1 s2.0 S0006899323002007 Main 2

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Brain Research 1814 (2023) 148429

Contents lists available at ScienceDirect

Brain Research
journal homepage: www.elsevier.com/locate/brainres

Investigation of the protective effect of long-term exercise on molecular


pathways and behaviours in scopolamine induced alzheimer’s
disease-like condition
Seda Kose a, *, Meltem Donmez Kutlu a, Samet Kara b, Sait Polat b, Kubra Akillioglu a
a
Cukurova University Medical Faculty, Department of Physiology, Division of Neurophysiology, Adana 01330, Turkey
b
Cukurova University Medical Faculty, Department of Histology and Embryology, Adana 01330, Turkey

A R T I C L E I N F O A B S T R A C T

Keywords: Despite research, the role of exercise in treatment and prevention of neurodegenerative diseases remains unclear.
Alzheimer’s disease Our study, investigated that protective effect of treadmill exercise on molecular pathways and cognitive be­
Amyloid beta haviours in a scopolamine-induced model of Alzheimer’s disease.
BDNF
For that purpose, male Balb/c mice subjected to exercise for 12 weeks. During the last 4 weeks of exercise,
Exercise
p-GSK3ßSer389
mice were given an injection of scopolamine (2 mg/kg). Following injection, open field test and Morris water
Scopolamine maze test were used to assess emotional-cognitive behaviour. Hippocampus and prefrontal cortex of mice were
isolated, and levels of BDNF, TrkB, and p-GSK3ßSer389 were assessed by western blotting, and levels of APP and
Aß-40 were analysed by immunohistochemistry.
In our study, scopolamine administration increased anxiety-like behaviour in open field test, while negatively
affecting spatial learning and memory in Morris water maze test. We found that exercise had a protective effect
against cognitive and emotional decline. Scopolamine decreased levels of p-GSK3ßSer389, BDNF in hippocampus
and prefrontal cortex.Whereas TrkB decreased in hippocampus and increased in prefrontal cortex. There was an
increase in p-GSK3ßSer389, BDNF, TrkB in the hippocampus, and p-GSK3ßSer389, BDNF in the prefrontal cortex in
the exercise + scopolamine group. Immunohistochemical analysis showed that scopolamine administration
increased APP and Aß-40 in hippocampus and prefrontal cortex in neuronal and perineuronal areas whereas Aß-
40 and APP were reduced in exercise + scopolamine groups.
In conclusion, long-term exercise may have a protective effect against scopolamine-induced impairments in
cognitive-emotional behaviour. It can be suggested that this protective effect is mediated by increased BDNF
levels and GSK3ßSer389 phosphorylation.

1. Introduction formed by amyloid beta (Aß) deposition. Aß is formed by two consecu­


tive cleavages of amyloid precursor protein (APP) by secretases (Chen
Alzheimer’s disease (AD) is one of the most common progressive et al., 2017). Aß plaques accumulate in the neocortex, posterior cingu­
neurodegenerative diseases in old age and accounts for 60–70% of de­ late, precuneus, and association areas of the limbic cortex (Miller-
mentia cases (Lane et al., 2018). An estimated 40 million people Thomas et al., 2016). Neurotoxic Aß plaques that accumulate outside the
worldwide are living with dementia. Most of them are over the age of 60 cell cause mitochondrial and synaptic damage and disrupt cell functions
(Scheltens et al., 2016). Today, the elderly population is growing as life by leading to hyperphosphorylation of the tau protein (Özkay et al.,
expectancy increases around the world due to widespread access to 2011). In addition, various cellular and molecular signalling pathways
healthcare and technological advances. It is estimated that the number triggered by inflammation lead to Aß accumulation (Akiyama and
of Alzheimer’s cases will triple by 2050 due to the growing elderly Barger, 2000). It has been reported that the synthesis and/or release of
population and the risk of developing dementia (Alzheimer’s, 2019). several inhibitory or excitatory neurotransmitters, particularly acetyl­
The most common pathological finding in AD is senile plaques choline (ACh), are altered in AD. ACh is known to play an important role

* Corresponding author at: Medical Faculty, University of Cukurova, 01330, Turkey.


E-mail address: kakillioglu@cu.edu.tr (S. Kose).

https://doi.org/10.1016/j.brainres.2023.148429
Received 16 February 2023; Received in revised form 17 May 2023; Accepted 27 May 2023
Available online 1 June 2023
0006-8993/© 2023 Elsevier B.V. All rights reserved.
S. Kose et al. Brain Research 1814 (2023) 148429

in learning, memory, and neuroplasticity (Kandimalla and Reddy, found evidence that GSK-3ß plays a role in the physiopathology of late-
2017). Slotkin et al. reported that decrease in choline acetyltransferase onset AD, its mechanisms of action have not been elucidated (Llorens-
(ChAT) enzyme activity and a decrease in ACh synthesis in AD (Slotkin Martin et al., 2014, Souder and Anderson, 2019). It is believed that the
et al., 1990). In summary, although the factors that cause AD are still discovery of GSK-3ß’s mechanisms of action on the pathways that lead to
under debate, cholinergic dysfunction, Aß accumulation, tau hyper­ AD will contribute to both a better understanding of the causes of the
phosphorylation, inflammation, DNA damage, and mitochondrial disease and the development of new treatments (Souder and Anderson,
damage all contribute to the development of the disease (Lane et al., 2019) Fig. 1a and b.
2018). Today, the fact that the benefits of pharmacological therapy in Alz­
Acetylcholine is abundant in the central nervous system. It is the heimer’s patients are very limited is leading researchers to investigate
main neurotransmitter active in the cerebral cortex, basal ganglia, and non-pharmacological methods (Zucchella et al., 2018). In parallel, it is
basal forebrain (Hampel et al., 2018). Acetylcholine is involved in many believed that exercise, which is a non-pharmacological method, may be
important physiological processes, such as learning memory, attention, an effective way to treat age-related neurodegenerative diseases, and
stress response, and sleep-wakefulness (Ferreira-Vieira et al. (2016)). research is underway in this area. For example, several studies have
Cholinergic loss in AD is due to the degeneration of cholinergic neurons shown that exercise can protect against neuronal damage through
in the nucleus basalis Meynert (NBM) and their axons that project to the BDNF, increase plasticity in the brain, and thus have a positive effect on
cerebral cortex. Nicotinic and muscarinic receptors in the cerebral cor­ cognitive disorders, (Koo et al., 2013, Vaynman et al., 2004, Xu et al.,
tex are also altered in AD (Hampel et al., 2018). The acquisition, storage, 2021). In a study using scopolamine to induce an amnesia model in
and use of new information depend on adequate cholinergic trans­ mice, it was shown that the spatial learning ability of scopolamine-
mission (Lim et al., 2015). Therefore, cognitive and emotional behav­ treated animals was impaired and that this impairment was improved
iour can be greatly affected by disruptions in cholinergic transmission. by treadmill exercise. The same study reported that treadmill exercise
The non-selective muscarinic receptor antagonist scopolamine has also improved spatial learning in healthy mice (Heo et al., 2014). The
been used experimentally in neuropsychopharmacology to induce aim of our study was to investigate the protective effect of long-term
cognitive deficits. As M1 and M5 receptors are more abundant in the exercise on emotional-cognitive behaviour and changes at the cellular
CNS, it is thought that the effects of scopolamine are mediated through level induced by scopolamine in the AD model. For this purpose, the
these receptors. (Klinkenberg and Blokland, 2010). Cholinergic trans­ effect of scopolamine, administered for 28 days to Balb/c mice that had
mission indicates its effects on emotional and cognitive functions mainly undergone 12 weeks of treadmill exercise, was assessed on cognitive
through the M1 receptor. Several studies have shown that scopolamine behaviour using the Morris water maze (MWM) test and on emotional
affects locomotor activity and impairs emotional and cognitive func­ behaviour and locomotor activity using an open field test. At the cellular
tions. A study has shown that infusing scopolamine into the amygdala level in the hippocampus and prefrontal cortex, BDNF, tyrosine receptor
increases locomotor activity. (Nomura et al., 1994). In another study, kinase-B (TrkB), and phosphorylated GSK-3ß Ser389 (p-GSK3ßSer389)
infusion of scopolamine into the hippocampus did not affect locomotor levels were assessed by Western blotting, and APP and Aß-40 levels were
activity but increased anxious behaviour (Smythe et al., 1996). It has analysed by immunohistochemistry. We chose the Balb/c mouse strain
been reported that rats given scopolamine showed hyperactivity in the for this study because there are some emotional behaviours that may be
open field test and memory impairment in the MWM test (Jafarian et al., specific to the Balb/c strain. For example, Balb/c mice show more
2019). In our study, the AD-like condition was induced by scopolamine anxiety-like behaviour and are susceptible to social defeat stress (Groves
(a non-selective muscarinic receptor antagonist). As mentioned in the et al., 2013, Mehta and Schmauss, 2011).
literature, this model is often used as a good model for learning and
memory impairment and causes Alzheimer’s-like physiopathological 2. Results
changes in the brain. In addition, the fact that scopolamine is easy to use,
has no serious side effects, and is cheaper than other methods is one of 2.1. Body weight
the reasons why it was preferred in our study.
Signaling pathways involving the activation of glycogen synthase In body weight measurements ANOVA found a significant difference
kinase-3ß (GSK-3ß) are one of the mechanisms proposed to explain the between weeks in the S-Saline [χ2 (11) = 152.3p < 0.001], E-Saline [χ2
physiopathological processes involved in the development of AD. (11) = 162.9p < 0.001], S-SCOP [χ2 (11) = 103.4p < 0.001], and E-
Glycogen synthase kinase-3 (GSK-3) is a structurally active serine- SCOP [χ2 (11) = 129.6p < 0.001] groups. There was a significant in­
threonine kinase found in all tissues. GSK-3ß is active in cells that are crease in body weight in all groups compared to the first week in all
not normally stimulated. GSK-3ß is rendered inactive by the phosphor­ weeks (p < 0.01).
ylation of its specific sites. This enzyme is rendered inactive by phos­ In body weight comparisons between groups, the second week [H
phorylation of Ser9, Ser21, and Ser389. GSK-3ß is involved in (3,53) = 8.32p = 0.04], third week [H (3,53) = 26.14p < 0.001], fourth
remodelling cell morphology by phosphorylating cytoskeletal proteins week [H (3,53) = 19.24p < 0.001], sixth week [H (3,53) = 9.24p =
that make up the cytoskeleton (microtubule-related proteins, tau, and 0.02], and seventh week [H (3,53) = 13.17p = 0.004] between groups
kinesin light chain) in the central nervous system, affecting functions significant difference was detected. A significant decrease was detected
such as synaptic plasticity, neurofilament stabilisation, and new synapse in the E-Saline group compared to the S-Saline group on the third (U =
formation (Hur and Zhou, 2010). At the same time, GSK-3ß can also 30.0p < 0.001 z = − 3.49) and, fourth (U = 59.0p = 0.02 z = − 2.28)
trigger processes that lead to the accumulation of Aß. GSK-3ß plays a role weeks. A significant decrease was found in the E-SCOP group on the
in the pathogenesis of AD by upregulating Aß production and sup­ third (U = 10.5 p < 0.001 z = − 3.49), fourth (U = 9.5 p < 0.001 z =
pressing acetylcholine synthesis (Tang, 2019). M1 receptors, which are − 3.54), sixth (U = 28.5 p = 0.01 z = − 2.39) and, seventh (U = 14.5 p =
abundant in the hippocampus and prefrontal cortex, have been shown to 0.001 z = − 3.27) weeks compared to the S-SCOP group. There was no
influence the cleavage process of APP. M1 receptor activation through significant difference between the groups in the first week [H (3,53) =
the PKC pathway leads to an increase in sAPPα and a decrease in Aß by 1.58p = 0.66], fifth week [H (3,53) = 6.19p = 0.10], eighth week [H
inhibiting BACE, while it reduces Aß-induced apoptosis and p-tau by (3,53) = 1.03p = 0.79], ninth week [H (3,53) = 3.43p = 0.33], tenth
inhibiting GSK-3ß (Fisher, 2012). GSK-3ß has also been shown to be week [H (3,53) = 3.94p = 0.26], eleventh week [H (3,53) = 3.09p =
involved in the conversion of short-term memory into long-term mem­ 0.37], and twelfth week [H (3,53) = 1.70p = 0.63] Fig. 2.
ory and the formation of new neurons (Llorens-Martin et al., 2014).
Studies in mouse models of AD have reported that abnormal activation
of GSK-3ß leads to the loss of dendritic spines. Although studies have

2
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 1. a) Muscarinic receptors of acetylcholine, b) possible effect of scopolamine on beta amyloid deposition.

3
S. Kose et al. Brain Research 1814 (2023) 148429

2.3. Morris water maze test

2.3.1. Training trials


In the Morris water maze test, ANOVA found a significant difference
between days in the S-Saline group escape latency for five days in the
training trials [χ2 (4) = 26.2p < 0.001]. Compared to the first day, the
escape latency was significantly reduced on the second (Z = − 2.3 p =
0.02), third (Z = − 2.02p = 0.04), fourth (Z = − 2.09p = 0.03), and fifth
(Z = − 3.58p < 0.001) days. In the E-Saline group, ANOVA detected a
significant difference between days in the escape latency [χ2 (4) = 25.7
p < 0.001]. A significant decrease was found on the fourth (Z = − 2.5 p
= 0.011) and fifth (Z = − 4.3 p < 0.001) days compared to the first day.
In the training trials, there was a significant difference between days in
the S-SCOP group in the escape latency [χ2 (4) = 17.5 p = 0.002]. In the
escape latency, a significant decrease was found in the time to find the
platform on the second (Z = − 2.7 p = 0.006), third (Z = − 4.0 p <
0,001), fourth (Z = − 2.7 p = 0.006), and fifth (Z = − 2.7 p = 0.007) days
compared to the first day. ANOVA detected a significant difference be­
Fig. 2. Body weight for twelve weeks. The data are presented as the mean ± tween days in the escape latency in the E-SCOP group [χ2 (4) = 10.8 p =
standard error. N = 11–15. For statistical analysis, see inside the text. 0.028]. Compared to the first day, a significant decrease was found in
the escape latency on the second (Z = − 2.6 p = 0.008), third (Z = − 3.3 p
2.2. Open field test = 0.001), fourth (Z = − 2.8 p = 0.004), and fifth (Z = − 2.6 p = 0.007)
days.
In the open field test, there was a significant drug [F (1,49) = 39.17p There was a significant difference between the groups in the escape
< 0.001] and exercise [F (1,49) = 7.18p = 0.01] effect in time spent in latency on the first [H (3,212) = 17.04p = 0.001], fourth [H (3,212) =
the center, but no drug × exercise [F (1,49) = 1.35p = 0.25] interaction. 10.04p = 0.01], and fifth [H (3,212) = 19.16p < 0.001] days of the
The time in the center was significantly reduced in the S-SCOP group training trials. When the E-Saline group was compared with the S-Saline
compared to the S-Saline group (p < 0.01). It increased significantly in group, no significant difference was found in the first (U = 1618.0p =
the E-Saline group compared to the S-Saline (p = 0.02) and E-SCOP (p < 0.3 z = − 1.0), second (U = 1760.5p = 0.8 z = − 0.2), third (U = 1701.0p
0.001) groups. No significant difference was found between the E-SCOP = 0.5 z = − 0.5), fourth (U = 1556.0p = 0.1 z = − 0.1) and fifth (U =
group and the S-SCOP group (p = 0.74) Fig. 3a. 1553.0p = 0.1 z = − 0.1) days. When the S-SCOP group was compared
In the open field test, significant drug effect [F (1,49) = 30.25p < with the S-Saline group, there was a significant increase in the escape
0.001], exercise effect [F (1,49) = 31.26p < 0.001] and drug × exercise latency on the first (U = 980.0p = 0.006 z = − 2.7), fourth (U = 992.0p
interaction [F (1,49) = 4.48p = 0.039] were found. Distance traveled = 0.02 z = − 2.2) and fifth (U = 840.0p = 0.001 z = − 3.2) days, but no
increased significantly in the E-Saline group compared to the S-Saline difference was found on the second (U = 1098.0p = 0.1 z = − 1.5) and
group (p = 0.049). It increased significantly in the E-SCOP group third (U = 1263.5p = 0.6 z = − 0.3) days. There was a significant in­
compared to the E-Saline (p < 0.001) and S-SCOP (p < 0.001) groups. crease in the escape latency on the first (U = 1058.5p = 0.008 z = − 2.6)
There was a tendency to increase in distance traveled in the S-SCOP and fifth (U = 1015.5p = 0.008 z = − 2.6) days when the E-SCOP group
group compared to the S-Saline group (p = 0.09) Fig. 3b. was compared with the E- Saline group, but there was no significant
There was a significant drug [F (1,49) = 4.06p < 0.05] and exercise difference on the second (U = 1436.5p = 0.9 z = − 0.0), third (U =
[F (1,49) = 43.79p < 0.001] effect on the rearing, but no drug × exercise 1434.0p = 0.9 z = − 0.0) and fourth (U = 1244.0p = 0.2 z = − 1.2) days.
[F (1,49) = 0.10p = 0.75] interaction. There was a significant increase in There was no significant difference in the escape latency on the first (U
the frequency of rearing in the E-Saline group compared to the S-Saline = 948.0p = 0.2 z = − 1.1), second (U = 910.5p = 0.2 z = − 1.2), third (U
(p < 0.001) group, and in the E-SCOP group compared to the S-SCOP (p = 1042.0p = 0.9 z = − 0.1), fourth (U = 876.5p = 0.1 z = − 1.5) and fifth
= 0.001) group. No significant difference was found in the S-SCOP group (U = 941.5p = 0.3 z = − 0.9) days when the E-SCOP group was
compared to the S-Saline group (p = 0.63) and in the E-SCOP group compared with the S-SCOP group Fig. 4a.
compared to the E-Saline group (p = 0.34) Fig. 3c. In the Morris water maze test, ANOVA found a significant difference
While there was a significant exercise effect [F (1,49) = 15.60p < between days in the S-Saline group in swimming speed for five days in
0.001] and drug × exercise interaction [F (1,49) = 4.85p < 0.05] on the the training trials [χ2 (4) = 17.14p < 0.01]. Swimming speed decreased
frequency of entry to the center, no drug effect [F (1,49) = 0.36p = 0.54] significantly on the third (Z = − 2.10p = 0.03), fourth (Z = − 3.22p =
was found. A significant increase was found in the frequency of entry to 0.001) and fifth (Z = − 3.34p = 0.001) days compared to the first day.
the center, in the E-SCOP group compared to the S-SCOP group (p = ANOVA detected a significant difference between days in swimming
0.001). No significant difference was found in the E-Saline and S-SCOP speed and duration in the E-Saline group [χ2 (4) = 32.22p < 0.001].
groups compared to the S-Saline group (p = 0.54, p = 0.68, respec­ Swimming speed decreased significantly on the third (Z = − 2.27p =
tively), and in the E-SCOP group compared to the E Saline group (p = 0.02), fourth (Z = − 3.90p < 0.001), and fifth (Z = − 4.63p < 0.001) days
0.20) Fig. 3d. compared to the first day. In the training trials, there was a significant
In the open field test, entry time to the center found a significant difference between the days in the S-SCOP group in swimming speed [χ2
exercise [F (1,49) = 33.70p < 0.001] effect, but no drug [F (1,49) = (4) = 9.8p = 0.043]. A decrease in swimming speed was detected on the
3.35p = 0.073] effect and drug × exercise [F (1,49) = 3.49p = 0.68] fourth day compared to the first day (Z = − 1.9p = 0.04). ANOVA found
interaction. Entry time to the center decreased in the E-Saline group no significant difference in swimming speed between days in the E-SCOP
compared to the S-Saline group, and in the E-SCOP group compared to group [χ2 (4) = 8.6p = 0.071].
the S-SCOP group (p < 0.05, p < 0.001, respectively). A trend to increase In the Morris water maze test, a significant difference was found
in entry time to the center was found in the S-SCOP group compared to between the groups on the first [H (3,212) = 16.50p = 0.001], third [H
the S-Saline group (p = 0.06). No significant difference was found in the (3,212) = 15.20p = 0.002], fourth [H (3,212) = 9.32p = 0.025] and fifth
E-SCOP group compared to the E-Saline group (p = 1.00) Fig. 3e. [H (3,212) = 21.43p < 0.001] days in swimming speed in the training
trials. When the E-Saline group was compared with the S-Saline group,

4
S. Kose et al. Brain Research 1814 (2023) 148429

a)
100

Time spent in the center (s)


80

60 ###

**
40

20 S-Saline
S-SCOP
E-Saline
E-SCOP
0

b) 6000 c)
###,^^^ 70

5000
60
***
Distance traveled (cm)

^^

Frequency of rearing
4000 50

*
40
3000

30
2000
20
S-Saline S-Saline
1000 S-SCOP S-SCOP
E-Saline 10 E-Saline
E-SCOP E-SCOP
0 0

d) 60
e) 40
^^
Frequency of entry to the center

50
Entry time to the center (s)

30

40

30 20

20
* ^^^

10
S-Saline S-Saline
10 S-SCOP S-SCOP
E-Saline E-Saline
E-SCOP E-SCOP

0 0

Fig. 3. a)time spent in the center (s). *p < 0,05, **p < 0,001 compared to S-Saline group, ###p < 0,001 compared to E-Saline group. b) The distance traveled (cm).
*p < 0,05 compared to S-Saline group, ###p < 0,001 compared to E-Saline group, ^^^p < 0,001 compared to S-SCOP group. c) Frequency of rearing. ***p < 0,001
compared to S-Saline group, ^^p < 0,01 compared to S-SCOP group. d) Frequency of entry to the center. ^^p < 0,01 compared to S-SCOP group. e) Entry time to the
center (s). *p < 0,05 compared to S-Saline group, ^^p < 0,01 compared to S-SCOP group. ANOVA was performed followed by Tukey HSD test. The data are presented
as the mean ± standard error. N = 11–15.

there was an increase in swimming speed on the first (U = 1309.0 p = the quadrant with the platform in by removing the platform was on the
0.01 z = − 2.5) day but no significant difference was found on the other 6th day. On the 6th day, a significant difference was found between the
days. When the S-SCOP group was compared with the S-Saline group, a groups when looking at the time spent on the target quadrant where the
significant increase in swimming speed was detected on the first (U = platform is located [H (3,53) = 21.52p < 0.001]. The time spent in the
935.0 p = 0.011 z = − 2.5), third (U = 975.0 p = 0.023 z = − 2.2), fourth target quadrant was significantly reduced in the S-SCOP group
(U = 946.0 p = 0.014 z = − 2.4) and fifth (U = 729.0 p < 0.001 z = − 3.8) compared to the S-Saline group (U = 7.0p < 0.001 z = − 3.9). There was
days. Compared to the E-Saline group, there was a significant increase in no significant difference in the time spent in the target quadrant in the E-
swimming speed on the third (U = 1049.0 p = 0.016 z = − 2.4) and fifth Saline group compared to the S-Saline group (U = 108.0p = 0.852 z =
(U = 1041.0 p = 0.014 z = − 2.4) days in the E-SCOP group but no − 0.1). A decreasing tendency was found in the E-SCOP group compared
significant difference was found on the other days. No significant dif­ to the E-Saline group (U = 51.5p = 0.06 z = − 1.8). There was a sig­
ference was found between the groups when the E-SCOP group was nificant increase in the time spent in the target quadrant in the E-SCOP
compared with the S-SCOP group Fig. 4b. group compared to the S-SCOP group (U = 19.5p = 0.004 z = − 2.8)
Fig. 5a.
2.3.2. Test There was no significant difference in swimming speed between the
Mice, whose training trials were completed for five days in the Morris groups on the test day [H (3,53) = 1.038p = 0.79] Fig. 5b.
water maze, were taken to the experiment from the opposite quadrant of

5
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 4. a) escape latency in the morris water maze (s). b) swimming speed (cm/s). *p < 0,05 **p < 0,01 compared to S-Saline group, #p < 0,05, ##p < 0,01
compared to E-Saline group. Friedman’s ANOVA test was performed follewed by Mann-Whitney U test. The data are presented as the mean ± standard error. N
= 11–15.

Fig. 5. a) time in target quadrant in the morris water maze (s). b) swimming speed (cm/s). ***p < 0,001 compared to S-Saline group, ^^p < 0,01 compared to S-SCOP
group. Kruskal Wallis was performed folewed by Mann Whitney U test. The data are presented as the mean ± standard error. N = 11–15.

2.4. Immunohistochemical analysis noted. This expression was particularly evident in the cytoplasm of
neurons. In addition, it was observed that there was no immunoreac­
The hippocampus and prefrontal cortex tissues were taken to tivity in glial cells Fig. 7b.
determine the expression of Aß-40 and APP was analysed by immuno­ When hippocampus tissue sections obtained from the E-Saline group,
histochemical method. No immune markers were observed in the hip­ were examined for Aß-40 immunoreactivity, it was observed that there
pocampus and prefrontal cortex sections where no primary antibody was no expression in the cytoplasm of neurons and glial cells. In addi­
was administered as a negative control Fig. 6a, 6b. tion, no immunoreactivity was observed in the perineuronal area Fig. 7c.
When the hippocampus tissue sections obtained from the S-Saline In the evaluation of hippocampus tissue sections obtained from the
group were evaluated for Aß-40 immunoreactivity, no immunostaining E-SCOP group in terms of Aß-40 expression, the presence of rather
was observed in the cytoplasm of neurons and glial cells. In addition, no minimal Aß-40 expression in the cytoplasm and perineuronal areas of
staining was observed in nerve fibers and perineuronal areas Fig. 7a. neurons located in the CA1 region was remarkable. In addition, while
In the evaluation of hippocampus tissue sections obtained from the S- immunoreactivity was not observed in many neuron cytoplasms in this
SCOP group for Aß-40 expression, the presence of pronounced Aß-40 region, Aß-40 expression was observed in a small number of neuron
expression in the cytoplasm of neurons and perineuronal areas was cytoplasms. However, no expression was observed in the cytoplasm of

Fig. 6. Immunohistochemical analysis of the prefrontal cortex and hippocampus. a) No immune markers were observed in hippocampus sections where no primary
antibody was administered as a negative control. b) No immune markers were observed in prefrontal cortex sections where no primary antibody was administered as
a negative control. Bar = 50 μm.

6
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 7. Immunohistochemical analysis of the hippocampus. a) Aß-40 immunoreactivity of the S-Saline group, No staining is observed in neurons (arrows), glial cells
(arrow heads) and perineuronal areas (*) in hippocampus b) Aß-40 immunoreactivity of the S-SCOP group, Strong expression of Aß-40 immunoreactivity in neuron
cytoplasm (ok) is observed. In addition to this, it appears that Aß-40 immunoreactivity is not expressed in glia cells (arrowhead), c) Aß-40 immunoreactivity of the E-
Saline Group, It is observed that Aß-40 immunoreactivity is not expressed in neuron cytoplasms (arrow) and glia cells (arrow head), d) Aß-40 immunoreactivity of the
E-SCOP group, Aß-40 immunoreactivity is observed in neurons, neuron cytoplasm and perineuronal areas (ok). It is observed that Aß-40 is not expressed in glia cells
(arrowhead), e) APP immunoreactivity of the S-Saline group, high expression of APP immunoreactivity is observed in the cytoplasm of neurons (arrow). In glia cells,
APP expression (arrow head) is not observed, f) APP immunoreactivity of the S-SCOP group, Strong expression of APP immunoreactivity in the cytoplasm of neurons
(arrow) is observed. However, APP is not expressed in glia cells (arrow head) or its weak expression is observed in some glia cells, g) APP immunoreactivity of the E-
Saline group, Strong expression of APP immunoreactivity is observed in the cytoplasm of neurons (arrow). Whereas in glia cells, there is no expression of APP or its
weak expression (arrowhead) is observed in some glial cells, h) APP immunoreactivity of the E-SCOP group, high expression of APP immunoreactivity in the
cytoplasm of neurons (arrow) is observed. However, APP is not expressed in glia cells (arrow head) or its weak expression is observed in some glia cells. Bar = 50 μm,
k) Aß-40, l) APP, **p < 0.01, ***p < 0.001 compared to S-Saline group, ^^^p < 0.001 compared to S-SCOP group ###p < 0.001 compared to E-Saline group. Mann
Whitney U test.

7
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 7. (continued).

glial cells Fig. 7d. obtained from the E-SCOP group for APP, the presence of pronounced
When the APP immunoreactivity of the hippocampus sections ob­ APP immunoreactivity was noted both in the cytoplasm of neurons and
tained from the S-Saline group were evaluated, significant APP immu­ in the perineuronal areas. No immunoreactivity was observed in the
noreactivity was observed in the cytoplasm of the neurons and the cytoplasm of glial cells Fig. 8h.
perineuronal areas. In contrast, APP immunoreactivity was not observed
in glial cells Fig. 7e.
In the examination of hippocampus tissue sections obtained from the 2.5. Western blot analysis
S-SCOP group for APP expression, significant immunoreactivity was
observed in the cytoplasm of neurons and perineuronal areas. However, A significant difference was found between the groups in the
no APP immunoreactivity was observed in glial cells Fig. 7f. expression of TrkB in the hippocampus [H (3, 16) = 13.29p = 0.004].
In the evaluation of hippocampus tissue sections obtained from the TrkB expression was significantly decreased in the S-SCOP group
E-Saline group in terms of APP immunoreactivity, the presence of compared to the S-Saline group (U = 0.0p = 0.01 z = − 2.4). TrkB
expression in the cytoplasm of neurons and perineuronal areas was expression increased significantly in the E-SCOP group compared to the
noted. However, no APP expression was observed in glial cells Fig. 7g. S-SCOP group (U = 0.0p = 0.02 z = − 2.3). TrkB expression increased
In the evaluation of hippocampus tissue sections obtained from the significantly in the E-SCOP group compared to the E-Saline group (U =
E-SCOP group in terms of APP immunoreactivity, the presence of sig­ 0.0p = 0.01 z = − 2.3). There was no significant difference in TrkB
nificant APP expression in the cytoplasm of neurons and perineuronal expression in the E-Saline group compared to the S-Saline group (U =
areas was noted. However, no APP immunoreactivity was observed in 4.0p = 0.21 z = − 1.2).
glial cells Fig. 7h. There was a significant difference between the groups in the
Prefrontal cortex tissue sections taken from the S-Saline group were expression of BNDF in the hippocampus [H(3, 16) = 11.13p = 0.011]. A
also evaluated for Aß-40 immunoreactivity. Aß-40 expression was not significant increase in BDNF expression was found in the E-Saline group
observed in the neuron cytoplasm and perineuronal area. In addition, no compared to the S-Saline group (U = 0.0p = 0.01 z = − 2.4). BDNF
Aß-40 immunoreactivity was observed in glial cells Fig. 8a. expression was significantly decreased in the S-SCOP group compared to
The evaluation of prefrontal cortex tissue sections obtained from the the S-Saline group (U = 0.0p = 0.014 z = − 2.4). BDNF expression
S-SCOP group for Aß-40 revealed the presence of Aß-40 expression in the increased significantly in the E-SCOP group compared to the S-SCOP
cytoplasm of neurons and perineuronal areas. While expression was not group (U = 1.0p = 0.01 z = − 2.3). There was no significant difference in
observed in the majority of glial cells, the presence of minimal immu­ BDNF expression in the E-SCOP group compared to the E-Saline group
noreactivity in a few glial cells was noted Fig. 8b. (U = 4.0p = 0.24 z = − 1.1).
Aß-40 immunoreactivity was evaluated in prefrontal cortex tissue Significant difference was found between groups in hippocampus p-
sections taken from mice belonging to the E-Saline group. In the eval­ GSK3ßSer389 expression [H (3, 16) = 12.98p = 0.005]. p-GSK3ßSer389
uation for Aß-40 immunoreactivity, there was no immunoreactivity in expression was significantly decreased in the S-SCOP group compared to
the cytoplasm of neurons and glial cells, and perineuronal areas Fig. 8c. the S-Saline group (U = 0.0p = 0.014 z = − 2.4). p-GSK3ßSer389
Immunohistochemical evaluation was performed for Aß-40 anti­ expression increased significantly in the E-SCOP group compared to the
bodies in prefrontal cortex tissues taken from mice belonging to the E- S-SCOP group (U = 0.0p = 0.02 z = − 2.3). There was no significant
SCOP group. In the examination of Aß-40 in the tissue sections taken, the difference in p-GSK3ßSer389 expression in the E-SCOP group compared to
presence of minimal expression of Aß-40 was observed in a small the E-Saline group (U = 6.0p = 0.56 z = − 0.5) Fig. 9a – d.
number of neurons. In addition, no expression was observed in glial cells Significant difference was found between groups in prefrontal cortex
Fig. 8d. TrkB expression [H(3, 16) = 7.8p = 0.04]. TrkB expression increased
In the examination of prefrontal cortex tissue sections obtained from significantly in the S-SCOP group compared to the S-Saline group (U =
the S-Saline group for APP, pronounced immunoreactivity of APP was 0.0p = 0.013 z = − 2.4). TrkB expression increased significantly in the E-
observed in the cytoplasm of neurons and the perineuronal space. In SCOP group compared to the E-Saline group (U = 0.0p = 0.01 z = − 2.3).
addition, no APP immunoreactivity was observed in glial cells in the There was no significant difference in TrkB expression in the E-Saline
prefrontal cortex Fig. 8e. group compared to the S-Saline group (U = 4.0p = 0.12 z = 0.0) and E-
In the examination of prefrontal cortex tissue sections obtained from SCOP group compared to S-SCOP (U = 8.0p = 1.00 z = 0.0).
the S-SCOP group for APP, significant APP expression was observed in There was also a significant difference between the groups in pre­
the cytoplasm of neurons and perineuronal areas. However, no APP frontal cortex BDNF expression [H(3, 16) = 10.07p = 0.018]. A signif­
immunoreactivity was observed in glial cells Fig. 8f. icant increase in BDNF expression was found in the E-Saline group
In contrast, the evaluation of prefrontal cortex tissue sections ob­ compared to the S-Saline group (U = 0.0p = 0.01 z = − 2.4). BDNF
tained from the E-Saline group for APP, showed significant APP expression was significantly decreased in the S-SCOP group compared to
expression in the cytoplasm of neurons and the perineuronal area. the S-Saline group (U = 0.0p = 0.014 z = − 2.4). BDNF expression
However, no immunoreactivity was observed in glial cells Fig. 8g. increased significantly in the E-SCOP group compared to the S-SCOP
However, in the evaluation of prefrontal cortex tissue sections group (U = 0.0p = 0.02 z = − 2.3). There was no significant difference in
BDNF expression in the E-SCOP group compared to the E-Saline group

8
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 8. Immunohistochemical analysis of the prefrontal cortex. a) Aß-40 immunoreactivity of the S-Saline group, It is observed that Aß-40 immunoreactivity is not
expressed in neuron cytoplasms (arrow) and glia cells (arrow head), b) Aß-40 immunoreactivity of the S-SCOP group, high expression of Aß-40 immunoreactivity in
neuron cytoplasm (ok) is observed. However, it is observed that Aß-40 immunoreactivity is not expressed in glia cells (arrow head), c) Aß-40 immunoreactivity of the
E-Saline group, It is observed that Aß-40 immunoreactivity is not expressed in neuron cytoplasms (arrow) and glia cells (arrow head), d) Aß-40 immunoreactivity of
the E-SCOP group, Weak expression of Aß-40 immunoreactivity in the cytoplasm and perineuronal area of some neurons is observed (arrow). Expression of this
antibody (arrow head) is not observed in glia cells, e) APP immunoreactivity of the S-Saline group, high expression of APP immunoreactivity in the cytoplasm of
neurons (arrow) is observed. However, it is observed that APP is not expressed in glia cells (arrowhead), f) APP immunoreactivity of the S-SCOP group, high
expression of APP immunoreactivity in the cytoplasm of neurons (arrow) is observed. However, it is observed that APP is not expressed in glia cells (arrow head), g)
APP immunoreactivity of the E-Saline group, Strong expression of APP immunoreactivity in the cytoplasm of neurons (arrow) is observed. However, APP is not
expressed in glia cells (arrow head) or its weak expression is observed in some glia cells, h) APP immunoreactivity of the E-SCOP group, Strong expression of APP
immunoreactivity in the cytoplasm of neurons (arrow) is observed. However, APP is not expressed in glia cells (arrow head) or poor expression of this antibody is
observed in some glia cells, Bar = 50 μm. k) Aß-40, l) APP, *p < 0.05, ***p < 0.001 compared to S-Saline group, ^^^p < 0.001 compared to S-SCOP group, ###p <
0.001 compared to E-Saline group. Mann Whitney U test.

9
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 8. (continued).

a) b) 1,8

1,6 ^,#
1,4

1,2

TrkB (fold change)


1,0

0,8
*
0,6

0,4 S-Saline
S-SCOP
0,2 E-Saline
E-SCOP
0,0

c) 1,2
d) 1,6
* ^ ^
1,4
1,0
(fold change)

1,2
BDNF (fold change)

0,8
* 1,0

0,6
0,8
Ser389

*
0,4 0,6
p-GSK3

S-Saline 0,4
S-Saline
0,2 S-SCOP
S-SCOP
E-Saline
E-SCOP 0,2 E-Saline
E-SCOP
0,0
0,0

Fig. 9. a)western blot image of hippocampus. b) TrkB fold change, c) BDNF fold change, d) p-GSK3ßSer389 fold change. *p < 0,05 compared to S-Saline group, ^p <
0,05 compared to S-SCOP group, #p < 0,01 compared to E-Saline group. Kruskal Wallis was performed followed by Mann Whitney U test. N = 4.

Fig. 10. a)western blot image of prefrontal cortex. b) TrkB fold change, c) BDNF fold change, d) p-GSK3ßSer389fold change, *p < 0,05 compared to S-Saline group, ^p
< 0,05 compared to S-SCOP group. ##p < 0.001 compared to E-Saline group. Kruskal Wallis was performed followed by Mann Whitney U test. N = 4.

10
S. Kose et al. Brain Research 1814 (2023) 148429

(U = 8.0p = 1.00 z = 0.0). NMDA receptor-mediated long-term potentiation (LTP), and its main­
There was a significant difference between the groups in the tenance (Kocahan and Doğan, 2017, Navakkode and Korte, 2012). In
expression of p-GSK3ßSer389 in the prefrontal cortex [H (3, 16) = 14.32p addition, cholinergic neurons reflected from the basal forebrain to the
= 0.002]. p-GSK3ßSer389 expression was significantly decreased in the S- hippocampus play an important role in the formation of learning and
SCOP group compared to the S-Saline group (U = 0.0p = 0.014 z = memory. Acetylcholine is known to act by facilitating NMDA receptor-
− 2.4). p-GSK3ßSer389 expression increased significantly in the E-SCOP mediated transmission in the hippocampus through muscarinic and
group compared to the S-SCOP group (U = 0.0p = 0.021 z = − 2.3). p- nicotinic receptors. Processes such as inhibition of GABA release,
GSK3ßSer389 expression was significantly decreased in the E-SCOP group blockade of potassium permeability, and activation of intracellular sig­
compared to the E-Saline group (U = 0.0p = 0.021 z = − 2.3) Fig. 10a – nalling pathways are reported to lower the membrane threshold for LTP
d. formation (Krnjevic et al., 1988). Gil-Bea et al. reported in their study
that increasing BDNF expression via muscarinic receptors is important
3. Discussion for memory formation in the hippocampus (Gil-Bea et al., 2011). They
suggest that BDNF activates the intracellular signalling pathways
The present study showed that long-term physical exercise may necessary for synaptic plasticity in neurons, such as the formation of new
protect against the decrease in memory performance and anxiety-like synapses through TrkB receptors with an autocrine effect. Although
behaviour observed with scopolamine administration in Balb/c mice. there are many studies in the literature reporting a decrease in BDNF
Our main findings were as follows: (1) scopolamine administration levels with scopolamine administration, the intracellular signalling
increased anxiety-like behaviour in the open field test, while negatively pathways involved in the reduction of BDNF levels due to a decrease in
affecting spatial learning and memory performance in Morris water muscarinic transmission have not been fully elucidated.
maze test; (2) long-term physical exercise had a protective effect against BDNF acts through the tyrosine kinase receptor (TrkB) and the pan-
cognitive and emotional behaviors impaired by scopolamine adminis­ neurotrophin receptor (p75) in the central nervous system. The effect of
tration; (3) scopolamine decreased levels of p-GSK3ßSer389, BDNF in BDNF on cognitive function is usually mediated by the TrkB receptor,
hippocampus and prefrontal cortex. Whereas TrkB decreased in hippo­ while it binds with low affinity to the p75 receptor. It is generally
campus and increased in prefrontal cortex; (4) there was an increase in thought that it exerts effects such as apoptosis and inhibition of neuronal
p-GSK3ßSer389, BDNF, TrkB in the hippocampus, and p-GSK3ßSer389, growth through this receptor. It is also known that this receptor localises
BDNF in the prefrontal cortex in the exercise + scopolamine group; (5) together with TrkB receptors (Bibel et al., 1999). Our study also looked
immunohistochemical analysis showed that scopolamine administration at the levels of TrkB receptors, which mediate the effect of BDNF. It was
increased APP and Aß-40 in hippocampus and prefrontal cortex in found that as the level of BDNF in the hippocampus decreased, the level
neuronal and perineuronal areas whereas Aß-40 and APP were reduced of TrkB also decreased. TrkB levels have been reported to decrease in
in exercise + scopolamine group. patients with Alzheimer’s disease (Ginsberg et al., 2006). It has been
Many studies in the literature have shown that the use of scopol­ reported that scopolamine administration reduces hippocampal BDNF
amine has a negative effect on spatial learning and memory performance and TrkB levels in mice and rats (Kim et al., 2021, Saikia et al., 2018). In
(Jafarian et al., 2019). The cholinergic system, which is highly the AD model induced by Aß25-35 in rats, it has been shown that there is a
concentrated in the thalamus, striatum, limbic system, and cortex, is decrease in BDNF and TrkB levels throughout the brain (Du et al., 2020).
involved in learning and memory, attention, and other higher brain TrkB triggers signalling pathways associated with functions such as
functions. Scopolamine, a non-selective blocker of muscarinic acetyl­ intracellular growth, differentiation, and plasticity through three
choline receptors, can cause atrophy and neurodegeneration in the rat distinct signalling pathways. These signaling pathways are Ras-mitogen-
brain. Scopolamine is known to cause deterioration in learning-memory, activated protein kinase (MAPK, MEK), phosphatidylinositol 3 kinase
and motor activity, mainly by blocking muscarinic receptors in the (PI3K), and phospholipase C-gamma (PLC-) (Minichiello, 2009). Akt,
hippocampus and cerebral cortex and increasing acetylcholinesterase also known as protein kinase B, plays an important role in cellular
activity (Klinkenberg and Blokland, 2010). In particular, the degenera­ functions such as cell proliferation, apoptosis, and glucose metabolism
tion of neurons from the Mynert nucleus in the basal forebrain to the (Song et al., 2005). While many of the studies have reported that Akt
hippocampus and cortex leads to a decline in cognitive function. Studies activation increases in AD, there are also studies that report that it de­
show that scopolamine administration has opposite effects on anxiety- creases (Griffin et al., 2005, Lee et al., 2009). Aß induces Akt by acti­
like behaviour. In addition to studies showing that it reduces anxiety- vating caspase-3, thereby activating GSK-3ß (Jo et al., 2011). Apoptotic
like behaviour, some studies suggest that it increases anxiety-like processes are triggered by the activation of GSK-3ß. In our study, we
behaviour which is in line with our findings (Furey and Drevets, 2006, found that TrkB expression increased despite a decrease in BDNF by a
Jafarian et al., 2019). mechanism we cannot explain in the prefrontal cortex. This may be
The administration of scopolamine to mice caused a decrease in related to the upregulation of TrkB due to the decrease in BDNF.
BDNF levels in the hippocampus and the prefrontal cortex, accompanied The Thr390 (human)/Ser389 (mouse) phosphorylation of GSK-3ß
by a deterioration in cognitive and emotional function. Many studies in occurs in a limited number of tissues compared to the phosphorylation
the literature show that scopolamine reduces BDNF levels (Safar et al., of Ser9 and Ser21. Ser9 and Ser21 phosphorylation are found in many
2016, Tang, 2019). BDNF mRNA levels have been shown to decrease in tissues, while Ser389 phosphorylation is found in the brain, thymus, and
Alzheimer’s patients in post-mortem examinations (Garzon et al., 2002). spleen. The target for Ser9 and Ser21 phosphorylation is cytoplasmic
A post-mortem study of Alzheimer’s patients with clinical mild cognitive GSK-3ß, whereas the primary target for Ser389 phosphorylation is nu­
impairment reported a correlation between BDNF levels in the parietal clear GSK-3ß. In addition, Ser9 phosphorylation of GSK-3ß is common to
cortex and decline in cognitive function (Peng et al., 2005). BDNF plays all brain regions, whereas Ser389 phosphorylation has been shown to be
an important role in neuronal development, growth, differentiation, and particularly prevalent in the hippocampus and cortex (Thornton et al.,
synaptic function in areas such as the cortex, hippocampus, and 2018). Thr390 (human)/Ser389 (mouse) phosphorylation of GSK-3ß by
cholinergic basal forebrain neurons during early development and in p38 MAPK has also been reported as an alternative pathway for GSK-3ß
adulthood (Marini et al., 2004). All of these regions are severely inactivation. It has been reported that p-GSK3ßSer389 is excessive in the
damaged in Alzheimer’s patients (Giannakopoulos et al., 1998). Studies brain and thymocytes, where p38 MAPK activity is high (Thornton et al.,
have reported that BDNF levels decrease when Aß proteins are applied to 2008). GSK-3ß has been reported to cause the excessive phosphorylation
normal nerve tissue (Rosa et al., 2021). Activity-dependent synaptic of the tau protein observed in AD and the accumulation of Aß plaques
plasticity is important in the cellular mechanisms of learning and (Wang et al., 2008). While GSK-3ß Ser9 phosphorylation hasbeen re­
memory. BDNF is known to be important for learning and memory, ported to decrease in AD, Ser389 phosphorylation has not been studied

11
S. Kose et al. Brain Research 1814 (2023) 148429

in the literature. In our study, we found that GSK-3ßSer389 phosphory­ enzyme adenyl cyclase. In some cells, M2 and M4 receptors trigger
lation was decreased in the hippocampus and prefrontal cortex in the intracellular signalling pathways involving phospholipase C. It has been
scopolamine-induced AD model. Thornton et al. reported in their study shown that activation of M1 and M3 receptors by acetylcholine increases
that learning-memory, locomotor activity, and anxiety-fear behaviours sAPPα secretion through the PKC pathway and also reduces Aß plaques
were not affected in Ser389 knockout mice, but unexpected fear be­ (Welt et al., 2015).
haviours were observed (Thornton et al., 2018). In our study, a decrease In AD, the degeneration of cholinergic neurons in the brain and a
in p-GSK3ßSer389 levels in the hippocampus and prefrontal cortex was decrease in the synthesis and release of ACh lead to a disruption of
observed, along with a decrease in spatial learning memory performance cholinergic transmission. ACh is responsible for regulating learning and
with scopolamine administration. cognitive functions that depend on the hippocampus. It acts on the
The p38 MAPK signalling pathway plays an important role in phys­ hippocampus and cerebral cortex in the brain, particularly through M1
iological processes such as neuronal plasticity and neurodegenerative receptors (Klinkenberg and Blokland, 2010). Studies in transgenic ani­
diseases. p38 MAPK is activated when it is phosphorylated by MKK6 and mals suggest that a cholinergic deficit plays a role in the accumulation of
MAPK3(MKK3) in the presence of cytokines, growth factors, and the Aß and the formation of phosphorylated tau (Ramos-Rodriguez et al.,
effects of cellular stress. Highly homologous isoforms of p38 MAPK 2013). It was first suggested by Nitsch et al. that stimulating M1 and M3
encoded by four different genes were expressed (p38a, p38ß, p38y, and receptors can increase sAPPα and thus prevent the formation of Aß
p38δ). The fact that they have different isoforms in different tissues al­ (Nitsch et al., 1992). M1 receptor activation increases sAPP via ADAM17
lows the p38 MAPK signalling pathway to have important functions in a (tumor necrosis factor-converting enzyme) and decreases Aß via PKC
wide range of physiological processes. These functions include apoptotic inhibition of BACE-1. It also reduces Aß-induced apoptosis and p-tau by
processes, inhibition of proliferation, and control of cell differentiation phosphorylating GSK-3ß (Fisher, 2012). Administration of a mAChR
(Zarubin and Han, 2005). The MAPK pathway has been shown to be antagonist such as scopolamine leads to an increase in Aß and hyper­
altered in AD. Phosphorylation of the ERK, JNK, and p38 forms of the phosphorylation of tau caused by GSK-3ß. It has been reported that M2
MAPK pathway is reported to be associated with cognitive decline in AD and M4 are ineffective in activating α-secretase and may even have an
(Ghasemi et al., 2014). Studies have shown that scopolamine adminis­ inhibitory effect on the release of these receptors sAPPα (Nitsch et al.,
tration increases the phosphorylation of p38, JNK, and ERK in the MAPK 1992).
signalling pathway (Moosavi et al., 2018, Pak et al., 2021). Our current In studies, increasing the inactive (phosphorylated) form of GSK-3ß
study did not assess p-p38 MAPK protein levels, but previous studies in BDNF-TrkB mediated signalling keeps the conversion of APP to Aß
have reported different results, such as that p-p38 MAPK activity in­ plaques under control. It has been reported that Aß plaques increase
creases, decreases, or does not change with scopolamine administration with an excessive increase in the active form of GSK-3ß due to a decrease
(Ban et al., 2020, Jahanmahin et al., 2019, Moosavi et al., 2018). It has in BDNF transmission. The active form of GSK-3ß has also been reported
been suggested that these differences may depend on the duration of to increase tau phosphorylation by activating tau kinases (Llorens-
scopolamine administration and the strain of animal used. In our study, Martin et al., 2014, Safar et al., 2016). In our study, increased Aß plaques
the decrease in p-GSK3ßSer389 levels may be due to decreased activation could also be explained by a decrease in phosphorylated GSK-3ß.
of the p38 MAPK pathway. Wang et al. showed in their study that BDNF In accordance with the literature, scopolamine did not affect body
increases the levels of p-p38 and phosphorylated AKT (Wang et al., weight. In mice that exercised, there was a significant decrease in body
2019). They proposed that BDNF phosphorylates p38 via the PI3K weight in the third and fourth weeks compared to sedentary subjects,
pathway using TrkB receptors. In our study, we found that BDNF and while a relative decrease was seen in other weeks (Baek et al., 2020,
TrkB levels decreased. This may have resulted in a decrease in phos­ Zhao et al., 2020).
phorylation of the p38 MAPK pathway and thus a decrease in p- In Balb/c mice that were exercised for 12 weeks, it was found that
GSK3ßSer389 levels. their anxiety-like behaviour decreased with an increase in center la­
In our study, it cannot be excluded that scopolamine administration tency, time spent in the center, frequency of entry to the center, and the
has an effect on changes in cognitive and emotional functions, a number of rearing in the open field test. There are many studies in the
decrease in BDNF-TrkB and p-GSK3ßSer389 levels, and various signalling literature that show the positive effects of physical exercise on emotional
pathways due to muscarinic receptor blockade. As expressed by and cognitive function, which is consistent with our findings (Pietrelli
Thornton and colleagues in their study, GSK-3ßSer389 phosphorylation et al., 2012). Physical exercise is known to have a positive effect on
may mediate the effects of scopolamine on anxiety and fear behaviour, learning and memory performance through cellular events such as
while other scopolamine-activated signalling pathways may also neurogenesis, increased LTP, and increased dendritic and synaptic
mediate its effects on cognitive function (Thornton et al., 2018). branching (van Praag et al., 1999). Studies report that the beneficial
In our study, we observed that scopolamine administration increased effects of exercise on the central nervous system are related to the
APP and Aß-40 expression in the hippocampus and prefrontal cortex in intracellular effects of BDNF (Russo-Neustadt et al., 1999). In our study,
neuronal and perineuronal areas. It has been reported that, physiologi­ we also found that treadmill exercise increased BDNF levels in the
cally, APP has a trophic effect on the central nervous system (Dawkins hippocampus and prefrontal cortex. It has been reported that BDNF
and Small, 2014). It has been reported that APP knockout mice have levels, which are increased by physical activity, cause neuronal plas­
smaller brains and that this affects neurogenesis (Wang et al., 2016). ticity by activating intracellular MAPK/ERK and phospholipase C sig­
APP has also been reported to play a role in LTP by regulating calcium nalling pathways through TrkB receptors (Minichiello, 2009, Ying et al.,
release (Kim et al., 2000). At physiological levels (picomolar), Aß has 2002). The reasons why BDNF levels increase in the brain during exer­
been reported to reduce potassium channel activity and apoptosis (Yu cise are not fully explained in the literature. The fact that BDNF crosses
et al., 2006). It has also been reported to increase acetylcholine release, the blood-brain barrier and is synthesised outside the brain, for example
protect neurons during aging, and be important for learning memory by by T and B lymphocytes, monocytes, and skeletal muscles, makes it
increasing synaptic plasticity (Morley et al., 2010). It is well known that difficult to identify the actual source of increased BDNF levels. The re­
in Alzheimer’s disease, there is an uncontrolled and excessive produc­ searchers suggest that in addition to BDNF released from peripheral
tion and accumulation of Aß. In rats, scopolamine (2 mg/kg) once a day tissues, several myokines (iris, cathepsin, etc.), glycolysis products
for 6 weeks has been shown to increase Aß plaques and decrease BDNF (lactate), ketone bodies (β-hydroxybutyrate) and cytokines (TNFα, IL-
levels in the hippocampus (Safar et al., 2016). The same study reported 1β, IL-18) cross the blood-brain barrier and regulate BDNF release in the
that scopolamine increased GSK-3ß mRNA and protein expression in the central nervous system (Delezie and Handschin, 2018, Tong et al.,
hippocampus. The M1, M3, and M5 receptors trigger intracellular sig­ 2012). Physical exercise has also been shown to increase choline uptake,
nalling pathways by activating PKC, while M1 and M4 activate the muscarinic receptor density, and the number of neurons expressing

12
S. Kose et al. Brain Research 1814 (2023) 148429

acetylcholine transferase in the hippocampus (Fordyce and Farrar, investigating the molecular and behavioural effects of long-term exer­
1991). cise, particularly in AD with cholinergic impairment. In addition, in our
In our study, it was found that p-GSKßSer389 levels did not change in study, not only the benefits of long-term exercise alone, but also the
the exercise group. Bayod et al. showed an increase in the level of p- benefits of this exercise in the adult period as a result of the changes in
GSK3ßSer9 in the hippocampus of rats that were exercised on a treadmill the developing brain were revealed. In short-term exercise models
5 times a week (for 30 min at a speed of 12 m/min) for 36 weeks (Bayod applied in the literature, mostly adult or old mice are used. Our study, in
et al., 2011). In a study investigating the effects of an enriched envi­ which mice that voluntarily received treadmill exercise as soon as they
ronment, such as physical exercise, it is reported that an increase in were separated from their mothers on day 21 were evaluated at 15
BDNF in the hippocampus causes an increase in the levels of p-GSK3ßSer9 weeks of age, differs in this respect. However, although short-term ex­
(Hu et al., 2013). ercise has been reported to increase BDNF and TrkB levels, the mecha­
The fact that the p-GSK3ßSer389 levels did not change with long-term nisms related to this have not been elucidated. In addition to BDNF and
physical exercise in our present study may be interpreted as an adap­ TrkB, we also analysed pGSK3ßSer389 levels, which have not been pre­
tation to long-term exercise. As mentioned above, although there are viously studied in the literature in mice subjected to long-term exercise,
studies on p-GSKßSer9 in the literature, p-GSKßSer389 has not been studied and tried to elucidate the mechanisms by which the positive effects of
in physical exercise. exercise occur. Although short- and long-term exercise has been shown
In our study, we found that long term physical exercise may protect to increase BDNF expression in the hippocampus in various animal
against the decrease in memory performance and anxiety-like behaviour models, it is emphasised that this effect of exercise is less in old animals
observed with scopolamine administration in Balb/c mice. Heo Y-M than in young animals (Adlard et al., 2005, Müller et al., 2020). Despite
et al. report that exerciseprotects against cognitive decline and induces the unclear mechanisms of how exercise modifies the processes of AD,
neurogenesis by increasing hippocampal BDNF levels in mice adminis­ the long preclinical period of the disease suggests that symptoms may be
tered 1 mg/kg scopolamine before treadmill exercise for 14 days (Heo significantly delayed by interventions such as exercise that are effective,
et al., 2014). Rosa et al. reported that the anxiety-like behaviour sustainable, patient-adaptable and have few side effects (Ribarič, 2022).
observed with cerebroventricular Aß-40 administration in mice was Studies with rodent models of AD have shown that exercise can alleviate
reduced by short-term treadmill exercise (Rosa et al., 2021). Although the symptoms of neurodegeneration (Belarbi et al., 2011, Nay et al.,
the beneficial effects of short-term exercise in AD have been shown in 2021). There are studies suggesting that exercise performed before the
the literature, there are less studies on the effects of long-term exercise. onset of AD-like neuropathology may slow the progression of the dis­
It has also been reported to increase cortical BDNF levels in trans­ ease. Results showing that exercise interventions after the development
genic mice that were given treadmill exercise for 12 weeks (5 days a of Aß pathology do not have a positive effect on cognitive and neuro­
week, 12 m/min) (Koo et al., 2013). In our study, we found that BNDF, pathological parameters are also reported (Richter et al., 2008, Zhao
TrkB, and p-GSK3ßSer389 levels decreased in the scopolamine treated et al., 2015). Ultimately, it was found that while the administration of
groups and increased in the exercise + scopolamine groups. Immuno­ scopolamine caused a deterioration in cognitive and emotional behav­
histochemical examination also showed that the increased expression of iour, prolonged physical exercise had a protective effect against these
Aß and APP in the prefrontal cortex and hippocampus decreased with negative scopolamine-induced behaviours. Scopolamine may have
scopolamine administration in the exercise groups. Physical activity has shown its effects on cognitive and emotional function by reducing the
been shown to have a beneficial effect on AD in studies for many years. levels of BDNF, TrkB, and p-GSK3ßSer389 in the hippocampus, BDNF and
Exercise has been reported to increase brain blood flow and hippo­ p-GSK3ßSer389 in the prefrontal cortex and increasing the levels of Aß-40
campal volume, reduce inflammation, oxidative stress, Aß plaques, and and APP in both areas. Long-term physical exercise may have a pro­
reduce neuroinflammation by preventing the occurrence of obesity and tective effect by increasing levels of BDNF, TrkB, and p-GSK3ßSer389 in
diabetes (Ribarič, 2022). As a result of these, it is suggested that exercise the prefrontal cortex and hippocampus and reducing levels of Aß-40 and
improves neurogenesis, synaptic plasticity, angiogenesis, and cognitive APP.
performance. Unfortunately, the cellular mechanisms and molecular
pathways underlying all these events are not fully understood. 4. Experimental procedure
Throughout the studies, it has been reported that conditions such as
decreased BDNF, oxidative stress, and inflammation cause the accu­ 4.1. Ethic
mulation of Aß plaques, while physical exercise reverses these negative
effects by increasing BDNF. In our study, physical exercise may have The experimental protocols were approved by the Ethics Committee
increased p-GSK3ßSer389 through the p38 MAPK pathway by increasing of Cukurova University Medical Sciences Experimental Application and
scopolamine-reduced BDNF levels, thereby reducing Aß plaque accu­ Research Center. Procedures in the study were performed in accordance
mulation. In addition, as mentioned in the literature, the accumulation with the NIH Guide for the Care and Use of Laboratory Animals (date:
of Aß plaques may be prevented by the effect of exercise on reducing August 12, 2020, and decision number: 2/5).
neuroinflammation. Moreover, since Ser389 phosphorylation uses nu­
clear GSK3ß as a substrate compared to Ser9 phosphorylation, is 4.2. Animals
expressed only in limited tissues, and the activation stimuli are different
compared to Ser9 phosphorylation, elucidation of this signaling In our study, male Balb/c mice were used at the age of 3 weeks (15
pathway will contribute to the understanding of the protective role of ±2 g). The experimental animals were provided by Cukurova University
physical exercise against the effects of scopolamine. Medical Sciences Experimental Application and Research Center, and
While p-GSK3ßSer389 was evaluated in our study, total GSK3ß could were housed in the animal care room in our department during the
not be evaluated. In addition, the p-p38 MAPK signalling pathway could experiment. The room temperature (22±1 ◦ C) and humidity (40–60%)
not be evaluated. These are accepted as the limitations of our study. The were kept under constant control during the 12-hour light/dark cycle of
small sample size of mice used in molecular studies is another limitation. the animal care room. Water and food were provided ad libitum to the
In the literature, the effects of long-term exercise on AD have mice.
generally been investigated in transgenic animal models and non-
transgenic models (Aβ or tau directly into the brain via intra­ 4.3. Exercise protocol
cerebroventricular (i.c.v.) or intrahippocampal injections) (García-Mesa
et al., 2011, Souza et al., 2013, Yang et al., 2022). The results of our Voluntary treadmill exercise was performed in mice in accordance
study are important because it is one of the limited number of studies with the exercise protocol previously described by Zhao et. al. Mice in

13
S. Kose et al. Brain Research 1814 (2023) 148429

the exercise group underwent a 6-day acclimatisation period before 4.5. Behavioural experiments
starting the 12-week exercise programme to get them used to the
treadmill. The treadmill was set at a walking speed of 5 m/min for the Mice were taken to the experiments on the last 8 days of the 28-day
first 2 days (days 1–2), then increased to 8 m/min on days 3–4 and 12 m/ injection period, 30 min after the injection. Before starting the behav­
min on days 5–6. The mice were then subjected to a 12-week exercise ioural tests, the mice underwent hand adaptation for 3 days. The mice
protocol that included 45 min of exercise per day, 5 days per week, from were first subjected to the open field test and, after a one-day rest, to the
6 to 8p.m. Each day, the mice were exercised at a series of speeds in the Morris water maze test.
following order: 5 m/min for 5 min, 8 m/min for 5 min, 12 m/min for
30 min, and 5 m/min for 5 min. Mice in sedentary control groups were 4.5.1. Open field test
placed on the static treadmill for the same amount of time (Zhao et al., The open field setup used in the test was made of black plexiglas,
2020) Fig. 11. 60x60x24, open at the top and surrounded by a 1 cm thick wall at the
sides. The area of the assembly adjacent to the wall is called the pe­
4.4. Drug riphery, and the remaining area is called the central area. The mice were
brought into the experimental room 3 min before the experiment to
In our study, the muscarinic acetylcholine receptor antagonist acclimatise to the environment in which the experiment would be
scopolamine (scopolamine hydrobromide trihydrate, 99%) was admin­ conducted, and then brought into the experiment. The room tempera­
istered during the last 4 weeks of a 12-week exercise programme to ture was set at 22±1 ◦ C and the illuminance at 100 lx. Mice were left in
induce an AD model. The administration dose of scopolamine was pre­ the open field from any corner, and their behaviour was recorded by a
pared by dilution with saline solution to 2 mg/kg. The prepared stock video-camera for 5 min. In the open field test, after the mice were left in
solution was kept at − 20 ◦ C in 1 ml Eppendorf tubes. The stock solution the open field, entry time to the center (sec), time spent in the center
was diluted with saline before injection. On the days of injection, it was (sec), frequency of entry to the center, distance travelled (cm), and the
kept at +4 ◦ C. number of rearing were recorded. In the open field test, the locomotor
In the scopolamine groups, 2 mg/kg (0.1 ml/10 g body weight vol­ activities of the mice were evaluated with the distance traveled variable,
ume) of scopolamine was administered intraperitoneally once a day and the anxiety-like behavior and exploratory behaviors of the mice
after exercise for 28 days (Hou et al., 2014, Safar et al., 2016, Satou were evaluated with the frequency of rearing, time spent in the center,
et al., 2018, Sharma and Gill, 2022). The control groups received the frequency of entry to the center, and entry time to the center variables.
same volume of saline intraperitoneally once a day. The body weights of
the animals were measured on the days of injection, and their weight 4.5.2. Morris water maze test
was also monitored weekly to monitor their somatic growth. The Morris water maze used in the test consists of a 95-cm-diameter,
Four different experimental groups (N = 69) were formed: sedentary- 45-cm-deep cylinder made of galvanised sheet metal with a black inte­
saline (S-Saline, n = 15), exercise-saline (E-Saline, n = 15), sedentary- rior. There is an escape platform (27 cm high and 10 cm in diameter)
scopolamine (S-SCOP, n = 11), and exercise-scopolamine (E-SCOP, n that can be moved inside the pool. During the experiments, the inside of
= 12). The mice were randomly divided into four groups. The the pool was filled with tap water to 1 cm above the platform level. The
randomization was performed according to a computer-generated water temperature was adjusted to 21±1 ◦ C. Black geometric shapes
randomization list. After exercise and scopolamine administration, the were hung on the walls of the laboratory, in contrast to the light-colored
mice were subjected to an open field test and Morris water maze test to wall, within the field of view of the mice, to provide a spatial cue to the
assess emotional and cognitive function, respectively. At the end of the mice swimming in the pool. The light in the pool is set at 50 lx.
injections, the prefrontal cortex and hippocampus of four mice from Mice were subjected to consecutive training trials 4 times a day for 5
each group that were not involved in the behavioural experiments were days in a Morris water maze. In each trial, mice left in the pool in one of
isolated. BDNF (Cat No.: 28205–1-AP), TrkB (Cat No.: STJ96109), p- the four imaginatively divided quadrants were given 60 s to find the
GSK3ßSer389 (Cat No.: 14850–1-AP), and beta actin (Cat No.: 20536–1- escape platform. Animals were removed from the pool after remaining
AP) levels were assessed by western blot analysis; APP and Aß-40 levels on the platform for 10 s. During the training trials, if the mice could not
were assessed by immunohistochemical analysis. find the platform for 60 s, they were guided by hand to spatially learn
the location of the platform and were removed from the pool by keeping
them on the platform for 10 s. The position of the platform was kept
fixed in the same quadrant for all training trials. Between training trials,
mice were kept in their cages at room temperature for 30 s.
On day 6, the test day of the experiment, the platform was removed
from the pool, and the experimental animals were left in the pool in the
quadrant opposite the quadrant where the platform had been before its
removal. For the test, mice were floated in the pool for 60 s, and the time
spent in the target quadrant was recorded by a camera. Spatial learning
and memory of mice were assessed using ETHOVISION XT (NOLDUS,
version 4.1) (Akıllıoğlu, 2010.). In the Morris water maze test, for the
first 5 days, the escape latency (sec) and the average swimming speed to
the platform (cm/sec) were recorded during the mice’s 60-second
training trials. On the 6th day of the experiment, the time spent in the
target quadrant for 60 s and the average swimming speed (cm/sec) were
recorded. On the first five days of the MWM test, mice were evaluated
for their spatial learning, and on the sixth day, for their memory.

4.6. Western blot analysis

4.6.1. Obtaining total protein samples


The prefrontal cortex and hippocampus of mice were isolated before
Fig. 11. Mice during treadmill exercise. western blotting to obtain proteins from the tissue samples to be used.

14
S. Kose et al. Brain Research 1814 (2023) 148429

The isolated tissues were kept at − 80 ◦ C. The prefrontal cortex and left at room temperature for 15 min. After 15 min, the sections were
hippocampus tissues were weighed on a sensitive scales. 3 ml of RIPA washed in distilled water and PBS, dried, and Avidin (ab93705, Abcam,
(for each 1 ml of RIPA, 10 μl PMSF, 10 μl sodium orthovanadate, and USA) was added and left for 20 min at room temperature. Amino­
10–20 μl protease inhibitor were added) was added per 1 g of tissue and ethylcarbozol (AEC, ab93705, Abcam, USA) solution was dripped onto
homogenised at 70–80 RPM for at least 5 min. The protein sample and the sections that had been passed through distilled water and PBS, left
the RIPA mixture were homogenised until creamy and transferred to 1 for 10 min, and then washed in tap water. Tissue sections were stained
ml Eppendorf tubes. Eppendorf tubes were centrifuged at 14,000 RPM with hematoxylin for 30 s before counterstaining. After staining, the
at +4 ◦ C for 10 min. The supernatant obtained by centrifugation was sections were washed in tap water and allowed to dry. Tissues were
stored at − 20 ◦ C. The total protein level of the samples was determined covered with a coverslip by dripping water-based sealant. Photographs
by the BCA (bicinchoninic acid) protein assay using albumin standards were taken using an Olympus BX53 light microscope (Japan).
of known protein levels. The immunohistochemical scoring method was used to investigate
3 μl of the gel electrophoresis standard was loaded into the first well the expression of APP and Aß-40. For immunohistochemical scoring, the
of the SDS-PAGE gel, and 30 μl of protein samples were loaded into each H-scoring system was used in 10 randomly selected areas of a 40-mm
of the other wells. The proteins were run at 80 V for the first 20 min and lens. The H-score was calculated by summing the products of the 4
then at 100 V for about 1–1.5 h until the proteins reached the end of the different staining intensities (3+++ (strong staining), 2++ (medium
membrane. At the end of the run, the gel was transferred to the PVDF staining), 1+ (weak staining), 0 (no staining)) together with the per­
membrane. After transfer to the membrane, the gel was gently shaken centage of positive cells. Differences in expression between groups were
for 1 h at +4 ◦ C in 20 ml of 5% blocking solution. After 1 h, the mem­ evaluated using the Mann-Whitney U test. p < 0.05 was considered
brane was washed 3 times for 5 min with a washing solution at +4 ◦ C in significant Fig. 12.
a shaker. Primary antibodies (ß-actin 1:1000, BDNF 1:500, TrkB 1:500,
and p-GSK3ßSer389 1:500) were applied immediately after washing. The 4.8. Statistical analysis
membrane was incubated with the primary antibody overnight at +4 ◦ C
on a shaker. After the primary antibody, the secondary antibody Data were expressed as mean ± standard error (SE). p < 0.05 was
(1:5000) was added to the membrane and incubated for 1 h at +4 ◦ C on a considered significant. Two-way ANOVA was used for normally
shaker. Enhanced Chemi Luminescence (ECL) solution was then used for distributed and parametric data, followed by Tukey’s test for pairwise
imaging. After exposure to the ECL solution, the radiation generated on group comparisons. Non-normally distributed and non-parametric data
the membrane was transferred to the 18x24 cm x-ray film, making the were analysed using the Kruskal-Wallis test, followed by the Mann-
protein bands visible. Quantitative analyses of band protein densities Whitney U test for pairwise group comparisons. As body weight and
were performed using the Image Lab program. The analysis was nor­ the Morris water maze test did not follow a normal distribution, Fried­
malised by dividing the specific protein density amounts by the band man’s ANOVA was used, followed by the Wilcoxon test for within-group
intensity amount of ß-actin protein. The amount of band intensity % comparisons and the Mann-Whitney U test for between-group compar­
change in comparison to the control group was expressed as: [(Prt C. isons. SPSS version 20.0 was used for statistical analysis.
sample/Prt C.ß-actin)/Prt C.control mean. X 100]. While the protein
density value of the control group was taken as 100 %, the protein fold 5. Conclusion
increase of the control group was taken as 1.
In our study, we found that physical exercise may protect against the
4.7. Immunohistochemical method memory impairment and anxiety-like behaviour observed with scopol­
amine induced Alzheimer’s Disease-like condition in Balb/c mice. There
In our study, 4 µm thick sections were taken from the hippocampus are many studies in the literature aimed at understanding the physio­
and prefrontal cortex tissues of 4 mice from each group for histological pathology of AD. In the scope of these studies, it is aimed to investigate
analyses. Histological sections of tissues were made on polylysine- the effects related to the decrease of BDNF and the accompanying
coated slides to determine APP and amyloid beta-40 (Aß-40) activity. disruption of intracellular signalling pathways. It is mainly reported that
The sections kept in the refrigerator were dried before the marking increased GSK-3ß mediate Aß plaque formation and excessive tau
process was started, then put back in the refrigerator and left for 15 min. phosphorylation, whereas Ser9 phosphorylation reduces these adverse
The sections, which were dried again after removal from the refriger­ effects. According to the results of our study, it can be suggested that the
ator, were deparaffinized in xylol in an oven at 60 ◦ C for 15 min. Tissue decrease in GSK3ßSer389 phosphorylation is important in the physiopa­
sections were then passed through the xylol series at room temperature. thology of AD, and the effects of physical exercise are mediated by
It was hydrated in a series of gradually decreasing degrees of alcohol and GSK3ßSer389 phosphorylation. In particular, the investigation of the p38
taken in distilled water. The sections were then placed in citrate solution MAPK/GSK3ßSer389 pathway, which is known to be active in specific
(pH: 6) and left in a water bath at 95 ◦ C for 45 min to reveal the antigens. tissues such as the brain and spleen, will be important in further studies
Sections cooled for 45 min at room temperature were washed in distilled to understand the role of this pathway in exercise and AD.
water and immersed in 3 % hydrogen peroxide (H2O2) for about 10 min
to prevent endogenous peroxides. Sections were washed in distilled 6. Limitations
water and phosphate buffer (PBS, pH: 7.2–7.4). After carefully wiping
and drying each section, the blocking solution (ab93705, Abcam, USA) Our study evaluated p-GSK3ßSer389, but due to budget limitations, we
was added to prevent non-specific binding. After 15 min at room tem­ were unable to evaluate total GSK3ß or the p-p38MAPK signalling
perature, the sections were passed through distilled water and phos­ pathway. Furthermore, the small sample size of mice used in molecular
phate buffer, and the surrounding tissue was dried. The primary studies was another limitation of our study. Despite these limitations, we
antibodies anti-APP (27320–1-AP, Proteintech, USA) at 1:200 dilution still believe that our research provides valuable insights into the area,
and anti-Aß-40 (NBP1 44047, Novus Biologicals, USA) at 1:200 dilution and that further research should be conducted to gain a better under­
were then applied. All primary antibody coated sections were stored standing of the mechanisms involved.
overnight at +4 ◦ C in a humid chamber. Instead of primary antibodies,
dilution solution was dripped onto sections taken for the negative con­ CRediT authorship contribution statement
trol. Sections treated with primary antibodies and kept at +4 ◦ C over­
night and then at room temperature for 1 h were then washed with Seda Kose: Validation, Investigation, Writing – original draft,
distilled water and PBS. Biotin (ab93705, Abcam, USA.) was applied and Writing – review & editing. Meltem Donmez Kutlu: Validation,

15
S. Kose et al. Brain Research 1814 (2023) 148429

Fig. 12. Timeline of the experiment.

Investigation, Writing – review & editing. Samet Kara: Validation, Akıllıoğlu K. Balb/c Farelerde Sinir Sisteminin Farklı Kritik “Gelişim Pencerelerinde”
NMDA Reseptör Blokajının Yetişkin Dönemde, Çevresel Faktörlere Bağlı Beynin
Investigation, Visualization, Resources. Sait Polat: Validation, Investi­
Bilişsel ve Duygusal İşlevleri Üzerine Etkisi. [Medical Speciality Thesis]. Adana,
gation, Visualization, Resources. Kubra Akillioglu: Conceptualization, Cukurova University. 2010.
Methodology, Validation, Formal analysis, Resources, Writing – original Akiyama, H., Barger, S., et al., 2000. Inflammation and Alzheimer’s disease. Neurobiol
draft, Writing – review & editing, Visualization, Project administration. Aging 21, 383–421. https://doi.org/10.1016/s0197-4580(00)00124-x.
Alzheimer’s A. 2019 Alzheimer’s disease facts and figures. Alzheimer’s & dementia
2019. 15. 321-387. https://doi.org/https://doi.org/10.1016/j.jalz.2019.01.010.
Declaration of Competing Interest Baek, S.Y., Li, F.Y., Kim, D.H., Kim, S.J., Kim, M.R., 2020. Enteromorpha prolifera
Extract Improves Memory in Scopolamine-Treated Mice via Downregulating
Amyloid-beta Expression and Upregulating BDNF/TrkB Pathway. Antioxidants
The authors declare that they have no known competing financial (Basel) 9. https://doi.org/10.3390/antiox9070620.
interests or personal relationships that could have appeared to influence Ban, J.Y., Park, H.K., Kim, S.K., 2020. Effect of Glycyrrhizic Acid on Scopolamine-
Induced Cognitive Impairment in Mice. Int Neurourol J 24, S48–S55. https://doi.
the work reported in this paper. org/10.5213/inj.2040154.077.
Bayod, S., del Valle, J., Canudas, A.M., Lalanza, J.F., Sanchez-Roige, S., Camins, A.,
Data availability Escorihuela, R.M., Pallàs, M., 2011. Long-term treadmill exercise induces
neuroprotective molecular changes in rat brain. Journal of Applied Physiology 111
(5), 1380–1390.
Data will be made available on request. Belarbi, K., Burnouf, S., Fernandez-Gomez, F.-J., Laurent, C., Lestavel, S., Figeac, M.,
Sultan, A., Troquier, L., Leboucher, A., Caillierez, R., Grosjean, M.-E., Demeyer, D.,
Obriot, H., Brion, I., Barbot, B., Galas, M.-C., Staels, B., Humez, S., Sergeant, N.,
Acknowledgments Schraen-Maschke, S., Muhr-Tailleux, A., Hamdane, M., Buée, L., Blum, D., 2011.
Beneficial effects of exercise in a transgenic mouse model of Alzheimer’s disease-like
Tau pathology. Neurobiology of disease 43 (2), 486–494.
This study is a part of the MSc thesis of Seda Köse. This study was
Bibel, M., Hoppe, E., Barde, Y.A., 1999. Biochemical and functional interactions between
supported by a research grant (TYL-2020-13222) from the Scientific the neurotrophin receptors trk and p75NTR. EMBO J 18, 616–622. https://doi.org/
Research Projects Unit of Cukurova University. 10.1093/emboj/18.3.616.
Chen, G.-F., Xu, T.-H., Yan, Y., Zhou, Y.-R., Jiang, Y.i., Melcher, K., Xu, H.E., 2017.
Amyloid beta: structure, biology and structure-based therapeutic development. Acta
Funding Pharmacol Sin 38 (9), 1205–1235.
Dawkins, E., Small, D.H., 2014. Insights into the physiological function of the beta-
amyloid precursor protein: beyond Alzheimer’s disease. J Neurochem 129, 756–769.
This work was supported by the Scientific Research Office of https://doi.org/10.1111/jnc.12675.
Cukurova University (I.U BAP, Project number: TYL-2020-13222). Delezie, J., Handschin, C., 2018. Endocrine Crosstalk Between Skeletal Muscle and the
Our study was presented partly at of the Turkish Society of Physio­ Brain. Frontiers in Neurology 9. https://doi.org/10.3389/fneur.2018.00698.
Du, Q., Zhu, X.Y., Si, J.R., 2020. Angelica polysaccharide ameliorates memory
logical Sciences, 46th National Physiology Congress, 8–10 October impairment in Alzheimer’s disease rat through activating BDNF/TrkB/CREB
2021, Virtual Meeting. The abstract of the presentation was printed in pathway. Experimental Biology and Medicine 245, 1–10. https://doi.org/10.1177/
“Acta Physiologica 2022; Volume 234, Supplement 724: PC05.” It also 1535370219894558.
Ferreira-Vieira, T.H., M. Guimaraes, I., R. Silva, F., M. Ribeiro, F., 2016. Alzheimer’s
presented partly at the Turkish Society of Physiological Sciences, 47th disease: targeting the cholinergic system. Current neuropharmacology 14 (1),
Turkish Physiology Congress, 1-4 November 2022, Antalya, Turkey. The 101–115.
abstract of the presentation was printed in “Acta Physiologica 2023; Fisher, A., 2012. Cholinergic modulation of amyloid precursor protein processing with
emphasis on M1 muscarinic receptor: perspectives and challenges in treatment of
Volume 237, Supplement 727: OC57.”
Alzheimer’s disease. Journal of neurochemistry 120, 22–33. https://doi.org/
This study was supported by a research grant (TYL-2020-13222) 10.1111/j.1471-4159.2011.07507.x.
from the Scientific Research Projects Unit of Cukurova University. Fordyce, D.E., Farrar, R.P., 1991. Effect of Physical-Activity on Hippocampal High-
Affinity Choline Uptake and Muscarinic Binding - a Comparison between Young and
Old F344 Rats. Brain Research 541, 57–62. https://doi.org/10.1016/0006-8993(91)
References 91073-A.
Furey, M.L., Drevets, W.C., 2006. Antidepressant efficacy of the antimuscarinic drug
Adlard, P.A., Perreau, V.M., Cotman, C.W., 2005. The exercise-induced expression of scopolamine - A randomized, placebo-controlled clinical trial. Archives of General
BDNF within the hippocampus varies across life-span. Neurobiol Aging 26, 511–520. Psychiatry 63, 1121–1129. https://doi.org/10.1001/archpsyc.63.10.1121.
https://doi.org/10.1016/j.neurobiolaging.2004.05.006.

16
S. Kose et al. Brain Research 1814 (2023) 148429

García-Mesa, Y., López-Ramos, J.C., Giménez-Llort, L., Revilla, S., Guerra, R., Gruart, A., Lee, H.K., Kumar, P., Fu, Q., Rosen, K.M., Querfurth, H.W., 2009. The Insulin/Akt
LaFerla, F.M., Cristòfol, R., Delgado-García, J.M., Sanfeliu, C., 2011. Physical Signaling Pathway Is Targeted by Intracellular beta-Amyloid. Molecular Biology of
exercise protects against Alzheimer’s disease in 3xTg-AD mice. Journal of the Cell 20, 1533–1544. https://doi.org/10.1091/mbc.E08-07-0777.
Alzheimer’s disease : JAD 24 (3), 421–454. Lim, Y.Y., Maruff, P., Schindler, R., Ott, B.R., Salloway, S., Yoo, D.C., Noto, R.B.,
Garzon, D., Yu, G., Fahnestock, M., 2002. A new brain-derived neurotrophic factor Santos, C.Y., Snyder, P.J., 2015. Disruption of cholinergic neurotransmission
transcript and decrease in brain-derived neurotrophic factor transcripts 1, 2 and 3 in exacerbates Aβ-related cognitive impairment in preclinical Alzheimer’s disease.
Alzheimer’s disease parietal cortex. J Neurochem 82, 1058–1064. https://doi.org/ Neurobiol Aging 36 (10), 2709–2715.
10.1046/j.1471-4159.2002.01030.x. Llorens-Martin, M., Jurado, J., Hernandez, F., Avila, J., 2014. GSK-3beta, a pivotal kinase
Ghasemi, R., Zarifkar, A., Rastegar, K., Maghsoudi, N., Moosavi, M., 2014. Repeated in Alzheimer disease. Front Mol Neurosci 7, 46. https://doi.org/10.3389/
intra-hippocampal injection of beta-amyloid 25–35 induces a reproducible fnmol.2014.00046.
impairment of learning and memory: Considering caspase-3 and MAPKs activity. Marini, A.M., Jiang, X., et al., 2004. Role of brain-derived neurotrophic factor and NF-
European Journal of Pharmacology 726, 33–40. https://doi.org/10.1016/j. kappaB in neuronal plasticity and survival: From genes to phenotype. Restor Neurol
ejphar.2013.11.034. Neurosci 22, 121–130.
Giannakopoulos, P., Gold, G., Michel, J.P., Bouras, C., 1998. Cellular vulnerability in Mehta, M., Schmauss, CJBn, 2011. Strain-specific cognitive deficits in adult mice exposed
brain aging and Alzheimer’s disease. Clinical correlates and molecular background. to early life stress. Behavioral Neuroscience 125, 29. https://doi.org/10.1037/
Ann Med Interne (Paris) 149, 187–191. a0021952.
Gil-Bea, F.J., Solas, M., Mateos, L., Winblad, B., Ramirez, M.J., Cedazo-Minguez, A., Miller-Thomas, M.M., Sipe, A.L., Benzinger, T.L., McConathy, J., Connolly, S.,
2011. Cholinergic Hypofunction Impairs Memory Acquisition Possibly Through Schwetye, K.E., 2016. Multimodality Review of Amyloid-related Diseases of the
Hippocampal Arc and BDNF Downregulation. Hippocampus 21, 999–1009. https:// Central Nervous System. Radiographics 36, 1147–1163. https://doi.org/10.1148/
doi.org/10.1002/hipo.20812. rg.2016150172.
Ginsberg, S.D., Che, S., Wuu, J., Counts, S.E., Mufson, E.J., 2006. Down regulation of trk Minichiello, L., 2009. TrkB signalling pathways in LTP and learning. Nat Rev Neurosci
but not p75NTR gene expression in single cholinergic basal forebrain neurons mark 10, 850–860. https://doi.org/10.1038/nrn2738.
the progression of Alzheimer’s disease. J Neurochem 97, 475–487. https://doi.org/ Moosavi, M., SoukhakLari, R., Moezi, L., Pirsalami, F., 2018. Scopolamine-induced
10.1111/j.1471-4159.2006.03764.x. passive avoidance memory retrieval deficit is accompanied with hippocampal
Griffin, R.J., Moloney, A., Kelliher, M., Johnston, J.A., Ravid, R., Dockery, P., MMP2, MMP-9 and MAPKs alteration. Eur J Pharmacol 819, 248–253. https://doi.
O’Connor, R., O’Neill, C., 2005. Activation of Akt/PKB, increased phosphorylation of org/10.1016/j.ejphar.2017.12.007.
Akt substrates and loss and altered distribution of Akt and PTEN are features of Morley, J.E., Farr, S.A., Banks, W.A., Johnson, S.N., Yamada, K.A., Xu, L., 2010.
Alzheimer’s disease pathology. Journal of Neurochemistry 93 (1), 105–117. A physiological role for amyloid-beta protein:enhancement of learning and memory.
Groves, N.J., Kesby, J.P., Eyles, D.W., McGrath, J.J., Mackay-Sim, A., Burne, T.H., 2013. Journal of Alzheimer’s disease : JAD 19, 441–449. https://doi.org/10.3233/JAD-
Adult vitamin D deficiency leads to behavioural and brain neurochemical alterations 2009-1230.
in C57BL/6J and BALB/c mice. Behavioural brain research 241, 120–131. https:// Müller, P., Duderstadt, Y., Lessmann, V., Müller, N.G., 2020. Lactate and BDNF: Key
doi.org/10.1016/j.bbr.2012.12.001. Mediators of Exercise Induced Neuroplasticity? J Clin Med 9. https://doi.org/
Hampel H, Mesulam MM, et al. The cholinergic system in the pathophysiology and 10.3390/jcm9041136.
treatment of Alzheimer’s disease. Brain 2018;141:1917-1933. https://doi.org/ Navakkode, S., Korte, M., 2012. Cooperation between cholinergic and glutamatergic
10.1093/brain/awy132. receptors are essential to induce BDNF-dependent long-lasting memory storage.
Heo, Y.M., Shin, M.S., Kim, S.H., Kim, T.W., Baek, S.B., Baek, S.S., 2014. Treadmill Hippocampus 22, 335–346. https://doi.org/10.1002/hipo.20902.
exercise ameliorates disturbance of spatial learning ability in scopolamine-induced Nay, K., Smiles, W.J., Kaiser, J., McAloon, L.M., Loh, K., Galic, S., Oakhill, J.S.,
amnesia rats. J Exerc Rehabil 10, 155–161. https://doi.org/10.12965/jer.140110. Gundlach, A.L., Scott, J.W., 2021. Molecular Mechanisms Underlying the Beneficial
Hou, XUE.-QIN., Wu, DIAN.-WEI., Zhang, CHUN.-XIA., Yan, RONG, Yang, CONG, Effects of Exercise on Brain Function and Neurological Disorders. Int J Mol Sci 22
Rong, CUI.-PING., Zhang, LEI, Chang, XIANG, Su, RU.-YU., Zhang, SHI.-JIE., (8), 4052.
He, WEN.-QING., Qu, ZHAO, Li, SHI, Su, ZI.-REN., Chen, YUN.-BO., Wang, Q.I., Nitsch, R.M., Slack, B.E., Wurtman, R.J., Growdon, J.H., 1992. Release of Alzheimer
Fang, SHU.-HUAN., 2014. Bushen-Yizhi formula ameliorates cognition deficits and amyloid precursor derivatives stimulated by activation of muscarinic acetylcholine
attenuates oxidative stress-related neuronal apoptosis in scopolamine-induced receptors. Science 258, 304–307. https://doi.org/10.1126/science.1411529.
senescence in mice. International Journal of Molecular Medicine 34 (2), 429–439. Nomura, Y., Nishiyama, N., Saito, H., Matsuki, N., 1994. Role of cholinergic
Hu YS, Long N, Pigino G, Brady ST, Lazarov O. Molecular Mechanisms of Environmental neurotransmission in the amygdala on performances of passive avoidance learning in
Enrichment: Impairments in Akt/GSK3 beta, Neurotrophin-3 and CREB Signaling. mice. Biol Pharm Bull 17, 490–494. https://doi.org/10.1248/bpb.17.490.
Plos One 2013;8. e64460. 10.1371/journal.pone.0064460. Özkay, Ü.D., Öztürk, Y., Can, Ö., 2011. Yaşlanan Dünyanın Hastalığı: Alzheimer
Hur, E.M., Zhou, F.Q., 2010. GSK3 signalling in neural development. Nature Reviews Hastalığı. SDÜ Tıp Fakültesi Dergisi 18, 35–42.
Neuroscience 11, 539–551. https://doi.org/10.1038/nrn2870. Pak, M.E., Oh, Y.-C., Park, Y.J., Kim, J.K., Shin, M.-G., Yoon, J.H., Go, Y., Morroni, F.,
Jafarian, S., Ling, K.H., Hassan, Z., Perimal-Lewis, L., Sulaiman, M.R., Perimal, E.K., 2021. Banhasasim-Tang Ameliorates Spatial Memory by Suppressing Oxidative
2019. Effect of zerumbone on scopolamine-induced memory impairment and Stress through Regulation of ERK/p38 Signaling in Hippocampus of Mice. Oxid Med
anxiety-like behaviours in rats. Alzheimer’s & Dementia: Translational Research & Cell Longev 2021, 1–12.
Clinical Interventions 5, 637–643. https://doi.org/10.1016/j.trci.2019.09.009. Peng, S., Wuu, J., Mufson, E.J., Fahnestock, M., 2005. Precursor form of brain-derived
Jahanmahin, A., Abbasnejad, Z., Haghparast, A., Ahmadiani, A., Ghasemi, R., 2019. The neurotrophic factor and mature brain-derived neurotrophic factor are decreased in
Effect of Intrahippocampal Insulin Injection on Scopolamine-induced Spatial the pre-clinical stages of Alzheimer’s disease. J Neurochem 93, 1412–1421. https://
Memory Impairment and Extracellular Signal-regulated Kinases Alteration. Basic doi.org/10.1111/j.1471-4159.2005.03135.x.
Clin Neurosci 10, 23–36. https://doi.org/10.32598/bcn.9.10.165. Pietrelli, A., Lopez-Costa, J., Goni, R., Brusco, A., Basso, N., 2012. Aerobic Exercise
Jo, J., Whitcomb, D.J., et al., 2011. A beta(1–42) inhibition of LTP is mediated by a Prevents Age-Dependent Cognitive Decline and Reduces Anxiety-Related Behaviors
signaling pathway involving caspase-3, Akt1 and GSK-3 beta. Nature Neuroscience in Middle-Aged and Old Rats. Neuroscience 202, 252–266. https://doi.org/10.1016/
14, 545–547. https://doi.org/10.1038/nn.2785. j.neuroscience.2011.11.054.
Kandimalla, R., Reddy, P.H., 2017. Therapeutics of Neurotransmitters in Alzheimer’s Ramos-Rodriguez, J.J., Pacheco-Herrero, M., Thyssen, D., Murillo-Carretero, M.I.,
Disease. Journal of Alzheimer’s disease: JAD 57, 1049–1069. https://doi.org/ Berrocoso, E., Spires-Jones, T.L., Bacskai, B.J., Garcia-Alloza, M., 2013. Rapid
10.3233/jad-161118. β-amyloid deposition and cognitive impairment after cholinergic denervation in
Kim, Y., Kim, J., He, M.T., Lee, A., Cho, E.J., 2021. Apigenin Ameliorates Scopolamine- APP/PS1 mice. Journal of Neuropathology & Experimental Neurology 72 (4),
Induced Cognitive Dysfunction and Neuronal Damage in Mice. Molecules 26. 272–285.
https://doi.org/10.3390/molecules26175192. Ribarič, S., 2022. Physical Exercise, a Potential Non-Pharmacological Intervention for
Kim, H.S., Park, C.H., et al., 2000. Carboxyl-terminal fragment of Alzheimer’s APP Attenuating Neuroinflammation and Cognitive Decline in Alzheimer’s Disease
destabilizes calcium homeostasis and renders neuronal cells vulnerable to Patients. Int J Mol Sci 23 (6), 3245.
excitotoxicity. FASEB J 14, 1508–1517. https://doi.org/10.1096/fj.14.11.1508. Richter, H., Ambrée, O., Lewejohann, L., Herring, A., Keyvani, K., Paulus, W., Palme, R.,
Klinkenberg, I., Blokland, A., 2010. The validity of scopolamine as a pharmacological Touma, C., Schäbitz, W.-R., Sachser, N., 2008. Wheel-running in a transgenic mouse
model for cognitive impairment: a review of animal behavioral studies. Neurosci model of Alzheimer’s disease: protection or symptom? Behavioural brain research
Biobehav Rev 34, 1307–1350. https://doi.org/10.1016/j.neubiorev.2010.04.001. 190 (1), 74–84.
Kocahan, S., Doğan, Z., 2017. Mechanisms of Alzheimer’s Disease Pathogenesis and Rosa, J.M., Camargo, A., Wolin, I.A.V., Kaster, M.P., Rodrigues, A.L.S., 2021. Physical
Prevention: The Brain, Neural Pathology, N-methyl-D-aspartate Receptors, Tau exercise prevents amyloid beta1-40-induced disturbances in NLRP3 inflammasome
Protein and Other Risk Factors. Clin Psychopharmacol Neurosci 15, 1–8. https://doi. pathway in the hippocampus of mice. Metab Brain Dis 36, 351–359. https://doi.org/
org/10.9758/cpn.2017.15.1.1. 10.1007/s11011-020-00646-8.
Koo, J.H., Kwon, I.S., Kang, E.B., Lee, C.K., Lee, N.H., Kwon, M.G., Cho, I.H., Cho, J.Y., Russo-Neustadt, A., Beard, R.C., Cotman, C.W., 1999. Exercise, antidepressant
2013. Neuroprotective effects of treadmill exercise on BDNF and PI3-K/Akt signaling medications, and enhanced brain derived neurotrophic factor expression.
pathway in the cortex of transgenic mice model of Alzheimer’s disease. Journal of Neuropsychopharmacology 21, 679–682. https://doi.org/10.1016/s0893-133x(99)
Exercise Nutrition & Biochemistry 17 (4), 151–160. 00059-7.
Krnjevic, K., Ropert, N., Casullo, J., 1988. Septohippocampal Disinhibition. Brain Safar, M.M., Arab, H.H., Rizk, S.M., El-Maraghy, S.A., 2016. Bone Marrow-Derived
Research 438, 182–192. https://doi.org/10.1016/0006-8993(88)91337-6. Endothelial Progenitor Cells Protect Against Scopolamine-Induced Alzheimer-Like
Lane, C.A., Hardy, J., Schott, J.M., 2018. Alzheimer’s disease. Eur J Neurol 25, 59–70. Pathological Aberrations. Mol Neurobiol 53, 1403–1418. https://doi.org/10.1007/
https://doi.org/10.1111/ene.13439. s12035-014-9051-8.
Saikia, B., Barua, ChandanaC, Sarma, J., Haloi, P., Tamuli, SarojiniM, Kalita, DhrubaJ,
Purkayastha, A., Barua, AchintaG, 2018. Zanthoxylum alatum ameliorates

17
S. Kose et al. Brain Research 1814 (2023) 148429

scopolamine-induced amnesia in rats: Behavioral, biochemical, and molecular Vaynman, S., Ying, Z., Gómez-Pinilla, FJJonr, 2004. Exercise induces BDNF and synapsin
evidence. Indian Journal of Pharmacology 50 (1), 30. I to specific hippocampal subfields. Journal of Neuroscience Research 76, 356–362.
Satou, T., Hanashima, Y., Mizutani, I., Koike, K.J.F., journal, f., 2018. The effect of https://doi.org/10.1002/jnr.20077.
inhalation of essential oil from Rosmarinus officinalis on scopolamine-induced Wang, S., Bolos, M., et al., 2016. Amyloid beta precursor protein regulates neuron
Alzheimer’s type dementia model mice. Flavour and fragrance journal 33, 230–234. survival and maturation in the adult mouse brain. Mol Cell Neurosci 77, 21–33.
https://doi.org/10.1002/ffj.3435. https://doi.org/10.1016/j.mcn.2016.09.002.
Scheltens, P., Blennow, K., Breteler, M.M.B., de Strooper, B., Frisoni, G.B., Salloway, S., Wang, H., Wei, Y., Pu, Y., Jiang, D., Jiang, X., Zhang, Y., Tao, J., 2019. Brain-derived
Van der Flier, W.M., 2016. Alzheimer’s disease. Lancet 388 (10043), 505–517. neurotrophic factor stimulation of T-type Ca(2+) channels in sensory neurons
Sharma, A., Gill, N., 2022. Therapeutic Potential of Dragea volubilis Leaf Extract against contributes to increased peripheral pain sensitivity. Sci Signal 12 (600). https://doi.
Scopolamine-induced Memory Impairment in Young and Aged Mice. International org/10.1126/scisignal.aaw2300.
Journal of Pharmaceutical Investigation 13 (1), 162–172. Wang, Y., Zhang, J.-X., Du, X.-X., Zhao, L.i., Tian, Q., Zhu, L.-Q., Wang, S.-H., Wang, J.-
Slotkin, T.A., Seidler, F.J., Crain, B.J., Bell, J.M., Bissette, G., Nemeroff, C.B., 1990. Z., 2008. Temporal correlation of the memory deficit with Alzheimer-like lesions
Regulatory changes in presynaptic cholinergic function assessed in rapid autopsy induced by activation of glycogen synthase kinase-3. Journal of Neurochemistry 106
material from patients with Alzheimer disease: implications for etiology and (6), 2364–2374.
therapy. Proc Natl Acad Sci U S A 87, 2452–2455. https://doi.org/10.1073/ Welt, T., Kulic, L., et al., 2015. Acute Effects of Muscarinic M1 Receptor Modulation on
pnas.87.7.2452. AbetaPP Metabolism and Amyloid-beta Levels in vivo: A Microdialysis Study.
Smythe, J.W., Murphy, D., Bhatnagar, S., Timothy, C., Costall, B., 1996. Muscarinic Journal of Alzheimer’s disease : JAD 46, 971–982. https://doi.org/10.3233/JAD-
antagonists are anxiogenic in rats tested in the black-white box. Pharmacology 150152.
Biochemistry and Behavior 54, 57–63. https://doi.org/10.1016/0091-3057(95) Xu, L., Zhu, L., Zhu, L., Chen, D., Cai, K., Liu, Z., Chen, A., 2021. Moderate Exercise
02130-2. Combined with Enriched Environment Enhances Learning and Memory through
Song, G., Ouyang, G.L., Bao, S.D., 2005. The activation of Akt/PKB signaling pathway BDNF/TrkB Signaling Pathway in Rats. Int J Environ Res Public Health 18 (16),
and cell survival. Journal of Cellular and Molecular Medicine 9, 59–71. https://doi. 8283.
org/10.1111/j.1582-4934.2005.tb00337.x. Yang, L., Wu, C., Li, Y., Dong, Y., Wu, C.-C., Lee, R.-C., Brann, D.W., Lin, H.W., Zhang, Q.,
Souder, D.C., Anderson, R.M., 2019. An expanding GSK3 network: implications for aging 2022. Long-term exercise pre-training attenuates Alzheimer’s disease–related
research. Geroscience 41, 369–382. https://doi.org/10.1007/s11357-019-00085-z. pathology in a transgenic rat model of Alzheimer’s disease. GeroScience 44 (3),
Souza, L.C., Filho, C.B., Goes, A.T.R., Fabbro, L.D., de Gomes, M.G., Savegnago, L., 1457–1477.
Oliveira, M.S., Jesse, C.R., 2013. Neuroprotective Effect of Physical Exercise in a Ying, S.-W., Futter, M., Rosenblum, K., Webber, M.J., Hunt, S.P., Bliss, T.V.P.,
Mouse Model of Alzheimer’s Disease Induced by β-Amyloid1–40 Peptide. Bramham, C.R., 2002. Brain-derived neurotrophic factor induces long-term
Neurotoxicity Research 24 (2), 148–163. potentiation in intact adult hippocampus: Requirement for ERK activation coupled to
Tang, K.S., 2019. The cellular and molecular processes associated with scopolamine- CREB and upregulation of Arc synthesis. Journal of Neuroscience 22 (5), 1532–1540.
induced memory deficit: A model of Alzheimer’s biomarkers. Life Sci 233, 116695. Yu, H.B., Li, Z.B., Zhang, H.X., Wang, X.L., 2006. Role of potassium channels in Abeta
https://doi.org/10.1016/j.lfs.2019.116695. (1–40)-activated apoptotic pathway in cultured cortical neurons. J Neurosci Res 84,
Thornton, T.M., Hare, B., et al., 2018. Failure to Inactivate Nuclear GSK3beta by Ser 1475–1484. https://doi.org/10.1002/jnr.21054.
(389)-Phosphorylation Leads to Focal Neuronal Death and Prolonged Fear Response. Zarubin, T., Han, J., 2005. Activation and signaling of the p38 MAP kinase pathway. Cell
Neuropsychopharmacology 43, 393–405. https://doi.org/10.1038/npp.2017.187. Res 15, 11–18. https://doi.org/10.1038/sj.cr.7290257.
Thornton, T.M., Pedraza-Alva, G., et al., 2008. Phosphorylation by p38 MAPK as an Zhao, G., Liu, H.L., Zhang, H., Tong, X.J., 2015. Treadmill exercise enhances synaptic
alternative pathway for GSK3 beta inactivation. Science 320, 667–670. https://doi. plasticity, but does not alter β-amyloid deposition in hippocampi of aged APP/PS1
org/10.1126/science.1156037. transgenic mice. Neuroscience 298, 357–366. https://doi.org/10.1016/j.
Tong, L.Q., Prieto, G.A., et al., 2012. Brain-Derived Neurotrophic Factor-Dependent neuroscience.2015.04.038.
Synaptic Plasticity Is Suppressed by Interleukin-1 beta via p38 Mitogen-Activated Zhao, N.a., Yan, Q.-W., Xia, J., Zhang, X.-L., Li, B.-X., Yin, L.-Y., Xu, B.o., 2020. Treadmill
Protein Kinase. Journal of Neuroscience 32, 17714–17724. https://doi.org/ Exercise Attenuates Aβ-Induced Mitochondrial Dysfunction and Enhances Mitophagy
10.1523/Jneurosci.1253-12.2012. Activity in APP/PS1 Transgenic Mice. Neurochem Res 45 (5), 1202–1214.
van Praag, H., Christie, B.R., Sejnowski, T.J., Gage, F.H., 1999. Running enhances Zucchella, C., Sinforiani, E., Tamburin, S., Federico, A., Mantovani, E., Bernini, S.,
neurogenesis, learning, and long-term potentiation in mice. Proc Natl Acad Sci U S A Casale, R., Bartolo, M., 2018. The Multidisciplinary Approach to Alzheimer’s Disease
96, 13427–13431. https://doi.org/10.1073/pnas.96.23.13427. and Dementia. A Narrative Review of Non-Pharmacological Treatment. Front Neurol
9, 1058. https://doi.org/10.3389/fneur.2018.01058.

18

You might also like