ArticlePDF Available

Tualang Honey and its Methanolic Fraction Improve LPS-induced Learning and Memory Impairment in Male Rats: Comparison with Memantine

Authors:

Abstract

Background Tualang honey (TH) has been shown to exert beneficial effects on learning and memory function in various animal models. However, its learning and memory effects in lipopolysaccharide (LPS) rat model have not been elucidated. Objective The present study aimed to investigate the cognitive-enhancing effects of TH and its methanolic fraction in comparison to the clinically approved N-methyl-D-aspartate (NMDA) receptor antagonist (memantine) using LPS rat model. Methods A total of ninety male Sprague Dawley rats were divided into 5 groups: (i) control, (ii) untreated LPS (iii) LPS treated with 200 mg/kg TH, (iv) LPS treated with 150 mg/kg methanol fraction of TH (MTH) and (v) LPS treated with 10 mg/kg memantine. All treatments were administered intraperitoneally once daily for 14 days. Morris water maze (MWM) and novel object recognition (NOR) tests were performed to assess spatial and recognition memory function. Results The present study confirmed that LPS significantly impairs spatial and recognition memory and alone treatment with TH or MTH improved spatial and recognition memory comparable to memantine. Conclusion Both TH and its methanolic fraction improved spatial and recognition memory of LPS rat model comparable to memantine. Thus, TH and its methanolic fraction have potential preventivetherapeutic effects for neurodegenerative diseases involving neuroinflammation.
Current Nutrition & Food Science
ISSN: 1573-4013
eISSN: 2212-3881
SCIENCE
BENTHAM
Send Orders for Reprints to reprints@benthamscience.net
Current Nutrition & Food Science, 2020, 16, 333-342
333
RESEARCH ARTICLE
Tualang Honey and its Methanolic Fraction Improve LPS-induced Learning
and Memory Impairment in Male Rats: Comparison with Memantine
Wan M.H.W. Yaacob1, Idris Long1, Rahimah Zakaria2,* and Zahiruddin Othman3
1School of Health Sciences, 2Department of Physiology, 3Department of Psychiatry, School of Medical Sciences, Universiti
Sains Malaysia Health Campus, 16150 Kubang Kerian, Malaysia
Abstract: Background: Tualang honey (TH) has been shown to exert beneficial effects on learning and
memory function in various animal models. However, its learning and memory effects in lipopolysaccha-
ride (LPS) rat model have not been elucidated.
Objective: The present study aimed to investigate the cognitive-enhancing effects of TH and its meth-
anolic fraction in comparison to the clinically approved N-methyl-D-aspartate (NMDA) receptor
antagonist (memantine) using LPS rat model.
Methods: A total of ninety male Sprague Dawley rats were divided into 5 groups: (i) control, (ii) untreat-
ed LPS (iii) LPS treated with 200 mg/kg TH, (iv) LPS treated with 150 mg/kg methanol fraction of TH
(MTH) and (v) LPS treated with 10 mg/kg memantine. All treatments were administered intraperitone-
ally once daily for 14 days. Morris water maze (MWM) and novel object recognition (NOR) tests were
performed to assess spatial and recognition memory function.
Results: The present study confirmed that LPS significantly impairs spatial and recognition memory and
alone treatment with TH or MTH improved spatial and recognition memory comparable to memantine.
Conclusion: Both TH and its methanolic fraction improved spatial and recognition memory of LPS rat
model comparable to memantine. Thus, TH and its methanolic fraction have potential preventive-
therapeutic effects for neurodegenerative diseases involving neuroinflammation.
A R T I C L E H I S T O R Y
Received: April 16, 2018
Revised: August 15, 2018
Accepted: November 09, 2018
DOI:
10.2174/1573401315666181130103456
Keywords: Learning and memory, lipopolysaccharide, methanolic function, morris water maze, novel object recognition, tua-
lang honey.
1. INTRODUCTION
Neuroinflammation is characterized by the microglia ac-
tivation along with the expression of major inflammatory
mediators in the brain tissue and its chronic state, giving rise
to the progression of neurodegenerative diseases including
Alzheimer’s disease (AD) and Parkinson’s disease [1].
Neuroinflammation can cause cognitive decline, even if it is
acutely stimulated by an immunostimulatory element such as
lipopolysaccharide (LPS) [2].
LPS is an endotoxin found in the outer membrane of
gram-negative bacteria. It induces systemic inflammatory
response syndrome via toll-like receptor (TLR) [3]. Binding
of LPS-TLR4 complex on the surface of microglia activates
several signal transduction pathways such as phosphoinosi-
tide 3-kinase/protein kinase B (PI3K/AKT), mitogen-
activated protein kinase (MAPK) and mammalian target of
rapamycin (mTOR), which finally lead to NF-κB activation.
*Address correspondence to this author at the Department of Physiology,
School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang
Kerian, Kelantan, Malaysia; Tel/Fax: +609-7676156, +609-7653370;
E-mails: rahimah@usm.my
NF-κB activation mediates production of pro-inflammatory
cytokines, chemokines and inducible enzymes such as induc-
ible nitric oxide synthase (iNOS) and cyclooxygenase-2
(COX-2), which altogether result in neuroinflammation [4,
5] and cognitive impairment [6].
To date, donepezil, rivastigmine, galantamine and me-
mantine are the US Food and Drug Administration (FDA)
approved treatments for AD. These drugs are symptomatic
and do not delay or stop the progression of the disease [7].
However, these drugs exhibit modest, but particularly relia-
ble, benefit for cognition, global status and functional ability
[8]. Thus, there is a need to continue our search for treatment
of AD.
Among the proposed mechanisms for AD, an antioxidant
complex or combination with CNS anti-inflammatory holds
the best promise of an effective AD treatment [9]. Tualang
honey (TH), a Malaysian multi floral jungle honey, is shown
to exhibit anti-inflammatory [10], antiproliferative [11],
antioxidant [12], anti-diabetic [13] and neuroprotective ef-
fects [14]. There is increasing evidence to support the cogni-
tive enhancer effects of TH in various animal models. Study
by Azman et al. [15] showed that treatment with 200 mg/kg
2212-3881/20 $65.00+.00 © 2020 Bentham Science Publishers
334 Current Nutrition & Food Science, 2020, Vol. 16, No. 3 Yaacob et al.
TH for 28 days ameliorate memory performance and
increase the levels of brain antioxidant enzymes against
noise stress-induced memory deficit in aged rats. TH intake
was able to improve both working and reference memory by
radial arm maze experiment as well as increase pyramidal
neurons count in hippocampal tissue of adult rats [16].
Saxena et al. [17] reported that TH reduced spatial memory
impairment and protected the hippocampal neurons from
damage in cerebral hypoperfusion-induced neurodegenera-
tion. These previous studies demonstrated that TH supple-
mentation enhanced neuronal proliferation in CA2, CA3 and
dentate gyrus region of the hippocampus in adult rats [14].
TH also mitigated the increased activity of acetylcholinester-
ase (AChE), and normalized the brain-derived neurotrophic
factor (BDNF), malondialdehyde and superoxide dismutase
levels in the hippocampus of stressed ovariectomized rats
[18-20].
Our group has investigated the antioxidant potentials of
four different samples of TH i.e. non irradiated TH, gamma
irradiated TH, methanolic fraction of TH and ethyl acetate
fraction of TH. We found its methanolic fraction exhibited
highest antioxidant potentials when compared among the
four TH samples tested (unpublished). We hypothesized that
TH, especially its methanolic fraction could protect from
LPS-induced learning and memory impairment. Therefore,
the present study aimed to investigate the cognitive enhanc-
ing effects of TH and its methanolic fraction in comparison
to the clinically approved N-methyl-D-aspartate (NMDA)
receptor antagonist (memantine) using LPS rat model.
2. MATERIALS AND METHODS
2.1. Honey Samples
TH from single batch of honey supplied by Federal Agri-
cultural Marketing Authorities (FAMA), Malaysia was used
in the present study. The honey was filtered to eliminate any
solid particles, concentrated on heat at 40°C and evaporated
to achieve a water content of about 20%. It was then gamma
() irradiated at 25 kGy at Steril Gamma (M) Sdn. Bhd. (Se-
langor, Malaysia) for sterilization. The final concentration of
the TH in a 230g/bottle was 1.3 g/mL.
2.2. Extraction of Honey
The honey was subjected to solid phase extraction (SPE)
to extract phenolic compounds as described by Kaškonienė
et al. [21] with slight modifications. The honey samples were
dissolved in five parts of acidified deionized water (pH 2
attained by using 2.0 M hydrochloric acid) until completely
fluid and then filtered through cotton wool to remove solid
particles. The solution was passed through preconditioned
C18 cartridges (6 mL x 1 g). The cartridges were precondi-
tioned by successively passing 6 mL each of methanol and
acidified water. The aqueous honey solution was applied to
the cartridge at a drop-wise flow rate to ensure efficient ad-
sorption of phenolic compounds. The cartridges were then
washed with acidified water to remove all sugars and other
polar constituents of honey. Later, the adsorbed compounds
were eluted with methanol and evaporated using a rotary
evaporator at 40°C. The methanolic fraction of Tualang hon-
ey (MTH) was used in the present study.
2.3. Animals
Ninety adult male Sprague Dawley rats of approximately
3 months old, with body weight of 270 ± 20g, were obtained
from Animal Research and Service Centre (ARASC), Uni-
versiti Sains Malaysia. The rats were allowed to acclimatize
to a new environment for one week prior to the start of the
experiment. They were maintained under standard laboratory
condition at room temperature of 21 ± 2°C with free access
to food and water. The photoperiod was the same throughout
the study with 12 h light-dark cycles. The protocols were
conducted in accordance with USM Guide for the Care and
Use of Laboratory Animals and approved by Animal Ethics
Committee [USM / Animal Ethics Approval I 2015/(95)
(605)].
2.4. Experimental Design
The rats were randomly divided into five groups (n=18
rats/group) as follows: (i) control rats, (ii) LPS-treated with
distilled water, (iii) LPS-treated with TH 200 mg/kg [15],
(iv) LPS-treated with MTH 150 mg/kg [22] and (v) LPS-
treated with memantine 10 mg/kg [23]. All treatments were
administered intraperitoneally once daily for 14 days, start-
ing from Day 1 to 14 [15]. LPS from E.coli 0111:B4 (Sigma-
Aldrich, St. Louis, MO) at the dose of 5 mg/kg [24] was in-
jected on Day 4. Body weight was measured twice; on Day 1
and 24. All rats were subjected to Morris water maze
(MWM) test on Day 15 and novel object recognition (NOR)
test on Day 21. After behavioral assessment and body
weighting, rats were sacrificed and brain tissues were imme-
diately collected and stored at -80°C for later use. The exper-
imental schedule is shown in Fig. (1).
2.5. Morris Water Maze (MWM)
The MWM test was performed on Day 15 as described
by Vorhees and Williams [25]. The acquisition training was
performed over the course of 5 days and the retention test
was conducted on Day 20. The apparatus consists of a circu-
lar water pool 150 cm in diameter and 60 cm in height. It
was filled with 23 ± 1°C water with a depth of 42 cm. The
water in the pool was made opaque by adding 1 kg tapioca
starch powder. A hidden circular platform (10 cm in diame-
ter), which was placed 1 cm below the water surface served
as the escape platform. The pool was divided into four quad-
rants: northeast (NE), northwest (NW), southeast (SE), and
southwest (SW) at equal distances on the rim. The platform
was placed in the center of the NE quadrant. Several visual
cues were positioned around the pool in plain sight of the
rats and these were exactly in the same position for all trials.
A digital camera was mounted to the ceiling straight above
the center of the pool and was connected to a computerized
recording system equipped with a tracking program (S-
MART 3.0: Panlab Co., Barcelona, Spain), which allowed
on- and off-line automated tracking of the paths taken by the
rats. All the trials were completed between 09.00 to 16.00 h
in a sound-attenuated laboratory.
Tualang Honey Improves Learning and Memory Impairment Current Nutrition & Food Science, 2020, Vol. 16, No. 3 335
Fig. (1). Experimental schedule.
2.5.1. Hidden Platform Trial for the Acquisition Test
The rats were gently placed into the water facing the wall
and were permitted to swim until they found the hidden plat-
form, which remained in a fixed position throughout the test.
The trials lasted for a maximum of 60s, and the escape laten-
cy was expressed by the swimming time to reach the
submerge platform in the pool. Upon reaching the hidden
platform, the rat was permitted to stay on it for at least 5s
before being withdrawn. If the rats failed to locate the hidden
platform within 60s they should be guided to the platform
and were placed on it for 10s for reinforcement before being
withdrawn. The latency was assigned as 60s amid one trial
and the next, the water in the pool was stirred to eliminate
olfactory traces of previous swim patterns. The entire sched-
ule was continued for 5 days and each animal had 4 trials for
training per day with 5-15 min inter-trial interval. The 4
starting points were randomized on each day of training.
Escape latencies and distance traveled by the rats during
training sessions were measured.
2.5.2. Probe Trial for the Retention Test
On Day 20, the probe test was performed with a cut-off
time of 60 s. Animal behaviors were analyzed with a grid
design consist of 4 quadrants (Fig. 2). This grid design, con-
structed with a computer-based image analyzer, was super-
imposed over the maze and viewed on a monitor. The num-
ber of target heading, distance traveled and time spent in
each of the quadrants were determined.
Fig. (2). Computerized grid design which used in the probe test.
Black circle representing the platform site.
2.6. Novel Object Recognition (NOR)
The NOR task was performed in an open field apparatus
(60 × 60 × 40 cm) painted in black color as described in a
previous study [26]. Before starting the training, all animals
had at least two free exploration sessions for contextual ha-
bituation, with no object inside the box for 5 min. During the
training session, two identical objects (A, A) were placed at
fixed distances within the square field. The rats were then
allowed to explore freely for 10 min. Time spent exploring
each object was recorded manually. After two successive
training sessions, the test sessions were conducted consisting
of two terms, short-term and long-term memory. The testing
interval of short-term and long-term memory was 2h and
24h respectively, after the last training session. This time one
of the objects was replaced with a new object (A, B). The
rats were allowed to explore freely for 10 min, and the
number of contacts with the original and novel object was
recorded.
All objects comprised of plastic toys (washable) and had
a height of about 4 cm. Objects presented similar textures,
colors and sizes, but distinctive shapes. The objects were
positioned in two adjacent corners, 10 cm from the side walls
to avoid accidental touching during the initial, thigmotaxic
exploration. The maze and objects were wiped cleaned with
20% alcohol after each test session to eliminate odor cues.
Object exploration was defined as directing the nose and
vibrissae to the object at a distance of less than 2 cm, as if
smelling it with caution; turning around, bumping or sitting
upon the object was not considered exploratory behaviors.
Total exploration time of the novel objects was recorded and
used to calculate a discrimination index: time spent with a
novel object (B) /total time exploring both objects (A + B)
during test sessions [27]. Prolong exploration time of the
novel object or better preference to novel object was consid-
ered as successful retention of memory for the familiar ob-
ject. The absence of any difference in the exploration of two
objects was interpreted as memory deficit [28].
2.7. Statistical Analysis
Study data were presented as means ± standard errors
(SEM). Differences between groups were evaluated using
paired Student’s t-test, one-way Analysis of variance
(ANOVA) and repeated measure ANOVA, where appropriate.

       
 
 
!"#%
&'*8
9;<=#%
%
>?
@

G%J%


 




336 Current Nutrition & Food Science, 2020, Vol. 16, No. 3 Yaacob et al.
Fig. (3). Mean body weight of experimental rats on Day 1 and 24 of the experiment. Data are expressed as means ± SEM. *p <0.05 vs. LPS
group; #p <0.05 vs. control group.
A probability value of less than 0.05 was used to indicate a
significant difference.
3. RESULTS
3.1. Effects on Bodyweight Changes
Body weight of each rat in each group was measured on
day 1 and 24 of the experiment (Fig. 3). Statistical analysis
revealed that there were no significant differences in means
of body weight of 5 groups on day 1 of the experiment.
However, a significant reduction in the mean body weight
gain of untreated LPS rats when compared with controls on
day 24 of the experiment (p <0.05). Interestingly, the LPS
rats treated with TH, MTH and memantine showed weight
gain comparable to controls. Their body weights were signif-
icantly higher when compared to the untreated LPS rats on
day 24 of the experiment (p <0.05), indicating that treatment
with TH, MTH and memantine could prevent LPS-induced
body weight loss.
3.2. Effects on Spatial Learning
Following 2 weeks of treatment, spatial learning of ex-
perimental rats was assessed by using MWM. Fig. (4A)
showed the total escape latency during the acquisition phase
for 5 days. The time per day for escape latency decreased
steadily in all the experimental groups. The LPS rats demon-
strated marked retardation in escape latency, indicating
memory deficit. LPS rats treated with TH and MTH rapidly
learned the location of the immersed hidden platform and
reached it within 23 and 22s on the fifth day of the trials,
respectively which was comparable to the group of rats treated
with memantine. The analysis of escape latency displayed
that the rats in the TH group had significantly decreased es-
cape latency on the third and fifth day (p <0.05) whereas the
rats in MTH group on first (p <0.05), fourth (p <0.05) and
fifth day (p <0.01), respectively as compared to the LPS
group. The distance traveled by each group was closely as-
sociated with the escape latencies. Rats treated with meman-
tine, TH and MTH showed a consistent decrease in searching
distance during the acquisition trial especially third day on-
wards when compared to rats in LPS group (p <0.05 on the
third, fourth and fifth day; Fig. (4B) indicates that TH and
MTH treatment could ameliorate LPS-induced spatial learn-
ing and memory deficit comparable to memantine.
3.3. Effects on Memory Retention
In order to determine the effects on memory, the probe
test behaviors were investigated on the sixth day by analyz-
ing the percentages of time and distance required to swim in
the ‘target quadrant’, where the escape platform had been
placed. The LPS rats showed severely impaired performanc-
es; they spent significantly (p <0.001) less time in the target
quadrant site compared to those of the control group. The
rats in TH and MTH groups spent significantly longer time
(p <0.05) than those in the LPS group. Similarly, TH and
MTH had significantly increased (p <0.05) swimming dis-
tance in the target quadrant as compared to the LPS group
(Fig. 5A & B). These results suggest that both TH and MTH
could ameliorate the LPS-induced poor memory retention
comparable to LPS rats treated with memantine. From the
trajectory view, it can be clearly seen that LPS-treated rats
swam in all of the four quadrants (aimless swimming) rather
than swimming mostly in the target quadrant. Whereas con-
trols and LPS rats treated with memantine, TH and MTH
swam mostly in the target quadrant (Fig. 5E). The number of
target crossing in the LPS group was statistically reduced (p
<0.001) compared to the control group, while TH treatment
significantly increased (p <0.05) the number of target cross-
ing compared to the LPS group (Fig. 5C). Both TH and
MTH groups were not significantly different from the other
groups in terms of the mean swimming speed, as computed
by dividing the total swimming distance by the latency (Fig.
5D).
3.4. Effects on Recognition Memory
Fig. (6) shows the effects of various treatments on object
recognition ability. No significant difference was observed in
the short-term memory of all rats. However, after 24 h of last









 

!
"#
$&
$&
'>
XZZ
Z
Tualang Honey Improves Learning and Memory Impairment Current Nutrition & Food Science, 2020, Vol. 16, No. 3 337
Fig. (4). Effects TH, MTH and memantine treatment on the escape latency to the hidden platform (A) and the distance traveled to the hidden
platform; during first to fifth day of acquisition trials (B). Data are expressed as means ± SEM. *p <0.05 and **p <0.01 vs. LPS group; #p
<0.05 and ##p <0.01 vs. control group.
Fig. (5) contd…




?

?
[%\!<J&%8
&8
!
"#
$&
$&
'>
Z
JJ
ZZ
Z
Z
JJ
J
ZZ
JJ
ZZ
Z
?

?

?

?

?
?
@%[%\!<J&8
&8
!
"#
$&
$&
'>
J
JJ
ZZ
Z
Z
J
Z
Z
Z
JJ
Z
Z
ZZ
Z
?

?

?

?

?
?
! "# $& $& '>
$>'Q\>$^
_Q>'!Q
XXX
ZZ
Z
Z
(A)
(B)
(A)
338 Current Nutrition & Food Science, 2020, Vol. 16, No. 3 Yaacob et al.
Fig. (5). Effects of TH, MTH and memantine treatment on memory retention of LPS rats. A) Percentage of time spent in target zone, B) per-
centage of distance travelled in target zone, C) number of target crossing, D) swimming speed, and E) swimming path; of experimental rats in
the probe trial without a platform. Data are expressed as means ± SEM. *p <0.05 and **p <0.01 vs. LPS group; ##p <0.01 and ###p <0.001 vs.
control group.
?

?

?

?

?
?
! "# $& $& '>
>`${!!\>$^
_Q>'!Q
XXX
ZZ
ZZ
|
|?
|
|?
|
|?
|
|?
! "# $& $& '>
$>
_Q>'!Q
Z
XX
ZZ
?

?

?
! "# $& $& '>
_Q>'!Q
! "# "#}$& "#}$& "#}'>
(B)
(C)
(D)
(E)
~>''>Q`'`)
Tualang Honey Improves Learning and Memory Impairment Current Nutrition & Food Science, 2020, Vol. 16, No. 3 339
Fig. (6). Effects of TH, MTH and memantine treatment on short-term and long-term recognition memory of LPS rats. Data are expressed as
means ± SEM. *p <0.05 and **p <0.01 vs. LPS group; ###p <0.001 vs. control group.
training session (long-term memory) LPS-induced rats had
significantly (p <0.001) lower discrimination index com-
pared to controls, indicating that LPS was associated with
poor recognition memory. Unlike untreated LPS rats, the
LPS rats that received TH and MTH for 14 days revealed
significantly (p <0.05) higher discrimination index which
was comparable to memantine group.
4. DISCUSSION
Honey has long been used as food as well as a natural
remedy for various diseases such as cardiovascular diseases,
inflammatory disorders, cancer, neurological degeneration,
infectious diseases and aging [29, 30]. Al-Himyari [31] had
proposed that honey intake is associated with a low risk of
AD and may prevent neurodegenerative process of the dis-
ease. However, based on the current literature search, the
data evaluating the efficacy of honey in learning and
memory models is still lacking.
Previous studies revealed body weight loss in rats or
mice that were subjected to the single administration of LPS
[32, 33]. This finding was consistent with our finding. We
used intraperitoneal (i.p.) injection of 5 mg/kg LPS to induce
neuroinflammation and memory impairment. Our data
demonstrated that the untreated LPS rats gradually gained
less body weight throughout the experimental period. LPS is
known to produce ‘sickness behaviors’ [34] which include
reduction in activity, reduction in exploration, decreased
social interaction, fever, reduction in consumption of food
and drink, hypersomnia, activation of the hypothalamic-
pituitary-adrenal (HPA) axis and activation of a sympathetic
system [35, 36]. The treated LPS groups displayed a restored
body weight gain which was comparable to controls, sug-
gesting that TH, MTH and memantine treatment inhibited
the LPS-induced ‘sickness behavior’ [37].
The present study confirmed that LPS significantly im-
pairs water maze learning and memory, as the escape latency
and the traveled distance to the hidden platform were in-
creased. These findings indicate a deficit in learning ability
and reference memory [38]. Similar findings were also noted
in other studies [32, 39]. The LPS-induced learning and
memory impairments were reversed by oral administration of
TH, MTH and memantine for two weeks. Previous study
demonstrated that subcutaneous memantine (10 mg/kg/day)
ameliorates the spatial memory impairments produced by
LPS alone in rats [40]. The study proposed that memantine
acts partly through its ability to reduce the effects of neuroin-
flammation, resulting in normal spatial learning. Thus, it is
possible that anti-inflammatory properties of Tualang honey
and its methanolic fraction [41] reduce the effects of neu-
roinflammation, leading to normal spatial learning.
The present study also confirmed that LPS significantly
impairs memory retention, as the time spent and distance in
the quadrant in which the platform was formerly located
were decreased. Rats treated with TH and MTH, however,
spent more time and distance in the target quadrant, suggest-
ing that both TH and its methanol extract could ameliorate
the LPS-induced poor memory retention comparable to me-
mantine. However, there were no differences in swimming
speed and ability to find the visible platform (in cued version
of water maze) among the experimental groups. The findings
may suggest that no changes were observed in motivation or
sensorimotor coordination in all the rats [42].
Additionally, rats that received TH and MTH exhibited a
preference for the novel object in the NOR task, as indicated
by the improvement in the percentage of discrimination in-
dex. The NOR task is based on spontaneous, natural explora-
tory behavior of rats to non-aversive stimuli and is a pure
working memory test free of confounds [43]. In the present
study, LPS rats demonstrated intact short-term but impaired
long-term recognition memory. This may suggest a failure in
memory consolidation and not to impairments in memory
encoding, attention or performance [44]. Nonetheless, our
results demonstrated that administration of TH and MTH
could ameliorate the LPS-induced decline in long-term
recognition memory comparable to memantine. The recogni-
tion memory improvement following treatment with TH has
also been observed in other animal models of memory defi-
cits [14, 15].



?


$''& "$''&
>`>'>>>_\
!
"#
$&
$&
'>
XXX
ZZ ZZ
Z
340 Current Nutrition & Food Science, 2020, Vol. 16, No. 3 Yaacob et al.
LPS-induced amnesia rodent model is one of the well-
established animal models of memory deficits [45]. LPS is a
potent endotoxin and is highly resistant to degradation by
mammalian enzymes thus providing a persistent inflammato-
ry stimulus that produces proinflammatory cytokines, chem-
okines and reactive oxygen species (ROS) [46]. Proinflam-
matory cytokines are key molecules that modulate immune
responses. Their lack of reversibility in chronic inflammation
would trigger dyshomeostasis [47]. Chronic inflammation is
characterized by long-standing microglia activation that sus-
tains the release of inflammatory mediators, leading to an
increase in ROS and nitrosative stress. This perpetuates the
inflammatory cycle [48], further prolonging the inflamma-
tion which in turn lead to cognitive impairment [49]. Previ-
ous studies demonstrated that LPS-induced inflammation in
the brain produces severe learning and memory deficits in a
variety of behavioral tasks [50-52].
In the present study, daily administration of TH and
MTH for 14 days attenuated the LPS-induced spatial and
recognition memory impairment in male rats comparable to
memantine. It is possible that TH and its methanolic frac-
tions act through their potent antioxidant free radical scav-
enging and anti-inflammatory properties [53] to improve
cognitive behaviors. Both antioxidant and anti-inflammatory
properties of honey could be the underlying cause for the
reduction in the damaging effect of oxidative stress-induced
neurodegeneration and cognitive impairment [54]. Further
studies are required to investigate the mechanism of action of
TH and its methanolic fraction on oxidative stress and neu-
roinflammation pathways in the rat brain to support our hy-
pothesis.
CONCLUSION
The present study demonstrates that TH and MTH were
able to reduce the LPS-induced spatial and recognition
memory impairment in male rats comparable to memantine.
Thus, TH and its methanolic fraction have a potential
preventive-therapeutic effect for neurodegenerative diseases
involving neuroinflammation.
ETHICS APPROVAL AND CONSENT TO
PARTICIPATE
The protocols were conducted in accordance to USM
Guide for the Care and Use of Laboratory Animals and ap-
proved by Animal Ethics Committee [USM / Animal Ethics
Approval I 2015/(95) (605)].
HUMAN AND ANIMAL RIGHTS
No humans were used in this research. All animal re-
search procedures followed were in accordance with the
Australian Code of Practice for the Care and Use of Animals
for Scientific Purposes (7th Edition, 2004).
CONSENT FOR PUBLICATION
Not applicable.
AVAILABILITY OF DATA AND MATERIALS
The authors confirm that the data supporting the findings
of this study are available within the article.
FUNDING
This study was supported by the Fundamental Research
Grant Scheme (FRGS), Malaysia with Grant no. 203/PPSP/
6171174.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Quintanilla RA, Orellana JA, von Bernhardi R. Understanding risk
factors for Alzheimer’s disease: interplay of neuroinflammation,
connexin-based communication and oxidative stress. Arch Med
Res 2012; 43(8): 632-44.
http://dx.doi.org/10.1016/j.arcmed.2012.10.016 PMID: 23142264
[2] Song JH, Lee JW, Shim B, et al. Glycyrrhizin alleviates neuroin-
flammation and memory deficit induced by systemic lipopoly-
saccharide treatment in mice. Molecules 2013; 18(12): 15788-803.
http://dx.doi.org/10.3390/molecules181215788 PMID: 24352029
[3] Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflam-
mation: the role and consequences. Neurosci Res 2014; 79: 1-12.
http://dx.doi.org/10.1016/j.neures.2013.10.004 PMID: 24144733
[4] Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mecha-
nisms underlying inflammation in neurodegeneration. Cell 2010;
140(6): 918-34.
http://dx.doi.org/10.1016/j.cell.2010.02.016 PMID: 20303880
[5] Park SE, Sapkota K, Kim S, Kim H, Kim SJ. Kaempferol acts
through mitogen-activated protein kinases and protein kinase
B/AKT to elicit protection in a model of neuroinflammation in
BV2 microglial cells. Br J Pharmacol 2011; 164(3): 1008-25.
http://dx.doi.org/10.1111/j.1476-5381.2011.01389.x
PMID: 21449918
[6] Shabab T, Khanabdali R, Moghadamtousi SZ, Kadir HA, Mohan
G. Neuroinflammation pathways: a general review. Int J Neurosci
2017; 127(7): 624-33.
http://dx.doi.org/10.1080/00207454.2016.1212854
PMID: 27412492
[7] Yiannopoulou KG, Papageorgiou SG. Current and future treat-
ments for Alzheimer’s disease. Ther Adv Neurol Disord 2013;
6(1): 19-33.
[8] Herrmann N, Chau SA, Kircanski I, Lanctôt KL. Current and
emerging drug treatment options for Alzheimer’s disease: a syste-
matic review. Drugs 2011; 71(15): 2031-65.
http://dx.doi.org/10.2165/11595870-000000000-00000
PMID: 21985169
[9] Summers WK. Current and future treatments of memory com-
plaints and Alzheimer’s disease. Therapy 2011; 8(5): 491-504.
http://dx.doi.org/10.2217/thy.11.60
[10] Ahmad I, Jimenez H, Yaacob NS, Yusuf N. Tualang honey protects
keratinocytes from ultraviolet radiation-induced inflammation and
DNA damage. Photochem Photobiol 2012; 88(5): 1198-204.
http://dx.doi.org/10.1111/j.1751-1097.2012.01100.x
PMID: 22276569
[11] Nurul Syazana MS, Halim AS, Gan SH, Shamsuddin S. Antiproli-
ferative effect of methanolic extraction of tualang honey on human
keloid fibroblasts. BMC Complement Altern Med 2011; 11: 82.
http://dx.doi.org/10.1186/1472-6882-11-82 PMID: 21943200
Tualang Honey Improves Learning and Memory Impairment Current Nutrition & Food Science, 2020, Vol. 16, No. 3 341
[12] Khalil MI, Alam N, Moniruzzaman M, Sulaiman SA, Gan SH.
Phenolic acid composition and antioxidant properties of Malaysian
honeys. J Food Sci 2011; 76(6): C921-8.
http://dx.doi.org/10.1111/j.1750-3841.2011.02282.x
PMID: 22417491
[13] Erejuwa OO, Gurtu S, Sulaiman SA, Ab Wahab MS, Sirajudeen
KN, Salleh MS. Hypoglycemic and antioxidant effects of honey
supplementation in streptozotocin-induced diabetic rats. Int J Vitam
Nutr Res 2010; 80(1): 74-82.
http://dx.doi.org/10.1024/0300-9831/a000008 PMID: 20533247
[14] Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Mohd Ismail ZI,
Muthuraju S. Tualang honey supplement improves memory per-
formance and hippocampal morphology in stressed ovariectomized
rats. Acta Histochem 2014; 116(1): 79-88.
http://dx.doi.org/10.1016/j.acthis.2013.05.004 PMID: 23810156
[15] Azman KF, Zakaria R, AbdAziz C, Othman Z, Al-Rahbi B. Tua-
lang honey improves memory performance and decreases depressi-
ve-like behavior in rats exposed to loud noise stress. Noise Health
2015; 17(75): 83-9.
http://dx.doi.org/10.4103/1463-1741.153388 PMID: 25774610
[16] Kamarulzaidi M, Mohd Yusoff M, Mohamed A, Hasan Adli D.
Tualang honey consumption enhanced hippocampal pyramidal
count and spatial memory performance of adult male rats. Sains
Malays 2016; 45(2): 215-20.
[17] Saxena A, Phyu H. IM A-A, Oothuman P. Improved spatial lear-
ning and memory performance following Tualang honey treatment
during cerebral hypoperfusion-induced neurodegeneration. J Transl
Sci 2016; 2(5): 264-71.
http://dx.doi.org/10.15761/JTS.1000150
[18] Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Enhan-
cement of BDNF concentration and restoration of the hypothala-
mic-pituitary-adrenal axis accompany reduced depressive-like be-
haviour in stressed ovariectomised rats treated with either Tualang
honey or estrogen. Sci World J 2014 2014; 2014: 310821.
http://dx.doi.org/10.1155/2014/310821
[19] Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. The
effects of Tualang honey supplement on medial prefrontal cortex
morphology and cholinergic system in stressed ovariectomised rats.
Int J Appl Res Nat Prod 2014; 7(3): 28-36.
[20] Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Protec-
tive effects of Tualang honey against oxidative stress and anxiety-
like behaviour in stressed ovariectomized rats. Int Sch Res Notices
2014 2014; 2014: 10.
http://dx.doi.org/10.1155/2014/521065http://dx.doi.org/10.1155/20
14/521065
[21] Kaškonienė V, Maruška A, Kornyšova O. Quantitative and qualita-
tive determination of phenolic compounds in honey. Cheminė
Technologija 2009; 3(52): 74-80.
[22] Wan Yaacob WMH, Long I, Zakaria R, Othman Z. Protective
effects of Tualang honey and its methanolic fraction against lipopo-
lysaccharide-induced oxidative damage in the rats’ hippocampus.
Health Environ J 2018; 9(Suppl. 1): 36.
[23] Lagrèze WA, Knörle R, Bach M, Feuerstein TJ. Memantine is
neuroprotective in a rat model of pressure-induced retinal ischemia.
Invest Ophthalmol Vis Sci 1998; 39(6): 1063-6.
PMID: 9579489
[24] Rose R, Banerjee A, Ramaiah SK. Characterization of a lipopoly-
saccharide mediated neutrophilic hepatitis model in Sprague Da-
wley rats. J Appl Toxicol 2007; 27(6): 602-11.
http://dx.doi.org/10.1002/jat.1243 PMID: 17370240
[25] Vorhees CV, Williams MT. Morris water maze: procedures for
assessing spatial and related forms of learning and memory. Nat
Protoc 2006; 1(2): 848-58.
http://dx.doi.org/10.1038/nprot.2006.116 PMID: 17406317
[26] Bevins RA, Besheer J. Object recognition in rats and mice: a one-
trial non-matching-to-sample learning task to study ‘recognition
memory’. Nat Protoc 2006; 1(3): 1306-11.
http://dx.doi.org/10.1038/nprot.2006.205 PMID: 17406415
[27] Kim JH, Wang Q, Choi JM, Lee S, Cho EJ. Protective role of caf-
feic acid in an Aβ25-35-induced Alzheimer’s disease model. Nutr
Res Pract 2015; 9(5): 480-8.
http://dx.doi.org/10.4162/nrp.2015.9.5.480 PMID: 26425277
[28] Carlini VP, Martini AC, Schiöth HB, Ruiz RD, Fiol de Cuneo M,
de Barioglio SR. Decreased memory for novel object recognition in
chronically food-restricted mice is reversed by acute ghrelin admi-
nistration. Neuroscience 2008; 153(4): 929-34.
http://dx.doi.org/10.1016/j.neuroscience.2008.03.015
PMID: 18434026
[29] Bogdanov S, Jurendic T, Sieber R, Gallmann P. Honey for nutrition
and health: a review. J Am Coll Nutr 2008; 27(6): 677-89.
http://dx.doi.org/10.1080/07315724.2008.10719745
PMID: 19155427
[30] Honey-health and therapeutic qualities. The National Honey Board:
Logmont, CO, USA 2003. Available from: http: //www.nhb.org/
[31] Al-Himyari FA. The use of honey as a natural preventive therapy
of cognitive decline and dementia in the middle east. Alzheimers
Dement 2009; 5(4): 247.
http://dx.doi.org/10.1016/j.jalz.2009.04.248
[32] Lee B, Sur B, Park J, et al. Ginsenoside rg3 alleviates lipopoly-
saccharide-induced learning and memory impairments by anti-
inflammatory activity in rats. Biomol Ther (Seoul) 2013; 21(5):
381-90.
http://dx.doi.org/10.4062/biomolther.2013.053 PMID: 24244826
[33] Zhang X-Y, Cao J-B, Zhang L-M, Li Y-F, Mi W-D. Deferoxamine
attenuates lipopolysaccharide-induced neuroinflammation and me-
mory impairment in mice. J Neuroinflammation 2015; 12: 20.
http://dx.doi.org/10.1186/s12974-015-0238-3 PMID: 25644393
[34] Klein SL, Nelson RJ. Activation of the immune-endocrine system
with lipopolysaccharide reduces affiliative behaviors in voles. Be-
hav Neurosci 1999; 113(5): 1042-8.
http://dx.doi.org/10.1037/0735-7044.113.5.1042 PMID: 10571486
[35] Hart BL. Biological basis of the behavior of sick animals. Neurosci
Biobehav Rev 1988; 12(2): 123-37.
http://dx.doi.org/10.1016/S0149-7634(88)80004-6 PMID: 3050629
[36] Pugh CR, Kumagawa K, Fleshner M, Watkins LR, Maier SF, Rudy
JW. Selective effects of peripheral lipopolysaccharide administra-
tion on contextual and auditory-cue fear conditioning. Brain Behav
Immun 1998; 12(3): 212-29.
http://dx.doi.org/10.1006/brbi.1998.0524 PMID: 9769157
[37] Castanon N, Bluthé RM, Dantzer R. Chronic treatment with the
atypical antidepressant tianeptine attenuates sickness behavior in-
duced by peripheral but not central lipopolysaccharide and inter-
leukin-1beta in the rat. Psychopharmacology (Berl) 2001; 154(1):
50-60.
http://dx.doi.org/10.1007/s002130000595 PMID: 11292006
[38] Frank-Cannon TC, Alto LT, McAlpine FE, Tansey MG. Does
neuroinflammation fan the flame in neurodegenerative diseases?
Mol Neurodegener 2009; 4: 47-7.
http://dx.doi.org/10.1186/1750-1326-4-47 PMID: 19917131
[39] Zhao W-X, Zhang J-H, Cao J-B, et al. Acetaminophen attenuates
lipopolysaccharide-induced cognitive impairment through an-
tioxidant activity. J Neuroinflammation 2017; 14(1): 17.
http://dx.doi.org/10.1186/s12974-016-0781-6 PMID: 28109286
[40] Rosi S, Vazdarjanova A, Ramirez-Amaya V, Worley PF, Barnes
CA, Wenk GL. Memantine protects against LPS-induced neuroin-
flammation, restores behaviorally-induced gene expression and
spatial learning in the rat. Neuroscience 2006; 142(4): 1303-15.
http://dx.doi.org/10.1016/j.neuroscience.2006.08.017
PMID: 16989956
[41] Devasvaran K, Yong Y-K. Anti-inflammatory and wound healing
properties of Malaysia Tualang honey. Curr Sci 2016; 110(1): 47-
51.
http://dx.doi.org/10.18520/cs/v110/i1/48-52
[42] Zarifkar A, Choopani S, Ghasemi R, et al. Agmatine prevents LPS-
induced spatial memory impairment and hippocampal apoptosis.
Eur J Pharmacol 2010; 634(1-3): 84-8.
http://dx.doi.org/10.1016/j.ejphar.2010.02.029 PMID: 20184876
[43] Lalonde R. The neurobiological basis of spontaneous alternation.
Neurosci Biobehav Rev 2002; 26(1): 91-104.
http://dx.doi.org/10.1016/S0149-7634(01)00041-0
PMID: 11835987
[44] Vogel-Ciernia A, Wood MA. Examining object location and object
recognition memory in mice. Curr Protoc Neurosci 2014; 69(8.31):
1-17.
http://dx.doi.org/10.1002/0471142301.ns0831s69
[45] Zakaria R, Wan Yaacob WMH, Othman Z, Long I, Ahmad AH,
Al-Rahbi B. Lipopolysaccharide-induced memory impairment in
342 Current Nutrition & Food Science, 2020, Vol. 16, No. 3 Yaacob et al.
rats: a model of Alzheimer’s disease. Physiol Res 2017; 66(4): 553-
65.
http://dx.doi.org/10.33549/physiolres.933480 PMID: 28406691
[46] Tanaka S, Ide M, Shibutani T, et al. Lipopolysaccharide-induced
microglial activation induces learning and memory deficits without
neuronal cell death in rats. J Neurosci Res 2006; 83(4): 557-66.
http://dx.doi.org/10.1002/jnr.20752 PMID: 16429444
[47] Ghosh S, Lertwattanarak R, Garduño JdeJ, et al. Elevated muscle
TLR4 expression and metabolic endotoxemia in human aging. J
Gerontol A Biol Sci Med Sci 2015; 70(2): 232-46.
http://dx.doi.org/10.1093/gerona/glu067 PMID: 24846769
[48] Tansey MG, McCoy MK, Frank-Cannon TC. Neuroinflammatory
mechanisms in Parkinson’s disease: potential environmental trig-
gers, pathways, and targets for early therapeutic intervention. Exp
Neurol 2007; 208(1): 1-25.
http://dx.doi.org/10.1016/j.expneurol.2007.07.004
PMID: 17720159
[49] Schmid CD, Melchior B, Masek K, et al. Differential gene expres-
sion in LPS/IFNgamma activated microglia and macrophages: in
vitro versus in vivo. J Neurochem 2009; 109(Suppl. 1): 117-25.
http://dx.doi.org/10.1111/j.1471-4159.2009.05984.x
PMID: 19393017
[50] Zhu B, Wang Z-G, Ding J, et al. Chronic lipopolysaccharide expo-
sure induces cognitive dysfunction without affecting BDNF ex-
pression in the rat hippocampus. Exp Ther Med 2014; 7(3): 750-4.
http://dx.doi.org/10.3892/etm.2014.1479 PMID: 24520281
[51] Hritcu L, Ciobica A, Stefan M, Mihasan M, Palamiuc L, Nabeshi-
ma T. Spatial memory deficits and oxidative stress damage follo-
wing exposure to lipopolysaccharide in a rodent model of Parkin-
son’s disease. Neurosci Res 2011; 71(1): 35-43.
http://dx.doi.org/10.1016/j.neures.2011.05.016 PMID: 21663772
[52] Joshi R, Garabadu D, Teja GR, Krishnamurthy S. Silibinin amelio-
rates LPS-induced memory deficits in experimental animals. Neu-
robiol Learn Mem 2014; 116(0): 117-31.
http://dx.doi.org/10.1016/j.nlm.2014.09.006 PMID: 25444719
[53] Vallianou V, Gounari P, Skourtis A, Panagos J, Kazazis C. Honey
and its anti-inflammatory, anti-bacterial and anti-oxidant proper-
ties. Gen Med (Los Angel) 2014; 2: 132.
http://dx.doi.org/10.4172/2327-5146.1000132
[54] Mijanur Rahman M, Gan SH, Khalil MI. Neurological effects of
honey: current and future prospects. Evid Based Complement Al-
ternat Med 2014; 2014: 958721.
http://dx.doi.org/10.1155/2014/958721 PMID: 24876885
... A growing body of evidence suggests that anti-in ammatory agents with neuroprotective qualities are potential tau-targeting therapies [5]. Numerous studies found that several anti-in ammatory drugs can reduce LPS-induced neuroin ammation [6,7]. Nevertheless, only a few research have a look into the precise mechanism of attenuation of LPS-induced neuroin ammation. ...
... During the rst 7 days of rats' habituation to the new environment, the rats were observed to detect abnormalities. If no abnormality was detected, the rats were randomly divided into ve groups (n = 10) as follows: (i) control, (ii) LPS 5 mg/kg [7], (iii) LPS treated with minocycline 25 mg/kg (15), (iv) LPS treated with minocycline 50 mg/kg (15) and (v) LPS treated with memantine 10 mg/kg [7]. Minocycline (USP, 12601 Twinbrook Pkwy, Rockville, MD) and memantine (USP, 12601 Twinbrook Pkwy, Rockville, MD) treatments were administered intraperitoneally for 14 days and LPS, obtained from E. coli 0111: B4 (Sigma-Aldrich, St. Louis, MO), was injected intraperitoneally once on day 5 of the experiment. ...
... During the rst 7 days of rats' habituation to the new environment, the rats were observed to detect abnormalities. If no abnormality was detected, the rats were randomly divided into ve groups (n = 10) as follows: (i) control, (ii) LPS 5 mg/kg [7], (iii) LPS treated with minocycline 25 mg/kg (15), (iv) LPS treated with minocycline 50 mg/kg (15) and (v) LPS treated with memantine 10 mg/kg [7]. Minocycline (USP, 12601 Twinbrook Pkwy, Rockville, MD) and memantine (USP, 12601 Twinbrook Pkwy, Rockville, MD) treatments were administered intraperitoneally for 14 days and LPS, obtained from E. coli 0111: B4 (Sigma-Aldrich, St. Louis, MO), was injected intraperitoneally once on day 5 of the experiment. ...
Preprint
Full-text available
Introduction The neuroinflammatory response was seen to impact the formation of phosphorylated tau protein in Alzheimer’s disease (AD). This study aims to investigate the molecular mechanism of minocycline in reducing phosphorylated tau protein formation in the hippocampus of lipopolysaccharide (LPS)-induced rats. Methods Fifty adult male Sprague Dawley (SD) rats were randomly allocated to 1 of 5 groups: control, LPS (5 mg/kg), LPS + minocycline (25 mg/kg), LPS + minocycline (50 mg/kg) and LPS + memantine (10 mg/kg). Minocycline and memantine were administered intraperitoneally (i.p) for two weeks, and LPS was injected i.p. once on day 5. ELISA was used to determine the level of phosphorylated tau protein in SD rats' hippocampal tissue. The density and expression of Toll-like receptor-4 (TLR-4), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-кβ), tumour necrosis factor-alpha (TNF-α), and cyclooxygenase (COX)-2 were determined using Western blot and immunohistochemistry. Results Minocycline, like memantine, prevented LPS-induced increase in phosphorylated tau protein level via reduced density and expression of TLR-4, NF-кβ, TNF-α and COX-2 proteins in rat hippocampal tissue. Interestingly, higher doses were shown to be more neuroprotective than lower doses. Conclusion This study suggests that minocycline suppresses the neuroinflammation signalling pathway and decreased phosphorylated tau protein formation induced by LPS in a dose-dependent manner. Minocycline has been recommended can be used as a preventative and therapeutic drug for neuroinflammatory diseases such as AD.
... Several studies have demonstrated the abilities of honey to promote cognitive functions in the inflammatory-induced model due to the presence of various compounds (Arshad et al., 2020;Yaacob et al., 2020;Adeniyi et al., 2022) Honey can improves anxiety, cognitive behavioral and locomotor activity dysfunctionalities in lipopolysaccharide-induced neuroinflammation model (Adeniyi et al., 2022). SBH and its phenolic content prevent neuroinflammation, act against neuronal damage caused by neurotoxins, and induce memory, learning, and cognitive function in the brain (Rao et al., 2016;Meo et al., 2017;Yaacob et al., 2018). ...
... Moreover, the high antioxidant activity of quercetin, a flavonoid compound in SBH, promotes memory performance and synaptic plasticity of hippocampal neuronal cells in the chronic lead exposure experimental model (Mijanur Rahman et al., 2014). Kelulut honey can improve spatial memory impairment in the LPS-induced rats model and anxiety in the metabolic syndrome rat model (Arshad et al., 2020;Yaacob et al., 2020). ...
Article
Full-text available
Tropical Meliponini bees produce stingless bee honey (SBH). Studies have shown beneficial properties, including antibacterial, bacteriostatic, anti-inflammatory, neurotherapeutic, neuroprotective, wound, and sunburn healing capabilities. High phenolic acid and flavonoid concentrations offer SBH its benefits. SBH can include flavonoids, phenolic acids, ascorbic acid, tocopherol, organic acids, amino acids, and protein, depending on its botanical and geographic origins. Ursolic acid, p-coumaric acid, and gallic acid may diminish apoptotic signals in neuronal cells, such as nuclear morphological alterations and DNA fragmentation. Antioxidant activity minimizes reactive oxygen species (ROS) formation and lowers oxidative stress, inhibiting inflammation by decreasing enzymes generated during inflammation. Flavonoids in honey reduce neuroinflammation by decreasing proinflammatory cytokine and free radical production. Phytochemical components in honey, such as luteolin and phenylalanine, may aid neurological problems. A dietary amino acid, phenylalanine, may improve memory by functioning on brain-derived neurotrophic factor (BDNF) pathways. Neurotrophin BDNF binds to its major receptor, TrkB, and stimulates downstream signaling cascades, which are crucial for neurogenesis and synaptic plasticity. Through BDNF, SBH can stimulate synaptic plasticity and synaptogenesis, promoting learning and memory. Moreover, BDNF contributes to the adult brain’s lasting structural and functional changes during limbic epileptogenesis by acting through the cognate receptor tyrosine receptor kinase B (TrkB). Given the higher antioxidants activity of SBH than the Apis sp. honey, it may be more therapeutically helpful. There is minimal research on SBH’s neuroprotective effects, and the related pathways contribute to it is unclear. More research is needed to elucidate the underlying molecular process of SBH on BDNF/TrkB pathways in producing neuroprotective effects.
... In addition, from the metabolic syndrome rats' elucidation, honey can normalize blood sugar and lower serum triglyceride and LDL levels, and behavioral tests support its effects on anxiety and memory (Arshad et al., 2020). Yaacob et al. (2020) found that the lipopolysaccharide (LPS) rat model greatly reduces spatial and recognition memory through honey consumption. Honey and its methanolic fraction has have potential therapeutic and preventative benefits for neuroinflammatory and neurodegenerative disorders (Yaacob et al., 2020). ...
... Yaacob et al. (2020) found that the lipopolysaccharide (LPS) rat model greatly reduces spatial and recognition memory through honey consumption. Honey and its methanolic fraction has have potential therapeutic and preventative benefits for neuroinflammatory and neurodegenerative disorders (Yaacob et al., 2020). ...
Article
Full-text available
Since ancient times, honey has been employed in many aspects of everyday life, the most popular of which is as a natural sweetener. Honey is used not only as a nutritional product but also in health as a supplement and in various applications, especially related to brain booster health. Brain health is the capacity to carry out all mental functions necessary for cognition, such as learning and judging, utilizing language, and recalling. This review presents the current trend of research on honey, particularly the interest in underlying mechanisms related to brain booster health. A total of 34 original articles addressing brain health from the consumption of honey were analyzed. We identified four main brain health benefits, which are memory booster, neuroprotective effect, anti-stress, and anti-nociceptive potentials with the proposed underlying mechanism. A lot of attention has been paid to the role that honey plays in brain health research, with the goal of examining the link between honey and brain health as well as the mechanism underlying it, the findings from this review may be potentially beneficial to develop new therapeutic roles for honey to help determine the best and most promising to benefit and boost overall brain health.
... Experimental design "The rats were separated into five groups (n=10 rats/ group): (i) control, (ii) LPS-treated with distilled water, (iii) LPS-treated with minocycline 25 mg/kg (11), (iv) LPS-treated with minocycline 50 mg/kg (10) and (v) LPStreated with memantine 10 mg/kg" (11 p. 334). The single dose (5 mg/kg) of intraperitoneal LPS (Sigma-Aldrich, St. Louis, MO. 297-473-0) injections was given on day 5 of the experiment (11). Minocycline and memantine (USP, Rockville, MD) were given intraperitoneally once daily for 14 days to the rats in the minocycline and memantine groups. ...
Article
Full-text available
Introduction: Minocycline has been demonstrated to have potent effects on neurologic structures and functions in several animal models. However, its neuroprotective properties following a single injection of lipopolysaccharide (LPS) in an adult rat model have not been clearly elucidated. This study investigated minocycline’s neuroprotective effects in the LPS-induced neuroinflammation rat model. Methods: Fifty adult male Sprague Dawley rats were split into five groups at random: (i) control, (ii) distilled water-treated LPS, (iii) 25 mg/kg minocycline-treated LPS, (iv) 50 mg/kg minocycline-treated LPS, and (v) 10 mg/kg memantine-treated LPS. On day 5, LPS (5 mg/kg) was given intraperitoneally once, whereas minocycline and memantine were given once daily for 14 days. Results: LPS was found to significantly induce β-amyloid peptide deposition and neuronal damage, and impair recognition memory, while administration of minocycline dose-dependently reversed these effects. These data suggest that LPS-induced recognition memory impairment by inducing β-amyloid peptide deposition and neuronal damage in the cortical and hippocampal areas. Furthermore, we compared minocycline with memantine administration, and these data suggested better effects in minocycline (50 mg/kg) and comparable effects between minocycline (25 mg/kg) and memantine (10 mg/kg) treatments in reducing β-amyloid peptide deposition, neuronal damage and recognition memory impairment induced by LPS. Conclusion: Minocycline may be a strong contender as an effective preventive-therapeutic drug for neuroinflammatory diseases such as Alzheimer’s disease (AD) based on these findings.
... Acetylcholinesterase (AChE) inhibitors (prevent the degradation of Ach and enhance ACh levels) including donepezil, rivastigmine, and galantamine cause nausea, anorexia, vomiting, and diarrhea. [5][6][7]. The glutamate antagonist memantine is another FDA-approved anti-Alzheimer medication that acts as an antagonist of nicotinic receptors (nAChRs) non-competitively that produces fast desensitization of nAChR activity [8] and lowers neuronal excitotoxicity [9]. ...
... The number of Animal Ethics Minocycline (USP, 12601Twinbrook Pkwy, Rockville, MD) and memantine (USP, 12601Twinbrook Pkwy, Rockville, MD) were administered intraperitoneally once daily for 2 weeks consequently, starting from day 1 to day14. LPS was obtained from E.coli 0111:B4 (Sigma-Aldrich, St. Louis, MO) and injected intraperitoneally once on day 5 at the dose of 5 mg/kg (Yaacob et al., 2018). After 2 weeks of treatment, all rats were subjected to OFT from day 15 to day 18. ...
Preprint
Full-text available
Introduction: Neuroinflammation following lipopolysaccharide (LPS) administration induces locomotor deficit and anxiety-like behavior. In this study, minocycline was compared to memantine, the NMDA receptor antagonist, for its effects on LPS-induced locomotor deficit and anxiety-like behavior in rats. Methodology: Adult male Sprague Dawley rats were administered either two different doses of minocycline (25 or 50 mg/kg/day, i.p.) or 10 mg/kg/day of memantine (i.p.) for 14 days four days prior to LPS (5 mg/kg, i.p.) injection. The locomotor activity and anxiety-like behavior were assessed using the open field test (OFT). The phosphorylated tau protein level was measured using ELISA while the expression and density of brain-derived neurotrophic factor (BDNF) and cAMP response element-binding protein (CREB) protein in the medial prefrontal cortex (mPFC) were measured using immunohistochemistry and western blot, respectively. Results: In the mPFC, minocycline treatment reduced the locomotor deficit and anxiety-like behavior, reduced phosphorylated tau protein level, and upregulated BDNF/CREB protein expression comparable to memantine, with the higher dose of minocycline having better benefits. Conclusion: Minocycline treatment attenuated LPS-induced locomotor deficit and anxiety-like behavior in rats, possibly via a decrease in phosphorylated tau protein levels and an increase in the expression of the BDNF/CREB proteins.
... Experimental design "The rats were separated into five groups (n=10 rats/ group): (i) control, (ii) LPS-treated with distilled water, (iii) LPS-treated with minocycline 25 mg/kg (11), (iv) LPS-treated with minocycline 50 mg/kg (10) and (v) LPStreated with memantine 10 mg/kg" (11 p. 334). The single dose (5 mg/kg) of intraperitoneal LPS (Sigma-Aldrich, St. Louis, MO. 297-473-0) injections was given on day 5 of the experiment (11). Minocycline and memantine (USP, Rockville, MD) were given intraperitoneally once daily for 14 days to the rats in the minocycline and memantine groups. ...
Article
Full-text available
Introduction: Minocycline has been demonstrated to have potent effects on neurologic structures and functions in several animal models. However, its neuroprotective properties following a single injection of lipopolysaccharide (LPS) in an adult rat model have not been clearly elucidated. This study investigated minocycline's neuroprotective effects in the LPS-induced neuroinflammation rat model. Methods: Fifty adult male Sprague Dawley rats were split into five groups at random: (i) control, (ii) distilled water-treated LPS, (iii) 25 mg/kg minocycline-treated LPS, (iv) 50 mg/kg minocycline-treated LPS, and (v) 10 mg/kg memantine-treated LPS. On day 5, LPS (5 mg/kg) was given intra-peritoneally once, whereas minocycline and memantine were given once daily for 14 days. Results: LPS was found to significantly induce β-amyloid peptide deposition and neuronal damage, and impair recognition memory, while administration of minocycline dose-dependently reversed these effects. These data suggest that LPS-induced recognition memory impairment by inducing β-amyloid peptide deposition and neuronal damage in the cortical and hip-pocampal areas. Furthermore, we compared minocycline with memantine administration, and these data suggested better effects in minocycline (50 mg/kg) and comparable effects between minocycline (25 mg/kg) and memantine (10 mg/kg) treatments in reducing β-amyloid peptide deposition, neuronal damage and recognition memory impairment induced by LPS. Conclusion: Minocycline may be a strong contender as an effective preventive-therapeutic drug for neuroinflammatory diseases such as Alzheimer's disease (AD) based on these findings.
Article
Introduction: The neuroinflammatory response was seen to impact the formation of phosphorylated tau protein in Alzheimer's disease (AD). This study aims to investigate the molecular mechanism of minocycline in reducing phosphorylated tau protein formation in the hippocampus of lipopolysaccharide (LPS)-induced rats. Methods: Fifty adult male Sprague Dawley (SD) rats were randomly allocated to 1 of 5 groups: control, LPS (5 mg/kg), LPS + minocycline (25 mg/kg), LPS + minocycline (50 mg/kg) and LPS + memantine (10 mg/kg). Minocycline and memantine were administered intraperitoneally (i.p) for two weeks, and LPS was injected i.p. once on day 5. ELISA was used to determine the level of phosphorylated tau protein in SD rats' hippocampal tissue. The density and expression of Toll-like receptor-4 (TLR-4), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-кβ), tumour necrosis factor-alpha (TNF-α), and cyclooxygenase (COX)-2 were determined using Western blot and immunohistochemistry. Results: Minocycline, like memantine, prevented LPS-induced increasein phosphorylated tau protein level suggested via reduced density and expression of TLR-4, NF-кβ, TNF-αand COX-2 proteins in rat hippocampal tissue. Interestingly, higher doses were shown to be more neuroprotective than lower doses. Conclusion: This study suggests that minocycline suppresses the neuroinflammation signalling pathway and decreased phosphorylated tau protein formation induced by LPS in a dose-dependent manner. Minocycline can be used as a preventative and therapeutic drug for neuroinflammatory diseases such as AD.
Preprint
Full-text available
Introduction Minocycline has been showed can ameliorates neuroinflammation that was encountered in many neurodegenerative diseases. This study aims to investigate the expression of inflammatory mediators in the rat medial prefrontal cortex (mPFC) after minocycline treatment in lipopolysaccharide (LPS) induced neuroinflammation rat model. Methods Adult male Sprague Dawley (SD) rats (N = 50) were divided into 5 groups: 1) control, 2) LPS (5 mg/kg), 3) LPS + minocycline (25 mg/kg), 4) LPS + minocycline (50 mg/kg) and 5) LPS + memantine (10 mg/kg). Intraperitoneal minocycline and memantine were given daily for 14 days, while LPS injection was given once on 5th day. Western blot and immunohistochemistry were used to assess density and expression of TLR-4, nuclear factor kappa-B (NF-kB), tumour necrosis factor (TNF)-α and cyclooxygenase (COX)-2 in the medial prefrontal cortex (mPFC) of rats. Results Findings displayed that minocycline significantly decreased expression and density of TLR-4, NF-kB, TNF-α and COX-2 proteins that were comparable to memantine in mPFC of SD rat injected with single intraperitoneal LPS. Interestingly, the anti-inflammatory effects of minocycline 50 mg/kg were significantly more than minocycline 25 mg/kg. Conclusion This study suggested that minocycline can modulated LPS-induced neuroinflammation in dose-dependent manner in the mPFC area. Thus, it is suggested that minocycline can be used as potential preventive-therapeutic drug for neuroinflammatory diseases such as depression and anxiety.
Conference Paper
Full-text available
Oxidative stress is a common pathogenic factor in neurodegenerative diseases. The present study was designed to investigate the protective effects of tualang honey (TH) and its methanolic fraction (MTH) against lipopolysaccharide (LPS)-induced oxidative stress in rats’ hippocampus. Thirty five male Sprague Dawley rats were divided into five groups of seven individuals (n = 7): (i) control rats, (ii) untreated LPS rats (5 mg/kg) (iii) LPS rats treated with TH 200 mg/kg, (iv) LPS rats treated with MTH 150 mg/kg and (v) LPS rats treated with memantine 10 mg/kg. All treatments were administered intraperitoneally once daily for 14 days. The rats were sacrificed and the hippocampus was carefully dissected out, homogenized and stored at -80C. Catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GPr), superoxide dismutase (SOD) and malondialdehyde (MDA) levels/activities were measured by using ELISA kits. Treatment with TH and MTH markedly reduced oxidative damage in rats’ hippocampus as evidence by restoration in the activities of antioxidant enzymes (CAT, GPx and GPr) and decreased in MDA level comparable to LPS rats treated with memantine (a control drug). However, no significant differences were detected in SOD level in all groups. As a conclusion, TH and MTH exert their protective effects against LPS-induced oxidative damage in rats’ hippocampus comparable to memantine.
Article
Full-text available
Alzheimer's disease (AD) is a primary cause of dementia in the middle-aged and elderly worldwide. Animal models for AD are widely used to study the disease mechanisms as well as to test potential therapeutic agents for disease modification. Among the non-genetically manipulated neuroinflammation models for AD, lipopolysaccharide (LPS)-induced animal model is commonly used. This review paper aims to discuss the possible factors that influence rats' response following LPS injection. Factors such as dose of LPS, route of administration, nature and duration of exposure as well as age and gender of animal used should be taken into account when designing a study using LPS-induced memory impairment as model for AD.
Article
Full-text available
Background Considerable evidence has shown that neuroinflammation and oxidative stress play an important role in the pathophysiology of postoperative cognitive dysfunction (POCD) and other progressive neurodegenerative disorders. Increasing evidence suggests that acetaminophen (APAP) has unappreciated antioxidant and anti-inflammatory properties. However, the impact of APAP on the cognitive sequelae of inflammatory and oxidative stress is unknown. The objective of this study is to explore whether APAP could have neuroprotective effects on lipopolysaccharide (LPS)-induced cognitive impairment in mice. Methods A mouse model of LPS-induced cognitive impairment was established to evaluate the neuroprotective effects of APAP against LPS-induced cognitive impairment. Adult C57BL/6 mice were treated with APAP half an hour prior to intracerebroventricular microinjection of LPS and every day thereafter, until the end of the study period. The Morris water maze was used to assess cognitive function from postinjection days 1 to 3. Animal behavioural tests as well as pathological and biochemical assays were performed to evaluate LPS-induced hippocampal damage and the neuroprotective effect of APAP. Results Mice treated with LPS exhibited impaired performance in the Morris water maze without changing spontaneous locomotor activity, which was ameliorated by treatment with APAP. APAP suppressed the accumulation of pro-inflammatory cytokines and microglial activation induced by LPS in the hippocampus. In addition, APAP increased SOD activity, reduced MDA levels, modulated glycogen synthase kinase 3β (GSK3β) activity and elevated brain-derived neurotrophic factor (BDNF) expression in the hippocampus. Moreover, APAP significantly decreased the Bax/Bcl-2 ratio and neuron apoptosis in the hippocampus of LPS-treated mice. Conclusions Our results suggest that APAP may possess a neuroprotective effect against LPS-induced cognitive impairment and inflammatory and oxidative stress via mechanisms involving its antioxidant and anti-inflammatory properties, as well as its ability to inhibit the mitochondrial permeability transition (MPT) pore and the subsequent apoptotic pathway.
Article
Full-text available
Honey has shown potential therapeutic properties in traditional medicine. This study investigated the cognition enhancing potential of Tualang honey in chronic cerebral hypoperfusion- induced neurodegeneration by permanent bilateral common carotid arteries ligation (2 vessels occlusion- 2VO) in rats. Previous studies have shown that rats subjected to 2VO (2 vessel occlusion) experienced cognitive deficits, oxidative stress and neuronal injury in the hippocampus of the brain. We studied the effect of Tualang honey on learning and memory using the Morris water maze (MWM) in rats. Rats were randomly divided into four groups (n=10); Sham control, untreated 2VO (2VO), honey treated 2VO (2VO+H) and honey treated group (H). Each group was again divided into two subgroups (n=5): one for long term memory assessment and another for short term memory and relearning tasks. After 10 weeks of treatment, all rat groups were tested for cognitive assessment by MWM (Morris water maze). 2VO+H rats had better spatial learning and memory performance than untreated 2VO rats in MWM tasks (p 0.05). This study shows that Malaysian Tualang honey might have therapeutic potential for the treatment of chronic cerebral hypoperfusion related neurodegenerative disorders such as Alzheimer’s disease (AD) in which cognitive impairment is profound.
Article
Full-text available
Activated microglial cells play an important role in immune and inflammatory responses in central nervous system and neurodegenerative diseases. Many pro-apoptotic pathways are mediated by signaling molecules that are produced during neuroinflammation. In glial cells, NF-κB, a transcription factor, initiates and regulates the expression of several inflammatory processes during inflammation which are attributed to the pathology of the several neurodegenerative diseases. In this review, we discuss the most important neuroinflammatory mediators with their pathways. Attenuating cytokines production and controlling microglial inflammatory response, which are the result of understanding neuroinflmmation pathways, are considered therapeutic strategies for treating neurodegenerative diseases with an inflammatory component.
Article
Full-text available
Inflammation is a biological response and a defence mechanism by the body to remove harmful stimuli followed by the healing process. Healing is a process of skin and other soft tissue repair at the site of injury. However, uncontrolled inflammation could lead to serious illnesses such as cancer and cardiovascular diseases, which cause great impact on public health and economy. This necessitates supplementation with anti-inflammatory properties to prevent or remove unnecessary inflammation and damage. For ages, Tualang honey (TH) has been used as a natural remedy for inflammation and wounds. TH also exhibits antioxidant, antibacterial and reproductive properties. This review collates the various studies on anti-inflammatory and wound-healing properties of TH. It also presents findings that indicate that honey may ameliorate ultraviolet-induced inflammation of the skin, chemical-induced inflammation of the eyes and oxidative stress on the eyes. Besides, wound-healing properties have also been highlighted here. These data suggest that TH might be a therapeutic agent in the management of inflammation and wound healing. However, there is a need to study the underlying mechanism of action of TH in vitro and in vivo, to develop a better understanding of its potential benefits.
Article
As a natural anti-oxidant source, Tualang honey, produced by wild bees nesting on the Tualang tree (Koompassia excelsa) is expected to have positive influence on health, including memory. This study investigated the effect of Tualang honey on the cell count of memory formation related hippocampal pyramidal neuron and on spatial memory performance (SMP) of rats using the radial arm maze (RAM). Sprague Dawley male rats (n=24), 7-8 weeks old were divided into two groups; experimental group group force-fed 1 mL/100 g body weight with 70% honey (HG); and the control group with 0.9% saline (CG) for 12 weeks. Nissl staining technique (with cresyl violet) was employed for neurohistological analysis of the hippocampal tissue. Six randomly selected rats from each group were used for the neuronal soma counting of pyramidal cell layer CA1, CA3a and CA3c regions. Two-way ANOVA analysis showed positively significant differences between treatment and control groups for SMP comparison of working memory and reference memory components, as well as the number of pyramidal neurons. Hence, this positive effects of Tualang honey, as demonstrated behaviorally and neurohistologically, supported report that Tualang honey could improve memory and deter hippocampal morphological impairments; possibly due to its high anti-oxidant properties.
Article
The effects of prenatal stress on immunocytochemical reaction on serotonin (5-HT) in the dorsal raphe nucleus (DRN) of the brainstem were investigated in 7-day-old male rat pups exposed to impact of pain in the formalin test (a control is an injection of saline). A strengthening effect of prenatal stress on pain behavior was revealed in animals in the formalin test. Prenatal stress decreased the number of 5-HT-immunoreactive neurons as compared with that in prenatally non-stressed animals in both pups with inflammatory pain and the controls. There were no differences in the number of 5-HT-immunoreactive cells between pups with inflammatory pain and saline in both prenatally non-stressed animals and prenatally stressed ones. The data suggest that stress of maternal separation during the experiment hides the difference in immunocytochemical result in the rat pups with inflammatory pain and in the control. Thus, it was demonstrated for the first time that in 7-day-old rat pups the 5-HT-ergic neurons in DRN of the brainstem are a target of the prenatal stress.
Article
Prenatal exposure to lipopolysaccharide (LPS) has been exploited to simulate brain disorder in animal model. Prenatal LPS-exposure has shown elevated levels of pro-inflammatory cytokines in the early stages of the postnatal period. This study determines the effect of prenatal LPS-exposure on oxidative stress (OS) in the distinct brain regions in the early postnatal stages. LPS (50 μg/kg, i.p.) and water for injection (100 μl, i.p.) were given to the experimental (n = 5) and control (n = 5) group of pregnant Swiss albino mice respectively on gestational day (GD)-16 and 17. Animals were decapitated on postnatal day (PnD) – 1, 7, 14 and 21 to assay levels of oxidative markers from 6 distinct brain regions. When compared with the control, prenatal LPS-exposure alters levels of OS markers: (i) on PnD-1, glutathione (GSH) level is raised and superoxide dismutase (SOD) activity is dropped, (ii) on PnD-7, advanced oxidation of protein product (AOPP) level is elevated, (iii) on PnD-14, lipid peroxidation (MDA) and activity of catalase (CAT) are enhanced, (iv) on PnD-21, increased MDA continued. The hippocampus (HC) and cerebellum (CB) were mostly susceptible to OS in the early postnatal development. Levels of MDA and activity of CAT enzyme were increased on PnD-14 in the cortex, HC and CB. Except MDA, all OS markers recovered and returned to the level of control animals on PnD-21. Taken together, these results suggest that prenatal LPS-exposure induces age- and region-specific OS in the early postnatal stage.
Article
Introduction: Hyperalgesia is considered as one of the marked signs of subchronic diabetes mellitus in patients that could affect their lifestyle. This study was designed to investigate the anti-nociceptive effect of chronic administration of quercetin in streptozotocin (STZ)-diabetic rats using formalin and hot tail immersion tests. Methods: Rats were divided into control, control or diabetic groups receiving sodium salicylate, untreated diabetic, and quercetin-treated control and diabetic groups. The treatment groups received i.p. administration of quercetin at a dose of 10 mg/kg for 6 weeks. Finally, hyperalgesia were assessed using standard formalin and hot tail immersion tests. Meanwhile, some markers of oxidative stress were also measured in brain tissue. Results: Quercetin or SS treatment of diabetic rats significantly reduced pain score in chronic phase of formalin test (p<0.05). Regarding hot tail immersion test, diabetic rats showed a significant reduction in tail flick latency as compared to control ones (p<0.05) and quercetin treatment of diabetic rats did significantly increase this latency relative to untreated diabetics (p<0.05). Quercetin treatment of diabetic rats also significantly decreased brain level of malondialdehyde (MDA) (p<0.05) and nitrite (p<0.05) and slightly increased activity of superoxide dismutase (SOD) relative to diabetics. Discussion: Taken together, chronic administration of quercetin could attenuate nociceptive score in chronic phase of formalin test in streptozotocin-diabetic rats and could also increase threshold of thermal nociception.