Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
413
Original Article
SMJ
Wipawan angnipon, Ph.D.*, Sukonthar Ngampramuan, Ph.D.*, Nopparat Suthprasertporn, Ph.D.*, Chanati
Jantrachotechatchawan, Ph.D.**, Patoomratana Tuchinda, Ph.D.***, Saksit Nobsathian, Ph.D.****
*Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom 73170, ailand, **Wolfson Centre
for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, United Kingdom, ***Department of Chemistry,
Faculty of Science, Mahidol University, Bangkok 10700, ailand, ****Nakhon Sawan Campus, Mahidol University, Phayuhakiri, Nakhon Sawan
60130, ailand.
Protective Roles of N-trans-feruloyltyramine Against
Scopolamine-Induced Cholinergic Dysfunction on
Cortex and Hippocampus of Rat Brains
ABSTRACT
Objective: To study the protective eects of N-trans-feruloyltyramine (NTF) on scopolamine-induced cholinergic
dysfunction, apoptosis, and inammation in rat brains.
Materials and Methods: Treatments were administered intraperitoneally (i.p.). Wistar rats (8-week-old) were
allocated into 4 groups (n = 3) as follows: scopolamine-only, NTF-only, NTF + scopolamine and control. Spatial
cognition was evaluated by Morris water maze. ROS assay and Western blot analyses were conducted in 3 brain
regions: the frontal cortex, hippocampus, and temporal cortex.
Results: NTF treatment inhibited scopolamine-induced memory impairment and signicantly attenuated scopolamine-
induced changes in the three brain regions. Investigated scopolamine-associated changes were as follows: increases
in ROS production and BACE1 level, decrease in ChAT level, increases in inammatory and apoptotic markers,
and activation of signaling pathway kinases related to inammation and apoptosis.
Conclusion: With its in vivo antioxidant, cholinergic-promoting, anti-apoptosis, and anti-inammatory biological
activities, NTF is a promising candidate to be further investigated as a potential treatment for Alzheimer’s-associated
neurodegeneration.
Keywords: Acetylcholine; Alzheimer’s disease; antioxidant; Morris water maze; N-trans-feruloyltyramine (Siriraj
Med J 2021; 73: 413-422)
Corresponding author: Wipawan angnipon
E-mail: wipawan.tha@mahidol.ac.th
Received 5 February 2021 Revised 20 April 2021 Accepted 22 April 2021
ORCID ID: http://orcid.org/0000-0001-7889-2801
http://dx.doi.org/10.33192/Smj.2021.55
INTRODUCTION
Alzheimer’s disease (AD) causes progressive and
irreversible deterioration of cognitive functions especially
memory.
1
e major pathological characteristics of the
human AD brain are extracellular aggregates of amyloid-β
(Aβ) and intracellular aggregates of hyperphosphorylated
tau, namely the senile plaques and the neurobrillary
tangles respectively.
2
The underlying mechanism of
sporadic AD involves cholinergic dysfunction including
degeneration of basal forebrain cholinergic neurons
and loss of hippocampal cholinergic bers.
3
Impaired
cholinergic transmission aects learning and memory,
cortical and hippocampal information processing, and
ultimately behaviors.
6
Scopolamine, an antagonist of a
muscarinic acetylcholine receptor,
4
induces cholinergic
dysfunction and cognitive impairment through oxidative
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
414
stress and neuroinflammation in animal models.
5
Intraperitoneal injection of scopolamine blocks cholinergic
neurotransmission, dysregulates cholinergic system, and
consequently impairs cognition in rodents.
N-trans-feruloyltyramine (NTF) was purified from
Polyalthia suberosa, a shrubby tree found throughout
southeast Asia and south China whose parts are used
in ai traditional medicine.
6
NTF has potent radical-
scavenging antioxidant property, especially from phenolic
hydroxyls. NTF and the other puried compound studied
by our group, N-benzylcinnamide (PT-3), have been
demonstrated to be eective in protecting rat cultured
cortical neurons against Aβ-induced cytotoxicity by
inhibiting ROS production, suppressing apoptotic caspase-3
and Bax, and in turn elevating anti-apoptotic Bcl-2.
7-8
As PT-3 that has elicited protective eects on primary
cortical neurons also showed promising outcome in
aged rat brain in vivo,
9
neuroprotective eects of NTF
in vivo had become our next primary study target.
In this research study, the animals were used in the
experiment, and they were calculated for the sample size
in accordance with the ethical guideline to protect the
unnecessary wastage of resources. Consistently, there are
several studies that have demonstrated the statistically
signicant results with a small sample size (n = 3).
9-11
Meanwhile, our preliminary results showed the protective
eects of NTF treatment against scopolamine-induced
cholinergic dysfunction, including, ROS production,
apoptosis, inammation, and associated signaling pathways
in several rat brain regions, specically the frontal cortex,
hippocampus and temporal cortex.
e frontal cortex integrity is correlated with the
higher reservation of cognitive performance in aging
populations.
12
e hippocampus and especially its associated
cholinergic signaling pathway play a crucial role in memory
formation.
13
e medial temporal lobe cortex or, in short,
the temporal cortex is reciprocally interconnected to the
hippocampus and it greatly involved in the hippocampus-
associated cognitive processes and cognitive decline such
as AD in humans
14
and ischemia-associated dementia in
rats.
15
Furthermore, ChAT activity in the temporal cortex
is positive correlated with cognitive preservation, but it is
negatively correlated with AD pathologies.
16
Importantly,
the frontal cortex, hippocampus, and temporal cortex are
three of the regions heavily aected by scopolamine
17,18
and aging
9
in the rat brain.
Herein, we studied the neuroprotective eects of
NTF treatment on scopolamine-associated cholinergic
dysfunction, namely, ROS production, apoptosis,
inammation, and relevant signaling pathways in the
rat frontal and temporal cortices and hippocampus.
MATERIALS AND METHODS
Reagents
Scopolamine (Sigma-Aldrich) and NTF were dissolved
in a vehicle solution of (v/v) 40% dimethylsulfoxide,
59% phosphate-buered saline (PBS), and 1% ethanol
and diluted in PBS for animal administration. NTF was
isolated from the acetone extract of Polyalthia suberosa
stems as previously described.
6
e test drug NTF was
in its form of pure compound. From 997.2 mg of the
semi-solid fraction, 88.1 mg (8.83 %) of NTF could be
puried by preparative layer chromatography.
6
Animal experiments
All experimental procedures were approved by
the Institute of Molecular Biosciences Animal Care and
Use Committee (MB-ACUC) (COA.NO.MB-ACUC
2016/002).
Twelve 8-week male Wistar rats (250-300 g) from the
National Laboratory Animal Center, Mahidol University,
were individually cared for in cages under 12 h light/
dark cycle, 22±1 °C temperature, 45-55 % humidity, and
ad libitum water and diet. e 12 rats were separated
into 4 groups, each with 3 animals, namely, scopolamine
treatment only, NTF treatment only, scopolamine plus
NTF treatment, and control. Following habituation for 5
days, animals were once per day injected intraperitoneally
(i.p.) for 14 days.
19
In the control group, animals were
injected with vehicle 1 h before the water maze test for 14
days. In the scopolamine-only group, animals were rst
injected for 7 days with vehicle followed by scopolamine
treatment on day 8 to day 14 (3.0 mg/kg BW) 1 h before
the water maze test. In the NTF treatment only group,
animals were injected with 1 ml aliquot of NTF (1.5
mg/kg BW) for 14 days 1 h before the water maze test.
In the scopolamine plus NTF treatment group, animals
were administered with NTF as described above for 7
days and then together with n day 8 to day 14 (3.0 mg/
kg BW) 1 h before the water maze test.
Rats were treated with NTF for 7 days prior to
MWM training because we would like to investigate the
neuroprotective eects of NTF against scopolamine. e
NTF eects as a memory enhancer can still remain during
the MWM training.
19
e dose of NTF was selected in
accordance with our previous ndings on the protective
roles of N-benzylcinnamide (PT-3).
9
In animal studies the
model of scopolamine has been performed using doses
between 0.5,1 or 3 mg/kg IP). e eect of scopolamine
to induce impairment of learning and memory is dose
dependent.
19-20
e highest dose of scopolamine exhibits
the greater severity of memory dysfunction. Scopolamine
was administered via the injection 1 hr before MWM
Thangnipon et al.
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
415
Original Article
SMJ
training because the half-life of scopolamine is 2.4±1.4
h for its elimination.
21
Central pharmacodynamic eects
of scopolamine peak between 1 and 3 h and disappear
aer 5–6 h.
22
Morris water maze test
Water maze tests were conducted to evaluate the
eects of NTF and scopolamine on the acquisition and
retention of spatial memory.
18
Visible platform trial
was performed on the day before the training trial.
Animals have to associate the relative location of the
visible platform with the provided visual cues. On the
following training trials, the platform was submerged
and invisible. e training trials were conducted over a
period of 7 days as follows: day 1, visible platform test;
days 2-6, hidden platform tests; and on the same day 6
aer hidden platform, probe trial test. ese consecutive 6
days correspond to the days 8-13 of the treatment period.
Rats in each group were put in the water facing dierent
quadrants, which were altered for each experiment,
and the escape latency dened as time spent to nd the
submerged platform was recorded. In the probe trial,
time spent in the target quadrant with the platform was
recorded.
e experimental schedule was designed to investigate
the protective eect of NTF as a memory enhancer in
rats. Rats underwent habituation for 5 days. Aer that,
NTF (1 ml, 1.5 mg/kg. i.p.) or vehicle was administrated
once a day for 7 days. en, 1 h prior to the rst trial
session, rats were administrated of veh, NTF, scopolamine
(3.0 mg/kg) or NTF+Scop (i.p) every MWM training
for 6 days, in accordance with the previously described
protocol.
23
ROS production assay
After animals were decapitated without prior
anesthesia, the 3 regions of the rat brains – the frontal
cortex, hippocampus, temporal cortex – were dissected
and frozen at -80°C. For the frontal cortex, the frontal
part of the brain was cut straight down coronally and
attached subcortical brain regions were removed. For
the hippocampus, the bilateral hippocampal tissues with
all subsectors were collected. For the temporal cortex, all
cortical regions encompassing the hippocampus or the
medial temporal lobe were dissected. Reactive oxygen
species (ROS) production was measured according to the
protocol of our previous work.
24
In brief, 2.4-3.0 mg of
brain tissue from each dissected region were homogenized
in 80-100 µl of ice-cold Locke’s buer, then 10 µl of
homogenate was adjusted to a concentration of 3 mg/ml
tissue, incubated with 10 µM 2',7'-dichlorouorescin
diacetate (DCFH-DA) (stock solution in 100% ethanol)
for 45 min at 37°C. Fluorescence of the oxidized product
2',7'-dichlorouorescein (DCF) (485 nm excitation and
535 nm emission) was measured every 10 minutes for
4 times in total at 37°C in a DTX 880 multimode plate
reader.
Western blot analysis
Rat brains were dissected into three regions: frontal
cortex, hippocampus and temporal cortex).
25
e brain
protein contents were extracted with the lysis buer,
centrifuged at 11,000 ×g for 15 min, and collected as
a supernatant. e protein samples were loaded onto
polyacrylamide gel. e proteins were separated by gel
electrophoresis and transferred onto (PVDF) membranes
where they were reacted to specic antibodies. On the
PVDF membrane, proteins of interest were identied using
a detection reagent. e protein bands were quantied
and analyzed with the same loading sample groups of
its own gel. PVDF membrane was incubated overnight
at 4°C with primary rabbit anti-Bax, -Bcl-2, -activated
caspase-3, -BACE1, -phospho-p38, -p38, -phospho-JNK,
-JNK, -phospho-ERK1/2, -ERK1/2, -phospho-Akt, -Akt,
-phospho-NF-κB p65 (Ser536), -NF-κB antibodies (1:1000
dilution) (Cell signaling), -IL-1β (1:1000) (Santa Cruz);
and primary mouse anti-ChAT (1:1000) (Millipore) and
-IL-6 (1:1000) (Santa Cruz); and rabbit anti-actin (1:2500)
(Cell Signaling) for normalization of protein loading.
Immunoreactive proteins were visualized by incubating
PVDF membranes with secondary horseradish peroxidase-
conjugated antibodies (1:1000) (Cell Signaling) at room
temperature for 1 h and subsequently with enhanced
chemical luminescence (ECL) reagents (Bio-Rad) before
signal exposure to Hyperlm
TM
(GE Healthcare). Intensities
of the visualized protein bands were measured by scanning
densitometry (Image J soware). e optical density
(OD) of protein bands on Hyperlm was determined
and normalized to the OD of β-actin and total phospho
proteins. e results were expressed as the percentage
of OD values by using the ImageJ soware.
Each gel contains four sample groups (Control,
Sco, NTF+sco, NTF). Each protein sample of three rats
on dierent brain regions, including frontal cortex (3),
hippocampus (3), and temporal cortex (3) was loaded
separately on dierent polyacrylamide gel. Four sample
groups of each region in each rat were underwent Western
blotting. Each blot was repeated for at least 3 times from
dierent batches of protein samples. e representative
blots were shown in gures.
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
416
Statistical analysis
Statistical analyses were performed using Prism
6.0 (GraphPad). Dierences in the escape latency from
the training/hidden platform trials were analyzed using
two-way analysis of variance (ANOVA). Dierences
between means of the 4 groups from all other experiments
were determined using one-way ANOVA with pair-
wise p-values corrected by Tukey’s posthoc test. Results
are deemed signicant at p < 0.05. In all Western blot
analyses, non-phospho proteins (ChAT, Bcl-2, Bax,
Activated caspase-3, IL-6, and IL-1β) were quantied and
normalized with β-Actin. Meanwhile, phosphoproteins
(p-NFκB, p-ERK, p-JNK, p-p38, p-Akt) were quantied
and normalized with the total protein counterparts.
RESULTS
Learning and memory in a Morris water maze test of
rats administered scopolamine with and without NTF
Control rats became proficient at locating the
submerged platform by day 5 of training compared to
day 1 (p < 0.05) in a hidden platform test (Fig 1A). On day
5 of training scopolamine-treated rats perform signicantly
less well than controls (p < 0.01), but, interestingly, rats
treated with scopolamine together NTF have prociencies
superior to those of controls, whereas NTF treatment
alone was no more eective than non-treated control.
In the probe trial tests, as expected, compared to control
rats, those in the scopolamine plus NTF treatment group
stay signicantly longer in the platform quadrant. ose
in the NTF only treatment group spent the same time.
Lastly as expected, those in the scopolamine-only group
stayed for a signicantly shorter duration (Fig 1B).
ROS production in brain tissues of rats administered
scopolamine with and without NTF
Scopolamine treatment for 7 days elevated ROS
level 2 folds in all three types of brain tissues (frontal
cortex, hippocampus, and temporal cortex) over control
levels (Fig 2). ROS levels were restored to control levels
by NTF administration (7 days prior and during the
scopolamine treatment period), while NTF alone (for
7 days) had no eect on ROS levels in all three types of
brain tissues.
Fig 1. NTF (1.5 mg/kg) was injected i.p. daily to rats 7 days before and together with scopolamine (3.0 mg/kg BW i.p.) 1 h before the 7-day
Morris water maze test; probe trial was conducted on day 7. Each data point (n=3) is expressed as mean ± SEM. A NTF prevented scopolamine
from elevating escape latency. *p < 0.05 scopolamine-treated with NTF versus (vs) the scopolamine group on day 3, **p < 0.01 scopolamine-
treated with NTF vs the scopolamine group on day 5. B NTF restored scopolamine-induced decrease in retention measured as time spent
in the target quadrant. *p < 0.05 scopolamine vs NTF, **p < 0.01 scopolamine vs scopolamine with NTF treated group.
Fig 2. Eects of NTF treatment in scopolamine-treated rats
on ROS. Production of ROS in brain homogenates were
measured by uorometric agent DCFH-DA. Each data bar
(n=3) is expressed as mean ± SEM. *p < 0.05, **p < 0.01 vs
control; #p < 0.05, ##p < 0.01 vs scopolamine-treated group.
Thangnipon et al.
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
417
Original Article
SMJ
Levels of cholinergic, apoptosis, and inammatory
proteins in brain tissues of rats administered scopolamine
with and without NTF
Administration of scopolamine to rats signicantly
decreases ChAT and increases BACE1 levels in the 3 brain
regions compared to control tissues (Fig 3A-C), and NTF
cotreatment was able to ameliorate these alterations.
However, NTF treatment alone had no eect on basal
levels of the two proteins.
Exposure to scopolamine causes a signicant increase
in apoptotic proteins, Bax and activated caspase-3, and
a corresponding decrease in anti-apoptotic Bcl-2 in the
three types of brain tissues compared to controls with
NTF treatment reversing these changes (Fig 4A-C).
However, these relative changes in apoptosis-related
proteins levels in the brain tissues of rats treated with
NTF alone varied with tissue types and thus impacting the
ability of NTF to restore the normal levels of apoptosis-
related proteins in the scopolamine-treated animals.
Scopolamine significantly increased levels of
cytokines – interleukin (IL)-6 and IL-1β – and of activated
transcription factor – phosphorylated NF-κB – which
were reversed by NTF. e extent of these phenomena
relative to control levels also depended on brain tissue
types (Fig 5A-C).
Activated cell signaling pathways in brain tissues of
rats administered scopolamine with and without NTF
We investigated the activation indicated by the
proportion of phosphorylated proteins of JNK, p38, ERK1/2,
and Akt. In the frontal cortex (Fig 6A), scopolamine
increased p-JNK and p-p38, decreased p-ERK1/2, and
increased p-Akt. NTF downregulated p-ERK1/2 but
upregulated p-Akt without aecting p-JNK or p-p38.
A combination of scopolamine and NTF could only
restore p-p38 to control level. In the hippocampus (Fig
6B), scopolamine elevated levels of p-JNK and p-p38,
depressed p-ERK1/2 and had no eect on p-Akt. NTF had
no eect on levels of p-JNK, increased p-p38, decreased
p-ERK1/2, and had no eect on p-Akt. A combination
of scopolamine and NTF could only restore p-ERK1/2
to control level and had no eect on the p-Akt level. In
the temporal cortex (Fig 6C), scopolamine raised levels
p-JNK and p-p38, lowered p-ERK1/2, and had no eect
on p-Akt. NTF had no eect on levels of p-JNK, p-p38,
or p-ERK1/2 but decreased p-Akt. A combination of
scopolamine and NTF restored levels of p-JNK, p-p38
and p-ERK1/2, and had no eect on p-Akt. Table 1
shows the quantitative protein values as the percentage
of change in expression relative to control groups in
Western blot analysis.
Fig 3. Eects of NTF on scopolamine-induced cholinergic dysfunction in rats on the levels of ChAT and BACE1 in: A frontal cortex, B
hippocampus, and C temporal cortex. Each data bar (n=3) is expressed as mean ± SEM. *p < 0.05, **p < 0.01 vs control group; ##p < 0.01,
###p < 0.001 vs scopolamine-treated group
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
418
Fig 4. Eects of NTF on scopolamine-induced apoptosis represented by the levels of Bax, Bcl-2, and activated caspase-3 in: A frontal cortex,
B hippocampus, and C temporal cortex. Each data bar (n=3) is expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs control
group; #p < 0.05, ###p < 0.001 vs scopolamine-treated group.
Fig 5. Eects of NTF on scopolamine-induced inammatory responses indicated by the levels of p-NF-κB, IL-6, and IL-1β in: A frontal
cortex, B hippocampus, and C temporal cortex. Each data bar (n=3) is expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 vs
control group; #p < 0.05, ##p < 0.01, ###p < 0.001 vs scopolamine-treated group.
DISCUSSION
Scopolamine has been demonstrated to increase
oxidative stress including ROS production in the mouse
cortex and hippocampus, which could be reversed by
pretreatment with sodium tanshinone IIA sulfonate,
a radical-scavenging antioxidant.
26
e current study
shows that administration of NTF was able to prevent
scopolamine-augmented ROS production in the frontal
and temporal cortices and hippocampus.
Oxidative stress contributes to the mechanisms of
multiple neurodegenerative disorders including AD.
27
Both oxidative damage and cholinergic dysfunction
have been implicated in cognitive impairment in rat
dementia models. Treatment with antioxidant Terminalia
chebula extract attenuated scopolamine-induced ROS
generation and reduction in ChAT and ACh levels in
mouse hippocampal tissue.
5
BACE1 is an enzyme necessary
for the production of Aβ42 and Aβ40 peptides in AD.
28
Consistent with our nding, an antioxidant ipriavone
decreased BACE1 expression in the hippocampus of a
scopolamine-treated rat.
29
Furthermore, NTF treatment alone
signicantly decreased apoptosis through downregulation
of activated caspase-3 and Bax and upregulation of
Bcl-2 in the hippocampus. Similarly, treatment with
an antioxidant sodium tanshinone IIA sulfonate in a
scopolamine-injected mouse signicantly decreased
Bax:Bcl-2 expression ratio and downregulated expression
of activated caspase-3 in the hippocampus and cortex.
26
Scopolamine also induces cytokines and NF-κB
phosphorylation;
30
all of these changes were restored
to normal levels in the rat cortex and hippocampus by
NTF as described in the current study. Pretreatment
Thangnipon et al.
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
419
Original Article
SMJ
with a water-soluble ginseng oligosaccharide extract
decreased scopolamine-induced expression of IL-1β and
IL-6 mRNAs in the murine hippocampus and signicantly
prevented scopolamine from inducing spatial memory
impairment during the water maze test.
31
In addition,
antioxidant isoavones from soybean-fermented food
tempeh restored ACh level and suppressed scopolamine-
induced IL-1β expression in the rat brains.
32
The effects of scopolamine treatment on phosphorylation
of the four signaling proteins (JNK, p38, ERK1/2, and Akt)
were consistent across the three brain regions. Whether
NTF could restore these changes could not be predicted
based on the eects of NTF treatment alone in each brain
tissue type. However, where scopolamine or NTF alone
resulted in similar eects to a cell signaling protein, e.g.
p-Akt (stimulated in the frontal cortex but unaected
in the temporal cortex and hippocampus), the results
of their combination were as expected. In scopolamine-
treated rats, NTF inhibited phosphorylation of JNK
and p38 in these three brain regions. Previous studies
have reported that JNK inhibitor SP600125 increased
the number of surviving cells aer ischemia in the rat
hippocampus
33
and that SB202190 p38 inhibitor provided
neuroprotection against cell apoptosis in mice.
34
Scopolamine-decreased p-ERK1/2 levels in the three
brain regions were accompanied by elevated BACE1
levels. Congruently, the expression and activity of BACE1
are negatively regulated by the ERK pathway.
35
Similar
to our study, treatment of an antioxidant ipriavone
restored ERK1/2 phosphorylation and decreased BACE-1
expression and subsequent Aβ pathologies in a scopolamine-
administered rat.
15
ERK1/2 inhibitor U0126 also blocked
alleviating eects of amyrin, a triterpene with antioxidant
activity, on scopolamine-induced memory decits.
36
CONCLUSION
is preliminary result demonstrates that NTF
was able to attenuate scopolamine-induced impairment
of the spatial cognitive performance, oxidative stress,
inflammation, and apoptosis in the brain cortex and
hippocampus, but its eects on cell signaling pathways
in these brain tissues remained ambiguous. e utility of
NTF as a potential preventive and therapeutic candidate
for AD requires further experimentation. In a future study,
Fig 6. Eects of NTF on scopolamine-induced dysfunction in signaling pathways indicated by the levels of p-JNK, p-p38, p-ERK1/2 and
p-Akt in: A frontal cortex, B hippocampus, and C temporal cortex. Each data bar (n=3) is expressed as mean ± SEM. *p < 0.05, **p < 0.01,
***p < 0.001 vs control group; ##p < 0.01, ###p < 0.001 vs scopolamine-treated group.
Volume 73, No.3: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
420
TABLE 1. Eects of NTF treatment on western blot protein expression in the frontal, hippocampus and temporal cortex in scopolamine-treated rats. Expression of target
proteins is represented as the percentage of control. Results are expressed as mean ± SEM of three independent experiments.
Frontal cortex Hippocampus Temporal cortex
NTF (1.5 mg/kg) - + + - + + - + +
Sco (3 mg/kg) + + - + + - + + -
ChAT 76.9 ± 0.90* 125.9 ± 4.70
###
101.1 ± 4.51 57.7 ± 1.41** 137.4 ± 0.73
###
141.6 ± 12.38** 52.9 ± 3.78** 119.4 ± 10.98
###
104.1 ± 4.11
BACE1 147.8 ± 4.45** 99.3 ± 2.43
##
114.3 ± 11.36 145.0 ± 1.23** 68.6 ± 9.74
###
80.7 ± 0.60 137.0 ± 6.40** 95.1 ± 8.43
##
81.8 ± 2.13
Bax 137.7 ± 0.67*** 112.33 ± 5.82
##
81.1 ± 2.90*** 188.5 ± 15.4*** 105.6 ± 7.41
###
42.3 ± 0.50** 143.0 ± 6.87*** 104.2 ± 0.19
###
76.2 ± 0.61*
Bcl-2 68.4 ± 4.11** 109.8 ± 6.23
##
98.9 ± 5.14 58.2 ± 4.01*** 189.7 ± 3.88
###
143.5 ± 5.11*** 54.0 ± 1.22*** 96.6 ± 1.94
###
119.8 ± 0.86***
Act. casp 3 211.2 ± 17.09*** 163.5 ± 1.85
#
97.2 ± 3.42 186.8 ± 2.32*** 112.2 ± 0.92
###
42.7 ± 0.83*** 148.1 ± 8.3** 83.4 ± 5.29
###
69.6 ± 5.92*
p-NFkB 224.8 ± 10.48*** 170.2 ± 9.14
##
105.3 ± 4.77 204.2 ± 7.93*** 115.1 ± 12.8
###
110.9 ± 3.90 130.1 ± 2.59* 91.1 ± 10.58
##
92.2 ± 5.04
IL-6 163.5 ± 4.31*** 133.7 ± 3.71
##
104.8 ± 4.28 153.6 ± 7.63*** 80.1 ± 2.50
###
53.9 ± 6.29** 160.2 ± 8.74*** 103.7 ± 6.28
###
60.6 ± 4.00**
IL-1β 190.5±5.52*** 118.7±15.4
##
117.2 ± 0.79 137.5 ± 2.36** 62.2 ± 3.73
###
40.7 ± 10.02*** 126.2 ± 9.26* 67.6 ± 5.94
###
63.4 ± 1.57**
p-JNK 120.2 ± 3.86** 77.8 ± 3.84
###
98.5 ± 3.10 133.6 ± 9.39* 70.8 ± 4.67
###
94.8 ± 7.94 159.7 ± 6.84*** 91.3 ± 2.56
###
85.6 ± 6.68
p38 171.2 ± 9.98** 91.9 ± 20.2
##
96.1 ± 3.33 145.2 ± 2.44*** 123.2 ± 2.26
##
107.9 ± 4.02 162.0 ± 4.81*** 109.0 ± 3.58
###
107.1 ± 3.53
p-ERK 36.9 ± 2.68* 124.7 ± 20.27
##
66.8 ± 14.29 55.1 ± 5.05** 93.4 ± 0.88
#
52.3 ± 11.29** 58.8 ± 4.43* 116.9 ± 3.28
##
101.7 ± 13.36
p-Akt 125.1 ± 16.05 134.0 ± 9.97 123.2 ± 9.56 90.3 ± 1.05 104.3 ± 3.20 103.2 ± 12.61 92.9 ± 4.52 55.5 ± 0.75
###
54.4 ± 2.02***
*p < 0.05, **p < 0.01, *** p < 0.001 compared to control;
#
p < 0.05,
##
p < 0.01,
###
p < 0.01 compared to scopolamine-treated group.
Thangnipon et al.
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
421
Original Article
SMJ
the eect of NTF on the altered activities of dierent cell
signaling pathways and their mechanisms in rat brain
tissues (frontal cortex, hippocampus, and temporal
cortex) are needed to be investigated and the sample size
should be expanded in order to acquire more conclusive
results.
ACKNOWLEDGEMENTS
We thanked Professor Prapon Wilairat for critical
and helpful suggestions on the manuscript. is project
was partly sponsored by a joint grant IRG5780009 between
Mahidol University and the ailand Research Fund
(TRF).
Conicts of Interest: All authors conrmed the absence
of potential conicts of interest as declared in the ICMJE
form.
REFERENCES
1. Gold CA, Budson AE. Memory loss in Alzheimer’s disease:
implications for development of therapeutics. Expert Rev
Neurother2008;8:1879-91.
2. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J
Med2010;362:329-44.
3. Schliebs R. Basal forebrain cholinergic dysfunction in Alzheimer’s
disease--interrelationship with β-amyloid, inammation and
neurotrophin signaling. Neurochem Res 2005;30:895-908.
4. Bajo R, Pusil S, López ME, Canuet L, Pereda E, Osipova D,
et al. Scopolamine eects on functional brain connectivity: a
pharmacological model of Alzheimer’s disease. Sci Rep 2015;5:
9748.
5. Kim MS, Lee DY, Lee J, Kim HW, Sung SH, Han JS, Jeon WK.
Terminalia chebula extract prevents scopolamine-induced
amnesia via cholinergic modulation and anti-oxidative eects
in mice. BMC Complement Altern Med 2018;18:136.
6. Tuchinda P, Pohmakotr M, Munyoo B, Reutrakul V, Santisuk T.
An azaanthracene alkaloid from Polyalthia suberosa. Phytochemistry
2000;53:1079-82.
7. angnipon W, Suwanna N, Kitiyanant N, Soi-Ampornkul R,
Tuchinda P, Munyoo B, et al. Protective role of N-trans-
feruloyltyramine against β-amyloid peptide-induced neurotoxicity
in rat cultured cortical neurons. Neurosci Lett 2012;513:229-
32.
8. Thangnipon W, Puangmalai N, Chinchalongporn V,
Jantrachotechatchawan C, Kitiyanant N, Soi-Ampornkul R, et
al. N-benzylcinnamide protects rat cultured cortical neurons
from β-amyloid peptide-induced neurotoxicity. Neurosci Lett
2013;556:20-25.
9. Thangnipon W, Suwanna N, Jantrachotechatchawan C,
Ngampramuan S, Tuchinda P, Nobsathian S. Protective roles
of N-benzylcinnamide on cortex and hippocampus of aged
rat brains. Arch Pharm Res 2015;38:1380-8.
10. Chen W, Guo J, Guo H, Kong X, Bai J, Long P. Protective
Eect of Vitamin C against Infancy Rat Corneal Injury Caused
by Acute UVB Irradiation. Biomed Res Int 2020;2020: 8089273.
11. Noviana R, Ilmiawan MI, Handini M. Synergistic Protective
Eect of Commercial Nigella Sativa Oil and Honey Combination
against Cisplatin-induced Nephrotoxicity in Rats. Jurnal Biotek
Medisiana Indonesia 2020;9:57-66.
12. Franzmeier N, Duering M, Weiner M, Dichgans M, Ewers M,
Alzheimer’s Disease Neuroimaging Initiative (ADNI). Le
frontal cortex connectivity underlies cognitive reserve in
prodromal Alzheimer’s disease. Neurology 2017;88:1054-61.
13. Ballinger EC, Ananth M, Talmage DA, Role LW. Basal forebrain
cholinergic circuits and signaling in cognition and cognitive
decline. Neuron 2016;91:1199-218.
14. Alafuzo I, Arzberger T, Al-Sarraj S, Bodi I, Bogdanovic N,
Braak H, et al. Staging of neurobrillary pathology in Alzheimer’s
disease: a study of the Brain Net Europe Consortium. Brain
Pathol 2008;18:484-96.
15. Pluta R, Kocki J, Ulamek-Koziol M, Petniak A, Gil-Kulik P,
Januszewski S, et al. Discrepancy in expression of β-secretase
and amyloid-β protein precursor in Alzheimer-related genes
in the rat medial temporal lobe cortex following transient
global brain ischemia. J Alzheimer Dis 2016;51:1023-31.
16. DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski
S, Bennett DA, et al. Upregulation of choline acetyltransferase
activity in hippocampus and frontal cortex of elderly subjects
with mild cognitive impairment. Ann Neurol 2002;51:145-55.
17. Hirokawa S, Nose M, Ishige A, Amagaya S, Oyama T, Ogihara
Y. Eect of Hachimi-jio-gan on scopolamine-induced memory
impairment and on acetylcholine content in rat brain. J
Ethnopharmacol 1996;50:77-84.
18. Piercey MF, Vogelsang GD, Franklin SR, Tang AH. Reversal of
scopolamine-induced amnesia and alterations in energy metabolism
by the nootropic piracetam: implications regarding identication
of brain structures involved in consolidation of memory traces.
Brain Res 1987;424:1-9.
19. Jang JY, Kim J, Shim J, Kim CY, Jang JH, Lee KW, Lee JH.
Decaeinated coee prevents scopolamine-induced memory
impairment in rats. Behav Brain Res 2013;245:113-9.
20. Rahimzadegan M, Soodi M. Comparison of Memory Impairment
and Oxidative Stress Following Single or Repeated Doses
Administration of Scopolamine in Rat Hippocampus. Basic
and Clinical Neuroscience 2018;9:4-14.
21. Ali-Melkkila T, Kanto J, Iisalo E. Pharmacokinetics and related
pharmacodynamics of anticholinergic drugs. Acta Anaesthesiol
Scand 1993;37:633-42.
22. Ebert U, Oertel R, Wesnes KA, Kirch, W. Eects of physostigmine
on scopolamine induced changes in quantitative
electroencephalogram and cognitive performance. Hum
Psychopharmacol 1998;13:199-210.
23. Semphuet T, Boongird A, Tantisira MH, Tiloksakulchai K,
Tapechum S, Pakaprot N. e Neuroprotective eect of Bacopa
monnieri against Pilocarpine-induced Status epilepticus in
rats. Siriraj Med J 2017;69:345-50.
24. Sattayasai J, Chaonapan P, Arkaravichie T, Soi-Ampornkul R,
Junnu S, Charoensilp P, et al. Protective eects of mangosteen
extract on H2O2-induced cytotoxicity in SK-N-SH cells and
scopolamine-induced memory impairment in mice. PLoS One
2013;8(12):e85053.
25. Katagiri T, Hatano N, Aihara M, Kawano H, Okamoto M, Liu Y,
et al. Proteomic analysis of proteins expressing in regions of rat
brain by a combination of SDS-PAGE with nano-liquid
chromatography-quadrupole-time of flight tandem mass
spectrometry. Proteome Sci 2010;8:41.
26. Xu QQ, Xu YJ, Yang C, Tang Y, Li L, Cai HB, et al. Sodium
Volume 73, No.6: 2021 Siriraj Medical Journal
https://he02.tci-thaijo.org/index.php/sirirajmedj/index
422
Tanshinone IIA sulfonate attenuates scopolamine-induced
cognitive dysfunctions via improving cholinergic system.
Biomed Res Int 2016;2016:9852536.
27. Ancelin ML, Christen Y, Ritchie K. Is antioxidant therapy a
viable alternative for mild cognitive impairment? Examination
of the evidence. Dement Geriatr Cogn Disord 2007;24:1-19.
28. Ghosh AK, Osswald HL. BACE1 (β-secretase) inhibitors for
the treatment of Alzheimer’s disease. Chem Soc Rev 2014;43:
6765-813.
29. Hafez HS, Ghareeb DA, Saleh SR, Abady MM, El Demellawy
MA, Hussien H, Abdel-Monem N. Neuroprotective eect of
ipriavone against scopolamine-induced memory impairment
in rats. Psychopharmacology (Berl) 2017;234:3037-53.
30. Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inammation.
Signal Transduct Target er 2017;2:17023.
31. Xu T, Shen X, Yu H, Sun L, Lin W, Zhang C. Water-soluble
ginseng oligosaccharides protect against scopolamine-induced
cognitive impairment by functioning as an antineuroinammatory
agent. J Ginseng Res 2016;40:211-9.
32. Ahmad A, Ramasamy K, Jaafar SM, Majeed AB, Mani V. Total
isoavones from soybean and tempeh reversed scopolamine-
induced amnesia, improved cholinergic activities and reduced
neuroinammation in brain. Food Chem Toxicol 2014;65:
120-8.
33. Guan QH, Pei DS, Zhang QG, Hao ZB, Xu TL, Zhang GY.
e neuroprotective action of SP600125, a new inhibitor of JNK,
on transient brain ischemia/reperfusion-induced neuronal
death in rat hippocampal CA1 via nuclear and non-nuclear
pathways. Brain Res 2005;1035:51-59.
34. Hildesheim J, Awwad RT, Fornace AJ Jr. p38 Mitogen-activated
protein kinase inhibitor protects the epidermis against the acute
damaging eects of ultraviolet irradiation by blocking apoptosis
and inammatory responses. J Invest Dermatol 2004;122:497-
502.
35. Tamagno E, Guglielmotto M, Giliberto L, Vitali A, Borghi R,
Autelli R, et al. JNK and ERK1/2 pathways have a dual opposite
eect on the expression of BACE1. Neurobiol Aging 2009;30:
1563-73.
36. Park SJ, Ahn YJ, Oh SR, Lee Y, Kwon G, Woo H, et al. Amyrin
attenuates scopolamine-induced cognitive impairment in
mice. Biol Pharm Bull 2014;37:1207-13.
Thangnipon et al.