Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 153(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34581724

ABSTRACT

A primary goal of molecular physiology is to understand how conformational changes of proteins affect the function of cells, tissues, and organisms. Here, we describe an imaging method for measuring the conformational changes of the voltage sensors of endogenous ion channel proteins within live tissue, without genetic modification. We synthesized GxTX-594, a variant of the peptidyl tarantula toxin guangxitoxin-1E, conjugated to a fluorophore optimal for two-photon excitation imaging through light-scattering tissue. We term this tool EVAP (Endogenous Voltage-sensor Activity Probe). GxTX-594 targets the voltage sensors of Kv2 proteins, which form potassium channels and plasma membrane-endoplasmic reticulum junctions. GxTX-594 dynamically labels Kv2 proteins on cell surfaces in response to voltage stimulation. To interpret dynamic changes in fluorescence intensity, we developed a statistical thermodynamic model that relates the conformational changes of Kv2 voltage sensors to degree of labeling. We used two-photon excitation imaging of rat brain slices to image Kv2 proteins in neurons. We found puncta of GxTX-594 on hippocampal CA1 neurons that responded to voltage stimulation and retain a voltage response roughly similar to heterologously expressed Kv2.1 protein. Our findings show that EVAP imaging methods enable the identification of conformational changes of endogenous Kv2 voltage sensors in tissue.


Subject(s)
Shab Potassium Channels , Spider Venoms , Animals , Carrier Proteins , Hippocampus/metabolism , Neurons/metabolism , Rats , Shab Potassium Channels/metabolism
2.
Front Neuroanat ; 15: 627368, 2021.
Article in English | MEDLINE | ID: mdl-34135737

ABSTRACT

Electron microscopy (EM)-based synaptology is a fundamental discipline for achieving a complex wiring diagram of the brain. A quantitative understanding of synaptic ultrastructure also serves as a basis to estimate the relative magnitude of synaptic transmission across individual circuits in the brain. Although conventional light microscopic techniques have substantially contributed to our ever-increasing understanding of the morphological characteristics of the putative synaptic junctions, EM is the gold standard for systematic visualization of the synaptic morphology. Furthermore, a complete three-dimensional reconstruction of an individual synaptic profile is required for the precise quantitation of different parameters that shape synaptic transmission. While volumetric imaging of synapses can be routinely obtained from the transmission EM (TEM) imaging of ultrathin sections, it requires an unimaginable amount of effort and time to reconstruct very long segments of dendrites and their spines from the serial section TEM images. The challenges of low throughput EM imaging have been addressed to an appreciable degree by the development of automated EM imaging tools that allow imaging and reconstruction of dendritic segments in a realistic time frame. Here, we review studies that have been instrumental in determining the three-dimensional ultrastructure of synapses. With a particular focus on dendritic spine synapses in the rodent brain, we discuss various key studies that have highlighted the structural diversity of spines, the principles of their organization in the dendrites, their presynaptic wiring patterns, and their activity-dependent structural remodeling.

3.
eNeuro ; 7(6)2020.
Article in English | MEDLINE | ID: mdl-33109633

ABSTRACT

Precise information on synapse organization in a dendrite is crucial to understanding the mechanisms underlying voltage integration and the variability in the strength of synaptic inputs across dendrites of different complex morphologies. Here, we used focused ion beam/scanning electron microscope (FIB/SEM) to image the dendritic spines of mice in the hippocampal CA1 region, CA3 region, somatosensory cortex, striatum, and cerebellum (CB). Our results show that the spine geometry and dimensions differ across neuronal cell types. Despite this difference, dendritic spines were organized in an orchestrated manner such that the postsynaptic density (PSD) area per unit length of dendrite scaled positively with the dendritic diameter in CA1 proximal stratum radiatum (PSR), cortex, and CB. The ratio of the PSD area to neck length was kept relatively uniform across dendrites of different diameters in CA1 PSR. Computer simulation suggests that a similar level of synaptic strength across different dendrites in CA1 PSR enables the effective transfer of synaptic inputs from the dendrites toward soma. Excitatory postsynaptic potentials (EPSPs), evoked at single spines by glutamate uncaging and recorded at the soma, show that the neck length is more influential than head width in regulating the EPSP magnitude at the soma. Our study describes thorough morphologic features and the organizational principles of dendritic spines in different brain regions.


Subject(s)
Dendrites , Synapses , Animals , Computer Simulation , Excitatory Postsynaptic Potentials , Mice , Neurons
4.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32817196

ABSTRACT

The aging process is accompanied by various neurophysiological changes, and the severity of neurodegenerative disorders such as Parkinson's disease (PD) increases with aging. However, the precise neuroanatomical changes that accompany the aging process in both normal and pathologic conditions remain unknown. This is in part because there is a lack of high-resolution imaging tool that has the capacity to image a desired volume of neurons in a high-throughput and automated manner. In the present study, focused ion beam/scanning electron microscopy (FIB/SEM) was used to image striatal neuropil in both wild-type (WT) mice and an A53T bacterial artificial chromosome (BAC) human α-synuclein (A53T-BAC-SNCA) transgenic (Tg) mouse model of PD, at 1, 3, 6, and 22 months of age. We demonstrated that spine density gradually decreases, and average spine head volume gradually increases with age in WT mice, suggesting a homeostatic balance between spine head volume and spine density. However, this inverse relationship between spine head volume and spine density was not observed in A53T-BAC-SNCA Tg mice. Taken together, our data suggest that PD is accompanied by an abnormality in the mechanisms that control synapse growth and maturity.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Corpus Striatum/metabolism , Dendritic Spines/metabolism , Mice , Mice, Transgenic , Parkinson Disease/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
5.
Biochem Biophys Res Commun ; 530(1): 130-135, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828274

ABSTRACT

Neurons in the central nervous system display a great diversity of synaptic architecture. While much of our knowledge on the excitatory synapse morphology derives from the prototypical asymmetric synapses, little has been studied about the atypical crest-type synapse that exists in the restricted brain regions. Here, we used focused ion beam scanning electron microscopy (FIB/SEM) to image a neuropil volume of interpeduncular nucleus (IPN) and manually reconstructed several dendrites to obtain an insight about the topography and quantitative features of crest synapses. Three-dimensional reconstruction showed numerous U-shaped structures protruding from the IPN dendrites. On either faces of the U-shaped structure, a pair of crest synapses are aligned in parallel such that there exists a positive correlation between the postsynaptic density (PSD) area of synapses that participate in pair formation. Interestingly, mitochondria are excluded from the site of crest synapses. Several presynaptic axons run through the hollow, cylindrical space of the U-shape grooves such that the plasma membrane of the axon and the dendrite are organized in a tight opposition without any intervening glial membrane. Unlike the peculiar dendritic morphology, IPN neurons possess typical somatic morphology with an oval, centrally located nucleus. In conclusion, our data reveals a hitherto unknown unique topographical feature of crest synapses in the IPN.


Subject(s)
Interpeduncular Nucleus/ultrastructure , Synapses/ultrastructure , Animals , Axons/ultrastructure , Dendrites/ultrastructure , Mice, Inbred C57BL , Microscopy, Electron, Scanning
6.
Brain ; 143(1): 249-265, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31816026

ABSTRACT

Parkinson's disease is one of the most common movement disorders and is characterized by dopaminergic cell loss and the accumulation of pathological α-synuclein, but its precise pathogenetic mechanisms remain elusive. To develop disease-modifying therapies for Parkinson's disease, an animal model that recapitulates the pathology and symptoms of the disease, especially in the prodromal stage, is indispensable. As subjects with α-synuclein gene (SNCA) multiplication as well as point mutations develop familial Parkinson's disease and a genome-wide association study in Parkinson's disease has identified SNCA as a risk gene for Parkinson's disease, the increased expression of α-synuclein is closely associated with the aetiology of Parkinson's disease. In this study we generated bacterial artificial chromosome transgenic mice harbouring SNCA and its gene expression regulatory regions in order to maintain the native expression pattern of α-synuclein. Furthermore, to enhance the pathological properties of α-synuclein, we inserted into SNCA an A53T mutation, two single-nucleotide polymorphisms identified in a genome-wide association study in Parkinson's disease and a Rep1 polymorphism, all of which are causal of familial Parkinson's disease or increase the risk of sporadic Parkinson's disease. These A53T SNCA bacterial artificial chromosome transgenic mice showed an expression pattern of human α-synuclein very similar to that of endogenous mouse α-synuclein. They expressed truncated, oligomeric and proteinase K-resistant phosphorylated forms of α-synuclein in the regions that are specifically affected in Parkinson's disease and/or dementia with Lewy bodies, including the olfactory bulb, cerebral cortex, striatum and substantia nigra. Surprisingly, these mice exhibited rapid eye movement (REM) sleep without atonia, which is a key feature of REM sleep behaviour disorder, at as early as 5 months of age. Consistent with this observation, the REM sleep-regulating neuronal populations in the lower brainstem, including the sublaterodorsal tegmental nucleus, nuclei in the ventromedial medullary reticular formation and the pedunculopontine nuclei, expressed phosphorylated α-synuclein. In addition, they also showed hyposmia at 9 months of age, which is consistent with the significant accumulation of phosphorylated α-synuclein in the olfactory bulb. The dopaminergic neurons in the substantia nigra pars compacta degenerated, and their number was decreased in an age-dependent manner by up to 17.1% at 18 months of age compared to wild-type, although the mice did not show any related locomotor dysfunction. In conclusion, we created a novel mouse model of prodromal Parkinson's disease that showed RBD-like behaviour and hyposmia without motor symptoms.


Subject(s)
Brain/metabolism , Disease Models, Animal , Mice , Olfaction Disorders/genetics , Parkinson Disease/genetics , Prodromal Symptoms , REM Sleep Behavior Disorder/genetics , alpha-Synuclein/genetics , Animals , Cell Count , Chromosomes, Artificial, Bacterial , Electroencephalography , Electromyography , Endopeptidase K/metabolism , Mice, Transgenic , Olfaction Disorders/physiopathology , Parkinson Disease/physiopathology , Polymorphism, Single Nucleotide , REM Sleep Behavior Disorder/physiopathology , Sleep , alpha-Synuclein/metabolism
7.
eNeuro ; 6(3)2019.
Article in English | MEDLINE | ID: mdl-31118204

ABSTRACT

The Golgi apparatus plays an indispensable role in posttranslational modification and transport of proteins to their target destinations. Although it is well established that the Golgi apparatus requires an acidic luminal pH for optimal activity, morphological and functional abnormalities at the neuronal circuit level because of perturbations in Golgi pH are not fully understood. In addition, morphological alteration of the Golgi apparatus is associated with several neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. Here, we used anatomical and electrophysiological approaches to characterize morphological and functional abnormalities of neuronal circuits in Golgi pH regulator (GPHR) conditional knock-out mice. Purkinje cells (PCs) from the mutant mice exhibited vesiculation and fragmentation of the Golgi apparatus, followed by axonal degeneration and progressive cell loss. Morphological analysis provided evidence for the disruption of basket cell (BC) terminals around PC soma, and electrophysiological recordings showed selective loss of large amplitude responses, suggesting BC terminal disassembly. In addition, the innervation of mutant PCs was altered such that climbing fiber (CF) terminals abnormally synapsed on the somatic spines of mutant PCs in the mature cerebellum. The combined results describe an essential role for luminal acidification of the Golgi apparatus in maintaining proper neuronal morphology and neuronal circuitry.


Subject(s)
Cerebellum/metabolism , Cerebellum/ultrastructure , Golgi Apparatus/ultrastructure , Neuronal Plasticity , Neurons/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Animals , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Disease Models, Animal , Female , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Male , Mice, Knockout , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Neurons/metabolism , Primary Cell Culture , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure
8.
Methods Mol Biol ; 1941: 17-27, 2019.
Article in English | MEDLINE | ID: mdl-30707424

ABSTRACT

A thorough understanding of the synaptic ultrastructure is necessary to bridge our current knowledge gap about the relationship between neuronal structure and function. Recent development of focused ion beam scanning electron microscopy (FIB/SEM) has made it possible to image neuronal structures with high speed and efficiency. Here, we present our routine protocol for correlative two-photon microscopy and FIB/SEM imaging of glutamatergic synapses. Femtosecond-pulsed near-infrared laser was used to create fiducial marks around the dendrite of interest in aldehyde-fixed tissues. Thereafter, samples were subjected to en bloc staining with rOTO (reduced osmium tetroxide-thiocarbohydrazide-osmium tetroxide), followed by lead aspartate and uranyl acetate to enhance tissue contrast. Reliable detection of postsynaptic density (PSD) and plasma membrane contours by the sample preparation protocol optimized for FIB/SEM allows us to precisely evaluate morphological features that shape glutamatergic synaptic transmission.


Subject(s)
Dendritic Spines/ultrastructure , Glutamic Acid/metabolism , Microscopy, Electron, Scanning/methods , Receptors, Glutamate/metabolism , Synapses/ultrastructure , Animals , Dendritic Spines/metabolism , Synapses/metabolism , Tissue Fixation/methods
9.
Hum Mol Genet ; 28(11): 1894-1904, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30689867

ABSTRACT

Parkinson's disease (PD) is characterized by dopaminergic (DA) cell loss and the accumulation of pathological alpha synuclein (asyn), but its precise pathomechanism remains unclear, and no appropriate animal model has yet been established. Recent studies have shown that a heterozygous mutation of glucocerebrosidase (gba) is one of the most important genetic risk factors in PD. To create mouse model for PD, we crossed asyn Bacterial Artificial Chromosome transgenic mice with gba heterozygous knockout mice. These double-mutant (dm) mice express human asyn in a physiological manner through its native promoter and showed an increase in phosphorylated asyn in the regions vulnerable to PD, such as the olfactory bulb and dorsal motor nucleus of the vagus nerve. Only dm mice showed a significant reduction in DA cells in the substantia nigra pars compacta, suggesting these animals were suitable for a prodromal model of PD. Next, we investigated the in vivo mechanism by which GBA insufficiency accelerates PD pathology, focusing on lipid metabolism. Dm mice showed an increased level of glucosylsphingosine without any noticeable accumulation of glucosylceramide, a direct substrate of GBA. In addition, the overexpression of asyn resulted in decreased GBA activity in mice, while dm mice tended to show an even further decreased level of GBA activity. In conclusion, we created a novel prodromal mouse model to study the disease pathogenesis and develop novel therapeutics for PD and also revealed the mechanism by which heterozygous gba deficiency contributes to PD through abnormal lipid metabolism under conditions of an altered asyn expression in vivo.


Subject(s)
Glucosylceramidase/genetics , Parkinson Disease/genetics , alpha-Synuclein/genetics , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Lipid Metabolism/genetics , Mice , Mice, Knockout , Mice, Transgenic , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pars Compacta/metabolism , Pars Compacta/pathology , Prodromal Symptoms
10.
Science ; 359(6376): 679-684, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29439241

ABSTRACT

Optogenetics has revolutionized the experimental interrogation of neural circuits and holds promise for the treatment of neurological disorders. It is limited, however, because visible light cannot penetrate deep inside brain tissue. Upconversion nanoparticles (UCNPs) absorb tissue-penetrating near-infrared (NIR) light and emit wavelength-specific visible light. Here, we demonstrate that molecularly tailored UCNPs can serve as optogenetic actuators of transcranial NIR light to stimulate deep brain neurons. Transcranial NIR UCNP-mediated optogenetics evoked dopamine release from genetically tagged neurons in the ventral tegmental area, induced brain oscillations through activation of inhibitory neurons in the medial septum, silenced seizure by inhibition of hippocampal excitatory cells, and triggered memory recall. UCNP technology will enable less-invasive optical neuronal activity manipulation with the potential for remote therapy.


Subject(s)
Brain/physiology , Deep Brain Stimulation/methods , Nanoparticles , Neurons/physiology , Optogenetics/methods , Animals , Light , Mice , Mice, Transgenic
11.
Front Mol Neurosci ; 11: 1, 2018.
Article in English | MEDLINE | ID: mdl-29403353

ABSTRACT

Voltage-gated K+ (Kv) channels play important roles in regulating neuronal excitability. Kv channels comprise four principal α subunits, and transmembrane and/or cytoplasmic auxiliary subunits that modify diverse aspects of channel function. AMIGO-1, which mediates homophilic cell adhesion underlying neurite outgrowth and fasciculation during development, has recently been shown to be an auxiliary subunit of adult brain Kv2.1-containing Kv channels. We show that AMIGO-1 is extensively colocalized with both Kv2.1 and its paralog Kv2.2 in brain neurons across diverse mammals, and that in adult brain, there is no apparent population of AMIGO-1 outside of that colocalized with these Kv2 α subunits. AMIGO-1 is coclustered with Kv2 α subunits at specific plasma membrane (PM) sites associated with hypolemmal subsurface cisternae at neuronal ER:PM junctions. This distinct PM clustering of AMIGO-1 is not observed in brain neurons of mice lacking Kv2 α subunit expression. Moreover, in heterologous cells, coexpression of either Kv2.1 or Kv2.2 is sufficient to drive clustering of the otherwise uniformly expressed AMIGO-1. Kv2 α subunit coexpression also increases biosynthetic intracellular trafficking and PM expression of AMIGO-1 in heterologous cells, and analyses of Kv2.1 and Kv2.2 knockout mice show selective loss of AMIGO-1 expression and localization in neurons lacking the respective Kv2 α subunit. Together, these data suggest that in mammalian brain neurons, AMIGO-1 is exclusively associated with Kv2 α subunits, and that Kv2 α subunits are obligatory in determining the correct pattern of AMIGO-1 expression, PM trafficking and clustering.

12.
Parasitol Res ; 117(1): 287-293, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29218443

ABSTRACT

This study aimed to determine the prevalence of intestinal parasites and its associated risk factors among school-going children in Kathmandu, Nepal. Between August and September 2016, a total of 333 stool samples were collected from children at five public schools. The collected samples were subjected to formol-ether concentration, followed by conventional microscopic examination for intestinal parasites. The overall prevalence of intestinal parasites was 24.3% (81/333), with Giardia spp. showing the highest prevalence of 18.9% (63/333). Samples positive for Giardia spp. by microscopy were further subjected to quantitative polymerase chain reaction (qPCR) for G. duodenalis, resulting in a positive ratio of 100%. The positive ratio of Giardia spp. was considerably high among children consuming tanker water (27.3%), jar water (21.0%), and tap water (17.5%). Our results demonstrated that G. duodenalis remains predominant in school-going children in Nepal.


Subject(s)
Giardia lamblia/isolation & purification , Giardiasis/epidemiology , Intestinal Diseases, Parasitic/epidemiology , Adolescent , Child , Feces/parasitology , Female , Giardia lamblia/genetics , Giardiasis/parasitology , Humans , Intestinal Diseases, Parasitic/parasitology , Male , Microscopy , Nepal/epidemiology , Prevalence , Risk Factors , Schools
13.
Brain Res Bull ; 129: 3-11, 2017 03.
Article in English | MEDLINE | ID: mdl-27491624

ABSTRACT

Principal neurons in multiple brain regions receive a vast majority of excitatory synaptic contacts on the tiny dendritic appendages called dendritic spines. These structures are believed to be the locus of memory storage in the brain. Indeed, neurological diseases leading to impairment in memory and cognitive capabilities are often associated with structural alteration of dendritic spines. While several landmark studies in the past have provided a great deal of information on the structure, function and molecular composition of prototypical mature dendritic spines, we still have a limited knowledge of nascent spines. In recent years there has been a surge of interest to understand the nascent spines and the increasing technical advances in the genetic, molecular and imaging methods have opened avenues for systematic and thorough investigation. In this review, by discussing studies from several labs including ours, we provide a systematic summary of the development, structure, molecular expression and function of nascent spines and highlight some of the potentially important and interesting research questions that remain to be answered.


Subject(s)
Dendritic Spines/physiology , Animals , Dendritic Spines/ultrastructure
14.
J Neurosci ; 35(44): 14922-42, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538660

ABSTRACT

The Kv2 family of voltage-gated potassium channel α subunits, comprising Kv2.1 and Kv2.2, mediate the bulk of the neuronal delayed rectifier K(+) current in many mammalian central neurons. Kv2.1 exhibits robust expression across many neuron types and is unique in its conditional role in modulating intrinsic excitability through changes in its phosphorylation state, which affect Kv2.1 expression, localization, and function. Much less is known of the highly related Kv2.2 subunit, especially in forebrain neurons. Here, through combined use of cortical layer markers and transgenic mouse lines, we show that Kv2.1 and Kv2.2 are localized to functionally distinct cortical cell types. Kv2.1 expression is consistently high throughout all cortical layers, especially in layer (L) 5b pyramidal neurons, whereas Kv2.2 expression is primarily limited to neurons in L2 and L5a. In addition, L4 of primary somatosensory cortex is strikingly devoid of Kv2.2 immunolabeling. The restricted pattern of Kv2.2 expression persists in Kv2.1-KO mice, suggesting distinct cell- and layer-specific functions for these two highly related Kv2 subunits. Analyses of endogenous Kv2.2 in cortical neurons in situ and recombinant Kv2.2 expressed in heterologous cells reveal that Kv2.2 is largely refractory to stimuli that trigger robust, phosphorylation-dependent changes in Kv2.1 clustering and function. Immunocytochemistry and voltage-clamp recordings from outside-out macropatches reveal distinct cellular expression patterns for Kv2.1 and Kv2.2 in intratelencephalic and pyramidal tract neurons of L5, indicating circuit-specific requirements for these Kv2 paralogs. Together, these results support distinct roles for these two Kv2 channel family members in mammalian cortex. SIGNIFICANCE STATEMENT: Neurons within the neocortex are arranged in a laminar architecture and contribute to the input, processing, and/or output of sensory and motor signals in a cell- and layer-specific manner. Neurons of different cortical layers express diverse populations of ion channels and possess distinct intrinsic membrane properties. Here, we show that the Kv2 family members Kv2.1 and Kv2.2 are expressed in distinct cortical layers and pyramidal cell types associated with specific corticostriatal pathways. We find that Kv2.1 and Kv2.2 exhibit distinct responses to acute phosphorylation-dependent regulation in brain neurons in situ and in heterologous cells in vitro. These results identify a molecular mechanism that contributes to heterogeneity in cortical neuron ion channel function and regulation.


Subject(s)
Neocortex/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Shab Potassium Channels/biosynthesis , Animals , Cells, Cultured , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/cytology , Organ Culture Techniques , Pyramidal Cells/cytology , Rats , Rats, Sprague-Dawley
15.
Cell Rep ; 10(2): 162-9, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25558061

ABSTRACT

Competition between synapses contributes to activity-dependent refinement of the nervous system during development. Does local competition between neighboring synapses drive circuit remodeling during experience-dependent plasticity in the cerebral cortex? Here, we examined the role of activity-mediated competitive interactions in regulating dendritic spine structure and function on hippocampal CA1 neurons. We found that high-frequency glutamatergic stimulation at individual spines, which leads to input-specific synaptic potentiation, induces shrinkage and weakening of nearby unstimulated synapses. This heterosynaptic plasticity requires potentiation of multiple neighboring spines, suggesting that a local threshold of neural activity exists beyond which inactive synapses are punished. Notably, inhibition of calcineurin, IP3Rs, or group I metabotropic glutamate receptors (mGluRs) blocked heterosynaptic shrinkage without blocking structural potentiation, and inhibition of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) blocked structural potentiation without blocking heterosynaptic shrinkage. Our results support a model in which activity-induced shrinkage signal, and not competition for limited structural resources, drives heterosynaptic structural and functional depression during neural circuit refinement.


Subject(s)
CA1 Region, Hippocampal/physiology , Dendrites/physiology , Synapses/physiology , Animals , Calcineurin/chemistry , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neuronal Plasticity , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism
16.
J Comp Neurol ; 522(15): 3555-74, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24962901

ABSTRACT

The Kv2.1 voltage-gated K+ channel is widely expressed throughout mammalian brain, where it contributes to dynamic activity-dependent regulation of intrinsic neuronal excitability. Here we show that somatic plasma membrane Kv2.1 clusters are juxtaposed to clusters of intracellular ryanodine receptor (RyR) Ca2+ -release channels in mouse brain neurons, most prominently in medium spiny neurons (MSNs) of the striatum. Electron microscopy-immunogold labeling shows that in MSNs, plasma membrane Kv2.1 clusters are adjacent to subsurface cisternae, placing Kv2.1 in close proximity to sites of RyR-mediated Ca2+ release. Immunofluorescence labeling in transgenic mice expressing green fluorescent protein in specific MSN populations reveals the most prominent juxtaposed Kv2.1:RyR clusters in indirect pathway MSNs. Kv2.1 in both direct and indirect pathway MSNs exhibits markedly lower levels of labeling with phosphospecific antibodies directed against the S453, S563, and S603 phosphorylation site compared with levels observed in neocortical neurons, although labeling for Kv2.1 phosphorylation at S563 was significantly lower in indirect pathway MSNs compared with those in the direct pathway. Finally, acute stimulation of RyRs in heterologous cells causes a rapid hyperpolarizing shift in the voltage dependence of activation of Kv2.1, typical of Ca2+ /calcineurin-dependent Kv2.1 dephosphorylation. Together, these studies reveal that striatal MSNs are distinct in their expression of clustered Kv2.1 at plasma membrane sites juxtaposed to intracellular RyRs, as well as in Kv2.1 phosphorylation state. Differences in Kv2.1 expression and phosphorylation between MSNs in direct and indirect pathways provide a cell- and circuit-specific mechanism for coupling intracellular Ca2+ release to phosphorylation-dependent regulation of Kv2.1 to dynamically impact intrinsic excitability.


Subject(s)
Brain/cytology , Brain/physiology , Neurons/cytology , Neurons/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Shab Potassium Channels/metabolism , Animals , Brain/ultrastructure , Cell Membrane/physiology , Cell Membrane/ultrastructure , Female , HEK293 Cells , Humans , Membrane Potentials/physiology , Mice, Knockout , Mice, Transgenic , Neurons/ultrastructure , Phosphorylation , Ryanodine Receptor Calcium Release Channel/genetics , Shab Potassium Channels/genetics
17.
J Neurosci ; 32(39): 13555-67, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-23015445

ABSTRACT

R-type calcium channels (RTCCs) are well known for their role in synaptic plasticity, but little is known about their subcellular distribution across various neuronal compartments. Using subtype-specific antibodies, we characterized the regional and subcellular localization of Ca(v)2.3 in mice and rats at both light and electron microscopic levels. Ca(v)2.3 immunogold particles were found to be predominantly presynaptic in the interpeduncular nucleus, but postsynaptic in other brain regions. Serial section analysis of electron microscopic images from the hippocampal CA1 revealed a higher density of immunogold particles in the dendritic shaft plasma membrane compared with the pyramidal cell somata. However, the labeling densities were not significantly different among the apical, oblique, or basal dendrites. Immunogold particles were also observed over the plasma membrane of dendritic spines, including both synaptic and extrasynaptic sites. Individual spine heads contained <20 immunogold particles, with an average density of ∼260 immunoparticles per µm(3) spine head volume, in accordance with the density of RTCCs estimated using calcium imaging (Sabatini and Svoboda, 2000). The Ca(v)2.3 density was variable among similar-sized spine heads and did not correlate with the density in the parent dendrite, implying that spines are individual calcium compartments operating autonomously from their parent dendrites.


Subject(s)
Calcium Channels, R-Type/metabolism , Calcium Channels, R-Type/ultrastructure , Cation Transport Proteins/metabolism , Cation Transport Proteins/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Calcium Channels, R-Type/chemistry , Calcium Channels, R-Type/deficiency , Cation Transport Proteins/chemistry , Cation Transport Proteins/deficiency , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Epitopes/metabolism , Female , Guinea Pigs , Imaging, Three-Dimensional , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Peptides/metabolism , Post-Synaptic Density/metabolism , Post-Synaptic Density/ultrastructure , Rats , Statistics as Topic , Statistics, Nonparametric , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
18.
J Comp Neurol ; 518(21): 4362-74, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20853512

ABSTRACT

T-type calcium channels play a pivotal role in regulating neural membrane excitability in the nervous system. However, the precise subcellular distributions of T-type channel subunits and their implication for membrane excitability are not well understood. Here we investigated the subcellular distribution of the α1G subunit of the calcium channel which is expressed highly in the mouse dorsal lateral geniculate nucleus (dLGN). Light microscopic analysis demonstrated that dLGN exhibits intense immunoperoxidase reactivity for the α1G subunit. Electron microscopic observation showed that the labeling was present in both the relay cells and interneurons and was found in the somatodendritic, but not axonal, domains of these cells. Most of the immunogold particles for the α1G subunit were either associated with the plasma membrane or the intracellular membranes. Reconstruction analysis of serial electron microscopic images revealed that the intensity of the intracellular labeling exhibited a gradient such that the labeling density was higher in the proximal dendrite and progressively decreased towards the distal dendrite. In contrast, the plasma membrane-associated particles were distributed with a uniform density over the somatodendritic surface of dLGN cells. The labeling density in the relay cell plasma membrane was about 3-fold higher than that of the interneurons. These results provide ultrastructural evidence for cell-type-specific expression levels and for uniform expression density of the α1G subunit over the plasma membrane of dLGN cells.


Subject(s)
Calcium Channels, T-Type/metabolism , Geniculate Bodies/metabolism , Protein Subunits/metabolism , Animals , Calcium Channels, T-Type/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Geniculate Bodies/cytology , Immunohistochemistry , Interneurons/metabolism , Interneurons/ultrastructure , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/genetics , Synapses/metabolism , Synapses/ultrastructure , Thalamus/cytology , Thalamus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...