and adrenocorticotropic hormone in the
plasma of the experimental groups. As shown in Figure 4a, a significant effect was demonstrated
across the groups (F(2,17) = 104.78; p < 0.001).The post hoc test revealed significant changes in the level
of corticosterone of the PNS group as compared with the control (p < 0.01) and PNS+ BME groups (p
< 0.01). However, exposure to BME treatment significantly reduced the PNS-induced elevation in
corticosterone of the PNS group, and therefore no significant difference between the control and PNS
+ BME groups (p = 0.078). Similarly, we found a significant increase in the level of adrenocorticotropic
hormone of the PNS group as compared with the level of the control group (F(2,37) = 64.16; p < 0.001).
Figure 3.
Exposure to BME treatment protects against PNS-induced memory impairment in ospring.
The behavioral profile in the Y-maze. (
a
) The BME exposed group (
a
) made more arm entries than
the PNS group (
b
) showed a higher percentage of alternations. Data are expressed as mean
±
SEM
(control n=7, PNS n=6, PNS +BME n=6). One-way ANOVA followed by Bonferroni post hoc test
(for all pairwise multiple comparisons, statistical significance is indicated by * p<0.05, ** p<0.01 and
*** p<0.001; NS—Not significant).
3.2. Hormonal Results
3.2.1. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Corticosterone and
Adrenocorticotropic Hormone
Subsequently, we estimated the level of corticosterone and adrenocorticotropic hormone in the
plasma of the experimental groups. As shown in Figure 4a, a significant eect was demonstrated across
the groups (F
(2,17)
=104.78; p<0.001).The post hoc test revealed significant changes in the level of
corticosterone of the PNS group as compared with the control (p<0.01) and PNS +BME groups (p<0.01).
However, exposure to BME treatment significantly reduced the PNS-induced elevation in corticosterone
of the PNS group, and therefore no significant dierence between the control and PNS +BME groups
(p=0.078). Similarly, we found a significant increase in the level of adrenocorticotropic hormone of
the PNS group as compared with the level of the control group (F
(2,37)
=64.16; p<0.001). The post
hoc analysis indicated a significant increase in the adrenocorticotropic hormone level of the PNS
group as compared with the control group (p<0.01) and the PNS +BME group (p<0.01). However,
there was no significant dierence between the PNS +BME and control groups (p=0.844) (Figure 4b).
Antioxidants 2020,9, 1229 9 of 24
The above results indicate that PNS increases the level of corticosterone and adrenocorticotropic
hormone but exposure to BME treatment had resilience eect on the PNS-induced CORT and ACTH
level, which could correlate with the observed anxiolytic behavior in the BME supplemented group.
Figure 4.
Maternal exposure to BME treatment and the resilience eect on the PNS-induced changes in
ospring plasma. (
a
) Corticosterone (CORT); (
b
) Adrenocorticotropic hormone (ACTH) level. Exposure
to BME treatment significantly reduced the PNS-induced CORT and ACTH level. Data are expressed
as mean
±
SEM (n=6 for each group). One-way ANOVA followed by Bonferroni post hoc test (for all
pairwise multiple comparisons, statistical significance is indicated by ** p<0.01; NS—Not significant).
3.2.2. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Changes on
Glucocorticoid Receptor
As shown in Figure 5(Supplementary Figure S3), the level of glucocorticoid receptor expression
was significantly dierent across the groups (F
(2,17)
=278.96, p<0.001). The post hoc comparison
suggested that level of glucocorticoid receptor was significantly higher in the PNS group as compared
with the control group (p<0.001) and the PNS +BME group (p<0.001). However, the level
of glucocorticoid receptor was significantly lower in the PNS group exposed to BME treatment as
compared with the control group (p<0.001). The observed results suggest that PNS alters glucocorticoid
receptor expression but exposure to BME treatment has a possible resilience eect on the PNS-induced
changes in glucocorticoid receptor.
Antioxidants 2020,9, 1229 10 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 11 of 26
Figure 5. Effect of exposure to BME treatment on PNS-induced changes in offspring’s glucocorticoid
receptor (GR) level in amygdale. (a) Representative Western blot showing that the level of GR varied
across the experimental group; (b) Estimated level of GR suggests that exposure to BME treatment
has a resilience effect on the PNS-induced changes. Data are expressed as mean ± SEM (n = 6/each
group). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple
comparisons, statistical significance is indicated by *** p < 0.001).
3.3. Western Blot Results
3.3.1. Effect of Exposure to BME Treatment on Prenatal Stress-Induced Neuronal Apoptosis
Furthermore, we examined the level of caspase-3 and Bcl-2 to measure the PNS-induced
neuronal damage and the effect of exposure to BME treatment. The Western blot analysis showed
that PNS induced the expression of caspase-3 (Figure 6a, Supplementary Figure S4). The analysis
revealed that the level of caspase-3 significantly varied across the groups (F
(2,17)
= 1714.3, p < 0.001).
The post hoc analysis demonstrated that level of caspase-3 was significantly higher in the PNS group
as compared with the control (p < 0.001) and the PNS + BME groups (p < 0.001). Exposure to the BME
treatment reduced the caspase-3 level, however, the level was significantly higher than in the control
group (p < 0.001) (Figure 6b). Furthermore, the level of Bcl-2 varied significantly across the
experimental groups (F
(2,17)
= 74.2, p < 0.001). The post hoc analysis indicated that the level of Bcl-2
was significantly lower in the PNS group than in the control (p < 0.01) and PNS + BME groups (p <
0.01). This showed that exposure to BME treatment elevated the Bcl-2 level, however, a significant
difference was not detected between the control and the PNS+ BME groups (p = 0.092) (Figure 6c).
Taken together, the observed results suggest that exposure to BME treatment protects against PNS-
induced neuronal damage by balancing the expression of caspase-3 and Bcl-2.
Figure 5.
Eect of exposure to BME treatment on PNS-induced changes in ospring’s glucocorticoid
receptor (GR) level in amygdale. (
a
) Representative Western blot showing that the level of GR varied
across the experimental group; (
b
) Estimated level of GR suggests that exposure to BME treatment has
a resilience eect on the PNS-induced changes. Data are expressed as mean
±
SEM (n=6/each group).
One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple comparisons, statistical
significance is indicated by *** p<0.001).
3.3. Western Blot Results
3.3.1. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Neuronal Apoptosis
Furthermore, we examined the level of caspase-3 and Bcl-2 to measure the PNS-induced neuronal
damage and the eect of exposure to BME treatment. The Western blot analysis showed that PNS
induced the expression of caspase-3 (Figure 6a, Supplementary Figure S4). The analysis revealed that
the level of caspase-3 significantly varied across the groups (F
(2,17)
=1714.3, p<0.001). The post hoc
analysis demonstrated that level of caspase-3 was significantly higher in the PNS group as compared
with the control (p<0.001) and the PNS +BME groups (p<0.001). Exposure to the BME treatment
reduced the caspase-3 level, however, the level was significantly higher than in the control group
(p<0.001) (Figure 6b). Furthermore, the level of Bcl-2 varied significantly across the experimental
groups (F
(2,17)
=74.2, p<0.001). The post hoc analysis indicated that the level of Bcl-2 was significantly
lower in the PNS group than in the control (p<0.01) and PNS +BME groups (p<0.01). This showed
that exposure to BME treatment elevated the Bcl-2 level, however, a significant dierence was not
detected between the control and the PNS+BME groups (p=0.092) (Figure 6c). Taken together,
the observed results suggest that exposure to BME treatment protects against PNS-induced neuronal
damage by balancing the expression of caspase-3 and Bcl-2.
Antioxidants 2020,9, 1229 11 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 12 of 26
Figure 6. Effect of exposure to BME treatment on PNS-induced changes on caspase-3 and Bcl-2 level
in amygdale. (a) Representative Western blot showing that the levels of caspase-3 and Bcl-2 varied
across the experimental groups. Quantitative estimation of caspase-3 (b) and Bcl-2 (c) showing that
exposure to BME treatment induces an opposite pattern of expression. Data are expressed as mean ±
SEM (n = 6/group). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple
comparisons, statistical significance is indicated by ** p < 0.01 and ***p < 0.001; NS—Not significant).
3.3.2. Effect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Synaptophysin
and Synaptotagmin-1
Furthermore, we found that exposure to BME treatment had a resilience effect on the PNS-
induced changes in synaptic proteins (Figure 7a, Supplementary Figure S5). In addition, the level of
synaptophysin was also altered by PNS. The observed changes in the level of synaptophysin was
significantly different across the groups (F
(2,17)
= 2097.45, p < 0.001). The post hoc comparison showed
that synaptophysin was significantly higher in the PNS group than that of the control group (p <
0.001) but significantly lower than that of the PNS + BME group (p < 0.001), whereas the level of
synaptophysin was significantly less than the control group (p < 0.001) (Figure 7b). One-way ANOVA
showed a significant difference across the experimental groups (F
(2,17)
= 528.20, p < 0.001), and the post
hoc comparison showed that the level of synaptotagmin-1 was significantly lower in the PNS group
than the control (p < 0.01) and PNS+ BME group (p < 0.001). However, the detected synaptotagmin-1
level in the control group was not significantly different from individuals exposed to BME treatment
(p = 0.08) (Figure 7c).
Figure 6.
Eect of exposure to BME treatment on PNS-induced changes on caspase-3 and Bcl-2 level in
amygdale. (
a
) Representative Western blot showing that the levels of caspase-3 and Bcl-2 varied across
the experimental groups. Quantitative estimation of caspase-3 (
b
) and Bcl-2 (
c
) showing that exposure
to BME treatment induces an opposite pattern of expression. Data are expressed as mean
±
SEM
(n=6/group). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple
comparisons, statistical significance is indicated by ** p<0.01 and *** p<0.001; NS—Not significant).
3.3.2. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Synaptophysin and
Synaptotagmin-1
Furthermore, we found that exposure to BME treatment had a resilience eect on the PNS-induced
changes in synaptic proteins (Figure 7a, Supplementary Figure S5). In addition, the level of
synaptophysin was also altered by PNS. The observed changes in the level of synaptophysin was
significantly dierent across the groups (F
(2,17)
=2097.45, p<0.001). The post hoc comparison showed
that synaptophysin was significantly higher in the PNS group than that of the control group (p<0.001)
but significantly lower than that of the PNS +BME group (p<0.001), whereas the level of synaptophysin
was significantly less than the control group (p<0.001) (Figure 7b). One-way ANOVA showed a
significant dierence across the experimental groups (F
(2,17)
=528.20, p<0.001), and the post hoc
comparison showed that the level of synaptotagmin-1 was significantly lower in the PNS group than
the control (p<0.01) and PNS+BME group (p<0.001). However, the detected synaptotagmin-1
level in the control group was not significantly dierent from individuals exposed to BME treatment
(p=0.08) (Figure 7c).
Antioxidants 2020,9, 1229 12 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 13 of 26
Figure 7. Exposure to BME treatment protects against PNS-induced changes in synaptic proteins. (a)
Representative Western blots showing that the level synaptophysin (SYP) and synaptotagmin-1 (SYT-
1) in the experimental groups; (b) SYP and (c) SYT-1 were significantly increased in the BME
supplemented group. Data are expressed as mean ± SEM (n = 6/each group). One-way ANOVA
followed by Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is
indicated by ** p < 0.01 and *** p < 0.001; NS—Not significant).
3.3.3. Effect of Exposure to BME Treatment on Prenatal Stress-Induced Changes on 5-HT1A and 5-
HT2C Receptors
Subsequently, we estimated the expression level of 5-HT1A and 5-HT2C receptors in the
experimental groups (Supplementary Figure S6). The one-way ANOVA analysis showed a significant
difference in the estimated level of 5-HT1A between the groups (F(2,17) = 44.94,p< 0.001). The post hoc
analysis indicated that 5-HT1A receptor expression was significantly lower in the PNS group than in
the control (p < 0.05) and PNS + BME groups (p < 0.01). In fact, the level of 5-HT1A was significantly
higher in the PNS + BME group than in the control group (p <0.01) (Figure 8a). When we examined
the 5-HT2C receptor, the level of expression between groups varied significantly (F(2,17) = 390.92,p<
0.001). The post hoc comparison revealed that the level of 5-HT2C receptor expression was elevated in
the PNS group as compared with the control (p < 0.01) and PNS + BME groups (p < 0.01). Exposure to
the BME treatment significantly reduced the PNS-induced changes in 5-HT2C receptor, and therefore
a significant difference was observed between the control and PNS + BME groups (p < 0.001) (Figure
8b). The obtained data demonstrates that exposure to BME treatment differentially regulates 5-HT1A
and 5-HT2C receptors, with a possible resilience effect on the PNS-induced impact on the serotonergic
system.
Figure 7.
Exposure to BME treatment protects against PNS-induced changes in synaptic proteins.
(
a
) Representative Western blots showing that the level synaptophysin (SYP) and synaptotagmin-1
(SYT-1) in the experimental groups; (
b
) SYP and (
c
) SYT-1 were significantly increased in the BME
supplemented group. Data are expressed as mean
±
SEM (n=6/each group). One-way ANOVA
followed by Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is
indicated by ** p<0.01 and *** p<0.001; NS—Not significant).
3.3.3. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Changes on 5-HT1A and
5-HT2C Receptors
Subsequently, we estimated the expression level of 5-HT
1A
and 5-HT
2C
receptors in the
experimental groups (Supplementary Figure S6). The one-way ANOVA analysis showed a significant
dierence in the estimated level of 5-HT
1A
between the groups (F
(2,17)
=44.94, p<0.001). The post hoc
analysis indicated that 5-HT
1A
receptor expression was significantly lower in the PNS group than in the
control (p<0.05) and PNS +BME groups (p<0.01). In fact, the level of 5-HT
1A
was significantly higher
in the PNS +BME group than in the control group (p<0.01) (Figure 8a). When we examined the 5-HT
2C
receptor, the level of expression between groups varied significantly (F
(2,17)
=390.92, p<0.001). The post
hoc comparison revealed that the level of 5-HT
2C
receptor expression was elevated in the PNS group as
compared with the control (p<0.01) and PNS +BME groups (p<0.01). Exposure to the BME treatment
significantly reduced the PNS-induced changes in 5-HT
2C
receptor, and therefore a significant dierence
was observed between the control and PNS +BME groups (p<0.001) (Figure 8b). The obtained data
demonstrates that exposure to BME treatment dierentially regulates 5-HT1A and 5-HT
2C
receptors,
with a possible resilience eect on the PNS-induced impact on the serotonergic system.
Antioxidants 2020,9, 1229 13 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 14 of 26
Figure 8. Exposure to BME treatment protects the PNS-induced alternations in the serotonergic
system. Quantitative real-time PCR analysis showing that exposure to BME treatment significantly
ameliorates the PNS-induced changes in (a) 5-HT
1A
and (b) 5-HT
2C
receptor expression. Data are
expressed as mean ± SEM (n = 6/group). One-way ANOVA followed by Bonferroni post hoc test (for
all pairwise multiple comparisons, statistical significance is indicated by * p < 0.05 and ** p < 0.01).
3.3.4. Effect of Exposure to BME Treatment on Gestational Stress-Induced Changes on Neurogranin
(Ng) and CaMKII Signaling
Subsequently, we examined the effect of PNS on activation of CaMKII. The western blot analysis
demonstrated that the level of CaMKII was altered (Figure 9a, Supplementary Figure S7). The one-
way ANOVA reported a significant difference across the variable groups (F
(2,17)
= 132.10, p < 0.001).
The post hoc comparison analysis showed that the level of p-CaMKII/CaMKII was significantly
reduced in the PNS group (p < 0.001) as compared with the control and PNS + BME groups (p < 0.001).
Furthermore, the level of p-CaMKII/CaMKII was significantly higher in the PNS + BME group than
in the control group (p < 0.001) (Figure 9b).
Figure 8.
Exposure to BME treatment protects the PNS-induced alternations in the serotonergic system.
Quantitative real-time PCR analysis showing that exposure to BME treatment significantly ameliorates
the PNS-induced changes in (
a
) 5-HT
1A
and (
b
) 5-HT
2C
receptor expression. Data are expressed as
mean
±
SEM (n=6/group). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise
multiple comparisons, statistical significance is indicated by * p<0.05 and ** p<0.01).
3.3.4. Eect of Exposure to BME Treatment on Gestational Stress-Induced Changes on Neurogranin
(Ng) and CaMKII Signaling
Subsequently, we examined the eect of PNS on activation of CaMKII. The western blot analysis
demonstrated that the level of CaMKII was altered (Figure 9a, Supplementary Figure S7). The one-way
ANOVA reported a significant dierence across the variable groups (F
(2,17)
=132.10, p<0.001). The post
hoc comparison analysis showed that the level of p-CaMKII/CaMKII was significantly reduced in the
PNS group (p<0.001) as compared with the control and PNS +BME groups (p<0.001). Furthermore,
the level of p-CaMKII/CaMKII was significantly higher in the PNS +BME group than in the control
group (p<0.001) (Figure 9b).
Antioxidants 2020,9, 1229 14 of 24
1
Figure 9.
Exposure to BME treatment protects against PNS-induced changes in the activation of CaMKII.
(
a
) Representative Western blot showing variations in the level of CaMKII/p-CaMKII and Ng/p-Ng.
Estimated ratio of phosphorylation of CaMKII (
b
) and Ng/p-Ng (
c
) in experimental groups. Data are
expressed as mean
±
SEM (n=6/each group). One-way ANOVA followed by Bonferroni post hoc test
(for all pairwise multiple comparisons, statistical significance is indicated by * p<0.05, ** p<0.01 and
*** p<0.001).
Next, we examined whether the neurogranin expression was associated with the CaMKII
expression. Our analysis revealed that the level of neurogranin expression among the groups was
significantly dierent (F
(2,17)
=94.08, p<0.001). In addition, the post hoc analysis showed that the level
of p-Ng/Ng in the PNS group was significantly lower than in the control (p<0.01) and PNS +BME
groups (p<0.05). The comparison analysis showed that the level of p-Ng/Ng was significantly lower
in the PNS +BME group than in the control group (p<0.05) (Figure 9c). These analyses suggest that
PNS reduced the level of phosphorylation of Ng and CaMKII but the exposure to BME treatment
reduced the eect.
3.3.5. Eect of Exposure to BME Treatment on Gestational Stress-Induced Changes on
N-Methyl-D-Aspartate Receptors (2A,2B)
Exposure to BME treatment protects against the PNS-induced alteration in N-methyl-D-aspartate
receptors (Figure 10a, Supplementary Figure S8). The Western blot analysis exhibited the dierential
expression pattern in the N-methyl-D-aspartate receptors among the experimental groups. One-way
ANOVA showed that the level of NR2A varied significantly across the experimental groups
(F
(2,17)
=719.59, p<0.001), and the post hoc analysis suggested that the level of NR2A was significantly
low in PNS group than in the control (p<0.001) and PNS +BME groups (p<0.001). BME treatment
protects against the PNS-induced changes in NR2A expression, however, the level of NR2A was
significantly low in the PNS +BME group (p<0.001) (Figure 10b). Similarly, the NR2B level was
Antioxidants 2020,9, 1229 15 of 24
also altered and the variations among the groups were significantly dierent (F
(2,17)
=72.00, p<0.01).
The post hoc analysis revealed that the NR2B level was significantly lower in the PNS group than
in the control (p<0.001) and PNS +BME groups (p<0.05). Exposure to BME treatment protected
against the PNS-induced eect, however, the NR2B levels in the control and PNS +BME groups were
significantly dierence (p<0.001) (Figure 10c). The observed results suggest that exposure to BME
treatment had a resilience eect on the PNS-induced changes in N-methyl-D-aspartate receptors.
1
Figure 10.
Exposure to BME treatment protects against the PNS-induced changes in NR2A and NR2B.
(
a
) Representative Western blots showing that the level of NR2A and NR2B; (
b
) NR2A and (
c
) NR2B
levels were significantly altered by exposure to BME treatment. Data are expressed as mean
±
SEM
(n=6/each group). One-way ANOVA followed by Bonferroni post hoc test (for all pairwise multiple
comparisons, statistical significance is indicated by * p<0.05 and *** p<0.001).
3.3.6. Eect of Exposure to BME Treatment on Postnatal Stress-Induced Changes in Postsynaptic
Density Protein-95
Furthermore, our analysis showed PNS-induced changes in expression of postsynaptic density
protein-95 (Figure 11; Supplementary Figure S9), which varied significantly across the experimental
groups (F
(2,17)
=88.62, p<0.001). The post hoc analysis indicated that the level of postsynaptic density
protein-95 in the PNS group was significantly lower than in the control (p<0.01), and PNS +BME
groups (p<0.05). Furthermore, exposure to BME treatment elevated the postsynaptic density protein-95
level, which was not significantly dierent from the control group (p=0.14) (Figure 11a). Taken together,
these results demonstrated that exposure to BME treatment elevated the expression of postsynaptic
density protein 95.
Antioxidants 2020,9, 1229 16 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 17 of 26
together, these results demonstrated that exposure to BME treatment elevated the expression of
postsynaptic density protein 95.
Figure 11. Effect of exposure to BME treatment on postsynaptic density protein 95 (PSD-95) in PNS
offspring. (a) Representative Western blot depicting the level of PSD-95 in the experimental groups;
(b) Exposure to BME treatment significantly increased the expression of PSD-95. Data are expressed
as mean ± SEM (n = 6/each group). One-way ANOVA followed by Bonferroni post hoc test (for all
pairwise multiple comparisons statistical significance is indicated by * p < 0.05 and ** p < 0.01; NS—
Not significant).
3.3.7. Effect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Expression of
Pro and Mature Brain-Derived Neurotrophic Factor
The level of pro and mature brain-derived neurotrophic factor was altered by PNS (Figure 12a;
Supplementary Figure S10). One-way ANOVA found that ratio of pro and mature brain-derived
neurotrophic factor varied significantly across the experimental groups (F(2,17) = 132.10, p < 0.001). The
post hoc analysis demonstrated that the estimated ratio of pro and mature BDNF in the PNS group
was significantly lower as compared with the control (p < 0.05) and PNS + BME groups (p < 0.05).
Interestingly, a significant difference was not detected between the control and PNS + BME groups
(p = 0.495) (Figure 12b). the observed results suggest that PNS alters the conversion of mature brain-
derived neurotrophic factor but the exposure to BME treatment has a resilience effect on the PNS-
induced effect on brain-derived neurotrophic factor maturation.
Figure 11.
Eect of exposure to BME treatment on postsynaptic density protein 95 (PSD-95) in
PNS ospring. (
a
) Representative Western blot depicting the level of PSD-95 in the experimental
groups; (
b
) Exposure to BME treatment significantly increased the expression of PSD-95. Data are
expressed as mean
±
SEM (n=6/each group). One-way ANOVA followed by Bonferroni post hoc test
(for all pairwise multiple comparisons statistical significance is indicated by * p<0.05 and ** p<0.01;
NS—Not significant).
3.3.7. Eect of Exposure to BME Treatment on Prenatal Stress-Induced Changes in Expression of Pro
and Mature Brain-Derived Neurotrophic Factor
The level of pro and mature brain-derived neurotrophic factor was altered by PNS (Figure 12a;
Supplementary Figure S10). One-way ANOVA found that ratio of pro and mature brain-derived
neurotrophic factor varied significantly across the experimental groups (F
(2,17)
=132.10, p<0.001).
The post hoc analysis demonstrated that the estimated ratio of pro and mature BDNF in the PNS
group was significantly lower as compared with the control (p<0.05) and PNS +BME groups
(p<0.05). Interestingly, a significant dierence was not detected between the control and PNS +BME
groups (p=0.495) (Figure 12b). the observed results suggest that PNS alters the conversion of mature
brain-derived neurotrophic factor but the exposure to BME treatment has a resilience eect on the
PNS-induced eect on brain-derived neurotrophic factor maturation.
Antioxidants 2020,9, 1229 17 of 24
Antioxidants 2020, 9, x FOR PEER REVIEW 18 of 26
Figure 12. Effect of exposure to BME treatment on pro and mature brain-derived neurotrophic factor
(BDNF) in PNS offspring.(a) Representative Western blots depicting the level of pro BDNF and
mature BDNF; (b) Exposure to BME treatment silences the PNS-induced changes in the ratio of
proand mature BDNF. Data are expressed as mean ± SEM (n = 6/each group). One-way ANOVA
followed by Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is
indicated by * p < 0.05; NS—Not significant).
4. Discussion
Prenatal stress can alter the architecture of the developing brain program [51], more specifically
the amygdala region [52] by a mother’s physiological and emotional environment [53]. The outcomes
of animal and human studies have linked maternal stress and epigenetic changes; PNS has been
known to alter the methylation pattern of GR, and thereby influence the HPA axis regulation in
offspring and induce depression and anxiety-like behavior [54]. In this study, we observed that PNS
induced anxiety-like behavior in offspring, which was similar to earlier reports in other models
[55,56] and we validated our social defeat observation model. However, the observed anxiolytic
behavior in the PNS + BME group showed that exposure to BME treatment had a resilience effect on
prenatal stress and reduced the anxiety-like behavior in their offspring, as reported in other anxiety
animal models [31,32]. Exposure to stress during gestation has been known to alter the level of stress
hormone in the dam and also in the amniotic fluid [57], which can induce alterations in the HPA axis
in offspring [58]. When the level of CORT and ACTH was estimated, we found that the level of CORT
and ACTH was elevated in individuals experiencing PNS, as reported earlier [7]. However, exposure
Figure 12.
Eect of exposure to BME treatment on pro and mature brain-derived neurotrophic factor
(BDNF) in PNS ospring. (
a
) Representative Western blots depicting the level of pro BDNF and
mature BDNF; (
b
) Exposure to BME treatment silences the PNS-induced changes in the ratio of proand
mature BDNF. Data are expressed as mean
±
SEM (n=6/each group). One-way ANOVA followed by
Bonferroni post hoc test (for all pairwise multiple comparisons, statistical significance is indicated by
*p<0.05; NS—Not significant).
4. Discussion
Prenatal stress can alter the architecture of the developing brain program [
51
], more specifically
the amygdala region [
52
] by a mother’s physiological and emotional environment [
53
]. The outcomes
of animal and human studies have linked maternal stress and epigenetic changes; PNS has been known
to alter the methylation pattern of GR, and thereby influence the HPA axis regulation in ospring
and induce depression and anxiety-like behavior [
54
]. In this study, we observed that PNS induced
anxiety-like behavior in ospring, which was similar to earlier reports in other models [
55
,
56
] and
we validated our social defeat observation model. However, the observed anxiolytic behavior in the
PNS +BME group showed that exposure to BME treatment had a resilience eect on prenatal stress and
reduced the anxiety-like behavior in their ospring, as reported in other anxiety animal models [
31
,
32
].
Exposure to stress during gestation has been known to alter the level of stress hormone in the dam and
also in the amniotic fluid [
57
], which can induce alterations in the HPA axis in ospring [
58
]. When the
level of CORT and ACTH was estimated, we found that the level of CORT and ACTH was elevated
in individuals experiencing PNS, as reported earlier [
7
]. However, exposure to BME treatment had
a resilience eect on the PNS-induced changes and the level of CORT and ACTH was close to the basal
level in their ospring. The adoptogenic properties of BME possibly have a resilience eect on the
Antioxidants 2020,9, 1229 18 of 24
PNS-induced eect on CORT and ACTH [
32
,
59
]. We observed that the level of GR was up regulated
in individuals who experienced PNS, and exposure to BME treatment resulted in down regulation
of GR, which might be due to GR-CORT interactions [
60
,
61
]. In addition, it has been documented
that stress induced an elevated level of CORT, overexpression of the GR known to activate oxidative
mediators [
62
,
63
], and then up-regulation of the potential regulatory molecule caspase-3 in respond to
the oxidative stress [
63
,
64
] and neuronal damage [
65
]. Caspase-3 is the key eector molecule in the
caspase family, which is involved in the final execution of apoptosis, and activates the apoptosis process
through the signal conduction pathway mediated by caspase-3 [
66
,
67
]. We observed that the level of
caspase-3 was up regulated by PNS. Furthermore, we noted that exposure to BME treatment lowered
the caspase-3 expression, suggesting that BME possibly reduced the oxidative stress, as reported
earlier [
32
,
68
]. Interestingly, anti-apoptotic Bcl-2 was reduced by PNS, while exposure to BME treatment
increased Bcl-2 expression, which has been known to suppress apoptosis [
69
,
70
]. Our data clearly
demonstrate that the trends of caspase-3 and Bcl-2 expression are opposite. The levels of caspase-3 and
Bcl-2 were dynamically balanced in this model. These results support the neuroprotective eects of
BME in the PNS-exposed ospring and are consistent with those of another models [71,72].
The HPA axis receives serotonergic input, and its feedback mechanism it a critical regulator
of behavior [
73
75
]. In line with earlier reports with other stress models [
74
,
76
,
77
], a reduced level
of SYP was observed in the individuals who experienced PNS and displayed anxiety-like behavior.
Whereas, exposure to BME treatment restored the level of SYP and behavior; the changes in level of
SYP possibly associated with modulation of the HPA axis [
75
,
77
]. Notably, the HPA axis mediated
changes in SYP have been known to drive the changes in SYT-1 [
78
,
79
]. Similarly, we found that PNS
induced reduction of SYT-1 expression, however, exposure to BME treatment restored the level of
SYT-1 and their behavior. Similar to other animal models, the observed lower level of SYT-1 was
possibly associated with an imbalance in the synaptic vesicle exocytosis, which has been implicated in
regulating synaptic transmission, synaptic strength, and plasticity [
80
,
81
]. Taken together, these results
demonstrate that expression of SYP and SYT may be associated with synaptic plasticity and decreased
long-term potentiation (LTP). Earlier studies have demonstrated that synaptic transmission could
be mediated by Ca
2+
dependent SYT-1 interaction with synaptic protein and 5-HT receptors [
82
,
83
].
In addition, activation/inhibition of 5-HT receptors regulates the CRH expression [
84
]. Stress-induced
changes in synaptic transmission possibly induce adverse eects on serotonergic systems (level of 5-HT,
5-HT receptors, and SERT), aecting fetal development [
85
,
86
]. We found that the level of 5-HT
1A
receptor expression was significantly decreased, whereas the 5-HT
2C
receptor expression was elevated.
In basolateral amygdale, the 5-HT
1A
receptor was involving in neural circuits and its activation reduced
fear-related anxiety [
87
]. PNS-induced reduction in the 5-HT
1A
receptor could reflect the reactive
anxiety-like behavior [
88
]. Exposure to BME treatment facilitates activation of the 5-HT
1A
receptor
and is possibly related to the observed anxiolytic behavior [
18
,
20
]. Earlier studies on stress model
have demonstrated that up regulation of the 5-HT
2C
receptor contributed to increased response to
anxiety [
21
]. The underlying mechanism in the dierential expression of the 5-HT receptor may be
possibly associated with stress-mediated elevated level of corticosterone [
89
,
90
]. It is worthwhile
mentioning that exposure to BME treatment and the resilience eect on the PNS-induced changes in
5-HT1A and 5-HT2C receptors were similar to the other stress models [91].
The CaMKII has been known to be more sensitive to 5-HT transmission, intracellular calcium
level, and its kinase activity mediated by phosphorylation [
20
,
21
]. As reported by Fumagalli et al. [
92
],
we observed that PNS did not alter the total CaMKII, but the level of phosphorylation was decreased.
However, exposure to BME treatment elevated the phosphorylation level of CaMKII [
93
], which
subsequently activated the target genes and may be linked with the observed anxiolytic behavior.
Earlier studies have shown that at low concentrations or in the absence of Ca
2+
, Ng could bind with
calmodulin (CaM), which was linked with the activity-dependent synaptic plasticity and LTP,a substrate
for learning and memory [
94
,
95
]. In this study, we observed the level of Ng was significantly low in
the PNS group, but BME treatment protected against the eect of PNS and the individuals showed less
Antioxidants 2020,9, 1229 19 of 24
anxiety-like behavior and memory. Supporting our observation, earlier studies demonstrated that stress
reduced the level of Ng and led to a reduction in LTP and a functional deficit in spatial learning [
96
,
97
].
The observed behavioral phenotype in this study could be linked with the PNS-induced reduction in
Ng and CaMKII.
As reported earlier, PNS down regulates the NMDA receptor expression (NR2A, 2B) [
98
,
99
],
possibly through the interaction of p-CaMKII [
23
]. It should be noted that exposure to BME treatment
and the resilience eect on the PNS-induced eects on NR2A and NR2B receptor expression may be
correlated with the observed anxiolytic behavior. Subsequently, we found that the PNS mediated
eect on PSD-95 and further exposure to BME treatment increased the level of PSD-95, possibly due to
the dierential eects mediated expression [
24
]. Exposure to the stress has been known to alter the
BDNF level either directly by glucocorticoid level or through the signaling molecules [
7
]. We found
that the level of proand mature BDNF was decreased in the amygdala of ospring that experienced
PNS, similar to another prenatal study report on hippocampus [
100
]. Stress reduced the proteolytic
conversion of pro-BDNF to mature BDNF; therefore, the lower level of BDNF estimated in individuals
who experienced PNS [
27
]. However, exposure to BME treatment increased the level of pro-BDNF,
as well as the mature BDNF level. The observed mechanism in the present study could be the eect of
BME treatment mediating BNDF level, as in earlier reports on other models [32,101].
5. Conclusions
In conclusion, behavioral data suggest that PNS induces anxiety-like behavior in ospring which
may be linked to an alteration in the HPA axis (CORT, ACTH, GR) and overexpression of GR, activating
oxidative stress and possibly the neuronal damage. Interestingly, exposure to BME treatment balances
caspase-3 and Bcl-2 expression to protect the neurons. In addition, the feedback mechanism of the
HPA axis and the serotonergic system alter synaptic proteins (synaptophysin, synaptotagmin-1),
5-HT receptors, phosphorylation of Ng and its interaction CaMKII, NMDA receptors (2A, 2B), PSD-95,
and conversion of pro and mature BDNF. Exposure to BME treatment protects against the PNS-induced
neuronal damage and changes in proand mature BDNF through the HPA axis and synaptic protein
mediated signaling, which may be linked with the observed anxiolytic behavior in ospring.
Supplementary Materials:
The following are available online at http://www.mdpi.com/2076-3921/9/12/1229/s1.
Supplementary Data-1: High performance liquid chromatography (HPLC) chromatogram of Bacopa monnieri
extract, Supplementary Information: Supplementary figures showing all the uncropped full Western blot images.
Supplementary Data for Western blot: Representative Western blots each band trace quantity.
Author Contributions:
K.E.R. designed the experiment; K.S. performed the experiment, K.S. and K.E.R. analyzed
the data and prepared the manuscript. All authors have read and agreed to the published version of the manuscript.
Funding:
K.S. is a recipient of a junior research fellowship through DST-Promotion of University Research
and Scientific Excellence (PURSE). DST-FIST is supporting the Instrumentation Facility at the Department of
Animal Science.
Acknowledgments: We thank Hemant Singh for his generous gift of Bacopa monnieri extract (CDRI-08).
Conflicts of Interest: The authors declare that they have no conflict of interest.
References
1.
Newport, D.J.; Stowe, Z.N.; Nemero, C.B. Parental depression, animal models of an adverse life event.
Am. J. Psychiatry 2002,159, 1265–1283. [CrossRef] [PubMed]
2.
Mueller, B.R.; Bale, T.L. Sex-specific programming of ospring emotionality after stress early in pregnancy.
J. Neurosci. 2008,28, 9055–9065. [CrossRef] [PubMed]
3.
Cabrera-Vera, T.M.; Battaglia, G. Prenatal exposure to fluoxetine (Prozac) produces site-specific and
age-dependent alterations in brain serotonin transporters in rat progeny, evidence from autoradiographic studies.
J. Pharmacol. Exp. Ther. 1998,286, 1474–1481. [PubMed]
4.
Forcelli, P.A.; Heinrichs, S.C. Teratogenic eects of maternal antidepressant exposure on neural substrates of
drug-seeking behavior in ospring. Addict. Biol. 2008,13, 52–62. [CrossRef] [PubMed]
Antioxidants 2020,9, 1229 20 of 24
5.
Zhang, J.; Fan, Y.; Li, Y.; Zhu, H.; Wang, L.; Zhu, M.Y. Chronic social defeat up-regulates expression of the
serotonin transporter in rat dorsal raphe nucleus and projection regions in a glucocorticoid-dependent manner.
J. Neurochem. 2012,123, 6. [CrossRef]
6.
Miyagawa, K.; Tsuji, M.; Ishii, D.; Takeda, K.; Takeda, H. Prenatal stress induces vulnerability to stress
together with the disruption of central serotonin neurons in mice. Behav. Brain Res.
2015
,277, 228–236.
[CrossRef] [PubMed]
7.
Wei, S.; Li, Z.; Ren, M.; Wang, J.; Gao, J.; Guo, Y.; Xu, K.; Li, F.; Zhu, D.; Zhang, H.; et al. Social defeat
stress before pregnancy induces depressive-like behaviours and cognitive deficits in adult male ospring,
correlation with neurobiological changes. BME Neurosci. 2018,19, 1–22. [CrossRef]
8.
Popoli, M.; Yan, Z.; McEwen, B.S.; Sanacora, G. The stressed synapse, the impact of stress and glucocorticoids
on glutamate transmission. Nat. Rev. Neurosci. 2012,13, 22–37. [CrossRef]
9. Weinstock, M. Prenatal stressors in rodents, eects on behavior. Neurobiol. Stress 2017,6, 3–13. [CrossRef]
10.
Wang, W.; Xu, J.; Li, L.; Wang, P.; Ji, X.; Ai, H.; Zhang, L.; Li, L. Neuroprotective eect of morroniside on focal
cerebral ischemia in rats. Brain Res. Bull. 2010,83, 196–201. [CrossRef]
11.
Sasaki, T.; Kitagawa, K.; Yagita, Y.; Sugiura, S.; Omura-Matsuoka, E.; Tanaka, S.; Matsushita, K.; Okano, H.;
Tsujimoto, Y.; Hori, M. Bcl2 enhances survival of newborn neurons in the normal and ischemic hippocampus.
Neurosci. Res. 2006,84, 1187–1196. [CrossRef] [PubMed]
12.
Price, M.L.; Curtis, A.L.; Kirby, L.G.; Valentino, R.J.; Lucki, I. Eects of corticotropin-releasing factor on brain
serotonergic activity. Neuropsychopharmacology 1998,18, 492–502. [CrossRef]
13.
Higuchi, Y.; Soga, T.; Parhar, I.S. Regulatory Pathways of Monoamine Oxidase A during Social Stress.
Front. Neurosci. 2017,11, 604. [CrossRef] [PubMed]
14.
Xu, Y.; Li, S.; Vernon, M.M.; Pan, J.; Chen, L.; Barish, P.A.; Zhang, Y.; Acharya, A.P.; Yu, J.;
Govindarajan, S.S.; et al. Curcumin prevents corticosterone-induced neurotoxicity and abnormalities
of neuroplasticity via 5-HT receptor pathway. J. Neurochem. 2011,118, 784–795. [CrossRef] [PubMed]
15.
Jiang, L.X.; Huang, G.D.; Su, F.; Wang, H.; Zhang, C.; Yu, X. Vortioxetine administration attenuates cognitive
and synaptic deficits in 5×FAD mice. Psychopharmacology 2020,237, 1233–1243. [CrossRef]
16.
Gassowska, M.; Baranowska-Bosiacka, I.; Moczydlowska, J.; Frontczak-Baniewicz, M.; Gewartowska, M.;
Struzynska, L.; Gutowska, I.; Chlubek, D.; Adamczyk, A. Perinatal exposure to lead (Pb) induces
ultrastructural and molecular alterations in synapses of rat ospring. Toxicology
2016
,373, 13–29.
[CrossRef] [PubMed]
17.
Cui, Y.; Cao, K.; Lin, H.; Cui, S.; Shen, C.; Wen, W.; Mo, H.; Dong, Z.; Bai, S.; Yang, L.; et al. Early-Life Stress
Induces Depression-Like Behavior and Synaptic-Plasticity Changes in a Maternal Separation Rat Model,
Gender Dierence and Metabolomics Study. Front. Pharmacol. 2020,11, 102. [CrossRef]
18.
Bader, L.R.; Carboni, J.D.; Burleson, C.A.; Cooper, M.A. 5-HT1A receptor activation reduces fear-related
behaviour following social defeat in Syrian hamsters. Pharmacol. Biochem. Behav.
2014
,122, 182–190. [CrossRef]
19.
Akatsu, S.; Ishikawa, C.; Takemura, K.; Ohtani, A.; Shiga, T. Eects of prenatal stress and neonatal
handling on anxiety, spatial learning and serotonergic system of male ospring mice. Neurosci. Res.
2015
,
101, 15–23. [CrossRef]
20.
Mikhalienko, V.A.; Butkevich, I.P. Prenatal stimulation of 5-HT 1A receptors improves adaptive behaviour in
prenatally stressed rats. Bull. Exp. Biol. Med. 2019,166, 306–330. [CrossRef]
21.
Heisler, L.K.; Zhou, L.; Bajwa, P.; Hsu, J.; Tecott, L.H. Serotonin 5-HT2C receptors regulate
anxiety-like behaviour. Genes Brain Behav. 2007,6, 491–496. [CrossRef]
22.
Purkayastha, S.; Ford, J.; Kanijilal, B.; Diallo, S.; Inigo, D.R.J.; Neuwirth, L.; El Idrissi, A.; Ahmed, Z.;
Wierasko, A.; Azmitia, E.C.; et al. Clozapine functions through the prefrontal cortex serotonin 1A receptor
to heighten neuronal activity via calmodulin kinase II–NMDA receptor interactions. J. Neurochem.
2012
,
120, 396–407. [CrossRef]
23.
Sanhueza, M.; Fernndez-Vilalobos, G.; Stein, I.S.; Kasumova, G.; Zhang, P.; Bayer, K.U.; Otmakhov, N.;
Hell, J.W.; Lisman, J. Role of the CaMKII/NMDA receptor complex in the maintenance of synaptic strength.
J. Neurosci. 2011,31, 9170–9178. [CrossRef] [PubMed]
24.
Incontro, S.; Diaz-Alonso, J.; Lafrati, J.; Vieria, M.; Asensio, C.S.; Sohal, V.S.; Roche, K.W.; Bender, K.J.;
Nicoll, R.A. The CaMKII/NMDA receptor complex control hippocampal synaptic transmission by
kinase-dependent and independent mechanism. Nat. Commun. 2018,9, 2069. [CrossRef] [PubMed]
Antioxidants 2020,9, 1229 21 of 24
25.
Xie, M.J.; Ishikawa, Y.; Yagi, H.; Iguchi, T.; Oka, Y.; Kuroda, K.; Iwata, K.; Kiyonari, H.; Matsuda, S.;
Matsuzaki, H.; et al. PIP3-PhIdb2 is crucial for LTP regulating synaptic NMDA and AMPA receptor density
and PSD95 turnover. Sci. Rep. 2019,9, 4305. [CrossRef]
26.
Yeh, C.M.; Huang, C.C.; Hsu, K.S. Prenatal stress alters hippocampal synaptic plasticity in young rat
ospring through preventing the proteolytic conversion of pro-brain-derived neurotrophic factor (BDNF) to
mature BDNF. J. Physiol. 2012,590, 991–1010. [CrossRef]
27.
Zheng, Y.; Fan, W.; Zhang, W.; Dong, E. Gestational stress induces depressive-like and anxiety phenotypes
through epigenetic regulation of BDNF expression in ospring hippocampus. Epigenetics
2016
,11, 150–162.
[CrossRef] [PubMed]
28.
Chatterji, N.; Rastogi, R.P.; Dhar, M.L. Chemical examination of Bacopa monniera Wettst. The constitution of
Bacoside A. Indian J. Chem. 1965,3, 24–29.
29.
Basu, N.; Rastogi, R.P.; Dhar, M.L. Chemical examination of Bacopa monniera, Wettst: Part III Bacoside B.
Indian J. Chem. 1967,5, 84–86.
30.
Singh, H.K.; Rastogi, R.P.; Srimal, R.C.; Dhawan, B.N. Eect of bacoside A and B on avoidance responses
in rats. Phytother. Res. 1988,2, 70–75. [CrossRef]
31.
Banerjee, R.; Hazra, S.; Ghosh, A.K.; Mondal, A.C. Chronic administration of Bacopa monniera increases
BDNF protein and mRNA expressions, a study in chronic unpredictable stress induced animal model
of depression. Psychiatry Investig. 2014,11, 3. [CrossRef]
32.
Kumar, S.; Mondal, A.C. Neuroprotective, neurotrophic and anti-oxidative role of Bacopa monnieri on CUS
induced model of depression in rat. Neurochem. Res. 2016,41, 11. [CrossRef]
33.
Calabrese, C.; Gregory, W.L.; Leo, M.; Kraemer, D.; Bone, K.; Oken, B. Eects of standardized Bacopa monnieri
extract on congnitive performance, anxiety, and depression in the elderly, a randomized, double–blind,
placebo-controlled trail. J. Altern. Complement. Med. 2008,14, 707–713. [CrossRef]
34.
Benson, S.; Downey, L.A.; Stough, C.; Wetherell, M.; Zangara, A.; Scholey, A. An acyte, double-blind,
placebo-controlled cross-over study of 320 mg and 640 mg doses of Bacopa monnieri (CDRI 08) on multitasking
stress reactivity and mood. Phytother. Res. 2014,28, 551–559. [CrossRef]
35.
Liu, J.; Mori, A. Stress, aging, and brain oxidative damage. Neurochem. Res.
1999
,24, 1479–1497. [CrossRef]
36. Plaza-Briceño, W.; Estay, S.F.; de la Fuente-Ortega, E.; Gutierrez, C.; Sanchez, G.; Hidalgo, C.; Chavez, A.E.;
Haeger, P.A. N-Methyl-d-Aspartate Receptor Modulation by Nicotinamide Adenine Dinucleotide Phosphate
Oxidase Type 2 Drives Synaptic Plasticity and Spatial Memory Impairments in Rats Exposed Pre- and
Postnatally to Ethanol. Antioxid. Redox Signal. 2020,32, 602–617. [CrossRef]
37.
Patel, S.K.; Singh, S.; Singh, H.K.; Singh, S.K. Eect of standardized extract of Bacopa monnieri (CDRI-08) on
testicular functions in adult male mice. J. Ethnopharmacol. 2017,197, 101–109. [CrossRef]
38.
Sfikakis, A.; Galanopoulou, P.; Konstandi, M.; Tsakayannis, D. Stress through handling for vaginal screening,
serotonin, and ACTH response to ether. Pharmacol. Biochem. Behav. 1996,53, 965–970. [CrossRef]
39.
Arakawa, H. Ethological approach to social isolation eects in behavioral studies of laboratory rodents.
Behav. Brain Res. 2018,341, 98–108. [CrossRef]
40. Lee, Y.A.; Kim, Y.J.; Goto, Y. Cognitive and aective alterations by prenatal and postnatal stress interaction.
Physiol. Behav. 2016,165, 146–153. [CrossRef]
41.
Walf, A.; Cheryl, A.F. The use of the elevated plus maze as as assay of anxiety-related behaviour in rodents.
Nat. Protoc. 2007,2, 322. [CrossRef]
42.
Conrad, C.D.; Galea, L.A.; Kuroda, Y.; McEwen, B.S. Chronic stress impairs rat spatial memory on the Y
maze and this eect is blocked by tianeptine pretreatment. Behav. Neurosci.
1996
,110, 1321–1334. [CrossRef]
43.
Mohri, M.; Rezapoor, H. Eects of heprin citrate and EDTA plasma biochemistry of sheep comparison with
serum. Res. Vet. Sci. 2009,86, 111–114. [CrossRef] [PubMed]
44.
Zapala, M.A.; Hovatta, I.; Ellison, J.A.; Wodicka, L.; Del Rio, J.A.; Tennant, R.; Tynan, W.; Broide, R.S.;
Helton, R.; Stoveken, B.S.; et al. Adult mouse brain gene expression patterns bear an embryologic imprint.
Proc. Natl. Acad. Sci. USA 2005,102, 10357–10362. [CrossRef] [PubMed]
45.
Preethi, J.; Singh, H.K.; Charles, P.D.; Rajan, K.E. Participation of microRNA124-CREB pathway, a parallel
memory enhancing mechanism of standardized extract of Bacopa moniera (BESEB CDRI-08). Neurochem. Res.
2012,10, 2167–2177. [CrossRef] [PubMed]
Antioxidants 2020,9, 1229 22 of 24
46.
Ganesh, A.; Bogdanowicz, W.; Haupt, M.; Marimuthu, G.; Rajan, K.E. Role of olfactory bulb serotonin in
olfactory learning in the greater short-nosed fruit bat, Cynopterus sphinx (Chiroptera, Pteropodidae). Brain Res.
2010,1352, 108–117. [CrossRef] [PubMed]
47.
Mariappan, S.; Bogdanowicz, W.; Raghuram, H.; Marimuthu, G.; Rajan, K.E. Structure of distress call,
implication for specificity and activation of dopaminergic system. J. Comp. Physiol. A
2016
,202, 55–65.
[CrossRef] [PubMed]
48. Bradford, M.M. A rapid and sensitive method for quantitation of microgram quantities of protein utilizing
the principle of protein-dye-binding. Anal. Biochem. 1976,72, 248–254. [CrossRef]
49.
Raguvarman, D.; Rajan, K.E. Environmental enrichment reduces anxiety by dierently activating serotonergic
and neuropeptide Y (NPY)-ergic system in Indian field mouse (Musbooduga), an animal model for
post-traumatic stress disorder. PLoS ONE 2015,10, e0127945. [CrossRef]
50.
Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and
the 2(-Delta DeltaC(T)) method. Methods 2001,25, 402–408. [CrossRef]
51.
Bock, J.; Rether, K.; Groger, N.; Xie, L.; Braun, K. Perinatal programming of emotional brain circuits,
An integrative from from system to moldecules. Front. Neurosci. 2014,8, 1–16. [CrossRef] [PubMed]
52.
Buss, C.; Davis, E.P.; Shahbaba, B.; Pruessner, J.C.; Head, K.; Sandman, C.A. Maternal cortisol over the course
of pregnancy and subsequent child amygdala and hippocampus volumes and aective problem. Proc. Natl.
Acad. Sci. USA 2012,109, E1312–E1319. [CrossRef] [PubMed]
53.
Beijers, R.M.; Buitelaar, J.K.; de Weerth, C. Mechanisms underlying the eects of prenatal psychological
stress on child outcomes, Beyond the HPA axis. Eur. Child. Adolesc. Psychiatry
2014
,23, 943–956. [CrossRef]
54.
Radtke, K.M.; Ruf, M.; Gunter, H.M.; Dohrmann, K.; Schauer, M.; Meyer, A.L.; Elbert, T. Transgenerational
impact of intimate partner violence on methylation in the promoter of the glucocorticod receptor.
Trans. Psychiatry 2011,1, e21. [CrossRef]
55.
Wilson, C.A.; Vazdarjanova, A.; Terry, A.V., Jr. Exposure to variable prenatal stress in rats, eets on
anxiety-related behaviours, innate and contextual fear and fear extinction. Behav. Brain Res.
2013
,
238, 279–288. [CrossRef]
56.
Matrisciano, F.; Tueting, P.; Dalal, I.; Kadriu, B.; Grayson, D.R.; Davis, J.M.; Nicoletti, F.; Guidotti, A.
Epigenetic modifications of GABAergic interneurons are associated with the schizophrenia-like phenotype
induced by prenatal stress in mice. Neuropharmacology 2013,68, 184–194. [CrossRef]
57.
Aziz, N.A.; Kendall, D.A.; Pardon, M.C. Prenatal exposure to chronic mild stress increases corticosterone
levels in the amniotic fluid and induces cognitive deficits in female ospring, improved by treatment with
the antidepressant drug amitriptyline. Behav. Brain Res. 2012,231, 29–39. [CrossRef]
58.
Drake, A.J.; Tang, J.I.; Nyirenda, M.J. Mechanisms underlying the role of glucocorticoids in the early life
programming of adult disease. Clin. Sci. 2007,113, 219–232. [CrossRef]
59.
Sheikh, N.; Ahmad, A.; Siripurapu, K.B.; Kuchibhotla, V.K.; Singh, S.; Palit, G. Eect of Bacopa monniera on
stress induced changes in plasma corticosterone and brain monoamines in rats. J. Ethnopharmacol.
2007
,111,
671–676. [CrossRef] [PubMed]
60.
Lightman, S.L.; Conway-Campbell, B.L. The crucial role of pulsatile activity of the HPA axis for continuous
dynamic equilibration. Nat. Rev. Neurosci. 2010,11, 710–718. [CrossRef] [PubMed]
61.
Hapgood, J.P.; Avenant, C.; Moliki, J.M. Glucocorticoid-independent modulation of GR activity: Implications
for immunotherapy. Pharmacol. Ther. 2016, 165. [CrossRef]
62.
P
é
rez-Nievas, B.G.; Garc
í
a-Bueno, B.; Caso, J.R.; Mench
é
n, L.; Leza, J.C. Corticosterone as a marker of
susceptibility to oxidative/nitrosative cerebral damage after stress exposure in rats. Psychoneuroendocrinology
2007,32, 703–711. [CrossRef]
63.
Orlovsky, M.A.; Dosenko, V.E.; Spiga, F.; Skiibo, G.C.; Lightman, S.L. Hippocampus remodeling by chronic
stress accompanied by GR, proteasome and caspase-3 overexpression. Brain Res.
2014
,1593, 83–94. [CrossRef]
64.
Filipovic, D.; Zlatkovic, J.; Inta, D.; Bjelobaba, I.; Stojiljkovic, M.; Gass, P. Chronic isolation stress predisposes
the frontal cortex but not the hippocampus to the potentially detrimental release of cytochrome c from
mitochondria and the activation of caspase-3. J. Neurosci. Res. 2011,89, 9. [CrossRef]
65.
Höschl, C.; Hajek, T. Hippocampal damage mediated by corticosteroids—A neuropsychiatric
research challenge. Eur. Arch. Psychiatry Clin. Neurosci. 2001,251, 81–88. [CrossRef]
66.
Zhang, D.; Wang, Z.; Sheng, C.; Peng, W.; Hui, S.; Gong, W.; Chen, S. Icarin prevents amyloid beta-induced
via the PI3K/Akt pathway in PC-12 cells. Evid. Based Complement. Altern. Med.
2015
,2015, 235265. [CrossRef]
Antioxidants 2020,9, 1229 23 of 24
67.
Means, J.C.; Venkatesan, A.; Gerdes, B.; Fan, J.Y.; Bjes, E.S.; Price, J.L. Drosophila spaghetti and doubletime link
the circadian clock and light to caspases, apoptosis and tauopathy. PLoS Genet.
2015
,11, e1005171. [CrossRef]
68.
Krishna, G.; Hosamani, R. Muralidhara Bacopa monnieri supplements oset paraquat-induced behavioural
phenotype and brain oxidative pathways in mice. Cent. Nerv. Syst. Agents Med. Chem.
2019
,
19, 57–66. [CrossRef]
69.
Siddiqui, W.A.; Ahad, A.; Ahsan, H. The mystery of BCL2 family, Bcl2 proteins and apoptosis, an update.
Arch. Toxicol. 2015,89, 289–317.
70.
Hata, A.N.; Engelman, J.A.; Faber, A.C. The BCL2 family, key mediators of the apoptotic response to targeted
anticancer therapeutics. Cancer Discov. 2015,5, 475–487. [CrossRef]
71.
Mondal, P.; Trigun, S.K. Bacopa monnieri Extract (CDRI-08) modulates the NMDA receptor subunits and
nNOS-apoptosis axis in cerebellum of hepatic encephalopathy rats. Evid. Based Complement. Altern. Med.
2015, 535013. [CrossRef]
72.
Singh, B.; Pandey, S.; Yadav, S.K.; Verma, R.; Singh, S.P.; Mahdi, A.A. Role of ethanolic extract of Bacopa
monnieri against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced mice model via inhibition
of apoptotic pathways of dopaminergic neurons. Brain Res. Bull. 2017,135, 120–128. [CrossRef] [PubMed]
73.
Andrews, M.H.; Matthews, S.G. Programming of the hypothalamo-pituitary-adrenal axis, serotonergic
involvement. Stress 2004,7, 15–27. [CrossRef] [PubMed]
74.
Gemmel, M.; Rayen, I.; Lotus, T.; van Donkelaar, E.; Steinbusch, H.W.; De Lacalle, S.; Kokras, N.;
Dalla, C.; Pawluski, J.L. Developmental fluoxetine and prenatal stress eects on serotonin, dopamine,
and synaptophysin density in the PFC and hippocampus of ospring at weaning. Dev. Psychobiol.
2016
,
58, 315–327. [CrossRef]
75.
Gemmel, M.; Kokras, N.; Dalla, C.; Pawluski, J.L. Perinatal fluoxetine prevents the eect of pre-gestational
maternal stress on 5-HT in the PFC, but maternal stress has enduring eects on mPFC synaptic structure
in ospring. Neuropharmacology 2018,128, 168–180. [CrossRef]
76.
Flak, J.N.; Ostrander, M.M.; Tasker, J.G.; Herman, J.P. Chronic stress-induced neurotransmitter plasticity in
the PVN. J. Comp. Neurol. 2009,517, 156–165. [CrossRef]
77.
Hescham, S.; Grace, L.; Kellaway, L.A.; Bugarith, K.; Russell, V.A. Eect of exercise on synaptophysin and
calcium/calmodulin-dependent protein kinase levels in prefrontal cortex and hippocampus of a rat model of
developmental stress. Metab. Brain Dis. 2009,24, 701–709. [CrossRef]
78.
Ge, J.F.; Qi, C.C.; Zhou, J.N. Imbalance of leptin pathway and hypothalamus synaptic plasticity markers are
associated with stress-induced depression in rats. Behav. Brain Res. 2013,249, 38–43. [CrossRef]
79.
Ge, J.F.; Xu, Y.Y.; Qin, G.; Peng, Y.N.; Zhang, C.F.; Liu, X.R.; Liang, L.C.; Wang, Z.Z.; Chen, F.H. Depression-like
Behavior Induced by Nesfatin-1 in Rats, Involvement of Increased Immune Activation and Imbalance of
Synaptic Vesicle Proteins. Front. Neurosci. 2015,9, 429. [CrossRef]
80.
Bhattacharyya, S.; Biou, V.; Xu, W.; Schluter, O.; Malenka, R.C. A critical role for PSD-95/AKAP interactions
in endocytosis of synaptic AMPA receptors. Nat. Neurosci. 2009,12, 172–181. [CrossRef]
81.
Chai, G.S.; Jiang, X.; Ni, Z.F.; Ma, Z.W.; Xie, A.J.; Cheng, X.S.; Wang, J.Z.; Liu, G.P. Betaine attenuates
Alzheimer-like pathological changes and memory deficits induced by homocysteine. J. Neurochem.
2013
,124,
388–396. [CrossRef] [PubMed]
82.
Zurawski, Z.; Rodriguez, S.; Hyde, K.; Alford, S.; Hamm, H.E. G
βγ
Binds to the Extreme C Terminus of
SNAP25 to Mediate the Action of Gi/o-Coupled G Protein-Coupled Receptors. Mol. Pharmacol.
2016
,89,
75–83. [CrossRef] [PubMed]
83.
Alford, S.; Hamm, H.; Rodriguez, S.; Zurawski, Z. G
βγ
SNARE Interactions and Their Behavioral Eects.
Neurochem. Res. 2019,44, 636–649. [CrossRef]
84.
Jørgensen, H.; Knigge, U.; Kjær, A.; Møller, M.; Warberg, J. Serotonergic stimulation of corticotropin-releasing
hormone and pro-opiomelanocortin gene expression. J. Neuroendocrinol.
2002
,14, 788–795. [CrossRef] [PubMed]
85.
Peters, D.A. Maternal stress increases fetal brain and neonatal cerebral cortex 5-hydroxytryptamine synthesis
in rats, a possible mechanism by which stress influences brain development. Pharmacol. Biochem. Behav.
1990
,
35, 943–947. [CrossRef]
86.
Van den Hove, D.L.A.; Lauder, J.M.; Scheepens, A.; Prickaerts, J.H.; Blanco, C.E.; Steinbusch, H.W.M.
Prenatal stress in the rat alters 5-HT1A receptor binding in the ventral hippocampus. Brain Res.
2006
,
1090, 29–34. [CrossRef]
Antioxidants 2020,9, 1229 24 of 24
87.
Morrison, K.E.; Cooper, M.A. A role for 5-HT1A receptors in the basolateral amygdala in the development of
conditioned defeat in Syrian hamsters. Pharmacol. Biochem. Behav. 2012,10, 3. [CrossRef]
88.
Akimova, E.; Lanzenberger, R.; Kasper, S. The serotonin-1A receptor in anxiety disorders. Biol. Psychiatry
2009,66, 7. [CrossRef]
89.
Fairchild, G.; Leitch, M.M.; Ingram,C.D. Acute and chronic effects of corticosterone on 5-HT1A receptor-mediated
autoinhibition in the rat dorsal raphe nucleus. Neuropharmacology 2003,45, 925–934. [CrossRef]
90.
Lanfumey, L.; Mongeau, R.; Cohen-Salmon, C.; Hamon, M. Corticosteroid-serotonin interactions in the
neurobiological mechanisms of stress-related disorders. Neurosci. Biobehav. Rev.
2008
,32, 1174–1184. [CrossRef]
91.
Krishnakumar, A.; Anju, T.R.; Abraham, P.M.; Paulose, C.S. Alteration in 5-HT 2C, NMDA receptor and IP3
in cerebral cortex of epileptic rats, restorative role of Bacopa monnieri.Neurochem. Res.
2015
,40, 216–225.
[CrossRef] [PubMed]
92.
Fumagalli, F.; Pasini, M.; Frasca, A.; Drago, F.; Racagni, G.; Riva, M.A. Prenatal stress alters glutamatergic
system responsiveness in adult rat prefrontal cortex. J. Neurochem.
2009
,109, 1733–1744. [CrossRef] [PubMed]
93.
Matsumoto, Y.; Morinobu, S.; Yamamoto, S.; Matsumoto, T.; Takei, S.; Fujita, Y.; Yamawaki, S.
Vorinostat ameliorates impaired fear extinction possibly via the hippocampal NMDA-CaMKII pathway in
an animal model of posttraumatic stress disorder. Psychopharmacology 2013,229, 1. [CrossRef] [PubMed]
94.
Gerendasy, D.G.; Sutclie, J.G. RC3/neurogranin, a postsynaptic calpacitin forsetting the response threshold
to calcium influxes. Mol. Neurobiol. 1997,15, 131–163. [CrossRef] [PubMed]
95.
Huang, K.P.; Huang, F.L.; Jager, T.; Li, J.; Reymann, K.G.; Balschun, D. Neurogranin/RC3 enhances
long-term potentiation and learning by promoting calcium-mediated signaling. J. Neurosci.
2004
,
24, 10660–10669. [CrossRef]
96.
Li, Q.; Cheng, D.; Chen, R.; Cai, Q.; Jia, N.; Su, Q.; Zhang, H.; Zhu, Z.; Zeng, J.; Li, H. Expression of
neurogranin in hippocampus of rat ospring exposed to restraint stress and pulsed magnetic fields. Brain Res.
2014,1570, 26–34. [CrossRef]
97.
Jones, K.J.; Templet, S.; Zemoura, K.; Kuzniewska, B.; Pena, F.X.; Hwang, H.; Lei, D.J.; Haensgen, H.;
Nguyen, S.; Saenz, C.; et al. Rapid, experience-dependent translation of neurogranin enables
memory encoding. Proc. Natl. Acad. Sci. USA 2018,115, E5805–E5814. [CrossRef]
98.
Huang, Y.; Shi, X.; Xu, H.; Yang, H.; Chen, T.; Chen, S.; Chen, X. Chronic unpredictable stress before
pregnancy reduce the expression of brain-derived neurotrophic factor and N-methyl-D-aspartate receptor in
hippocampus of ospring rats associated with impairment of memory. Neurochem. Res.
2010
,35, 7. [CrossRef]
99.
Sun, H.; Guan, L.; Zhu, Z.; Li, H. Reduced levels of NR1 and NR2A with depression-like behaviour in
dierent brain regions in prenatally stressed juvenile ospring. PLoS ONE 2013,8, e81775. [CrossRef]
100.
Badihian, N.; Daniali, S.S.; Kelishadi, R. Transcriptional and epigenetic changes of brain derived neurotrophic
factor following prenatal stress: A systematic review of animal studies. Neurosci. Biobehav. Rev.
2019
. [CrossRef]
101.
Preethi, J.; Singh, H.K.; Rajan, K.E. Possible involvement of standardized Bacopa monniera extract (CDRI-08)
in epigenetic regulation of reelin and brain-derived neurotropic factor to enhance memory. Front. Pharmacol.
2016,7, 166. [CrossRef] [PubMed]
Publisher’s Note:
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional
aliations.
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Subsequently, radiopharmaceutical analysis confirmed the biodistribution of bacosides in the brain [30]. Earlier, we have shown that supplementation of BME improved memory by acting through the serotonergic system [31], regulates signaling molecules through microRNA (miR)-124 [32], epigenetic modifications [33,34], and regulates the synaptic plasticity [35]. Earlier studies reported that B. monnieri supplementation enhances cognitive performance in patients with neurodegenerative disorders [36][37][38][39]. ...
... Earlier studies reported that B. monnieri supplementation enhances cognitive performance in patients with neurodegenerative disorders [36][37][38][39]. Studies in animal models and in elderly people demonstrated that supplementation of B. monnieri extract (CDRI-08) reduces oxidative stress, depression, and anxiety-like behaviors [35,[40][41][42]. Carnosine (β-alanyl-L-histidine) is synthesized from β-alanine and L-histidine by the enzyme carnosine synthase [43] and is an effective scavenger of reactive oxygen species and aldehydes. ...
... The pregnant rats were subjected to social defeat (SD) to induce social stress during GD-16 to GD-18 in the specially designed SD cage to induce maternal stress (MS) [35,48]. SD cage constructed with two chambers [intruder chamber (IC), resident chamber (RC)] with equal size (30 × 30 cm). ...
Article
Full-text available
Maternal stress (MS) during gestation is known to increase the risk for the development of behavioural and physiological disorders and advances cellular aging. In this study, we investigated whether the supplementation of standardized Bacopa monnieri extract (CDRI-08/BME) or l-Carnosine (L-C) to the mother exposed to social stress during gestation modify the effect on their offspring's neurobehaviour, antioxidant defence gene expression, telomere length, and telomere biology. To test this, timed pregnant rats were subjected to social stress during the gestational day (GD) 16–18. A subset of stressed pregnant rats received either BME [80 mg/kg in 0.5% gum acacia (per-orally; p.o)] or L-C [1 mg/kg (p.o)] every day from GD-10 to until their pup's attained postnatal day (PND)-23. We observed that MS induced anxiety-like behaviour, altered inter-limb coordination, antioxidant defence genes [Superoxide dismutase (SOD1,2), Catalase (CAT), Glutathione peroxidase-3 (GPX3)], telomerase reverse transcriptase (TERT), shelterin complex subunits (TRF1, RAP1B, POT1) protein level and shorten telomere length. Notably, supplementation of BME/L-C dampens the MS, thus the effect on neurobehaviour, antioxidant defence gene expression, and telomere biology is minimized in their offspring. Together, our results suggest that supplementation of BME/L-C during gestation dampens the MS and reduced oxidative stress-mediated changes in telomere shortening/biology and associated neurobehaviour in offspring born following MS.
... In the present context, it is evident (Figure 4) that MoHE pathogenesis is associated with the declining levels of both BDNF and TrkB in the DG region, which, however, are found to be restored back to their normal levels due to the treatment with CDRI-08. The finding gets indirect support from the BM treatment-led normalization of memory impairments in prenatally stressed rats by modulating BDNF signalling (Sivasangari & Rajan, 2020), but there is no evidence involving AN. Thus, the findings of this article, by presenting a concordant pattern of restoration of all the markers/molecular players of the sequential steps of AN in DG, are the first to demonstrate restoration of the MoHE-associated compromised AN by CDRI-08. ...
Article
Full-text available
Modulation of in vivo adult neurogenesis (AN) is an evolving concept in managing neurodegenerative diseases. CDRI‐08, a bacoside‐enriched fraction of Bacopa monnieri , has been demonstrated for its neuroprotective actions, but its effect on AN remains unexplored. This article describes the status of AN by monitoring neuronal stem cells (NSCs) proliferation, differentiation/maturation markers and BDNF–TrkB levels (NSCs signalling players) vs. the level of neurodegeneration and their modulations by CDRI‐08 in the hippocampal dentate gyrus (DG) of male rats with moderate grade hepatic encephalopathy (MoHE). For NSC proliferation, 10 mg/kg b.w. 5‐bromo‐2′‐deoxyuridine (BrdU) was administered i.p. during the last 3 days, and for the NSC differentiation study, it was given during the first 3 days to the control, the MoHE (developed by 100 mg/kg b.w. of thioacetamide i.p. up to 10 days) and to the MoHE male rats co‐treated with 350 mg/kg b.w. CDRI‐08. Compared with the control rats, the hippocampus DG region of MoHE rats showed significant decreases in the number of Nestin ⁺ /BrdU ⁺ and SOX2 ⁺ /BrdU ⁺ (proliferating) and DCX ⁺ /BrdU ⁺ and NeuN ⁺ /BrdU ⁺ (differentiating) NSCs. This was consistent with a similar decline in BDNF ⁺ /TrkB ⁺ NSCs. However, all these NSC marker positive cells were observed to be recovered to their control levels, with a concordant restoration of total cell numbers in the DG of the CDRI‐08‐treated MoHE rats. The findings suggest that the restoration of hippocampal AN by CDRI‐08 is consistent with the recovery of BDNF–TrkB‐expressing NSCs in the MoHE rat model of neurodegeneration.
... Sustained hyperactivity of the HPA axis under stress could lead to the release of Glu-based excitatory neurotransmitters [57]. The large accumulation of Glu in the synaptic gap would stimulate the over-activation of NMDAR, resulting in the inward flow of calcium ions, which induced neurotoxicity and neuronal dysfunction [58]. ...
Article
Full-text available
Depression is a complex and prevalent mental illness. Cherry leaf is a traditional Chinese herbal medicine, which has confirmed to exert a certain antidepressant effect, but its potential neural regulation mechanism is not clear. This paper aims to investigate the improved action of cherry leaf decoction (CLD) on chronic unpredictable mild stress (CUMS) rats and its potential neural regulation mechanism by verifying the role and function of NMDAR regulatory target α2δ-1 in depression due to CUMS. Male SD rats were subjected to random stressors persisting for 5 weeks to establish the CUMS depression rat model. CLD could effectively alleviate depression-like behaviors of CUMS rats in behavioral tests including sucrose preference test, forced swimming test, tail suspension test and open field test. After the administration of the CLD, the expression of corticotropic-releasing hormone (CRH) in the hypothalamus was inhibited. Moreover, the levels of CRH, adrenal cortical hormone (ACTH) and corticosterone (CORT) in serum also decreased significantly. CUMS upregulated the expressions of α2δ-1, N-methyl-d-aspartate receptor 1 (NR1), NR2A and NR2B, and enhanced the binding ability to of α2δ-1 and NR1, which were reversed by CLD. The results demonstrated that CLD could ameliorate depression-like behaviors due to CUMS, which was related to the fact that CLD down-regulated α2δ-1 level and interfered with α2δ-1 binding to NR1, thereby reducing NMDAR expression and ultimately inhibiting HPA axis activity.
Article
Full-text available
Bacopa monnieri (L.) Wettst. (Plantaginaceae) (BM), known as “brahmi”, is a reputed medicinal plant, particularly in Ayurvedic medicine. Since BM has been traditionally used for memory dysfunction, experimental studies at the pre-clinical level support the nootropic effect of the plant relevant to Alzheimer’s disease (AD) demonstrated through various mechanisms. It has been reported that saponins (e.g. bacosides, bacopasides, and bacopasaponins) are largely responsible for the memory-enhancing and other neuropharmacological effects of BM. Bacosides, which are non-polar molecules that can easily cross the blood-brain barrier, are reported to directly lead to anti-inflammatory and antioxidant effects in the brain. BM extracts rich in bacosides are considered advantageous due to their higher nootropic efficacy. The findings suggest that only bacoside enrichment increases antiamnesic activity; other components also contribute to the extract with a synergism. Adverse side effects of BM in humans have rarely been reported. In the current review, we aimed to scrutinize in vivo data derived from the studies related to the cognitive-enhancement effect of BM with a special focus on AD.
Article
Full-text available
Long-term Memory (LTM) is formed by sequential changes in the different brain regions due to synaptic plasticity changes. This synaptic plasticity changes formed in the brain due to the acquittance of unexposed information and its retrieval due to learning and memory formation (LMF). In a normal condition, LMF uses RNA and protein synthesis machinery to form LTM, which lasts till the end of an organism's lifetime. Formed LTM shows sequential changes happening in the presynaptic and postsynaptic neurons. Stated sequential changes are initiated when the released neurotransmitter binds with the postsynaptic neuronal receptors and activates the brain's ERK - CREB neuronal signaling pathway. Based on the previous findings, the present study was designed to study the interrelationship between cognitive impairment and oral/gut dysbiosis with the help of a probiotic strain (Lactobacillus plantarum). Two phases of behavioural analysis (first and second phase) were used to identify the effect of oral microbial infusions on impaired LTM formation and its reversal using restoration of dysbiosed gut/oral microbiota. The first phase of behavioural analysis (FPBA) reported that oral microbial infusion plays a major role in developing oral/gut dysbiosis, which results in impaired cognitive functions. Further, formed oral/gut microbiota dysbiosis was reversed with the help of probiotic strain in the second phase of behavioural analysis (SPBA). Thus, a comparative two-phase behavioural analysis revealed that probiotics may play a significant role in reversing induced cognitive decline. The outcome of the present study also proved that probiotic treatment might play a major role in the reversal of dysbiosed microbiota in the oral cavity and the gut.
Article
Background Alzheimer's disease (AD) is the most prevalent dementia, affecting a large number of populations. Despite being under scrutiny for decades, an effective therapeutic option is still not available. Methods and results This study explores the therapeutic role of a nootropic herb Bacopa monnieri (BM) in AD‐like pathological conditions produced by injecting preformed amyloid‐β 42 (Aβ 42 ) fibril bilaterally into hippocampus of Wistar rats, and ethanolic extract of BM is orally administered for 4 weeks. Assessment of behavioral changes reveals that BM treatment ameliorates Aβ 42 ‐induced cognitive impairment and compromised explorative behavior. Supplementation of BM also reduces oxidative stress biomarkers, proinflammatory cytokines, and cholinesterase activity in the AD rats. Additionally, BM treatment restores Bcl‐2‐associated X protein (Bax)/ B‐cell lymphoma 2 (Bcl‐2) imbalance, increases neurotrophic factors expression, and prevents neurodegeneration validated by quantifying Nissl‐positive hippocampal neurons. Interestingly, BM administration eliminates amyloid plaques in the hippocampal region and normalizes the Aβ 42 ‐induced increase in phospho‐tau and total tau expression. Mechanistic investigations reveal that BM interacts with glycogen synthase kinase (GSK‐3β) and restores Wnt/β‐catenin signaling. Conclusion BM has been used in diet as a nootropic herb for several centuries. This study highlights the anti‐Alzheimer activity of BM from the behavioral to the molecular level by modulating mitochondrial dysfunction, and GSK‐3β mediates the Wnt/β‐catenin signaling pathway.
Article
Full-text available
Neurodegenerative diseases, including Alzheimer’s, Parkinson’s, and Huntington’s, are a growing global health challenge with no definitive cure. Oxidative stress is implicated in these disorders, and antioxidants have emerged as a promising avenue for addressing them. Research has shown the potential of natural antioxidants to combat oxidative stress in neurodegenerative disorders, but clinical trials have often failed to treat patients effectively. However, natural extracts have shown diverse molecular activities beyond their antioxidant capabilities, indicating their potential for prevention and disease management. This review will explore in vitro and in vivo research studies to highlight the promising prospects of natural antioxidants and their therapeutic applications in Neurodegenerative conditions.
Chapter
Advances in the treatment of neurodegenerative diseases (NDs) are nominal. Currently available therapies are merely symptomatic treatments that cannot prevent the development of the disease. Several herbs have been found very useful for managing neurological diseases. There are immense possibilities to discover a more successful line of ND treatment. Phytochemicals from medicinal plants may play a vital role in maintaining the chemical balance of the brain by affecting the capacity of receptors for the major inhibitory neurotransmitters. A few plants have already gained popularity for the potential treatment of NDs. This volume highlights the therapeutic role of medicinal plants and their scientific validation for improving neuronal health. It presents 15 chapters that cover the herbal treatment of NDs, including Parkinson's disease and Alzheimer's disease. The contents cover a range of pharmaceutical agents like sirtuins, berberine, rosmarinic acid and resveratrol. The book serves as a reference for pharmacology and herbal medicine scholars as well as healthcare workers interested in information about alternative and complementary therapies for neurological disorders.
Chapter
Advances in the treatment of neurodegenerative diseases (NDs) are nominal. Currently available therapies are merely symptomatic treatments that cannot prevent the development of the disease. Several herbs have been found very useful for managing neurological diseases. There are immense possibilities to discover a more successful line of ND treatment. Phytochemicals from medicinal plants may play a vital role in maintaining the chemical balance of the brain by affecting the capacity of receptors for the major inhibitory neurotransmitters. A few plants have already gained popularity for the potential treatment of NDs. This volume highlights the therapeutic role of medicinal plants and their scientific validation for improving neuronal health. It presents 15 chapters that cover the herbal treatment of NDs, including Parkinson's disease and Alzheimer's disease. The contents cover a range of pharmaceutical agents like sirtuins, berberine, rosmarinic acid and resveratrol. The book serves as a reference for pharmacology and herbal medicine scholars as well as healthcare workers interested in information about alternative and complementary therapies for neurological disorders.
Article
Full-text available
Schizophrenia (SZ) is a deleterious brain disorder affecting cognition, emotion and reality perception. The most widely accepted neurochemical-hypothesis is the imbalance of neurotransmitter-systems. Depleted GABAergic-inhibitory function might produce a regionally-located dopaminergic and glutamatergic-storm in the brain. The dopaminergic-release may underlie the positive psychotic-symptoms while the glutamatergic-release could prompt the primary negative symptoms/cognitive deficits. This may occur due to excessive synaptic-pruning during the neurodevelopmental stages of adolescence/early adulthood. Thus, although SZ is not a neurodegenerative disease, it has been suggested that exaggerated dendritic-apoptosis could explain the limited neuroprogression around its onset. This apoptotic nature of SZ highlights the potential therapeutic action of anti-apoptotic drugs, especially at prodromal stages. If dysregulation of apoptotic mechanisms underlies the molecular basis of SZ, then anti-apoptotic molecules could be a prodromal therapeutic option to halt or prevent SZ. In fact, risk alleles related in apoptotic genes have been recently associated to SZ and shared molecular apoptotic changes are common in the main neurodegenerative disorders and SZ. PRISMA-guidelines were considered. Anti-apoptotic drugs are commonly applied in classic neurodegenerative disorders with promising results. Despite both the apoptotic-hallmarks of SZ and the widespread use of anti-apoptotic targets in neurodegeneration, there is a strikingly scarce number of studies investigating anti-apoptotic approaches in SZ. We analyzed the anti-apoptotic approaches conducted in neurodegeneration and the potential applications of such anti-apoptotic therapies as a promising novel therapeutic strategy, especially during early stages.
Article
Full-text available
More than 300 million people suffer from depressive disorders globally. People under early-life stress (ELS) are reportedly vulnerable to depression in their adulthood, and synaptic plasticity can be the molecular mechanism underlying such depression. Herein, we simulated ELS by using a maternal separation (MS) model and evaluated the behavior of Sprague–Dawley (SD) rats in adulthood through behavioral examination, including sucrose preference, forced swimming, and open-field tests. The behavior tests showed that SD rats in the MS group were more susceptible to depression- and anxiety-like behaviors than did the non-MS (NMS) group. Nissl staining analysis indicated a significant reduction in the number of neurons at the prefrontal cortex and hippocampus, including the CA1, CA2, CA3, and DG regions of SD rats in the MS group. Immunohistochemistry results showed that the percentages of synaptophysin-positive area in the prefrontal cortex and hippocampus (including the CA1, CA2, CA3, and DG regions) slice of the MS group significantly decreased compared with those of the NMS group. Western blot analysis was used to assess synaptic-plasticity protein markers, including postsynaptic density 95, synaptophysin, and growth-associated binding protein 43 protein expression in the cortex and hippocampus. Results showed that the expression levels of these three proteins in the MS group were significantly lower than those in the NMS group. LC–MS/MS analysis revealed no significant differences in the peak areas of sex hormones and their metabolites, including estradiol, testosterone, androstenedione, estrone, estriol, and 5β-dihydrotestosterone. Through the application of nontargeted metabolomics to the overall analysis of differential metabolites, pathway-enrichment results showed the importance of arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and the phenylalanine, tyrosine, and tryptophan biosynthesis pathways. In summary, the MS model caused adult SD rats to be susceptible to depression, which may regulate synaptic plasticity through arginine and proline metabolism; pantothenate and CoA biosyntheses; glutathione metabolism; and phenylalanine, tyrosine, and tryptophan biosyntheses.
Article
Full-text available
Rationale and objectiveVortioxetine has been reported to exhibit a variety of neurobiological functions and neuroprotective effects. In the present study, we aimed to investigate the effects of vortioxetine on cognitive performance in a transgenic mouse model of Alzheimer’s disease (AD).Methods We administered vortioxetine (10 mg/kg, i.p., every day, for approximately 6 weeks), which acts on multiple 5-serotonin (5-HT) receptors, to 3.5-month-old 5×FAD mice. Subsequently, we used the open field (OF) test to detect anxiety-like behavior in the mice. The novel object recognition (NOR) test and Morris water maze (MWM) were used to assess the cognitive states of the 5×FAD mice. We also measured the levels of insoluble amyloid plaques and soluble β-amyloid (Aβ) plaques. Finally, we explored the expression levels of postsynaptic density protein 95 (PSD95), synaptophysin (SYP), and synaptotagmin-1 (SYT1) in the hippocampus of the mice.ResultsThe administration of vortioxetine effectively reversed the reduction in anxiety-type behaviors in 5×FAD mice and improved the impairment in recognition memory and spatial reference memory. However, we did not find that vortioxetine decreased or delayed the formation of amyloid plaques or Aβ. Interestingly, we found a significant increase in the expression levels of PSD95, SYP, and SYT1 in the 5×FAD mice after vortioxetine treatment compared with the control group.Conclusion These results demonstrate that vortioxetine may improve cognitive impairment in 5×FAD mice. The role in cognitive improvement may be related to the beneficial effects of vortioxetine on synaptic function.
Article
Full-text available
The essential involvement of phosphoinositides in synaptic plasticity is well-established, but incomplete knowledge of the downstream molecular entities prevents us from understanding their signalling cascades completely. Here, we determined that Phldb2, of which pleckstrin-homology domain is highly sensitive to PIP3, functions as a phosphoinositide-signalling mediator for synaptic plasticity. BDNF application caused Phldb2 recruitment toward postsynaptic membrane in dendritic spines, whereas PI3K inhibition resulted in its reduced accumulation. Phldb2 bound to postsynaptic scaffolding molecule PSD-95 and was crucial for localization and turnover of PSD-95 in the spine. Phldb2 also bound to GluA1 and GluA2. Phldb2 was indispensable for the interaction between NMDA receptors and CaMKII, and the synaptic density of AMPA receptors. Therefore, PIP3-responsive Phldb2 is pivotal for induction and maintenance of LTP. Memory formation was impaired in our Phldb2−/− mice.
Article
Full-text available
Background: Parkinson's Disease (PD) is characterized by alterations in cerebellum and basal ganglia functioning with corresponding motor deficits and neuropsychiatric symptoms. Involvement of oxidative dysfunction has been implicated for the progression of PD, and environmental neurotoxin exposure could influence such behavior and psychiatric pathology. Assessing dietary supplementation strategies with naturally occurring phytochemicals to reduce behavioral anomalies associated with neurotoxin exposure would have major clinical importance. The present investigation assessed the influence of Bacopa monneri (BM) on behaviors considered to reflect anxiety-like state and motor function as well as selected biochemical changes in brain regions of mice chronically exposed to ecologically relevant herbicide, paraquat (PQ). Materials & methods: Male mice (4-week old, Swiss) were daily provided with oral supplements of standardized BM extract (200 mg/kg body weight/day; 3 weeks) and PQ (10 mg/kg, i.p. three times a week; 3 weeks). Results: We found that BM supplementation significantly reversed the PQ-induced reduction of exploratory behavior, gait abnormalities (stride length and mismatch of paw placement) and motor impairment (rotarod performance). In a separate study, BM administration prevented the reduction in dopamine levels and reversed cholinergic activity in brain regions important for motor (striatum) pathology. Further, in mitochondria, PQ-induced decrease in succinate dehydrogenase (SDH) activity and energy charge (MTT reduction), was restored with BM supplementation. Conclusion: These findings suggest that BM supplementation mitigates paraquat-induced behavioral deficits and brain oxidative stress in mice. However, further investigations would enable us to identify specific molecular mechanism by which BM influences behavioural pathology.
Article
Full-text available
Background: Epidemiological surveys and studies with animal models have established a relationship between maternal stress and affective disorders in their offspring. However, whether maternal depression before pregnancy influences behaviour and related neurobiological mechanisms in the offspring has not been studied. Results: A social defeat stress (SDS) maternal rat model was established using the resident-intruder paradigm with female specific pathogen-free Wistar rats and evaluated with behavioural tests. SDS maternal rats showed a significant reduction in sucrose preference and locomotor and exploratory activities after 4 weeks of stress. In the third week of the experiment, a reduction in body weight gain was observed in SDS animals. Sucrose preference, open field, the elevated-plus maze, light-dark box, object recognition, the Morris water maze, and forced swimming tests were performed using the 2-month-old male offspring of the female SDS rats. Offspring subjected to pre-gestational SDS displayed enhanced anxiety-like behaviours, reduced exploratory behaviours, reduced sucrose preference, and atypical despair behaviours. With regard to cognition, the offspring showed significant impairments in the retention phase of the object recognition test, but no effect was observed in the acquisition phase. These animals also showed impairments in recognition memory, as the discrimination index in the Morris water maze test in this group was significantly lower for both 1 h and 24 h memory retention compared to controls. Corticosterone, adrenocorticotropic hormone, and monoamine neurotransmitters levels were determined using enzyme immunoassays or radioimmunoassays in plasma, hypothalamus, left hippocampus, and left prefrontal cortex samples from the offspring of the SDS rats. These markers of hypothalamic-pituitary-adrenal axis responsiveness and the monoaminergic system were significantly altered in pre-gestationally stressed offspring. Brain-derived neurotrophic factor (BDNF), cyclic adenosine monophosphate response element binding protein (CREB), phosphorylated CREB (pCREB), and serotonin transporter (SERT) protein levels were evaluated using western blotting with right hippocampus and right prefrontal cortex samples. Expression levels of BDNF, pCREB, and SERT in the offspring were also altered in the hippocampus and in the prefrontal cortex; however, there was no effect on CREB. Conclusion: We conclude that SDS before pregnancy might induce depressive-like behaviours, cognitive deficits, and neurobiological alterations in the offspring.
Article
Full-text available
Significance De novo protein synthesis is critical for memory formation. We found that protein synthesis during acquisition is transiently required for contextual memory formation. We identified one candidate gene, Nrgn (encoding protein neurogranin, Ng) with enhanced translation upon novel-context exposure, and found that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. Furthermore, fragile-X mental retardation protein interacts with the 3′UTR of the Nrgn mRNA, which is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Together, these results indicate that experience-dependent and acute translation of Ng in the hippocampus during memory acquisition enables durable context memory encoding.
Article
Full-text available
CaMKII is one of the most studied synaptic proteins, but many critical issues regarding its role in synaptic function remain unresolved. Using a CRISPR-based system to delete CaMKII and replace it with mutated forms in single neurons, we have rigorously addressed its various synaptic roles. In brief, basal AMPAR and NMDAR synaptic transmission both require CaMKIIα, but not CaMKIIβ, indicating that, even in the adult, synaptic transmission is determined by the ongoing action of CaMKIIα. While AMPAR transmission requires kinase activity, NMDAR transmission does not, implying a scaffolding role for the CaMKII protein instead. LTP is abolished in the absence of CaMKIIα and/or CaMKIIβ and with an autophosphorylation impaired CaMKIIα (T286A). With the exception of NMDAR synaptic currents, all aspects of CaMKIIα signaling examined require binding to the NMDAR, emphasizing the essential role of this receptor as a master synaptic signaling hub.
Article
Aims: Pre- and/or early postnatal ethanol exposure (prenatal alcohol exposure [PAE]) impairs synaptic plasticity as well as memory formation, but the mechanisms underlying these effects remain unclear. Both long-term potentiation (LTP) and spatial memory formation in the hippocampus involve the nicotinamide adenine dinucleotide phosphate oxidase type 2 (NOX2) enzyme. Previous studies have reported that N-methyl-d-aspartate receptor (NMDAR) activation increases NOX2-mediated superoxide generation, resulting in inhibition of NMDAR function, but whether NOX2 impacts NMDAR function in PAE animals leading to impaired LTP and memory formation remains unknown. We aim to evaluate whether the NOX2-NMDAR complex is involved in the long-lasting deleterious effects of PAE on hippocampal LTP and memory formation. Results: Here we provide novel evidence that PAE animals display impaired NMDAR-dependent LTP in the cornus ammonis field 1 (CA1) and NMDAR-mediated LTP in the dentate gyrus (DG). Moreover, PAE rats displayed increased NMDAR-mediated transmission in both hippocampal areas. Interestingly, NOX2 pharmacological inhibition restored NMDAR-mediated transmission and LTP in the CA1, but not in the DG. PAE also induced overexpression of NOX2 and CaMKII isoforms, but did not modify the content or the redox state of the N-methyl-d-aspartate receptor subunit-1 (NR1) subunit of NMDAR in both areas of the hippocampus. In addition, adolescent PAE rats orally fed the antioxidant and free radical scavenger apocynin exhibited significantly improved spatial memory acquisition. Innovation and Conclusion: By showing in PAE animals NOX2 overexpression and increased NMDAR-mediated transmission, which might lead to impaired synaptic plasticity and memory formation in a region-specific manner, we provide an important advance to our current understanding of the cellular mechanisms underlying PAE-dependent defective hippocampal function.
Article
Gestational period plays critical role in neuropsychological development. One of the genes that undergoes changes by prenatal stress (PNS) exposure, is the gene coding brain derived neurotrophic factor (BDNF). Studies have reported different patterns of change following PNS in BDNF, which emphasizes the complexity of the issue. In this review, systematic search of PubMed, Scopus, Web of Science and Cochrane CENTRAL databases was performed. Primary searches resulted in 2132 studies and finally 43 studies were found to meet the inclusion criteria. Transcriptional and epigenetic changes of BDNF gene in the brain were recorded. Decreased or unchanged BDNF total mRNA and BDNF mature protein, with hypermethylation of the coding exons were the most reported changes. However, stress paradigm, gender of the fetus and the day of sacrifice were found to significantly affect the results. Hippocampus and prefrontal cortex are the most vulnerable regions. They can show long lasting and persistent transcriptional and epigenetics changes of BDNF gene following PNS. Further studies evaluating the importance of these findings in humans are essential.
Article
Various types of adaptive behavior during the prepubertal period were analyzed in the offspring of rats receiving chronic injections of serotonin (5-HT) reuptake inhibitor fluoxetine, 5-HT1A receptor agonist buspirone, or their combination starting from gestation day 9 and subjected to immobilization stress from the 15th day of pregnancy until delivery. Prenatal stress increased pain sensitivity, prolonged inflammatory pain response, and increased the levels of anxiety and depression. Chronic administration of drugs acting through 5-HT1A receptors to pregnant rats improved the studied behavioral parameters in their offspring. Differences in the pain sensitivity were found between the effect of drug combination and each of them separately, and in the level of depression between combined administration and fluoxetine alone.