Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 413: 113447, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34224763

ABSTRACT

Several studies have shown that oxytocin (OXT) modulates social behavior. Similarly, monoamines such as dopamine (DA) play a role in regulating social behavior. Previous studies have demonstrated that the soluble N-ethylmaleimide-sensitive fusion attachment protein receptor (SNARE) protein syntaxin 1A (STX1A) regulates the secretion of OXT and monoamines, and that STX1A gene knockout (STX1A KO) mice exhibit atypical social behavior, such as deficient social recognition, due to reduced OXT release. In this study, we analyzed the neural mechanism regulating social behavior by OXT and/or DA using STX1A KO mice as a model animal. We found that OXT directly induced DA release from cultured DA neurons through OXT and V1a receptors. In STX1A KO mice, the atypical social behavior was partially improved by OXT administration, which was inhibited by D1 receptor blockade. In addition, the atypical social behavior in STX1A KO mice was partially improved by facilitation of DAergic signaling with the DA reuptake inhibitor GBR12909. Moreover, the amelioration by GBR12909 was inhibited by OXTR blockade. These results suggest that the reciprocal interaction between the DAergic and OXTergic neuronal systems in the CNS may be important in regulating social behavior.


Subject(s)
Behavioral Symptoms/metabolism , Central Nervous System/metabolism , Chemotactic Factors/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Social Behavior , Syntaxin 1/metabolism , Animals , Behavioral Symptoms/drug therapy , Cells, Cultured , Central Nervous System/drug effects , Disease Models, Animal , Dopamine Antagonists/pharmacology , Mice , Mice, Knockout , Oxytocin/pharmacology , Piperazines/pharmacology , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Oxytocin/antagonists & inhibitors , Syntaxin 1/deficiency
2.
J Neurochem ; 156(5): 604-613, 2021 03.
Article in English | MEDLINE | ID: mdl-32858780

ABSTRACT

De novo heterozygous mutations in the STX1B gene, encoding syntaxin 1B, cause a familial, fever-associated epilepsy syndrome. Syntaxin 1B is an essential component of the pre-synaptic neurotransmitter release machinery as a soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein that regulates the exocytosis of synaptic vesicles. It is also involved in regulating the functions of the SLC6 family of neurotransmitter transporters that reuptake neurotransmitters, including inhibitory neurotransmitters, such as γ-aminobutyric acid (GABA) and glycine. The purpose of the present study was to elucidate the molecular mechanisms underlying the development of febrile seizures by examining the effects of syntaxin 1B haploinsufficiency on inhibitory synaptic transmission during hyperthermia in a mouse model. Stx1b gene heterozygous knockout (Stx1b+/- ) mice showed increased susceptibility to febrile seizures and drug-induced seizures. In cultured hippocampal neurons, we examined the temperature-dependent properties of neurotransmitter release and reuptake by GABA transporter-1 (GAT-1) at GABAergic neurons using whole-cell patch-clamp recordings. The rate of spontaneous quantal GABA release was reduced in Stx1b+/- mice. The hyperthermic temperature increased the tonic GABAA current in wild-type (WT) synapses, but not in Stx1b+/- synapses. In WT neurons, recurrent bursting activities were reduced in a GABA-dependent manner at hyperthermic temperature; however, this was abolished in Stx1b+/- neurons. The blockade of GAT-1 increased the tonic GABAA current and suppressed recurrent bursting activities in Stx1b+/- neurons at the hyperthermic temperature. These data suggest that functional abnormalities associated with GABA release and reuptake in the pre-synaptic terminals of GABAergic neurons may increase the excitability of the neural circuit with hyperthermia.


Subject(s)
Body Temperature/physiology , Extracellular Fluid/metabolism , Seizures/metabolism , Synapses/metabolism , Syntaxin 1/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Animals, Newborn , Cells, Cultured , Hippocampus/metabolism , Hyperthermia/genetics , Hyperthermia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pentylenetetrazole/toxicity , Seizures/chemically induced , Seizures/genetics , Synapses/genetics , Syntaxin 1/genetics
3.
Front Neural Circuits ; 15: 787692, 2021.
Article in English | MEDLINE | ID: mdl-34987361

ABSTRACT

Activation-induced manganese-enhanced MRI (AIM-MRI) is an attractive tool for non-invasively mapping whole brain activities. Manganese ions (Mn2+) enter and accumulate in active neurons via calcium channels. Mn2+ shortens the longitudinal relaxation time (T1) of H+, and the longitudinal relaxation rate R1 (1/T1) is proportional to Mn2+ concentration. Thus, AIM-MRI can map neural activities throughout the brain by assessing the R1 map. However, AIM-MRI is still not widely used, partially due to insufficient information regarding Mn2+ dynamics in the brain. To resolve this issue, we conducted a longitudinal study looking at manganese dynamics after systemic administration of MnCl2 by AIM-MRI with quantitative analysis. In the ventricle, Mn2+ increased rapidly within 1 h, remained high for 3 h, and returned to near control levels by 24 h after administration. Microdialysis showed that extracellular Mn returned to control levels by 4 h after administration, indicating a high concentration of extracellular Mn2+ lasts at least about 3 h after administration. In the brain parenchyma, Mn2+ increased slowly, peaked 24-48 h after administration, and returned to control level by 5 days after a single administration and by 2 weeks after a double administration with a 24-h interval. These time courses suggest that AIM-MRI records neural activity 1-3 h after MnCl2 administration, an appropriate timing of the MRI scan is in the range of 24-48 h following systemic administration, and at least an interval of 5 days or a couple of weeks for single or double administrations, respectively, is needed for a repeat AIM-MRI experiment.


Subject(s)
Magnetic Resonance Imaging , Manganese , Animals , Brain/diagnostic imaging , Chlorides , Ions , Longitudinal Studies , Mice
4.
Eur J Neurosci ; 46(12): 2867-2874, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29139159

ABSTRACT

In neuronal plasma membrane, two syntaxin isoforms, HPC-1/syntaxin 1A (STX1A) and syntaxin 1B (STX1B), are predominantly expressed as soluble N-ethylmaleimide-sensitive fusion attachment protein receptors, also known as t-SNAREs. We previously reported that glutamatergic and GABAergic synaptic transmissions are impaired in Stx1b null mutant (Stx1b-/- ) mice but are almost normal in Stx1a null mutant (Stx1a-/- ) mice. These observations suggested that STX1A and STX1B have distinct functions in fast synaptic transmission in the central nervous system (CNS). Interestingly, recent studies indicated that Stx1a-/- or Stx1a+/- mice exhibit disruption in the monoaminergic system in the CNS, causing unusual behaviour that is similar to neuropsychological alterations observed in psychiatric patients. Here, we studied whether STX1B contributes to the regulation of monoaminergic system and if STX1B is related to neuropsychological properties in human neuropsychological disorders similar to STX1A. We found that monoamine release in vitro was normal in Stx1b+/- mice unlike Stx1a-/- or Stx1a+/- mice, but the basal extracellular dopamine (DA) concentration in the ventral striatum was increased. DA secretion in the ventral striatum is regulated by GABAergic neurons, and Stx1b+/- mice exhibited reduced GABA release both in vitro and in vivo, disrupting the DAergic system in the CNS of these mice. We also found that Stx1b+/- mice exhibited reduced pre-pulse inhibition (PPI), which is believed to represent one of the prominent schizotypal behavioural profiles of human psychiatric patients. The reduction in PPI was rescued by DA receptor antagonists. These observations indicated that STX1B contributes to excess activity of the DAergic system through regulation of GABAergic transmission.


Subject(s)
GABAergic Neurons/metabolism , Synaptic Potentials , Syntaxin 1/genetics , Animals , Cells, Cultured , Dopamine/metabolism , Dopamine Antagonists/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Male , Mice , Mice, Inbred C57BL , Neural Inhibition , Syntaxin 1/metabolism , Ventral Striatum/cytology , Ventral Striatum/metabolism , Ventral Striatum/physiology , gamma-Aminobutyric Acid/metabolism
5.
Neurosci Lett ; 644: 5-9, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28235601

ABSTRACT

Autism spectrum disorder (ASD) is highly heritable and encompasses a various set of neuropsychiatric disorders with a wide-ranging presentation. HPC-1/syntaxin1A (STX1A) encodes a neuronal plasma membrane protein that regulates the secretion of neurotransmitters and neuromodulators. STX1A gene ablated mice (null and heterozygote mutant) exhibit abnormal behavioral profiles similar to human autistic symptoms, accompanied by reduction of monoamine secretion. To determine whether copy number variation of STX1A gene and the change of its expression correlate with ASD as in STX1A gene ablated mice, we performed copy number assay and real-time quantitative RT-PCR using blood or saliva samples from ASD patients. We found that some ASD patients were haploid for the STX1A gene similar to STX1A heterozygote mutant mice. However, copy number of STX1A gene was normal in the parents and siblings of ASD patients with STX1A gene haploidy. In ASD patients with gene haploidy, STX1A mRNA expression was reduced to about half of their parents. Thus, a part of ASD patients had haploidy of STX1A gene and lower STX1A gene expression.


Subject(s)
Autism Spectrum Disorder/genetics , Syntaxin 1/genetics , Adolescent , Animals , Child , Female , Gene Dosage , Haploidy , Humans , Male , Mice , Mice, Mutant Strains , Pedigree , Young Adult
6.
J Neurochem ; 138(1): 117-23, 2016 07.
Article in English | MEDLINE | ID: mdl-27059771

ABSTRACT

HPC-1/syntaxin1A (STX1A), a neuronal soluble N-ethylmaleimide-sensitive fusion attachment protein receptor, contributes to neural function in the CNS by regulating transmitter release. Recent studies reported that STX1A is associated with human neuropsychological disorders, such as autism spectrum disorder and attention deficit hyperactivity disorder. Previously, we showed that STX1A null mutant mice (STX1A KO) exhibit neuropsychological abnormalities, such as fear memory deficits, attenuation of latent inhibition, and unusual social behavior. These observations suggested that STX1A may be involved in the neuropsychological basis of these abnormalities. Here, to study the neural basis of social behavior, we analyzed the profile of unusual social behavior in STX1A KO with a social novelty preference test, which is a useful method for quantification of social behavior. Interestingly, the unusual social behavior in STX1A KO was partially rescued by intracerebroventricular administration of oxytocin (OXT). In vivo microdialysis studies revealed that the extracellular OXT concentration in the CNS of STX1A KO was significantly lower compared with wild-type mice. Furthermore, dopamine-induced OXT release was reduced in STX1A KO. These results suggested that STX1A plays an important role in social behavior through regulation of the OXTergic neural system. Dopamine (DA) release is reduced in CNS of syntaxin1A null mutant mice (STX1A KO). Unusual social behavior was observed in STX1A KO. We found that oxytocin (OXT) release, which was stimulated by DA, was reduced and was rescued the unusual social behavior in STX1A KO was rescued by OXT. These results indicated that STX1A plays an important role in promoting social behavior through regulation of DA-induced OXT release in amygdala.


Subject(s)
Amygdala/metabolism , Oxytocin/metabolism , Social Behavior Disorders/genetics , Social Behavior Disorders/pathology , Syntaxin 1/deficiency , Amygdala/drug effects , Analysis of Variance , Animals , Benzoxazines/pharmacology , Disease Models, Animal , Dopamine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Exploratory Behavior/physiology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Ovariectomy , Oxytocin/pharmacology , Piperazines/pharmacology , Piperidines/pharmacology , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Syntaxin 1/genetics
7.
PLoS One ; 9(2): e90004, 2014.
Article in English | MEDLINE | ID: mdl-24587181

ABSTRACT

Two syntaxin 1 (STX1) isoforms, HPC-1/STX1A and STX1B, are coexpressed in neurons and function as neuronal target membrane (t)-SNAREs. However, little is known about their functional differences in synaptic transmission. STX1A null mutant mice develop normally and do not show abnormalities in fast synaptic transmission, but monoaminergic transmissions are impaired. In the present study, we found that STX1B null mutant mice died within 2 weeks of birth. To examine functional differences between STX1A and 1B, we analyzed the presynaptic properties of glutamatergic and GABAergic synapses in STX1B null mutant and STX1A/1B double null mutant mice. We found that the frequency of spontaneous quantal release was lower and the paired-pulse ratio of evoked postsynaptic currents was significantly greater in glutamatergic and GABAergic synapses of STX1B null neurons. Deletion of STX1B also accelerated synaptic vesicle turnover in glutamatergic synapses and decreased the size of the readily releasable pool in glutamatergic and GABAergic synapses. Moreover, STX1A/1B double null neurons showed reduced and asynchronous evoked synaptic vesicle release in glutamatergic and GABAergic synapses. Our results suggest that although STX1A and 1B share a basic function as neuronal t-SNAREs, STX1B but not STX1A is necessary for the regulation of spontaneous and evoked synaptic vesicle exocytosis in fast transmission.


Subject(s)
Exocytosis/physiology , Neurons/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Syntaxin 1/metabolism , Animals , Evoked Potentials/physiology , Gene Expression Regulation , Glutamic Acid/metabolism , Male , Mice , Mice, Knockout , Neurons/cytology , Synaptic Vesicles/metabolism , Syntaxin 1/genetics , gamma-Aminobutyric Acid/metabolism
8.
Nat Prod Commun ; 9(1): 101-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24660475

ABSTRACT

Increases in the number of patients with dementia involving Alzheimer's disease (AD) are seen as a grave public health problem. In neurodegenerative disorders involving AD, biological stresses, such as oxidative and inflammatory stress, induce neural cell damage. Asparagus (Asparagus officinalis) is a popular vegetable, and an extract prepared from this reportedly possesses various beneficial biological activities. In the present study, we investigated the effects of enzyme-treated asparagus extract (ETAS) on neuronal cells and early cognitive impairment of senescence-accelerated mouse prone 8 (SAMP8) mice. The expression of mRNAs for factors that exert cytoprotective and anti-apoptotic functions, such as heat-shock protein 70 and heme oxygenase-1, was upregulated in NG108-15 neuronal cells by treatment with ETAS. Moreover, when release of lactate dehydrogenase from damaged NG108-15 cells was increased for cells cultured in medium containing either the nitric oxide donor sodium nitroprusside or the hypoxia mimic reagent cobalt chloride, ETAS significantly attenuated this cell damage. Also, when contextual fear memory, which is considered to be a hippocampus-dependent memory, was significantly impaired in SAMP8 mice, ETAS attenuated the cognitive impairment. These results suggest that ETAS produces cytoprotective effects in neuronal cells and attenuates the effects on the cognitive impairment of SAMP8 mice.


Subject(s)
Asparagus Plant , Cognitive Dysfunction/drug therapy , Neuroprotective Agents/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Animals , Cell Line, Tumor , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/metabolism , Male , Mice , Rats
9.
J Neurochem ; 130(4): 514-25, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24666284

ABSTRACT

Two types of syntaxin 1 isoforms, HPC-1/syntaxin 1A (STX1A) and syntaxin 1B (STX1B), are thought to have similar functions in exocytosis of synaptic vesicles. STX1A(-/-) mice which we generated previously develop normally, possibly because of compensation by STX1B. We produced STX1B(-/-) mice using targeted gene disruption and investigated their phenotypes. STX1B(-/-) mice were born alive, but died before postnatal day 14, unlike STX1A(-/-) mice. Morphologically, brain development in STX1B(-/-) mice was impaired. In hippocampal neuronal culture, the cell viability of STX1B(-/-) neurons was lower than that of WT or STX1A(-/-) neurons after 9 days. Interestingly, STX1B(-/-) neurons survived on WT or STX1A(-/-) glial feeder layers as well as WT neurons. However, STX1B(-/-) glial feeder layers were less effective at promoting survival of STX1B(-/-) neurons. Conditioned medium from WT or STX1A(-/-) glial cells had a similar effect on survival, but that from STX1B(-/-) did not promote survival. Furthermore, brain-derived neurotrophic factor (BDNF) or neurotrophin-3 supported survival of STX1B(-/-) neurons. BDNF localization in STX1B(-/-) glial cells was disrupted, and BDNF secretion from STX1B(-/-) glial cells was impaired. These results suggest that STX1A and STX1B may play distinct roles in supporting neuronal survival by glia. Syntaxin 1A (STX1A) and syntaxin 1B (STX1B) are thought to have similar functions as SNARE proteins. However, we found that STX1A and STX1B play distinct roles in neuronal survival using STX1A(-/-) mice and STX1B(-/-) mice. STX1B was important for neuronal survival, possibly by regulating the secretion of neurotrophic factors, such as BDNF, from glial cells.


Subject(s)
Neurons/physiology , Syntaxin 1/physiology , Animals , Blotting, Western , Brain/growth & development , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/pharmacology , Cell Survival/genetics , Cell Survival/physiology , Immunoenzyme Techniques , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Munc18 Proteins/metabolism , Neuroglia/physiology , Neurotrophin 3/biosynthesis , Neurotrophin 3/pharmacology , Real-Time Polymerase Chain Reaction , Syntaxin 1/genetics , Transfection
10.
Neurosci Lett ; 558: 186-91, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24269977

ABSTRACT

Pregabalin is widely used as an analgesic for the treatment of neuropathic pain. In the present experiments using mouse spinal slices, we recorded electrically evoked glutamatergic excitatory postsynaptic currents (eEPSCs) from superficial dorsal horn neurons. Pregabalin reduced the amplitude of eEPSCs, and increased the paired pulse ratio. Pregabalin also inhibited the frequency of spontaneously occurring miniature EPSCs without affecting their amplitude. Partial ligation of the sciatic nerve increased the expression of the calcium channel α2δ-1 subunit, and increased the presynaptic inhibitory action of pregabalin. Intrathecal injection of antisense oligodeoxynucleotide against the α2δ-1 subunit, decreased the expression of α2δ-1 mRNA in the spinal dorsal horn, and decreased pregabalin's action. These results provide further evidence that pregabalin exerts its presynaptic inhibitory action via binding with the α2δ subunit in a state-dependent manner. Furthermore, presynaptic actions of pregabalin were attenuated in knockout mice lacking the protein syntaxin 1A, a component of the synaptic vesicle release machinery, indicating that syntaxin 1A is required for pregabalin to exert its full presynaptic inhibitory action. These observations might suggest that direct and/or indirect interactions with the presynaptic proteins composing the release machinery underlie at least some part of pregabalin's presynaptic actions.


Subject(s)
Analgesics/pharmacology , Posterior Horn Cells/drug effects , Syntaxin 1/genetics , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Oligonucleotides, Antisense/pharmacology , Posterior Horn Cells/physiology , Pregabalin , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Sciatic Nerve/injuries , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/pharmacology
11.
Proc Natl Acad Sci U S A ; 110(48): 19420-5, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218571

ABSTRACT

In preparation for the metabolic demands of pregnancy, ß cells in the maternal pancreatic islets increase both in number and in glucose-stimulated insulin secretion (GSIS) per cell. Mechanisms have been proposed for the increased ß cell mass, but not for the increased GSIS. Because serotonin production increases dramatically during pregnancy, we tested whether flux through the ionotropic 5-HT3 receptor (Htr3) affects GSIS during pregnancy. Pregnant Htr3a(-/-) mice exhibited impaired glucose tolerance despite normally increased ß cell mass, and their islets lacked the increase in GSIS seen in islets from pregnant wild-type mice. Electrophysiological studies showed that activation of Htr3 decreased the resting membrane potential in ß cells, which increased Ca(2+) uptake and insulin exocytosis in response to glucose. Thus, our data indicate that serotonin, acting in a paracrine/autocrine manner through Htr3, lowers the ß cell threshold for glucose and plays an essential role in the increased GSIS of pregnancy.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/pharmacology , Signal Transduction/physiology , Animals , Female , Glucose/metabolism , Immunoblotting , Immunohistochemistry , Insulin Secretion , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Pregnancy , Receptors, Serotonin, 5-HT3/genetics
12.
Br J Nutr ; 110(9): 1549-58, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-23537529

ABSTRACT

Recently, the ability of polyphenols to reduce the risk of dementia and Alzheimer's disease (AD) has attracted a great deal of interest. In the present study, we investigated the attenuating effects of oligomerised lychee fruit-derived polyphenol (OLFP, also called Oligonol) on early cognitive impairment. Male senescence-accelerated mouse prone 8 (SAMP8) mice (4 months old) were given OLFP (100 mg/kg per d) for 2 months, and then conditioned fear memory testing was conducted. Contextual fear memory, which is considered hippocampus-dependent memory, was significantly impaired in SAMP8 mice compared with non-senescence-accelerated mice. OLFP attenuated cognitive impairment in SAMP8 mice. Moreover, the results of real-time PCR analysis that followed DNA array analysis in the hippocampus revealed that, compared with SAMP8 mice, the mRNA expression of Wolfram syndrome 1 (Wfs1) was significantly higher in SAMP8 mice administered with OLFP. Wfs1 reportedly helps to protect against endoplasmic reticulum (ER) stress, which is thought to be one of the causes for AD. The expression of Wfs1 was significantly up-regulated in NG108-15 neuronal cells by the treatment with OLFP, and the up-regulation was inhibited by the treatment of the cells with a c-Jun N-terminal kinase-specific inhibitor rather than with an extracellular signal-regulated kinase inhibitor. Moreover, OLFP significantly attenuated the tunicamycin-induced expression of the ER stress marker BiP (immunoglobulin heavy chain-binding protein) in the cells. These results suggest that OLFP has an attenuating effect on early cognitive impairment in SAMP8 mice, and diminishes ER stress in neuronal cells.


Subject(s)
Catechin/analogs & derivatives , Cognition Disorders/drug therapy , Endoplasmic Reticulum Stress/drug effects , Litchi/chemistry , Memory Disorders/drug therapy , Phenols/therapeutic use , Phytotherapy , Polyphenols/therapeutic use , Aging , Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Animals , Catechin/pharmacology , Catechin/therapeutic use , Cognition Disorders/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Fruit/chemistry , Heat-Shock Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory/drug effects , Memory Disorders/metabolism , Mice , Mice, Inbred Strains , Neurons/drug effects , Neurons/metabolism , Phenols/pharmacology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Polyphenols/pharmacology , RNA, Messenger/metabolism
13.
J Neurosci ; 32(1): 381-9, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22219298

ABSTRACT

The membrane protein HPC-1/syntaxin 1A is believed to play a key role in synaptic vesicle exocytosis, and it was recently suggested to be required for synaptic plasticity. Despite evidence for the function of HPC-1/syntaxin 1A in synaptic plasticity, the underlying cellular mechanism is unclear. We found that although fast synaptic transmission and long-term depression were unaffected, HPC-1/syntaxin 1A knock-out (STX1A(-/-)) mice showed impaired long-term potentiation (LTP) in response to theta-burst stimulation in CA1 hippocampal slices. The impairment in LTP was rescued by the application of forskolin, an adenylyl cyclase activator, or more robust stimulation, suggesting that cAMP/protein kinase A signaling was suppressed in these mice. In addition, catecholamine release from the hippocampus was significantly reduced in STX1A(-/-) mice. Because HPC-1/syntaxin 1A regulates exocytosis of dense-core synaptic vesicles, which contain neuromodulatory transmitters such as noradrenaline, dopamine and 5-HT, we examined the effect of neuromodulatory transmitters on LTP induction. Noradrenaline and dopamine enhanced LTP induction in STX1A(-/-) mice, whereas catecholamine depletion reduced LTP induction in wild-type mice. Theses results suggest that HPC-1/syntaxin 1A regulates catecholaminergic systems via exocytosis of dense-core synaptic vesicles, and that deletion of HPC-1/syntaxin 1A causes impairment of LTP induction.


Subject(s)
CA1 Region, Hippocampal/metabolism , Catecholamines/deficiency , Long-Term Potentiation/genetics , Synapses/metabolism , Syntaxin 1/deficiency , Animals , CA1 Region, Hippocampal/drug effects , Catecholamines/pharmacology , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Synapses/drug effects , Syntaxin 1/antagonists & inhibitors , Syntaxin 1/genetics
14.
Biochem Biophys Res Commun ; 416(1-2): 125-9, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22093827

ABSTRACT

It is well known that exercise prevents and reduces cognitive impairment. In the present study, we focused on exercise training as a tool to prevent cognitive impairment, and searched for novel molecules that may relate to the prevention of cognitive impairment in the hippocampus. Two-month-old senescence-accelerated mouse prone-8 (SAMP8) mice were subjected to voluntary exercise training by running on a wheel for 4 months, and were then assigned a conditioned fear memory test. Moreover, various mRNA levels in the hippocampus were examined by DNA array analysis and real-time PCR. Contextual fear memory in SAMP8 control mice was significantly impaired compared with that in non-senescence mice. Exercise training definitely attenuated such cognitive impairment. The results of real-time PCR analysis that was conducted following DNA array analysis in the hippocampus revealed that, compared with SAMR8 control mice, the expression levels of leucine zipper transcription factor-like protein 1 (Lztfl1) mRNA were significantly higher in SAMP8 mice subjected to exercise training. In addition, the overexpression of Lztfl1 promoted neurite outgrowth in Neuro 2a cells. These results suggest that exercise has a preventive effect on cognitive impairment in SAMP8 mice, and that exercise-induced increase in Lztfl1 induces neurite outgrowth.


Subject(s)
Cognition Disorders/prevention & control , Neurites/physiology , Physical Conditioning, Animal , Transcription Factors/biosynthesis , Animals , Hippocampus/metabolism , Male , Memory , Mice , Mice, Mutant Strains , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism
15.
J Mol Histol ; 42(5): 483-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21822613

ABSTRACT

HPC-1/syntaxin 1A (STX1A) is abundantly expressed in neurons. STX1A is believed to regulate exocytosis in synaptic vesicles. In our recent studies, STX1A knockout (KO) mice showed normal development, and basal synaptic transmission in cultured hippocampal neurons appeared to be normal. However, behavioral abnormalities were observed in STX1A KO mice. In the normal rodent retina, the STX1A protein is expressed in two synaptic layers (plexiform layers). Here, to evaluate the effects of the loss of STX1A on retinal structure, we examined the retinal layer structure in STX1A KO mice using hematoxylin staining and immunostaining. We found that the general layer structures in the retina were preserved in all genotypes. However, the outer plexiform layer (OPL) was significantly thicker in KO and heterozygous mutant (HT) mice compared with that in wild-type (WT) mice. No significant differences were observed in the thicknesses of the other layers. Immunostaining for protein kinase C α showed that the alignment of rod bipolar cell bodies in the inner nuclear layer (INL) was slightly disrupted in HT and KO retinas. Furthermore, the dendrites of these cells in the OPL of KO mice were sparse, compared to those in WT mice. Our results show that STX1A KO mice have increased thickness of the OPL and changes in the morphology of the INL that may contribute to the change in OPL thickness. We suggest that STX1A may play a role in the structural formation of the INL and OPL in the retina.


Subject(s)
Retina/cytology , Retina/metabolism , Syntaxin 1/deficiency , Animals , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Protein Kinase C/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/enzymology , Retinal Neurons/cytology , Retinal Neurons/metabolism , Staining and Labeling , Syntaxin 1/metabolism
16.
J Biol Chem ; 286(37): 32244-50, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21768089

ABSTRACT

The Rab27 effector granuphilin/Slp4 is essential for the stable attachment (docking) of secretory granules to the plasma membrane, and it also inhibits subsequent fusion. Granuphilin is thought to mediate these processes through interactions with Rab27 on the granule membrane and with syntaxin-1a on the plasma membrane and its binding partner Munc18-1. Consistent with this hypothesis, both syntaxin-1a- and Munc18-1-deficient secretory cells, as well as granuphilin null cells, have been observed to have a deficit of docked granules. However, to date there has been no direct comparative analysis of the docking defects in those mutant cells. In this study, we morphometrically compared granule-docking states between granuphilin null and syntaxin-1a null pancreatic ß cells derived from mice having the same genetic background. We found that loss of syntaxin-1a does not cause a significant granule-docking defect, in contrast to granuphilin deficiency. Furthermore, we newly generated granuphilin/syntaxin-1a double knock-out mice, characterized their phenotypes, and found that the double mutant mice represent a phenocopy of granuphilin null mice and do not represent phenotypes of syntaxin-1a null mice, including their granule-docking behavior. Because granuphilin binds to syntaxin-2 and syntaxin-3 as well as syntaxin-1a, it likely mediates granule docking through interactions with those multiple syntaxins on the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Secretory Vesicles/metabolism , Syntaxin 1/metabolism , Vesicular Transport Proteins/metabolism , Animals , Biological Transport/physiology , Cell Line , Cell Membrane/genetics , Insulin/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Knockout , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Secretory Vesicles/genetics , Syntaxin 1/genetics , Vesicular Transport Proteins/genetics
17.
Eur J Neurosci ; 32(1): 99-107, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20576034

ABSTRACT

HPC-1/syntaxin 1A (STX1A) is thought to regulate the exocytosis of synaptic vesicles in neurons. In recent human genetic studies, STX1A has been implicated in neuropsychological disorders. To examine whether STX1A gene ablation is responsible for abnormal neuropsychological profiles observed in human psychiatric patients, we analysed the behavioral phenotype of STX1A knockout mice. Abnormal behavior was observed in both homozygotes (STX1A(-/-)) and heterozygotes (STX1A(+/-)) in a social interaction test, a novel object exploring test and a latent inhibition (LI) test, but not in a pre-pulse inhibition test. Interestingly, attenuation of LI, which is closely related to human schizotypic symptoms, was restored by administration of the selective serotonin reuptake inhibitor, fluoxetine, but not by the dopamine reuptake inhibitor, GBR12935, or the noradrenalin reuptake inhibitor, desipramine. We also observed that LI attenuation was restored by DOI (a 5-HT(2A) receptor agonist), but not by 8-OH-DPAT (a 5-HT(1A) receptor agonist), mCPP (a 5-HT(2C) receptor agonist), SKF 38393 (a D(1) receptor agonist), quinpirole (a D(2)/D(3) receptor agonist) or haloperidol (a D(2)/D(3) receptor antagonist). Thus, attenuation of LI is mainly caused by disruption of 5-HT-ergic systems via 5-HT(2A) receptors. In addition, 5-HT release from hippocampal and hypothalamic slices was significantly reduced. Therefore, ablation of STX1A may cause disruption of 5-HT-ergic transmission and induce abnormal behavior.


Subject(s)
Behavior, Animal/physiology , Serotonin/metabolism , Syntaxin 1/genetics , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Amphetamines/pharmacology , Animals , Behavior, Animal/drug effects , Desipramine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Fluoxetine/pharmacology , Haloperidol/pharmacology , Hippocampus/cytology , Hippocampus/metabolism , Humans , Hypothalamus/cytology , Hypothalamus/metabolism , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Piperazines/pharmacology , Quinpirole/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Social Behavior
18.
J Neurosci Methods ; 175(1): 125-32, 2008 Oct 30.
Article in English | MEDLINE | ID: mdl-18775748

ABSTRACT

We developed a novel strategy for conditional silencing of synaptic transmission in specific neuronal types in transgenic animals. We generated a recombinant protein termed immuno-tetanus toxin (ITet), which contains a monoclonal antibody variable region for human interleukin-2 receptor alpha-subunit (IL-2Ralpha) fused to tetanus toxin light chain. ITet was designed to transiently suppress transmitter release from target neurons genetically engineered to express human IL-2Ralpha via proteolytic cleavage of vesicle-associated membrane protein-2 (VAMP-2). The in vivo actions of ITet were investigated by using mutant mice that express IL-2Ralpha in striatal neurons under the control of the gene encoding dopamine D(2) receptor. Unilateral ITet injection into the striatum induced rotational behavior in the mutant mice and the rotations gradually reversed to the normal level. The behavioral alteration was accompanied by a transient decrease in the striatal VAMP-2 level and depolarization-evoked transmitter release in synaptic target region. However, ITet injection caused no structural change in striatal cells and nerve terminals in the mutants. These data indicate that ITet acts on striatal neurons bearing human IL-2Ralpha and temporally reduces their VAMP-2 content, thereby causing the blockade of transmitter release. Our ITet technology provides a useful approach for inducible and reversible control of synaptic transmission in specific neuronal types in the brain.


Subject(s)
Neurons/drug effects , Neurotransmitter Agents/metabolism , Tetanus Toxin/pharmacology , Analysis of Variance , Animals , Antibodies/pharmacology , Behavior, Animal/drug effects , Chromatography, High Pressure Liquid/methods , Corpus Striatum/cytology , Functional Laterality , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Transgenic , Potassium Chloride/pharmacology , Receptors, Dopamine D2/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Tetanus Toxin/chemistry , Time Factors , Vesicle-Associated Membrane Protein 2/metabolism , gamma-Aminobutyric Acid/metabolism
19.
J Cell Biol ; 177(4): 695-705, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17502420

ABSTRACT

The mechanism of glucose-induced biphasic insulin release is unknown. We used total internal reflection fluorescence (TIRF) imaging analysis to reveal the process of first- and second-phase insulin exocytosis in pancreatic beta cells. This analysis showed that previously docked insulin granules fused at the site of syntaxin (Synt)1A clusters during the first phase; however, the newcomers fused during the second phase external to the Synt1A clusters. To reveal the function of Synt1A in phasic insulin exocytosis, we generated Synt1A-knockout (Synt1A(-/-)) mice. Synt1A(-/-) beta cells showed fewer previously docked granules with no fusion during the first phase; second-phase fusion from newcomers was preserved. Rescue experiments restoring Synt1A expression demonstrated restoration of granule docking status and fusion events. Inhibition of other syntaxins, Synt3 and Synt4, did not affect second-phase insulin exocytosis. We conclude that the first phase is Synt1A dependent but the second phase is not. This indicates that the two phases of insulin exocytosis differ spatially and mechanistically.


Subject(s)
Exocytosis/physiology , Insulin/metabolism , Animals , Cells, Cultured , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Microscopy, Fluorescence
20.
J Neurosci ; 26(21): 5767-76, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16723534

ABSTRACT

The protein HPC-1/syntaxin 1A is abundantly expressed in neurons and localized in the neuronal plasma membrane. It forms a complex with SNAP-25 (25 kDa synaptosomal-associated protein) and VAMP-2 (vesicle-associated membrane protein)/synaptobrevin called SNARE (a soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor) complex, which is considered essential for synaptic vesicle exocytosis; thus, HPC-1/syntaxin 1A is considered crucial for synaptic transmission. To examine the physiological function of HPC-1/syntaxin 1A in vivo, we produced knock-out (KO) mice by targeted gene disruption. Although HPC-1/syntaxin 1A expression was completely depleted without any effect on the expression of other SNARE proteins, the KO mice were viable. They grew normally, were fertile, and displayed no difference in appearance compared with control littermate. In cultured hippocampal neurons derived from the KO mice, the basic synaptic transmission in vitro was normal. However, the mutant mice had impaired long-term potentiation in the hippocampal slice. Also, although KO mice exhibited normal spatial memory in the hidden platform test, consolidation of conditioned fear memory was impaired. Interestingly, the KO mice had impaired conditioned fear memory extinction. These observations suggest that HPC-1/syntaxin 1A may be closely related to synaptic plasticity.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Synaptic Transmission/physiology , Syntaxin 1/metabolism , Animals , Mice , Syntaxin 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...