Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Pharm Biol ; 58(1): 447-453, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32432948

ABSTRACT

Context: There are numerous pharmacological activities for Ruta graveolens and its bioactive constituent, rutin, on learning and memory.Objective: This study aimed to examine the effect of R. graveolens and rutin on memory in rats.Materials and methods: In this study animals were treated with the hydroalcholic extract of R. graveolens and rutin by IP injection for 10 days. Behavioural and biochemical tests as well as HPLC analysis and antioxidant activity of extract have been evaluated.Results: R. graveolens extract and rutin significantly increased learning and improved spatial memory, as well as secondary latency; moreover, there were significant increases in the serum and brain antioxidant capacity as well as the level of TBARS in serum and brain tissues. Results also showed that R. graveolens has significant DPPH radical scavenging effect (IC50: 159.17 ± 1.56 µg/mL). The HPLC analysis of extract showed that caffeic acid (19.92 ± 0.01), rutin (40.15 ± 0.01), and apigenin (0.84 ± 0.01) mg/g of dry extract are the main components of the extract.Discussion and conclusion: Regarding the effects of R. graveolens extract and rutin on animal brain cells, memory function, and learning, additional studies, including clinical trials, might be beneficial in producing natural supplementary drugs from this herb.


Subject(s)
Avoidance Learning/drug effects , Brain/drug effects , Ruta , Rutin/pharmacology , Spatial Memory/drug effects , Animals , Avoidance Learning/physiology , Brain/metabolism , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Rutin/isolation & purification , Rutin/therapeutic use , Spatial Memory/physiology
2.
Int Immunopharmacol ; 66: 242-250, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30500621

ABSTRACT

Experiencing psychosocial adversities in early life such as maternal separation (MS) increases the risk of psychiatric disorders. Immune-inflammatory responses have imperative roles in the pathophysiology of psychiatric disorders. MS relatively changes the composition of intestinal microbiota leading to an overactivation of the hypothalamic-pituitary-adrenal (HPA) axis, and subsequently increases the corticosterone level. In this study, we aimed to evaluate the role of corticosterone in behavioral changes and microbiota modifications in a mouse model of MS afflicted neuroinflammatory response in the hippocampus. For this purpose, 180 min of MS stress was applied to mice at postnatal day (PND) 2-14 followed by behavioral tests including forced swimming test (FST), splash test, open field test (OFT) and elevated plus maze (EPM) at PND 50-52. For evaluating the role of corticosterone, mice were subjected to adrenalectomy. Using real-time RT-PCR, the expression of inflammatory genes was determined in the hippocampus and colon tissues. We found that MS provoked depressive- and anxiety-like behaviors in adult male mice. In addition, MS was able to active a neuroimmune response in the hippocampus, motivate inflammation and histopathologic changes in the colon tissue and modify the composition of gut microbiota as well. Interestingly, our findings showed that adrenalectomy (decline in the corticosterone level), could modulate the above-mentioned negative effects of MS. In conclusion, our results demonstrated that overactivation of HPA axis and the subsequent increased level of corticosterone could act, possibly, as the deleterious effects of MS on behavior, microbiota composition changes and activation of neuroimmune response.


Subject(s)
Attention Deficit Disorder with Hyperactivity/metabolism , Corticosterone/metabolism , Gastrointestinal Microbiome/physiology , Hippocampus/physiology , Inflammation/metabolism , Stress, Physiological/physiology , Animals , Animals, Newborn , Attention Deficit Disorder with Hyperactivity/psychology , Behavior, Animal , Disease Models, Animal , Female , Male , Maternal Deprivation , Mice , Mice, Inbred Strains , Neuroimmunomodulation , Pituitary-Adrenal System , Problem Behavior
3.
Iran J Pharm Res ; 14(4): 1197-206, 2015.
Article in English | MEDLINE | ID: mdl-26664387

ABSTRACT

Some animal models have been used to study Alzheimer's disease (AD). AD is an irreversible progressive neurodegenerative disease and the most common cause of dementia. Animal studies have shown that there is a relation between decrease in cholinergic functions in the nucleus basalis of Meynert (NBM) and loss of learning capability and memory. The aim of this study was to investigate the effect of Rheum ribes extract (RR) on memory deficit in one of the rat models of AD. Plant (1500gr) was collected from Saman (kahkesh) region of Chaharmahal Va Bakhtiari province in Iran. RR hydro-alcoholic extracts were prepared using maceration method. Rat model of Alzheimer was induced by Nucleus Basalis of Meynert lesions (NBML). Animals (n = 32) received extracts for 20 days and then passive avoidance and Morris water maze tasks were performed for memory evaluation. FRAP and HPLC methods were used for measurement of the antioxidant and Malondialdehyde (MDA) levels in blood. In water maze experiment, probe trial results showed that NBML group spent significantly less time in target quadrant, in which the platform was located on the preceding day. In addition, the time spent in target quadrant was significantly increased in NBML + RR groups (250 and 500 mg/kg) compared to the NBML group. In passive avoidance task, mean initial latency time and step-though latency were significantly decreased in NBML group. RR extracts significantly prolonged step-through latency in NBML + RR groups. Results of this study suggest that Rheum ribes extracts can improve memory deficits induced by bilateral NBM lesions in rats.

5.
Nat Neurosci ; 10(9): 1125-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17660813

ABSTRACT

Using targeted mouse mutants and pharmacologic inhibition of alphaCaMKII, we demonstrate that the alphaCaMKII protein, but not its activation, autophosphorylation or its ability to phosphorylate synapsin I, is required for normal short-term presynaptic plasticity. Furthermore, alphaCaMKII regulates the number of docked vesicles independent of its ability to be activated. These results indicate that alphaCaMKII has a nonenzymatic role in short-term presynaptic plasticity at hippocampal CA3-CA1 synapses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Hippocampus/cytology , Neuronal Plasticity/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Synapses/physiology , Synaptic Transmission/physiology , Analysis of Variance , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Electric Stimulation , Enzyme Activation/genetics , Excitatory Postsynaptic Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Mutagenesis/physiology , Neurons/radiation effects , Patch-Clamp Techniques/methods , Phosphorylation , Presynaptic Terminals/ultrastructure , Synapses/ultrastructure , Synapsins/metabolism , Synaptic Transmission/genetics , Synaptic Vesicles/physiology
6.
Nat Neurosci ; 10(3): 280-2, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17259980

ABSTRACT

Angelman syndrome (AS) is a severe neurological disorder characterized by mental retardation, motor dysfunction and epilepsy. We show that the molecular and cellular deficits of an AS mouse model can be rescued by introducing an additional mutation at the inhibitory phosphorylation site of alphaCaMKII. Moreover, these double mutants no longer show the behavioral deficits seen in AS mice, suggesting that these deficits are the direct result of increased inhibitory phosphorylation of alphaCaMKII.


Subject(s)
Angelman Syndrome/complications , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mental Disorders/etiology , Mental Disorders/therapy , Angelman Syndrome/genetics , Angelman Syndrome/pathology , Animals , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Conditioning, Classical/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/genetics , Female , Freezing Reaction, Cataleptic/physiology , Hippocampus/physiopathology , In Vitro Techniques , Male , Maze Learning/physiology , Mental Disorders/genetics , Mental Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Motor Activity/physiology , Phosphorylation , Phosphotransferases/metabolism , Reaction Time , Time Factors , Ubiquitin-Protein Ligases/genetics
7.
J Lipid Res ; 47(3): 673-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16371647

ABSTRACT

Sphingomyelin (SM) and phosphatidylcholine (PC) are two major phospholipids on plasma lipoproteins. Their concentration is classically measured by lipid extraction, thin-layer chromatography, and phosphate determination on separated SM or PC spots. Here, we describe two rapid, specific, and sensitive enzymatic measurements for both phospholipids. Plasma was incubated with bacterial sphingomyelinase (for SM measurement) or bacterial PC-specific phospholipase D (for PC measurement), alkaline phosphatase, choline oxidase, peroxidase, N-ethyl-N-(2-hydroxy-3-sulfopropyl)-3,5-dimethoxyaniline, and 4-aminoantipyrine for 45 min. A blue dye, with an optimal absorption at 595 nm, was generated. PC levels did not influence SM measurement and vice versa. The linear range for the SM measurement was 0.5-5 microg, and that for PC was 2.5-20 microg. The mean percentage recovery was 98.0 +/- 5.2% for SM and 96.6 +/- 3.8% for PC. The interassay coefficient of variation of the assay was 1.7 +/- 0.05% for SM and 3.1 +/- 0.13% for PC. These two new methods are amenable to automation and can be adapted for large-scale, high-throughput assays.


Subject(s)
Phosphatidylcholines/blood , Sphingomyelins/blood , Dose-Response Relationship, Drug , Humans , Sensitivity and Specificity
8.
Biochim Biophys Acta ; 1737(1): 44-51, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16216550

ABSTRACT

Sphingolipids play a very important role in cell membrane formation, signal transduction, and plasma lipoprotein metabolism, and all these functions may have an impact on atherosclerotic development. Serine palmitoyl-CoA transferase (SPT) is the key enzyme in sphingolipid biosynthesis. To evaluate in vivo SPT activity and its role in sphingolipid metabolism, we applied homologous recombination to embryonic stem cells, producing mice with long chain base 1 (Sptlc1) and long chain base 2 (Sptlc2), two subunits of SPT, gene deficiency. Homozygous Sptlc11 and Sptlc2 mice are embryonic lethal, whereas heterozygous versions of both animals (Sptlc1(+/-), Sptlc2(+/-)) are healthy. Analysis showed that, compared with WT mice, Sptlc1(+/-) and Sptlc2(+/-) mice had: (1) decreased liver Sptlc1 and Sptlc2 mRNA by 44% and 57% (P<0.01 and P<0.0001, respectively); (2) decreased liver Sptlc1 mass by 50% and Sptlc2 mass by 70% (P<0.01 and P<0.01, respectively), moreover, Sptlc1 mass decreased by 70% in Sptlc2(+/-) mouse liver, while Sptlc2 mass decreased by 53% in Sptlc1(+/-) mouse liver (P<0.001 and P<0.01, respectively); (3) decreased liver SPT activity by 45% and 60% (P<0.01, respectively); (4) decreased liver ceramide (22% and 39%, P<0.05 and P<0.01, respectively) and sphingosine levels (22% and 31%, P<0.05 and P<0.01, respectively); (5) decreased plasma ceramide (45% and 39%, P<0.01, respectively), sphingosine-1-phosphate (31% and 32%, P<0.01, respectively) and sphingosine levels (22.5% and 25%, P<0.01, respectively); (6) dramatically decreased plasma lysosphingomyelin (17-fold and 16-fold, P<0.0001, respectively); and (7) no change of plasma sphingomyelin, triglyceride, total cholesterol, phospholipids, and liver sphingomyelin levels. These results indicated that both Sptlc1 and Sptlc2 interactions are necessary for SPT activity in vivo, and that SPT activity directly influences plasma sphingolipid levels. Furthermore, manipulation of SPT activity might well influence the course of such diseases as atherosclerosis.


Subject(s)
Serine C-Palmitoyltransferase/deficiency , Sphingolipids/metabolism , Animals , Ceramides/blood , Chimera , Female , Genetic Vectors , Heterozygote , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Serine C-Palmitoyltransferase/biosynthesis , Sphingolipids/blood , Sphingomyelins/metabolism
9.
J Neurosci ; 25(42): 9721-34, 2005 Oct 19.
Article in English | MEDLINE | ID: mdl-16237176

ABSTRACT

Molecular and cellular studies of the mechanisms underlying mammalian learning and memory have focused almost exclusively on postsynaptic function. We now reveal an experience-dependent presynaptic mechanism that modulates learning and synaptic plasticity in mice. Consistent with a presynaptic function for endogenous H-ras/extracellular signal-regulated kinase (ERK) signaling, we observed that, under normal physiologic conditions in wild-type mice, hippocampus-dependent learning stimulated the ERK-dependent phosphorylation of synapsin I, and MEK (MAP kinase kinase)/ERK inhibition selectively decreased the frequency of miniature EPSCs. By generating transgenic mice expressing a constitutively active form of H-ras (H-rasG12V), which is abundantly localized in axon terminals, we were able to increase the ERK-dependent phosphorylation of synapsin I. This resulted in several presynaptic changes, including a higher density of docked neurotransmitter vesicles in glutamatergic terminals, an increased frequency of miniature EPSCs, and increased paired-pulse facilitation. In addition, we observed facilitated neurotransmitter release selectively during high-frequency activity with consequent increases in long-term potentiation. Moreover, these mice showed dramatic enhancements in hippocampus-dependent learning. Importantly, deletion of synapsin I, an exclusively presynaptic protein, blocked the enhancements of learning, presynaptic plasticity, and long-term potentiation. Together with previous invertebrate studies, these results demonstrate that presynaptic plasticity represents an important evolutionarily conserved mechanism for modulating learning and memory.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/biosynthesis , Learning/physiology , MAP Kinase Signaling System/physiology , Neuronal Plasticity/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Synapsins/biosynthesis , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Presynaptic Terminals/enzymology , Proto-Oncogene Proteins p21(ras)/biosynthesis , Proto-Oncogene Proteins p21(ras)/genetics , Synapsins/genetics
10.
Nat Neurosci ; 8(10): 1292-4, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16136042

ABSTRACT

L7-PKCI transgenic mice, which lack parallel fiber-Purkinje cell long-term depression (LTD), were tested with two different mazes to dissociate the relative importance of declarative and procedural components of spatial navigation. We show that L7-PKCI mice are deficient in acquisition of an adapted goal-oriented behavior, part of the procedural component of the task. This supports the hypothesis that cerebellar LTD may subserve a general sensorimotor adaptation process shared by motor and spatial learning functions.


Subject(s)
Adaptation, Physiological/physiology , Cerebellum/cytology , Long-Term Synaptic Depression/physiology , Perceptual Disorders/physiopathology , Purkinje Cells/physiology , Spatial Behavior/physiology , Adaptation, Physiological/genetics , Analysis of Variance , Animals , Behavior, Animal , Escape Reaction/physiology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Perceptual Disorders/genetics , Protein Kinase C/genetics , Psychomotor Performance/physiology , Reaction Time/genetics , Time Factors
11.
Biochim Biophys Acta ; 1735(2): 130-4, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15967715

ABSTRACT

Sphingomyelin (SM) plays a very important role in cell membrane formation and plasma lipoprotein metabolism. All these functions may have an impact on atherosclerotic development. To investigate the relationship between SM metabolism and atherosclerosis, we utilized a sphingolipid-rich diet to feed LDL receptor gene knockout (LDLr KO) mice and studied lipid metabolism and atherosclerosis in the mice. After 3 months of a sphingolipid-rich diet, we found a significant increase in SM, cholesterol, and SM/phosphatidylcholine (PC) ratio (50%, P<0.001; 62%, P<0.01; and 45%, P<0.01, respectively), compared to chow fed diet. HDL-lipids were not significantly altered. Non-HDL-SM, non-HDL-C, and non-HDL-SM/non-HDL-PC ratio were significantly increased (115%, P<0.001; 106%, P<0.001; and 106%, P<0.01, respectively). FPLC confirmed the results. SDS-PAGE showed an increase of apoB48 and apoB100, but no changes of apoAI. Moreover, we found that an SM-rich diet significantly increased atherosclerotic lesion area in both root assay and en face assay, compared to chow diet (58,210+/-15,300 microm(2) vs. 9670+/-2370 microm(2), P<0.001; 5.9+/-3.1% vs. 1.1+/-0.9%, P<0.001). These results indicate that the enrichment of sphingolipids in diet has proatherogenic properties.


Subject(s)
Arteriosclerosis/etiology , Sphingolipids/administration & dosage , Sphingomyelins/blood , Animals , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Diet, Atherogenic , Female , Mice , Mice, Knockout , Mutation , Receptors, LDL/genetics , Sphingomyelins/metabolism
12.
J Biol Chem ; 280(11): 10284-9, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15590644

ABSTRACT

Sphingolipids play a very important role in cell membrane formation, signal transduction, and plasma lipoprotein metabolism, all of which may well have an impact on the development of atherosclerosis. To investigate the relationship between sphingolipid metabolism and atherosclerosis, we utilized myriocin to inhibit mouse serine palmitoyl-CoA transferase (SPT), the key enzyme for sphingolipid biosynthesis. We injected 8-week-old apoE-deficient mice with myriocin (0.3 mg/kg/every other day, intraperitoneal) for 60 days. On a chow diet, myriocin treatment caused a significant decrease (50%) in liver SPT activity (p < 0.001), significant decreases in plasma sphingomyelin, ceramide, and sphingosine-1-phosphate levels (54, 32, and 73%, respectively) (p < 0.0001), and a significant increase in plasma phosphatidylcholine levels (91%) (p < 0.0001). Plasma total cholesterol and triglyceride levels demonstrated no significant changes, but there was a significant decrease in atherosclerotic lesion area (42% in root and 36% in en face assays) (p < 0.01). On a high fat diet, myriocin treatment caused marked decreases in plasma sphingomyelin, ceramide, and sphingosine-1-phosphate levels (59, 66, and 81%, respectively) (p < 0.0001), and a marked increase in plasma phosphatidylcholine levels (100%) (p < 0.0001). Total cholesterol and triglyceride demonstrated no significant changes, but there was a significant decrease in atherosclerotic lesion area (39% in root and 37% in en face assays) (p < 0.01). These results indicate that, apart from cholesterol levels, sphingolipids have an effect on atherosclerotic development and that SPT has proatherogenic properties. Thus, inhibition of SPT activity could be an alternative treatment for atherosclerosis.


Subject(s)
Apolipoproteins E/metabolism , Arteriosclerosis/metabolism , Fatty Acids, Monounsaturated/pharmacology , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Acyltransferases/antagonists & inhibitors , Acyltransferases/chemistry , Animal Feed , Animals , Aorta/metabolism , Arteriosclerosis/blood , Arteriosclerosis/pathology , Ceramides/blood , Cholesterol/metabolism , Fatty Acids, Monounsaturated/chemistry , Immunosuppressive Agents/pharmacology , Lipid Metabolism , Lipids/blood , Lipoproteins/metabolism , Lysophospholipids/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Phosphatidylcholines/blood , Serine C-Palmitoyltransferase , Signal Transduction , Sphingolipids/blood , Sphingosine/metabolism , Time Factors , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...