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










Publication year range
1.
STAR Protoc ; 2(4): 100904, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34723214

ABSTRACT

Dendritic spine enlargement by synaptic activation is thought to increase synaptic efficacy underlying learning and memory. This process requires forces generated by actin polymerization and actin-adhesion coupling (clutch coupling). Here, we describe a protocol to monitor actin filament retrograde flow and actin polymerization within spines using a standard epi-fluorescence microscope. In combination with chemical long-term potentiation, this protocol allows us to quantify clutch coupling efficiency and actin polymerization rate, which are essential variables for generating forces for activity-dependent spine enlargement. For complete details on the use and execution of this protocol, please refer to Kastian et al. (2021).


Subject(s)
Actins/metabolism , Dendritic Spines , Hippocampus/cytology , Long-Term Potentiation/physiology , Neurophysiology/methods , Actin Cytoskeleton/metabolism , Animals , Cells, Cultured , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Dendritic Spines/physiology , Mice , Polymerization
2.
J Neurochem ; 158(3): 673-693, 2021 08.
Article in English | MEDLINE | ID: mdl-34107066

ABSTRACT

Dendritic spines are small, actin-rich protrusions that act as the receiving sites of most excitatory inputs in the central nervous system. The remodeling of the synapse architecture is mediated by actin cytoskeleton dynamics, a process precisely regulated by the small Rho GTPase family. Wnt ligands exert their presynaptic and postsynaptic effects during formation and consolidation of the synaptic structure. Specifically, Wnt5a has been identified as an indispensable synaptogenic factor for the regulation and organization of the postsynaptic side; however, the molecular mechanisms through which Wnt5a induces morphological changes resulting from actin cytoskeleton dynamics within dendritic spines remain unclear. In this work, we employ primary rat hippocampal cultures and HT22 murine hippocampal neuronal cell models, molecular and pharmacological tools, and fluorescence microscopy (laser confocal and epifluorescence) to define the Wnt5a-induced molecular signaling involved in postsynaptic remodeling mediated via the regulation of the small Rho GTPase family. We report that Wnt5a differentially regulates the phosphorylation of Cofilin in neurons through both Ras-related C3 botulinum toxin substrate 1 and cell division cycle 42 depending on the subcellular compartment and the extracellular calcium levels. Additionally, we demonstrate that Wnt5a increases the density of dendritic spines and promotes their maturation via Ras-related C3 botulinum toxin substrate 1. Accordingly, we find that Wnt5a requires the combined activation of small Rho GTPases to increase the levels of filamentous actin, thus promoting the stability of actin filaments. Altogether, these results provide evidence for a new mechanism by which Wnt5a may target actin dynamics, thereby regulating the subsequent morphological changes in dendritic spine architecture.


Subject(s)
Actin Depolymerizing Factors/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Neurons/metabolism , Wnt-5a Protein/metabolism , rho GTP-Binding Proteins/metabolism , Actin Depolymerizing Factors/analysis , Animals , Cell Line , Cells, Cultured , Dendritic Spines/chemistry , Enzyme Activation/physiology , Female , Hippocampus/chemistry , Hippocampus/cytology , Neurons/chemistry , Pregnancy , Rats , Rats, Sprague-Dawley , Wnt-5a Protein/analysis , rho GTP-Binding Proteins/analysis
3.
Neurosci Lett ; 725: 134909, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32169587

ABSTRACT

The hippocampus has a well-known role in mediating learning and memory, and its function can be directly regulated by both stress and glucocorticoid receptor activation. Hippocampal contributions to learning are thought to be dependent on changes in the plasticity of synapses within specific subregions, and these functional changes are accompanied by morphological changes in the number and shape of dendritic spines, the physical correlates of these glutamatergic synapses. Serum- and glucocorticoid-inducible kinase 1 (SGK1) regulates dendritic spine morphology in the prefrontal cortex, and modulation of SGK1 expression in mouse hippocampus regulates learning. However, the role of SGK1 in dendritic spine morphology within the CA1 and dentate gyrus regions of the hippocampus are unknown. Thus, herpes simplex viral vectors expressing GFP and various SGK1 constructs, including wild type SGK1, a catalytically inactive version of SGK1 (K127Q), and a phospho-defective version of SGK1 (S78A), were infused into the hippocampus of adult mice and confocal fluorescent microscopy was used to visualize dendritic spines. We show that increasing expression of SGK1 in the dentate gyrus increased the total number of spines, driven primarily by an increase in mushroom spines, while decreasing SGK1 activity (K127Q) in the CA1 region increased the total number of dendritic spines, driven by a significant increase in mushroom and stubby spines. The differential effects of SGK1 in these regions may be mediated by the interactions of SGK1 with multiple pathways required for spine formation and stability. As the formation of mature synapses is a crucial component of learning and memory, this indicates that SGK1 is a potential target in the pathway underlying stress-associated changes in cognition and memory.


Subject(s)
Dendritic Spines/metabolism , Hippocampus/metabolism , Immediate-Early Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Animals , Dendritic Spines/chemistry , Enzyme Activation/physiology , Genetic Vectors/administration & dosage , Genetic Vectors/analysis , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/analysis , Hippocampus/chemistry , Immediate-Early Proteins/analysis , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/analysis
4.
Nat Commun ; 11(1): 570, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31996679

ABSTRACT

Localization of the N-methyl-D-aspartate type glutamate receptor (NMDAR) to dendritic spines is essential for excitatory synaptic transmission and plasticity. Rather than remaining trapped at synaptic sites, NMDA receptors undergo constant cycling into and out of the postsynaptic density. Receptor movement is constrained by protein-protein interactions with both the intracellular and extracellular domains of the NMDAR. The role of extracellular interactions on the mobility of the NMDAR is poorly understood. Here we demonstrate that the positive surface charge of the hinge region of the N-terminal domain in the GluN1 subunit of the NMDAR is required to maintain NMDARs at dendritic spine synapses and mediates the direct extracellular interaction with a negatively charged phospho-tyrosine on the receptor tyrosine kinase EphB2. Loss of the EphB-NMDAR interaction by either mutating GluN1 or knocking down endogenous EphB2 increases NMDAR mobility. These findings begin to define a mechanism for extracellular interactions mediated by charged domains.


Subject(s)
Dendritic Spines , Receptor, EphB2/chemistry , Receptor, EphB2/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Biophysics , Dendritic Spines/chemistry , Dendritic Spines/genetics , Dendritic Spines/metabolism , Glycosylation , HEK293 Cells , Humans , Ion Channels , Mice , Models, Molecular , Nervous System/chemistry , Nervous System/metabolism , Neurons/chemistry , Neurons/metabolism , Neurosciences , Protein Conformation , Protein Interaction Domains and Motifs , Receptor, EphB2/genetics , Tyrosine/chemistry , Tyrosine/metabolism
5.
J Neurosci ; 39(48): 9598-9610, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31628182

ABSTRACT

Activation of the membrane estrogen receptor G-protein-coupled estrogen receptor (GPER) in ovariectomized mice via the GPER agonist G-1 mimics the beneficial effects of 17ß-estradiol (E2) on hippocampal CA1 spine density and memory consolidation, yet the cell-signaling mechanisms mediating these effects remain unclear. The present study examined the role of actin polymerization and c-Jun N-terminal kinase (JNK) phosphorylation in mediating effects of dorsal hippocampally infused G-1 on CA1 dendritic spine density and consolidation of object recognition and spatial memories in ovariectomized mice. We first showed that object learning increased apical CA1 spine density in the dorsal hippocampus (DH) within 40 min. We then found that DH infusion of G-1 increased both CA1 spine density and phosphorylation of the actin polymerization regulator cofilin, suggesting that activation of GPER may increase spine morphogenesis through actin polymerization. As with memory consolidation in our previous work (Kim et al., 2016), effects of G-1 on CA1 spine density and cofilin phosphorylation depended on JNK phosphorylation in the DH. Also consistent with our previous findings, E2-induced cofilin phosphorylation was not dependent on GPER activation. Finally, we found that infusion of the actin polymerization inhibitor, latrunculin A, into the DH prevented G-1 from increasing apical CA1 spine density and enhancing both object recognition and spatial memory consolidation. Collectively, these data demonstrate that GPER-mediated hippocampal spinogenesis and memory consolidation depend on JNK and cofilin signaling, supporting a critical role for actin polymerization in the GPER-induced regulation of hippocampal function in female mice.SIGNIFICANCE STATEMENT Emerging evidence suggests that G-protein-coupled estrogen receptor (GPER) activation mimics effects of 17ß-estradiol on hippocampal memory consolidation. Unlike canonical estrogen receptors, GPER activation is associated with reduced cancer cell proliferation; thus, understanding the molecular mechanisms through which GPER regulates hippocampal function may provide new avenues for the development of drugs that provide the cognitive benefits of estrogens without harmful side effects. Here, we demonstrate that GPER increases CA1 dendritic spine density and hippocampal memory consolidation in a manner dependent on actin polymerization and c-Jun N-terminal kinase phosphorylation. These findings provide novel insights into the role of GPER in mediating hippocampal morphology and memory consolidation, and may suggest first steps toward new therapeutics that more safely and effectively reduce memory decline in menopausal women.


Subject(s)
Actins/metabolism , CA1 Region, Hippocampal/metabolism , Dendritic Spines/metabolism , Memory Consolidation/physiology , Polymerization , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Actins/analysis , Animals , CA1 Region, Hippocampal/chemistry , Dendritic Spines/chemistry , Female , Mice , Mice, Inbred C57BL , Receptors, Estrogen/analysis , Receptors, G-Protein-Coupled/analysis
6.
Curr Protoc Neurosci ; 88(1): e69, 2019 06.
Article in English | MEDLINE | ID: mdl-31216393

ABSTRACT

The Golgi-Cox method has been one of the most effective techniques for studying the morphology of neuronal dendrites and dendritic spines. However, the reliability and time-consuming process of Golgi-Cox staining have been major obstacles to the widespread application of this technique. To overcome these shortcomings and to promote this invaluable technique, we developed the FD Rapid GolgiStain™ Kit based on the principle of the methods described by Ramón-Moliner in 1970 and Glaser and Van der Loos in 1981. The kit significantly improves and simplifies the Golgi-Cox technique. This kit is reliable for visualizing morphological details of neurons, allowing for analysis of various parameters of dendritic morphology-such as dendritic length and branching pattern and dendritic spine number, shape, and size-in both animal and postmortem human brains. A 40-min instructional video for tissue freezing, cryosectioning, and staining is provided. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Dendritic Spines/chemistry , Golgi Apparatus/chemistry , Neurons/chemistry , Staining and Labeling/methods , Animals , Dendrites/chemistry , Dendrites/physiology , Dendritic Spines/physiology , Golgi Apparatus/physiology , Humans , Neurons/physiology , Silver Staining/methods
7.
Proc Natl Acad Sci U S A ; 116(19): 9586-9591, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31028150

ABSTRACT

Cells in the brain act as components of extended networks. Therefore, to understand neurobiological processes in a physiological context, it is essential to study them in vivo. Super-resolution microscopy has spatial resolution beyond the diffraction limit, thus promising to provide structural and functional insights that are not accessible with conventional microscopy. However, to apply it to in vivo brain imaging, we must address the challenges of 3D imaging in an optically heterogeneous tissue that is constantly in motion. We optimized image acquisition and reconstruction to combat sample motion and applied adaptive optics to correcting sample-induced optical aberrations in super-resolution structured illumination microscopy (SIM) in vivo. We imaged the brains of live zebrafish larvae and mice and observed the dynamics of dendrites and dendritic spines at nanoscale resolution.


Subject(s)
Brain/diagnostic imaging , Neuroimaging , Animals , Brain/anatomy & histology , Dendrites/chemistry , Dendritic Spines/chemistry , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Zebrafish
8.
Cereb Cortex ; 29(2): 505-516, 2019 02 01.
Article in English | MEDLINE | ID: mdl-29293918

ABSTRACT

SEC14 and Spectrin domain-1 (Sestd1) is a synapse protein that exhibits a striking shift from the presynaptic to postsynaptic space as neurons mature postnatally in the mouse hippocampus. Hippocampal pyramidal neurons from mice with global genetic deletion of Sestd1 have reduced dendrite arbors, spines, and excitatory synapses. Electrophysiologically this correlates with cell-autonomous reductions in both AMPA- and NMDA-excitatory postsynaptic currents in individual hippocampal neurons from which Sestd1 has been deleted in vivo. These neurodevelopmental and functional deficits are associated with increased activation of the Rho family GTPases Rac1 and RhoA. Co-immunoprecipitation and mass spectrometry reveal that the Breakpoint Cluster Region protein, a Rho GTPase activating protein (GAP), forms complexes with Sestd1 in brain tissue. This complements earlier findings that Sestd1 can also partner with other Rho family GAPs and guanine nucleotide exchange factors. Our findings demonstrate that Sestd1 is a developmentally dynamic synaptic regulator of Rho GTPases that contributes to dendrite and excitatory synapse formation within differentiating pyramidal neurons of the forebrain.


Subject(s)
Carrier Proteins/metabolism , Dendritic Spines/metabolism , Neuropeptides/metabolism , Prosencephalon/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Synapses/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carrier Proteins/analysis , Dendrites/chemistry , Dendrites/metabolism , Dendritic Spines/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurogenesis/physiology , Neuropeptides/analysis , Organ Culture Techniques , Prosencephalon/chemistry , Prosencephalon/growth & development , Proto-Oncogene Proteins c-bcr/analysis , Synapses/chemistry , rac1 GTP-Binding Protein/analysis
9.
Brain Stimul ; 11(4): 856-859, 2018.
Article in English | MEDLINE | ID: mdl-29674117

ABSTRACT

BACKGROUND: Electroconvulsive therapy (ECT) is a rapid and effective treatment for major depressive disorder. Chronic stress-induced depression causes dendrite atrophy and deficiencies in brain-derived neurotrophic factor (BDNF), which are reversed by anti-depressant drugs. Electroconvulsive seizures (ECS), an animal model of ECT, robustly increase BDNF expression and stimulate dendritic outgrowth. OBJECTIVE: The present study aims to understand cellular and molecular plasticity mechanisms contributing to the efficacy of ECS following chronic stress-induced depression. METHODS: We quantify Bdnf transcript levels and dendritic spine density and morphology on cortical pyramidal neurons in mice exposed to vehicle or corticosterone and receiving either Sham or ECS treatment. RESULTS: ECS rescues corticosterone-induced defects in spine morphology and elevates Bdnf exon 1 and exon 4-containing transcripts in cortex. CONCLUSIONS: Dendritic spine remodeling and induction of activity-induced BDNF in the cortex represent important cellular and molecular plasticity mechanisms underlying the efficacy of ECS for treatment of chronic stress-induced depression.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Dendritic Spines/metabolism , Depression/metabolism , Depression/therapy , Electroconvulsive Therapy/methods , Seizures/metabolism , Animals , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/genetics , Cerebral Cortex/chemistry , Cerebral Cortex/metabolism , Dendritic Spines/chemistry , Depression/genetics , Disease Models, Animal , Gene Expression , Male , Mice , Seizures/genetics
10.
Neuron ; 97(5): 1126-1136.e10, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29429935

ABSTRACT

Most synaptic excitatory connections are made on dendritic spines. But how the voltage in spines is modulated by its geometry remains unclear. To investigate the electrical properties of spines, we combine voltage imaging data with electro-diffusion modeling. We first present a temporal deconvolution procedure for the genetically encoded voltage sensor expressed in hippocampal cultured neurons and then use electro-diffusion theory to compute the electric field and the current-voltage conversion. We extract a range for the neck resistances of 〈R〉=100±35MΩ. When a significant current is injected in a spine, the neck resistance can be inversely proportional to its radius, but not to the radius square, as predicted by Ohm's law. We conclude that the postsynaptic voltage cannot only be modulated by changing the number of receptors, but also by the spine geometry. Thus, spine morphology could be a key component in determining synaptic transduction and plasticity.


Subject(s)
Dendritic Spines/physiology , Hippocampus/physiology , Models, Neurological , Pyramidal Cells/physiology , Synapses/physiology , Animals , Dendritic Spines/chemistry , Hippocampus/chemistry , Hippocampus/cytology , Pyramidal Cells/chemistry , Synapses/chemistry
11.
Neuron ; 97(5): 1110-1125.e14, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29478916

ABSTRACT

Compartmentalization of calcium-dependent plasticity allows for rapid actin remodeling in dendritic spines. However, molecular mechanisms for the spatio-temporal regulation of filamentous actin (F-actin) dynamics by spinous Ca2+-transients are still poorly defined. We show that the postsynaptic Ca2+ sensor caldendrin orchestrates nano-domain actin dynamics that are essential for actin remodeling in the early phase of long-term potentiation (LTP). Steep elevation in spinous [Ca2+]i disrupts an intramolecular interaction of caldendrin and allows cortactin binding. The fast on and slow off rate of this interaction keeps cortactin in an active conformation, and protects F-actin at the spine base against cofilin-induced severing. Caldendrin gene knockout results in higher synaptic actin turnover, altered nanoscale organization of spinous F-actin, defects in structural spine plasticity, LTP, and hippocampus-dependent learning. Collectively, the data indicate that caldendrin-cortactin directly couple [Ca2+]i to preserve a minimal F-actin pool that is required for actin remodeling in the early phase of LTP.


Subject(s)
Calcium Signaling/physiology , Calcium-Binding Proteins/deficiency , Dendritic Spines/metabolism , Long-Term Potentiation/physiology , Synaptic Potentials/physiology , Animals , COS Cells , Calcium-Binding Proteins/genetics , Cells, Cultured , Chlorocebus aethiops , Dendritic Spines/chemistry , Dendritic Spines/genetics , HEK293 Cells , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Rats , Rats, Wistar
12.
Adv Exp Med Biol ; 1006: 105-118, 2017.
Article in English | MEDLINE | ID: mdl-28865017

ABSTRACT

Developmental changes in the expression and localization of drebrin has been mainly analyzed in chick embryo and young rat by various anti-drebrin polyclonal and monoclonal antibodies. Immunoblot analysis demonstrated that the adult drebrin isoform (drebrin A) is restricted to neural tissues, while the embryonic drebrin isoforms (drebrin E1 and E2 in chicken and drebrin E in mammals) are found in a wide variety of tissues. In the developing brain, drebrin E (including chicken drebrin E2) is expressed in newly generated neurons. During neuronal migration, drebrin E is distributed ubiquitously within the neurons. Once drebrin A is expressed in the developing neuron, drebrin E is no longer present within the cell soma and accumulates in the growth cone of growing processes, resulting in the cessation of neuronal migration. The limited subcellular localization of drebrin A, which is possibly regulated by a drebrin A-specific mechanism, is likely to affect the localization of drebrin E. In the adult brain, drebrin is mainly localized in dendritic spines, but in some nuclei, drebrin can be detected in neuronal somata as well as dendritic spines. The fact that the developmental changes in drebrin expression highly correlate in time with the sensitive period of visual cortical plasticity in kittens suggests that synaptic plasticity depends on drebrin.


Subject(s)
Neuronal Plasticity , Neurons/metabolism , Neuropeptides/isolation & purification , Visual Cortex/diagnostic imaging , Animals , Cats , Chick Embryo , Chickens/metabolism , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Microscopy , Neurons/chemistry , Neurons/ultrastructure , Neuropeptides/biosynthesis , Neuropeptides/metabolism , Rats , Visual Cortex/chemistry , Visual Cortex/metabolism
13.
Mol Pharmacol ; 89(4): 413-24, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26769413

ABSTRACT

Angiotensin IV (Ang IV) and related peptide analogs, as well as nonpeptide inhibitors of insulin-regulated aminopeptidase (IRAP), have previously been shown to enhance memory and cognition in animal models. Furthermore, the endogenous IRAP substrates oxytocin and vasopressin are known to facilitate learning and memory. In this study, the two recently synthesized 13-membered macrocyclic competitive IRAP inhibitors HA08 and HA09, which were designed to mimic the N terminus of oxytocin and vasopressin, were assessed and compared based on their ability to bind to the IRAP active site, and alter dendritic spine density in rat hippocampal primary cultures. The binding modes of the IRAP inhibitors HA08, HA09, and of Ang IV in either the extended or γ-turn conformation at the C terminus to human IRAP were predicted by docking and molecular dynamics simulations. The binding free energies calculated with the linear interaction energy method, which are in excellent agreement with experimental data and simulations, have been used to explain the differences in activities of the IRAP inhibitors, both of which are structurally very similar, but differ only with regard to one stereogenic center. In addition, we show that HA08, which is 100-fold more potent than the epimer HA09, can enhance dendritic spine number and alter morphology, a process associated with memory facilitation. Therefore, HA08, one of the most potent IRAP inhibitors known today, may serve as a suitable starting point for medicinal chemistry programs aided by MD simulations aimed at discovering more drug-like cognitive enhancers acting via augmenting synaptic plasticity.


Subject(s)
Cystinyl Aminopeptidase/antagonists & inhibitors , Cystinyl Aminopeptidase/metabolism , Dendritic Spines/metabolism , Disulfides/metabolism , Macrocyclic Compounds/metabolism , Animals , Cells, Cultured , Crystallography , Cystinyl Aminopeptidase/analysis , Dendritic Spines/chemistry , Disulfides/pharmacology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Female , HEK293 Cells , Humans , Macrocyclic Compounds/pharmacology , Pregnancy , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
14.
Cell Rep ; 12(2): 326-34, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26146087

ABSTRACT

A diverse array of neuromodulators governs cellular function in the prefrontal cortex (PFC) via the activation of G-protein-coupled receptors (GPCRs). However, these functionally diverse signals are carried and amplified by a relatively small assortment of intracellular second messengers. Here, we examine whether two distinct Gαi-coupled neuromodulators (norepinephrine and GABA) act as redundant regulators of glutamatergic synaptic transmission. Our results reveal that, within single dendritic spines of layer 5 pyramidal neurons, alpha-2 adrenergic receptors (α2Rs) selectively inhibit excitatory transmission mediated by AMPA-type glutamate receptors, while type B GABA receptors (GABA(B)Rs) inhibit NMDA-type receptors. We show that both modulators act via the downregulation of cAMP and PKA. However, by restricting the lifetime of active Gαi, RGS4 promotes the independent control of these two distinct target proteins. Our findings highlight a mechanism by which neuromodulatory microdomains can be established in subcellular compartments such as dendritic spines.


Subject(s)
Dendritic Spines/metabolism , Prefrontal Cortex/metabolism , Receptors, Glutamate/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Spines/chemistry , Down-Regulation/drug effects , Excitatory Postsynaptic Potentials , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Norepinephrine/pharmacology , RGS Proteins/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/metabolism , Receptors, Glutamate/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/pharmacology
15.
J Biomed Opt ; 20(1): 010501, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25585023

ABSTRACT

We report a novel fluorescence imaging approach to imaging nonfluorescence-labeled biological tissue samples. The method was demonstrated by imaging neurons in Golgi-Cox-stained and epoxy-resin-embedded samples through the excitation of the background fluorescence of the specimens. The dark neurons stood out clearly against background fluorescence in the images, enabling the tracing of a single dendritic spine using both confocal and wide-field fluorescence microscopy. The results suggest that the reported fluorescence imaging method would provide an effective alternative solution to image nonfluorescence-labeled samples, and it allows tracing the dendritic spine structure of neurons.


Subject(s)
Dendritic Spines/chemistry , Fluorescent Dyes/chemistry , Microscopy, Fluorescence/methods , Pyramidal Cells/cytology , Animals , Male , Mice , Microscopy, Confocal , Pyramidal Cells/chemistry
16.
J Neurosci ; 34(50): 16698-712, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505322

ABSTRACT

Stoichiometric labeling of endogenous synaptic proteins for high-contrast live-cell imaging in brain tissue remains challenging. Here, we describe a conditional mouse genetic strategy termed endogenous labeling via exon duplication (ENABLED), which can be used to fluorescently label endogenous proteins with near ideal properties in all neurons, a sparse subset of neurons, or specific neuronal subtypes. We used this method to label the postsynaptic density protein PSD-95 with mVenus without overexpression side effects. We demonstrated that mVenus-tagged PSD-95 is functionally equivalent to wild-type PSD-95 and that PSD-95 is present in nearly all dendritic spines in CA1 neurons. Within spines, while PSD-95 exhibited low mobility under basal conditions, its levels could be regulated by chronic changes in neuronal activity. Notably, labeled PSD-95 also allowed us to visualize and unambiguously examine otherwise-unidentifiable excitatory shaft synapses in aspiny neurons, such as parvalbumin-positive interneurons and dopaminergic neurons. Our results demonstrate that the ENABLED strategy provides a valuable new approach to study the dynamics of endogenous synaptic proteins in vivo.


Subject(s)
Fluorescent Dyes/analysis , Guanylate Kinases/analysis , Membrane Proteins/analysis , Microscopy, Fluorescence, Multiphoton/methods , Animals , CA1 Region, Hippocampal/chemistry , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Disks Large Homolog 4 Protein , Fluorescent Dyes/metabolism , Guanylate Kinases/metabolism , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques
17.
Sci Rep ; 4: 6353, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25220605

ABSTRACT

Learning and memory depend on morphological and functional changes to neural spines. Non-muscle myosin 2b regulates actin dynamics downstream of long-term potentiation induction. However, the mechanism by which myosin 2b is regulated in the spine has not been fully elucidated. Here, we show that filamin A-interacting protein (FILIP) is involved in the control of neural spine morphology and is limitedly expressed in the brain. FILIP bound near the ATPase domain of non-muscle myosin heavy chain IIb, an essential component of myosin 2b, and modified the function of myosin 2b by interfering with its actin-binding activity. In addition, FILIP altered the subcellular distribution of myosin 2b in spines. Moreover, subunits of the NMDA receptor were differently distributed in FILIP-expressing neurons, and excitation propagation was altered in FILIP-knockout mice. These results indicate that FILIP is a novel, region-specific modulator of myosin 2b.


Subject(s)
Carrier Proteins/physiology , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIB/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Actins/metabolism , Animals , Blotting, Western , Cells, Cultured , Immunoenzyme Techniques , Immunoprecipitation , Long-Term Potentiation , Mice , Mice, Inbred ICR , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Protein Binding , Rats
18.
J Neurosci ; 34(30): 9891-904, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25057192

ABSTRACT

Spinocerebellar ataxia type 5 (SCA5), a dominant neurodegenerative disease characterized by profound Purkinje cell loss, is caused by mutations in SPTBN2, a gene that encodes ß-III spectrin. SCA5 is the first neurodegenerative disorder reported to be caused by mutations in a cytoskeletal spectrin gene. We have developed a mouse model to understand the mechanistic basis for this disease and show that expression of mutant but not wild-type ß-III spectrin causes progressive motor deficits and cerebellar degeneration. We show that endogenous ß-III spectrin interacts with the metabotropic glutamate receptor 1α (mGluR1α) and that mice expressing mutant ß-III spectrin have cerebellar dysfunction with altered mGluR1α localization at Purkinje cell dendritic spines, decreased mGluR1-mediated responses, and deficient mGluR1-mediated long-term potentiation. These results indicate that mutant ß-III spectrin causes mislocalization and dysfunction of mGluR1α at dendritic spines and connects SCA5 with other disorders involving glutamatergic dysfunction and synaptic plasticity abnormalities.


Subject(s)
Disease Models, Animal , Mutation/genetics , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/genetics , Spectrin/genetics , Spinocerebellar Ataxias/genetics , Animals , Cerebellum/chemistry , Cerebellum/pathology , Dendritic Spines/chemistry , Dendritic Spines/pathology , Female , Humans , Male , Mice , Mice, Transgenic , Receptors, Metabotropic Glutamate/metabolism , Spinocerebellar Ataxias/physiopathology
19.
J Vis Exp ; (87)2014 May 12.
Article in English | MEDLINE | ID: mdl-24894563

ABSTRACT

In the mammalian cortex, neurons form extremely complicated networks and exchange information at synapses. Changes in synaptic strength, as well as addition/removal of synapses, occur in an experience-dependent manner, providing the structural foundation of neuronal plasticity. As postsynaptic components of the most excitatory synapses in the cortex, dendritic spines are considered to be a good proxy of synapses. Taking advantages of mouse genetics and fluorescent labeling techniques, individual neurons and their synaptic structures can be labeled in the intact brain. Here we introduce a transcranial imaging protocol using two-photon laser scanning microscopy to follow fluorescently labeled postsynaptic dendritic spines over time in vivo. This protocol utilizes a thinned-skull preparation, which keeps the skull intact and avoids inflammatory effects caused by exposure of the meninges and the cortex. Therefore, images can be acquired immediately after surgery is performed. The experimental procedure can be performed repetitively over various time intervals ranging from hours to years. The application of this preparation can also be expanded to investigate different cortical regions and layers, as well as other cell types, under physiological and pathological conditions.


Subject(s)
Dendritic Spines/physiology , Microscopy, Fluorescence, Multiphoton/methods , Animals , Cerebral Cortex/ultrastructure , Dendritic Spines/chemistry , Mice , Microscopy, Fluorescence, Multiphoton/instrumentation , Skull/surgery , Synapses/chemistry
20.
Biochim Biophys Acta ; 1838(10): 2391-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24915021

ABSTRACT

The development of dendritic spines with specific geometry and membrane composition is critical for proper synaptic function. Specific spine membrane architecture, sub-spine microdomains and spine head and neck geometry allow for well-coordinated and compartmentalized signaling, disruption of which could lead to various neurological diseases. Research from neuronal cell culture, brain slices and direct in vivo imaging indicates that dendritic spine development is a dynamic process which includes transition from small dendritic filopodia through a series of structural refinements to elaborate spines of various morphologies. Despite intensive research, the precise coordination of this morphological transition, the changes in molecular composition, and the relation of spines of various morphologies to function remain a central enigma in the development of functional neuronal circuits. Here, we review research so far and aim to provide insight into the key events that drive structural change during transition from immature filopodia to fully functional spines and the relevance of spine geometry to function.


Subject(s)
Dendritic Spines , Membrane Microdomains , Nervous System Diseases , Pseudopodia , Signal Transduction , Animals , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Pseudopodia/chemistry , Pseudopodia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...