Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pharmacol Sci ; 153(3): 175-182, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37770159

ABSTRACT

We previously found that pituitary adenylate cyclase-activating polypeptide (PACAP)-deficient (PACAP-/-) mice exhibit dendritic spine morphology impairment and neurodevelopmental disorder (NDD)-like behaviors such as hyperactivity, increased novelty-seeking behavior, and deficient pre-pulse inhibition. Recent studies have indicated that rodent models of NDDs (e.g., attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder) show abnormalities in the axon initial segment (AIS). Here, we revealed that PACAP-/- mice exhibited a longer AIS length in layer 2/3 pyramidal neurons of the primary somatosensory barrel field compared with wild-type control mice. Further, we previously showed that a single injection of atomoxetine, an ADHD drug, improved hyperactivity in PACAP-/- mice. In this study, we found that repeated treatments of atomoxetine significantly improved AIS abnormality along with hyperactivity in PACAP-/- mice. These results suggest that AIS abnormalities are associated with NDDs-like behaviors in PACAP-/- mice. Thus, improvement in AIS abnormalities will be a novel drug therapy for NDDs.

2.
Biochem Biophys Res Commun ; 631: 146-151, 2022 11 26.
Article in English | MEDLINE | ID: mdl-36194909

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved pleiotropic neuropeptide, implicated in emotional stress responses and anxiety-related disorders. Here, we examined whether our recently developed small-molecule non-peptide PACAP receptor antagonists could ameliorate anxiety-like behaviors induced by acute restraint stress in mice. The antagonists PA-9 and its derivative PA-915 improved anxiety-like behaviors in mice subjected to restraint stress. An anxiolytic effect was observed with single acute dose, suggesting their fast-acting properties. PA-915 demonstrated a statistically significant anxiolytic effect whereas fluoxetine did not. These results indicate the potential of PAC1 antagonists as a novel treatment for anxiety.


Subject(s)
Anti-Anxiety Agents , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Fluoxetine , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide
3.
Biochem Biophys Res Commun ; 519(3): 626-632, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31540692

ABSTRACT

Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder, characterized by impaired social interaction, repetitive behavior and restricted interests. Although the molecular etiology of ASD remains largely unknown, recent studies have suggested that de novo mutations are significantly involved in the risk of ASD. We and others recently identified spontaneous de novo mutations in PKD2, a protein kinase D family member, in sporadic ASD cases. However, the biological significance of the de novo PKD2 mutations and the role of PKD2 in brain development remain unclear. Here, we performed functional analysis of PKD2 in cortical neuron development using in utero electroporation. PKD2 is highly expressed in cortical neural stem cells in the developing cortex and regulates cortical neuron development, including the neuronal differentiation of neural stem cells and migration of newborn neurons. Importantly, we determined that the ASD-associated de novo mutations impair the kinase activity of PKD2, suggesting that the de novo PKD2 mutations can be a risk factor for the disease by loss of function of PKD2. Our current findings provide novel insight into the molecular and cellular pathogenesis of ASD.


Subject(s)
Autism Spectrum Disorder/enzymology , Cerebral Cortex/metabolism , Neurons/metabolism , TRPP Cation Channels/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Embryonic Development , HEK293 Cells , Humans , Neurons/cytology
4.
J Pharmacol Sci ; 140(4): 321-324, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31257060

ABSTRACT

Since induced pluripotent stem cells (iPSCs) were generated from mice and humans by Professor Shinya Yamanaka et al. in 2006 and 2007, respectively, a variety of human-derived cells have been generated, including myocardial, liver, retinal pigment epithelial, and neuronal cells. These iPSCs are now used not only in clinical research focusing on regeneration and transplantation in diverse medical fields, but also in molecular and cellular pathological studies. Importantly, by using human-derived iPSCs, it has become possible to conduct drug discovery research that more accurately models the pathology of human diseases. In research on psychiatric disorders, iPSC-related technologies, which have enabled the use of neuronal cells that carry the genetic information of the patients, will be important for elucidating not only the molecular and cellular etiology of psychiatric disorders but also the molecular mechanisms of drug action in these disorders. This review outlines the pharmacological research of psychiatric disorders that utilizes iPSC-related technologies.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Mental Disorders/physiopathology , Animals , Cell Culture Techniques/methods , Drug Discovery/methods , Humans , Neurons/physiology
5.
Int J Neuropsychopharmacol ; 20(5): 410-421, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28034961

ABSTRACT

Background: Isolation-reared mice show social encounter-induced hyperactivity with activation of prefrontal serotonergic and dopaminergic systems, but it is not known whether this stress response is observed in other pathological conditions. Here we examined whether the social encounter stimulation induces abnormal behavior during withdrawal in chronic methamphetamine-treated mice. Methods: To induce methamphetamine-induced behavioral sensitization, male mice were injected with methamphetamine (1 mg/kg) once daily for 7 days. Results: The encounter with an intruder elicited hyperactivity 24 h after the last injection of methamphetamine in methamphetamine-sensitized mice. This response was observed even as long as 2 weeks after withdrawal of methamphetamine. The encounter increased c-Fos expression in the prefrontal cortex, dorsal raphe nucleus and ventral tegmental area in methamphetamine-sensitized mice, while it did not in control mice. Furthermore, the encounter increased extracellular serotonin (5-HT) and dopamine, but not noradrenaline, levels in the prefrontal cortex in methamphetamine-sensitized mice. Local injection of 5,7-dihydroxytryptamine and 6-hydroxydopamine into the prefrontal cortex attenuated encounter-induced hyperactivity in methamphetamine-sensitized mice and it markedly decreased prefrontal 5-HT and dopamine levels, respectively. Pharmacological analysis showed that the encounter-induced hyperactivity is mediated by dopamine D1 receptors and 5-HT2A receptors and attenuated by anxiolytics and antidepressants such as diazepam, osemozotan and selective 5-HT reuptake inhibitors. The effect of paroxetine was blocked by the 5-HT3 receptor antagonist azasetron. Conclusions: The present study shows that psychological stress elicits hyperactivity with activation of prefrontal 5-HT and dopamine systems in methamphetamine-dependent mice and suggests that the abnormal behavior is associated with anxiety and depression.


Subject(s)
Central Nervous System Stimulants/toxicity , Dopamine/metabolism , Hyperkinesis/chemically induced , Methamphetamine/toxicity , Prefrontal Cortex/metabolism , Serotonin/metabolism , 5,7-Dihydroxytryptamine/toxicity , Animals , Desipramine/therapeutic use , Dopamine Agents/pharmacology , Enzyme Inhibitors/therapeutic use , Hyperkinesis/drug therapy , Locomotion/drug effects , Male , Mice , Microdialysis , Oxidopamine/toxicity , Prefrontal Cortex/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Serotonin Agents/pharmacology , Social Behavior , Time Factors
6.
Horm Behav ; 96: 130-136, 2017 11.
Article in English | MEDLINE | ID: mdl-28942000

ABSTRACT

Recent studies have reported that oxytocin ameliorates behavioral abnormalities in both animal models and individuals with autism spectrum disorders (ASD). However, the mechanisms underlying the ameliorating effects of oxytocin remain unclear. In this study, we examined the effects of intranasal oxytocin on impairments in social interaction and recognition memory in an ASD mouse model in which animals are prenatally exposed to valproic acid (VPA). We found that a single intranasal administration of oxytocin restored social interaction deficits for up to 2h in mice prenatally exposed to VPA, but there was no effect on recognition memory impairments. Additionally, administration of oxytocin across 2weeks improved prenatal VPA-induced social interaction deficits for at least 24h. In contrast, there were no effects on the time spent sniffing in control mice. Immunohistochemical analysis revealed that intranasal administration of oxytocin increased c-Fos expression in the paraventricular nuclei (PVN), prefrontal cortex, and somatosensory cortex, but not the hippocampal CA1 and CA3 regions of VPA-exposed mice, suggesting the former regions may underlie the effects of oxytocin. These findings suggest that oxytocin attenuates social interaction deficits through the activation of higher cortical areas and the PVN in an ASD mouse model.


Subject(s)
Autistic Disorder/chemically induced , Oxytocin/therapeutic use , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/psychology , Recognition, Psychology/drug effects , Social Learning/drug effects , Valproic Acid/adverse effects , Animals , Autistic Disorder/drug therapy , Autistic Disorder/pathology , Autistic Disorder/psychology , Behavior, Animal/drug effects , Disease Models, Animal , Female , Hippocampus/drug effects , Interpersonal Relations , Male , Mice , Mice, Inbred ICR , Oxytocin/pharmacology , Prefrontal Cortex/drug effects , Pregnancy , Proto-Oncogene Proteins c-fos/metabolism , Social Behavior
7.
Neurochem Res ; 41(10): 2574-2584, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27300699

ABSTRACT

Valproic acid (VPA) is a multi-target drug and an inhibitor of histone deacetylase (HDAC). We have previously demonstrated that prenatal exposure to VPA at embryonic day 12.5 (E12.5), but not at E14.5, causes autism-like behavioral abnormalities in male mouse offspring. We have also found that prenatal VPA exposure causes transient histone hyperacetylation in the embryonic brain, followed by decreased neuronal cell numbers in the prefrontal and somatosensory cortices after birth. In the present study, we examined whether prenatal HDAC inhibition affects neuronal maturation in primary mouse cortical neurons. Pregnant mice were injected intraperitoneally with VPA (500 mg/kg) and the more selective HDAC inhibitor trichostatin A (TSA; 500 µg/kg) at E12.5 or E14.5, and primary neuronal cultures were prepared from the cerebral cortices of their embryos. Prenatal exposure to VPA at E12.5, but not at E14.5, decreased total number, total length, and complexity of neuronal dendrites at 14 days in vitro (DIV). The effects of VPA weakened at 21 DIV. Exposure to TSA at E12.5, but not at E14.5, also delayed maturation of cortical neurons. In addition, real-time quantitative PCR revealed that the prenatal exposure to TSA decreased neuroligin-1 (Nlgn1), Shank2, and Shank3 mRNA levels and increased contactin-associated protein-like 2 mRNA level. The delay in neuronal maturation was also observed in Nlgn1-knockdown cells, which were transfected with Nlgn1 siRNA. These findings suggest that prenatal HDAC inhibition causes changes in gene expression of autism-related molecules linked to a delay of neuronal maturation.


Subject(s)
Autistic Disorder/genetics , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylase Inhibitors/pharmacology , Neurogenesis/drug effects , Neurons/drug effects , Prenatal Exposure Delayed Effects/metabolism , Animals , Autistic Disorder/pathology , Female , Histone Deacetylases/metabolism , Histones/metabolism , Mice , Neurons/metabolism , Pregnancy , Valproic Acid/pharmacology
8.
Int J Neuropsychopharmacol ; 18(11): pyv062, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26025781

ABSTRACT

BACKGROUND: Reduced motivation is an important marker of psychiatric disorders, including depression. We describe the female encounter test, a novel method of evaluating reward-seeking behavior in mice. METHODS: The test apparatus consists of three open chambers, formed with partitions that allow the animal to move freely from one chamber to another. A test male mouse is habituated in the apparatus, and subsequently a female and male mouse are introduced into a wire-mesh box in the left and right chamber, respectively. The time the test male mouse spends in the female or male area is measured for 10 min. RESULTS: All six strains of mice tested showed a significant preference for female encounters. The preference was observed in 7-30-week-old mice. The preference was blocked by castration of the resident male test mouse, and was not affected by the phase of the menstrual cycle of the female intruder. The preference was impaired in mouse models of depression, including social isolation-reared, corticosterone-treated, and lipopolysaccharide-treated mice. The impairment was alleviated by fluvoxamine in isolation-reared and lipopolysaccharide-treated mice, and it was improved by the metabotropic glutamate 2/3 receptor antagonist LY341495 in corticosterone-treated mice. Encounter with a female, but not male, mouse increased c-Fos expression in the nucleus accumbens shell of test male mice. Furthermore, both the preference and encounter-induced increases in c-Fos expression were blocked by dopamine D1 and D2 receptor antagonists. CONCLUSIONS: These findings indicate that motivation in adult male mice can be easily evaluated by quantitating female encounters.


Subject(s)
Mice/psychology , Motivation , Psychological Tests , Reward , Social Behavior , Amino Acids/pharmacology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Castration , Choice Behavior/drug effects , Choice Behavior/physiology , Corticosterone , Depressive Disorder/diagnosis , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Disease Models, Animal , Equipment Design , Estrous Cycle , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluvoxamine/pharmacology , Lipopolysaccharides , Male , Mice/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Social Isolation , Xanthenes/pharmacology
9.
Behav Pharmacol ; 26(7 Spec No): 681-90, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25932719

ABSTRACT

We have recently found that isolation-reared mice show hyperactivity during an encounter with an intruder. However, it is not known whether encounter-induced hyperactivity may model some aspects of psychiatric disorders. The present study examined the pharmacological profile of encounter-induced hyperactivity in isolation-reared mice. Encounter-induced hyperactivity was reduced by acute administration of various antidepressants including the tricyclic antidepressant desipramine (10 mg/kg), the selective serotonin (5-HT) reuptake inhibitors fluvoxamine (10 mg/kg) and paroxetine (10 mg/kg), the 5-HT/noradrenaline reuptake inhibitors venlafaxine (10 mg/kg) and duloxetine (10 mg/kg), the antipsychotic drug risperidone (0.01 mg/kg), the 5-HT2 antagonist ritanserin (1 mg/kg), and the glucocorticoid receptor antagonist RU-43044 (30 mg/kg). The α2 adrenoceptor agonist clonidine (0.03 mg/kg) and the 5-HT4 receptor agonist BIMU8 (30 mg/kg) also reduced encounter-induced hyperactivity. The effect of desipramine was blocked by the α2 adrenoceptor antagonist idazoxan (0.3 mg/kg). The effect of fluvoxamine was blocked by the 5-HT4 receptor antagonist GR125487 (3 mg/kg), but not the 5-HT1A receptor antagonist WAY100635 (1 mg/kg), the 5-HT3 receptor antagonist azasetron (3 mg/kg), or the 5-HT6 receptor antagonist SB399885 (3 mg/kg). The effect of venlafaxine was blocked by the simultaneous administration of idazoxan (0.3 mg/kg) and GR125487 (3 mg/kg), but not by either compound alone. These findings suggest that encounter-induced hyperactivity in isolation-reared mice is a robust model for testing the pharmacological profile of antidepressants, although the range of antidepressants tested is limited and some non-antidepressants are also effective. The present study also shows a key role of α2 and 5-HT4 receptors in the antidepressant effect in this model.


Subject(s)
Motor Activity/drug effects , Social Behavior , Social Isolation/psychology , Animals , Animals, Outbred Strains , Benzimidazoles/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Desipramine/pharmacology , Duloxetine Hydrochloride/pharmacology , Fluvoxamine/pharmacology , Hydroxycorticosteroids/pharmacology , Idazoxan/pharmacology , Indoles/pharmacology , Male , Mice , Neuropsychological Tests , Oxazines/pharmacology , Paroxetine/pharmacology , Piperazines/pharmacology , Psychotropic Drugs/pharmacology , Pyridines/pharmacology , Risperidone/pharmacology , Ritanserin/pharmacology , Sulfonamides/pharmacology , Venlafaxine Hydrochloride/pharmacology
10.
J Pharmacol Sci ; 127(2): 232-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25727963

ABSTRACT

We have found that fluvoxamine-induced increases in prefrontal dopamine release are enhanced by adrenalectomy/castration and 5-HT1A receptors are involved in the enhancement. This study examined which 5-HT1A autoreceptors or postsynaptic receptor play a key role in the enhancement in mice. Adrenalectomy/castration-induced enhancement of fluvoxamine-induced increase in the dopamine release was not blocked by local perfusion with the 5-HT1A receptor antagonist WAY100635 (10 µM), while it was blocked by systemic administration of WAY100635 at low dose (0.1 mg/kg) which blocked preferentially autoreceptor-mediated responses. These finding suggests that 5-HT1A autoreceptors play a key role in the enhancement of prefrontal dopamine release.


Subject(s)
Adrenalectomy , Castration , Dopamine/metabolism , Fluvoxamine/pharmacology , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Male , Mice, Inbred Strains , Piperazines/administration & dosage , Piperazines/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Serotonin 5-HT1 Receptor Antagonists/administration & dosage , Serotonin 5-HT1 Receptor Antagonists/pharmacology
11.
Int J Neuropsychopharmacol ; 17(12): 1957-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24818616

ABSTRACT

Galantamine, an inhibitor of acetylcholinesterase, promotes hippocampal neurogenesis, but the exact mechanism for this is not known. In the present study, we examined the mechanisms underlying the effects of acute galantamine on neurogenesis in the mouse hippocampus. Galantamine (3 mg/kg) increased the number of 5-bromo-2'-deoxyuridine (BrdU)-positive cells in the subgranular zone of the dentate gyrus. This effect was blocked by the muscarinic receptor antagonist scopolamine and the preferential M1 muscarinic receptor antagonist telenzepine, but not by the nicotinic receptor antagonists mecamylamine and methyllycaconitine. Galantamine did not alter the ratio of neuronal nuclei (NeuN)- or glial fibrillary acidic protein (GFAP)-positive cells to BrdU-labeled cells in the subgranular zone and granule cell layer. Galantamine (1, 3 mg/kg) promoted the survival of 2-wk-old newly divided cells in mice in the granule cell layer of the dentate gyrus, whereas it did not affect the survival of newly divided cells at 1 and 4 wk. Galantamine-induced increases in cell survival were blocked by the α7 nicotinic receptor antagonist methyllycaconitine, but not by scopolamine. Bilateral injection of recombinant IGF2 into the dentate gyrus of the hippocampus mimicked the effects of galantamine. The effects of galantamine were blocked by direct injection of the IGF1 receptor antagonist JB1. These findings suggest that galantamine promotes neurogenesis via activation of the M1 muscarinic and α7 nicotinic acetylcholine receptors. The present study also suggests that IGF2 is involved in the effects of galantamine on the survival of 2-wk-old immature cells in the granule cell layer.


Subject(s)
Galantamine/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Receptor, Muscarinic M1/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cell Survival/drug effects , DNA-Binding Proteins , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/physiology , Insulin-Like Growth Factor II/metabolism , Male , Mice , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Nuclear Proteins/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptor, Muscarinic M1/antagonists & inhibitors , Recombinant Proteins/metabolism , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
12.
Int J Neuropsychopharmacol ; 17(6): 883-93, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24405605

ABSTRACT

We recently showed that social encounter stimulation induces hyperactivity in mice reared in social isolation from early life and this is associated with the transient activation of prefrontal dopaminergic and serotonergic systems. In the present study, we examined the effect of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor antagonist 2, 3-dioxo-6-nitro-1, 2, 3, 4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX) on encounter-induced behavioural and neurochemical changes to study the role of the receptor in abnormal behaviours in isolation-reared mice. The encounter to an intruder mouse induced hyperactivity with transient increases in prefrontal dopamine and serotonin levels in isolation-reared mice. NBQX attenuated the encounter-induced hyperactivity and the associated neurochemical changes in isolation-reared mice. In addition, NBQX reduced aggressive behaviour and cognitive impairment in isolation-reared mice, but did not affect depressive-like behaviour or spontaneous hyper-locomotion in these animals. The AMPA receptor agonist (S)-AMPA increased prefrontal dopamine and serotonin release, and this effect was higher in isolation-reared mice than in the group-reared mice, suggesting higher prefrontal AMPA receptor activity in isolation-reared mice. Furthermore, isolation rearing increased the expression of AMPA receptor subunits (GluR1, GluR2 and GluR3) and GluR1 Ser845 phosphorylation in the prefrontal cortex, but not in the hippocampus or nucleus accumbens. Taken together, these results suggest that an increase in AMPA receptor activity in the prefrontal cortex contributes to some, but not all, abnormal behaviours in isolation-reared mice.


Subject(s)
Hyperkinesis/physiopathology , Prefrontal Cortex/physiopathology , Receptors, AMPA/metabolism , Social Behavior , Social Isolation , Aggression/drug effects , Aggression/physiology , Aggression/psychology , Animals , Animals, Outbred Strains , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Depression/drug therapy , Depression/etiology , Depression/physiopathology , Dopamine/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Hippocampus/physiopathology , Hyperkinesis/psychology , Male , Mice , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Prefrontal Cortex/drug effects , Quinoxalines/pharmacology , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Serotonin/metabolism , Social Isolation/psychology
13.
Neurochem Res ; 39(5): 825-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24634253

ABSTRACT

Spontaneously hypertensive rats (SHRs) are used as a model for attention-deficit/hyperactivity disorder (ADHD), since SHRs are hyperactive and show defective sustained attention in behavioral tasks. The psychostimulants amphetamine and methylphenidate and the selective norepinephrine reuptake inhibitor atomoxetine are used as ADHD medications. The effects of high K(+) stimulation or psychostimulants on brain norepinephrine or dopamine release in SHRs have been previously studied both in vitro and in vivo, but the effects of atomoxetine on these neurotransmitters have not. The present study examined the effects of administration of atomoxetine on extracellular norepinephrine, dopamine, and serotonin levels in the prefrontal cortex of juvenile SHRs and Wistar-Kyoto (WKY) rats. Baseline levels of prefrontal norepinephrine, dopamine, and serotonin were similar in SHRs and WKY rats. Systemic administration of atomoxetine (3 mg/kg) induced similar increases in prefrontal norepinephrine and dopamine, but not serotonin, levels in both strains. Furthermore, there was no difference in high K(+)-induced increases in extracellular norepinephrine, dopamine, and serotonin levels in the prefrontal cortex between SHRs and WKY rats. These findings indicate that monoamine systems in the prefrontal cortex are similar between SHRs and WKY rats.


Subject(s)
Dopamine/physiology , Norepinephrine/physiology , Prefrontal Cortex/metabolism , Propylamines/pharmacology , Serotonin/physiology , Animals , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/physiopathology , Disease Models, Animal , Male , Potassium/administration & dosage , Prefrontal Cortex/drug effects , Rats, Inbred SHR , Rats, Inbred WKY
14.
J Pharmacol Sci ; 125(3): 237-41, 2014.
Article in English | MEDLINE | ID: mdl-24942324

ABSTRACT

Post-weaning social isolation in mice induces behavioral abnormalities such as hyperactivity, aggression, depression- and anxiety-like behaviors, deficits of prepulse inhibition, and reduced pain sensitivity to the noxious stimuli. Then, this mouse is considered to be a model of psychiatric disorders including schizophrenia. We have found that serotonin (5-HT)(1A)-receptor ligands attenuate these abnormalities, suggesting the pharmacological role of the receptor in treatment of psychiatric disorders. Furthermore, we have recently found that isolation-reared mice show social encounter-induced hyperactivity, a novel phenotype of the abnormal behaviors, and the hyperactivity is triggered by activation of the serotonergic system from the dorsal raphe to the frontal cortex. This review summarizes the effects of 5-HT(1A) receptor ligands on aggressive behavior, deficits of prepulse inhibition, reduced pain sensitivity to the noxious stimuli, and encounter-induced hyperactivity in social isolation-reared mice. These findings suggest that the 5-HT(1A) receptor is a potential target molecule for treatment of psychiatric disorders and pain.


Subject(s)
Behavior, Animal/physiology , Mental Disorders/etiology , Receptor, Serotonin, 5-HT1A/physiology , Social Isolation/psychology , Weaning , Animals , Mental Disorders/genetics , Mental Disorders/therapy , Mice , Molecular Targeted Therapy
15.
Int J Neuropsychopharmacol ; 16(1): 91-103, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22093185

ABSTRACT

Maternal use of valproic acid (VPA) during pregnancy has been implicated in the aetiology of autism spectrum disorders in children, and rodents prenatally exposed to VPA showed behavioural alterations similar to those observed in humans with autism. However, the exact mechanism for VPA-induced behavioural alterations is not known. To study this point, we examined the effects of prenatal exposure to VPA and valpromide, a VPA analog lacking histone deacetylase inhibition activity, on behaviours, cortical pathology and histone acetylation levels in mice. Mice exposed to VPA at embryonic day 12.5 (E12.5), but not at E9 and E14.5, displayed social interaction deficits, anxiety-like behaviour and memory deficits at age 4-8 wk. In contrast to male mice, the social interaction deficits (a decrease in sniffing behaviour) were not observed in female mice at age 8 wk. The exposure to VPA at E12.5 decreased the number of Nissl-positive cells in the middle and lower layers of the prefrontal cortex and in the lower layers of the somatosensory cortex at age 8 wk. Furthermore, VPA exposure caused a transient increase in acetylated histone levels in the embryonic brain, followed by an increase in apoptotic cell death in the neocortex and a decrease in cell proliferation in the ganglionic eminence. In contrast, prenatal exposure to valpromide at E12.5 did not affect the behavioural, biochemical and histological parameters. Furthermore, these findings suggest that VPA-induced histone hyperacetylation plays a key role in cortical pathology and abnormal autism-like behaviours in mice.


Subject(s)
Autistic Disorder/chemically induced , Autistic Disorder/metabolism , Histones/metabolism , Interpersonal Relations , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Valproic Acid/adverse effects , Acetylation/drug effects , Animals , Autistic Disorder/psychology , Cell Movement/drug effects , Cell Movement/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred ICR , Pregnancy , Prenatal Exposure Delayed Effects/psychology
16.
Behav Pharmacol ; 24(4): 328-31, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23751518

ABSTRACT

Recent clinical studies have shown that serotonin-norepinephrine reuptake inhibitors such as venlafaxine and duloxetine are effective against symptoms of attention-deficit/hyperactivity disorder such as inattention, oppositionality, and hyperactivity. We have recently found that these serotonin-norepinephrine reuptake inhibitors, like methylphenidate, reduced the hyperactivity in spontaneously hypertensive rats (SHR), an animal model of attention-deficit/hyperactivity disorder. The present study investigated whether the α2-adrenoceptor and the dopamine-D1 receptor are involved in the behavioral effects of methylphenidate and venlafaxine in SHR. Adolescent male SHR showed greater horizontal locomotion in a familiar open field than male Wistar Kyoto and Wistar rats, and methylphenidate (0.3 mg/kg) and venlafaxine (30 mg/kg) reduced horizontal locomotion in SHR, but not Wistar Kyoto or Wistar rats. The effects of methylphenidate and venlafaxine were blocked by idazoxan (an α2-adrenoceptor antagonist), but not by SCH23390 (a dopamine-D1 receptor antagonist). These findings suggest that the α2-adrenoceptor plays a key role in the effects of methylphenidate and venlafaxine on enhanced locomotion in SHR.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Central Nervous System Stimulants/therapeutic use , Cyclohexanols/therapeutic use , Methylphenidate/therapeutic use , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic Agents/pharmacology , Analysis of Variance , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Exploratory Behavior/drug effects , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Time Factors , Venlafaxine Hydrochloride
17.
Behav Pharmacol ; 24(1): 74-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23268987

ABSTRACT

Previous studies suggest that metabotropic glutamate 2/3 receptors are involved in psychiatric disorders. In this study, we examined the effects of the selective metabotropic glutamate 2/3 (mGlu2/3) receptor agonist MGS0028 on behavioral abnormalities in mice lacking the pituitary adenylate cyclase-activating polypeptide (PACAP), an experimental model of psychiatric disorders such as schizophrenia and attention-deficit/hyperactivity disorder. We found that PACAP-deficient mice showed impairments in the novel object recognition test and these impairments were improved by MGS0028 (0.1 mg/kg). Similarly, MGS0028 improved hyperactivity and jumping behaviors, but did not reverse increased immobility times in the forced swim test in PACAP-deficient mice. These results suggest that MGS0028 may be a potential, novel treatment for psychiatric disorders.


Subject(s)
Antipsychotic Agents/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Bridged Bicyclo Compounds/therapeutic use , Dicarboxylic Acids/therapeutic use , Memory Disorders/drug therapy , Nootropic Agents/therapeutic use , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Metabotropic Glutamate/agonists , Animals , Attention Deficit Disorder with Hyperactivity/metabolism , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Male , Memory Disorders/metabolism , Mice , Mice, Mutant Strains , Molecular Targeted Therapy , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Metabotropic Glutamate/metabolism , Recognition, Psychology/drug effects
18.
J Pharmacol Sci ; 121(4): 351-4, 2013.
Article in English | MEDLINE | ID: mdl-23538676

ABSTRACT

Nitric oxide (NO) induces cytotoxicity in neuronal and glial cells via activation of the Na(+)/Ca(2+) exchanger (NCX). This study examined the role of the predominant brain-specific NCX splice variant NCX1.5 in NO-induced cytotoxicity in the HEK293 cell expression system. Cells were transfected with the plasmid construct pcDNA3.1/V5-His containing full-length rat NCX1.5 cDNA. There was no difference in the cytotoxic effects of the NO donors sodium nitroprusside and S-nitroso-N-acetylpenicillamine between control and transfected cells. These results suggest that NO cytotoxicity is not dependent on NCX1.5.


Subject(s)
Brain/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression/genetics , Gene Expression/physiology , Genetic Variation/genetics , Nitric Oxide/toxicity , Protein Splicing/genetics , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Male , Nitric Oxide Donors/toxicity , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/physiology
19.
J Neurosci ; 31(36): 12963-71, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21900575

ABSTRACT

Recurrent seizure activity has been shown to induce a variety of permanent structural changes in the brain. Matrix metalloproteinases (MMPs) function to promote neuronal plasticity, primarily through cleavage of extracellular matrix proteins. Here, we investigated the role of MMP-9 in the development of pentylenetetrazole (PTZ)-induced kindled seizure in mice. Repeated treatment with PTZ (40 mg/kg) produced kindled seizure, which was accompanied by enhanced MMP-9 activity and expression in the hippocampus. No change in MMP-9 activity was observed in the hippocampi of mice with generalized tonic seizure following single administration of PTZ (60 mg/kg). MMP-9 colocalized with the neuronal marker NeuN and the glial marker GFAP in the dentate gyrus of the kindled mouse hippocampus. Coadministration of diazepam or MK-801 with PTZ inhibited the development of kindling and the increased MMP-9 levels in the hippocampus. Marked suppression of kindled seizure progression in response to repeated PTZ treatment was observed in MMP-9((-/-)) mice compared with wild-type mice, an observation that was accompanied by decreased hippocampal levels of mature brain-derived neurotrophic factor. Microinjecting the BDNF scavenger TrkB-Fc into the right ventricle before each PTZ treatment significantly suppressed the development of kindling in wild-type mice, whereas no effect was observed in MMP-9((-/-)) mice. On the other hand, bilateral injections of pro-BDNF into the hippocampal dentate gyrus significantly enhanced kindling in wild-type mice but not MMP-9((-/-)) mice. These findings suggest that MMP-9 is involved in the progression of behavioral phenotypes in kindled mice because of conversion of pro-BDNF to mature BDNF in the hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Convulsants , Hippocampus/metabolism , Kindling, Neurologic/physiology , Matrix Metalloproteinase 9/physiology , Pentylenetetrazole , Protein Precursors/metabolism , Seizures/enzymology , Animals , Anticonvulsants/pharmacology , Blotting, Western , Brain-Derived Neurotrophic Factor/administration & dosage , Diazepam/pharmacology , Dizocilpine Maleate/pharmacology , Electrophoresis, Polyacrylamide Gel , Fear/psychology , Hippocampus/enzymology , Male , Matrix Metalloproteinase 9/metabolism , Memory/physiology , Mice , Mice, Inbred ICR , Mice, Knockout , Microinjections , Nerve Tissue Proteins/pharmacology , Neuroprotective Agents/pharmacology , Protein Kinases/pharmacology , Protein Precursors/administration & dosage , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Seizures/chemically induced
20.
J Pharmacol Sci ; 118(4): 543-6, 2012.
Article in English | MEDLINE | ID: mdl-22447305

ABSTRACT

We have recently shown that prenatal valproic acid (VPA) exposure causes autism spectrum disorders-like behavioral abnormalities and Nissl-positive cell loss in both prefrontal and somatosensory cortices in male mice. We have also found that VPA-induced social interaction deficits are observed in male but not female offspring. This study demonstrated that the exposure to VPA at embryonic day 12.5 significantly decreased Nissl-positive cell numbers in the prefrontal cortex, but not in the somatosensory cortex, in female offspring. These findings suggest that VPA-induced morphological abnormalities in the somatosensory cortex may be involved in the sex-dependent social interaction deficits.


Subject(s)
Prefrontal Cortex/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Sex Characteristics , Somatosensory Cortex/drug effects , Valproic Acid/toxicity , Animals , Female , Male , Mice , Mice, Inbred ICR , Prefrontal Cortex/embryology , Prefrontal Cortex/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Somatosensory Cortex/embryology , Somatosensory Cortex/pathology
SELECTION OF CITATIONS
SEARCH DETAIL