Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
2.
Cell Physiol Biochem ; 29(1-2): 189-96, 2012.
Article in English | MEDLINE | ID: mdl-22415088

ABSTRACT

BACKGROUND/AIMS: We have earlier found that indomethacin activates CaMKII, as a novel action distinct from COX inhibition. To explore further indomethacin actions, the present study focused upon PKC and examined the effect of indomethacin on α7 ACh receptor responses and hippocampal synaptic transmission through PKC. METHODS: We recorded currents through α7 ACh receptors expressed in Xenopus oocytes, quantified PKC activity in the in situ and cell-free PKC assay, and monitored field excitatory postsynaptic potentials (fEPSPs) and miniature excitatory postsynaptic currents (mEPSCs) from the CA1 region of rat hippocampal slices. RESULTS: Indomethacin potentiated α7 ACh receptor currents in a bell-shaped concentration (100 nM-1 mM)-dependent manner, and the potentiating effect was inhibited by the PKC inhibitor GF109203X. Indomethacin activated PKC in a concentration (1-100 µM)-dependent manner for cultured rat hippocampal neurons. Additionally, indomethacin (100 µM) significantly activated PKC-ε under the cell-free conditions. Indomethacin (100 µM) induced a transient huge increase in the fEPSP slope followed by persistent increase, and the former effect was attenuated by the α7 ACh receptor antagonist α-bungarotoxin or GF109203X. Indomethacin (100 µM) also increased the rate of nicotine-evoked mEPSCs, and the effect was prevented by α-bungarotoxin or GF109203X. CONCLUSION: The results of the present study show that indomethacin activates PKC, possibly PKC-e in the brain, thereby potentiating α7 ACh receptor responses to stimulate presynaptic glutamate release, which in part contributes to facilitation of hippocampal transmission. This extends our knowledge about diverse indomethacin actions.


Subject(s)
Indomethacin/pharmacology , Protein Kinase C/metabolism , Receptors, Nicotinic/metabolism , Animals , Bungarotoxins/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Nicotinic/chemistry , Xenopus/growth & development , Xenopus/metabolism , alpha7 Nicotinic Acetylcholine Receptor
3.
J Cell Physiol ; 227(3): 919-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21503887

ABSTRACT

The present study examined the effect of indomethacin (IM), a cyclooxygenase inhibitor, on learning and memory functions. IM activated Ca(2+) /calmodulin-dependent protein kinase II (CaMKII) in cultured rat hippocampal neurons. IM (100 µM) significantly increased the rate of spontaneous AMPA receptor-mediated miniature excitatory postsynaptic currents elicited from CA1 pyramidal neurons of rat hippocampal slices, without affecting the amplitude, and enhanced extracellular high K(+) (20 mM)-induced glutamate release from rat hippocampal slices, indicating that IM stimulates presynaptic glutamate release. Those IM effects were clearly inhibited by the CaMKII inhibitor KN-93. IM persistently facilitated synaptic transmission monitored from the CA1 region of rat hippocampal slices in a concentration (1-100 µM)-dependent manner that was also abolished by KN-93. In the water maze test, IM (1 mg/kg, i.p.) enhanced spatial learning and memory ability for normal rats, and ameliorated scopolamine-induced spatial learning and memory impairment or age-related spatial learning and memory deterioration for senescence-accelerated mouse-prone 8 mice. In the test to learn 15 numbers consisting of three patterns of five digit number for healthy human subjects, oral intake with IM (25 mg/kg) significantly raised the scores of correct number arrangements that subjects memorized 5 min and 3 days after the test. The results of the present study indicate that IM could enhance learning and memory potential by facilitating hippocampal synaptic transmission as a result from stimulating presynaptic glutamate release under the control of CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Indomethacin/administration & dosage , Indomethacin/metabolism , Maze Learning/drug effects , Memory/drug effects , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Humans , Indomethacin/pharmacology , Male , Maze Learning/physiology , Memory/physiology , Mice , Middle Aged , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Organ Culture Techniques , Rats , Rats, Wistar , Xenopus , Young Adult
4.
Cell Physiol Biochem ; 27(1): 37-44, 2011.
Article in English | MEDLINE | ID: mdl-21325820

ABSTRACT

BACKGROUND/AIMS: The mechanism underlying extracellular adenosine-induced caspase-independent apoptosis in HuH-7 human hepatoma cells is not fully understood. The present study investigated the role for apoptosis-inducing factor (AIF)-homologous mitochondrion-associated inducer of death (AMID) in the pathway. METHODS: To see the implication of AMID in adenosine-induced HuH-7 cell apoptosis, real-time reverse transcription-polymerase chain reaction (RT-PCR), immunofluorescent cytochemistry, time-laps GFP monitoring, cell cycle analysis, flow cytometry, Western blotting, cell viability assay, and TUNEL staining were carried out. RESULTS: Adenosine upregulated AMID expression in HuH-7 cells, and translocated AMID from the cytosol into the nucleus. Adenosine induced HuH-7 cell apoptosis, and the effect was further enhanced by overexpressing AMID. Adenosine-induced HuH-7 cell apoptosis, alternatively, was inhibited by knocking-down AMID. CONCLUSION: The results of the present study provide evidence for AMID as a critical factor for adenosine-induced caspase-independent HuH-7 cell apoptosis.


Subject(s)
Adenosine/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Caspases/metabolism , Mitochondrial Proteins/metabolism , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Humans , Mitochondrial Proteins/analysis , Mitochondrial Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation
5.
Lipids Health Dis ; 10: 25, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21272376

ABSTRACT

In the water maze test, oral administration with 1,2-dilynoleoyl-sn-glycero-3-phosphocholine (DLPhtCho)(5 mg/kg) alone or DLPhtCho (5 mg/kg) plus 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPhtCho)(5 mg/kg) significantly shortened the prolonged acquisition latency for rats intraperitoneally injected with scopolamine, with more efficient effect than (POPhtCho)(5 mg/kg) alone, arachidonic acid (AA)(5 mg/kg) alone, docosahexaenoic acid (DHA)(5 mg/kg) alone, or 1-palmitoyl-2-linoleil-sn-glycero-3-phosphoserine (PLPhtSer)(5 mg/kg) alone. POPhtCho (5 mg/kg) alone or DLPhtCho (5 mg/kg) plus POPhtCho (5 mg/kg) also significantly shortened the prolonged retention latency for rats intraperitoneally injected with scopolamine, but otherwise no significant effect was obtained with DLPhtCho (5 mg/kg) alone, AA (5 mg/kg) alone, DHA (5 mg/kg) alone, or PLPhtSer (5 mg/kg) alone. Oral co-administration with DLPhtCho (5 mg/kg) and POPhtCho (5 mg/kg) significantly shortened the acquisition latency for rats untreated with scopolamine as compared with the latency for administration with polyethylene glycol (PEG), DLPhtCho alone at doses of 5 and 10 mg/kg, or POPhtCho alone at doses of 5 and 10 mg/kg, while no efficient effect on the retention latency was obtained. To assess the effect of DLPhtCho and POPhtCho on cognitive functions for humans, Mini Mental State Examination (MMSE) test was performed in subjects with cognitive disorders (the average MMSE score, 15). Oral co-intake with DLPhtCho (50 mg) and POPhtCho (45 mg) once after breakfast everyday raised the score to over 20, corresponding to normal cognitive functions, throughout 5 months after intake, and the increase in the score was significantly greater than that for oral intake with DLPhtCho (100 mg/day) alone or POPhtCho (90 mg/kg) alone. Taken together, the results of the present study show that co-intake with DLPhtCho and POPhtCho could enhance learning and memory ability and improve cognitive disorders for both the animals and humans with a promising efficacy.


Subject(s)
Cognition Disorders/drug therapy , Memory Disorders/drug therapy , Nootropic Agents/therapeutic use , Phosphatidylcholines/therapeutic use , Aged , Aged, 80 and over , Animals , Cognition Disorders/prevention & control , Disease Models, Animal , Drug Combinations , Female , Humans , Male , Maze Learning/drug effects , Memory Disorders/prevention & control , Middle Aged , Nootropic Agents/pharmacology , Phosphatidylcholines/pharmacology , Rats , Rats, Wistar , Scopolamine
6.
Behav Brain Funct ; 6: 52, 2010 Sep 13.
Article in English | MEDLINE | ID: mdl-20831835

ABSTRACT

BACKGROUND: Accumulating evidence has pointed that a variety of lipids could exert their beneficial actions against dementia including Alzheimer disease and age-related cognitive decline via diverse signaling pathways. Endoplasmic reticulum (ER) stress-induced neuronal apoptosis, on the other hand, is a critical factor for pathogenesis of neurodegenerative diseases such as Alzheimer disease and Parkinson disease, senile dementia, and ischemic neuronal damage. The present study examined the effects of 1,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine (DLPhtEtn), a phospholipid, on ER stress-induced neuronal death and age-related cognitive disorders. METHODS: PC-12 cell viability was assayed before and after treatment with amyloid-ß(1-40) peptide or thapsigargin in the presence and absence of DLPhtEtn. A series of behavioral tests were performed for senescence-accelerated mouse-prone 8 (SAMP8) mice after 7-month oral administration with polyethylene glycol (PEG) or DLPhtEtn and then, the number of hippocampal neurons was counted. RESULTS: Amyloid-ß(1-40) peptide or thapsigargin is capable of causing ER stress-induced apoptosis. DLPhtEtn (30 µM) significantly inhibited PC-12 cell death induced by amyloid-ß(1-40) peptide or thapsigargin. In the water maze test, oral administration with DLPhtEtn (1 mg/kg) for 7 months (three times a week) significantly shortened the prolonged retention latency for SAMP8 mice. In contrast, DLPhtEtn had no effect on the acquisition and retention latencies in both the open field test and the passive avoidance test for SAMP8 mice. Oral administration with DLPhtEtn (1 mg/kg) for 7 months prevented a decrease in the number of hippocampal neurons for SAMP8 mice. CONCLUSION: The results of the present study show that DLPhtEtn ameliorates age-related spatial memory decline without affecting motor activities or fear memory, possibly by protecting hippocampal neuronal death. DLPhtEtn, thus, might exert its beneficial action against senile dementia and neurodegenerative diseases such as Alzheimer disease.


Subject(s)
Cell Death/drug effects , Cell Survival/drug effects , Hippocampus/drug effects , Memory Disorders/pathology , Memory Disorders/prevention & control , Neurons/pathology , Phosphatidylethanolamines/pharmacology , Age Factors , Amyloid beta-Peptides/pharmacology , Animals , Disease Models, Animal , Drug Administration Schedule , Hippocampus/pathology , Male , Mice , Mice, Inbred Strains , PC12 Cells , Peptide Fragments/pharmacology , Phosphatidylethanolamines/administration & dosage , Rats , Thapsigargin/pharmacology
7.
Neurochem Res ; 35(5): 782-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20165912

ABSTRACT

The present study investigated the role of O-linked beta-N-acetylglucosamine (O-GlcNAc) glycosylation (O-GlcNAcylation) in AMPA receptor trafficking. Alloxan, an inhibitor of O-GlcNAc transferase, potentiated responses of AMPA receptors composed of the GluR1 subunit expressed in Xenopus oocytes. No potentiating effect of alloxan was obtained with mutant GluR1 (S831A) receptor lacking CaMKII phosphorylation site. Alloxan facilitated basal synaptic transmission to approximately 120% of basal levels and enhanced Schaffer collateral-CA1 long-term potentiation (LTP) in rat hippocampal slices, especially in the late phase of the LTP. Alloxan stimulated translocation of the GluR1 and GluR2 subunit from the cytosol towards the plasma membrane in rat hippocampal slices with the LTP, although it had no effect on subcellular distribution of the NR1 subunit. Taken together, the results of the present study show that alloxan regulates AMPA receptor trafficking by inhibiting O-GlcNAcylation, to modulate hippocampal synaptic transmission and synaptic plasticity.


Subject(s)
Alloxan/pharmacology , N-Acetylglucosaminyltransferases/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission/drug effects , Animals , Glycosylation/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Long-Term Potentiation/drug effects , Oocytes , Rats , Xenopus
8.
Behav Brain Res ; 206(1): 151-4, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19716848

ABSTRACT

Long-term potentiation (LTP) was monitored from the CA1 region of the intact rat hippocampus by delivering high frequency stimulation (HFS) to the Schaffer collateral commissural pathway. Intraventricular injection with mutant amyloid beta(1-42) peptide lacking glutamate-22 (Abeta(1-42)E22Delta), favoring oligomerization, 10 min prior to HFS, inhibited expression of LTP, with the potency more than wild-type amyloid beta(1-42) peptide. Intraperitoneal injection with the linoleic acid derivative 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA) 70 min prior to HFS neutralized mutant Abeta(1-42)E22Delta peptide-induced LTP inhibition. In the water maze test, continuous intraventricular injection with mutant Abeta(1-42)E22Delta peptide for 14 days prolonged the acquisition latency as compared with that for control, with the potency similar to wild-type Abeta(1-42) peptide, and intraperitoneal injection with DCP-LA shortened the prolonged latency to control levels. The results of the present study indicate that DCP-LA neutralizes mutant Abeta(1-42)E22Delta peptide-induced impairment of LTP and spatial learning.


Subject(s)
Amyloid beta-Peptides/administration & dosage , CA1 Region, Hippocampal/drug effects , Caprylates/pharmacology , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Analysis of Variance , Animals , CA1 Region, Hippocampal/physiopathology , Electric Stimulation , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Injections, Intraventricular , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Rats , Rats, Wistar , Spatial Behavior/drug effects , Spatial Behavior/physiology
9.
Behav Brain Res ; 206(1): 47-51, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19729040

ABSTRACT

Expression of Schaffer collateral-CA1 long-term potentiation (LTP) and long-term depression (LTD) was not affected in hippocampal slices from wild-type mice pretreated with lipopolysaccharide (0.25mg/kg, i.p.), to increase interleukin-18 (IL-18) concentrations in the brain. For IL-18 knock-out (IL-18 KO) mice, the LTP was still expressed, the extent being similar to that for wild-type mice. In the open-field test to assess motor activity, rearing activity for IL-18 KO mice was significantly suppressed as compared with that for wild-type mice, without significant difference in the locomotion activity between two groups. In the passive avoidance test to assess fear memory, the retention latency for IL-18 KO mice was much shorter than for wild-type mice, without significant difference in the acquisition latency between two groups. In the water maze test, the acquisition latency for IL-18 KO mice significantly prolonged as compared with that for wild-type mice, without significant difference in the retention latency between two groups. For IL-18 KO mice, intraventricular injection with IL-18 for 4 days (total, 240 fg) prior to water maze task shortened the prolonged acquisition latency, reaching a level similar to that for wild-type mice. The results of the present study, thus, suggest that IL-18 is a critical regulator for exploratory activity, fear memory, and spatial learning.


Subject(s)
Anxiety/physiopathology , Interleukin-18/physiology , Maze Learning/physiology , Motor Activity/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Avoidance Learning/physiology , Brain/physiology , CA1 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials/physiology , Exploratory Behavior/physiology , Injections, Intraventricular , Interleukin-18/administration & dosage , Interleukin-18/genetics , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spatial Behavior , Synaptic Transmission/physiology
10.
Int J Alzheimers Dis ; 2011: 431320, 2010 Dec 19.
Article in English | MEDLINE | ID: mdl-21234376

ABSTRACT

Oligomers of 40- or 42-mer amyloid ß-protein (Aß40, Aß42) cause cognitive decline and synaptic dysfunction in Alzheimer's disease. We proposed the importance of a turn at Glu22 and Asp23 of Aß42 to induce its neurotoxicity through the formation of radicals. Recently, a novel deletion mutant at Glu22 (E22Δ) of Aß42 was reported to accelerate oligomerization and synaptotoxicity. To investigate this mechanism, the effects of the E22Δ mutation in Aß42 and Aß40 on the transformation of ß-sheets, radical production, and neurotoxicity were examined. Both mutants promoted ß-sheet transformation and the formation of radicals, while their neurotoxicity was negative. In contrast, E22P-Aß42 with a turn at Glu22 and Asp23 exhibited potent neurotoxicity along with the ability to form radicals and potent synaptotoxicity. These data suggest that conformational change in E22Δ-Aß is similar to that in E22P-Aß42 but not the same, since E22Δ-Aß42 exhibited no cytotoxicity, unlike E22P-Aß42 and wild-type Aß42.

11.
Behav Brain Res ; 204(1): 129-32, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19482045

ABSTRACT

1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPhtCho) (1 microM) enhanced long-term depression (LTD), a synaptic plasticity relevant to learning and memory, in the CA1 region of rat hippocampal slices, where expression of the alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor subunit GluR1 on the plasma membrane was decreased. In the water maze test, oral administration with POPhtCho (5 mg/kg) significantly shortened the prolonged retention latency for rats intraperitoneally injected with scopolamine (1 mg/kg), while the acquisition latency was not affected. For humans with mild cognitive impairment and dementia (average of Mini Mental State Examination score, 18), oral intake with POPhtCho (300 mg/day, once after breakfast) everyday raised the score to over 20, corresponding to normal cognitive functions, throughout 6 months after intake. The results of the present study, thus, indicate that POPhtCho could ameliorate cognitive disorders, possibly by enhancing LTD.


Subject(s)
Central Nervous System Agents/pharmacology , Cognition Disorders/drug therapy , Dementia/drug therapy , Hippocampus/drug effects , Long-Term Synaptic Depression/drug effects , Phosphatidylcholines/pharmacology , Aged , Aged, 80 and over , Animals , Central Nervous System Agents/administration & dosage , Cognition Disorders/chemically induced , Excitatory Postsynaptic Potentials/drug effects , Female , Hippocampus/metabolism , Humans , In Vitro Techniques , Male , Maze Learning/drug effects , Middle Aged , Phosphatidylcholines/administration & dosage , Psychiatric Status Rating Scales , Rats , Rats, Wistar , Receptors, AMPA/metabolism
12.
J Cell Physiol ; 221(1): 183-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19492412

ABSTRACT

The linoleic acid derivative 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA) activated Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) by inhibiting protein phosphatase-1 (PP-1). DCP-LA induced a transient huge facilitation of synaptic transmission monitored from the CA1 region of rat hippocampal slices, which was largely inhibited by the CaMKII inhibitor KN-93. DCP-LA potentiated kainate-evoked whole-cell membrane currents for Xenopus oocytes expressing alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors composed of the GluR1, GluR3, GluR1/GluR2, GluR1/GluR3, and GluR1/GluR2/GluR3 subunits, and the potentiation was significantly inhibited by KN-93. A similar potentiation was still found with mutant GluR1 (S831A) receptor lacking CaMKII phosphorylation site. The GluR1 and GluR2 subunits formed AMPA receptors in the rat hippocampus, and DCP-LA increased expression of both the subunits on the plasma membrane. The DCP-LA action was blocked by KN-93 and the exocytosis inhibitor botulinum toxin type A, but not by the endocytosis inhibitor phenylarsine oxide. DCP-LA, thus, appears to activate CaMKII through PP-1 inhibition, that stimulates AMPA receptor exocytosis to increase expression of the receptors on the plasma membrane, responsible for potentiate AMPA receptor responses and facilitation of hippocampal synaptic transmission.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caprylates/pharmacology , Exocytosis/drug effects , Protein Phosphatase 1/antagonists & inhibitors , Receptors, AMPA/metabolism , Animals , Enzyme Activation/drug effects , Hippocampus/drug effects , Hippocampus/enzymology , Male , Rats , Rats, Wistar , Synaptic Transmission/drug effects
13.
Life Sci ; 84(9-10): 263-6, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19100751

ABSTRACT

AIMS: The present study was conducted to understand the role of 1,2-dilynoleoyl-sn-glycero-3-phosphocholine (DLPhtCho) in cognitive functions. MAIN METHODS: Two-electrode voltage-clamp was made to Xenopus oocytes expressing rat alpha7 acetylcholine (ACh) receptors. Field excitatory postsynaptic potentials (fEPSPs) were monitored from the CA1 region of rat hippocampal slices. Water maze test was carried out to assess spatial learning and memory for rats. KEY FINDINGS: In the oocyte expression system, DLPhtCho at a concentration of 10 microM potentiated ACh-evoked currents to approximately 190% of basal amplitudes 70 min after 10-min treatment. In contrast, 1-stearoyl-2-lynoleoyl-sn-glycero-3-phosphocholine (SLPhtCho), 1-palmitoyl-2-linoleoyl-sn-glycero-3-phosphocholine (PLPhtCho), and 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPhtCho) had no effect on the currents. DLPhtCho (10 microM) enhanced slope of fEPSPs to about 150% of basal levels at 70-min treatment, that is inhibited by alpha-bungarotoxin, an inhibitor of alpha7 ACh receptors, while no enhancement was obtained with SLPhtCho, PLPhtCho, or POPhtCho. In the water maze test, oral administration with DLPhtCho (5 mg/kg) significantly shortened the prolonged acquisition latency for rats intraperitoneally injected with scopolamine (1 mg/kg). SIGNIFICANCE: The results of the present study show that DLPhtCho improves scopolamine-induced learning and memory deficits, possibly by facilitating hippocampal synaptic transmission under the control of alpha7 ACh receptors. DLPhtCho, therefore, could be developed as a beneficial anti-dementia drug.


Subject(s)
Cholinergic Antagonists , Maze Learning/drug effects , Memory/drug effects , Phosphatidylcholines/pharmacology , Receptors, Nicotinic/metabolism , Scopolamine , Animals , Cholinergic Antagonists/metabolism , Cholinergic Antagonists/pharmacology , Male , Oocytes/cytology , Oocytes/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Scopolamine/metabolism , Scopolamine/pharmacology , Spatial Behavior/drug effects , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
14.
Neurosci Lett ; 448(2): 208-11, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18983890

ABSTRACT

Olprinone, an inhibitor of cyclic nucleotide phosphodiesterase III, inhibited an increase in intracellular Ca(2+) concentrations for acutely dissociated rat hippocampal pyramidal neurons induced by extracellular high K(+) (35 mM) depolarization. Olprinone (100 microM) significantly reduced spontaneous glutamate release from rat hippocampal slices. Furthermore, olprinone significantly decreased the rate of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated miniature excitatory postsynaptic currents (AMPA-mEPSCs) monitored from CA1 pyramidal neurons of rat hippocampal slices, and the effect was blocked by KT5823, an inhibitor of protein kinase G (PKG), but not by H-89, an inhibitor of protein kinase A (PKA). In the PKA assay using PC-12 cells, olprinone did not activate PKA. Taken together, the results of the present study show that olprinone attenuates intracellular Ca(2+) rise through voltage-sensitive Ca(2+) channels and inhibits presynaptic glutamate release via a cGMP/PKG pathway.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Imidazoles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Pyridones/pharmacology , Signal Transduction , Animals , Calcium/metabolism , Carbazoles/pharmacology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/metabolism , In Vitro Techniques , Isoquinolines/pharmacology , Male , Membrane Potentials/physiology , Phosphodiesterase 3 Inhibitors , Potassium/metabolism , Protein Kinase Inhibitors/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology
15.
Ann Neurol ; 63(3): 377-87, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18300294

ABSTRACT

OBJECTIVE: Soluble oligomers of amyloid beta (Abeta), rather than amyloid fibrils, have been proposed to initiate synaptic and cognitive dysfunction in Alzheimer's disease (AD). However, there is no direct evidence in humans that this mechanism can cause AD. Here, we report a novel amyloid precursor protein (APP) mutation that may provide evidence to address this question. METHODS: A Japanese pedigree showing Alzheimer's-type dementia was examined for mutations in APP, PSEN1, and PSEN2. In addition, 5,310 Japanese people, including 2,121 patients with AD, were screened for the novel APP mutation. The pathogenic effects of this mutation on Abeta production, degradation, aggregation, and synaptotoxicity were also investigated. RESULTS: We identified a novel APP mutation (E693Delta) producing variant Abeta lacking gulutamate-22 (E22Delta) in Japanese pedigrees showing Alzheimer's-type dementia and AD. Although the secretion of total Abeta was markedly reduced by this mutation, the variant Abeta was more resistant to proteolytic degradation. The mutant peptides showed the unique aggregation property of enhanced oligomerization but no fibrillization, and inhibited hippocampal long-term potentiation more potently than wild-type peptide in rats in vivo. Consistent with the nonfibrillogenic property of the variant Abeta, a very low amyloid signal was observed in the patient's brain on positron emission tomography using Pittsburgh compound-B. INTERPRETATION: The E693Delta mutation has been suggested as a cause of dementia because of enhanced formation of synaptotoxic Abeta oligomers. Our findings may provide genetic validation in humans for the emerging hypothesis that the synaptic and cognitive impairment in AD is primarily caused by soluble Abeta oligomers.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Genetic Variation/genetics , Adult , Aged , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/genetics , Asian People/genetics , Female , Genetic Markers/genetics , Haplotypes/genetics , Humans , Male , Middle Aged , Mutation/genetics , Pedigree
16.
Cryobiology ; 53(3): 330-5, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17026979

ABSTRACT

The cryoprotective effect of intracellular free high-mannose oligosaccharides (HMOS) on mammalian cells and proteins was examined by monitoring PC-12 cell viability and assaying protein kinase C (PKC)-epsilon activity. 1-Deoxymannojirimycin, an inhibitor of alpha-mannosidase, to cause an increase in intracellular free HMOS, significantly rescued PC-12 cells with 2-h freezing insult at -15 degrees C in a concentration (1-50mM)- and pretreatment time (48-72h)-dependent manner, as compared with unpretreated cells; full rescue from freezing injury was obtained with 1-deoxymannojirimycin at more than 25mM for 48-h pretreatment and more than 3mM for 72- and 96-h pretreatment. For PC-12 cells pretreated with 1-deoxymannojirimycin at 1mM for 72h, thawed cell viability after more than 8-w cryopreservation at -80 degrees C in 10% (v/v) dimethyl sulfoxide was much higher than that for cells without pretreatment. PKC-epsilon activity was well preserved after 16-h cryopreservation at -20 degrees C in the presence of mannose 9-N-acetylglucosamine 2 (Man9-GlcNAc2) (1 mM), an HMOS, while the activity was reduced to 15% without Man9-GlcNAc2. Collectively, the results of the present study suggest that intracellular free HMOS is a key molecule to protect mammalian cells and proteins from freezing injury; in other words, HMOS could be a new target for cryopreservation of mammalian cells and proteins.


Subject(s)
Cryopreservation , Mannans/metabolism , Oligosaccharides/metabolism , 1-Deoxynojirimycin/pharmacology , Animals , Cell Survival/drug effects , Cryoprotective Agents/pharmacology , Enzyme Inhibitors/pharmacology , Intracellular Fluid/metabolism , Mannans/pharmacology , Mannose/chemistry , Oligosaccharides/chemistry , PC12 Cells , Protein Kinase C-epsilon/metabolism , Rats , alpha-Mannosidase/antagonists & inhibitors
17.
J Lipid Res ; 47(6): 1146-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16520488

ABSTRACT

This study examined the effect of 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA), a newly synthesized linoleic acid derivative with cyclopropane rings instead of cis-double bonds, on protein kinase C (PKC) activity. In the in situ PKC assay with reverse-phase high-performance liquid chromatography, DCP-LA significantly activated PKC in PC-12 cells in a concentration-dependent (10 nM-100 microM) manner, with the maximal effect at 100 nM, and the DCP-LA effect was blocked by GF109203X, a PKC inhibitor, or a selective inhibitor peptide of the novel PKC isozyme PKC-epsilon. Furthermore, DCP-LA activated PKC in HEK-293 cells that was inhibited by the small, interfering RNA against PKC-epsilon. In the cell-free PKC assay, of the nine isozymes examined here, DCP-LA most strongly activated PKC-epsilon, with >7-fold potency over other PKC isozymes, in the absence of dioleoyl-phosphatidylserine and 1,2-dioleoyl-sn-glycerol; instead, the DCP-LA action was inhibited by dioleoyl-phosphatidylserine. DCP-LA also activated PKC-gamma, a conventional PKC, but to a much lesser extent compared with that for PKC-epsilon, by a mechanism distinct from PKC-epsilon activation. Thus, DCP-LA serves as a selective activator of PKC-epsilon, possibly by binding to the phosphatidylserine binding site on PKC-epsilon. These results may provide fresh insight into lipid signaling in PKC activation.


Subject(s)
Caprylates/pharmacology , Phosphatidylserines/metabolism , Protein Kinase C-epsilon/metabolism , Animals , Base Sequence , Binding Sites , Caprylates/metabolism , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Linoleic Acids/chemistry , Linoleic Acids/metabolism , Linoleic Acids/pharmacology , PC12 Cells , Phosphatidylserines/chemistry , Phosphatidylserines/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
18.
Neurotoxicology ; 27(4): 458-67, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16469385

ABSTRACT

Extracellular adenosine reduced viability of RCR-1 rat astrocytoma cells in a dose (0.3-10mM)- and treatment time (24-72h)-dependent manner. In the apoptosis assay using propidium iodide (PI) and annexin V, treatment with adenosine (1mM) for 72h increased the population of PI-negative/annexin V-positive cells, that is related to early apoptosis, and that of PI-positive/annexin V-positive cells, that is related to late apoptosis/secondary necrosis. In addition, nuclei of cells treated with adenosine (1mM) for 72h were reactive to an antibody against single-stranded DNA. Adenosine activated caspase-3, -8 and -9, but mitochondrial membrane potentials were not affected. Adenosine-induced RCR-1 cell death was significantly inhibited by 8-CPT, an antagonist of A(1) adenosine receptors, and forskolin, an adenylate cyclase activator. SQ22536, an adenylate cyclase inhibitor, alternatively, exhibited an effect similar to adenosine. CHA, an agonist of A(1) adenosine receptors, activated caspase-3 and -9, but not caspase-8. Adenosine-induced cytotoxicity of RCR-1 cells was also significantly inhibited by dipyridamole, an inhibitor of adenosine transporter, and AMDA, an inhibitor of adenosine kinase. AICAR, an activator of AMP-activated protein kinase (AMPK), reduced RCR-1 cell viability, but synergistic effect was not obtained with co-treatment with adenosine and AICAR. AICAR activated caspase-3 and -9, but not caspase-8. An additive inhibition was found in the co-presence of 8-CPT and dipyridamole. Extracellular adenosine, thus, appears to activate caspase-9 followed by the effector caspase, caspase-3, at least via two independent pathways linked to A(1) adenosine receptor-mediated adenylate cyclase inhibition and adenosine uptake into cells/conversion to AMP/activation of AMPK, possibly regardless of mitochondrial damage, thereby leading to RCR-1 cell death, dominantly by apoptosis. Moreover, caspase-8 activation could again contribute to adenosine-induced cytotoxicity, although the underlying mechanism is currently unknown. Collectively, the results of the present study may represent a new pathway for caspase activation relevant to diverse adenosine signals in cell death.


Subject(s)
Adenosine/physiology , Astrocytoma/physiopathology , Caspases/metabolism , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/physiology , Receptor, Adenosine A1/physiology , Signal Transduction/physiology , AMP-Activated Protein Kinases , Adenosine/pharmacology , Analysis of Variance , Animals , Cell Death/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry/methods , Immunohistochemistry/methods , Mitochondrial Membranes/drug effects , Models, Biological , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Tetrazolium Salts , Theophylline/analogs & derivatives , Theophylline/pharmacology , Thiazoles
19.
Neuroreport ; 17(1): 105-8, 2006 Jan 23.
Article in English | MEDLINE | ID: mdl-16361960

ABSTRACT

In the water-maze test, the linoleic acid derivative, 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA) (1 mg/kg, intraperitoneally), significantly shortened the prolonged latency for accelerated-senescence-prone mice 8 (SAMP8), reaching a level similar to the latency for accelerated-senescence-resistant mice 1 (SAMR1) as control. In the open-field test to assess motor activity, it was confirmed that the DCP-LA effect is not due to increased motor activity. In the passive avoidance test to assess fear memory, DCP-LA had no effect on the latency of acquisition and retention for SAMP8. The results of the present study, thus, suggest that DCP-LA could improve age-related learning impairment by enhancing cognitive functions.


Subject(s)
Aging/physiology , Caprylates/therapeutic use , Learning Disabilities/drug therapy , Linoleic Acid/chemistry , Aging/genetics , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Disease Models, Animal , Exploratory Behavior/drug effects , Learning Disabilities/etiology , Male , Maze Learning/drug effects , Mice , Mice, Neurologic Mutants , Motor Activity/drug effects , Reaction Time/drug effects
20.
Pain ; 120(1-2): 106-112, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16360272

ABSTRACT

The present study examined noradrenaline-induced modulation of ATP-evoked currents in dorsal root ganglion (DRG) neurons after sciatic nerve injury (transection). ATP (10 microM) generated fast/mixed type of whole-cell membrane currents, possibly as mediated via P2X(3)/P2X(3)-like receptors, and slow type of the currents, possibly as mediated via P2X(2/3) receptors, in acutely dissociated L4/5 DRG neurons, without significant difference between sham and injury group. For sham group, noradrenaline (10 microM) enhanced fast/mixed type of ATP-evoked currents in ipsilateral DRG neurons, that is not inhibited by H-7, a broad inhibitor of protein kinases, but otherwise it had no effect on slow type of the currents. For injury group, noradrenaline (10 microM) significantly potentiated slow type of ATP-evoked currents in ipsilateral DRG neurons, that is abolished by H-7 or GF109203X, a selective inhibitor of protein kinase C (PKC), while it depressed fast/mixed type of the currents. In the analysis of real-time reverse transcription-polymerase chain reaction, an increase in the mRNAs for alpha(1b), alpha(2a), alpha(2d), and beta(2) adrenergic receptors was found with the ipsilateral DRGs after sciatic nerve injury. Collectively, the results of the present study suggest that noradrenaline potentiates P2X(2/3) receptor currents by activating PKC via alpha(1) adrenergic receptors linked to G(q) protein, perhaps dominantly alpha(1b) adrenergic receptors, in DRG neurons after sciatic nerve injury. This may account for a nociceptive pathway in response to noradrenergic sprouting after peripheral nerve injury.


Subject(s)
Afferent Pathways/metabolism , Ganglia, Spinal/metabolism , Norepinephrine/metabolism , Posterior Horn Cells/metabolism , Receptors, Purinergic P2/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Adaptation, Physiological , Animals , Cells, Cultured , Male , Rats , Rats, Wistar , Receptors, Adrenergic , Receptors, Purinergic P2X
SELECTION OF CITATIONS
SEARCH DETAIL