Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Neuropsychopharmacol ; 25(9): 786-793, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35882205

ABSTRACT

BACKGROUND: Fragile X syndrome (FXS) is a genetic condition that causes a range of developmental problems, including intellectual disability, aggressive behavior, anxiety, abnormal sensory processing, and cognitive impairment. Despite intensive preclinical research in Fmr1-targeted transgenic mice, an effective treatment for FXS has yet to be developed. We previously demonstrated that ASP5736, a 5-Hydroxytryptamine (serotonin) receptor 5A receptor antagonist, ameliorated scopolamine-induced working memory deficits in mice, reference memory impairment in aged rats, and methamphetamine-induced positive symptoms and phencyclidine-induced cognitive impairment in animal models of schizophrenia. We hypothesized that ASP5736 may be effective for ameliorating similar behavior deficits in male Fmr1-targeted transgenic rats as a preclinical model of FXS. METHODS: We evaluated the effect of acute oral administration of ASP5736 on the abnormal behavior of hyperactivity (0.01, 0.1 mg/kg), prepulse inhibition (0.01, 0.03, 0.1 mg/kg), and the novel object recognition task (0.1 mg/kg) in Frmr1-knockout (KO) rats. RESULTS: Fmr1-KO rats showed body weight gain, hyperactivity, abnormal sensory motor gating, and cognitive impairment. ASP5736 (0.1 mg/kg) reversed the hyperactivity and ameliorated the sensory motor gating deficits (0.03-0.1 mg/kg). ASP5736 (0.01 mg/kg) also improved cognitive impairment. CONCLUSIONS: ASP5736 is a potential drug candidate for FXS. Further studies are needed to confirm its clinical efficacy.


Subject(s)
Fragile X Syndrome , Methamphetamine , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/therapeutic use , Fragile X Syndrome/drug therapy , Fragile X Syndrome/genetics , Guanidines , Isoquinolines , Male , Memory Disorders/drug therapy , Mice , Mice, Knockout , Phencyclidine/therapeutic use , Rats , Rats, Transgenic , Receptors, Serotonin , Scopolamine/therapeutic use , Serotonin , Serotonin Antagonists/pharmacology
2.
Bioorg Med Chem ; 28(11): 115455, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32307259

ABSTRACT

Gamma-secretase modulators (GSMs) selectively lower amyloid-ß42 (Aß42) and are therefore potential disease-modifying drugs for Alzheimer's disease (AD). Here, we report the discovery of imidazopyridine derivatives as GSMs with oral activity on not only Aß42 levels but also cognitive function. Structural optimization of the biphenyl group and pyridine-2-amide moiety of compound 1a greatly improved GSM activity and rat microsomal stability, respectively. 5-{8-[(3,4'-Difluoro[1,1'-biphenyl]-4-yl)methoxy]-2-methylimidazo[1,2-a]pyridin-3-yl}-N-methylpyridine-2-carboxamide (1o) showed high in vitro potency and brain exposure, induced a robust reduction in brain Aß42 levels, and exhibited undetectable inhibition of cytochrome p450 enzymes. Moreover, compound 1o showed excellent efficacy against cognitive deficits in AD model mice. These findings suggest that compound 1o is a promising candidate for AD therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Cognitive Dysfunction/drug therapy , Disease Models, Animal , Imidazoles/pharmacology , Pyridines/pharmacology , Administration, Oral , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Cognitive Dysfunction/metabolism , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Imidazoles/chemistry , Male , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
Eur Neuropsychopharmacol ; 28(5): 620-629, 2018 05.
Article in English | MEDLINE | ID: mdl-29571967

ABSTRACT

The 5-HT5A receptor is arguably the least understood 5-HT receptor. Despite widespread expression in human and rodent brains it lacks specific ligands. Our previous results suggest that 5-HT5A receptor antagonists may be effective against cognitive impairment in schizophrenia. In this study, using behavioral, immunohistochemical, electrophysiological and microdialysis techniques, we examined the mechanism by which ASP5736, a novel and selective 5-HT5A receptor antagonist, exerts a positive effect in animal models of cognitive impairment. We first confirmed the effect of ASP5736 on cognitive deficits in rats treated subchronically with phencyclidine hydrochloride (PCP) using an attentional set shifting task. Subsequently, we identified 5-HT5A receptors in dopaminergic (DAergic) neurons and parvalbumin (PV)-positive interneurons in the ventral tegmental area (VTA) and in PV-positive interneurons in the medial prefrontal cortex (mPFC). Burst firing of the DAergic cells in the parabrachial pigmental nucleus (PBP) in the VTA, which predominantly project to the mPFC, was significantly enhanced by treatment with ASP5736. In contrast, ASP5736 exerted no significant effect on either the firing rate or burst firing in the DA cells in the paranigral nucleus (PN), that project to the nucleus accumbens (N. Acc.). ASP5736 increased the release of DA and gamma-aminobutyric acid (GABA) in the mPFC of subchronically PCP-treated rats. These results support our hypothesis that ASP5736 might block the inhibitory 5-HT5A receptors on DAergic neurons in the VTA that project to the mPFC, and interneurons in the mPFC, and thereby improve cognitive impairment by preferentially enhancing DAergic and GABAergic neurons in the mPFC.


Subject(s)
Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Guanidines/pharmacology , Isoquinolines/pharmacology , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Schizophrenia/complications , Schizophrenia/drug therapy , Action Potentials/physiology , Animals , Cognitive Dysfunction/chemically induced , Discrimination, Psychological/drug effects , Dopamine/metabolism , Dopaminergic Neurons/physiology , Interneurons/physiology , Male , Phencyclidine , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Rats , Schizophrenic Psychology , Serotonin Antagonists/pharmacology , Ventral Tegmental Area/physiology , gamma-Aminobutyric Acid/metabolism
4.
Eur J Pharmacol ; 810: 26-35, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28552344

ABSTRACT

KCNH3 (BEC1) is a member of the ether-à-go-go (KCNH) family of voltage-gated K+ channels. The aim of this study was to determine the pharmacological profiles in vitro and in vivo of a KCNH3 inhibitor N-(4-fluorophenyl)-N'-phenyl-N''-(pyrimidin-2-ylmethyl)-1,3,5-triazine-2,4,6-triamine (ASP2905). We analyzed the effects of ASP2905 on channel activity in vitro and its neuropharmacological properties in young and aged rats as well as in mice. ASP2905 potently inhibited potassium currents in CHO cells expressing KCNH3 (IC50 = 9.0nM). In contrast, ASP2905 (≤ 10µM) minimally bound with low affinities to 55 transmembrane proteins. ASP2905 (0.1µM, 1µM) decreased the frequency of spontaneous inhibitory postsynaptic currents in cultured rat hippocampal neurons. In mice, ASP2905 reversed the disruption of spontaneous alternation behavior induced by MK-801 and scopolamine (minimum effective dose of ASP2905: 0.0625mg/kg, po). ASP2905 ameliorated the cognitive deficits of aged rats in step-through passive avoidance (0.0313 and 0.0625mg/kg, po) and Morris water-maze tasks (0.01mg/kg, po) and effectively penetrated the brain. The mean plasma and brain concentrations of ASP2905 reached their maxima (Cmax = 0.399ng/ml and 1.77ng/g, respectively) 1h after a single oral administration and then decreased (t1/2 = 1.5-1.6h) (brain plasma ratio = 2.7-4.9). The present study suggests that ASP2905 is a selective, orally administered inhibitor of KCNH3, which can enhance cognitive performance.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Pyrimidines/pharmacology , Triazines/pharmacology , Animals , Behavior, Animal/drug effects , CHO Cells , Cognition/drug effects , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Inhibitory Postsynaptic Potentials/drug effects , Male , Maze Learning/drug effects , Mice , Neurochemistry , Rats
5.
J Pharmacol Sci ; 127(3): 362-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25837935

ABSTRACT

Despite the human 5-HT5A receptor being cloned in 1994, the biological function of this receptor has not been extensively characterized due to a lack of specific ligands. We recently reported that the selective 5-HT5A receptor antagonist ASP5736 ameliorated cognitive impairment in several animal models of schizophrenia. Given that areas of the brain with high levels of 5-HT5A receptor expression, such as the hippocampus and cerebral cortex, have important functions in cognition and memory, we evaluated the chemically diverse, potent and brain-penetrating 5-HT5A receptor antagonists ASP5736, AS2030680, and AS2674723 in rodent models of cognitive dysfunction associated with dementia. Each of these compounds exhibited a high affinity for recombinant 5-HT5A receptors that was comparable to that of the non-selective ligand of this receptor, lysergic acid diethylamide (LSD). Although each compound had a low affinity for other receptors, 5-HT5A was the only receptor for which all three compounds had a high affinity. Each of the three compounds ameliorated scopolamine-induced working memory deficit in mice and improved reference memory impairment in aged rats at similar doses. Further, ASP5736 decreased the binding of LSD to 5-HT5A receptors in the olfactory bulb of rats in a dose-dependent manner and occupied 15%-50% of brain 5-HT5A receptors at behaviorally effective doses. These results indicate that the 5-HT5A receptor is involved in learning and memory and that treatment with 5-HT5A receptor antagonists might be broadly effective for cognitive impairment associated with not only schizophrenia but also dementia.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/psychology , Guanidines/therapeutic use , Isoquinolines/therapeutic use , Memory Disorders/drug therapy , Receptors, Serotonin/metabolism , Scopolamine/pharmacology , Serotonin Antagonists/therapeutic use , Aging , Animals , Brain/metabolism , Cognition Disorders/chemically induced , Dementia/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Guanidines/pharmacology , Isoquinolines/pharmacology , Lysergic Acid Diethylamide/metabolism , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Mice, Inbred Strains , Olfactory Bulb/metabolism , Rats, Wistar , Schizophrenia/drug therapy , Serotonin Antagonists/pharmacology
6.
Eur Neuropsychopharmacol ; 24(10): 1698-708, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25108314

ABSTRACT

We recently identified ASP5736, (N-(diaminomethylene)-1-(3,5-difluoropyridin-4-yl)-4-fluoroisoquinoline-7-carboxamide (2E)-but-2-enedioate), a novel antagonist of 5-HT5A receptor, and here describe the in vitro and in vivo characterization of this compound. ASP5736 exhibited a high affinity for the human 5-HT5A receptor (Ki = 3.6 ± 0.66 nM) and antagonized 5-carboxamidotryptamine (5-CT)-induced Ca(2+) influx in human cells stably expressing the 5-HT5A receptor with approximately 200-fold selectivity over other receptors, including other 5-HT receptor subtypes, enzymes, and channels except human 5-HT2c receptor (Ki = 286.8 nM) and 5-HT7 receptor (Ki = 122.9 nM). Further, ASP5736 dose-dependently antagonized the 5-CT-induced decrease in cAMP levels in HEK293 cells stably expressing the 5-HT5A receptor. We then evaluated the effects of ASP5736 on cognitive impairments in several animal models of schizophrenia. Working memory deficit in MK-801-treated mice and visual learning deficit in neonatally phencyclidine (PCP)-treated mice were both ameliorated by ASP5736. In addition, ASP5736 also attenuated MK-801- and methamphetamine (MAP)-induced hyperactivity in mice without causing sedation, catalepsy, or plasma prolactin increase. The addition of olanzapine did not affect ASP5736-induced cognitive enhancement, and neither the sedative nor cataleptogenic effects of olanzapine were worsened by ASP5736. These results collectively suggest that ASP5736 is a novel and potent 5-HT5A receptor antagonist that not only ameliorates positive-like symptoms but also cognitive impairments in animal models of schizophrenia, without adverse effects. Present studies also indicate that ASP5736 holds potential to satisfy currently unmet medical needs for the treatment of schizophrenia by either mono-therapy or co-administered with commercially available antipsychotics.


Subject(s)
Antipsychotic Agents/pharmacology , Guanidines/pharmacology , Isoquinolines/pharmacology , Schizophrenia/drug therapy , Serotonin Antagonists/pharmacology , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Calcium/metabolism , Catalepsy/drug therapy , Catalepsy/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Cyclic AMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Guanidines/chemistry , Guanidines/pharmacokinetics , HEK293 Cells , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Male , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Motor Activity/physiology , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Schizophrenia/physiopathology , Serotonin Antagonists/chemistry , Serotonin Antagonists/pharmacokinetics
7.
Neuropharmacology ; 79: 412-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24373902

ABSTRACT

γ-Secretase is the enzyme responsible for the intramembranous proteolysis of various substrates, such as amyloid precursor protein (APP) and Notch. Amyloid-ß peptide 42 (Aß42) is produced through the sequential proteolytic cleavage of APP by ß- and γ-secretase and causes the synaptic dysfunction associated with memory impairment in Alzheimer's disease. Here, we identified a novel cyclohexylamine-derived γ-secretase modulator, {(1R*,2S*,3R*)-3-[(cyclohexylmethyl)(3,3-dimethylbutyl)amino]-2-[4-(trifluoromethyl)phenyl]cyclohexyl}acetic acid (AS2715348), that may inhibit this pathological response. AS2715348 was seen to reduce both cell-free and cellular production of Aß42 without increasing levels of APP ß-carboxyl terminal fragment or inhibiting Notch signaling. Additionally, the compound increased Aß38 production, suggesting a shift of the cleavage site in APP. The inhibitory potency of AS2715348 on endogenous Aß42 production was similar across human, mouse, and rat cells. Oral administration with AS2715348 at 1 mg/kg and greater significantly reduced brain Aß42 levels in rats, and no Notch-related toxicity was observed after 28-day treatment at 100 mg/kg. Further, AS2715348 significantly ameliorated cognitive deficits in APP-transgenic Tg2576 mice. Finally, AS2715348 significantly reduced brain Aß42 levels in cynomolgus monkeys. These findings collectively show the promise for AS2715348 as a potential disease-modifying drug for Alzheimer's disease.


Subject(s)
Acetates/pharmacology , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Brain/drug effects , Cyclohexylamines/pharmacology , Neuroprotective Agents/pharmacology , Acetates/adverse effects , Acetates/pharmacokinetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cognition/drug effects , Cyclohexylamines/adverse effects , Cyclohexylamines/pharmacokinetics , Disease Models, Animal , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Molecular Structure , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Nootropic Agents/adverse effects , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Notch/metabolism
8.
Eur J Pharmacol ; 703(1-3): 53-61, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23276665

ABSTRACT

Alzheimer's disease is characterized by a progressive decline in cognitive function and involves ß-amyloid (Aß) in its pathogenesis. To characterize cognitive deficits associated with Aß accumulation, we analyzed PS1/APP mice overexpressing mutant presenilin-1 (PS1, M146L; line 6.2) and amyloid precursor protein (APP, K670N/M671L; line Tg2576), a mouse model of Alzheimer's disease with accelerated Aß production. Age-dependent changes in working and spatial memory behaviors were investigated using Y-maze and Morris water maze tasks, respectively, in female PS1/APP mice at ages of 2, 4, 6, and 12 months. Significant deficits in working and spatial memory were observed from 4 and 6 months of age, respectively. Acute single-dose administrations of memantine, a low-to-moderate-affinity N-methyl-d-aspartate (NMDA) antagonist, showed improvements in working memory deficits at 4 months of age, whereas donepezil, an acetylcholinesterase (AChE) inhibitor, did not. However, both drugs improved spatial memory dysfunction at 6 months of age at therapeutically relevant doses. No age-related dramatic changes were observed in expression levels of several proteins relating to memory dysfunction and also the mechanisms of donepezil and memantine in the cerebral cortex of PS1/APP mice until 6 months of age. Taken together, these results suggest dysfunctions in cholinergic and/or glutamatergic transmissions may be involved in the cognitive deficits associated with Aß toxicity. Since donepezil and memantine have been widely used for treating patients of Alzheimer's disease, these results also suggest that cognitive deficits in PS1/APP mice assessed in the Y-maze and Morris water maze tasks are a useful animal model for evaluating novel Alzheimer's disease therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Indans/therapeutic use , Memantine/therapeutic use , Memory Disorders/drug therapy , Nootropic Agents/therapeutic use , Piperidines/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Cholinesterase Inhibitors/blood , Cholinesterase Inhibitors/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Donepezil , Female , Indans/blood , Indans/pharmacology , Maze Learning , Memantine/blood , Memantine/pharmacology , Memory/drug effects , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Nootropic Agents/blood , Nootropic Agents/pharmacology , Piperidines/blood , Piperidines/pharmacology , Receptors, AMPA/metabolism
9.
J Neurochem ; 125(3): 465-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23240999

ABSTRACT

Given that amyloid-ß 42 (Aß42) is believed to be a culprit in Alzheimer's disease (AD), reducing Aß42 production should be a potential therapeutic approach. γ-Secretase modulators (GSMs) cause selective reduction of Aß42 or both reduction of Aß42 and Aß40 without affecting total Aß through shifting the γ-cleavage position in amyloid precursor protein. We recently reported on GSM-2, one of the second-generation GSMs, that selectively reduced brain Aß42 level and significantly ameliorated cognitive deficits in plaque-free 5.5-month-old Tg2576 AD model mice. Here, we investigated the effects of GSM-2 on 10-, 14-, and 18-month-old mice which had age-dependent increase in amyloid plaques. Eight-day treatment with GSM-2 significantly ameliorated cognitive deficits measured by Y-maze task in the mice of any age. However, GSM-2 reduced brain soluble Aß42 only in 10-month-old mice. In contrast, GSM-2 markedly reduced newly synthesized soluble Aß42 in both 10- and 18-month-old mice with similar efficacy when measured using the stable isotope-labeling technique, suggesting that nascent Aß42 plays a more significant role than plaque-associated soluble Aß42 in the cognitive deterioration of Tg2576 mice. These findings further indicate the potential utility of approach to reducing Aß42 synthesis in AD therapeutic regimens.


Subject(s)
Alzheimer Disease/complications , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Cognition Disorders/metabolism , Enzyme Inhibitors/therapeutic use , Peptide Fragments/metabolism , Acetates/pharmacology , Acetates/therapeutic use , Age Factors , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/therapeutic use , Chromatography, Liquid , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme-Linked Immunosorbent Assay , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mass Spectrometry , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/immunology , Piperidines/pharmacology , Piperidines/therapeutic use
10.
Eur J Pharmacol ; 685(1-3): 59-69, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22542656

ABSTRACT

Hypofunction of brain N-methyl-d-aspartate (NMDA) receptors has been implicated in psychiatric disorders such as schizophrenia and Alzheimer's disease. Inhibition of glycine transporter-1 (GlyT1) is expected to increase glycine, a co-agonist of the NMDA receptor and, consequently, to facilitate NMDA receptor function. We have identified ASP2535 (4-[3-isopropyl-5-(6-phenyl-3-pyridyl)-4H-1,2,4-triazol-4-yl]-2,1,3-benzoxadiazole) as a novel GlyT1 inhibitor, and here describe our in vitro and in vivo characterization of this compound. ASP2535 potently inhibited rat GlyT1 (IC(50)=92 nM) with 50-fold selectivity over rat glycine transporter-2 (GlyT2). It showed minimal affinity for many other receptors except for µ-opioid receptors (IC(50)=1.83 µM). Oral administration of ASP2535 dose-dependently inhibited ex vivo [(3)H]-glycine uptake in mouse cortical homogenate, suggesting good brain permeability. This profile was confirmed by pharmacokinetic analysis. We then evaluated the effect of ASP2535 on animal models of cognitive impairment in schizophrenia and Alzheimer's disease. Working memory deficit in MK-801-treated mice and visual learning deficit in neonatally phencyclidine (PCP)-treated mice were both attenuated by ASP2535 (0.3-3mg/kg, p.o. and 0.3-1mg/kg, p.o., respectively). ASP2535 (1-3mg/kg, p.o.) also improved the PCP-induced deficit in prepulse inhibition in rats. Moreover, the working memory deficit in scopolamine-treated mice and the spatial learning deficit in aged rats were both attenuated by ASP2535 (0.1-3mg/kg, p.o. and 0.1mg/kg, p.o., respectively). These studies provide compelling evidence that ASP2535 is a novel and centrally-active GlyT1 inhibitor that can improve cognitive impairment in animal models of schizophrenia and Alzheimer's disease, suggesting that ASP2535 may satisfy currently unmet medical needs for the treatment of these diseases.


Subject(s)
Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Oxadiazoles/pharmacology , Schizophrenia/drug therapy , Triazoles/pharmacology , Administration, Oral , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Male , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Mice , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Permeability , Rats , Rats, Wistar , Schizophrenia/physiopathology , Triazoles/administration & dosage , Triazoles/pharmacokinetics
11.
J Neurosci ; 32(6): 2037-50, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323718

ABSTRACT

γ-Secretase inhibitors (GSIs) reduce amyloid-ß (Aß) peptides but inevitably increase the ß-C-terminal fragment (ß-CTF) of amyloid precursor protein (APP), potentially having undesirable effects on synapses. In contrast, γ-secretase modulators (GSMs) reduce Aß42 without increasing ß-CTF. Although the Aß-lowering effects of these compounds have been extensively studied, little effort has been made to investigate their effects on cognition. Here, we compared the effects of two GSIs--(2S)-2-hydroxy-3-methyl-N-[(2S)-1-{[(1S)-3-methyl-2-oxo-2,3,4,5-tetrahydro-1H-3-benzazepin-1-yl]amino}-1-oxopropan-2-yl]butanamide (LY450139, semagacestat) and (2R)-2-[[(4-chlorophenyl)sulfonyl][[2-fluoro-4-(1,2,4-oxazol-3-yl)phenyl]methyl]amino-5,5,5-trifluoropentanamide (BMS-708163)--and a second-generation GSM [{(2S,4R)-1-[(4R)-1,1,1-trifluoro-7-methyloctan-4-yl]-2-[4-(trifluoromethyl)phenyl]piperidin-4-yl}acetic acid (GSM-2)] on spatial working memory in APP-transgenic (Tg2576) and nontransgenic mice using the Y-maze task. While acute dosing with either GSI ameliorated memory deficits in 5.5-month-old Tg2576 mice, these effects disappeared after 8 d subchronic dosing. Subchronic dosing with either GSI rather impaired normal cognition in 3-month-old Tg2576 mice, with no inhibition on the processing of other γ-secretase substrates, such as Notch, N-cadherin, or EphA4, in the brain. LY450139 also impaired normal cognition in wild-type mice; however, the potency was 10-fold lower than that in Tg2576 mice, indicating an APP-dependent mechanism likely with ß-CTF accumulation. Immunofluorescence studies revealed that the ß-CTF accumulation was localized in the presynaptic terminals of the hippocampal stratum lucidum and dentate hilus, implying an effect on presynaptic function in the mossy fibers. In contrast, both acute and subchronic dosing with GSM-2 significantly ameliorated memory deficits in Tg2576 mice and did not affect normal cognition in wild-type mice. We demonstrated a clear difference between GSI and GSM in effects on functional consequences, providing new insights into strategies for developing these drugs against Alzheimer's disease.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Azepines/pharmacology , Cognition/drug effects , Cognition/physiology , Protease Inhibitors/pharmacology , Alanine/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Female , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic
12.
Proc Natl Acad Sci U S A ; 105(16): 6133-8, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18413613

ABSTRACT

The G protein-coupled receptor (GPCR) family is highly diversified and involved in many forms of information processing. SREB2 (GPR85) is the most conserved GPCR throughout vertebrate evolution and is expressed abundantly in brain structures exhibiting high levels of plasticity, e.g., the hippocampal dentate gyrus. Here, we show that SREB2 is involved in determining brain size, modulating diverse behaviors, and potentially in vulnerability to schizophrenia. Mild overexpression of SREB2 caused significant brain weight reduction and ventricular enlargement in transgenic (Tg) mice as well as behavioral abnormalities mirroring psychiatric disorders, e.g., decreased social interaction, abnormal sensorimotor gating, and impaired memory. SREB2 KO mice showed a reciprocal phenotype, a significant increase in brain weight accompanying a trend toward enhanced memory without apparent other behavioral abnormalities. In both Tg and KO mice, no gross malformation of brain structures was observed. Because of phenotypic overlap between SREB2 Tg mice and schizophrenia, we sought a possible link between the two. Minor alleles of two SREB2 SNPs, located in intron 2 and in the 3' UTR, were overtransmitted to schizophrenia patients in a family-based sample and showed an allele load association with reduced hippocampal gray matter volume in patients. Our data implicate SREB2 as a potential risk factor for psychiatric disorders and its pathway as a target for psychiatric therapy.


Subject(s)
Brain/pathology , Genetic Predisposition to Disease/genetics , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Schizophrenia/genetics , Schizophrenia/pathology , Alleles , Amino Acid Sequence , Animals , Behavior, Animal , Evolution, Molecular , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Molecular Sequence Data , Organ Size/genetics , Polymorphism, Single Nucleotide , Schizophrenic Psychology
13.
J Pharmacol Exp Ther ; 323(2): 708-19, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17684118

ABSTRACT

Central adenosine A(2A) receptor is a promising target for drugs to treat Parkinson's disease (PD), and the central blockade of adenosine A(1) receptor improves cognitive function. In the present study, we investigated the effect of a novel adenosine A(1) and A(2A) dual antagonist, 5-[5-amino-3-(4-fluorophenyl) pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), in animal models of PD and cognition. The binding affinities of ASP5854 for human A(1) and A(2A) receptors were 9.03 and 1.76 nM, respectively, with higher specificity and no species differences. ASP5854 also showed antagonistic action on A(1) and A(2A) agonist-induced increases of intracellular Ca(2+) concentration. ASP5854 ameliorated A(2A) agonist 2-[p-(2-carboxyethyl) phenethylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680)- and haloperidol-induced catalepsy in mice, with the minimum effective doses of 0.32 and 0.1 mg/kg, respectively, and it also improved haloperidol-induced catalepsy in rats at doses higher than 0.1 mg/kg. In unilateral 6-hydroxydopamine-lesioned rats, ASP5854 significantly potentiated l-dihydroxyphenylalanine (L-DOPA)-induced rotational behavior at doses higher than 0.032 mg/kg. ASP5854 also significantly restored the striatal dopamine content reduced by 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment in mice at doses higher than 0.1 mg/kg. Furthermore, in the rat passive avoidance test, ASP5854 significantly reversed the scopolamine-induced memory deficits, whereas the specific adenosine A(2A) antagonist 8-((E)-2-(3,4-dimethoxyphenyl)ethenyl)-1,3-diethyl-7-methyl-3,7-dihydro-1H-purine-2,6-dione (KW-6002; istradefylline) did not. Scopolamine- or 5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate) (MK-801)-induced impairment of spontaneous alternation in the mouse Y-maze test was ameliorated by ASP5854, whereas KW-6002 did not exert improvement at therapeutically relevant dosages. These results demonstrate that the novel, selective, and orally active dual adenosine A(1) and A(2A) receptors antagonist ASP5854 improves motor impairments, is neuroprotective via A(2A) antagonism, and also enhances cognitive function through A(1) antagonism.


Subject(s)
Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Aminopyridines/therapeutic use , Cognition Disorders/drug therapy , MPTP Poisoning/drug therapy , Piperazines/therapeutic use , Adenosine/analogs & derivatives , Adenosine/pharmacology , Aminopyridines/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dopamine/analysis , Dopamine/metabolism , Haloperidol/pharmacology , Humans , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Phenethylamines/pharmacology , Piperazines/pharmacology , Rats , Rats, Inbred F344 , Rats, Wistar , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Scopolamine/pharmacology
14.
J Biol Chem ; 277(43): 41254-8, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12185074

ABSTRACT

Glutathione peroxidase catalyzes the reduction of hydrogen peroxide and organic hydroperoxide by glutathione and functions in the protection of cells against oxidative damage. Glutathione peroxidase exists in several forms that differ in their primary structure and localization. We have also shown that selenoprotein P exhibits a glutathione peroxidase-like activity (Saito, Y., Hayashi, T., Tanaka, A., Watanabe, Y., Suzuki, M., Saito, E., and Takahashi, K. (1999) J. Biol. Chem. 274, 2866-2871). To understand the physiological significance of the diversity among these enzymes, a comparative study on the peroxide substrate specificity of three types of ubiquitous glutathione peroxidase (cellular glutathione peroxidase, phospholipid hydroperoxide glutathione peroxidase, and extracellular glutathione peroxidase) and of selenoprotein P purified from human origins was done. The specific activities and kinetic parameters against two hydroperoxides (hydrogen peroxide and phosphatidylcholine hydroperoxide) were determined. We next examined the thiol specificity and found that thioredoxin is the preferred electron donor for selenoprotein P. These four enzymes exhibit different peroxide and thiol specificities and collaborate to protect biological molecules from oxidative stress both inside and outside the cells.


Subject(s)
Glutathione Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Proteins/metabolism , Sulfhydryl Compounds/metabolism , Kinetics , Selenoprotein P , Selenoproteins , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...