Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Food Funct ; 13(16): 8474-8488, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35861716

ABSTRACT

The rich and diverse phytoconstituents of wheatgrass have established it as a natural antioxidant and detoxifying agent. The anti-inflammatory potential of wheatgrass has been studied extensively. However, the neuroprotective potential of wheatgrass has not been studied in depth. In this study, we investigated the neuroprotective response of wheatgrass against age-related scopolamine-induced amnesia in mice. Scopolamine is an established anticholinergic drug that demonstrates the behavioural and molecular characteristics of Alzheimer's disease. In the current study, wheatgrass extracts (prepared from 5 and 7 day old plantlets) were administered to scopolamine-induced memory deficit mice. The Morris water maze (MWM) and Y-maze tests demonstrated that wheatgrass treatment improves the behavior and simultaneously enhances the memory of amnesic mice. We further evaluated the expression of neuroinflammation related genes and proteins in the hippocampal region of mice. Wheatgrass significantly upregulated the mRNA and protein expression of neuroprotective markers such as BDNF and CREB in scopolamine-induced mice. Simultaneously, wheatgrass also downregulated the expression of inflammatory markers such as TNF-α and tau genes in these mice. The treatment of scopolamine-induced memory impaired mice with wheatgrass resulted in an elevation in the level of the phosphorylated form of ERK and Akt proteins. Wheatgrass treatment of mice also regulated the phosphorylation of tau protein and simultaneously prevented its aggregation in the hippocampal region of the brain. Overall, this study suggests the therapeutic potential of wheatgrass in the treatment of age-related memory impairment, possibly through the involvement of ERK/Akt-CREB-BDNF pathway and concomitantly ameliorating the tau-related pathogenesis.


Subject(s)
Neuroprotective Agents , Scopolamine , Acetylcholinesterase/metabolism , Amnesia/chemically induced , Amnesia/drug therapy , Amnesia/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Maze Learning , Memory Disorders/drug therapy , Mice , Neuroprotective Agents/therapeutic use , Plant Extracts/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Scopolamine/adverse effects , Scopolamine/metabolism
2.
Metab Brain Dis ; 36(5): 901-910, 2021 06.
Article in English | MEDLINE | ID: mdl-33651274

ABSTRACT

Memory disorders are a result of a number of factors, of which elevated brain oxidative stress and acetylcholinesterase (AChE) activity are significant hallmarks. A number of Citrus species have cognition-enhancing capacity mediated by their antioxidant and anti-cholinesterase activities. This study was designed to assess the cognitive-enhancing, antioxidant and anticholinesterase potentials of Citrus reticulata var. kinnow (CR) leaf extracts. CR extracts were examined by bioactivity guided fractionation using in-vitro DPPH and Ellman assays to determine antioxidant and AChE inhibitory capacity. The most active component was further evaluated for memory improvement effects using mouse model of scopolamine induced amnesia. Passive shock avoidance test and elevated plus maze test were employed to determine cognitive functions while brain biochemical parameters were measured to establish the neuroprotective mechanism. The methanol extract (ME) showed marked AChE inhibitory and antioxidant activities, therefore, it was fractionated. Comparative analysis of all obtained fractions revealed that ethylacetate fraction (EAF) was most active. Both ME and EAF improved cognitive dysfunction caused by scopolamine in mice by reducing TBARS levels and brain AChE activity. TLC densitometric studies showed appreciable levels of naringenin in ME (0.32 % w/w) and EAF (1.14 % w/w). The observed memory enhancement effects of ME and EAF could be attributed to their ability to inhibit AChE activity and antioxidant effects due to presence of flavonoids.


Subject(s)
Amnesia/drug therapy , Citrus , Cognition/drug effects , Memory/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Acetylcholinesterase/metabolism , Amnesia/chemically induced , Amnesia/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Female , Male , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Plant Leaves
3.
Mol Med ; 26(1): 107, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33183243

ABSTRACT

BACKGROUND: The expression of SIN3A is closely correlated with electroacupuncture (EA) treatment efficacy of scopolamine-induced amnesia (SIA), but its underlying mechanisms remain to be further explored. METHODS: Quantitative real-time PCR was performed to analyze the expression of candidate microRNAs (miRNAs) and SIN3A mRNA in a rat model of SIA. Western blot was carried out to evaluate the differential expression of SIN3A proteins under different circumstances. Luciferase assay was used to explore the inhibitory role of certain miRNAs in SIN3A expression. A novel object recognition (NOR) test was performed to assess the memory function of SIA rats undergoing EA treatment. Immunohistochemistry was carried out to evaluate the expression of SIN3A in the hippocampus of SIA rats. RESULTS: Rno-miR-183-5p, rno-miR-34c-3p and rno-miR-210-3p were significantly up-regulated in SIA rats treated with EA. In addition, rno-miR-183-5p and rno-miR-210-3p exerted an inhibitory effect on SIN3A expression. EA treatment of SIA rats effectively restored the dysregulated expression of rno-miR-183-5p, rno-miR-210-3p and SIN3A. EA treatment also promoted the inhibited expression of neuronal IEGs including Arc, Egr1, Homer1 and Narp in the hippocampus of SIA rats. Accordingly, the NOR test also confirmed the effect of EA treatment on the improvement of memory in SIA rats. CONCLUSION: In summary, the findings of this study demonstrated that scopolamine-induced amnesia was associated with downregulated expression of miR-210/miR-183 and upregulated expression of SIN3A. Furthermore, treatment with EA alleviated scopolamine-induced amnesia in rats and was associated with upregulated expression of miR-210/miR-183 and downregulated expression of SIN3A.


Subject(s)
Amnesia/etiology , Electroacupuncture , MicroRNAs/genetics , Scopolamine/adverse effects , Sin3 Histone Deacetylase and Corepressor Complex/genetics , 3' Untranslated Regions , Amnesia/diagnosis , Amnesia/metabolism , Amnesia/therapy , Animals , Cell Line , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Electroacupuncture/methods , Gene Expression Regulation , Memory , Protein Binding , Rats
4.
J Nutr Biochem ; 82: 108394, 2020 08.
Article in English | MEDLINE | ID: mdl-32454411

ABSTRACT

Omega-3 polyunsaturated fatty acids (PUFA) are critical for optimal brain health and are involved in psychiatric and neurological ailments. Here, we report the effects of higher endogenous omega-3 PUFA on memory impairment in the hippocampus by studying mice with transgenic expression of the fat-1 gene that converts omega-6 to omega-3 PUFA. We performed Y-maze and passive avoidance tests to evaluate the memory function of fat-1 mice treated with scopolamine. Fat-1 mice showed induced alternation in the Y-maze test and increased latency in the passive avoidance test. The effects of scopolamine on hippocampal neurogenesis were confirmed by increases in the number of Ki-67- and DCX-positive cells in the fat-1 mice. Western blotting revealed increased brain-derived neurotrophic factor (BDNF) and phosphorylated cAMP response element-binding protein levels, and lower scopolamine-induced apoptosis based on the cleaved-caspase 3 protein level in fat-1 mice. These findings suggest that higher endogenous omega-3 PUFA prevented granular cell loss, increased BDNF signaling, and decreased apoptosis signaling in scopolamine-treated fat-1 mice. These processes may underlie granular cell survival and suggest potential therapeutic targets for memory impairment.


Subject(s)
Amnesia/metabolism , Cadherins/metabolism , Fatty Acids, Omega-3/pharmacology , Hippocampus/drug effects , Memory/drug effects , Scopolamine/adverse effects , Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Amnesia/chemically induced , Amnesia/drug therapy , Animals , Apoptosis/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Doublecortin Protein , Fatty Acids, Omega-3/administration & dosage , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology
5.
Behav Brain Res ; 356: 18-40, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30118774

ABSTRACT

Alzheimer's disease (AD) is an enervating and chronic progressive neurodegenerative disorder, occurring frequently in the elderly and adversely affecting intellectual capabilities and the cognitive processes. Bergenin possesses efficacious antioxidant, antiulcerogenic, anti-HIV, hepatoprotective, neuroprotective, anti-inflammatory and immunomodulatory activity along with antinociceptive effect and wound healing properties. Previous studies have shown that bergenin has in vitro bovine adrenal tyrosine hydroxylase inhibitory activity, mushroom tyrosinase inhibitory activities, ß-secretase (BACE-1) enzyme inhibitory activity and prevented neuronal death in the primary culture of rat cortical neurons. Protein tyrosine phosphatase-1B (PTP1B) is an intriguing target for anticancer and antidiabetic drugs and has recently been implicated to act as a positive regulator of neuroinflammation. Bergenin is also found to inhibit human protein tyrosine phosphatase-1B (hPTP1B) in vitro. Thus, bergenin was screened by molecular docking study using GOLD suite (version 5.2), CCDC for predicting its activity against targets of AD management like acetylcholinesterase (AChE) (1B41), butyrylcholinesterase (BuChE) (1P0I), Tau protein kinase 1 (GSK-3ß) (1J1B), BACE-1 (1FKN) wherein the GOLD score and fitness of bergenin were comparable to those of standard drugs like donepezil, galanthamine, physostigmine, etc. Bergenin demonstrated dose-dependent inhibition of both AChE and BuChE in vitro and found to be safe up to 50 µM when screened in vitro on SH-SY5Y cell lines by cytotoxicity studies using MTT and Alamar blue assays. It also led to dose-dependent prevention of NMDA induced toxicity in these cells. Pretreatment with bergenin (14 days) in rats at three dose levels (20, 40 and 80 mg/kg; p.o.) significantly (p < 0.01) and dose-dependently alleviated amnesia induced by scopolamine (2 mg/kg, i.p.). The therapeutic effect of bergenin supplementation for 28 days, at three dose levels, was also evaluated in streptozotocin (3 mg/kg, ICV, unilateral) induced AD model in Wistar rats using Morris water maze and Y maze on 7th, 14th, 21st and 28th days. STZ caused significant (p < 0.001) cognitive impairment and cholinergic deficit and increased oxidative stress in rats. Bergenin could significantly ameliorate STZ induced behavioral deficits, inhibit the AChE and BuChE activity in parallel with an increase in the diminished GSH levels in a dose-dependent fashion. The histopathological investigations were also supportive of this datum. The bergenin treatment at 80 mg/kg led to significant (p < 0.05) abatement of the raised Aß-1-42 levels and alleviated the perturbed p- tau levels leading to significantly low (p < 0.01) levels of p-tau in brain homogenates of rats as compared to ICV STZ injected rats. In conclusion, the observed effects might be attributed to the cholinesterase inhibitory activity of bergenin coupled with its antioxidant effect, anti-inflammatory activity and reduction of Aß-1-42 and p-tau levels which could have collectively helped in the attenuation of cognitive deficits. The current findings of the study are indicative of the promising preventive and ameliorative potential of bergenin in the management of AD through multiple targets.


Subject(s)
Alzheimer Disease/drug therapy , Benzopyrans/metabolism , Benzopyrans/pharmacology , Acetylcholinesterase/metabolism , Alzheimer Disease/pathology , Amnesia/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Butyrylcholinesterase/metabolism , Cell Line , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Molecular Docking Simulation/methods , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Streptozocin/pharmacology
6.
Biomed Pharmacother ; 108: 374-390, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30227331

ABSTRACT

Bergenia ciliata (Haw) Sternb. possess immunomodulatory, anti-inflammatory, antioxidant, anti-urolithiatic, wound healing, anti-malarial, anti-diabetic and anti-cancer properties. Moreover, the methanolic extracts of the rhizomes of the plant were found to demonstrate beneficial neuroprotective effects in the intracerebroventricular streptozotocin-induced model in rats. Thus, the present study was undertaken to further explore the neuroprotective potential of the aqueous (BA) and methanolic extracts (BM) of B. ciliata through various in-vitro and in-vivo studies. Both the extracts at all tested concentrations i.e. 50-50,000 ng/mL did not cause any significant reduction of cell viability of SH-SY5Y cells when tested for 48 h when assessed through MTT and resazurin metabolism- based cell viability assays. The pre-treatment with the extracts could confer significant (p < 0.001) and dose-dependent protective effects against NMDA induced injury in SH-SY5Y cells. BM [IC50: 5.7 and 5.19 µg/mL for acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) respectively] led to more potent inhibition of both the enzymes as compared to BA (IC50: 227.12 and 23.25 µg/mL for AChE and BuChE respectively). BM also proved to be a 1.85-fold better scavenger of the DPPH free radicals as compared to BA. Thus, BM was taken further for the evaluation of the beneficial effects of 14-day pre-treatment in rats in the scopolamine (2 mg/kg, i.p.) induced amnesia model at 125, 250 and 500 mg/kg, p.o. BM pre-treatment at 250 and 500 mg/kg could significantly ameliorate the cognitive impairment (p < 0.001), inhibit AChE (p < 0.001) and BuChE (p < 0.05) activity, restore GSH levels (p < 0.05) in serum and brain homogenates and recover the morphology of hippocampal neurons back to normal. Moreover, the BM administration at 500 mg/kg also showed beneficial effects through the significant (p < 0.05) reduction of Aß1-42, phosphorylated tau (p-tau) and GSK-3ß immunoreactivity in the brain homogenates of the intracerebroventricularly streptozotocin (ICV STZ) injected rats as observed from the results of the ELISA assays. The outcomes of the study unveiled that BM exerts its beneficial effects through prevention of NMDA induced excitotoxic cell death, dual cholinesterase inhibition, antioxidant activity coupled with the reduction of the immunoreactivity for the Aß1-42, p-tau and GSK-3ß indicating its potential to be screened further for various other models to determine the exact mechanism of action.


Subject(s)
Amnesia/drug therapy , Cognitive Dysfunction/drug therapy , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Acetylcholinesterase/metabolism , Amnesia/chemically induced , Amnesia/metabolism , Animals , Antioxidants/pharmacology , Brain/drug effects , Butyrylcholinesterase/metabolism , Cell Line , Cell Survival/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Humans , Male , N-Methylaspartate/pharmacology , Neurons/drug effects , Rats , Rats, Wistar , Scopolamine/pharmacology , Streptozocin/pharmacology
7.
Biomed Pharmacother ; 105: 1344-1352, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30021372

ABSTRACT

Centella asiatica (CA) has been used by Ayurvedic medical practitioners in India for almost 3000 years. The neuropharmacological properties of CA and its constituents have been studied extensively. Anti-oxidant, free radical scavenging and cholinergic modulatory activities are the reported mechanisms of action for its efficacy in memory disorders. Its medicinal values are mainly attributed to the presence of several triterpenes, namely asiatic acid, madecassic acid, asiaticoside, and madecassoside. The present study was aimed to investigate the role of these triterpenes content in CA extract on the antioxidant, cholinesterase modulation and anti-amnesic properties. The fractions of CA extract enriched for (CAE-EF) and depleted/freed of (CAE-FF) triterpenes contents were compared with methanolic extract (CAE). Both in vitro and in vivo methods for evaluation of antioxidant and anticholinergic activities were used. In vitro, free radical scavenging assays (ABTS, DPPH, NO, NORAC, and ORAC) and cholinesterase (AChE and BuChE) inhibition assays were used. For evaluation of anti-amnesic effect, scopolamine induced amnesia in rats, as the acute model of memory loss was used. Following behavioural assessments (MWM, PA, EPM), biomarkers of oxidative stress (reduced GSH, MDA and SOD activity) and cholinesterase (AChE and BuChE) status were also estimated in cerebral cortex and hippocampus of rat brain. The methanolic extract (CAE) was found to perform best among all three fractions for in vitro free radical scavenging, cholinesterase inhibition, improvement of scopolamine-induced amnesia and also in vivo antioxidant effect and cholinesterase inhibitory activities. Interestingly triterpenes free fraction (CAE-FF) showed better antioxidant activity than triterpenes enriched fraction (CAE-EF) along with comparable anti-amnesic effect. This indicates that triterpenes are not solely responsible for antioxidant activity, cholinesterase inhibitory and anti-amnesic effect of CA.


Subject(s)
Amnesia/drug therapy , Antioxidants/pharmacology , Centella/chemistry , Cholinergic Antagonists/pharmacology , Cholinesterase Inhibitors/pharmacology , Triterpenes/pharmacology , Amnesia/metabolism , Animals , Cholinesterases/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Methanol/chemistry , Oxidants/pharmacology , Oxidative Stress/drug effects , Pentacyclic Triterpenes/pharmacology , Plant Extracts , Rats , Rats, Wistar , Scopolamine/pharmacology
8.
Indian J Pharmacol ; 50(1): 30-38, 2018.
Article in English | MEDLINE | ID: mdl-29861525

ABSTRACT

OBJECTIVE: Hydroethanolic extract of Zanthoxylum alatum seeds (HEZA) in scopolamine-induced amnesia was investigated for memory enhancing activity. MATERIALS AND METHODS: Radial arm maze (RAM) test was performed to evaluate the behavioral activity. Rats were treated with HEZA (50, 100, and 200 mg/kg, p. o.) and tacrine (3 mg/kg. i. p.) for 14 days. Scopolamine (0.4 mg/kg) was injected i. p. into rats after 45 min of drug administration on the 14th day. The messenger RNA (mRNA)/protein profile of few markers (acetylcholinesterase [AChE], heme oxygenase-1 [HO-1], nuclear factor-kappa B [NFκB], nuclear factor erythroid 2-related factor 2 [Nrf2], protein phosphatase 2A[PP2A], Tau, brain-derived neurotrophic factor [BDNF], tropomyosin-related kinase B [TrkB], Bcl-2-associated X protein [Bax], and Caspase-3) were also measured by polymerase chain reaction (PCR) and immunoblotting assay. Brain cytokines (tumor necrosis factor alpha [TNF-α], interleukin [IL]-1 ß, and IL-10) in hippocampus were evaluated using commercially available enzyme-linked immunosorbent assay kits. RESULTS: HEZA exhibited anti-amnesic activity as indicated by a significant reduction in the working memory error and reference memory error in RAM. Pretreatment with HEZA significantly down-regulated the expression of AChE, NFκB, Tau, Bax, and Caspase-3 with simultaneous up-regulation of Nrf2, HO-1, PP2A, BDNF, and TrkB genes in the hippocampal tissues similar to tacrine when compared with scopolamine-treated rats. Pretreatment with HEZA attenuated scopolamine-induced elevation of TNF-α, IL-1 ß, levels in hippocampus and reversed diminished IL-10 concentrations towards normal levels in the brain. CONCLUSION: Zanthoxylum alatum seeds could probably counteract amnesia. Since its use is mainly reported as a stimulant and tonic, this novel activity could be a boon for the scientists to explore more in this direction.


Subject(s)
Amnesia/drug therapy , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Zanthoxylum , Acetylcholinesterase/genetics , Amnesia/chemically induced , Amnesia/metabolism , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cytokines/metabolism , Heme Oxygenase (Decyclizing)/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/toxicity , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/toxicity , Protein Phosphatase 2/genetics , Rats, Wistar , Receptor, trkB/metabolism , Scopolamine , Seeds , Toxicity Tests, Acute , bcl-2-Associated X Protein/metabolism , tau Proteins/genetics
9.
BMC Complement Altern Med ; 18(1): 136, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29716575

ABSTRACT

BACKGROUND: Terminalia chebula Retz. (Combretaceae) is a traditional herbal medicine that is widely used in the treatment of diabetes, immunodeficiency diseases, and stomach ulcer in Asia. However, the anti-amnesic effect of T. chebula has not yet been investigated. The present study was designed to determine whether T. chebula extract (TCE) alleviates amnesia induced by scopolamine in mice. We also investigated possible mechanisms associated with cholinergic system and anti-oxidant effects. METHODS: TCE (100 or 200 mg/kg) was orally administered to mice for fourteen days (days 1-14), and scopolamine was intraperitoneally injected to induce memory impairment for seven days (days 8-14). Learning and memory status were evaluated using the Morris water maze. Hippocampal levels of acetylcholine (ACh), acetylcholinesterase (AChE) and choline acetyltransferase (ChAT) were measured ex vivo. Levels of reactive oxygen species (ROS), nitric oxide (NO), and malondialdehyde (MDA) in the hippocampus were also examined. RESULTS: In the Morris water maze task, TCE treatment reversed scopolamine-induced learning and memory deficits in acquisition and retention. TCE reduced hippocampal AChE activities and increased ChAT and ACh levels in the scopolamine-induced model. Moreover, TCE treatment suppressed scopolamine-induced oxidative damage by ameliorating the increased levels of ROS, NO, and MDA. CONCLUSION: These findings suggest that TCE exerts potent anti-amnesic effects via cholinergic modulation and anti-oxidant activity, thus providing evidence for its potential as a cognitive enhancer for amnesia.


Subject(s)
Amnesia/metabolism , Antioxidants/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Scopolamine/adverse effects , Terminalia/chemistry , Acetylcholine/analysis , Acetylcholine/metabolism , Acetylcholinesterase/analysis , Acetylcholinesterase/metabolism , Amnesia/chemically induced , Amnesia/prevention & control , Animals , Hippocampus/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL
10.
BMC Complement Altern Med ; 17(1): 367, 2017 Jul 17.
Article in English | MEDLINE | ID: mdl-28716085

ABSTRACT

BACKGROUND: Curcuma longa L. is a well-known medicinal plant that has been used for its anti-cancer, neuroprotective, and hepatoprotective effects. However, the neuroprotective effect of fermented C. longa (FCL) has not been reported. Therefore, in this study, the effectiveness of FCL for the regulation of memory dysfunction was investigated in two brain cell lines (rat glioma C6 and murine microglia BV2) and scopolamine-treated mice. METHODS: C. longa powder was fermented by 5% Lactobacillus plantarum K154 containing 2% (w/v) yeast extract at 30 °C for 72 h followed by sterilization at 121 °C for 15 min. The protective effects of fermented C. longa (FCL) on oxidative stress induced cell death were analyzed by MTT assay in C6 cells. The anti-inflammatory effects of FCL were investigated by measuring the production of nitric oxide (NO) and prostaglandin E2 (PGE2) as well as the expression levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-stimulated BV2 cells. The step-through passive avoidance test, Morris water maze test, acetylcholinesterase (AChE) activity, and expression of cAMP response element-binding protein (CREB) and brain-derived neurotropic factor (BDNF) were employed to determine the effects of FCL on scopolamine-induced memory deficit in mice. The contents of curcuminoids were analyzed through LC/MS. RESULTS: Pretreatment with FCL effectively prevented the cell death induced by oxidative stress in C6 cells. Moreover, FCL inhibited the production NO and PGE2 via the inhibition of iNOS and COX-2 expression in BV2 cells. FCL significantly attenuated scopolamine-induced memory impairment in mice and prevented scopolamine-induced AChE activity in the hippocampus. Additionally, FCL reversed the reduction of CREB and BDNF expression. The curcuminoids content in FCL was 1.44%. CONCLUSION: FCL pretreatment could alleviate scopolamine-induced memory impairment in mice, as well as oxidative stress and inflammation in C6 and BV2 cells, respectively. Thus, FCL might be a useful material for preventing impairment of learning and memory.


Subject(s)
Amnesia/drug therapy , Brain/drug effects , Curcuma/chemistry , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Phytotherapy , Acetylcholinesterase/metabolism , Amnesia/chemically induced , Amnesia/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Curcumin/analysis , Curcumin/pharmacology , Curcumin/therapeutic use , Cyclic AMP Response Element-Binding Protein/metabolism , Fermentation , Inflammation/chemically induced , Inflammation/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Memory Disorders , Mice, Inbred ICR , Neuroprotective Agents/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Scopolamine
11.
Food Funct ; 8(4): 1460-1467, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28275781

ABSTRACT

Aqueous extracts from Asparagus officinalis L. stems (AEAS) are rich in polysaccharides, gamma-amino butyric acid (GABA), and steroidal saponin. This study was designed to investigate the effects of AEAS on learning, memory, and acetylcholinesterase-related activity in a scopolamine-induced model of amnesia. Sixty ICR mice were randomly divided into 6 groups (n = 10) including the control group (CT), scopolamine group (SC), donepezil group (DON), low, medium, and high dose groups of AEAS (LS, MS, HS; 1.6 mL kg-1, 8 mL kg-1, 16 mL kg-1). The results showed that 8 mL kg-1 of AEAS used in this study significantly reversed scopolamine-induced cognitive impairments in mice in the novel object recognition test (P < 0.05) and the Y-maze test (P < 0.05), and also improved the latency to escape in the Morris water maze test (P < 0.05). Moreover, it significantly increased acetylcholine and inhibited acetylcholinesterase activity in the hippocampus, which was directly related to the reduction in learning and memory impairments. It also reversed scopolamine-induced reduction in the hippocampal brain-derived neurotrophic factor (BDNF) and the cAMP response element-binding protein (CREB) mRNA expression. AEAS protected against scopolamine-induced memory deficits. In conclusion, AEAS protected learning and memory function in mice by enhancing the activity of the cholinergic nervous system, and increasing BDNF and CREB expression. This suggests that AEAS has the potential to prevent cognitive impairments in age-related diseases, such as Alzheimer's disease.


Subject(s)
Amnesia/drug therapy , Asparagus Plant/chemistry , Memory/drug effects , Plant Extracts/administration & dosage , Scopolamine/administration & dosage , Amnesia/chemically induced , Amnesia/metabolism , Amnesia/psychology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred ICR
12.
Neurochem Res ; 42(5): 1333-1344, 2017 May.
Article in English | MEDLINE | ID: mdl-28097466

ABSTRACT

The present study was designed to explore the beneficial effects of successive 10 days administration of Trachyspermum ammi seed's powder (TASP) along with diet (at the dose of 0.5%, 1.0% and 2.0% w/w) on learning and memory of mice. A total of 306 mice divided in 51 equal groups were employed in the study. Passive avoidance paradigm (PAP) and Object recognition Task (ORT) were employed as exteroceptive models. The brain acetylcholinesterase activity (AChE), serum cholesterol, brain monoaldehyde (MDA), brain reduced glutathione (GSH) and brain nitrite were estimated and Alprazolam, Scopolamine and Electroshock induced amnesia was employed to describe the actions. Treatment of TASP significantly increased step down latency of PAA and significantly increased discrimination index of ORT in groups with or without amnesia when compared to respective control groups. Furthermore, TASP administration resulted in significant fall in brain AChE activity, brain MDA level and brain nitrite level with simultaneous rise in brain GSH level, thereby decreased oxidative damage. A significant decrease in serum cholesterol was also observed. Ajowan supplementation may prove a remedy for the management of cognitive disorders owing to have pro-cholinergic, antioxidant and hypo-lipidemic activities.


Subject(s)
Alprazolam/toxicity , Amnesia/drug therapy , Apiaceae , Electroshock/adverse effects , Plant Extracts/therapeutic use , Scopolamine/toxicity , Acetylcholinesterase/metabolism , Amnesia/etiology , Amnesia/metabolism , Animals , Dietary Supplements , Dose-Response Relationship, Drug , Glutathione/antagonists & inhibitors , Glutathione/metabolism , Male , Mice , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Seeds
13.
PLoS One ; 11(8): e0159823, 2016.
Article in English | MEDLINE | ID: mdl-27483466

ABSTRACT

We evaluated the neuropharmacological effects of Gongjin-Dan (GJD) on the memory impairment caused by scopolamine injection. BALB/c mice were orally treated with GJD (100, 200, or 400 mg/kg, daily) or tacrine (THA, 10 mg/kg) for 10 days, and scopolamine (2 mg/kg) was injected intraperitoneally. The radial arm maze and passive avoidance tests were performed to evaluate the animal's learning and memory. Scopolamine increased the task completing time, the number of total errors (reference and working memory error) in the radial arm maze task, and the latency time in the passive avoidance test, which were significantly ameliorated by treatment with GJD. The GJD treatment also attenuated the scopolamine-induced hyperactivation of acetylcholinesterase activity, and suppression of the expression of brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and their receptors in the hippocampus. These effects of GJD were supported by both the doublecortin (DCX)-positive staining and Nissl staining, which were used to measure hippocampal neurogenesis and atrophy, respectively. These findings strongly suggest that GJD exerts a potent anti-amnesic effect, and its underlying mechanism might involve the modulation of cholinergic activity.


Subject(s)
Amnesia/chemically induced , Amnesia/drug therapy , Hippocampus/drug effects , Memory/drug effects , Plant Extracts/therapeutic use , Scopolamine , Acetylcholinesterase/metabolism , Amnesia/metabolism , Amnesia/physiopathology , Animals , Avoidance Learning/drug effects , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Disease Models, Animal , Doublecortin Protein , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Nerve Growth Factor/analysis , Nerve Growth Factor/metabolism
14.
Biosci Biotechnol Biochem ; 80(1): 166-71, 2016.
Article in English | MEDLINE | ID: mdl-26287580

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease induced by cholinergic neuron damage or amyloid-beta aggregation in the basal forebrain region and resulting in cognitive disorder. We previously reported on the neuroprotective effects of Betula platyphylla bark (BPB) in an amyloid-beta-induced amnesic mouse model. In this study, we obtained a cognitive-enhancing compound by assessing results using a scopolamine-induced amnesic mouse model. Our results show that oral treatment of mice with BPB and betulin significantly ameliorated scopolamine-induced memory deficits in both passive avoidance and Y-maze tests. In the Morris water maze test, administration of BPB and betulin significantly improved memory and cognitive function indicating the formation of working and reference memories in treated mice. Moreover, betulin significantly increased glutathione content in mouse hippocampus, and the increase was greater than that from betulinic acid treatment. We conclude that BPB and its active component betulin have potential as therapeutic, cognitive enhancer in AD.


Subject(s)
Amnesia/drug therapy , Betula/chemistry , Memory/drug effects , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Triterpenes/pharmacology , Administration, Oral , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amnesia/chemically induced , Amnesia/metabolism , Amnesia/physiopathology , Animals , Avoidance Learning/drug effects , Cognition/drug effects , Disease Models, Animal , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Male , Maze Learning/drug effects , Mice , Neuroprotective Agents/isolation & purification , Nootropic Agents/isolation & purification , Pentacyclic Triterpenes , Plant Bark/chemistry , Plant Extracts/chemistry , Scopolamine , Triterpenes/isolation & purification , Betulinic Acid
15.
Mol Neurobiol ; 53(3): 1760-1769, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25744565

ABSTRACT

Our earlier report on scopolamine-induced amnesia and its improvement by pre-treatment with i-Extract (alcoholic extract of Ashwagandha leaf) suggested that the i-Extract mediated nootropic effect may involve neuronal immediate early gene, Arc. With a hypothesis that the i-Extract induced expression of Arc protein may cause augmentation in Arc function, we examined the effect of i-extract on a major function of Arc protein, i.e. F-actin expansion, using Arc antisense oligodeoxynucleotides (ODN). Stereotaxic infusion of Arc antisense ODN in the CA1 region of hippocampus decreased the level of Arc protein as demonstrated by immunoblotting. However, this decrease was attenuated when treated with i-Extract prior to infusion of Arc antisense ODN. We noted a significant decrease in the polymerization of F-actin during scopolamine-induced amnesia as well as Arc antisense ODN infusion that was restored rather enhanced when pre-treated with i-Extract in both the cases. We also compared the corresponding changes between CA1 (the infusion site) and CA3 (neighbouring site of infusion) regions of hippocampus, and found more pronounced effects in CA1 than in the CA3 region. The extent of F-actin polymerization, as revealed by changes in the dendritic spine architecture through Golgi staining, showed that both scopolamine as well as Arc antisense ODN disrupted the spine density and mushroom-shaped morphology that was again regained if pre-treated with i-Extract. In conclusion, the findings reveal that the Arc helps in polymerization of F-actin and subsequent changes in the morphology of dendritic spines after pre-treatment with i-Extract in scopolamine-induced amnesic mice, suggesting an important role of Arc in scopolamine-induced amnesia and its recovery by i-Extract.


Subject(s)
Alcohols/chemistry , Amnesia/drug therapy , Amnesia/metabolism , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Actins/metabolism , Animals , Blotting, Western , Dendritic Spines/drug effects , Dendritic Spines/pathology , Down-Regulation/drug effects , Fluorescent Antibody Technique , Hippocampus/pathology , Male , Mice , Neuroprotective Agents/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polymerization , Staining and Labeling , Stereotaxic Techniques
16.
Neurology ; 85(24): 2107-15, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26567269

ABSTRACT

OBJECTIVE: To improve current understanding of the mechanisms behind thalamic amnesia, as it is unclear whether it is directly related to damage to specific nuclei, in particular to the anterior or mediodorsal nuclei, or indirectly related to lesions of the mammillothalamic tract (MTT). METHODS: We recruited 12 patients with a left thalamic infarction and 25 healthy matched controls. All underwent a comprehensive neuropsychological assessment of verbal and visual memory, executive functions, language, and affect, and a high-resolution structural volumetric MRI scan. Thalamic lesions were manually segmented and automatically localized with a computerized thalamic atlas. As well as comparing patients with controls, we divided patients into subgroups with intact or damaged MTT. RESULTS: Only one patient had a small lesion of the anterior nucleus. Most of the lesions included the mediodorsal (n = 11) and intralaminar nuclei (n = 12). Patients performed worse than controls on the verbal memory tasks, but the 5 patients with intact MTT who showed isolated lesions of the mediodorsal nucleus (MD) only displayed moderate memory impairment. The 7 patients with a damaged MTT performed worse on the verbal memory tasks than those whose MTT was intact. CONCLUSIONS: Lesions in the MTT and in the MD result in memory impairment, severely in the case of MTT and to a lesser extent in the case of MD, thus highlighting the roles played by these 2 structures in memory circuits.


Subject(s)
Amnesia/diagnosis , Amnesia/etiology , Cerebral Infarction/complications , Cerebral Infarction/diagnosis , Mammillary Bodies/pathology , Mediodorsal Thalamic Nucleus/pathology , Adult , Aged , Amnesia/metabolism , Cerebral Infarction/metabolism , Female , Humans , Male , Mammillary Bodies/metabolism , Mediodorsal Thalamic Nucleus/metabolism , Middle Aged , Nerve Net/metabolism , Nerve Net/pathology , Neural Pathways/metabolism , Neural Pathways/pathology , Thalamus/metabolism , Thalamus/pathology
17.
Anesth Analg ; 121(3): 661-666, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26287296

ABSTRACT

BACKGROUND: Hyperpolarization-activated, cyclic nucleotide-gated (HCN) subtype 1 (HCN1) channels have been identified as targets of ketamine to produce hypnosis. Volatile anesthetics also inhibit HCN1 channels. However, the effects of HCN1 channels on volatile anesthetics in vivo are still elusive. This study uses global and conditional HCN1 knockout mice to evaluate how HCN1 channels affect the actions of volatile anesthetics. METHODS: Minimum alveolar concentrations (MACs) of isoflurane and sevoflurane that induced immobility (MAC of immobility) and/or hypnosis (MAC of hypnosis) were determined in wild-type mice, global HCN1 knockout (HCN1) mice, HCN1 channel gene with 2 lox-P sites flanking a region of the fourth exon of HCN1 (HCN1) mice, and forebrain-selective HCN1 knockout (HCN1: cre) mice. Immobility of mice was defined as no purposeful reactions to tail-clamping stimulus, and hypnosis was defined as loss of righting reflex. The amnestic effects of isoflurane and sevoflurane were evaluated by fear-potentiated startle in these 4 strains of mice. RESULTS: All MAC values were expressed as mean ± SEM. For MAC of immobility of isoflurane, no significant difference was found among wild-type, HCN1, HCN1, and HCN1: cre mice (all ~1.24%-1.29% isoflurane). For both HCN1 and HCN1: cre mice, the MAC of hypnosis for isoflurane (each ~1.05% isoflurane) was significantly increased over their nonknockout controls: HCN1 versus wild-type (0.86% ± 0.03%, P < 0.001) and HCN1: cre versus HCN1 mice (0.84% ± 0.03%, P < 0.001); no significant difference was found between HCN1 and HCN1: cre mice. For MAC of immobility of sevoflurane, no significant difference was found among wild-type, HCN1, HCN1, and HCN1: cre mice (all ~2.6%-2.7% sevoflurane). For both HCN1 and HCN1: cre mice, the MAC of hypnosis for sevoflurane (each ~1.90% sevoflurane) was significantly increased over their nonknockout controls: HCN1 versus wild-type (1.58% ± 0.05%, P < 0.001) and HCN1: cre versus HCN1 mice (1.56% ± 0.05%, P < 0.001). No significant difference was found between HCN1 and HCN1: cre mice. By fear-potentiated startle experiments, amnestic effects of isoflurane and sevoflurane were significantly attenuated in HCN1 and HCN1: cre mice (both P < 0.002 versus wild-type or HCN1 mice). No significant difference was found between HCN1 and HCN1: cre mice. CONCLUSIONS: Forebrain HCN1 channels contribute to hypnotic and amnestic effects of volatile anesthetics, but HCN1 channels are not involved in the immobilizing actions of volatile anesthetics.


Subject(s)
Amnesia/chemically induced , Amnesia/metabolism , Anesthetics, Inhalation/adverse effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/deficiency , Hypnotics and Sedatives/adverse effects , Immobilization , Potassium Channels/deficiency , Animals , Immobilization/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prosencephalon/drug effects , Prosencephalon/metabolism
18.
BMC Complement Altern Med ; 15: 55, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25888276

ABSTRACT

BACKGROUND: Tong Luo Jiu Nao (TLJN), a modern formula of Chinese medicine extracts on the basis of Traditional Chinese Medicine theory, has been used to treat dementia. The present study aimed to investigate its ameliorating effects on Aß1-40-induced cognitive impairment in rats using a series of novel reward-directed instrumental learning (RDIL) tasks, and to determine its possible mechanism of action. METHODS: Rats were pretreated with TLJN extract (0.9 and 1.8 g/kg, p.o.) for 10 daysbefore surgery, and were trained to gain reward reinforcement by lever pressing at the meantime. Thereafter, rats received a bilateral microinjection of Aß1-40 in CA1 regions of the hippocampus. Cognitive performance was evaluated with the goal directed (higher response ratio) and habit (visual signal discrimination and extinction) learning tasks, as well as on the levels of biochemical parameters and molecules. RESULTS: Our findings first demonstrated that TLJN can improve Aß1-40-induced amnesia in RDIL via enhancing the comprehension of action-outcome association and the utilization of cue information to guide behavior. Then, its ameliorating effects should attribute to the modulation of ERK/CaMKII/CREB signaling in the hippocampus. CONCLUSION: TLJN can markedly enhance cognitions of Aß1-40 microinjection animal model in adaptive behavioral tasks. It has the potential, possibly as complementary and alternative therapy, to prevent and/or delay the deterioration of cognitive impairment in AD.


Subject(s)
Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Cognition/drug effects , Drugs, Chinese Herbal/therapeutic use , Hippocampus/drug effects , Learning/drug effects , Phytotherapy , Adaptation, Psychological , Alzheimer Disease/metabolism , Amnesia/drug therapy , Amnesia/metabolism , Amyloid beta-Peptides/adverse effects , Amyloid beta-Peptides/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognition Disorders/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dementia , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Feedback, Psychological/drug effects , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Male , Medicine, Chinese Traditional , Peptide Fragments/adverse effects , Peptide Fragments/metabolism , Rats, Wistar , Signal Transduction
19.
Neurosci Lett ; 556: 5-9, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24103378

ABSTRACT

A number of tremorogenic ß-carboline alkaloids such as harmane are naturally present in the human food chain. They are derived from medicinal plants such as Peganum harmala that have been used as folk medicine in anticancer therapy. In the present study, effects of the histaminergic system of the dorsal hippocampus (CA1) on harmane-induced amnesia were examined. One-trial step-down was used to assess memory retention in adult male mice. The results showed that pre-training intra-CA1 administration of histamine (5µg/mouse), ranitidine (H2 receptor antagonist; at the doses of 0.25 and 0.5µg/mouse) and pyrilamine (H1 receptor antagonist; at the dose of 5µg/mouse) decreased memory formation. Pre-training intraperitoneal (i.p.) administration of harmane (12mg/kg) also decreased memory formation. Moreover, pre-training intra-CA1 injection of a sub-threshold dose of histamine (2.5µg/mouse) could reverse harmane (12mg/kg, i.p.)-induced impairment of memory. On the other hand, pre-training intra-CA1 injection of sub-threshold doses of ranitidine (0.0625µg/mouse) and pyrilamine (2.5µg/mouse) increased harmane-induced impairment of memory. In conclusion, the present findings suggest the involvement of the CA1 histaminergic system in harmane-induced impairment of memory formation.


Subject(s)
CA1 Region, Hippocampal/drug effects , Harmine/analogs & derivatives , Memory/drug effects , Receptors, Histamine H1/metabolism , Receptors, Histamine H2/metabolism , Amnesia/chemically induced , Amnesia/metabolism , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , CA1 Region, Hippocampal/metabolism , Harmine/toxicity , Male , Memory/physiology , Mice
20.
Neurobiol Learn Mem ; 106: 177-84, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012642

ABSTRACT

Arc (Activity-regulated cytoskeletal-associated protein) is a member of the immediate-early gene (IEG) family protein. Because of its critical role in learning and memory, it is widely considered to be an important protein in synaptic plasticity and related neurobiological functions. Alcoholic extract of Ashwagandha leaves (i-Extract) was recently shown to have preventive and therapeutic potential for scopolamine-induced amnesia and glutamate-induced excitotoxicity. In the present study, we investigated the involvement of Arc in scopolamine-induced amnesia and its recovery by i-Extract with particular focus to the changes in Arc expression in the hippocampus and cerebral cortex of mice. Morris water maze test showed that spatial learning and memory of mice were drastically reduced by scopolamine administration but improved with i-Extract treatment as compared to control and scopolamine-challenged mice. Molecular analysis revealed a remarkable decline in Arc expression in both hippocampus and cerebral cortex of amnesic mice, which was recovered after i-Extract treatment. Interestingly, Arc expression showed better recovery in the hippocampus than the cerebral cortex and the pre-treatment with i-Extract was more effective than the post-treatment. These findings suggest that Arc may be involved in i-Extract mediated recovery from amnesia.


Subject(s)
Amnesia/metabolism , Cytoskeletal Proteins/metabolism , Hippocampus/drug effects , Maze Learning/drug effects , Nerve Tissue Proteins/metabolism , Plant Extracts/pharmacology , Withania , Amnesia/chemically induced , Amnesia/drug therapy , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Hippocampus/metabolism , Male , Mice , Scopolamine
SELECTION OF CITATIONS
SEARCH DETAIL