Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(4): 103040, 2023 04.
Article in English | MEDLINE | ID: mdl-36803960

ABSTRACT

A hippocampal mossy fiber synapse implicated in learning and memory is a complex structure in which a presynaptic bouton attaches to the dendritic trunk by puncta adherentia junctions (PAJs) and wraps multiply branched spines. The postsynaptic densities (PSDs) are localized at the heads of each of these spines and faces to the presynaptic active zones. We previously showed that the scaffolding protein afadin regulates the formation of the PAJs, PSDs, and active zones in the mossy fiber synapse. Afadin has two splice variants: l-afadin and s-afadin. l-Afadin, but not s-afadin, regulates the formation of the PAJs but the roles of s-afadin in synaptogenesis remain unknown. We found here that s-afadin more preferentially bound to MAGUIN (a product of the Cnksr2 gene) than l-afadin in vivo and in vitro. MAGUIN/CNKSR2 is one of the causative genes for nonsyndromic X-linked intellectual disability accompanied by epilepsy and aphasia. Genetic ablation of MAGUIN impaired PSD-95 localization and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic (AMPA) receptor surface accumulation in cultured hippocampal neurons. Our electrophysiological analysis revealed that the postsynaptic response to glutamate, but not its release from the presynapse, was impaired in the MAGUIN-deficient cultured hippocampal neurons. Furthermore, disruption of MAGUIN did not increase the seizure susceptibility to flurothyl, a GABAA receptor antagonist. These results indicate that s-afadin binds to MAGUIN and regulates the PSD-95-dependent cell surface localization of the AMPA receptor and glutamatergic synaptic responses in the hippocampal neurons and that MAGUIN is not involved in the induction of epileptic seizure by flurothyl in our mouse model.


Subject(s)
Microfilament Proteins , Receptors, AMPA , Synapses , Animals , Mice , Disks Large Homolog 4 Protein/metabolism , Flurothyl , Hippocampus/metabolism , Microfilament Proteins/metabolism , Mossy Fibers, Hippocampal/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Transcription Factors/metabolism
2.
Development ; 150(4)2023 02 15.
Article in English | MEDLINE | ID: mdl-36458527

ABSTRACT

Ramified, polarized protoplasmic astrocytes interact with synapses via perisynaptic astrocyte processes (PAPs) to form tripartite synapses. These astrocyte-synapse interactions mutually regulate their structures and functions. However, molecular mechanisms for tripartite synapse formation remain elusive. We developed an in vitro co-culture system for mouse astrocytes and neurons that induced astrocyte ramifications and PAP formation. Co-cultured neurons were required for astrocyte ramifications in a neuronal activity-dependent manner, and synaptically-released glutamate and activation of astrocytic mGluR5 metabotropic glutamate receptor were likely involved in astrocyte ramifications. Astrocytic Necl2 trans-interacted with axonal Necl3, inducing astrocyte-synapse interactions and astrocyte functional polarization by recruiting EAAT1/2 glutamate transporters and Kir4.1 K+ channel to the PAPs, without affecting astrocyte ramifications. This Necl2/3 trans-interaction increased functional synapse number. Thus, astrocytic Necl2, synaptically-released glutamate and axonal Necl3 cooperatively formed tripartite glutamatergic synapses in vitro. Studies on hippocampal mossy fiber synapses in Necl3 knockout and Necl2/3 double knockout mice confirmed these previously unreported mechanisms for astrocyte-synapse interactions and astrocyte functional polarization in vivo.


Subject(s)
Glutamic Acid , Synapses , Mice , Animals , Synapses/physiology , Mice, Knockout , Glutamic Acid/pharmacology , Astrocytes/physiology , Mossy Fibers, Hippocampal
3.
Transl Psychiatry ; 11(1): 548, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34697299

ABSTRACT

An increasing body of evidence suggests that impaired synapse development and function are associated with schizophrenia; however, the underlying molecular pathophysiological mechanism of the disease remains largely unclear. We conducted a family-based study combined with molecular and cellular analysis using induced pluripotent stem cell (iPSC) technology. We generated iPSCs from patients with familial schizophrenia, differentiated these cells into neurons, and investigated the molecular and cellular phenotypes of the patient's neurons. We identified multiple altered synaptic functions, including increased glutamatergic synaptic transmission, higher synaptic density, and altered splicing of dopamine D2 receptor mRNA in iPSC-derived neurons from patients. We also identified patients' specific genetic mutations using whole-exome sequencing. Our findings support the notion that altered synaptic function may underlie the molecular and cellular pathophysiology of schizophrenia, and that multiple genetic factors cooperatively contribute to the development of schizophrenia.


Subject(s)
Induced Pluripotent Stem Cells , Schizophrenia , Cell Differentiation , Humans , Neurons , Receptors, Dopamine D2/genetics , Schizophrenia/genetics
4.
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
5.
Sci Rep ; 7(1): 13409, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042611

ABSTRACT

The roles of calcium-calmodulin-dependent protein kinase II-alpha (CaMKIIα) in the expression of long-term synaptic plasticity in the adult brain have been extensively studied. However, how increased CaMKIIα activity controls the maturation of neuronal circuits remains incompletely understood. Herein, we show that pyramidal neurons without CaMKIIα activity upregulate the rate of spine addition, resulting in elevated spine density. Genetic elimination of CaMKIIα activity specifically eliminated the observed maturation-dependent suppression of spine formation. Enhanced spine formation was associated with the stabilization of actin in the spine and could be reversed by increasing the activity of the small GTPase Rap1. CaMKIIα activity was critical in the phosphorylation of synaptic Ras GTPase-activating protein (synGAP), the dispersion of synGAP from postsynaptic sites, and the activation of postsynaptic Rap1. CaMKIIα is already known to be essential in learning and memory, but our findings suggest that CaMKIIα plays an important activity-dependent role in restricting spine density during postnatal development.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Differentiation/genetics , Dendritic Spines/metabolism , Neurons/cytology , Neurons/metabolism , rap1 GTP-Binding Proteins/genetics , Animals , Biomarkers , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Fluorescent Antibody Technique , Hippocampus , Mice , Models, Biological , Neuronal Plasticity , Phosphorylation , Protein Binding , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , rap1 GTP-Binding Proteins/metabolism
6.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28197549

ABSTRACT

Inhibitory synapses are established during development but continue to be generated and modulated in strength in the mature nervous system. In the spinal cord and brainstem, presynaptically released inhibitory neurotransmitter dominantly switches from GABA to glycine during normal development in vivo. While presynaptic mechanisms of the shift of inhibitory neurotransmission are well investigated, the contribution of postsynaptic neurotransmitter receptors to this shift is not fully elucidated. Synaptic clustering of glycine receptors (GlyRs) is regulated by activation-dependent depolarization in early development. However, GlyR activation induces hyperpolarization after the first postnatal week, and little is known whether and how presynaptically released glycine regulates postsynaptic receptors in a depolarization-independent manner in mature developmental stage. Here we developed spinal cord neuronal culture of rodents using chronic strychnine application to investigate whether initial activation of GlyRs in mature stage could change postsynaptic localization of GlyRs. Immunocytochemical analyses demonstrate that chronic blockade of GlyR activation until mature developmental stage resulted in smaller clusters of postsynaptic GlyRs that could be enlarged upon receptor activation for 1 h in the mature stage. Furthermore, live cell-imaging techniques show that GlyR activation decreases its lateral diffusion at synapses, and this phenomenon is dependent on PKC, but neither Ca2+ nor CaMKII activity. These results suggest that the GlyR activation can regulate receptor diffusion and cluster size at inhibitory synapses in mature stage, providing not only new insights into the postsynaptic mechanism of shifting inhibitory neurotransmission but also the inhibitory synaptic plasticity in mature nervous system.


Subject(s)
Inhibitory Postsynaptic Potentials/physiology , Neurons/metabolism , Protein Transport/physiology , Receptors, Glycine/metabolism , Spinal Cord/cytology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Female , Glycine Agents/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Photobleaching , Protein Transport/drug effects , Receptors, Glycine/genetics , Strychnine/pharmacology , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
7.
Nat Commun ; 5: 4742, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25144834

ABSTRACT

Developmental deficits in neuronal connectivity are considered to be present in patients with autism spectrum disorders (ASDs). Here we examine this possibility by using in vivo spine imaging in the early postnatal cortex of ASD mouse models. Spines are classified by the presence of either the excitatory postsynaptic marker PSD-95 or the inhibitory postsynaptic marker gephyrin. ASD mouse models show consistent upregulation in the dynamics of PSD-95-positive spines, which may subsequently contribute to stable synaptic connectivity. In contrast, spines receiving inputs from the thalamus, detected by the presence of gephyrin clusters, are larger, highly stable and unaffected in ASD mouse models. Importantly, two distinct mouse models, human 15q11-13 duplication and neuroligin-3 R451C point mutation, show highly similar phenotypes in spine dynamics. This selective impairment in dynamics of PSD-95-positive spines receiving intracortical projections may be a core component of early pathological changes and be a potential target of early intervention.


Subject(s)
Autistic Disorder/physiopathology , Dendritic Spines/pathology , Animals , Autistic Disorder/etiology , Biomarkers/analysis , Biomarkers/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Dendritic Spines/physiology , Disease Models, Animal , Disks Large Homolog 4 Protein , Female , Green Fluorescent Proteins , Guanylate Kinases/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Post-Synaptic Density , Pregnancy , Synapses/physiology
8.
Nat Commun ; 4: 1440, 2013.
Article in English | MEDLINE | ID: mdl-23385585

ABSTRACT

Dendritic morphogenesis and formation of synapses at appropriate dendritic locations are essential for the establishment of proper neuronal connectivity. Recent imaging studies provide evidence for stabilization of dynamic distal branches of dendrites by the addition of new synapses. However, molecules involved in both dendritic growth and suppression of synapse maturation remain to be identified. Here we report two distinct functions of doublecortin-like kinases, chimeric proteins containing both a microtubule-binding domain and a kinase domain in postmitotic neurons. First, doublecortin-like kinases localize to the distal dendrites and promote their growth by enhancing microtubule bundling. Second, doublecortin-like kinases suppress maturation of synapses through multiple pathways, including reduction of PSD-95 by the kinase domain and suppression of spine structural maturation by the microtubule-binding domain. Thus, doublecortin-like kinases are critical regulators of dendritic development by means of their specific targeting to the distal dendrites, and their local control of dendritic growth and synapse maturation.


Subject(s)
Dendrites/enzymology , Protein Serine-Threonine Kinases/metabolism , Synapses/enzymology , Animals , Animals, Newborn , Dendritic Spines/enzymology , Doublecortin-Like Kinases , Excitatory Postsynaptic Potentials , Gene Knockdown Techniques , Glutamic Acid/metabolism , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Mice , Mice, Knockout , Microscopy, Fluorescence , Phenotype , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , Protein Transport , RNA, Small Interfering/metabolism , Receptors, AMPA/metabolism , Signal Transduction , Subcellular Fractions/enzymology
9.
Mol Cell Neurosci ; 49(2): 184-95, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22146684

ABSTRACT

Axonal varicosities and dendritic spines at excitatory synapses are dynamic structures essential for synaptic plasticity, whereas the behavior of inhibitory synapses during development and plasticity remains largely unknown. To investigate the morphology and dynamics of inhibitory synapses, we used two distinct pre- and postsynaptic fluorescent probes: one is a yellow fluorescent protein, Venus, incorporated into vesicular GABA transporter (VGAT) gene as a specific marker of presynaptic inhibitory neurons and the other red fluorescent protein (mCherry)-tagged gephyrin, a postsynaptic scaffolding protein, as a postsynaptic marker. Using primary culture of mouse hippocampal neurons and confocal laser-scanning microscopy, we established a system by which close contacts of Venus-positive axonal varicosities with mCherry-labeled gephyrin clusters in the dendritic shafts of dissociated hippocampal pyramidal neurons could be clearly visualized. Time-lapse imaging revealed that: (1) the presynaptic varicosities actively moved with marked changes in their shapes, and the postsynaptic scaffolding protein gephyrin clusters underwent coordinated movements in a tight association with the presynaptic varicosities, (2) the extents of morphological changes and movements depended on the developmental stages, reaching a stable level as the inhibitory synaptic connections matured, and (3) the motility indexes of the varicosity and its counterpart gephyrin cluster were well correlated. Furthermore, action potential blockade with tetrodotoxin treatment reduced the varicosity size, gephyrin cluster mobility as well as the amplitude of GABAergic synaptic currents in pyramidal neurons. Such a neural activity-dependent dynamic change in GABAergic synaptic morphology is likely to play a critical role in the regulatory mechanism underlying the formation and plasticity of inhibitory synapses.


Subject(s)
Hippocampus/cytology , Synapses/ultrastructure , Animals , Axons/metabolism , Axons/ultrastructure , Carrier Proteins/metabolism , Cells, Cultured , Dendritic Spines/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Synapses/metabolism , Synapses/physiology , Time-Lapse Imaging/methods , Vesicular Inhibitory Amino Acid Transport Proteins/physiology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
10.
J Neurochem ; 105(4): 1384-93, 2008 May.
Article in English | MEDLINE | ID: mdl-18331582

ABSTRACT

The NMDA receptor regulates spine morphological plasticity by modulating Rho GTPases. However, the molecular mechanisms for NMDA receptor-mediated regulation of Rho GTPases remain elusive. In this study, we show that p250GAP, an NMDA receptor-associated RhoGAP, regulates spine morphogenesis by modulating RhoA activity. Knock-down of p250GAP increased spine width and elevated the endogenous RhoA activity in primary hippocampal neurons. The increased spine width by p250GAP knock-down was suppressed by the expression of a dominant-negative form of RhoA. Furthermore, p250GAP is involved in NMDA receptor-mediated RhoA activation. In response to NMDA receptor activation, exogenously expressed green fluorescent protein (GFP)-tagged p250GAP was redistributed. Thus, these data suggest that p250GAP plays an important role in NMDA receptor-mediated regulation of RhoA activity leading to spine morphological plasticity.


Subject(s)
Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , GTPase-Activating Proteins/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Dendritic Spines/enzymology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Mice , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/genetics
11.
J Neurosci ; 26(29): 7693-706, 2006 Jul 19.
Article in English | MEDLINE | ID: mdl-16855097

ABSTRACT

Organization and dynamic remodeling of postsynaptic density (PSD) are thought to be critical in postsynaptic signal transduction, but the underlying molecular mechanisms are not well understood. We show here that four major scaffolding molecules, PSD-95, GKAP, Shank, and PSD-Zip45, show distinct instability in total molecular content per synapse. Fluorescence recovery after photobleaching also confirmed their distinct turnover rates. Among the PSD molecules examined, PSD-95 was most stable, but its elimination did not influence the dynamics of its direct binding partner GKAP. Multiple interactions of scaffolding molecules with the actin cytoskeleton have suggested their importance in both maintenance and remodeling of the PSD. Indeed, acute pharmacological disruption of F-actin rapidly eliminated the dynamic fraction of GKAP, Shank, and PSD-Zip45, without changing synaptic localization of PSD-95. GKAP content in synapses increased after pharmacological enhancement of neuronal activity, whereas Shank and PSD-Zip45 content showed reduction. Inhibition of F-actin dynamics prevented activity-dependent redistribution of all three scaffolds. We also assessed involvement of glutamate receptors in the regulation of PSD dynamics. Genetic manipulations eliminating either NMDA receptors or metabotropic glutamate receptors did not primarily influence mobility of their binding scaffolds. These results collectively indicate a critical role of filamentous actin in determining the extent of dynamic reorganization in PSD molecular composition.


Subject(s)
Actins/physiology , Synaptic Membranes/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Disks Large Homolog 4 Protein , Fluorescence Recovery After Photobleaching , Guanylate Kinases , Homer Scaffolding Proteins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Microscopy, Confocal , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/physiology , Protein Structure, Tertiary/physiology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Receptors, N-Methyl-D-Aspartate/deficiency , SAP90-PSD95 Associated Proteins , Synaptic Membranes/metabolism , Tissue Distribution , Transfection
12.
Cell ; 110(4): 429-41, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12202033

ABSTRACT

The adult brain is extremely vulnerable to various insults. The recent discovery of neural progenitors in adult mammals, however, raises the possibility of repairing damaged tissue by recruiting their latent regenerative potential. Here we show that activation of endogenous progenitors leads to massive regeneration of hippocampal pyramidal neurons after ischemic brain injury. Endogenous progenitors proliferate in response to ischemia and subsequently migrate into the hippocampus to regenerate new neurons. Intraventricular infusion of growth factors markedly augments these responses, thereby increasing the number of newborn neurons. Our studies suggest that regenerated neurons are integrated into the existing brain circuitry and contribute to ameliorating neurological deficits. These results expand the possibility of novel neuronal cell regeneration therapies for stroke and other neurological diseases.


Subject(s)
Brain Ischemia/drug therapy , Growth Substances/pharmacology , Hippocampus/growth & development , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/metabolism , Stem Cells/metabolism , Animals , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Dendrites/drug effects , Dendrites/metabolism , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Growth Substances/therapeutic use , Hippocampus/drug effects , Hippocampus/ultrastructure , Male , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Regeneration/drug effects , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/ultrastructure , Neuronal Plasticity/drug effects , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/ultrastructure , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology , Stem Cells/drug effects , Stem Cells/ultrastructure , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...