Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Front Mol Neurosci ; 17: 1379089, 2024.
Article in English | MEDLINE | ID: mdl-38628370

ABSTRACT

Protein phosphorylation, a key regulator of cellular processes, plays a central role in brain function and is implicated in neurological disorders. Information on protein phosphorylation is expected to be a clue for understanding various neuropsychiatric disorders and developing therapeutic strategies. Nonetheless, existing databases lack a specific focus on phosphorylation events in the brain, which are crucial for investigating the downstream pathway regulated by neurotransmitters. To overcome the gap, we have developed a web-based database named "Kinase-Associated Neural PHOspho-Signaling (KANPHOS)." This paper presents the design concept, detailed features, and a series of improvements for KANPHOS. KANPHOS is designed to support data-driven research by fulfilling three key objectives: (1) enabling the search for protein kinases and their substrates related to extracellular signals or diseases; (2) facilitating a consolidated search for information encompassing phosphorylated substrate genes, proteins, mutant mice, diseases, and more; and (3) offering integrated functionalities to support pathway and network analysis. KANPHOS is also equipped with API functionality to interact with external databases and analysis tools, enhancing its utility in data-driven investigations. Those key features represent a critical step toward unraveling the complex landscape of protein phosphorylation in the brain, with implications for elucidating the molecular mechanisms underlying neurological disorders. KANPHOS is freely accessible to all researchers at https://kanphos.jp.

2.
Front Mol Neurosci ; 17: 1376762, 2024.
Article in English | MEDLINE | ID: mdl-38516040

ABSTRACT

The unraveling of the regulatory mechanisms that govern neuronal excitability is a major challenge for neuroscientists worldwide. Neurotransmitters play a critical role in maintaining the balance between excitatory and inhibitory activity in the brain. The balance controls cognitive functions and emotional responses. Glutamate and γ-aminobutyric acid (GABA) are the primary excitatory and inhibitory neurotransmitters of the brain, respectively. Disruptions in the balance between excitatory and inhibitory transmission are implicated in several psychiatric disorders, including anxiety disorders, depression, and schizophrenia. Neuromodulators such as dopamine and acetylcholine control cognition and emotion by regulating the excitatory/inhibitory balance initiated by glutamate and GABA. Dopamine is closely associated with reward-related behaviors, while acetylcholine plays a role in aversive and attentional behaviors. Although the physiological roles of neuromodulators have been extensively studied neuroanatomically and electrophysiologically, few researchers have explored the interplay between neuronal excitability and cell signaling and the resulting impact on emotion regulation. This review provides an in-depth understanding of "cell signaling crosstalk" in the context of neuronal excitability and emotion regulation. It also anticipates that the next generation of neurochemical analyses, facilitated by integrated phosphorylation studies, will shed more light on this topic.

3.
Expert Rev Proteomics ; 20(11): 291-298, 2023.
Article in English | MEDLINE | ID: mdl-37787112

ABSTRACT

INTRODUCTION: Since the emergence of the cholinergic hypothesis of Alzheimer's disease (AD), acetylcholine has been viewed as a mediator of learning and memory. Donepezil improves AD-associated learning deficits and memory loss by recovering brain acetylcholine levels. However, it is associated with side effects due to global activation of acetylcholine receptors. Muscarinic acetylcholine receptor M1 (M1R), a key mediator of learning and memory, has been an alternative target. The importance of targeting a specific pathway downstream of M1R has recently been recognized. Elucidating signaling pathways beyond M1R that lead to learning and memory holds important clues for AD therapeutic strategies. AREAS COVERED: This review first summarizes the role of acetylcholine in aversive learning, one of the outputs used for preliminary AD drug screening. It then describes the phosphoproteomic approach focused on identifying acetylcholine intracellular signaling pathways leading to aversive learning. Finally, the intracellular mechanism of donepezil and its effect on learning and memory is discussed. EXPERT OPINION: The elucidation of signaling pathways beyond M1R by phosphoproteomic approach offers a platform for understanding the intracellular mechanism of AD drugs and for developing AD therapeutic strategies. Clarifying the molecular mechanism that links the identified acetylcholine signaling to AD pathophysiology will advance the development of AD therapeutic strategies.


Subject(s)
Acetylcholine , Alzheimer Disease , Humans , Acetylcholine/pharmacology , Acetylcholine/therapeutic use , Receptor, Muscarinic M1/metabolism , Donepezil/pharmacology , Donepezil/therapeutic use , Signal Transduction , Alzheimer Disease/drug therapy
4.
Nat Commun ; 14(1): 3293, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280197

ABSTRACT

Lissencephaly-1 (LIS1) is associated with neurodevelopmental diseases and is known to regulate the molecular motor cytoplasmic dynein activity. Here we show that LIS1 is essential for the viability of mouse embryonic stem cells (mESCs), and it governs the physical properties of these cells. LIS1 dosage substantially affects gene expression, and we uncovered an unexpected interaction of LIS1 with RNA and RNA-binding proteins, most prominently the Argonaute complex. We demonstrate that LIS1 overexpression partially rescued the extracellular matrix (ECM) expression and mechanosensitive genes conferring stiffness to Argonaute null mESCs. Collectively, our data transforms the current perspective on the roles of LIS1 in post-transcriptional regulation underlying development and mechanosensitive processes.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase , Argonaute Proteins , Embryonic Stem Cells , Microtubule-Associated Proteins , Animals , Mice , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Survival , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Pluripotent Stem Cells , Protein Interaction Maps , Argonaute Proteins/metabolism
5.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232945

ABSTRACT

Dopamine regulates emotional behaviors, including rewarding and aversive behaviors, through the mesolimbic dopaminergic pathway, which projects dopamine neurons from the ventral tegmental area to the nucleus accumbens (NAc). Protein phosphorylation is critical for intracellular signaling pathways and physiological functions, which are regulated by neurotransmitters in the brain. Previous studies have demonstrated that dopamine stimulated the phosphorylation of intracellular substrates, such as receptors, ion channels, and transcription factors, to regulate neuronal excitability and synaptic plasticity through dopamine receptors. We also established a novel database called KANPHOS that provides information on phosphorylation signals downstream of monoamines identified by our kinase substrate screening methods, including dopamine, in addition to those reported in the literature. Recent advances in proteomics techniques have enabled us to clarify the mechanisms through which dopamine controls rewarding and aversive behaviors through signal pathways in the NAc. In this review, we discuss the intracellular phosphorylation signals regulated by dopamine in these two emotional behaviors.


Subject(s)
Dopamine , Ventral Tegmental Area , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neurotransmitter Agents/metabolism , Nucleus Accumbens/metabolism , Phosphorylation , Receptors, Dopamine/metabolism , Transcription Factors/metabolism , Ventral Tegmental Area/metabolism
6.
Cell Rep ; 40(10): 111309, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36070693

ABSTRACT

Dysfunctional dopamine signaling is implicated in various neuropsychological disorders. Previously, we reported that dopamine increases D1 receptor (D1R)-expressing medium spiny neuron (MSN) excitability and firing rates in the nucleus accumbens (NAc) via the PKA/Rap1/ERK pathway to promote reward behavior. Here, the results show that the D1R agonist, SKF81297, inhibits KCNQ-mediated currents and increases D1R-MSN firing rates in murine NAc slices, which is abolished by ERK inhibition. In vitro ERK phosphorylates KCNQ2 at Ser414 and Ser476; in vivo, KCNQ2 is phosphorylated downstream of dopamine signaling in NAc slices. Conditional deletion of Kcnq2 in D1R-MSNs reduces the inhibitory effect of SKF81297 on KCNQ channel activity, while enhancing neuronal excitability and cocaine-induced reward behavior. These effects are restored by wild-type, but not phospho-deficient KCNQ2. Hence, D1R-ERK signaling controls MSN excitability via KCNQ2 phosphorylation to regulate reward behavior, making KCNQ2 a potential therapeutical target for psychiatric diseases with a dysfunctional reward circuit.


Subject(s)
Dopamine , KCNQ2 Potassium Channel , Mental Disorders , Nerve Tissue Proteins , Animals , Dopamine/metabolism , KCNQ2 Potassium Channel/metabolism , Mental Disorders/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Receptors, Dopamine D1/metabolism , Reward
7.
Mol Psychiatry ; 27(8): 3479-3492, 2022 08.
Article in English | MEDLINE | ID: mdl-35665767

ABSTRACT

Acetylcholine is a neuromodulator critical for learning and memory. The cholinesterase inhibitor donepezil increases brain acetylcholine levels and improves Alzheimer's disease (AD)-associated learning disabilities. Acetylcholine activates striatal/nucleus accumbens dopamine receptor D2-expressing medium spiny neurons (D2R-MSNs), which regulate aversive learning through muscarinic receptor M1 (M1R). However, how acetylcholine stimulates learning beyond M1Rs remains unresolved. Here, we found that acetylcholine stimulated protein kinase C (PKC) in mouse striatal/nucleus accumbens. Our original kinase-oriented phosphoproteomic analysis revealed 116 PKC substrate candidates, including Rac1 activator ß-PIX. Acetylcholine induced ß-PIX phosphorylation and activation, thereby stimulating Rac1 effector p21-activated kinase (PAK). Aversive stimulus activated the M1R-PKC-PAK pathway in mouse D2R-MSNs. D2R-MSN-specific expression of PAK mutants by the Cre-Flex system regulated dendritic spine structural plasticity and aversive learning. Donepezil induced PAK activation in both accumbal D2R-MSNs and in the CA1 region of the hippocampus and enhanced D2R-MSN-mediated aversive learning. These findings demonstrate that acetylcholine stimulates M1R-PKC-ß-PIX-Rac1-PAK signaling in D2R-MSNs for aversive learning and imply the cascade's therapeutic potential for AD as aversive learning is used to preliminarily screen AD drugs.


Subject(s)
Acetylcholine , p21-Activated Kinases , Animals , Mice , Protein Kinase C , Donepezil/pharmacology , Brain
8.
Neurochem Res ; 47(9): 2757-2772, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35624196

ABSTRACT

The structural plasticity of dendritic spines plays a critical role in NMDA-induced long-term potentiation (LTP) in the brain. The small GTPases RhoA and Ras are considered key regulators of spine morphology and enlargement. However, the regulatory interaction between RhoA and Ras underlying NMDA-induced spine enlargement is largely unknown. In this study, we found that Rho-kinase/ROCK, an effector of RhoA, phosphorylated SynGAP1 (a synaptic Ras-GTPase activating protein) at Ser842 and increased its interaction with 14-3-3ζ, thereby activating Ras-ERK signaling in a reconstitution system in HeLa cells. We also found that the stimulation of NMDA receptor by glycine treatment for LTP induction stimulated SynGAP1 phosphorylation, Ras-ERK activation, spine enlargement and SynGAP1 delocalization from the spines in striatal neurons, and these effects were prevented by Rho-kinase inhibition. Rho-kinase-mediated phosphorylation of SynGAP1 appeared to increase its dissociation from PSD95, a postsynaptic scaffolding protein located at postsynaptic density, by forming a complex with 14-3-3ζ. These results suggest that Rho-kinase phosphorylates SynGAP1 at Ser842, thereby activating the Ras-ERK pathway for NMDA-induced morphological changes in dendritic spines.


Subject(s)
Dendritic Spines , Long-Term Potentiation , ras GTPase-Activating Proteins , 14-3-3 Proteins/metabolism , Animals , Dendritic Spines/metabolism , HeLa Cells , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , N-Methylaspartate/metabolism , ras GTPase-Activating Proteins/metabolism , rhoA GTP-Binding Protein
9.
J Neurochem ; 160(3): 325-341, 2022 02.
Article in English | MEDLINE | ID: mdl-34878647

ABSTRACT

The nucleus accumbens (NAc) plays critical roles in emotional behaviors, including aversive learning. Aversive stimuli such as an electric foot shock increase acetylcholine (ACh) in the NAc, and muscarinic signaling appears to increase neuronal excitability and aversive learning. Muscarinic signaling inhibits the voltage-dependent potassium KCNQ current which regulates neuronal excitability, but the regulatory mechanism has not been fully elucidated. Phosphorylation of KCNQ2 at threonine 217 (T217) and its inhibitory effect on channel activity were predicted. However, whether and how muscarinic signaling phosphorylates KCNQ2 in vivo remains unclear. Here, we found that PKC directly phosphorylated KCNQ2 at T217 in vitro. Carbachol and a muscarinic M1 receptor (M1R) agonist facilitated KCNQ2 phosphorylation at T217 in NAc/striatum slices in a PKC-dependent manner. Systemic administration of the cholinesterase inhibitor donepezil, which is commonly used to treat dementia, and electric foot shock to mice induced the phosphorylation of KCNQ2 at T217 in the NAc, whereas phosphorylation was suppressed by an M1R antagonist. Conditional deletion of Kcnq2 in the NAc enhanced electric foot shock induced aversive learning. Our findings indicate that muscarinic signaling induces the phosphorylation of KCNQ2 at T217 via PKC activation for aversive learning.


Subject(s)
Avoidance Learning/physiology , KCNQ2 Potassium Channel/metabolism , Nerve Tissue Proteins/metabolism , Nucleus Accumbens/metabolism , Parasympathetic Nervous System/physiology , Protein Kinase C/metabolism , Receptors, Muscarinic/physiology , Animals , Carbachol/pharmacology , Cholinesterase Inhibitors/pharmacology , Donepezil/pharmacology , KCNQ2 Potassium Channel/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Nerve Tissue Proteins/genetics , Phosphorylation , Receptor, Muscarinic M2/drug effects
10.
Cells ; 11(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35011609

ABSTRACT

Protein phosphorylation plays critical roles in a variety of intracellular signaling pathways and physiological functions that are controlled by neurotransmitters and neuromodulators in the brain. Dysregulation of these signaling pathways has been implicated in neurodevelopmental disorders, including autism spectrum disorder, attention deficit hyperactivity disorder and schizophrenia. While recent advances in mass spectrometry-based proteomics have allowed us to identify approximately 280,000 phosphorylation sites, it remains largely unknown which sites are phosphorylated by which kinases. To overcome this issue, previously, we developed methods for comprehensive screening of the target substrates of given kinases, such as PKA and Rho-kinase, upon stimulation by extracellular signals and identified many candidate substrates for specific kinases and their phosphorylation sites. Here, we developed a novel online database to provide information about the phosphorylation signals identified by our methods, as well as those previously reported in the literature. The "KANPHOS" (Kinase-Associated Neural Phospho-Signaling) database and its web portal were built based on a next-generation XooNIps neuroinformatics tool. To explore the functionality of the KANPHOS database, we obtained phosphoproteomics data for adenosine-A2A-receptor signaling and its downstream MAPK-mediated signaling in the striatum/nucleus accumbens, registered them in KANPHOS, and analyzed the related pathways.


Subject(s)
Brain/metabolism , Databases, Protein , Neurons/metabolism , Protein Kinases/metabolism , Animals , Calcium Channels/metabolism , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Phosphoproteins/metabolism , Phosphorylation , Receptor, Adenosine A2A/metabolism , Substrate Specificity
11.
Cell Struct Funct ; 45(2): 143-154, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32641624

ABSTRACT

Prickle2 has been identified in genetic studies of subjects with autism spectrum disorder (ASD) and epilepsy, but the pathological mechanism of Prickle2 remains to be fully understood. Proteomic analysis of Prickle2 with mass spectrometry revealed twenty-eight Prickle2 interactors, including immunoglobulin superfamily member 9b (Igsf9b), in the brain. Here, because Igsf9 family proteins are associated with psychiatric diseases and seizures, we studied the physiological interaction between Prickle2 and Igsf9b. Prickle2 colocalized with Igsf9b in cultured hippocampal neurons. Knockdown of Prickle2 affected the subcellular localization of Igsf9b. Interestingly, Igsf9b localized along axonal processes in a pattern opposite to the ASD-related molecule ANK3/AnkG. AnkG is a major component of the axon initial segment (AIS), where a variety of ASD and epilepsy susceptibility proteins accumulate. Igsf9b-knockdown neurons displayed altered AnkG localization. Prickle2 depletion caused defects in AnkG and voltage-gated Na+ channel localization, resulting in altered network activity. These results support the idea that Prickle2 regulates AnkG distribution by controlling the proper localization of Igsf9b. The novel function of Prickle2 in AIS cytoarchitecture provides new insights into the shared pathology of ASD and epilepsy.Key words: Prickle2, Igsf9b, axon initial segment, neuronal excitability, ASD.


Subject(s)
Axons/metabolism , Hippocampus/metabolism , LIM Domain Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Ankyrins/genetics , Ankyrins/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Epilepsy/genetics , Epilepsy/metabolism , LIM Domain Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
12.
Stem Cell Reports ; 13(4): 684-699, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31543469

ABSTRACT

Mutations in the microtubule-associated protein tau (MAPT) gene are known to cause familial frontotemporal dementia (FTD). The R406W tau mutation is a unique missense mutation whose patients have been reported to exhibit Alzheimer's disease (AD)-like phenotypes rather than the more typical FTD phenotypes. In this study, we established patient-derived induced pluripotent stem cell (iPSC) models to investigate the disease pathology induced by the R406W mutation. We generated iPSCs from patients and established isogenic lines using CRISPR/Cas9. The iPSCs were induced into cerebral organoids, which were dissociated into cortical neurons with high purity. In this neuronal culture, the mutant tau protein exhibited reduced phosphorylation levels and was increasingly fragmented by calpain. Furthermore, the mutant tau protein was mislocalized and the axons of the patient-derived neurons displayed morphological and functional abnormalities, which were rescued by microtubule stabilization. The findings of our study provide mechanistic insight into tau pathology and a potential for therapeutic intervention.


Subject(s)
Alleles , Amino Acid Substitution , Frontotemporal Dementia/etiology , Induced Pluripotent Stem Cells/metabolism , Mutation , tau Proteins/genetics , Calpain/metabolism , Disease Progression , Disease Susceptibility , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/physiopathology , Humans , Induced Pluripotent Stem Cells/cytology , Mitochondria/metabolism , Neurons/metabolism , Phosphorylation , Phosphotransferases/metabolism , tau Proteins/metabolism
13.
Curr Protoc Chem Biol ; 11(1): e60, 2019 03.
Article in English | MEDLINE | ID: mdl-30615307

ABSTRACT

Protein phosphorylation plays a critical role in the regulation of cellular function. Information on protein phosphorylation and the responsible kinases is important for understanding intracellular signaling. A method for in vivo screening of kinase substrates named KIOSS (kinase-oriented substrate screening) has been developed. This protocol provides a method that utilizes phosphoprotein-binding modules such as 14-3-3 protein, the pin1-WW domain, and the chek2-FHA domain as biological filters to successfully enrich phosphorylated proteins related to intracellular signaling rather than housekeeping and/or structural proteins. More than 1000 substrate candidates for PKA, PKC, MAPK, and Rho-kinase in HeLa cells, as well as phosphorylation downstream of D1R, NMDAR, adenosine A2a receptor, PKA, PKC, MAPK, and Rho-kinase in mouse brain slice cultures have been identified by this method. An online database named KANPHOS (Kinase-Associated Neural Phospho-Signaling) provides the phosphorylation signals identified by these studies, as well as those previously reported in the literature. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Protein Kinases/metabolism , Substrate Specificity , Animals , HeLa Cells , Humans , Mice , Phosphorylation , Tumor Cells, Cultured
14.
J Biochem ; 165(4): 301-307, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30590682

ABSTRACT

Accumulating information on eukaryotic protein phosphorylation implies a large and complicated phospho-signalling network in various cellular processes. Although a large number of protein phosphorylation sites have been detected, their physiological consequences and the linkage between each phosphorylation site and the responsible protein kinase remain largely unexplored. To understand kinase-oriented phospho-signalling pathways, we have developed novel substrate screening technologies. In this review, we described the in vitro and in vivo screening methods named kinase-interacting substrate screening analysis and kinase-oriented substrate screening analysis, respectively.


Subject(s)
Phosphoproteins/metabolism , Protein Kinases/metabolism , Proteomics , Signal Transduction , Animals , Humans , Phosphoproteins/genetics , Protein Kinases/genetics
15.
Neuron ; 89(3): 550-65, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26804993

ABSTRACT

Dopamine (DA) type 1 receptor (D1R) signaling in the striatum presumably regulates neuronal excitability and reward-related behaviors through PKA. However, whether and how D1Rs and PKA regulate neuronal excitability and behavior remain largely unknown. Here, we developed a phosphoproteomic analysis method to identify known and novel PKA substrates downstream of the D1R and obtained more than 100 candidate substrates, including Rap1 GEF (Rasgrp2). We found that PKA phosphorylation of Rasgrp2 activated its guanine nucleotide-exchange activity on Rap1. Cocaine exposure activated Rap1 in the nucleus accumbens in mice. The expression of constitutively active PKA or Rap1 in accumbal D1R-expressing medium spiny neurons (D1R-MSNs) enhanced neuronal firing rates and behavioral responses to cocaine exposure through MAPK. Knockout of Rap1 in the accumbal D1R-MSNs was sufficient to decrease these phenotypes. These findings demonstrate a novel DA-PKA-Rap1-MAPK intracellular signaling mechanism in D1R-MSNs that increases neuronal excitability to enhance reward-related behaviors.


Subject(s)
Dopamine/metabolism , Phosphoproteins/metabolism , Proteome/metabolism , Proteomics , Receptors, Dopamine D1/metabolism , Reward , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , Action Potentials/physiology , Animals , Benzazepines/pharmacology , Cocaine/pharmacology , Colforsin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/pharmacology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Guanine Nucleotide Exchange Factors/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Neurons/physiology , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , rap1 GTP-Binding Proteins/genetics
16.
Cell Rep ; 14(1): 11-21, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26725111

ABSTRACT

Unveiling the molecular and cellular mechanisms underlying memory has been a challenge for the past few decades. Although synaptic plasticity is proven to be essential for memory formation, the significance of "single-cell memory" still remains elusive. Here, we exploited a primary culture system for the analysis of C. elegans neurons and show that a single thermosensory neuron has an ability to form, retain, and reset a temperature memory. Genetic and proteomic analyses found that the expression of the single-cell memory exhibits inter-individual variability, which is controlled by the evolutionarily conserved CaMKI/IV and Raf pathway. The variable responses of a sensory neuron influenced the neural activity of downstream interneurons, suggesting that modulation of the sensory neurons ultimately determines the behavioral output in C. elegans. Our results provide proof of single-cell memory and suggest that the individual differences in neural responses at the single-cell level can confer individuality.


Subject(s)
Caenorhabditis elegans/metabolism , Memory/physiology , Nerve Net/physiology , Neuronal Plasticity/physiology , Signal Transduction/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , raf Kinases/metabolism
17.
Nat Neurosci ; 18(5): 698-707, 2015 May.
Article in English | MEDLINE | ID: mdl-25821909

ABSTRACT

Disrupted-in-schizophrenia 1 (DISC1) is a susceptibility gene for major psychiatric disorders, including schizophrenia. DISC1 has been implicated in neurodevelopment in relation to scaffolding signal complexes. Here we used proteomic analysis to screen for DISC1 interactors and identified several RNA-binding proteins, such as hematopoietic zinc finger (HZF), that act as components of RNA-transporting granules. HZF participates in the mRNA localization of inositol-1,4,5-trisphosphate receptor type 1 (ITPR1), which plays a key role in synaptic plasticity. DISC1 colocalizes with HZF and ITPR1 mRNA in hippocampal dendrites and directly associates with neuronal mRNAs, including ITPR1 mRNA. The binding potential of DISC1 for ITPR1 mRNA is facilitated by HZF. Studies of Disc1-knockout mice have revealed that DISC1 regulates the dendritic transport of Itpr1 mRNA by directly interacting with its mRNA. The DISC1-mediated mRNA regulation is involved in synaptic plasticity. We show that DISC1 binds ITPR1 mRNA with HZF, thereby regulating its dendritic transport for synaptic plasticity.


Subject(s)
Hippocampus/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Proteins/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , 3' Untranslated Regions/genetics , Animals , Biological Transport , Cytoplasmic Granules/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Protein Binding , Protein Interaction Mapping , RNA Interference , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Schizophr Bull ; 41(3): 744-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25332407

ABSTRACT

BACKGROUND: Nuclear distribution E homolog 1 (NDE1), located within chromosome 16p13.11, plays an essential role in microtubule organization, mitosis, and neuronal migration and has been suggested by several studies of rare copy number variants to be a promising schizophrenia (SCZ) candidate gene. Recently, increasing attention has been paid to rare single-nucleotide variants (SNVs) discovered by deep sequencing of candidate genes, because such SNVs may have large effect sizes and their functional analysis may clarify etiopathology. METHODS AND RESULTS: We conducted mutation screening of NDE1 coding exons using 433 SCZ and 145 pervasive developmental disorders samples in order to identify rare single nucleotide variants with a minor allele frequency ≤5%. We then performed genetic association analysis using a large number of unrelated individuals (3554 SCZ, 1041 bipolar disorder [BD], and 4746 controls). Among the discovered novel rare variants, we detected significant associations between SCZ and S214F (P = .039), and between BD and R234C (P = .032). Furthermore, functional assays showed that S214F affected axonal outgrowth and the interaction between NDE1 and YWHAE (14-3-3 epsilon; a neurodevelopmental regulator). CONCLUSIONS: This study strengthens the evidence for association between rare variants within NDE1 and SCZ, and may shed light into the molecular mechanisms underlying this severe psychiatric disorder.


Subject(s)
Bipolar Disorder/genetics , Child Development Disorders, Pervasive/genetics , Microtubule-Associated Proteins/genetics , Schizophrenia/genetics , Adult , Exons , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
19.
Glia ; 61(5): 679-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23382131

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) iplays a crucial role in the antiviral responses of Type I interferons (IFNs). The role of IFITM3 in the central nervous system (CNS) is, however, largely unknown, despite the fact that its expression is increased in the brains of patients with neurologic and neuropsychiatric diseases. Here, we show the role of IFITM3 in long-lasting neuronal impairments in mice following polyriboinosinic-polyribocytidylic acid (polyI:C, a synthetic double-stranded RNA)-induced immune challenge during the early stages of development. We found that the induction of IFITM3 expression in the brain of mice treated with polyI:C was observed only in astrocytes. Cultured astrocytes were activated by polyI:C treatment, leading to an increase in the mRNA levels of inflammatory cytokines as well as Ifitm3. When cultured neurons were treated with the conditioned medium of polyI:C-treated astrocytes (polyI:C-ACM), neurite development was impaired. These polyI:C-ACM-induced neurodevelopmental abnormalities were alleviated by ifitm3(-/-) astrocyte-conditioned medium. Furthermore, decreases of MAP2 expression, spine density, and dendrite complexity in the frontal cortex as well as memory impairment were evident in polyI:C-treated wild-type mice, but such neuronal impairments were not observed in ifitm3(-) (/) (-) mice. We also found that IFITM3 proteins were localized to the early endosomes of astrocytes following polyI:C treatment and reduced endocytic activity. These findings suggest that the induction of IFITM3 expression in astrocytes by the activation of the innate immune system during the early stages of development has non-cell autonomous effects that affect subsequent neurodevelopment, leading to neuropathological impairments and brain dysfunction, by impairing endocytosis in astrocytes.


Subject(s)
Astrocytes/pathology , Membrane Proteins/physiology , Neurons/pathology , Animals , Animals, Newborn , Astrocytes/immunology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cytokines/biosynthesis , Cytokines/genetics , Endocytosis/drug effects , Endocytosis/immunology , Female , Immunity, Innate/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Neurons/immunology , Poly I-C/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects
20.
Hum Mol Genet ; 20(23): 4666-83, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21903668

ABSTRACT

Disrupted-In-Schizophrenia 1 (DISC1) is a promising candidate gene for susceptibility to psychiatric disorders, including schizophrenia. DISC1 appears to be involved in neurogenesis, neuronal migration, axon/dendrite formation and synapse formation; during these processes, DISC1 acts as a scaffold protein by interacting with various partners. However, the lack of Disc1 knockout mice and a well-characterized antibody to DISC1 has made it difficult to determine the exact role of DISC1 in vivo. In this study, we generated mice lacking exons 2 and 3 of the Disc1 gene and prepared specific antibodies to the N- and C-termini of DISC1. The Disc1 mutant mice are viable and fertile, and no gross phenotypes, such as disorganization of the brain's cytoarchitecture, were observed. Western blot analysis revealed that the DISC1-specific antibodies recognize a protein with an apparent molecular mass of ~100 kDa in brain extracts from wild-type mice but not in brain extracts from DISC1 mutant mice. Immunochemical studies demonstrated that DISC1 is mainly localized to the vicinity of the Golgi apparatus in hippocampal neurons and astrocytes. A deficiency of full-length Disc1 induced a threshold shift in the induction of long-term potentiation in the dentate gyrus. The Disc1 mutant mice displayed abnormal emotional behavior as assessed by the elevated plus-maze and cliff-avoidance tests, thereby suggesting that a deficiency of full-length DISC1 may result in lower anxiety and/or higher impulsivity. Based on these results, we suggest that full-length Disc1-deficient mice and DISC1-specific antibodies are powerful tools for dissecting the pathophysiological functions of DISC1.


Subject(s)
Behavior, Animal , Exons/genetics , Gene Targeting , Nerve Tissue Proteins/genetics , Aging/drug effects , Aging/pathology , Amines/metabolism , Animals , Antibodies/immunology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal/drug effects , Clozapine/pharmacology , Gene Expression Regulation, Developmental/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/ultrastructure , Immunohistochemistry , Maze Learning/drug effects , Mice , Mice, Inbred Strains , Nerve Tissue Proteins/immunology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phenotype , Synaptic Transmission/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL