You are on page 1of 10

Downloaded from https://iranpaper.

ir

Brain Research Bulletin 170 (2021) 264–273

Contents lists available at ScienceDirect

Brain Research Bulletin


journal homepage: www.elsevier.com/locate/brainresbull

Effects of vanillic acid on Aβ1-40-induced oxidative stress and learning and


memory deficit in male rats
Nesa Ahmadi a, b, Samaneh Safari a, b, Naser Mirazi b, Seyed Asaad Karimi a, c,
Alireza Komaki a, c, *, 1
a
Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
b
Department of Biology, Faculty of Basic Sciences, Bu-Ali Sina University, Hamadan, Iran
c
Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran

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

Keywords: Alzheimer’s disease (AD) is a neurodegenerative disease, in which the accumulation of β-amyloid (Aβ) peptide in
β-Amyloid the extracellular space causes a progressive reduction in cognitive performance. Aβ stimulates active oxygen
Vanillic acid species generation leading to oxidative stress and neural cell death. Vanillic Acid (VA) is the oxidant form of
Oxidative stress
vanillin widely found in vanilla beans. VA has many properties, such as suppressing apoptosis and eliminating
Hippocampus
the harmful effects of oxidative stress in animal models. The VA effects on impaired learning and memory in Aβ
Learning and memory
Alzheimer’s disease rats were assessed. Forty adults male Wistar rats were assigned to the following five groups in random: the
control, sham (received saline (vehicle) via intracerebroventricular (ICV) injection), Aβ (received Aβ1–40 via ICV
injection), VA (50 mg/kg by oral gavage once a day through four weeks), and Aβ + VA (50 mg/kg) groups. Open
field test, novel object recognition (NOR) test, Morris water maze (MWM) test, and passive avoidance learning
(PAL) task were performed, and finally, we determined the malondialdehyde (MDA), total antioxidant capacity
(TAC) and total oxidant status (TOS) levels. Aβ decreased the cognitive memory in NOR, spatial memory in
MWM, and passive avoidance memory in PAL tests. In contrast, VA improved learning and memory in the treated
group. Aβ significantly increased MDA and TOS and decreased TAC levels, whereas VA treatment significantly
reversed TAC, TOS and MDA levels. In conclusion, VA decreased the Aβ effects on learning and memory by
suppressing oxidative stress and can be regarded as a neuroprotective substance in AD.

1. Introduction neurofibrillary tangles within select regions including hippocampus and


cortex (Braak and Braak, 1990). The disease is divided into familial cases
Alzheimer’s disease (AD) is an age-dependent and progressive and sporadic cases (Piaceri et al., 2013). In both cases, Aβ peptides,
neurodegenerative disease, affecting the central nervous system (CNS), produced from amyloid precursor protein (APP) by β and γ-secretases,
and eventually leading to cognitive and memory impairment (Burns and are major contributors to the pathology of the disease (Palop and Mucke,
Iliffe, 2009; Mendiola-Precoma et al., 2016). Considering the increasing 2010). Severe atrophy of the hippocampus due to neuronal cell death, is
rate of older people worldwide, the number of AD cases it is estimated at one of the major hallmarks of AD progression (Wirths and Bayer, 2010).
114 million by 2050 (Querfurth and LaFerla, 2010). Increased in the In addition to neurofibrillary tangles and Aβ plaques, deficits in
prevalence of AD can lead to medical costs and death in cases over 65 cholinergic function (Giacobini, 2003), neuroinflammation (Heneka
years (Alzheimer’s; Prince et al., 2015). AD is mainly characterized by et al., 2015), sex steroid hormones deficiencies (Vest and Pike, 2013),
the presence of neurofibrillary tangles and β-amyloid (Aβ) plaques in the excitotoxicity (Hynd et al., 2004), altered synaptic function (Bell and
cortex and hippocampus (Samandouras et al., 2006; Tanzi and Bertram, Claudio Cuello, 2006), abnormal mitochondrial dynamics and synaptic
2005). Abnormal phosphorylation and hyperphosphorylation of the degeneration (Reddy et al., 2012), and oxidative stress (Markesbery,
cytoskeletal protein tau within neurons leading to the formation of 1997) are considerate to contribute to the pathology of AD.

* Corresponding author at: Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/
518, Iran.
E-mail address: Komaki@umsha.ac.ir (A. Komaki).
1
URL:umsha.ac.ir

https://doi.org/10.1016/j.brainresbull.2021.02.024
Received 7 October 2020; Received in revised form 14 February 2021; Accepted 24 February 2021
Available online 27 February 2021
0361-9230/© 2021 Elsevier Inc. All rights reserved.
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

Clinically AD is characterized by progressive cognitive decline and antioxidant effects of VA and the association between AD and
abnormalities in behavior (Larson et al., 1992; Wilson et al., 2011) and is enhancement in oxidative stress and a decrease in the antioxidant status,
the leading cause of dementia in the elderly, presenting itself clinically we hypothesized that VA will act as a neuroprotective factor against
by progressive loss of memory and learning (Larson et al., 1992). The Aβ1− 42 induced memory impairment. Accordingly, we designed the
involvement of subcortical structures, including the "hippocampus" in present study for evaluating the possible therapeutic effectiveness of VA
learning and memory processes has been well documented (Bird and as a therapeutic or protective agent in Aβ1− 42 -related memory and
Burgess, 2008; Jarrard, 1993). oxidative stress deficit in male rats.
Oxidative stress reflects the imbalance between the oxidant/antiox­
idant status, with progressive generation and accumulation of reactive 2. Materials and methods
oxygen species (ROS) in healthy cells. Oxidative stress is involved in
many pathological conditions, such as nervous system diseases (Jenner, 2.1. Ethics statement
2003; Sayre et al., 2001). Evidence suggest that oxidative stress is an
early event in AD, occurring prior to plaques or tangle pathology, and The experiments on rats were performed under animal care and use
therefore may play a major pathogenic role in the disease. The AD brain guidelines proved by the institutional ethics committee, Hamadan
also shows evidence of ROS-induced damage (Pereira et al., 2012), and University of Medical Sciences, according to the National Institutes of
patients with AD have damaged, oxidized DNA, RNA, protein and lipid Health Guide for Care and Use of Laboratory Animals. Minimize
products caused by ROS that can be used as possible disease progression suffering was considered.
markers (Ahmad et al., 2017). Considering the well-known association
between oxidative stress and learning and memory deficit (Alzoubi 2.2. Animals and experimental design
et al., 2018; Su et al., 2008), it seems that therapeutic strategies directed
at controlling the excessive production of pro-oxidant factors may be Forty male Wistar rats (200–220 g) provided by the laboratory ani­
valuable to control neurodegeneration in AD and dementia (Agostinho mal house of Hamadan University of Medical Sciences were studied.
et al., 2010). Therefore, there is a lot of attention to the use and devel­ Animals (four rats per cage) were housed an animal house provided with
opment of antioxidant therapies to manage cognitive decline during AD. controlled temperature (20–25 ◦ C/ a 12-h light/dark cycle/50–70 %
Vanillic acid (VA) appears to have considerable antioxidant activity humidity) and water and food were available. After one week of adap­
(Kumpulainen, 1999). Vanillic acid (VA, a famous flavonoid, is abun­ tation, animals were randomly divided into the following groups (n =
dant in multifarious nuts, fruits, and herbs, like honey, and its crude type 6–8 animals in each group): group 1 (control; treated with normal saline
is taken from the roots of an indigenous herb called Angelica Sinensis by oral gavage once a day for 4 weeks), group 2 (sham; intra­
(Fig. 1) (Singh et al., 2015). It is also detected in black rice, where it cerebroventricular (ICV) injection of phosphate-buffered saline (PBS)
peaked at maturity. It is normally used as a food additive (Huang et al., and receiving normal saline by oral gavage once a day for 4 weeks),
2019; Shao et al., 2014). Several pharmacologic effects, including group 3 (Aβ; ICV Aβ1− 42 injections), group 4 (VA; receiving 50 mg/kg of
inhibiting the snake venom activity, as well as antimicrobial, VA by oral gavage once a day for 4 weeks), and group 5 (Aβ + VA; ICV
anti-inflammatory, and antioxidant properties have been reported for Aβ1− 42 injections and receiving VA by oral gavage once a day for 4
VA (Mattila and Kumpulainen, 2002; Tai et al., 2012). The antioxidant weeks). Administration of VA was done by oral gavage, once a day
mechanisms of VA include exacerbating antioxidative status and ATPase through four weeks after the Aβ injection, followed by performing the
activity, free radical scavenging activity, reducing power, the inhibition considered tests. Dose of VA was selected according to the previous
of lipid peroxidation, and increase in catalase and superoxide dismutase studies (Leal et al., 2011; Singh et al., 2015). The experimental design
(SOD) activity while inhibiting cholinergic enzymatic activity and and schedule of the behavioral tests are shown in Fig. 2. Images of
reactivating oxidative-deactivated purine metabolism and pentose behavioral tests are shown in Fig. 3.
phosphate pathways (Chou et al., 2010; Salau et al., 2020). Vanillic acid
has been reported to have cognitive enhancing effects against 2.3. ICV injection of Aβ1− 42 and neurosurgical procedure
streptozotocin-induced neurodegeneration by downregulation of neu­
roinflammatory process and exerting specific anti-inflammatory and We dissolved Aβ1− 42 (1 mg; Tocris Bioscience Bristol, UK) in 1 mL of
antioxidant effects (Singh et al., 2015). PBS as a vehicle and incubated for 7 days at 37 ◦ C before use that results
Although AD symptoms can temporarily be controlled by some in producing amyloid fibrils (with neurotoxic activity) (Asadbegi et al.,
medications and therapeutic methods, no permanent treatment is 2018; Lorenzo and Yankner, 1994). Before transferring animals to a
available. Accordingly, identifying new therapeutic options to slow stereotaxic apparatus (Stoelting Co., USA), anesthetization was done
down or prevent the AD progression is of great importance. Given the through i.p. injection of ketamine (100 mg/kg) and xylazine (10 mg/ kg)
(Karimi et al., 2019). The skull was exposed over the ventricular area
according to the coordinates as follows: 2 mm lateral to the midline, 1.2
mm posterior to bregma, and 4 mm ventral to the cortex surface (Pax­
inos and Watson, 2005). The microsyringes (Hamilton Laboratory
Products, USA) (5 μL) were used for injections through 5 min and per­
formed gently (1 μL/ min). The syringes were kept in the injection site
for 5 min following the injection and then gently removed. PBS was
injected into the Sham-operated group. The rats were recovered for 7
days (Asadbegi et al., 2017). In our experiment, Congo Red staining was
performed to confirm the formation of Aβ plaque in the rats’ brain. For
this purpose, serial coronal sections of 5 μm thickness were obtained at
the level of the hippocampus. Then the slides were studied by optic
microscope and Image J software. Fig. 4 shows the Aβ plaques (red
spots) in the coronal sections of the hippocampus region.

Fig. 1. Chemical structure of the vanillic acid.

265
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

Fig. 2. Experimental design and schedule of the behavioral tests.

Fig. 3. Images of behavioral tests. The Open-Field test (A) was used to assess locomotor activity. The Novel Object Recognition (B) task was used to evaluate
recognition memory. Morris water maze (C), and step-through apparatus (D) were used to evaluate the spatial and passive learning and memory, respectively.

2.4. Behavioral tests a 70 % alcohol solution before placing other rats for preventing olfactory
perception by other animals (Asadbegi et al., 2016; Ganji et al., 2017).
2.4.1. Open field test (locomotion)
The open-field test (OFT) measures locomotor activity, motor 2.4.2. Novel object recognition test (NOR)
impairment, and anxiety in rodents. OFT was first developed to measure NOR was first developed by Ennanceur and Delacour, according to
emotions in rats (Prut and Belzung, 2003). The rats were transferred to the spontaneous behavior of animals for recognizing a novel object in a
the testing room in their home cages and allowed to habituate through familiar environment (Ennaceur and Delacour, 1988). Recognition
30 min prior to testing. The apparatus was made of a black round arena memory as a type of declarative memory evaluates an animal’s capa­
(48 × 41.5 × 36 cm) elevated 60 cm above the floor. The floor can be bility for judging or discriminating between objects, considering visual
seen with a grid of 16 squares. Using a video camera, the paths traveled and tactile data (Antunes and Biala, 2012; Ennaceur and Delacour,
by animals were recorded within 10 min. The numbers of squares 1988). In this research, the NOR test was used for evaluating the VA
crossed using four paws were calculated. The OFT box was washed using effects on cognitive flexibility in a rat model of Aβ neurotoxicity. In brief,

266
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

Fig. 4. Photomicrographs of Congo Red staining in rat hippocampus sections. Staining the Congo Red was done to confirm the presence of Aβ plaque in the
hippocampus (straight arrows) from studied groups. The scale bar is 100 μm.

the apparatus (a black wooden box) (48 × 41.5 × 36 cm) was elevated ethanol solution) for eliminating the remaining odors (Hashemi-Firouzi
60 cm above the floor. During the habituation (before performing the et al., 2018).
test), the rats were allowed to freely explore the box with no object
through 10 min, and then it was subjected to a training phase (30 min 2.4.3. Morris water maze (MWM) task
following habituation), in which two identical plastic cubbies (length: 7 The MWM task was applied for measuring learning and spatial
cm, width: 3.5 cm, height: 8.5 cm) were located in the box. Twenty-four memory. It includes a water-filled (to a depth of 25 cm/ 22 ± 1 ◦ C) black
hours later, the animals were subjected to the retention test, where a circular pool (180 cm in diameter and 60 cm in height. The apparatus
novel (unfamiliar) object (length: 4 cm, width: 5.5 cm, height: 8.5 cm) was placed in a soundproof, dim lighted room; however, there were
was replaced with one of the objects. Using a video camera, the rats’ different visual cues. The pool has four quadrants: north (N), south (S),
behaviors were recorded, and the time taken to explore each object was east (E), and west (W), as well as a fixed hidden platform just below (i.e.,
determined within 10 min. Exploration was considered as the rat placing ~ 1.0 cm) the surface of the water. The training trial took for 4 days at
its nose within 2 cm from the object. The discrimination index (DI) was roughly the same time each day with two blocks with four trials (90 s). A
determined as the time taken to explore the novel object, divided by the 30 s interval was considered between two trials on the platform. The
whole exploration time (Takuma et al., 2014). At the end of each ses­ animals could rest through 5 min between two consecutive blocks. A
sion, using a wet tissue paper the box and objects were cleaned (10 % video camera (Nikon, USA) attached to a computer above the pool was

267
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

used for recording the time taken to reach the hidden platform (the 2.6. Statistical analysis
escape latency). One day following the spatial acquisition phase (fifth
day), the retention phase, the probe trial was conducted, and during the The GraphPad Prism software v. 8.0 was applied for statistical ana­
trial, the platform was removed from the pool, and animals were lyses using one-way and two-way analysis of variance (ANOVA) fol­
allowed to swim for 60 s. The swim speed, escape latency, time spent in lowed by Tukey’s post-hoc test for multiple comparisons. The results are
the target quadrant, and distance traveled were recorded in each trial, In presented as the mean ± S.E.M at a probability level of <0.05.
the visible platform trials, the covering of the platform was done using a
sheet of aluminum foil (Moridi et al., 2020; Rezvani-Kamran et al., 2017; 3. Result
Zarrinkalam et al., 2016).
3.1. Aβ and VA effects on locomotion (in OFT)
2.4.4. Passive avoidance learning (PAL) task
The experimental groups were found with no significant difference in
2.4.4.1. Apparatus. The step-through apparatus was applied to estimate locomotor activity. There were not significant differences in distance
passive avoidance learning and memory. It a two-compartment box traveled [F (4, 36) = 2.417, P = 0.0665, one-way ANOVA, Fig. 5A] and
(bright and dark) with the same size (20 × 20 × 30 cm) isolated through mean velocity [F (4, 39) = 2.155, P = 0.0923, one-way ANOVA, Fig. 5A]
a guillotine door. The bright compartment was made of transparent among experimental groups. Thus, these results confirmed that treat­
plastic, whereas the dark one made from dark opaque plastic. A shock ment with the VA did not affect locomotion.
generator was used for electrifying the floor of the dark chamber. An
insulated stimulator delivered electric shocks (50 Hz, 1 mA, 1.5 s) to the
grid floor of the dark chamber (Barzegar et al., 2015; Komaki et al., 3.2. Aβ and VA effects on recognition memory (in NOR test)
2015).
Fig.6 indicates DI related to the experimental groups. According to
2.4.4.2. Training. Habituation was done 30 min prior to the experi­ the one-way ANOVA results, the groups were found with a significant
ments. The groups had two trials of habituation to the apparatus. difference in the DI [F (4, 43) = 38.78, P < 0.0001, Fig. 6]. Based on the
Accordingly, an animal was located in the light chamber facing away Tukey’s post-hoc test results, the DI of Aβ rats was significantly lower
from the door, followed by raising the door after 30 s. The rats prefer compared with the control and sham groups (P < 0.0001). post hoc
dark environments compared to lit places. Following entering the dark comparisons showed the memory restoring effect of VA agent against
chamber, the door was closed, and 30 s later, the rat was taken from the Aβ. DI increased in the Aβ + VA group compared to the Aβ group (P <
dark chamber placing in the home cage. After an interval of 30 min, the 0.0001). No significant differences were observed in the DI in the Aβ +
habituation was repeated. At the same interval, the first acquisition trial VA group and the control and sham groups based on the one-way
was conducted. ANOVA results (P = 0.8953 and P = 0.0852, respectively).
In the training trial, each rat was located in the light chamber, and 30
s later, the guillotine door was opened. The rat was free for entering the
dark chamber, and 2 min later, a mild electrical shock was applied and 3.3. Aβ and VA effects on memory acquisition (in MWM task training)
the animal was returned to the home cage. We recorded entrance latency
to the dark compartment (step-through latency, STLa) when the rat put The escape latency (i.e., time to reach the hidden platform) and
four paws on the grid floor of the dark chamber. The experiment was swimming distance were used to assess the acquisition of the MWM task.
repeated after 2 min. Finally, a foot shock was received by the rat when There were significant differences in escape latency between experi­
reentered the dark chamber; however, training was finished as the rat mental groups (Treatment effect: F (2.423, 16.96) = 29.28, P < 0.0001,
remained in the light chamber through 120 s (consecutive). The entries Day effect; F (2.148, 15.04) = 42.32, P < 0.0001, repeated measures
number into the dark compartment was noted (Habibitabar et al., 2020; two-way ANOVA, Fig. 7). According to the results obtained from four
Khodamoradi et al., 2015). training days, the escape latency increased in the Aβ group compared
with the other experimental groups. VA improved Aβ-induced impair­
2.4.4.3. Retention. In this step, 24 h following the training trial, the ment in memory acquisition. The escape latency decreased in the VA +
retention test was conducted. Each rat was placed in the light chamber, Aβ group compared with the Aβ group.
followed by raising the door 5 s later, and the step-through latency in the In addition, there were statistically significant differences in traveled
retention trial (STLr), as well as the time spent in the dark compartment distance between experimental groups (Treatment effect: F (3.258,
(TDC) were noted for 300 s. The test was finished as the rat either 91.22) = 27.72, P < 0.0001, Day effect; F (3, 28) = 159.8, P < 0.0001,
entered the dark chamber or stayed in the light chamber for 300 s, repeated measures two-way ANOVA, Fig. 8). Total traveled distance to
showing retention of the passive avoidance response. The retention trial reach platform during four days of hidden platform training reveals Aβ
was free of applying electric shock (Barzegar et al., 2015; Shekarian animals travel further before reaching the platform. The traveled dis­
et al., 2020). tance decreased in the VA + Aβ group compared with the Aβ group.

2.5. Biochemical parameters


3.4. Aβ and VA effects on spatial reference memory (in MWM task probe
After the experiments, anesthetization was done through ketamine test)
(100 mg/kg)/ xylazine (10 mg/ kg) i.p. injection (ethyl carbamate, 1.8
g/kg; i.p.). Blood sampling (4 mL) was done from the vena cava vein and To assess reference memory at the end of learning, a probe trial was
transferred into heparinized tubes. The samples were centrifuged (3500 conducted 24 h after the last training trial on day 5. During this test, the
rpm/ 10 min 4 ◦ C), the serum was frozen at − 80 ◦ C and sent for platform was removed and the time spent in each quadrant of the MWM
biochemistry assessment. In the end, malondialdehyde (MDA), total was recorded. There were significant differences in the time spent in
antioxidant capacity (TAC) and total oxidant status (TOS) levels were target zone between the experimental groups [F (4, 35) = 8.387, P <
also measured (Habibitabar et al., 2020; Komaki et al., 2019; West, 0.0001, one-way ANOVA, Fig. 9]. Aβ animals spent less time in the
1993). target zone in compare with other animals. As shown in Fig. 9, animals
in Aβ + VA group spent more time in the target zone in compare with Aβ
animals.

268
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

Fig. 5. Effect of Vanillic acid and Aβ on locomotion in open field test. The experimental groups were found with no significant difference in locomotor activity.
There were not significant differences in distance traveled (A) and mean velocity (B) among experimental groups. Data are expressed as mean ± SEM.

Fig. 8. Aβ and VA effects on traveled distance (during four days of hidden


platform training in MWM task). There were significant differences in trav­
eled distance to find the hidden platform between the experimental groups. The
traveled distance increased in the Aβ group compared with the other experi­
Fig. 6. Effect of Vanillic acid on Aβ-induced impairment in novel object- mental groups, and decreased in the VA + Aβ group compared with the Aβ
recognition test. Discrimination index of Aβ rats was significantly lower group. Data are presented as mean ± SEM. Aβ vs. control on third day * (P <
than the other groups. Vanillic acid increased the discrimination index of Aβ 0.05) and fourth day **** (P < 0.0001), and vs. sham on third day && (P <
rats. Data are expressed as mean ± SEM. *** P < 0.001. 0.01) and fourth day &&&& (P < 0.0001). Aβ group vs. VA on the third (P <
0.05) and fourth days ## (P < 0.0001), and Aβ vs. Aβ + VA group on third and
fourth days #### (P < 0.0001).

Fig. 7. Aβ and VA effects on escape latency (during four days of hidden


platform training in MWM task). There were significant differences in latency
to find the hidden platform between the experimental groups. The escape la­
tency increased in the Aβ group compared with the other experimental groups,
and decreased in the VA + Aβ group compared with the Aβ group. Data are
presented as mean ± SEM. On the first day: Aβ vs. control *** (P < 0.001) and
sham && (P < 0.01), on the second day: Aβ vs. control *** (P < 0.001) and Fig. 9. Measurement of the spatial reference memory in the probe test. Aβ
sham && (P < 0.01), on the third day: Aβ vs. control *** (P < 0.001) and sham animals spent less time in the target zone in compare with other animals. An­
&&&& (P < 0.0001), and on the fourth day: Aβ vs. control **** (P < 0.0001) imals in Aβ + VA group spent more time in the target zone in compare with Aβ
and sham &&&& (P < 0.0001). Also, on the first day: Aβ vs. VA # (P < 0.05), animals. Data are presented as mean ± SEM. * p < 0.05, ** p < 0.01, **** p
second day: ## (P < 0.01), third day ### (P < 0.001), and fourth day ### < 0.0001.
(P < 0.001). A significant difference was detected between the Aβ vs. Aβ + VA
on the first day # (P < 0.05), second day ## (P < 0.01), third and fourth days 3.5. Aβ and VA effects on the PAL acquisition and retention
### (P < 0.001).
There was no significant difference in the STLa among the experi­
mental groups in the acquisition trial (before receiving the electrical

269
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

shock) [F (4, 35) = 0.6970, P = 0.5992, one-way ANOVA, Fig. 10A]. (4, 24) = 26.35, P < 0.0001, One-way ANOVA, Fig. 11C). Aβ increased
This result indicates that the exploratory behavior of the different TOS in compare with control group (P < 0.0001). Treatment of Aβ an­
groups of rats in the dark did not differ. Also, significant differences were imals with VA decreased TOS (P < 0.0001).
not observed among the different experimental groups with respect to
the number of trials to acquisition criterion [F (4, 35) = 2.333, P = 4. Discussion
0.0748, one-way ANOVA, Fig. 10B].
The retention test was conducted 24 h after the training. It revealed a The major findings of the present study are as follows: In Aβ rats, VA
significant difference in the STLr among the groups [F (4, 35) = 39.94, P 1) decreased plasma oxidative stress, 2) improved impairment in
< 0.0001, One-way ANOVA, Fig. 10C]. Specifically, STLr in the Aβ acquisition and retrieval phases of reference memory (in MWM test), 3)
group was significantly less than the control group (P < 0.0001). STLr ameliorated the impairment in the PAL acquisition and retention. Aβ
increased in the Aβ + VA group compared to the Aβ group (P < 0.0001). and treatment with the VA did not affect locomotion in OF test.
In addition, there was also a statistically significant difference in TDC It has been shown that oxidative stress is involved in the neuropa­
between experimental animals [F (4, 35) = 13.28, P < 0.0001, one-way thology of AD (Markesbery, 1997). Heat shock proteins (HSPs) are
ANOVA, Fig. 10D]. Aβ group spent more time in the dark compartment abnormally present in the neuropathological lesions of AD, suggesting
in comparison with the control animals (P < 0.0001). TDC decreased in that oxidative stress may play significant roles in this disorder (Cam­
the Aβ + VA group compared to the Aβ group (P < 0.0001). All results panella et al., 2018). HSPs are one of the main markers of oxidative
are summarized in Fig. 10. stress. Pappolla et al. by immunohistochemistry revealed a very pro­
nounced increase of superoxide dismutase (SOD) and hemoxygenase-1
(HO-1) in neurons surrounding amyloid deposits in transgenic mouse
3.6. Aβ and VA effects on biomarkers of oxidative stress model of AD (Pappolla et al., 1998). Also, inducing oxidative stress in
primary chick brain neurons by exposure to sublethal doses of H2O2
MDA is the most common indicator of oxidative stress in various increases levels of total secreted endogenous Aβ (Goldsbury et al., 2008).
health conditions (Habibitabar et al., 2020; Khoubnasabjafari et al., Oxidative stress and amyloid toxicity leave neurons chemically vulner­
2015; Weber et al., 2017). The impact of Aβ and VA administration on able (Verri et al., 2012), and plaque formation in certain areas of the
MDA concentration in serum was evaluated. There was a significant CNS causes neuroinflammation and the induction of free radicals, which
difference in the case of MDA among the experimental groups of rats [F leads to the destruction of areas such as the amygdala, hippocampus and
(4, 34) = 9.370, P < 0.0001, One-way ANOVA, Fig. 11A]. As illustrated cortex (Mancuso et al., 2007). Reducing oxidative damage and con­
in Fig. 11A, Aβ increased MDA concentration in serum compared with trolling the excessive production of ROS can be considered a promising
the control group (P < 0.0001, Fig. 11A). Treatment of Aβ animals with strategy for therapeutic intervention in AD and control neuro­
VA decreased MDA concentration (P < 0.01). In addition, there was a degeneration (Dumont et al., 2009). The antioxidative activity of
significant difference in the case of TAC among the experimental groups vanillic acid is well documented (Sang et al., 2002; Shyamala et al.,
of rats [F (4, 28) = 6.239, P = 0.0010, One-way ANOVA, Fig. 11B). Aβ 2007).
decreased TAC (P = 0.0018, Fig. 11B). Treatment of Aβ animals with VA Our data in the present study showed that Aβ1− 42-treated rat
increased total TAC (P = 0.0146). Furthermore, there was a significant exhibited a significant alteration in MDA, TAC, and TOS. Our results are
difference in the case of TOS among the experimental groups of rats [F

Fig. 10. Aβ and VA effects on the PAL acquisition and retention. There was no significant difference in the STLa (A) and number of trials to acquisition criterion
(B) among the experimental groups in the acquisition trial (before receiving the electrical shock). But Aβ decreased STLr (C) and increased TDC (D) in the retention
test. VA reversed the effects of Aβ on STLr and TDC. Data are presented as mean ± SEM. **** p < 0.0001.

270
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

We used OFT for locomotor activity, NOR test for cognitive memory,
MWM test for spatial memory, PAL task for passive avoidance learning
and memory, and finally, oxidative stress biomarkers as possible
mechanisms in the memory-enhancing effects of VA against Aβ. Before
behavioral tests, the motor activity of rats in OFT was evaluated. VA and
surgery did not affect locomotor activity. Therefore, it can be concluded
that the results of behavioral tests are independent of the rats’ motor
activity. In our research, the ICV injection of Aβ1− 40 reduced spatial
memory in the Aβ group. Choi et al., reported that the ICV injection of
Aβ1− 40 in mice destroyed the spatial memory in the MWM test, which
can be due to the accumulation of these peptides in the brain (Choi et al.,
2001). Besides, it has demonstrated that the administration of Aβ leads
to a decrease in antioxidant capacity leading to impairment in synaptic
plasticity, which occurs during learning and memory (Asadbegi et al.,
2018). Furthermore, it has been reported that the Aβ1− 40 injection can
induce memory impairment detected by the PAL task (Jellinger, 2002).
In other word, VA administration in Aβ rats improved their passive
avoidance memory, which can because of the antioxidant properties of
VA.
Consistent with the results of our study, Amin et al. reported that VA
significantly ameliorated cognitive deficits with increased glutathione
(GSH) levels in brain tissues and increased nuclear factor erythroid 2-
related factor 2 (Nrf2) / HO-1 expression in Aβ1− 42-treated mice
(Amin et al., 2017). It has been suggested that the beneficial therapeutic
effects of vanillic acid are through the positive regulation of Akt /
GSK-3β / Nrf2 signaling pathways (Amin et al., 2017). Recent studies
have suggested that Akt/Nrf2 signaling pathways play a central role in
the development process of AD (Hong and An, 2018; Wang et al., 2013),
and activation of Nrf2 provides neuroprotection in AD (Khodagholi
et al., 2010). Previous works have shown a reduction in Nrf2 levels in AD
(Choudhry et al., 2012; Kanninen et al., 2009), and reported that
intrahippocampal injection of a lentiviral vector expressing Nrf2 im­
proves spatial learning in a mouse model of AD (Kanninen et al., 2009).
It has been shown that VA treatment significantly increases Nrf2 protein
expression in the brains of Aβ1− 42-treated mice (Amin et al., 2017).
Based on the above, it can be concluded that one possible mechanism in
the memory-enhancing effects of VA against Aβ, is an increase in Nrf2
protein expression. On the other hand, activation of Akt / Nrf2 signaling
has been reported to reduce learning and memory deficits in APP/PS1
mice (Hong and An, 2018). VA-induced activation in the Akt pathway
can also be considered as one of the possible mechanisms in the
memory-enhancing effects of VA against Aβ (Amin et al., 2017).
Vanillic acid also reduces β-site APP-cleaving enzyme 1 (BACE-1)
expression in the Aβ1− 42-treated mice (Amin et al., 2017). β-secretase 1,
also known as BACE-1 is the key enzyme responsible for APP processing
and Aβ production (Liang et al., 2010), and is associated with neuronal
loss and dysfunction and memory impairment (Haass and Selkoe, 2007;
Yang et al., 2003).
In AD, neuroinflammation as much as plaques and tangles is
involved in AD pathogenesis (Heneka et al., 2015). Neuroinflammation
often results in enduring memory impairment and has been reported as a
Fig. 11. Effect of Aβ and VA on serum levels of malondialdehyde (MDA) part of the neuropathogenesis of cognitive impairment (Allison and
(A), total antioxidant capacity (TAC) (B), total oxidant status (TOS) (C). Aβ Ditor, 2014). VA treatment has been shown to reverse the increased
increased MDA and TOS and decreased TAC. While treatment with VA signif­ expression of inflammatory markers induced by Aβ1− 42 (Amin et al.,
icantly reversed the effects of Aβ on these parameters. Data presented as means 2017), preventing neuroinflammation and memory impairment in the
± S.E.M. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Aβ1− 42-treated mice.
VA-induced elevation in the synaptophysin and post-synapse density
consistent with other previous reports (Abdul et al., 2008). High levels 95 (PSD95) protein can also be considered as one of the possible
of MDA have been suggested as an essential factor in the pathogenesis mechanisms in the memory-enhancing effects of VA against Aβ (Amin
and neuronal damage of AD patients (Aybek et al., 2007). Here, in our et al., 2017). Animal and human studies have shown that PSD95 and
work Aβ increased MDA concentration, and treatment of Aβ animals synaptophysin expression decreases in AD (Amin et al., 2017; Canas
with VA decreased MDA concentration. The study by Amin et al. showed et al., 2009; Tu et al., 2014). PSD95 controls AMPA receptor incorpo­
that the increase in oxidative stress in the brains of Aβ1− 42-treated mice ration during long-term potentiation (LTP) and experience-driven syn­
was significantly reduced by VA treatment (Amin et al., 2017). VA aptic plasticity (Ehrlich and Malinow, 2004). LTP is a principal form of
treatment reduces ROS induction and prevents the depletion of endog­ synaptic plasticity in the mammalian brain, thought to underlie learning
enous reduced glutathione (GSH) levels (Verri et al., 2012). and memory (Bliss and Collingridge, 1993). Therefore, in mice treated

271
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

with Aβ1− 42, decreased synaptophysin and PSD95 levels were associated Agostinho, P.A., Cunha, R., Oliveira, C., 2010. Neuroinflammation, oxidative stress and
the pathogenesis of Alzheimer’s disease. Curr. Pharm. Des. 16, 2766–2778.
with memory impairment (Amin et al., 2017).
Ahmad, W., Ijaz, B., Shabbiri, K., Ahmed, F., Rehman, S., 2017. Oxidative toxicity in
VA has also been shown to improve spatial learning and memory diabetes and Alzheimer’s disease: mechanisms behind ROS/RNS generation.
retention by preventing oxidative stress, and well known to reduce J. Biomed. Sci. 24, 76.
acetylcholinesterase (AChE), tumor necrosis factor α (TNF-α) and Allison, D.J., Ditor, D.S., 2014. The common inflammatory etiology of depression and
cognitive impairment: a therapeutic target. J. Neuroinflammation 11, 1–12.
corticosterone by improving antioxidants to provide neuroprotection Alzoubi, K.H., Mayyas, F.A., Mahafzah, R., Khabour, O.F., 2018. Melatonin prevents
and could be an effective therapeutic agent for the treatment of neuro­ memory impairment induced by high-fat diet: role of oxidative stress. Behav. Brain
degenerative disorders (Singh et al., 2015). Res. 336, 93–98.
Amin, F.U., Shah, S.A., Kim, M.O., 2017. Vanillic acid attenuates Aβ 1-42-induced
AD involves a substantial loss of the elements of the cholinergic oxidative stress and cognitive impairment in mice. Sci. Rep. 7, 1–15.
system (Kása et al., 1997). Deficits in cholinergic function (Giacobini, Antunes, M., Biala, G., 2012. The novel object recognition memory: neurobiology, test
2003) contribute to the pathology of AD, affecting cognition, behavior procedure, and its modifications. Cogn. Process. 13, 93–110.
Asadbegi, M., Yaghmaei, P., Salehi, I., Ebrahim-Habibi, A., Komaki, A., 2016.
and activities of daily living (White and Ruske, 2002). VA has also been Neuroprotective effects of metformin against Aβ-mediated inhibition of long-term
shown to reduce AChE activity in the brain of STZ-treated mice (Singh potentiation in rats fed a high-fat diet. Brain Res. Bull. 121, 178–185.
et al., 2015). Decreased AChE activity increases acetylcholine levels and Asadbegi, M., Yaghmaei, P., Salehi, I., Komaki, A., Ebrahim-Habibi, A., 2017.
Investigation of thymol effect on learning and memory impairment induced by
compensates for cholinergic system defects, which ultimately lead to intrahippocampal injection of amyloid beta peptide in high fat diet-fed rats. Metab.
improved learning and memory (Canas et al., 2009). In the clinic, Brain Dis. 32, 827–839.
pharmacological inhibition of AChE improves memory (Weinstock et al., Asadbegi, M., Komaki, A., Salehi, I., Yaghmaei, P., Ebrahim-Habibi, A., Shahidi, S.,
Sarihi, A., Asl, S.S., Golipoor, Z., 2018. Effects of thymol on amyloid-β-induced
2001) and cholinesterase inhibitors are used as potential anti-Alzheimer
impairments in hippocampal synaptic plasticity in rats fed a high-fat diet. Brain Res.
drugs (Weinstock et al., 2001). Bull. 137, 338–350.
In the current study, our results showed a significant reduction in Aybek, H., Ercan, F., Aslan, D., Şahiner, T., 2007. Determination of malondialdehyde,
memory function as evidenced by MWM, PAL and NOR test perfor­ reduced glutathione levels and APOE4 allele frequency in late-onset Alzheimer’s
disease in Denizli, Turkey. Clin. Biochem. 40, 172–176.
mance. We also observed that VA treatment (30 mg/kg, p.o, 4 weeks) Barzegar, S., Komaki, A., Shahidi, S., Sarihi, A., Mirazi, N., Salehi, I., 2015. Effects of
improved memory, as shown by the reduction in escape latency and cannabinoid and glutamate receptor antagonists and their interactions on learning
swimming distance, and the increased amount of time spent in the target and memory in male rats. Pharmacol. Biochem. Behav. 131, 87–90.
Bell, K.F.S., Claudio Cuello, A., 2006. Altered synaptic function in Alzheimer’s disease.
quadrant during the probe test. In the PAL test, we observed a lower TDC Eur. J. Pharmacol. 545, 11–21.
and a higher STLr following VA treatment. Vanillic acid treatment Bird, C.M., Burgess, N., 2008. The hippocampus and memory: insights from spatial
ameliorated the effects of Aβ1− 42 on discrimination index in NOR test. processing. Nat. Rev. Neurosci. 9, 182–194.
Bliss, T.V., Collingridge, G.L., 1993. A synaptic model of memory: long-term potentiation
The observed improvement in memory function associated with VA in the hippocampus. Nature 361, 31.
treatment demonstrates its neuroprotective effect against Aβ1− 42- Braak, H., Braak, E., 1990. Alzheimer’s disease: striatal amyloid deposits and
induced memory impairment. neurofibrillary changes. J. Neuropathol. Exp. Neurol. 49, 215–224.
Burns, A., Iliffe, S., 2009. Alzheimer’s disease. BMJ 338, b158. Vol., ed.^eds.
Campanella, C., Pace, A., Caruso Bavisotto, C., Marzullo, P., Marino Gammazza, A.,
5. Conclusion Buscemi, S., Palumbo Piccionello, A., 2018. Heat shock proteins in Alzheimer’s
disease: role and targeting. Int. J. Mol. Sci. 19, 2603.
Canas, P.M., Porciúncula, L.O., Cunha, G.M., Silva, C.G., Machado, N.J., Oliveira, J.M.,
In summary, our results demonstrated that the reversal of cognitive Oliveira, C.R., Cunha, R.A., 2009. Adenosine A2A receptor blockade prevents
deficits by VA treatment in Aβ1− 42-treated rats might result from the synaptotoxicity and memory dysfunction caused by β-amyloid peptides via p38
antioxidant activity of VA; vanillic acid treatment was associated with mitogen-activated protein kinase pathway. J. Neurosci. 29, 14741–14751.
Choi, S.H., Park, C.H., Koo, J.W., Seo, J.-H., Kim, H.-S., JEONG, S.-J., Lee, J.-H., Kim, S.
an increased in TAC and a decreased in TOS and MDA. Accordingly, VA- S., Suh, Y.-H., 2001. Memory impairment and cholinergic dysfunction by centrally
based natural therapeutic agents are applicable for preventing and administered Aβ and carboxyl-terminal fragment of Alzheimer’s APP in mice. Faseb
treating oxidative stress-associated disorders, like AD. However, more J. 15, 1816–1818.
Chou, T.H., Ding, H.Y., Hung, W.J., Liang, C.H., 2010. Antioxidative characteristics and
studies and clinical trials should be conducted to measure the VA
inhibition of α-melanocyte-stimulating hormone-stimulated melanogenesis of
effectiveness in AD in humans. vanillin and vanillic acid from Origanum vulgare. Exp. Dermatol. 19, 742–750.
Choudhry, F., Howlett, D.R., Richardson, J.C., Francis, P.T., Williams, R.J., 2012. Pro-
oxidant diet enhances β/γ secretase-mediated APP processing in APP/PS1 transgenic
CRediT authorship contribution statement mice. Neurobiol. Aging 33, 960–968.
Dumont, M., Wille, E., Stack, C., Calingasan, N.Y., Beal, M.F., Lin, M.T., 2009. Reduction
Nesa Ahmadi: Conceptualization, Methodology, Investigation. of oxidative stress, amyloid deposition, and memory deficit by manganese
superoxide dismutase overexpression in a transgenic mouse model of Alzheimer’s
Samaneh Safari: Formal analysis, Software. Naser Mirazi: Writing -
disease. Faseb J. 23, 2459–2466.
review & editing, Data curation. Seyed Asaad Karimi: Writing - original Ehrlich, I., Malinow, R., 2004. Postsynaptic density 95 controls AMPA receptor
draft, Resources, Validation. Alireza Komaki: Supervision, Conceptu­ incorporation during long-term potentiation and experience-driven synaptic
alization, Writing - review & editing, Resources. plasticity. J. Neurosci. 24, 916–927.
Ennaceur, A., Delacour, J., 1988. A new one-trial test for neurobiological studies of
memory in rats. 1: behavioral data. Behav. Brain Res. 31, 47–59.
Ganji, A., Salehi, I., Nazari, M., Taheri, M., Komaki, A., 2017. Effects of Hypericum
Declaration of Competing Interest scabrum extract on learning and memory and oxidant/antioxidant status in rats fed a
long-term high-fat diet. Metab. Brain Dis. 32, 1255–1265.
There are no conflicts of interest to declare. Giacobini, E., 2003. Cholinergic function and Alzheimer’s disease. Int. J. Geriatr. Psych
18, S1–S5.
Goldsbury, C., Whiteman, I.T., Jeong, E.V., Lim, Y.A., 2008. Oxidative stress increases
Acknowledgments levels of endogenous amyloid-β peptides secreted from primary chick brain neurons.
Aging Cell 7, 771–775.
Haass, C., Selkoe, D.J., 2007. Soluble protein oligomers in neurodegeneration: lessons
The current study was funded (Grant No.: IR.BASU.REC.1398.030) from the Alzheimer’s amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 8, 101–112.
by Faculty of Basic Sciences, Bu-Ali Sina University, Hamadan, Iran. The Habibitabar, E., Moridi, H., Shateri, H., Karimi, S.A., Salehi, I., Komaki, A., Sarihi, A.,
authors are grateful to the staff of the Neurophysiology Research Center, 2020. Chronic NaHS treatment improves spatial and passive avoidance learning and
memory and anxiety-like behavior and decreases oxidative stress in rats fed with a
Hamadan University of Medical Sciences for supporting this study.
high-fat diet. Brain Res. Bull.
Hashemi-Firouzi, N., Shahidi, S., Soleimani-Asl, S., Komaki, A., 2018. 5-
References Hydroxytryptamine receptor 6 antagonist, SB258585 exerts neuroprotection in a rat
model of Streptozotocin-induced Alzheimer’s disease. Metab. Brain Dis. 33,
1243–1253.
Abdul, H.M., Sultana, R., Clair, D.K.S., Markesbery, W.R., Butterfield, D.A., 2008.
Oxidative damage in brain from human mutant APP/PS-1 double knock-in mice as a
function of age. Free Radic. Biol. Med. 45, 1420–1425.

272
Downloaded from https://iranpaper.ir

N. Ahmadi et al. Brain Research Bulletin 170 (2021) 264–273

Heneka, M.T., Carson, M.J., El Khoury, J., Landreth, G.E., Brosseron, F., Feinstein, D.L., Prince, M., Wimo, A., Guerchet, M., Ali, G., Wu, Y., Prina, M., 2015. The global impact of
Jacobs, A.H., Wyss-Coray, T., Vitorica, J., Ransohoff, R.M., 2015. dementia. World Alzheimer Report 1–82.
Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 14, 388–405. Prut, L., Belzung, C., 2003. The open field as a paradigm to measure the effects of drugs
Hong, Y., An, Z., 2018. Hesperidin attenuates learning and memory deficits in APP/PS1 on anxiety-like behaviors: a review. Eur. J. Pharmacol. 463, 3–33.
mice through activation of Akt/Nrf2 signaling and inhibition of RAGE/NF-κB Querfurth, H.W., LaFerla, F.M., 2010. Alzheimer’s disease. N. Engl. J. Med. 362,
signaling. Arch. Pharm. Res. 41, 655–663. 329–344.
Huang, X., Xi, Y., Mao, Z., Chu, X., Zhang, R., Ma, X., Ni, B., Cheng, H., You, H., 2019. Reddy, P.H., Tripathi, R., Troung, Q., Tirumala, K., Reddy, T.P., Anekonda, V.,
Vanillic acid attenuates cartilage degeneration by regulating the MAPK and PI3K/ Shirendeb, U.P., Calkins, M.J., Reddy, A.P., Mao, P., Manczak, M., 2012. Abnormal
AKT/NF-κB pathways. Eur. J. Pharmacol. 859, 172481. mitochondrial dynamics and synaptic degeneration as early events in Alzheimer’s
Hynd, M.R., Scott, H.L., Dodd, P.R., 2004. Glutamate-mediated excitotoxicity and disease: implications to mitochondria-targeted antioxidant therapeutics. Biochim.
neurodegeneration in Alzheimer’s disease. Neurochem. Int. 45, 583–595. Biophys. Acta. (BBA) - Mol. Basis Disease 1822, 639–649.
Jarrard, L.E., 1993. On the role of the hippocampus in learning and memory in the rat. Rezvani-Kamran, A., Salehi, I., Shahidi, S., Zarei, M., Moradkhani, S., Komaki, A., 2017.
Behav. Neural Biol. 60, 9–26. Effects of the hydroalcoholic extract of Rosa damascena on learning and memory in
Jellinger, K., 2002. Alzheimer disease and cerebrovascular pathology: an update. male rats consuming a high-fat diet. Pharm. Biol. 55, 2065–2073.
J. Neural Transm. 109, 813–836. Salau, V.F., Erukainure, O.L., Ibeji, C.U., Olasehinde, T.A., Koorbanally, N.A., Islam, M.
Jenner, P., 2003. Oxidative stress in Parkinson’s disease. Ann. Neurol. 53 (Suppl 3), S., 2020. Vanillin and vanillic acid modulate antioxidant defense system via
S26–36 discussion S36-8. amelioration of metabolic complications linked to Fe 2+-induced brain tissues
Kanninen, K., Heikkinen, R., Malm, T., Rolova, T., Kuhmonen, S., Leinonen, H., Ylä- damage. Metab. Brain Dis. 1–12.
Herttuala, S., Tanila, H., Levonen, A.-L., Koistinaho, M., 2009. Intrahippocampal Samandouras, G., Teddy, P., Cadoux-Hudson, T., Ansorge, O., 2006. Amyloid in
injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse neurosurgical and neurological practice. J. Clin. Neurosci. 13, 159–167.
model of Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 106, 16505–16510. Sang, S., Lapsley, K., Jeong, W.-S., Lachance, P.A., Ho, C.-T., Rosen, R.T., 2002.
Karimi, S.A., Salehi, I., Shykhi, T., Zare, S., Komaki, A., 2019. Effects of exposure to Antioxidative phenolic compounds isolated from almond skins (Prunus amygdalus
extremely low-frequency electromagnetic fields on spatial and passive avoidance Batsch). J. Agric. Food Chem. 50, 2459–2463.
learning and memory, anxiety-like behavior and oxidative stress in male rats. Behav. Sayre, L.M., Smith, M.A., Perry, G., 2001. Chemistry and biochemistry of oxidative stress
Brain Res. 359, 630–638. in neurodegenerative disease. Curr. Med. Chem. 8, 721–738.
Kása, P., Rakonczay, Z., Gulya, K., 1997. The cholinergic system in Alzheimer’s disease. Shao, Y., Xu, F., Sun, X., Bao, J., Beta, T., 2014. Phenolic acids, anthocyanins, and
Prog. Neurobiol. 52, 511–535. antioxidant capacity in rice (Oryza sativa L.) grains at four stages of development
Khodagholi, F., Eftekharzadeh, B., Maghsoudi, N., Rezaei, P.F., 2010. Chitosan prevents after flowering. Food Chem. 143, 90–96.
oxidative stress-induced amyloid β formation and cytotoxicity in NT2 neurons: Shekarian, M., Komaki, A., Shahidi, S., Sarihi, A., Salehi, I., Raoufi, S., 2020. The
involvement of transcription factors Nrf2 and NF-κB. Mol. Cell. Biochem. 337, protective and therapeutic effects of vinpocetine, a PDE1 inhibitor, on oxidative
39–51. stress and learning and memory impairment induced by an intracerebroventricular
Khodamoradi, N., Komaki, A., Salehi, I., Shahidi, S., Sarihi, A., 2015. Effect of vitamin E (ICV) injection of amyloid beta (aβ) peptide. Behav. Brain Res. 383, 112512.
on lead exposure-induced learning and memory impairment in rats. Physiol. Behav. Shyamala, B., Naidu, M.M., Sulochanamma, G., Srinivas, P., 2007. Studies on the
144, 90–94. antioxidant activities of natural vanilla extract and its constituent compounds
Khoubnasabjafari, M., Ansarin, K., Jouyban, A., 2015. Reliability of malondialdehyde as through in vitro models. J. Agric. Food Chem. 55, 7738–7743.
a biomarker of oxidative stress in psychological disorders. BioImpacts: BI. 5, 123. Singh, J.C.H., Kakalij, R.M., Kshirsagar, R.P., Kumar, B.H., Komakula, S.S.B., Diwan, P.
Komaki, A., Karimi, S.A., Salehi, I., Sarihi, A., Shahidi, S., Zarei, M., 2015. The treatment V., 2015. Cognitive effects of vanillic acid against streptozotocin-induced
combination of vitamins E and C and astaxanthin prevents high-fat diet induced neurodegeneration in mice. Pharm. Biol. 53, 630–636.
memory deficits in rats. Pharmacol. Biochem. Behav. 131, 98–103. Su, B., Wang, X., Nunomura, A., Moreira, P.I., Lee, H.-g., Perry, G., Smith, M.A., Zhu, X.,
Komaki, H., Faraji, N., Komaki, A., Shahidi, S., Etaee, F., Raoufi, S., Mirzaei, F., 2019. 2008. Oxidative stress signaling in Alzheimer’s disease. Curr. Alzheimer Res. 5,
Investigation of protective effects of coenzyme Q10 on impaired synaptic plasticity 525–532.
in a male rat model of Alzheimer’s disease. Brain Res. Bull. 147, 14–21. Tai, A., Sawano, T., Ito, H., 2012. Antioxidative properties of vanillic acid esters in
Kumpulainen, J., 1999. Natural Antioxidants and Anticarcinogens in Nutrition. Vol.. multiple antioxidant assays. Biosci. Biotechnol. Biochem. 76, 314–318.
Woodhead Publishing. Takuma, K., Hara, Y., Kataoka, S., Kawanai, T., Maeda, Y., Watanabe, R., Takano, E.,
Larson, E.B., Kukull, W.A., Katzman, R.L., 1992. Cognitive impairment: dementia and Hayata-Takano, A., Hashimoto, H., Ago, Y., 2014. Chronic treatment with valproic
Alzheimer’s disease. Annu. Rev. Public Health 13, 431–449. acid or sodium butyrate attenuates novel object recognition deficits and
Leal, L., Pierdoná, T., Góes, J., Fonsêca, K., Canuto, K., Silveira, E., Bezerra, A., Viana, G., hippocampal dendritic spine loss in a mouse model of autism. Pharmacol. Biochem.
2011. A comparative chemical and pharmacological study of standardized extracts Behav. 126, 43–49.
and vanillic acid from wild and cultivated Amburana cearensis AC Smith. Tanzi, R.E., Bertram, L., 2005. Twenty years of the Alzheimer’s disease amyloid
Phytomedicine. 18, 230–233. hypothesis: a genetic perspective. Cell 120, 545–555.
Liang, B., Duan, B.-Y., Zhou, X.-P., Gong, J.-X., Luo, Z.-G., 2010. Calpain activation Tu, S., Okamoto, S.-i., Lipton, S.A., Xu, H., 2014. Oligomeric Aβ-induced synaptic
promotes BACE1 expression, amyloid precursor protein processing, and amyloid dysfunction in Alzheimer’s disease. Mol. Neurodegener. 9, 1–12.
plaque formation in a transgenic mouse model of Alzheimer disease. J. Biol. Chem. Verri, M., Pastoris, O., Dossena, M., Aquilani, R., Guerriero, F., Cuzzoni, G., Venturini, L.,
285, 27737–27744. Ricevuti, G., Bongiorno, A., 2012. Mitochondrial Alterations, Oxidative Stress and
Lorenzo, A., Yankner, B.A., 1994. Beta-amyloid neurotoxicity requires fibril formation Neuroinflammation in Alzheimer’s Disease. Vol., ed.^eds. SAGE Publications Sage
and is inhibited by congo red. Proc. Natl. Acad. Sci. U.S.A. 91, 12243–12247. UK, London, England.
Mancuso, C., Scapagini, G., Curro, D., Giuffrida Stella, A.M., De Marco, C., Butterfield, D. Vest, R.S., Pike, C.J., 2013. Gender, sex steroid hormones, and Alzheimer’s disease.
A., Calabrese, V., 2007. Mitochondrial dysfunction, free radical generation and Horm. Behav. 63, 301–307.
cellular stress response in neurodegenerative disorders. Front Biosci. 12, 1107–1123. Wang, C.-Y., Xie, J.-W., Xu, Y., Wang, T., Cai, J.-H., Wang, X., Zhao, B.-L., An, L.,
Markesbery, W.R., 1997. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic. Wang, Z.-Y., 2013. Trientine reduces BACE1 activity and mitigates amyloidosis via
Biol. Med. 23, 134–147. the AGE/RAGE/NF-κB pathway in a transgenic mouse model of Alzheimer’s disease.
Mattila, P., Kumpulainen, J., 2002. Determination of free and total phenolic acids in Antioxid. Redox Signal. 19, 2024–2039.
plant-derived foods by HPLC with diode-array detection. J. Agric. Food Chem. 50, Weber, D., Stuetz, W., Buerkle, A., Grune, T., 2017. Oxidative stress biomarkers in the
3660–3667. MARK-AGE Study. Free Radic. Biol. Med. 112, 58.
Mendiola-Precoma, J., Berumen, L., Padilla, K., Garcia-Alcocer, G., 2016. Therapies for Weinstock, M., KIRSCHBAUM-SLAGER, N., Lazarovici, P., Bejar, C., Youdim, M.B.,
prevention and treatment of Alzheimer’s disease. Biomed Res. Int. 2016. Shoham, S., 2001. Neuroprotective effects of novel cholinesterase inhibitors derived
Moridi, H., Sarihi, A., Habibitabar, E., Shateri, H., Salehi, I., Komaki, A., Karimi, J., from rasagiline as potential anti-Alzheimer drugs. Ann. N. Y. Acad. Sci. 939,
Karimi, S.A., 2020. Effects of post-training administration of LY341495, as an 148–161.
mGluR2/3 antagonist on spatial memory deficit in rats fed with high-fat diet. IBRO West, M.J., 1993. Regionally specific loss of neurons in the aging human hippocampus.
Rep. 9, 241–246. Neurobiol. Aging 14, 287–293.
Palop, J.J., Mucke, L., 2010. Amyloid-β–induced neuronal dysfunction in Alzheimer’s White, K.G., Ruske, A.C., 2002. Memory deficits in Alzheimer’s disease: the encoding
disease: from synapses toward neural networks. Nat. Neurosci. 13, 812–818. hypothesis and cholinergic function. Psychon. Bull. Rev. 9, 426–437.
Pappolla, M., Chyan, Y., Omar, R., Hsiao, K., Perry, G., Smith, M., Bozner, P., 1998. Wilson, R.S., Leurgans, S.E., Boyle, P.A., Bennett, D.A., 2011. Cognitive decline in
Evidence of oxidative stress and in vivo neurotoxicity of beta-amyloid in a transgenic prodromal Alzheimer disease and mild cognitive impairment. Arch. Neurol. 68,
mouse model of Alzheimer’s disease: a chronic oxidative paradigm for testing 351–356.
antioxidant therapies in vivo. Am. J. Pathol. 152, 871. Wirths, O., Bayer, T.A., 2010. Neuron loss in transgenic mouse models of Alzheimer’s
Paxinos, G., Watson, C., 2005. The Rat Brain in Stereotaxic Coordinates. Elsevier disease. Int. J. Alzheimers Dis. 2010.
Academic Press, Burlington (MA). Vol., ed.^eds. San Diego (CA), London (UK). Yang, L.-B., Lindholm, K., Yan, R., Citron, M., Xia, W., Yang, X.-L., Beach, T., Sue, L.,
Pereira, M.D., Ksiazek, K., Menezes, R., 2012. Oxidative Stress in Neurodegenerative Wong, P., Price, D., 2003. Elevated β-secretase expression and enzymatic activity
Diseases and Ageing. Vol., ed.^eds. Hindawi.. detected in sporadic Alzheimer disease. Nat. Med. 9, 3–4.
Piaceri, I., Nacmias, B., Sorbi, S., 2013. Genetics of familial and sporadic Alzheimer’s Zarrinkalam, E., Heidarianpour, A., Salehi, I., Ranjbar, K., Komaki, A., 2016. Effects of
disease. Front Biosci. (Elite Ed) 5, 167–177. endurance, resistance, and concurrent exercise on learning and memory after
morphine withdrawal in rats. Life Sci. 157, 19–24.

273

You might also like