Skip to main content
  • Article
  • Published:

Acer okamotoanum improves cognition and memory function in Aβ25–35-induced Alzheimer’s mice model

Abstract

We studied the effect of ethyl acetate (EA) fraction from Acer okamotoanum on cognitive improvement and protective abilities in amyloid beta (Aβ)25–35 peptide-injected Alzheimer’s disease (AD) mice. EA was oral administration at 100 and 200 mg/kg/day during the 14 days. We studied the protective effect of EA against AD on the basis of behavioral tests including T-maze test, Novel object recognition test, and Morris water maze test. Control group injected with Aβ25–35 showed significant impairments in memory function. But the oral administration of EA (EA 100 and EA 200 groups) improved the cognition and memory function. In addition, EA against Aβ25–35 peptide has been shown to inhibit lipid peroxidation levels and nitric oxide production in tissues. Acetylcholinesterase (AChE) was elevated in the brain by Aβ25–35 peptide, whereas administration of EA (EA 100 and EA 200 groups) significantly decreased AChE level. Our results indicated that EA improves learning and long-term memory against Aβ25–35 peptide–caused deficit through attenuation of oxidative stress.

Introduction

Alzheimer disease (AD) is the major familiar chronic neurodegenerative disease that progresses with age. The characteristics of the brain of an AD patient brain include amyloid-β peptide (Aβ) accumulation, mitochondrial deficits, neurofibrillary tangles, synapse depletion, and oxidative stress (Pi et al. 2012), resulting in progressive memory loss and cognitive impairment (Paban et al. 2014). In particular, Aβ accumulation and oxidative stress are determining factors of neurodegeneration in AD. Neurodegeneration caused by Aβ leads to cognitive impairment, neuro-inflammation (Bicca et al. 2015), and neuronal apoptosis (Park et al. 2009). Aβ originates from proteolytic cleavage of amyloid precursor protein (APP) by β- and γ-secretases via amyloidogenic pathway, and it accumulates in AD brains owing to excessive production and impaired clearance. The Aβ accumulation leads to neurodegeneration and apoptosis, which impair synaptic function and long-term potentiation (Esmaeili et al. 2015). The hydrolysis of acetylcholine by acetylcholinesterase (AChE) occurs within and around the amyloid peptides in the brain of AD patients. AChE expression promotes the Aβ fibril formation and leads to Aβ toxicity (Kwon et al. 2011).

The genus Acer is divided into 15 types in Korea and includes typical maple trees widely distributed in the mountainous regions (Kim et al. 1998). In particular, Acer okamotoanum (A. okamotoanum), which is indigenous to Korea and widely distributed in Ulleung Island was reported to have health benefits (Yim et al. 1981). A. okamotoanum is rich in flavonoids and phenolic compounds, such as quercetin, kaempferol, and gallic acid (Qadir et al. 2007; Jeong et al. 2009). Several studies demonstrated that the leaf, branch, and root of A. okamotoanum have useful biological properties, such as anti-HIV-1 integrase, anti-cancer, anti-oxidant, skin whitening, and anti-herpetic effects (Jeong et al. 2010; Woo et al. 1997). In addition, A. okamotoanum was reported to affect alcohol detoxification (Yoo et al. 2011) through reduction decline of the levels of ethanol and acetaldehyde in the blood. However, the effects of the A. okamotoanum on cognitive impairment associated with AD were not yet studied.

Therefore, we investigated the influence of A. okamotoanum on cognitive impairment and neuroprotective function in ICR mice model induced by intracerebroventricular injection of Aβ25–35 peptides.

Materials and methods

Plant materials

Acer okamotoanum was obtained from Ulleung Island by Korea National Arboretum and deposited at the Herbarium of our department.

Drugs

25–35, malondialdehyde (MDA), acetylcholine iodide, and 5,5′-dithiobis (2-nitrobenzoic acid) (DTNB) were purchased from Sigma Aldrich (Saint Louis, Missouri, USA). Dimethyl sulfoxide and sodium chloride (NaCl) were obtained from Bio Basics Inc. (Ontario, Canada). Thiobarbituric acid (TBA) was acquired from Lancaster Synthesis (Ward Hill, USA).

Arrangement of the ethyl acetate (EtOAc) fraction from A. okamotoanum (EA)

Freeze-dried A. okamotoanum was extracted with methanol under reflux and repeated 5 times at 3-h intervals. The extract was concentrated using a rotary evaporator and suspended in distilled water (DW). Extract was suspended in DW and partitioned with EtOAc.

Mice model and experimental design

The male 5-week-old ICR mice (weighing 24.0–26.0 g) were gained from the Orient Inc. (Seongnam, Korea). The experimental models were housed 4 per plastic cages fed with a standard diet and free drinking edible water. Also, they were sustained under the constant temperature (20 ± 2 °C), 12-h light–dark illumination period. The mice management was conducted in accordance with the animal care and use guidelines (approval No. PNU-2014-0056) approved by the Pusan National University-Institutional Animal Care and Use Committee (PNU-IACUC). The experimental models were separated into 4 groups, consisting of six individuals each: normal group = 0.9% NaCl-lesioned + intraorally water; control group = Aβ25–35-lesioned + intraorally water; EA 100 group = Aβ25–35-lesioned + intraorally EA (100 mg/kg/day); EA 200 group = Aβ25–35-lesioned + intraorally EA (200 mg/kg/day) during the 14 days using a zonde. As shown in Fig. 1, the behavioral experiments design.

Fig. 1
figure 1

Experimental schedule of behavioral tests for mice injected with Aβ25–35

Induction of AD by Aβ25–35-injection to brain of ICR mice

25–35 peptide was solubilized in saline solution to a final concentration of 1 mg/ml. The solution was incubated for 3 days (37 °C) to obtain the aggregated form (Maurice et al. 1996). The aggregated Aβ25–35 peptide was injected into the experimental models hippocampus in accordance with the process constituted by Laursen and Belknap (1986). Experimental models in the control, EA 100 and 200 groups were anesthetized with CO2 and injected with aggregated 5 nmol of Aβ25–35 peptide (5 μl) into the left lateral ventricle at a rate of 1 μl/min. Normal group was injected with saline solution (0.9% NaCl) in the same area. All the injections were performed using a 10-μl Hamilton syringe (26-gauge needle, 2.2 mm depth).

Morris water maze test

Classical water maze test procedure was based on a method described by Morris (1984). Water maze apparatus that was composed of circular water tank pool (diameter, 80 cm; height, 50 cm) was randomly divided into quadrants. An escape platform (8 cm in diameter) was submerged 1 cm underneath the water surface in the middle of 1 quadrant. The water temperature was maintained at 22 ± 1 °C, and then white poster color added to water in order that make hidden the platform. The platform position was not changed during the training period. 4 stickers on the walls of the apparatus provided visual cues for navigation. The experimental models were instructed to locate the obscure platform in three times per day for consecutive 3 days. In every trial, the experimental models were released in the water pool facing a different sticker and permitted to swim within 60 s. After successfully reaching the platform, they were allowed to remain on the platform for 15 s. If the mice failed to find the platform, the experimenter released the mice on the platform and permitted to stay on it for 15 s. The memory retention session (probe trial test) was conducted after 3 days of training. In the primary test on day 4, the experiment was carried out in a manner as described based on Morris (1984). In the secondary test, the platform was removed and then the mice were placed in the water and allowed to swim within 60 s to look for the platform. The time spent in the target quadrant was set down. In the tertiary test, time the mice spent to reach the platform in transparent water was counted. The percentage of occupancy of the target quadrant was calculated as the percentage of time spent in the target quadrant during a 60 s trial.

Novel object recognition test

This test was conducted according to the process constituted by Bevins and Besheer (2006), in a rectangular black open box (40 cm × 30 cm × 20 cm). During first session (training session), two identical objects (plastic models) were placed at the opposite sides in the box. The mice were released at the center of the box and permitted to explore liberally during the 10 min. In this session, the number of touch times for each object was set down. The experimental models were released again into the same field after 24 h. However, in this session (test session), one of the original objects was changed with a novel (another plastic model). In test session, mice were permitted to search and explore freely during the 10 min, and all trials in the object recognition test were recorded.

T-maze test

This test was based on the process described by Montgomery (1952). T-maze apparatus was composed of a stem and two arms made of black boards (length of start and goal stems, 50 cm; width, 13 cm; height of walls, 20 cm). The apparatus composed of a start position, left route, and right route with a black door to divide the routes from one another. Experimental models were released in the start position, and the number of total entry times in the right route of the T-maze was measured for each mouse for 10 min. Experimental models were released back into the same apparatus for 24 h after the training session. The mice were permitted to explore freely through the maze within 10 min, and the number of total entry times was measured (test session). The percentage of space perceptive alternations was calculated as the ratio (%) of the number of entries into the left or right route to the number of total route entries, multiplied by 100.

Determination of lipid peroxidation

The lipid peroxidation levels in organs were conducted by the method of Ohkawa et al. (1979). After the finish of behavioral experiments, experimental models were anesthetized using CO2 gas. The brains, livers, and kidneys of the mice were directly isolated and placed on ice. Followed by dissection, these tissues were homogenized. The obtained homogenate was blended with 1% phosphoric acid and 0.67% TBA solution. After boiling for 45 min, the mixture was cooled on the ice and 2 ml of 1-butanol was mixed prior to centrifugation at 13,000×g for 10 min (4 °C), absorbance was measured at 540 nm. The lipid peroxidation levels were determined and calculated using the standard curve of MDA content.

Estimation of reduced nitric oxide (NO)

Reduced NO level in brain, liver and kidney were assayed by Schmidt et al. (1992). The 150 μl of supernatant from the lipid peroxidation process was blended with 130 μl of DW, and then mixed solvent 20 μl was blended to the identical amount of phosphoric acid and 0.1% N-(1-naphthyl)ethylenediamine dihydrochloride solvent and the absorbance of the mixture was assayed at 540 nm. The manufacture of NO level was calculated with the standard curve of sodium nitrite content.

AChE inhibitory activity assay

AChE activity assay was performed as described by Ellman et al. (1961). The reaction solution included 4 ml of (0.1 M) phosphate buffer saline, 100 μl of DTNB, and 200 μl of acetylthiocholine iodide mixture, which were blended and incubated during the 30 min (25 °C). After 5 min, samples (400 μl) were added to the reaction mixtures and then the absorbance was measured at 412 nm.

Statistical analysis

Our data are expressed as mean ± SD (n = 6). All statistical significances were performed using a one-way ANOVA, followed by Duncan’s post hoc tests. Comparisons with P < 0.05 were considered statistical significance.

Results

Body weight change

This study did not show any dramatic difference in the body weights among these four groups, normal, control, EA 100, and EA 200, during experimental periods (data not shown).

Morris water maze test

The influence to EA on spatial memory was examined in the water maze. In the training session, normal group immediately learned the hidden platform location by swimming across the water tank pool and promptly reached the hidden platform. EA 100 and EA 200 groups reached the platform on a period of 3 days with less time. In the final test, the groups administered with EA 100 and EA 200 significantly reduced the time to reach platform. The EA 200 group dramatically reduced time to reach the platform from 58.5 to 18.0 s (Fig. 2). When the platform was removed, the staying time in the target quadrant was shorter in Aβ25–35-injected control group than normal group (Fig. 3). However, of the EA administration increased the staying time in the target quadrant. It indicates that the EA administration protected against the deficit of memory role induced by Aβ25–35. In addition, Fig. 4(A) and (B) indicated that EA administration did not affect the visual and exercise ability. There were no significance differences among all of groups in latency to reach the exposed platform (Fig. 4(A)). On the other hand, EA-treated groups took shorter to find the platform than the control groups, when the platform was hidden (Fig. 4(B)).

Fig. 2
figure 2

Effect of EA on spatial learning in the Morris water maze test. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–cThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test

Fig. 3
figure 3

Effect of EA on memory impairment induced by Aβ25–35 injection in the Morris water maze test. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–bThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test

Fig. 4
figure 4

Effects of EA on the performance of Aβ25–35-treated mice in finding the hidden (A) and exposed (B) platforms in the Morris water maze test. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–cThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test. NS No significant

Novel object recognition test

The effect of EA on novel object recognition impairment was shown in Fig. 5. There was no significant difference in exploratory preference among the groups in the training session. After 24 h of training session, cognitive ability of the normal group toward the novel object was significantly higher than familiar object recognition, whereas the Aβ25–35-injected control group did not show significant difference on object recognition ability between familiar and novel object. However, the EA 100 and EA 200 groups increased novel object cognitive ability to 60.7 and 65.1%, respectively. It indicated that administration of EA from A. okamotoanum evidently improved object recognition ability.

Fig. 5
figure 5

Effect of EA on objective recognition test. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–dThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test. * The object cognitive abilities for familiar and novel objects are significantly different as determined by Student’ s t test (p < 0.05)

T-maze test

Influence of EA on short-term memory was examined using the T-maze test (Fig. 6). Aβ25–35-injected control group did not show the spatial perception toward the novel route. However, EA 100- and EA 200-treated groups showed a higher preference for the novel route to 61.4 and 61.1%, respectively. It indicated that oral administration of EA from A. okamotoanum enhanced the ability to spatial perception toward novel route.

Fig. 6
figure 6

Spatial alternation test in the T-maze test. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–bThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test. * The object cognitive abilities for familiar and novel routes are significantly different as determined by Student’ s t test (p < 0.05)

Evaluation of lipid peroxidation

The results of the preventive activity of EA against lipid peroxidation caused by Aβ25–35 are shown in Fig. 7. Aβ25–35-injected control group raised lipid peroxide values in the brain. Compared with the normal group, the MDA value of the control group was raised dramatically by 75.1 nmol/mg protein in the brain. However, the administration of EA 100 and EA 200 groups was 40.0 and 40.2 nmol/mg protein, respectively. Additionally, the MDA values in the kidney of normal and control groups were 63.5 and 87.3 nmol/mg protein, respectively. On the other hand, the levels were reduced 53.0 and 53.1 nmol/mg protein in EA 100 and EA 200 groups, respectively. Moreover, the MDA concentration in the liver increased in control group to 47.7 nmol/mg protein, compared with that of normal group, 19.6 nmol/mg protein. The hepatic MDA levels in the EA 100 and EA 200 groups decreased to 13.9 and 10.9 nmol/mg protein, respectively. These results demonstrated that EA from A. okamotoanum significantly restored the level of MDA in the brains, kidneys, and livers of Aβ25–35-injected experimental models.

Fig. 7
figure 7

Effect of EA administration on lipid peroxidation in mice brain (A), kidney (B), and liver (C). The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–cThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test

NO scavenging activity

Figure 8 shows the effects of EA toward NO formation in the brains, kidneys, and livers of mice-injected Aβ25–35. The value of NO in the brain of the normal group was 13.3 μmol/L/mg protein, while that of the Aβ25–35-injected control group was 22.4 μmol/L/mg protein. However, the brain NO values of the EA 100 and EA 200 groups showed 9.1 and 5.3 μmol/L/mg protein, respectively. NO formation in the kidney of the control group was 43.2 μmol/L/mg protein, but NO levels were decreased to 36.6 and 34.0 μmol/L/mg protein by oral administration of EA 100 and EA 200, respectively. The liver NO formation of the control group was also improved compared to the normal group showing 23.8 and 12.9 μmol/L/mg protein, respectively. However, the EA 100 and EA 200 groups led to the significant decrease in NO levels from 23.8 to 16.8 and 20.9 μmol/L/mg protein, respectively. It was indicated that oral administration of EA 100 and EA 200 significantly inhibited NO formation in the brain, kidney, and liver.

Fig. 8
figure 8

Effect of EA administration on Aβ25–35 induced NO production in mice brain (A), kidney (B), and liver (C). The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–cThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test

AChE activity

As shown in Fig. 9, we investigated the effect of EA from A. okamotoanum on AChE activity in the left brain. Aβ25–35-injected control group demonstrated that dramatic increase in the AChE activity in the left brain compared with normal group. On the other hand, EA 100- and EA 200-oral administered groups significantly inhibited AChE activity compared with the control group in the brain. It indicated that EA from A. okamotoanum has a excellent prevention influence on AChE activity in brain.

Fig. 9
figure 9

Effect of EA administration on AChE inhibitory activity in mice brain. The groups are defined as follows: normal = 0.9% NaCl injection + oral administration of water; control = Aβ25–35 injection + oral administration of water; EA100 = Aβ25–35 injection + oral administration of EA (100 mg/kg/day); EA200 = Aβ25–35 injection + oral administration of EA (200 mg/kg/day). Values are represented as mean ± SD. a–bThe different letters represent significant differences (p < 0.05) by Duncan’s multiple range test

Discussion

AD is the most familiar form of dementia and it is an ascending worldwide public health issue (Brookmeyer and Gray 2000). AD patients were characterized by continuing memory deficit and cognitive impairment, including personality changes, aphasia, and apraxia (Paban et al. 2014). AD neuropathological features include deposits of Aβ peptides, neurofibrillary tangles, neuronal and synaptic degeneration, and neuronal inflammation (Rozemuller et al. 1989). Aβ is created by sequential endoproteolytic cleavage of APP by β- and γ-secretase, and penetrates a dynamic equilibrium among soluble and deposited forms (De Mattos et al. 2002). The most prominent Aβ type in the AD patient brain is Aβ1-40, which can be divided into a more toxic fragment, Aβ25–35 (Kubo et al. 2002). Aβ25–35 has related to induce the hydrogen peroxide production and lipid peroxide in the brain (Yatin et al. 2000). Therefore, Aβ25–35 injection into the intracerebroventricular region of mice causes numerous adverse effects, such as impairment of learning, memory, and cognition. In the current study, Aβ25–35 was injected into the brain of mice to induce AD, and then the neuroprotective activities of EA from A. okamotoanum on learning, memory, and cognitive impairment were evaluated using the Morris water maze, novel object recognition test, and T-maze.

Morris water maze test is a well-established method for the assessment of standard learning and long-term memory (Morris 1984). In the current study, the normal group showed rapid decrease in time to reach the hidden platform by training. However, the control group injected with Aβ25–35 did not decrease the latency time to reach the platform. It indicated that the injection of Aβ25–35 led to long-term memory impairment. However, EA 100 and EA 200 administration significantly shortened the latency time to reach the hidden platform, opposed to that of control group. On the contrary, the time to reach the exposed platform did not show any significant difference among groups. It indicated that the improvement of long-term spatial cognition impaired by Aβ25–35 was not involved in swimming and visual ability, but it was not related to the memory function.

25–35-injection led to the impairment of object recognition ability. Mori et al. (2011) reported that injection of Aβ25–35 into mice led to an impairment of object recognition ability. Our finding also demonstrated that Aβ25–35-injected control group did not show significant difference on recognition between familiar and novel object. In contrast, EA 100 and EA 200 groups showed higher novel objective recognition. It indicated that injection of Aβ25–35-induced deficits in recognition of novel object. However, EA from A. okamotoanum improved the Aβ25–35-induced recognition impairment.

T-maze test results showed that EA 100 and EA 200 groups display significantly higher preference for the novel route, compared to Aβ25–35-injected control group. In addition, EA could prevent memory and cognitive deficits in AD mice model.

Aβ causes oxidative stress by inducing lipid peroxidation, reactive oxygen species, and reactive nitrogen species, such as NO (Markesbery and Carney 1999; Avdulov et al. 1997; Butterfield and Lauderback 2002). It induced the formation of senile plaques, mitochondrial DNA damage, and thus might cause neuronal dysfunction and cell death (Reddy 2006). Eventually, oxidative stress induces the pathogenesis of the neurodegenerative disorders, such as AD (Christen 2000; Yin et al. 2013).

NO contributes to various neurodegenerative disorders, including AD (Stepanichev et al. 2008). Torreilles et al. (1999) reported that the oxidative damage due to the overproduction of NO contributes to Aβ-caused neurotoxicity and memory impairment. In addition, NO in the brain contributes to the pathologies of AD (Aliev et al. 2009). Lipid peroxidation products of several aldehydes, including MDA and 4-hydroxy-2,3-nonenal levels were elevated by oxidative stress. Among them, MDA was considered as a key marker of the lipid peroxidation in the AD model (Esterbauer et al. 1991). Thus, NO and MDA represent important markers of oxidative stress in the AD model. Several studies reported increased MDA (Cotman and Su 1996), NO (Butterfield 1997), carbonyls, and heme oxygenase-1 (Smith et al. 1994, 1996) in the AD model. The present study showed that the level of MDA and NO in mice brain, kidney, and liver were elevated by Aβ25–35 injection. Choi et al. (2013) demonstrated that Aβ25–35 injection in mice resulted in a dramatic improvement in MDA and NO level in mice brain, kidney, and liver, suggesting that Aβ25–35-induced oxidative stress and neurodegeneration. However, oral administration of EA had an prevention influence on MDA and NO production in brain, kidney, and liver of AD mice. It indicated that EA alleviated the oxidative stress and neurodegeneration induced by Aβ25–35.

AChE was found to be involved in the Aβ of senile plaques, pre-Aβ dispersed sediment, and cerebral blood vessels (Geula and Mesulam 1989). AChE is accelerate the lump of Aβ peptide into fibrils deposited in the AD patients’ brain and forms an AChE complex with Aβ. This Aβ–AChE complex has higher toxic effect than Aβ peptide (Melo et al. 2003). Moreover, the Aβ protein also regulates AChE expression and glycosylation in cell, transgenic mice, and AD patient brain. It indicated that AChE creates a vicious cycle, leading to an increased Aβ accumulation (Parnetti et al. 2002). Several reports have shown that AChE activity raised within and encompassing amyloid plaques in AD brain (Ulrich et al. 1990; Chiapinotto Spiazzi et al. 2015). Nakdook et al. (2010) similarly showed that injection of Aβ25–35 caused the increased AChE activity in the cortical and hippocampal regions. The present study also demonstrated that AChE activity in the brain was raised in Aβ25–35-induced mice compared with that of the normal group. Furthermore, in the groups administered with EA, the AChE activity in brain was dramatically lower than that of the control group. It was demonstrated that EA from A. okamotoanum significantly inhibited Aβ25–35-induced AChE activity.

Acer okamotoanum has several biological active compounds. Kim et al. (1998) demonstrated that A. okamotoanum contains quercetin 3-O-(2′′,6′′-O-digalloyl)-β-d-galactoside, flavonol glycoside gallate ester, six kinds of flavonol glycosides, and three kinds of phenolic compounds. These active compounds have the beneficial function against HIV-1 and oxidative stress. In addition, Yun et al. (2001) reported that A. okamotoanum was important functional plant that contains antioxidants, several coumarinolignans, and cleomiscosins A and C. These compounds exerted strong antioxidant effects against lipid peroxidation and LDL oxidation (Thuong et al. 2005). However, no previous report has demonstrated that whether A. okamotoanum protects against improves Aβ25–35-induced cognition impairment. This study is the initial report on the effects of A. okamotoanum against AD. In conclusion, our results indicate that EA improved learning and long-term memory ability against Aβ25–35–induced impairment. Moreover, the present study suggests the potential of EA from A. okamotoanum as a promising dietary agent with protective effect against AD.

References

  • Aliev G, Palacios HH, Lipsitt AE, Fischbach K, Lamb BT, Obrenovich ME, Morales L, Gasimov E, Bragin V (2009) Nitric oxide as an initiator of brain lesions during the development of Alzheimer disease. Neurotox Res 16:293–305

    Article  CAS  Google Scholar 

  • Avdulov NA, Chochina SV, Igbavboa U, O’Hare EO, Schroeder F, Cleary JP, Wood WG (1997) Amyloid beta-peptides increase annular and bulk fluidity and induce lipid peroxidation in brain synaptic plasma membranes. J Neurochem 68:2086–2091

    Article  CAS  Google Scholar 

  • Bevins RA, Besheer J (2006) Object recognition in rats and mice: a one-trial non-matching to-sample learning task to study ‘recognition memory’. Nat Protoc 1:1306–1311

    Article  Google Scholar 

  • Bicca MA, Costa R, Loch Neckel G, Figueiredo CP, Medeiros R, Calixto JB (2015) B2 receptor blockage prevents Aβ-induced cognitive impairment by neuroinflammation inhibition. Behav Brain Res 278:482–491

    Article  CAS  Google Scholar 

  • Brookmeyer R, Gray S (2000) Methods for projecting the incidence and prevalence of chronic diseases in aging populations: application to Alzheimer’s disease. Stat Med 19:1481–1493

    Article  CAS  Google Scholar 

  • Butterfield DA (1997) Beta-amyloid-associated free radical oxidative stress and neurotoxicity: implications for Alzheimer’s disease. Chem Res Toxicol 10:495–506

    Article  CAS  Google Scholar 

  • Butterfield DA, Lauderback CM (2002) Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress. Free Radic Biol Med 32:1050–1060

    Article  CAS  Google Scholar 

  • Chiapinotto Spiazzi C, Bucco Soares M, Pinto Izaguirry A, Musacchio Vargas L, Zanchi MM, Frasson Pavin N, Ferreira Affeldt R, Seibert Ludtke D, Prigol M, Santos FW (2015) Selenofuranoside ameliorates memory Loss in Alzheimer-like sporadic dementia: AChE activity, oxidative stress, and inflammation involvement. Oxid Med Cell Longev 2015:1–9

    Article  Google Scholar 

  • Choi JY, Cho EJ, Lee HS, Lee JM, Yoon YH, Lee SH (2013) Tartary buckwheat improves cognition and memory function in an in vivo amyloid-β-induced Alzheimer model. Food Chem Toxicol 53:105–111

    Article  CAS  Google Scholar 

  • Christen Y (2000) Oxidative stress and Alzheimer disease. Am J Clin Nutr 71:621–629

    Google Scholar 

  • Cotman CW, Su JH (1996) Mechanisms of neuronal death in Alzheimer’s disease. Brain Pathol 6:493–506

    Article  CAS  Google Scholar 

  • De Mattos RB, Bales KR, Cummins DJ, Paul SM, Holtzman DM (2002) Brain to plasma amyloid-β efflux: a measure of brain amyloid burden in a mouse model of Alzheimer’s disease. Science 295:2264–2267

    Article  Google Scholar 

  • Ellman GL, Courtney KD, Andres VJ, Feather Stone RM (1961) A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 7:88–95

    Article  CAS  Google Scholar 

  • Esmaeili TP, Moosavi SM, Shabani M, Haghani M (2015) Erythropoietin improves synaptic plasticity and memory deficits by decrease of the neurotransmitter release probability in the rat model of Alzheimer’s disease. Pharmacol Biochem Behav 130:15–21

    Article  Google Scholar 

  • Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11:81–128

    Article  CAS  Google Scholar 

  • Geula C, Mesulam M (1989) Special properties of cholinesterases in the cerebral cortex of Alzheimer’s disease. Brain Res 498:185–189

    Article  CAS  Google Scholar 

  • Jeong MH, Ha JH, Oh SH, Kim SS, Lee HJ, Kang HY, Lee HY (2009) Comparison of biological activities of Acer mono and A. okamotoanum extracts by water extraction and low temperature high pressure extraction. Korean J Med Crop Sci 17:407–414

    Google Scholar 

  • Jeong MH, Kim SS, Kim JS, Lee HJ, Choi GP, Lee HY (2010) Skin whitening and skin immune activities of different Parts of Acer mono and Acer okamotoanum. J Korean For Soc 99:470–478

    Google Scholar 

  • Kim HJ, Woo ER, Shin CG, Park HK (1998) A new flavonol glycoside gallate ester from Acer okamotoanum and its inhibitory activity against human immunodeficiency virus-1 (HIV-1) integrase. J Nat Prod 61:145–148

    Article  CAS  Google Scholar 

  • Kubo T, Nishimura S, Kumagae Y, Kaneko I (2002) In vivo conversion of racemized beta-amyloid ([D-Ser 26] Aβ1–40) to truncated and toxic fragments ([D-Ser 26]Aβ25–35/40) and fragment presence in the brains of Alzheimer’s patients. J Neurosci Res 70:474–483

    Article  CAS  Google Scholar 

  • Kwon SH, Lee HK, Kim JA, Hong SI, Kim SY, Jo TH, Park YI, Lee CK, Kim YB, Lee SY, Jang CG (2011) Neuroprotective effects of Eucommia ulmoides Oliv. Bark on amyloid beta25–35-induced learning and memory impairments in mice. Neurosci Lett 487:123–127

    Article  CAS  Google Scholar 

  • Laursen SE, Belknap JK (1986) Intracerebroventricular injections in mice. Some methodological refinements. J Pharmacol Methods 16:355–357

    Article  CAS  Google Scholar 

  • Markesbery WR, Carney JM (1999) Oxidative alterations in Alzheimer’s disease. Brain Pathol 9:133–146

    Article  CAS  Google Scholar 

  • Maurice T, Lockhart BP, Privat A (1996) Amnesia induced in mice by centrally administered β-amyloid peptides involves cholinergic dysfunction. Brain Res 706:181–193

    Article  CAS  Google Scholar 

  • Melo JB, Agostinho P, Oliveira CR (2003) Involvement of oxidative stress in the enhancement of acetylcholinesterase activity induced by amyloid β-peptide. Neurosci Res 45:117–127

    Article  CAS  Google Scholar 

  • Montgomery KC (1952) A test of two explanations of spontaneous alternation. J Comp Physiol Psychol 45:287–293

    Article  CAS  Google Scholar 

  • Mori K, Obaram Y, Moriya T, Inatomi S, Nakahata N (2011) Effects of Hericium erinaceus on amyloid β (25–35) peptide-induced learning and memory deficits in mice. Biomed Res 32:67–72

    Article  CAS  Google Scholar 

  • Morris R (1984) Developments of a water-maze procedure for studying a spatial learning in the rat. J Neurosci Methods 11:47–60

    Article  CAS  Google Scholar 

  • Nakdook W, Khongsombat O, Taepavarapruk P, Taepavarapruk N, Ingkaninan K (2010) The effects of Tabernaemontana divaricata root extract on amyloid β-peptide 25–35 peptides induced cognitive deficits in mice. J Ethnopharmacol 130:122–126

    Article  Google Scholar 

  • Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95:351–358

    Article  CAS  Google Scholar 

  • Paban V, Manrique C, Filali M, Maunoir Regimbal S, Fauvelle F, Alescio Lautier B (2014) Therapeutic and preventive effects of methylene blue on Alzheimer’s disease pathology in a transgenic mouse model. Neuropharmacology 76:68–79

    Article  CAS  Google Scholar 

  • Park HJ, Kim SS, Kang S, Rhim H (2009) Intracellular Aβ and C99 aggregates induce mitochondria-dependent cell death in human neuroglioma H4 cells through recruitment of the 20S proteasome subunits. Brain Res 1273:1–8

    Article  CAS  Google Scholar 

  • Parnetti L, Amici S, Lanari A, Romani C, Antognelli C, Andreasen N, Minthon L, Davidsson P, Pottel H, Blennow K, Gallai V (2002) Cerebrospinal fluid levels of biomarkers and activity of acetylcholinesterase (AChE) and butyrylcholinesterase in AD patients before and after treatment with different AChE inhibitors. Neurol Sci 23:95–96

    Article  Google Scholar 

  • Pi R, Mao X, Chao X, Cheng Z, Liu M, Duan X, Ye M, Chen X, Mei Z, Liu P, Li W, Han Y (2012) Tacrine-6-ferulic acid, a novel multifunctional dimer, inhibits amyloid-β-mediated Alzheimer’s disease-associated pathogenesis in vitro and in vivo. PLoS ONE 7:e3192

    Google Scholar 

  • Qadir SA, Kim CH, Kwon MC, Lee HJ, Kang HY, Choi DH, Lee HY (2007) Comparison of anticancer and immune modulatory activities in the different parts for the Acer mono Max. and Acer okamotoanum. Korean J Med Crop Sci 15:405–410

    Google Scholar 

  • Reddy PH (2006) Amyloid precursor protein-mediated free radicals and oxidative damage: implications for the development and progression of Alzheimer’s disease. J Neurochem 96:1–13

    Article  CAS  Google Scholar 

  • Rozemuller JM, Eikelenboom P, Stam FC, Beyreuther K, Masters CL (1989) A4 protein in Alzheimer’s disease: primary and secondary cellular events in extracellular amyloid deposition. J Neuropathol Exp Neurol 48:674–691

    Article  CAS  Google Scholar 

  • Schmidt HH, Warner TD, Nakane M, Forstermann U, Murad F (1992) Regulation and sub cellular location of nitrogen oxide synthases in RAW264.7 macrophages. Mol Pharmacol 41:615–624

    CAS  Google Scholar 

  • Smith MA, Kutty RK, Richey PL, Yan SD, Stern D, Chader GJ, Wiggert B, Petersen RB, Perry G (1994) Heme oxygenase-1 is associated with the neurofibrillary pathology of Alzheimer’s disease. Am J Pathol 145:42–47

    CAS  Google Scholar 

  • Smith MA, Richey PL, Sayre LM, Anderson VE, Beal MF, Kowall N (1996) Test for oxidative damage in Alzheimer’s. Nature 382:120–121

    Article  CAS  Google Scholar 

  • Stepanichev MY, Onufriev MV, Yakovlev AA, Khrenov AI, Peregud DI, Vorontsova ON, Lazareva NA, Gulyaeva NV (2008) Amyloid-β (25–35) increases activity of neuronal NO-synthase in rat brain. Neurochem Int 52:1114–1124

    Article  CAS  Google Scholar 

  • Thuong PT, Na MK, Su ND, Seong RS, Lee YM, Sok DE, Bae KH (2005) Inhibitory effect of coumarins from Weigela subsessilis on low density lipoprotein oxidation. Biol Pharm Bull 28:1095–1097

    Article  CAS  Google Scholar 

  • Torreilles F, Salman Tabcheh S, Guerin MC, Torreilles J (1999) Neurodegenerative disorders: the role of peroxynitrite. Brain Res Rev 30:153–163

    Article  CAS  Google Scholar 

  • Ulrich J, Meier Ruge W, Probst A, Meier E, Ipsen S (1990) Senile plaques: staining for acetylcholinesterase and A4 protein: a comparative study in the hippocampus and entorhinal cortex. Acta Neuropathol 80:624–628

    Article  CAS  Google Scholar 

  • Woo ER, Kim HJ, Kwak JK, Lim YK, Park SK, Kim HS, Lee CK, Park H (1997) Anti-herpetic activity of various medicinal plant extracts. Arch Pharm Res 20:58–67

    Article  CAS  Google Scholar 

  • Yatin SM, Varadarajan S, Butterfield DA (2000) Vitamin E prevents Alzheimer’s amyloid β-peptide (1-42)-induced neuronal protein oxidation and reactive oxygen species production. J Alzheimers Dis 2:123–131

    CAS  Google Scholar 

  • Yim YJ, Lee EB, Kim SH (1981) Vegetation of Ulneung and Dokdo Island. Korean Assoc Conserv Nat 19:97–112

    Google Scholar 

  • Yin Y, Liu Y, Zhuang J, Pan X, Li P, Yang Y, Li YP, Zhao ZQ, Huang LQ, Zhao ZX (2013) Melatonin in Alzheimer’s disease. Int J Mol Sci 14:14575–14593

    Article  Google Scholar 

  • Yoo YM, Jung EM, Kang HY, Choi IG, Choi KC, Jeung EB (2011) The sap of Acer okamotoanum decreases serum alcohol levels after acute ethanol ingestion in rats. Int J Mol Sci 28:489–495

    Google Scholar 

  • Yun BS, Lee IK, Ryoo IJ (2001) Coumarins with monoamine oxidase inhibitory activity and antioxidative coumarinolignans from Hibiscus syriacus. J Nat Prod 64:1238–1240

    Article  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2015R1D1A1A01058868), Republic of Korea.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Sanghyun Lee or Eun Ju Cho.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, S.Y., Lee, J., Lee, D.G. et al. Acer okamotoanum improves cognition and memory function in Aβ25–35-induced Alzheimer’s mice model. Appl Biol Chem 60, 1–9 (2017). https://doi.org/10.1007/s13765-016-0244-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13765-016-0244-x

Keywords