HOME JOURNALS CONTACT

Pharmacologia

Year: 2015 | Volume: 6 | Issue: 5 | Page No.: 192-212
DOI: 10.17311/pharmacologia.2015.192.212
Precautionary Ellagic Acid Treatment Ameliorates Chronically Administered Scopolamine Induced Alzheimer’s Type Memory and Cognitive Dysfunctions in Rats
Ramandeep Kaur, Sidharth Mehan, Deepa Khanna, Sanjeev Kalra and Shaba Parveen

Abstract: Background and Objective: The neuroprotective ability of Ellagic Acid (EA) as a constructive herbal drug to impede cholinergic dysfunctions and oxidative stress in Alzheimer’s Disease (AD) in chronically administered scopolamine induced Alzheimer’s type dementia in rats was evaluated. Methodology: Alzheimer’s type dementia was induced by chronically administered intraperitoneal injection of scopolamine (0.7 mg kg-1) to rats for period of 7 days. The EA (25 and 50 mg kg-1) and Donepezil (0.5 mg kg-1) were administrated to rats orally daily for a period of 13 days. Memory-related behavioral parameters were evaluated using the Elevated plus Maze (EPM) for 2 days and Morris Water Maze (MWM) for 5 days. At the end of protocol schedule i.e., day 14, biochemical parameters were estimated like AChE, MDA, GSH, catalase and SOD to evaluate the neuroprotective action of EA via AChE inhibition and antioxidant activity. Result: Chronically injected scopolamine treatment increased the transfer latency in EPM, escape latency time and shortened time spent in the target quadrant in MWM; these effects were reversed by EA. Scopolamine-mediated changes in malondialdehyde (MDA) and AChE activity were significantly attenuated by EA in rats. Recovery of antioxidant capacities, including reduced glutathione (GSH) content and the activities of SOD and catalase was also evident in EA treated rats. Conclusion: The present findings sufficiently encourage that EA has a major role in the neuroprotection in chronically injected Scopolamine induced Alzheimer type dementia. The EA can be used as an effectual herbal treatment to prevent cholinergic dysfunctions and oxidative stress associated with Alzheimer type dementia.

Fulltext PDF Fulltext HTML

How to cite this article
Ramandeep Kaur, Sidharth Mehan, Deepa Khanna, Sanjeev Kalra and Shaba Parveen, 2015. Precautionary Ellagic Acid Treatment Ameliorates Chronically Administered Scopolamine Induced Alzheimer’s Type Memory and Cognitive Dysfunctions in Rats. Pharmacologia, 6: 192-212.

Keywords: acetylcholinesterase, oxidative stress, polyphenols and Neuroinflammation

INTRODUCTION

Alzheimer’s Disease (AD) is a severe neurodegenerative disorder that gradually results in loss of memory and impairment of cognitive functions in the elderly1-5. In 2014, an estimated 5.2 million people of all ages have AD in U.S. This includes an estimated 5 million people of age 65 and older and approximately 200,000 individuals under age 65 who have younger-onset of Alzheimer’s6 disease. The pathological features of AD include extracellular amyloid deposition and intra-neuronal neurofibrillary tangles (NFTs) of hyperphosphorylated microtubule-associated tau protein7-9. The deposition of amyloid plaques is the primary event that leads to an inflammatory reaction, NFTs formation and ultimately cause neuronal death10-12. The mechanisms of neuronal cell loss in AD have not yet been fully elucidated but increased oxidative stress and inflammation are considered important mediators of neuronal damage in AD13-17.

Many naturally occurring compounds have been proposed as potential therapies to slow or prevent the progression of AD, mostly by acting as antioxidants18-24, but also with some direct anti-amyloid actions18,23,25-30. Recent studies have suggested the positive effects of dietary antioxidants as an aid in potentially reducing somatic cell and neuronal damage by free radicals18-21,31-34. The beneficial health effects of plant-derived products have been largely attributed to polyphenolic compounds, as well as vitamins, minerals and dietary fibers18,19,35.

Ellagic acid (EA), a non flavonoid polyphenol, plays an essential role in explaining the sensory properties of fruits, food and beverages which exhibit this phyto-constituent36-40.

The EA has been well proven to contain anti-oxidant41-46, anti-inflammatory47-51, anti-proliferative,52-56 antidiabetic57-59 and cardioprotective properties60,61.

Neuroprotection can be a property of EA as it prevents both neuro-oxidation and neuroinflammation62-68. Moreover, by in vitro studies it was observed that EA inhibits β-secretase (BACE1), thus inhibiting Aβ-fibrillation and decrease AChE activity4,69-71. Recent studies suggested that glucose metabolism is affected during AD72-75. The EA stimulated GLUT4 translocation primary factor responsible for insulin induced glucose uptake and maintain glucose homeostasis76,77. The EA also shows modulation of monoaminergic system (serotonergic and noradrenergic systems) and GABAergic system78-80. Cognitive impairment in AD patients correlates with disturbance in various neurotransmitters, as the ratio of excitatory-inhibitory neurotransmitter levels disturb, cytotoxic damage to neurons and glia occurs and norepinephrine and serotonin levels declined81-91. Further, Gamma-Amino Butyric Acid (GABA) increases the formation of soluble receptor for advanced glycation end products (RAGE) and decreases the levels of full-length RAGE, lowering the Aβ uptake and inflammatory mediated reactions92,93.

Scopolamine, an antimuscarinic agent, competitively antagonizes the effect of acetylcholine on the muscarinic receptors by occupying postsynaptic receptor sites with high affinity and increases AChE activity in the cortex and hippocampus94-103. Scopolamine abolishes cerebral blood flow due to cholinergic hypofunction104-107. Scopolamine additionally triggers ROS, inducing free radical injury and an increase in a scopolamine-treated group brain MDA levels and deterioration in antioxidant status108-112. Scopolamine induces neuro-inflammation by promoting high level of oxidative stress and pro inflammatory cytokines in the hippocampus113-119. Scopolamine is proven to increase levels of APP and Tau protein. Chronic administration of scopolamine led to marked histopathological alterations in the cerebral cortex, including neuronal degeneration30,120-122. Scopolamine administration has been used both in healthy human volunteers and in animals as a model of dementia to determine the effectiveness of potential new therapeutic agents for Alzheimer’s disease123-128 (Fig. 1).

Donepezil, a reversible inhibitor of AChE, is neuroprotective due to not only activation of cholinergic transmission but also by reducing the amount of the toxic form of amyloid β fibrils129-136. Donepezil ameliorated the scopolamine induced memory impairment by reducing AChE activity and oxidative stress and restoring cerebral circulation137-143. With this background, EA might show neuroprotection via inhibiting neuronal dysfunctions. There is major requirement to determine therapeutic potential for EA in cases of AD with suitable behavioral and biochemical markers. This study was an attempt to investigate the neuroprotective effect of EA, potential of doses for the treatment of Alzheimer’s disease.

MATERIAL AND METHOD

Chemicals: EA was purchased from Yucca Interprises, Mumbai, India and suspended in saline solution. Scopolamine hydrochloride was purchased from Sigma-Aldrich, St, Louis, MO, USA. Donepezil was obtained from Ranbaxy Pvt. Limited, Mumbai, India and both scopolamine and donepezil were dissolved in saline solution. All reagents used in this study were of analytical grade and high purity.

Animals: Male Wistar rats (weighing 220-250 g, aged 8-10 months) obtained from the Animal House of the Institute were employed in the studies. The animals were kept in polyacrylic cages with wire mesh top and soft bedding. They were kept under standard husbandry conditions of 12 h reverse light cycle with food and water ad libitum, maintained at 22±2°C. The experimental protocol was approved by Institutional Animal Ethics Committee (IAEC) as per the guidelines of Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Government of India (RITS/IAEC/2013/01/01). Animals were acclimatized to laboratory conditions prior to experimentation.

Drug administration: EA was administered by oral (p.o.) route in dose of 25 and 50 mg kg-1. Scopolamine was administered by intraperitoneal (i.p.) route in dose of 0.7 mg kg-1. Donepezil was administered by oral (p.o.) route in dose of 0.5 mg kg-1.

Six groups (each group consist six rats) were employed in the present study. (1) Group 1: Normal Control (2) Group 2: Scopolamine Control (0.7 mg kg-1, i.p.) (3) Group 3: EA perse (50 mg kg-1, p.o.) 25 mg kg-1, p.o.+Scopolamine (0.7 mg kg-1, i.p.) (4) Group 6: EA 50 mg kg-1, p.o.+Scopolamine (0.7 mg kg-1, i.p.). After a 5 day habituation period, rats were given EA (25 and 50 mg kg-1, p.o.) and Donepezil (0.5 mg kg-1, p.o.) for total of 13 days. EA alone was treated for 6 days and then scopolamine (0.7 mg kg-1, i.p.) was administered together with EA for another 7 days. Rats underwent locomotor activity (LMA) for 2 days i.e., 6th day and 13th day, MWM test for 5 days i.e., 7th day to 11th day. The day after completion of Morris Water Maze (MWM), the Elevated plus Maze (EPM) was conducted for 2 days i.e., 12th-13th day. The day after EPM, the rats were sacrificed and biochemical parameters were estimated (Fig. 2).

Elevated plus maze: Elevated plus Maze (EPM) served as the behavioral model (where in the stimulus existed outside the body) to evaluate learning and memory in rats. It consists of two opened arms (50x10 cm) and two covered arms (50x40x10 cm). The arms were extended from central platform (10x10 cm) and the maze was kept elevated to a height of 50 cm from the floor. The EPM was conducted for 2 days i.e., 12th-13th day of protocol schedule. Each animal was kept at the end of an open arm, facing away from the central platform on 12th day.

Transfer Latency (TL) which was taken as the time taken by the animal to move into any one of the covered arms with all its four legs, recorded on 12th day i.e., acquisition trial144. If the rat did not enter into one of the covered arms within 120 sec then it was gently pushed into one of the two covered arms and the transfer latency was assigned as 120 sec. The rats were allowed to explore the maze for 10 sec and then were returned to its home cage. TL was again examined 24 h after the first trial on 13th day of protocol schedule i.e., retention latency.

Spatial navigation task in morris water maze: Morris water maze employed in the present study was a model to evaluate spatial learning and memory. Escape from water itself acts as motivation and eliminates the use of other motivational stimuli such as food and water deprivation. Water provides uniform environment and eliminates interference due to olfactory clues145. Animals were trained to swim to a platform in a circular pool (180 cm diameterx60 cm) located in a sound attenuated dark test room. The pool was filled with water (28±2°C) to a depth of 40 cm. A movable circular platform, 9 cm in diameter and mounted on a column, was placed in the pool 2 cm below the water level for Escape Latency Time (ELT) while during Time Spent in the Target Quadrant (TSTQ), the platform was removed. Four equally spaced locations around the edge of the pool (N, S, E and W) were used to divide the pool into 4 quadrants and one of them is used as start point which was same during all trials. The pool was filled with opaque water to prevent visibility of the platform in the pool. The escape platform was placed in the middle of one of the random quadrants of the pool and kept in the same position throughout the experiments. Animals received a training session consisting of day 7-10 and ELT was recorded. ELT defined as the time taken by the animal to locate the hidden platform. ELT was noted as an index of learning. Each animal was subjected to single trial for four consecutive days (starting form 7th day of EA administration to 10th day), during which they were allowed to escape on the hidden platform and to remain there for 20 sec. If the rats failed to find the platform within 120 sec, it was guided gently onto the platform and allowed to remain there for 20 sec.

On fifth day (i.e., 11th day of EA administration) the platform was removed. Rats were placed in water maze and allowed to explore the maze for 120 sec. Time spent in three quadrants, that is, Q1, Q2 and Q3 was recorded and TSTQ in search of the missing platform provided as an index of retrieval. Care was taken not to disturb the relative location of water maze with respect to other objects in the laboratory.

Assessment of locomotor activity: Gross behavioral activity was assessed by digital actophotometer on 6th day and 13th day of protocol schedule to rule out any interference in locomotor activity by drugs which may affect the process of learning and memory, in before and after of MWM task. Each animal was observed over a period of 5 min in a square (30 cm) closed arena equipped with infrared light-sensitive photocells and values expressed as counts per 5 min146. The beams in the actophotometer, cut by the animal, were taken as measure of movements. The apparatus was placed in a darkened, sound-attenuated and ventilated testing room.

Preparation of brain homogenate: On 14th day of protocol schedule, Animals were sacrificed by decapitation, brains removed and rinsed with ice cold isotonic saline solution. Brain tissue samples were then homogenized with 10 times (w/v) ice cold 0.1 M phosphate buffer (pH 7.4). The homogenate was centrifuged at 10,000x g for 15 min, supernatant was separated and aliquots were used for biochemical estimations146.

Protein estimation: The protein content was measured by using Agappe protein estimation kit (Biuret method).

Estimation of acetylcholinesterase levels: The quantitative measurement of AChE activity in brain was performed according to the method described by Ellman et al. (1961)147. The enzymatic activity in the supernatant was expressed as nmol per mg protein.

Estimation of malondialdehyde: The quantitative measurement of MDA-end product of lipid peroxidation-in brain homogenate was performed according to the method of Wills (1966)148. The concentration of MDA was expressed as nmol per mg protein.

Estimation of reduced glutathione: The GSH in brain was estimated according to the method described by Ellman et al. (1959)149. The concentration of glutathione in the supernatant expressed as μmol per mg protein.

Estimation of superoxide dismutase activity: The SOD activity was measured according to the method described by Misra and Frodvich (1972)150. The activity of SOD was expressed as % activity.

Estimation of catalase activity: Catalase activity was measured by the method of Aebi (1974)151. The activity of catalase was expressed as % activity.

Statistical analysis: All the results and data were expressed as Mean±Standard deviation. Data was analyzed using two way ANOVA followed by Post hoc test bonferroni and one way ANOVA followed by Post hoc test tukey’s multi-comparison test. The p<0.05 was considered as statistically significant.

RESULTS

Effect of Ellagic acid on rats in elevated plus maze: On 12th day of protocol schedule, acquisition latency was recorded. Retention was observed as Transfer Latency (TL) on 13th day to evaluate learning and memory in rats using EPM. On 12th and 13th day Scopolamine administered rats showed remarkable increase (113±9.380 and 106.5±11.148 sec) in TL, when compared to normal (64±4.242 and 36.833±6.765 sec) and EA perse rats (63.333±10.385 and 32.833±3.311 sec). During experiment, EA perse administration did not reveal any change, when compared to normal rats in TL. Donepezil, a well established standard drug for AD considerably decrease (65.5±13.003 and 21.666±5.085 sec) TL, when compared to Scopolamine managed rats and reversed the memory impairment induced by Scopolamine. Administration of EA at the dose of 25 mg kg-1, p.o. exhibit notable decrease (72.00±8.049 and 39.333±6.186 sec) in TL, when compared to Scopolamine treated rats. The EA (50 mg kg-1, p.o.) administration also decreases (69.333±8.041 and 25.333±3.881 sec) TL, when differentiate to Scopolamine handled rats and there were expressively variation was found in between treatment doses of EA 25 and 50 mg kg-1, p.o. indicating improved retention memory (Fig. 3).

Effect of ellagic acid on rats in spatial navigation task using Morris water maze: On 7th-10th day of 14 day protocol schedule, Escape Latency Time (ELT) was observed. On 7th day, there were no significant changes observed in Scopolamine (94.33±13.125 sec) treated rats, when compared to normal (89±9.859 sec) and EA perse governed (86.33±13.937 sec) rats. The EA perse administration did not show any significant change when compared to normal rats. Moreover, Donepezil treated rats did not show any considerable changes (88±9.033 sec), when compared to Scopolamine responded rats.

In the treatment groups, administration of EA did not confirm notable changes (96.33±10.053; 88.66±10.689 sec) in ELT at 25 and 50 mg kg-1, p.o. when compared to Scopolamine treated rats. There were no changes found in ELT between treatment doses of EA 25 and 50 mg kg-1, p.o.

Comparison data of 8th day, 9th day and 10th day ELT in MWM, showed that Scopolamine administered rats manifest remarkable increase (92±8.173, 85.33±12.75 and 83.33±8.664 sec) in ELT, when collate to normal (76.33±7.840, 29.16±7.808 and 15.33±3.723 sec) and EA perse (67.33±5.645, 29.33±8.710 and 15±2.898 sec) rats. EA perse administration did not show any significant difference, when compared to normal rats during ELT. Donepezil served rats outstandingly decreased (51±10.158, 26.16±6.40 and 10.83±4.622 sec) ELT when compared to Scopolamine dosed rats. EA at 25 mg kg-1, p.o. proved remarkable decreased (79±10.807, 60.83±8.658 and 38.16±9.703 sec) in the ELT, when compared to Scopolamine employed rats. EA at the dose 50 mg kg-1, p.o. significantly decreased (65.33±11.707, 43±9.838 and 24.5±8.312 sec) the ELT, when compared to Scopolamine and EA 25 mg kg-1, p.o. treated rats, indicating remarkable improvement in learning (Fig. 4).

On 11th day of protocol schedule, TSTQ was performed. Time Spent in Target Quadrant (TSTQ) in search of missing platform provided as an index of retrieval. Scopolamine treated rats showed remarkable decrease (7.667±3.077 sec) in TSTQ when compared to normal (45.17±8.060 sec) and EA perse treated (43.83±6.242 sec) rats.


In perse group of EA, there were no changes during TSTQ when compared to normal group. Further, Donepezil served rats improved (46.17±5.345 sec) memory when compared to Scopolamine treated rats.

EA (25 mg kg-1, p.o.) administration showed remarkable increase (19.50±1.517 sec) in TSTQ when compared to Scopolamine treated rats. EA (50 mg kg-1, p.o.) administration indicated improvement (32.00±8.149 sec) in memory function when compared with Scopolamine governed rats. Moreover, markedly difference was also observed in between treatment doses of EA (Fig. 5).

Effect of ellagic acid on rats in locomotor activity: On 6th and 13th day of protocol schedule, locomotor activity was observed to rule out any interference in locomotor activity by treatment drugs. Scopolamine employed rats did not reveal any significant changes (281.333±15.318 and 274.833±5.344) in locomotor activity when compared to normal (263.833±17.474 and 274.5±21.314) and EA perse (270.666±18.250 and 274.5±4.764) rats. EA perse administration also did not show any considerable change in locomotor activity at 50 mg kg-1, p.o. when compared to normal rats. Donepezil treated also showed trivially changes (267.5±21.314 and 274.833±5.344) when compared to Scopolamine treated rats. EA 25 mg kg-1, p.o. (266.833±15.458 and 270.833±20.692) and 50 mg kg-1, p.o. (274.5±4.764 and 283.5±16.208) administration did not showed any notable changes in locomotor activity of rats when differentiate to Scopolamine treated rats, indicating there were no effect on locomotor activity (Fig. 6).

Effect of ellagic acid on acetylcholinesterase levels: Prolongation of availability of acetylcholine has been used to enhancing cholinergic function. This prolongation may be achieved by inhibiting AChE.


Scopolamine administered rats significantly increased (415.0±19.62) the AChE level when compared to normal (136.8±4.956) and EA perse (137.2±4.167) rats. EA perse administration did not show any appreciable changes in AChE level at the dose of 50 mg kg-1, p.o. when compared to normal rats. Donepezil treated rats appreciably decreased (231.0±7.668) the AChE level in contrast to Scopolamine dosed rats. EA (25 mg kg-1, p.o.) showed remarkably diminished the AChE level (360.8±15.96) when compared to Scopolamine rats. Administration of EA (50 mg kg-1, p.o.) significantly reduced (311.7±17.63) the AChE level when compared to Scopolamine employed rats. Moreover, expressive distinction was present in between treatment doses of EA (Fig. 7).

Effect of ellagic acid on malondialdehyde levels: MDA is an indicator of lipid peroxidation. Scopolamine administration increased (42.50±3.082) the MDA level when compared to normal (19.88±0.960) and EA perse (19.15±1.841) rats. Further, EA perse administration did not show any considerable changes in MDA levels when compared to normal rats. Donepezil appreciably decreased (23.12±0.511) the MDA level when compared to Scopolamine managed rats. EA (25 mg kg-1, p.o.) administration showed remarkably decrease (33.57±3.347) in MDA level when compared to Scopolamine treated rats. EA administered rats at the dose of 50 mg kg-1, p.o significantly decreased (27.97±2.089) in MDA level when compared to Scopolamine and EA 25 mg kg-1, p.o. treated rats (Fig. 8).

Effect of ellagic acid on reduced glutathione levels: Reduced GSH is a marker of cellular antioxidant and provide protection against oxidative stress. Scopolamine governed rats remarkably decreased (2.067±0.417) the GSH level when compared to normal (9.833±0.776) and EA perse treated (9.733±0.799) rats. EA perse administration did not show any considerable changes in GSH levels in contrast to normal rats. Donepezil outstandingly increase (7.767±0.361) the GSH levels when compared to Scopolamine treated rats. The EA (25 mg kg-1, p.o.) administration exhibited remarkable increase (5.250±0.575) in GSH level when compared to Scopolamine treated rats. The EA (50 mg kg-1, p.o.) showed significantly increase (6.317±0.386) in GSH level when compared to Scopolamine treated rats. Moreover, in between treatment doses of EA, significance difference was present (Fig. 9).

Effect of Ellagic acid on superoxide dismutase activity: The SOD is an antioxidant enzyme which plays a key role in detoxifying superoxide anions. Scopolamine administered rats significantly decreased (27.33±3.386) the SOD levels in brain homogenate when compared to normal (100.0±0.0) and EA perse (95.83±2.639) rats.


EA perse administration did not reveal any considerable change in SOD activity when collated to normal rats.

Donepezil expressively increase (82.00±3.950) SOD activity when compared to Scopolamine treated rats. In treatment group, EA (25 mg kg-1, p.o.) administration showed remarkable increase (59.17±8.060) in SOD activity when compared to Scopolamine treated rats. EA (50 mg kg-1, p.o.) administration showed significantly increase (71.33±4.033) in SOD activity when compared to Scopolamine treated rats and a remarkable disparity was found in between EA treated groups (Fig. 10).

Effect of ellagic acid on catalase activity: Catalase is also an antioxidant enzyme which has capability to detoxify oxidative free radicals. Scopolamine treated rats manifested remarkable decrease (36.50±4.461) in catalase activity in brain homogenate when differentiated to normal (100.0±0.0) and EA perse treated (95.50±1.871) rats. The EA perse administration did not show any considerable changes in catalase activity when compared to normal rats. Donepezil significantly increase (81.67±4.033) in catalase activity when compared to Scopolamine treated (36.50±4.461) rats. The EA (25 mg kg-1, p.o.) remarkably increased (59.17±4.579) the catalase activity when compared to Scopolamine treated rats. The EA (50 mg kg-1, p.o.) administration exhibited significantly increase (73.67±3.559) in catalase activity when compared to Scopolamine and EA 25 mg kg-1, p.o. treated rats (Fig. 11).

DISCUSSION

Clinically AD is characterized by an insidious degradation of memory, associated with functional decline and neurobehavioral disturbances152,153. Despite the availability of various treatment strategies, the severity and prevalence of this disease are not yet under control. Therefore, alternative and complementary medicines including herbal supplements, phytochemicals and extracts are being utilized in the management of AD154-160. The current hypothesis about the mechanisms by which neurons come into necrotic or apoptotic processes has led to believe that the therapeutic use of natural antioxidants may be beneficial in aging and neurodegenerative disorders161-163.

In the present study, the effect of improving memory deficit of EA was evaluated using chronically administered scopolamine induced Alzheimer’s type dementia in rats.

It is well known that scopolamine as a cholinergic receptor antagonist has been shown to impair learning and memory processing95,97,100,103. Scopolamine produces deficits in acquisition, immediate retention and working memory164-169.

The current study has revealed that long term administered scopolamine significantly increased the levels of lipid peroxidation products such as MDA and decreased the levels of antioxidants viz., GSH, SOD and catalase. The increase in oxidative stress was found to be associated with increase in AChE activity and spatial cognitive deficit. Present findings are in tune with previous reports99,102,111,118.

Scopolamine induced Alzheimer’s type dementia model has been widely used to provide a pharmacological model of memory dysfunction for screening potential cognition enhancing agents99,110-112,170. The cognitive-enhancing activity of EA on chronically administered scopolamine induced memory impairments in rats was investigated by using behavioral and biochemical parameters.

During elevated plus maze, decrease in retention latency indicated improvement of memory and vice versa142,171-173. In EPM, it was shown that long term injected scopolamine also drastically increase in TL, demonstrating that the central cholinergic neuronal system plays an important role in learning acquisition. EA dose-dependently decreased TL prolongation induced by scopolamine. These results suggested that the neuroprotective effect of EA on scopolamine-induced memory impairment may be related to mediation of the cholinergic nervous system. In order to confirm the effects of EA, MWM was used to test spatial learning in rats, where scopolamine treated rats were taking more time to reach at the hidden platform which shows memory impairments in this spatial task. EA treated rats impressively reduced the escape latency prolonged by scopolamine. Moreover, EA exhibited appreciable improvement of cognitive performance as indicated by significant decrease in ELT. It is important to notice that MWM test investigating spatial learning and memory has been used in detecting changes of the central cholinergic system174-178. If the animals spent more time in target quadrant where the platform had previously been placed during the training session, this would indicate that the animals learned from prior experience with the MWM test, showing the spatial memory improvement. Scopolamine treated rats decreased TSTQ, on the other side EA treated rats expressively increased the TSTQ. Both the test doses viz., 25 and 50 mg kg-1, p.o. significantly attenuated these behavioral changes in rats with chronically administered scopolamine induced memory and cognitive impairment.

Along with EPM and MWM, Locomotor activity also was investigated using actophotometer to determine any modulation in locomotor activity by treatment drugs which may affect locomotion in EPM and MWM. However, no significant difference in locomotor activity was observed in any of the animal groups. These results suggest that there was not any sedative effect or interference in EPM and MWM locomotion. Therefore, transfer latency in EPM, escape latency and TSTQ in MWM were purely result of improved memory. Therefore, EA can repair the long-term memory in chronically injected scopolamine-induced memory impairments.

To investigate the effect of EA on cholinergic function, that governs vital aspects of memory and other cognitive functions, brain acetylcholinesterase activity was measured in the present study. The hippocampus, amygdala and cortical regions of the brain are mainly involved in cholinergic transmission to monitor learning and memory processing and seem to be more prone to oxidative damage9,179-181. Moreover, oxidative damage to the rat synapse in these regions of brain has been reported to contribute to cognitive deficits182,183. The AD is characterized by alterations at the level of various neurotransmitters. The most severely affected is the cholinergic system which is responsible for the storage and retrieval of items in memory and its degradation correlates well with the severity of cognitive and memory impairment10,184.

In this study, scopolamine was found to significantly elevate AChE activity, an enzyme responsible for degradation of ACh which is in tune with earlier reports102,118. This increase in AChE activity was significantly restored dose dependently by EA. These observations suggest the modulation of cholinergic neurotransmission and/or prevention of cholinergic neuronal loss.

Recently, many studies have reported that memory impairments is associated to oxidative damage in the scopolamine-induced dementia in rats110-112. Moreover, many clinical studies have reported that oxidative stress is closely involved in the pathogenesis of AD13,185-188.

Lipid peroxidation is an important indicator of neurodegenration of brain. Unlike other body membranes, neuronal membranes contain a very high percentage of long chain polyunsaturated fatty acids because they are used to construct complex structures needed for high rates of signal transfer. The ROS are generated continuously in nervous tissues during normal metabolism and neuronal activity. The brain is subjected to free radical induced lipid peroxidation because it uses one-third of the inspired oxygen189,190. Lipids and proteins, the major structural and functional components of the cell membrane are the target of oxidative modification by free radicals in neurodegenerative disorders191. Extensive evidence exists on lipid peroxidation and protein oxidation leading to loss of membrane integrity, an important factor in acceleration of aging and age-related neurodegenerative disorders. Oxidative stress has been implicated in the pathogenesis of AD in humans192-194.

In the present study, scopolamine-injection in rats significantly induced peroxidation of lipids and proteins and reduced antioxidant defense indicating increased oxidative stress. MDA is an end product of lipid peroxidation and is a measure of free radical generation and scopolamine injected rats showed extensive lipid peroxidation as evidenced by increase in MDA levels. In order to evaluate the effect of EA on lipid peroxidation in brain, MDA level was assessed. MDA level was remarkably increased by scopolamine and EA dose-dependently reduced MDA level, indicating the reduced peroxidation of lipids.

Lipid peroxidation may enhance due to depletion of GSH content in the brain which is often considered as the first line of defense of the cell by this endogenous antioxidant against oxidative stress191,195-197. Evidence has been presented that the neuronal defense against H2O2 which is the most toxic molecule to the brain, is mediated primarily by the glutathione system198-200. The GSH is a tri-peptide, an endogenous antioxidant found in all animal cells in variable amounts and is a very accurate indicator of oxidative stress197. Consistent with previous studies, in present study, scopolamine treatment significantly decreased the GSH levels. Further, co-administration of EA markedly improved GSH levels.

The most important antioxidant enzymes are SOD and catalase. The SOD plays a key role in detoxifying superoxide anions which otherwise damages the cell membranes and macromolecules. Scopolamine administration showed a significant reduction in enzymatic activity of SOD and catalase. On the other side, Catalase has the capability to detoxify H2O2 radicals. Release of H2O2 promotes the formation of numerous other oxidant species that greatly contributes for oxidative stress leading to the pathogenesis of AD189,201. Scopolamine treatment was found to be decreased SOD and catalase activities. Treatment of rats with EA significantly preserved the activities of SOD and catalase.

It has been well documented that persistent administration of scopolamine in response to degradation of ACh and increase the level of AChE enzyme, further responsible for the production of oxidative stress and pro-inflammatory mediators viz., cytokines and further activation of these cells99,110-112. A strong and long lasting administration of scopolamine has been demonstrated to cause cholinergic dysfunction while inhibition of this scopolamine mediated abnormalities has shown to reverse cholinergic dysfunction as well as inhibit the release of oxidative and inflammatory markers99,103,112. The results of the present study suggest that chronic administration of EA perse did not have any significant effect on cognitive performance in normal animals. But, EA treatment groups at the dose of 25 and 50 mg kg-1, p.o. showed marked improvement in cognitive tasks when compared to scopolamine treated rats suggesting the significant role of ACh in long lasting administrated scopolamine mediated cognitive dysfunction. Reports also support that ACh is involved in memory acquisition and retention10,155,202,203. Moreover, scopolamine injection drastically impaired memory retention, resembling Alzheimer’s dementia103,112. The same has been reported to be attenuated by pretreatment with herbal supplements and extracts and phytochemicals156-158,160.

The presented data in this study also suggests that EA possesses potent antioxidant activity by scavenging ROS and exerting a neuro-protective effect against oxidative damage induced by long term administration of scopolamine (Fig. 12).

Predominant role of AChE inhibition, antioxidant activity reveal an important contributory factor to the beneficial effects of EA against dementia. Higher dose of Ellagic acid i.e., 50 mg kg-1, p.o. was found more neuroprotective in all behavioral and biochemical evaluations. At lastly, the neuroprotective effects of EA might result from the regulation of AChE and the anti-oxidative defense system. These results suggest that EA can be used as a constructive herbal drug to impede cholinergic dysfunctions and oxidative stress in AD.

CONCLUSION

It was concluded that long term injected scopolamine could persuade Alzheimer’s type dementia via increase AChE levels and oxidative stress like bio-markers. Scopolamine mediated Alzheimer’s type dementia is mainly associated with cognitive and memory impairments in behavioral models like elevated plus maze and morris water maze. Ellagic acid diminished the acetylcholinesterase level and improves the anti-oxidant defense system. Further, Ellagic acid down turned the cognitive impairments induced by scopolamine. Like Donepezil, Ellagic acid reversed the scopolamine induced Alzheimer’s type dementia in rats. Therefore, Ellagic Acid can be used as an effectual herbal treatment to prevent cholinergic dysfunctions and oxidative stress associated with Alzheimer’s type dementia.

On the basis of this study, the major bio-markers of Alzheimer’s disease like amyloid beta, inflammatory cytokines and histopathological changes can be further evaluated according to current protocol schedule to confirm and justify the strong evidence of Ellagic acid in long term injected scopolamine mediated dementia.

ACKNOWLEDGMENTS

We express our gratitude to Chairman Dr. Rajender Sra and Secretary Dr. Om Parkash, Rajendra Institute of Technology and Sciences, Sirsa, Haryana, India for their inspiration. We also special thanks to Mr. Raghuvir Singh for their constant support.

REFERENCES

  • Chong, Z.Z., F. Li and K. Maiese, 2005. Stress in the brain: Novel cellular mechanisms of injury linked to Alzheimer's disease. Brain Res. Rev., 49: 1-21.
    CrossRef    Direct Link    


  • Walsh, D.M., I. Klyubin, G.M. Shankar, M. Townsend and J.V. Fadeeva et al., 2005. The role of cell-derived oligomers of Abeta in Alzheimer's disease and avenues for therapeutic intervention. Biochem. Soc. Trans., 33: 1087-1090.
    PubMed    


  • Jellinger, K.A., 2006. Alzheimer 100-highlights in the history of Alzheimer research. J. Neural Trans., 113: 1603-1623.
    CrossRef    Direct Link    


  • Muthaiyah, B., M.M. Essa, V. Chauhan and A. Chauhan, 2011. Protective effects of walnut extract against amyloid beta peptide-induced cell death and oxidative stress in PC12 cells. Neurochem. Res., 36: 2096-2103.
    CrossRef    Direct Link    


  • Anand, R., K.D. Gill and A.A. Mahdi, 2014. Therapeutics of Alzheimer's disease: Past, present and future. Neuropharmacology, 76: 27-50.
    Direct Link    


  • Alzheimer's Association, 2014. 2014 Alzheimer's disease facts and figures. Alzheimer's Dementia, 10: e47-e92.
    CrossRef    Direct Link    


  • Anderson, D.C., 2013. Alzheimer's disease biomarkers: More than molecular diagnostics. Drug Dev. Res., 74: 92-111.
    CrossRef    Direct Link    


  • Blennow, K., H. Zetterberg and A.M. Fagan, 2012. Fluid biomarkers in Alzheimer disease. Cold Spring Harb. Perspect. Med., Vol. 2
    CrossRef    


  • Mehan, S., H. Meena, D. Sharma and R. Sankhla, 2011. JNK: A stress-activated protein kinase therapeutic strategies and involvement in Alzheimer's and various neurodegenerative abnormalities. J. Mol. Neurosci., 43: 376-390.
    CrossRef    PubMed    Direct Link    


  • Kihara, T. and S. Shimohama, 2004. Alzheimer's disease and acetylcholine receptors. Acta Neurobiol. Exp., 64: 99-106.
    Direct Link    


  • Verdile, G., S. Fuller, C.S. Atwood, S.M. Laws, S.E. Gandy and R.N. Martins, 2004. The role of beta amyloid in Alzheimer's disease: Still a cause of everything or the only one who got caught? Pharmacol. Res., 50: 397-409.
    CrossRef    PubMed    Direct Link    


  • Anandatheerthavarada, H.K., G. Biswas, M.A. Robin and N.G. Avadhani, 2003. Mitochondrial targeting and a novel transmembrane arrest of Alzheimer's amyloid precursor protein impairs mitochondrial function in neuronal cells. J. Cell Boil., 161: 41-54.
    Direct Link    


  • Marcus, D.L., C. Thomas, C. Rodriguez, K. Simberkoff, J.S. Tsai, J.A. Strafaci and L. Michael, 1989. Freedman Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer's disease. Exp. Neurol., 150: 40-44.
    Direct Link    


  • Nunomura, A., G. Perry, G. Aliev, K. Hirai and A. Takeda et al., 2001. Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol., 60: 759-767.
    PubMed    Direct Link    


  • Sarkar, P.K., 2005. Degeneration and death of neurons in adult neurodegenerative diseases. Curr. Sci.-Bangalore, 89: 764-767.
    Direct Link    


  • Heneka, M.T. and M.K. O'Banion, 2007. Inflammatory processes in Alzheimer's disease. J. Neuroimmunol., 184: 69-91.
    CrossRef    Direct Link    


  • Galasko, D. and T.J. Montine, 2010. Biomarkers of oxidative damage and inflammation in Alzheimer's disease. Biomark. Med., 4: 27-36.
    Direct Link    


  • Engelhart, M.J., M.I. Geerlings, A. Ruitenberg, J.C. van Swieten, A. Hofman, J.C. Witteman and M.M. Breteler, 2002. Dietary intake of antioxidants and risk of Alzheimer disease. J. Am. Med. Assoc., 287: 3223-3229.
    CrossRef    PubMed    Direct Link    


  • Morris, M.C., D.C. Evans, J.L. Bienias, C.C. Tangney and D.A. Bennett et al., 2002. Dietary intake of antioxidant nutrients and the risk of incident Alzheimer disease in a biracial community study. J. Am. Med. Assco., 287: 3230-3237.
    PubMed    


  • Dai, Q., A.R. Borenstein, Y. Wu, J.C. Jackson and E.B. Larson, 2006. Fruit and vegetable juices and Alzheimer's disease: The Kame project. Am. J. Med., 119: 751-759.
    CrossRef    Direct Link    


  • Mancuso, C., T.E. Bates, D.A. Butterfield, S.C. Calafato and C. Cornelius et al., 2007. Natural antioxidants in Alzheimer's disease. Expert Opin. Invest. Drugs, 16: 1921-1931.
    CrossRef    PubMed    Direct Link    


  • Staehelin, H.B., 2008. Neuronal protection by bioactive nutrients. Int. J. Vitamin Nutr. Res., 78: 282-285.
    CrossRef    Direct Link    


  • Harvey, B.S., I.F. Musgrave, K.S. Ohlsson, A. Fransson and S.D. Smid, 2011. The green tea polyphenol (-)-epigallocatechin-3-gallate inhibits amyloid-β evoked fibril formation and neuronal cell death in vitro. Food Chem., 129: 1729-1736.
    Direct Link    


  • Obulesu, M. and D.M. Rao, 2011. Effect of plant extracts on Alzheimer's disease: An insight into therapeutic avenues. J. Neurosci. Rural Pract., 2: 56-61.
    CrossRef    Direct Link    


  • Bastianetto, S., C. Ramassamy, S. Dore, Y. Christen and J. Poirier et al., 2000. The Ginkgo biloba extract (EGb 761) protects hippocampal neurons against cell death induced by β-amyloid. Eur. J. Neurosci., 12: 1882-1890.
    CrossRef    PubMed    ISI    


  • Choi, Y.T., C.H. Jung, S.R. Lee, J.H. Bae and W.K. Baek et al., 2001. The green tea polyphenol (-)-epigallocatechin gallate attenuates β-amyloid-induced neurotoxicity in cultured hippocampal neurons. Life Sci., 70: 603-614.
    Direct Link    


  • Li, M.H., J.H. Jang, B. Sun and Y.J. Surh, 2004. Protective effects of oligomers of grape seed polyphenols against beta-amyloid-induced oxidative cell death. Ann. N.Y. Acad. Sci., 1030: 317-317.


  • Mishra, S. and K. Palanivelu, 2008. The effect of curcumin (turmeric) on Alzheimer's disease: An overview. Ann. Indian Acad. Neurol., 11: 13-19.
    CrossRef    Direct Link    


  • Craggs, L. and R.N. Kalaria, 2010. Revisiting dietary antioxidants, neurodegeneration and dementia. Neuroreport, 22: 1-3.
    PubMed    Direct Link    


  • Choi, D.Y., Y.J. Lee, S.Y. Lee, Y.M. Lee and H.H. Lee et al., 2012. Attenuation of scopolamine-induced cognitive dysfunction by obovatol. Arch. Pharm. Res., 35: 1279-1286.
    CrossRef    Direct Link    


  • Lim, G.P., T. Chu, F. Yang, W. Beech, S.A. Frautschy and G.M. Cole, 2001. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J. Neurosci., 21: 8370-8377.
    PubMed    


  • Thomas, P., Y.J. Wang, J.H. Zhong, S. Kosaraju, N.J. O'Callaghan, X.F. Zhou and M. Fenech, 2009. Grape seed polyphenols and curcumin reduce genomic instability events in a transgenic mouse model for Alzheimer's disease. Mutat. Res./Fundam. Mol. Mech. Mutagen., 661: 25-34.
    CrossRef    PubMed    Direct Link    


  • Fernandez-Fernandez, L., G. Comes, I. Bolea, T. Valente and J. Ruiz et al., 2012. LMN diet, rich in polyphenols and polyunsaturated fatty acids, improves mouse cognitive decline associated with aging and Alzheimer's disease. Behav. Brain Res., 228: 261-271.
    Direct Link    


  • Gomez-Pinilla, F. and T.T. Nguyen, 2012. Natural mood foods: The actions of polyphenols against psychiatric and cognitive disorders. Nutr. Neurosci., 15: 127-133.
    CrossRef    Direct Link    


  • Anhe, F.F., Y. Desjardins, G. Pilon, S. Dudonne, M.I. Genovese, F.M. Lajolo and A. Marette, 2013. Polyphenols and type 2 diabetes: A prospective review. Pharm. Nut., 1: 105-114.
    Direct Link    


  • Hakkinen, S., M. Heinonen, S. Karenlampi, H. Mykkanen, J. Ruuskanen and A.R. Torronen, 1999. Screening of selected flavonoids and phenolic acids in 19 berries. Food Res. Int., 32: 345-353.
    CrossRef    Direct Link    


  • Gil, M.I., F.A. Tomas-Barberan, B. Hess-Pierce, D.M. Holcroft and A.A. Kader, 2000. Antioxidant activity of pomegranate Juice and its relationship with phenolic composition and processing. J. Agric. Food Chem., 48: 4581-4589.
    CrossRef    PubMed    Direct Link    


  • Hartman, R.E., A. Shah, A.M. Fagan, K.E. Schwetye and M. Parsadanian et al., 2006. Pomegranate juice decreases amyloid load and improves behavior in a mouse model of Alzheimer's disease. Neurobiol. Dis., 24: 506-515.
    CrossRef    Direct Link    


  • Nantitanon, W., S. Yotsawimonwat and S. Okonogi, 2010. Factors influencing antioxidant activities and total phenolic content of guava leaf extract. LWT-Food Sci. Technol., 43: 1095-1103.
    CrossRef    Direct Link    


  • Landete, J.M., 2011. Ellagitannins, ellagic acid and their derived metabolites: A review about source, metabolism, functions and health. Food Res. Int., 44: 1150-1160.
    CrossRef    Direct Link    


  • Singh, K., A.K. Khanna and R. Chandan, 1999. Hepatoprotective activity of ellagic acid against carbon tetrachloride induced hepatotoxicity in rats. Ind. J. Exp. Biol., 37: 1025-1026.
    Direct Link    


  • Atessahin, A., A.O. Ceribasi, A. Yuce, O. Bulmus and G. Cikim, 2007. Role of ellagic acid against cisplatin-induced nephrotoxicity and oxidative stress in rats. Basic Clin. Pharmacol. Toxicol., 100: 121-126.
    CrossRef    Direct Link    


  • Yüce, A., A. Ateşşahin, A.O. Çeribaşı and M. Aksakal, 2007. Ellagic acid prevents cisplatin-induced oxidative stress in liver and heart tissue of rats. Basic Clin. Pharmacol. Toxicol., 101: 345-349.
    CrossRef    PubMed    Direct Link    


  • Chao, P.C., C.C. Hsu and M.C. Yin, 2009. Anti-inflammatory and anti-coagulatory activities of caffeic acid and ellagic acid in cardiac tissue of diabetic mice. Nutr. Metab. (Lond), Vol. 6.
    CrossRef    


  • Ozkaya, A., S. Celik, A. Yuce, Z. Sahin and O. Yilmaz, 2010. The effects of ellagic acid on some biochemical parameters in the liver of rats against oxidative stress induced by aluminum. Kafkas Univ. Vet. Fak. Derg., 16: 263-268.
    Direct Link    


  • Turk, G., M. Sonmez, A.O. Ceribasi, A. Yuce and A. Atessahin, 2010. Attenuation of cyclosporine A-induced testicular and spermatozoal damages associated with oxidative stress by ellagic acid. Int. Immunopharmacol., 10: 177-182.
    Direct Link    


  • Papoutsi, Z., E. Kassi, I. Chinou, M. Halabalaki, L.A. Skaltsounis and P. Moutsatsou, 2008. Walnut extract (Juglans regia L.) and its component ellagic acid exhibit anti-inflammatory activity in human aorta endothelial cells and osteoblastic activity in the cell line KS483. Brit. J. Nutr., 99: 715-722.
    CrossRef    Direct Link    


  • Bae, J.Y., J.S. Choi, S.W. Kang, Y.J. Lee, J. Park and Y.H. Kang, 2010. Dietary compound ellagic acid alleviates skin wrinkle and inflammation induced by UV-B irradiation. Exp. Dermatol., 19: e182-e190.
    CrossRef    Direct Link    


  • Umesalma, S. and G. Sudhandiran, 2010. Differential inhibitory effects of the polyphenol ellagic acid on inflammatory mediators NF‐κB, iNOS, COX‐2, TNF‐α, and IL‐6 in 1, 2‐dimethylhydrazine‐induced rat colon carcinogenesis. Basic Clin. Pharmacol. Toxicol., 107: 650-655.
    Direct Link    


  • Rosillo, M.A., M. Sanchez-Hidalgo, A. Cardeno, M. Aparicio-Soto, S. Sanchez-Fidalgo, I. Villegas and C.A. de la Lastra, 2012. Dietary supplementation of an ellagic acid-enriched pomegranate extract attenuates chronic colonic inflammation in rats. Pharmacol. Res., 66: 235-242.
    CrossRef    Direct Link    


  • Favarin, D.C., M.M. Teixeira, E.L. de Andrade, C.F. Alves and J.E.L. Chica et al., 2013. Anti-inflammatory effects of ellagic acid on acute lung injury induced by acid in mice. Mediators Inflamm.
    CrossRef    


  • Malik, A., S. Afaq, M. Shahid, K. Akhtar and A. Assiri, 2011. Influence of ellagic acid on prostate cancer cell proliferation: A caspase dependent pathway. Asian J. Trop. Med., 4: 550-555.
    CrossRef    PubMed    


  • Srigopalram, S., S. Ilavenil and I.A. Jayraaj, 2012. Apoptosis associated inhibition of DEN-induced hepatocellular carcinogenesis by ellagic acid in experimental rats. Biomed. Prevent. Nutr., 2: 1-8.
    Direct Link    


  • Umesalma, S. and G. Sudhandiran, 2011. Ellagic acid prevents rat colon carcinogenesis induced by 1, 2 dimethyl hydrazine through inhibition of AKT-phosphoinositide-3 kinase pathway. Eur. J. Pharmacol., 660: 249-258.
    CrossRef    Direct Link    


  • Qiu, Z., B. Zhou, L. Jin, H. Yu and L. Liu et al., 2013. In vitro antioxidant and antiproliferative effects of ellagic acid and its colonic metabolite, urolithins, on human bladder cancer T24 cells. Food Chem. Toxicol., 59: 428-437.
    CrossRef    PubMed    Direct Link    


  • Zhao, M., S.N. Tang, J.L. Marsh, S. Shankar and R.K. Srivastava, 2013. Ellagic acid inhibits human pancreatic cancer growth in Balb c nude mice. Can. Lett., 337: 210-217.
    CrossRef    PubMed    Direct Link    


  • Malini, P., G. Kanchana and M. Rajadurai, 2011. Antibiabetic efficacy of ellagic acid in streptozotocininduced diabetes mellitus in albino Wistar rats. Asian J. Pharmaceut. Clin. Res., 4: 124-128.
    Direct Link    


  • You, Q., F. Chen, X. Wang, Y. Jiang and S. Lin, 2012. Anti-diabetic activities of phenolic compounds in muscadine against alpha-glucosidase and pancreatic lipase. LWT-Food Sci. Technol., 46: 164-168.
    CrossRef    Direct Link    


  • Akileshwari, C., G. Raghu, P. Muthenna, N.H. Mueller, P. Suryanaryana, J.M. Petrash and G.B. Reddy, 2014. Bioflavonoid ellagic acid inhibits aldose reductase: Implications for prevention of diabetic complications. J. Funct. Foods, 6: 374-383.
    Direct Link    


  • Kannan, M.M. and S.D. Quine, 2013. Ellagic acid inhibits cardiac arrhythmias, hypertrophy and hyperlipidaemia during myocardial infarction in rats. Metabolism, 62: 52-61.
    CrossRef    PubMed    Direct Link    


  • Rani, U.P., R. Kesavan, R. Ganugula, T. Avaneesh, U.P. Kumar, G.B. Reddy and M. Dixit, 2013. Ellagic acid inhibits PDGF-BB-induced vascular smooth muscle cell proliferation and prevents atheroma formation in streptozotocin-induced diabetic rats. J. Nutr. Biochem., 24: 1830-1839.
    Direct Link    


  • Hassoun, E.A., J. Vodhanel and A. Abushaban, 2004. The modulatory effects of ellagic acid and vitamin E succinate on TCDD-induced oxidative stress in different brain regions of rats after subchronic exposure. J. Biochem. Mol. Toxicol., 18: 196-203.
    CrossRef    PubMed    


  • Pavlica, S. and R. Gebhardt, 2005. Protective effects of ellagic and chlorogenic acids against oxidative stress in PC12 cells. Free Radic. Res., 39: 1377-1390.
    CrossRef    PubMed    Direct Link    


  • Shukitt-Hale, B., F.C. Lau, A.N. Carey, R.L. Galli, E.L. Spangler, D.K. Ingram and J.A. Joseph, 2008. Blueberry polyphenols attenuate kainic acid-induced decrements in cognition and alter inflammatory gene expression in rat hippocampus. Nutr. Neurosci., 11: 172-182.
    CrossRef    PubMed    Direct Link    


  • Tan, H.P., D.Z. Wong, S.K. Ling, C.H. Chuah and H.A. Kadir, 2012. Neuroprotective activity of galloylated cyanogenic glucosides and hydrolysable tannins isolated from leaves of Phyllagathis rotundifolia. Fitoterapia, 83: 223 -229.
    CrossRef    PubMed    Direct Link    


  • Uzar, E., H. Alp, M.U. Cevik, U. Firat, O. Evliyaoglu, A. Tufek and Y. Altun, 2012. Ellagic acid attenuates oxidative stress on brain and sciatic nerve and improves histopathology of brain in streptozotocin-induced diabetic rats. Neurol. Sci., 33: 567-574.
    CrossRef    PubMed    Direct Link    


  • Gaire, B.P., N. Jamarkattel-Pandit, D. Lee, J. Song and J.Y. Kim et al., 2013. Terminalia chebula extract protects OGD-R induced PC12 cell death and inhibits LPS induced microglia activation. Molecules, 18: 3529-3542.
    CrossRef    Direct Link    


  • Rojanathammanee, L., K.L. Puig and C.K. Combs, 2013. Pomegranate polyphenols and extract inhibit nuclear factor of activated t-cell activity and microglial activation in vitro and in a transgenic mouse model of Alzheimer disease. J. Nutr., 143: 597-605.
    PubMed    Direct Link    


  • Feng, Y., S.G. Yang, X.T. Du, X. Zhang and X.X. Sun et al., 2009. Ellagic acid promotes Aβ42 fibrillization and inhibits Aβ42-induced neurotoxicity. Biochem. Biophys. Res. Commun., 390: 1250-1254.
    CrossRef    Direct Link    


  • Wilson, G.N., G.A. Mickley and K.M. Matera, 2010. The efficacy of ellagic acid in attenuating neurophysiological and cognitive-behavioral symptoms associated with infusion of amyloid-beta (Aβ) peptide fragments in adult rats. Baldwin-Wallace Coll. J. Res. Creat. Stud., 3: 15-30.


  • Sheean, P., M.K. Rout, R.J. Head and L.E. Bennett, 2012. Modulation of in vitro activity of zymogenic and mature recombinant human β-secretase by dietary plants. FEBS J., 279: 1291-1305.
    CrossRef    PubMed    Direct Link    


  • Messier, C. and M. Gagnon, 1996. Glucose regulation and cognitive functions: Relation to Alzheimer's disease and diabetes. Behav. Brain Res., 75: 1-11.
    Direct Link    


  • Beal, M.F., 2000. Energetics in the pathogenesis of neurodegenerative diseases. Trends Neurosci., 23: 298-304.
    Direct Link    


  • Dhingra, D., M. Parle and S.K. Kulkarni, 2003. Effect of combination of insulin with dextrose, D(-) fructose and diet on learning and memory in mice. Indian J. Pharmacol., 35: 151-156.
    Direct Link    


  • Mehan, S., R. Arora, D. Sharma, H. Meena, G. Sharma and T. Vyas, 2011. Dementia: A complete literature review on various mechanisms involves in pathogenesis and an intracerebroventricular streptozotocin induced Alzheimer's disease. Int. J. Pharm. Prof. Res., 2: 406-421.


  • Poulose, N., C.N.V. Prasad, P.A.N. Haridas and G. Anilkumar, 2011. Ellagic acid stimulates glucose transport in adipocytes and muscles through AMPK mediated pathway. J. Diabetes Metabol.
    CrossRef    


  • Makino-Wakagi, Y., Y. Yoshimura, Y. Uzawa, N. Zaima, T. Moriyama and Y. Kawamura, 2012. Ellagic acid in pomegranate suppresses resistin secretion by a novel regulatory mechanism involving the degradation of intracellular resistin protein in adipocytes. Biochem. Biophys. Res. Commun., 417: 880-885.
    CrossRef    PubMed    Direct Link    


  • Dhingra, D. and R. Chhillar, 2012. Antidepressant-like activity of ellagic acid in unstressed and acute immobilization-induced stressed mice. Pharmacol. Rep., 64: 796-807.
    CrossRef    PubMed    Direct Link    


  • Girish, C., V. Raj, J. Arya and S. Balakrishnan, 2013. Involvement of the GABAergic system in the anxiolytic-like effect of the flavonoid ellagic acid in mice. Eur. J. Pharmacol., 710: 49-58.
    CrossRef    PubMed    Direct Link    


  • Girish, C., V. Raj, J. Arya and S. Balakrishnan, 2012. Evidence for the involvement of the monoaminergic system, but not the opioid system in the antidepressant-like activity of ellagic acid in mice. Eur. J. Pharmacol., 682: 118-125.
    CrossRef    PubMed    Direct Link    


  • Friedman, J.I., D.N. Adler and K.L. Davis, 1999. The role of norepinephrine in the pathophysiology of cognitive disorders: Potential applications to the treatment of cognitive dysfunction in schizophrenia and Alzheimer's disease. Biol. Psychiatry, 46: 1243-1252.
    PubMed    Direct Link    


  • Brambilla, P., J. Perez, F. Barale, G. Schettini and J.C. Soares, 2003. GABAergic dysfunction in mood disorders. Mol. Psychiatry, 8: 721-737.
    CrossRef    PubMed    Direct Link    


  • Tatton, W., D. Chen, R. Chalmers-Redman, L. Wheeler, R. Nixon and N. Tatton, 2003. Hypothesis for a common basis for neuroprotection in glaucoma and Alzheimer's disease: Anti-apoptosis by alpha-2-adrenergic receptor activation. Surv. Ophthalmol., 48: S25-S37.
    PubMed    Direct Link    


  • Wenk, G.L., K. McGann, B. Hauss-Wegrzyniak and S. Rosi, 2003. The toxicity of tumor necrosis factor-alpha upon cholinergic neurons within the nucleus basalis and the role of norepinephrine in the regulation of inflammation: Implications for Alzheimer's disease. Neuroscience, 121: 719-729.
    CrossRef    PubMed    


  • Choudary, P.V., M. Molnar, S.J. Evans, H. Tomita and J.Z. Li et al., 2005. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc. Natl. Acad. Sci., 102: 15653-15658.
    CrossRef    PubMed    Direct Link    


  • Ciranna, L., 2006. Serotonin as a modulator of glutamate- and GABA-mediated neurotransmission: implications in physiological functions and in pathology. Curr. Neuropharmacol., 4: 101-114.
    PubMed    


  • Madsen, K., W.J. Neumann, K. Holst, L. Marner and M.T. Haahr et al., 2011. Cerebral serotonin 4 receptors and amyloid-β in early Alzheimer's disease. J. Alzheimers Dis., 26: 457-466.
    CrossRef    PubMed    


  • Xu, Y., J. Yan, P. Zhou , J. Li, H. Gao, Y. Xia and Q. Wang, 2012. Neurotransmitter receptors and cognitive dysfunction in Alzheimer's disease and Parkinson's disease. Prog. Neurobiol., 97: 1-13.
    CrossRef    PubMed    Direct Link    


  • Chalermpalanupap, T., B. Kinkead, W.T. Hu, M.P. Kummer and T. Hammerschmidt et al., 2013. Targeting norepinephrine in mild cognitive impairment and Alzheimer's disease. Alzheimers Res. Ther., Vol. 5.
    CrossRef    


  • Yu, J.T., N.D. Wang, T. Ma, H. Jiang, J. Guan and L. Tan, 2011. Roles of β-adrenergic receptors in Alzheimer's disease: Implications for novel therapeutics. Brain Res. Bull., 84: 111-117.
    CrossRef    PubMed    Direct Link    


  • Coutellier, L., P.M. Ardestani and M. Shamloo, 2014. β1-adrenergic receptor activation enhances memory in Alzheimer's disease model. Ann. Clin. Trans. Neurol., 1: 348-360.
    CrossRef    Direct Link    


  • Bierhaus, A., S. Schiekofer, M. Schwaninger, M. Andrassy and P.M. Humpert et al., 2001. Diabetes-associated sustained activation of the transcription factor nuclear factor-κB. Diabetes, 50: 2792-2808.
    CrossRef    Direct Link    


  • Cheng, X., J. Wu, M. Geng and J. Xiong, 2014. The role of synaptic activity in the regulation of amyloid beta levels in Alzheimer's disease. Neurobiol. Aging, 35: 1217-1232.
    CrossRef    Direct Link    


  • Spencer, Jr. D.G. and H. Lal, 1983. Effects of anticholinergic drugs on learning and memory. Drug Dev. Res., 3: 489-502.
    CrossRef    Direct Link    


  • Chen, K.C., M.G. Baxter and J.S. Rodefer, 2004. Central blockade of muscarinic cholinergic receptors disrupts affective and attentional set-shifting. Eur. J. Neurosci., 20: 1081-1088.
    CrossRef    Direct Link    


  • Wang, D., R. Yu and Y.Q. Lu, 2005. Protective effect of Pregnenolone sulfate against scopolamine induced memory impairment in an experimental animal model. Med. Hypotheses Res., 2: 295-302.


  • Terry, Jr. A.V., 2006. Muscarinic Receptor Antagonists in Rats. In: Animal Models of Cognitive Impairment, Levin, E.D. and J.J. Buccafusco (Eds.). CRC Press, Boca Raton, FL., ISBN-13: 9781420004335


  • Lee, Y.K., D.Y. Yuk, T.I. Kim, Y.H. Kim and K.T. Kim et al., 2009. Protective effect of the ethanol extract of Magnolia officinalis and 4-o-methylhonokiol on scopolamine-induced memory impairment and the inhibition of acetylcholinesterase activity. J. Nat. Med., 63: 274-282.
    CrossRef    Direct Link    


  • Kwon, S.H., H.K. Lee, J.A. Kim, S.I. Hong and H.C. Kim et al., 2010. Neuroprotective effects of chlorogenic acid on scopolamine-induced amnesia via anti-acetylcholinesterase and anti-oxidative activities in mice. Eur. J. Pharmacol., 649: 210-217.
    CrossRef    PubMed    Direct Link    


  • Liem-Moolenaar, M., P. de Boer, M. Timmers, R.C. Schoemaker and J.G. van Hasselt et al., 2011. Pharmacokinetic-pharmacodynamic relationships of central nervous system effects of scopolamine in healthy subjects. Br. J. Clin. Pharmacol., 71: 886-898.
    CrossRef    Direct Link    


  • Sahraei, M.S., E. Jafarzadeh and E.Z. Karimivaghef, 2012. Investigation of the scopolamine effect on acetylcholinesterase activity. Res. Pharmaceut. Sci., 7: 152-152.
    Direct Link    


  • Arafa, N.M.S., M. Abdel-Rahman and R.A.H.A. Mahmoud, 2013. Prophylactic effect of Hypericum perforatum L. extract in scopolamine rat model of cognitive dysfunction. Open Conf. Proc. J., 4: 23-30.


  • Kwon, S.H., S.X. Ma, H.J. Joo, S.Y. Lee and C.G. Jang, 2013. Inhibitory effects of Eucommia ulmoides Oliv. Bark on scopolamine-induced learning and memory deficits in mice. Biomol. Ther. (Seoul), 21: 462-469.
    CrossRef    Direct Link    


  • Tsukada, H., S. Yamazaki, A. Noda, T. Inoue and N. Matsuoka et al., 1999. FK960 [N-(4-acetyl-1-piperazinyl)-p-fluorobenzamide monohydrate], a novel potential antidementia drug, restores the regional cerebral blood flow response abolished by scopolamine but not by HA-966: A positron emission tomography study with unanesthetized rhesus monkeys. Brain Res., 832: 118-123.
    Direct Link    


  • Tsukada, H., T. Kakiuchi, I. Ando and Y. Ouchi, 1997. Functional activation of cerebral blood flow abolished by scopolamine is reversed by cognitive enhancers associated with cholinesterase inhibition: A positron emission tomography study in unanesthetized monkeys. J. Pharmacol. Exp. Ther., 281: 1408-1414.
    Direct Link    


  • Pachauri, S.D., S. Tota, K. Khandelwal, P.R.P. Verma and C. Nath et al., 2012. Protective effect of fruits of Morinda citrifolia L. on scopolamine induced memory impairment in mice: A behavioral, biochemical and cerebral blood flow study. J. Ethnopharmacol., 139: 34-41.
    CrossRef    PubMed    Direct Link    


  • Tota, S., C. Nath, A.K. Najmi, R. Shukla and K. Hanif, 2012. Inhibition of central angiotensin converting enzyme ameliorates scopolamine induced memory impairment in mice: Role of cholinergic neurotransmission, cerebral blood flow and brain energy metabolism. Behav. Brain Res., 232: 66-76.
    CrossRef    Direct Link    


  • Hebell, R., O. Shalev, W. Roker and B.H. Bank, 1986. Inhibition of erythrocyte Ca2+ ATPase by activated oxygen through thiol and lipid dependent mechanism. Biochim. Biophy. Acta, 862: 8-16.


  • El-Sherbiny, D.A., A.E. Khalifa, A.S. Attia and E.D. Eldenshary, 2003. Hypericum perforatum extract demonstrates antioxidant properties against elevated rat brain oxidative status induced by amnestic dose of scopolamine. Pharmacol. Biochem. Behav., 76: 525-533.
    CrossRef    PubMed    Direct Link    


  • Fan, Y., J. Hu, J. Li, Z. Yang and X. Xin et al., 2005. Effect of acidic oligosaccharide sugar chain on scopolamine-induced memory impairment in rats and its related mechanisms. Neurosci. Lett., 374: 222-226.
    CrossRef    PubMed    Direct Link    


  • Jeong, E.J., K.Y. Lee, S.H. Kim, S.H. Sung and Y.C. Kim, 2008. Cognitive-enhancing and antioxidant activities of iridoid glycosides from Scrophularia buergeriana in scopolamine-treated mice. Eur. J. Pharmacol., 588: 78-84.
    CrossRef    


  • Hancianu, M., O. Cioanca, M. Mihasan and L. Hritcu, 2013. Neuroprotective effects of inhaled lavender oil on scopolamine-induced dementia via anti-oxidative activities in rats. Phytomedicine, 20: 446-452.
    Direct Link    


  • Jain, N.K., C. Patil, S.K. Kulkarni and A. Singh, 2002. Modulatory role of cyclooxygenase inhibitors in aging- and scopolamine or lipopolysaccharide-induced cognitive dysfunction in mice. Behav. Brain Res., 133: 369-376.
    CrossRef    Direct Link    


  • Kim, S., D.H. Kim, J.J. Choi, J.G. Lee and C.H. Lee et al., 2009. Forsythiaside, a constituent of the fruits of Forsythia suspense, ameliorates scopolamine-induced memory impairment in mice. Biomol. Ther., 17: 249-255.
    Direct Link    


  • Lee, B.B., I.S. Shim, H.J. Lee and D.H. Hahm, 2011. Rehmannia glutinosa ameliorates scopolamine-induced learning and memory impairment in rats. J. Microbiol. Biotechnol., 21: 874-883.
    Direct Link    


  • Lee, B., B. Sur, I. Shim, H. Lee and D.H. Hahm, 2012. Phellodendron amurense and its major alkaloid compound, berberine ameliorates scopolamine-induced neuronal impairment and memory dysfunction in rats. Korean J. Physiol. Pharmacol., 16: 79-89.
    CrossRef    Direct Link    


  • Jang, Y.J., J. Kim, J. Shim, C.Y. Kim, J.H. Jang, K.W. Lee and H.J. Lee, 2013. Decaffeinated coffee prevents scopolamine-induced memory impairment in rats. Behav. Brain Res., 245: 113-119.
    Direct Link    


  • Ahmad, A., K. Ramasamy, S.M. Jaafar, A.B.A. Majeed and V. Mani, 2014. Total isoflavones from soybean and tempeh reversed Scopolamine-induced amnesia, improved cholinergic activities and reduced neuroinflammation in brain. Food Chem. Toxicol., 65: 120-128.
    CrossRef    Direct Link    


  • Abd-El-Fattah, M.A., N.F. Abdelakader and H.F. Zaki, 2014. Pyrrolidine dithiocarbamate protects against scopolamine-induced cognitive impairment in rats. Eur. J. Pharmacol., 723: 330-338.
    CrossRef    PubMed    Direct Link    


  • Haroutunian, V., N. Greig, X.F. Pei, T. Utsuki and R. Gluck et al., 1997. Pharmacological modulation of Alzheimer's β-amyloid precursor protein levels in the CSF of rats with forebrain cholinergic system lesions. Brain. Res. Mol. Brain Res., 46: 161-168.
    PubMed    


  • Liskowsky, W. and R. Schliebs, 2006. Muscarinic acetylcholine receptor inhibition in transgenic Alzheimer-like Tg2576 mice by scopolamine favours the amyloidogenic route of processing of amyloid precursor protein. Int. J. Dev. Neurosci., 24: 149-156.
    Direct Link    


  • Bihaqi, S.W., A.P. Singh and M. Tiwari, 2012. Supplementation of Convolvulus pluricaulis attenuates scopolamine-induced increased tau and Amyloid Precursor Protein (AβPP) expression in rat brain. Indian J. Pharmacol., 44: 593-598.
    CrossRef    Direct Link    


  • Preston, G.C., C. Brazell, C. Ward, P. Broks, M. Traub and S.M. Stahl, 1988. The scopolamine model of dementia: Determination of central cholinomimetic effects of physostigmine on cognition and biochemical markers in man. J. Psychopharmacol., 2: 67-79.
    CrossRef    Direct Link    


  • Wesnes, K., R. Anand and T. Lorscheid, 1990. Potential of moclobemide to improve cerebral insufficiency identified using a scopolamine model of aging and dementia. Acta Psychiatr. Scand., 360: 71-72.
    CrossRef    Direct Link    


  • Molchan, S.E., A.M. Mellow, B.A. Lawlor, H.J. Weingartner, R.M. Cohen, M.R. Cohen and T. Sunderland, 1990. TRH attenuates scopolamine-induced memory impairment in humans. Psychopharmacology, 100: 84-89.
    CrossRef    Direct Link    


  • Lines, C.R., J.H. Ambrose, A. Heald and M. Traub, 1993. A double-blind, placebo-controlled study of the effects of eptastigmine on scopolamine-induced cognitive deficits in healthy male subjects. Human Psychopharmacol.: Clin. Exp., 8: 271-278.
    CrossRef    Direct Link    


  • Gattu, M., K.L. Boss, A.V. Terry Jr. and J.J. Buccafusco, 1997. Reversal of scopolamine-induced deficits in navigational memory performance by the seed oil of Celastrus paniculatus. Pharmacol. Biochem. Behav., 57: 793-799.
    Direct Link    


  • Buccafusco, J.J., 2009. The Revival of Scopolamine Reversal for the Assessment of Cognition-Enhancing Drugs. In: Methods of Behavior Analysis in Neuroscience, Buccafusco, J.J. (Ed.). 2nd Edn., CRC Press, Boca Raton, FL


  • Rogers, J. and L.F. Lue, 2001. Microglial chemotaxis, activation and phagocytosis of amyloid β-peptide as linked phenomena in Alzheimer's disease. Neurology, 39: 333-340.
    Direct Link    


  • Sugimoto, H., Y. Yamanishi, Y. Iimura and Y. Kawakami, 2000. Donepezil Hydrochloride (E2020) and other acetylcholinesterase inhibitors. Curr. Med. Chem., 7: 303-339.
    PubMed    


  • Bartolini, M., C. Bertucci, V. Cavrini and V. Andrisano, 2003. β-Amyloid aggregation induced by human acetylcholinesterase: Inhibition studies. Biochem. Pharmacol., 65: 407-416.
    Direct Link    


  • Kimura, M., S. Akasofu, H. Ogura and K. Sawada, 2005. Protective effect of donepezil against Aβ(1-40) neurotoxicity in rat septal neurons. Brain Res., 1047: 72-84.
    Direct Link    


  • Kimura, M., H, Komatsu, H. Ogura and K. Sawada, 2005. Comparison of donepezil and memantine for protective effect against amyloid-β (1-42) toxicity in rat septal neurons. Neurosci. Lett., 391: 17-21.
    Direct Link    


  • Reale, M., C. Iarlori, F. Gambi, C. Feliciani, L. Isabella and D. Gambi, 2006. The acetylcholinesterase inhibitor, donepezil, regulates a Th2 bias in Alzheimer's disease patients. Neuropharmacology, 50: 606-613.
    PubMed    Direct Link    


  • Molino, I., L. Colucci, A.M. Fasanaro, E. Traini and F. Amenta, 2013. Efficacy of memantine, donepezil, or their association in moderate-severe Alzheimer's disease: A review of clinical trials. Sci. World J.
    CrossRef    


  • Yatabe, Y., M. Hashimoto, K. Kaneda, K. Honda, Y. Ogawa, S. Yuuki and M. Ikeda, 2013. Efficacy of increasing donepezil in mild to moderate Alzheimer's disease patients who show a diminished response to 5 mg donepezil: A preliminary study. Psychogeriatrics, 13: 88-93.
    CrossRef    Direct Link    


  • Schwarz, R.D., M.J. Callahan, R.E. Davis, J.C. Jaen and H. Tecle, 1997. Development of M1 subtype selective muscarinic agonists for Alzheimer's disease: Translation of in vitro selectivity into in vivo efficacy. Drug Dev. Res., 40: 133-143.
    CrossRef    Direct Link    


  • Riedel, G., S.H. Kang, D.Y. Choi and B. Platt, 2009. Scopolamine-induced deficits in social memory in mice: Reversal by donepezil. Behav. Brain Res., 204: 217-225.
    Direct Link    


  • Lindner, M.D., J.B. Hogan, D.B. Hodges Jr., A.F. Orie and P. Chen et al., 2006. Donepezil primarily attenuates scopolamine-induced deficits in psychomotor function, with moderate effects on simple conditioning and attention and small effects on working memory and spatial mapping. Psychopharmacology, 188: 629-640.
    CrossRef    Direct Link    


  • Agrawal, R., E. Tyagi, R. Shukla and C. Nath, 2008. Effect of insulin and melatonin on acetylcholinesterase activity in the brain of amnesic mice. Behav. Brain Res., 189: 381-386.
    CrossRef    PubMed    Direct Link    


  • Snyder, P.J., M.M. Bednar, J.R. Cromer and P. Maruff, 2005. Reversal of scopolamine-induced deficits with a single dose of donepezil, an acetylcholinesterase inhibitor. Alzheimer's Dementia, 1: 126-135.
    Direct Link    


  • Sumanth, M., H. Sowmya, S.V. Nagaraj and K. Narasimharaju, 2010. Efficacy of donepezil and galantamine in retrograde amnesia. Asian J. Pharm. Clin. Res., 3: 23-25.
    Direct Link    


  • Alkalay, A., G.D. Rabinovici, G. Zimmerman, N. Agarwal and D. Kaufer et al., 2013. Plasma acetylcholinesterase activity correlates with intracerebral β-amyloid load. Curr. Alzheimer Res., 10: 48-56.
    CrossRef    PubMed    Direct Link    


  • Sharma, M. and Y.K. Gupta, 2001. Intracerebroventricular injection of streptozotocin in rats produces both oxidative stress in the brain and cognitive impairment. Life Sci., 68: 1021-1029.
    Direct Link    


  • Morris, R., 1984. Developments of a water-maze procedure for studying spatial learning in the rat. J. Neurosci. Methods, 11: 47-60.
    CrossRef    PubMed    Direct Link    


  • Kumar, A., S. Dogra and A. Prakash, 2009. Neuroprotective effects of Centella asiatica against intracerebroventricular colchicine-induced cognitive impairment and oxidative stress. Int. J. Alzheimer's Dis.,
    CrossRef    


  • Ellman, G.L., K.D. Courtney, V. Andres Jr. and R.M. Featherstone, 1961. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol., 7: 88-90.
    CrossRef    Direct Link    


  • Will, E.D., 1966. Mechanism of lipid peroxide formation in animal tissues. Biochem. J., 99: 667-676.
    Direct Link    


  • Ellman, G.L., 1959. Tissue sulfhydryl groups. Arch. Biochem. Biophys., 82: 70-77.
    CrossRef    PubMed    Direct Link    


  • Misra, H.P. and I. Fridovich, 1972. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J. Biol. Chem., 247: 3170-3175.
    CrossRef    PubMed    Direct Link    


  • Aebi, H., S.R. Wyss, B. Scherz and F. Skvaril, 1974. Heterogeneity of erythrocyte catalase II. Isolation and characterization of normal and variant erythrocyte catalase and their subunits. Eur. J. Biochem., 48: 137-145.
    Direct Link    


  • Annicchiarico, R., A. Federici, C. Pettenati and C. Caltagirone, 2007. Rivastigmine in Alzheimer's disease: Cognitive function and quality of life. Ther. Clin. Risk Manage., 3: 1113-1123.
    Direct Link    


  • Fisher, A., 2008. Cholinergic treatments with emphasis on m1 muscarinic agonists as potential disease-modifying agents for Alzheimer's disease. Neurotherapeutics, 5: 433-442.
    CrossRef    Direct Link    


  • Raskind, M.A., E.R. Peskind, T. Wessel and W. Yuan, 2000. Galantamine in AD A 6-month randomized, placebo-controlled trial with a 6-month extension. Neurology, 54: 2261-2268.
    Direct Link    


  • Rockwood, K., J. Mintzer, L. Truyen, T. Wessel and D. Wilkinson, 2001. Effects of a flexible galantamine dose in Alzheimer's disease: A randomised, controlled trial. J. Neurol. Neurosurgery Psychiatry, 71: 589-595.
    CrossRef    Direct Link    


  • Mahadevan, S. and Y. Park, 2008. Multifaceted therapeutic benefits of Ginkgo biloba L.: Chemistry, efficacy, safety and uses. J. Food Sci., 73: R14-R19.
    CrossRef    PubMed    Direct Link    


  • Goswami, S., A. Saoji, N. Kumar, V. Thawani, M. Tiwari and M. Thawani, 2011. Effect of Bacopa monnieri on cognitive functions in Alzheimer's disease patients. Int. J. Collaborat Res. Int. Med. Public Health, 3: 285-293.
    Direct Link    


  • Hajiaghaee, R. and S. Akhondzadeh, 2012. Herbal medicine in the treatment of Alzheimer's disease. J. Med. Plants, 11: 2-7.


  • Downey, L.A., J. Kean, F. Nemeh, A. Lau and A. Poll et al., 2013. An acute, double-blind, placebo-controlled crossover study of 320 mg and 640 mg doses of a special extract of Bacopa monnieri (CDRI 08) on sustained cognitive performance. Phytother. Res., 27: 1407-1413.
    CrossRef    Direct Link    


  • Canevelli, M., N. Adali, E. Kelaiditi, C. Cantet, P.J. Ousset and M. Cesari, 2014. Effects of Gingko biloba supplementation in Alzheimer's disease patients receiving cholinesterase inhibitors: Data from the ICTUS study. Phytomedicine, 21: 888-892.
    Direct Link    


  • Di Matteo, V. and E. Esposito, 2003. Biochemical and therapeutic effects of antioxidants in the treatment of Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Curr. Drug Targets CNS Neurol. Disord., 2: 95-107.
    PubMed    


  • McGhie, T.K., M.C. Walton, L.E. Barnett, R. Vather and H. Martin et al., 2007. Boysenberry and blackcurrant drinks increased the plasma antioxidant capacity in an elderly population but had little effect on other markers of oxidative stress. J. Sci. Food Agric., 87: 2519-2527.
    CrossRef    Direct Link    


  • Zhou, C., Y. Huang and S. Przedborski, 2008. Oxidative stress in Parkinson's disease: A mechanism of pathogenic and therapeutic significance. Ann. New York Acad. Sci., 1147: 93-104.
    CrossRef    Direct Link    


  • Beninger, R.J., K. Jhamandas, R.J. Boegman and S.R. El-Defrawy, 1986. Effects of scopolamine and unilateral lesions of the basal forebrain on T-maze spatial discrimination and alternation in rats. Pharmacol. Biochem. Behav., 24: 1353-1360.
    Direct Link    


  • Smith, G., 1988. Animal models of Alzheimer's disease: Experimental cholinergic denervation. Brain Res. Rev., 13: 103-118.
    Direct Link    


  • Ennaceur, A. and K. Meliani, 1992. Effects of physostigmine and scopolamine on rat's performances in object-recognition and radial-maze tests. Psychopharmacology, 109: 321-330.
    CrossRef    Direct Link    


  • Wolff, M., N. Benhassine, P. Costet, R. Hen, L. Segu and M.C. Buhot, 2003. Delay-dependent working memory impairment in young-adult and aged 5-HT1BKO mice as assessed in a radial-arm water maze. Learn. Memory, 10: 401-409.
    Direct Link    


  • Carballo-Marquez, A., A. Vale-Martinez, G. Guillazo-Blanch, M. Torras-Garcia, N. Boix-Trelis and M. Marti-Nicolovius, 2007. Differential effects of muscarinic receptor blockade in prelimbic cortex on acquisition and memory formation of anodor-reward task. Learn. Mem., 14: 616-624.
    PubMed    


  • Halder, S., A.K. Mehta, R. Kar, M. Mustafa, P.K. Mediratta and K.K. Sharma, 2011. Clove oil reverses learning and memory deficits in scopolamine-treated mice. Planta Med., 77: 830-834.
    CrossRef    Direct Link    


  • Ebert, U. and W. Kirch, 1998. Scopolamine model of dementia: Electroencephalogram findings and cognitive performance. Eur. J. Clin. Invest., 28: 944-949.
    CrossRef    PubMed    Direct Link    


  • Itoh, J., T. Nabeshima and T. Kameyama, 1990. Utility of an elevated plus-maze for the evaluation of memory in mice: Effects of nootropics, scopolamine and electroconvulsive shock. Psychopharmacology, 101: 27-33.
    PubMed    Direct Link    


  • Miyazaki, S., M. Imaizumi and K. Onodera, 1995. Ameliorating effects of histidine on scopolamine-induced learning deficits using an elevated plus-maze test in mice. Life Sci., 56: 1563-1570.
    Direct Link    


  • Kruk-Słomka, M., B. Budzynska and G. Biała, 2012. Involvement of cholinergic receptors in the different stages of memory measured in the modified elevated plus maze test in mice. Pharmacol. Rep., 64: 1066-1080.
    Direct Link    


  • D'Hooge, R. and P.P. De Deyn, 2001. Applications of the morris water maze in the study of learning and memory. Brain Res. Rev., 36: 60-90.
    CrossRef    PubMed    Direct Link    


  • Wisman, L.A., G. Sahin, M. Maingay, G. Leanza and D. Kirik, 2008. Functional convergence of dopaminergic and cholinergic input is critical for hippocampus-dependent working memory. J. Neurosci., 28: 7797-7807.
    Direct Link    


  • Hosseini-Sharifabad, A., S. Mohammadi-Eraghi, K. Tabrizian, M. Soodi and T. Khorshidahmad et al., 2011. Effects of training in the Morris water maze on the spatial learning acquisition and VAChT expression in male rats. Daru: J. Fac. Pharm. Tehran Univ. Med. Sci., 19: 166-172.
    PubMed    Direct Link    


  • Ma, Y., S. Wang, Y. Tian, L. Chen, G. Li and J. Mao, 2013. Disruption of persistent nociceptive behavior in rats with learning impairment. PloS One, Vol. 8.
    CrossRef    


  • Wan, D., L. Xue, H. Zhu and Y. Luo, 2013. Catalpol induces neuroprotection and prevents memory dysfunction through the cholinergic system and BDNF. Evid. Based Complementary Alternative Med., Vol. 2013.
    CrossRef    


  • Francis, P.T., A.M. Palmer, M. Snape and G.K. Wilcock, 1999. The cholinergic hypothesis of Alzheimer's disease: A review of progress. J. Neurol. Neurosurg. Psychiatry, 66: 137-147.
    PubMed    Direct Link    


  • Deshmukh, R., V. Sharma, S. Mehan, N. Sharma and K.L. Bedi, 2009. Amelioration of intracerebroventricular streptozotocin induced cognitive dysfunction and oxidative stress by vinpocetine: A PDE1 inhibitor. Eur. J. Pharmacol., 620: 49-56.
    PubMed    Direct Link    


  • Sharma, V. and R. Deshmukh, 2012. Tumor necrosis factor and alzheimer's disease: A cause and consequence relationship. Bull. Clin. Psychopharmacol., 22: 86-97.
    Direct Link    


  • Pratico, D. and N. Delanty, 2000. Oxidative injury in diseases of the central nervous system: Focus on Alzheimer's disease. Am. J. Med., 109: 577-585.
    CrossRef    PubMed    Direct Link    


  • Zana, M., Z. Janka and J. Kalman, 2007. Oxidative stress: A bridge between Down's syndrome and Alzheimer's disease. Neurobiol. Aging, 28: 648-676.
    Direct Link    


  • Yan, Z. and J. Feng, 2004. Alzheimer's disease: Interactions between cholinergic functions and β-amyloid. Curr. Alzheimer Res., 1: 241-248.
    CrossRef    Direct Link    


  • Smith, M.A., C.A. Rottkamp, A. Nunomura, A.K. Raina and G. Perry, 2000. Oxidative stress in Alzheimer's disease. Biochim. Biophys. Acta Mol. Basis Dis., 1502: 139-144.
    CrossRef    PubMed    Direct Link    


  • McGrath, L.T., B.M. McGleenon, B. Brennan, D. McColl, S. McILroy and A.P. Passmore, 2001. Increased oxidative stress in Alzheimer's disease as assessed with 4-hydroxynonenal but not malondialdehyde. Q. J. Med., 94: 485-490.
    CrossRef    Direct Link    


  • Perry, G., A.D. Cash and M.A. Smith, 2002. Alzheimer disease and oxidative stress. BioMed Res. Int., 2: 120-123.
    CrossRef    


  • Gella, A. and N. Durany, 2009. Oxidative stress in Alzheimer disease. Cell Adhesion Migration, 3: 88-93.
    CrossRef    Direct Link    


  • Balu, M., P. Sangeetha, D. Haripriya and C. Panneerselvam, 2005. Rejuvenation of antioxidant system in central nervous system of aged rats by grape seed extract. Neurosci. Lett., 383: 295-300.
    Direct Link    


  • Attrey, D.P., A.K. Singh, T. Naved and B. Roy, 2012. Effect of seabuckthorn extract on scopolamine induced cognitive impairment. Indian J. Exp. Biol., 50: 690-695.
    Direct Link    


  • Lobo, V., A. Patil, A. Phatak and N. Chandra, 2010. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev., 4: 118-126.
    CrossRef    PubMed    Direct Link    


  • Rahman, K., 2007. Studies on free radicals, antioxidants and co-factors. Clin. Interv. Aging, 2: 219-236.
    Direct Link    


  • Mangialasche, F., M.C. Polidori, R. Monastero, S. Ercolani, C. Camarda, R. Cecchetti and P. Mococci, 2009. Biomarkers of oxidative and nitrosative damage in Alzheimer's disease and mild cognitive impairment. Ageing Res. Rev., 8: 285-305.
    CrossRef    


  • Uttara, B., A.V. Singh, P. Zamboni and R.T. Mahajan, 2009. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol., 7: 65-74.
    CrossRef    PubMed    Direct Link    


  • Younes, M. and C.P. Siegers, 1981. Mechanistic aspects of enhanced lipid peroxidation following glutathione depletion in vivo. Chem. Biol. Interact., 34: 257-266.
    CrossRef    


  • Casini, A.F., A. Pompella and M. Comporti, 1985. Liver glutathione depletion induced by bromobenzene, iodobenzene and diethylmaleate poisoning and its relation to lipid peroxidation and necrosis. Am. J. Pathol., 118: 225-237.
    Direct Link    


  • Schuessel, K., S. Leutner, N.J. Cairns, W.E. Muller and A. Eckert, 2004. Impact of gender on upregulation of antioxidant defence mechanisms in Alzheimer's disease brain. J. Neural Transmission, 111: 1167-1182.
    CrossRef    Direct Link    


  • Desagher, S., J. Glowinski and J. Premont, 1996. Astrocytes protect neurons from hydrogen peroxide toxicity. J. Neurosci., 16: 2553-2562.
    Direct Link    


  • Mulier, B., I. Rahman, T. Watchorn, K. Donaldson, W. MacNee and P.K. Jeffery, 1998. Hydrogen peroxide-induced epithelial injury: The protective role of intracellular nonprotein thiols (NPSH). Eur. Respiratory J., 11: 384-391.
    Direct Link    


  • Dringen, R., J.M. Gutterer and J. Hirrlinger, 2000. Glutathione metabolism in brain metabolic interaction between astrocytes and neurons in the defense against reactive oxygen species. Eur. J. Biochem., 267: 4912-4916.
    CrossRef    Direct Link    


  • Mann, H., M.T. McCoy, J. Subramaniam, H. Van Remmen and J.L. Cadet, 1997. Overexpression of superoxide dismutase and catalase in immortalized neural cells: Toxic effects of hydrogen peroxide. Brain Res., 770: 163-168.
    Direct Link    


  • Blockland, A., 1995. Acetylcholine: A neurotransmitter for learning and memory? Brain Res. Rev., 21: 285-300.
    Direct Link    


  • Oda, Y., 1999. Choline acetyltransferase: The structure, distribution and pathologic changes in the central nervous system. Pathology, 49: 921-937.
    CrossRef    Direct Link    

  • © Science Alert. All Rights Reserved