Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Pharmacol Sci ; 144(2): 76-82, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32736867

ABSTRACT

Astrocytes, comprising the primary glial-cell type, are involved in the formation and maturation of synapses, and thus contribute to sustainable synaptic transmission between neurons. Given that the animals in higher phylogenetic tree have brains with a higher density of glial cells with respect to neurons, there is a possibility that the relative astrocytic density directly influences synaptic transmission. However, the notion has not been tested thoroughly. Here we addressed it, by using a primary culture preparation where single hippocampal neurons are surrounded by a variable but a countable number of cortical astrocytes in dot-patterned microislands, and recording synaptic transmission by patch-clamp electrophysiology. Neurons with a higher astrocytic density showed a higher amplitude of the evoked excitatory postsynaptic current than that of neurons with a lower astrocytic density. The size of the readily releasable pool of synaptic vesicles per neuron was significantly larger. The frequency of spontaneous synaptic transmission was higher, but the amplitude was unchanged. The number of morphologically identified glutamatergic synapses was comparable, but the percentage of functional ones was increased, indicating a lower ratio of presynaptically silent synapses. Taken together, the higher astrocytic density enhanced excitatory synaptic transmission by increasing the fraction of functional synapses through presynaptic un-silencing.


Subject(s)
Astrocytes/physiology , Brain/cytology , Neurons/physiology , Synapses/physiology , Synaptic Transmission , Animals , Astrocytes/pathology , Cells, Cultured , Excitatory Postsynaptic Potentials , Female , Mice, Inbred ICR , Neurons/pathology , Phylogeny , Pregnancy
2.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32699089

ABSTRACT

Herpes simplex virus (HSV) is a neuroinvasive virus that has been used as a model organism for studying common properties of all herpesviruses. HSV induces host organelle rearrangement and forms multiple, dispersed assembly compartments in epithelial cells, which complicates the study of HSV assembly. In this study, we show that HSV forms a visually distinct unitary cytoplasmic viral assembly center (cVAC) in both cancerous and primary neuronal cells that concentrates viral structural proteins and is a major site of capsid envelopment. The HSV cVAC also concentrates host membranes that are important for viral assembly, such as Golgi- and recycling endosome-derived membranes. Finally, we show that HSV cVAC formation and/or maintenance depends on an intact microtubule network and a viral tegument protein, pUL51. Our observations suggest that the neuronal cVAC is a uniquely useful model to study common herpesvirus assembly pathways and cell-specific pathways for membrane reorganization.IMPORTANCE Herpesvirus particles are complex and contain many different proteins that must come together in an organized and coordinated fashion. Many viruses solve this coordination problem by creating a specialized assembly factory in the host cell, and the formation of such factories provides a promising target for interfering with virus production. Herpes simplex virus 1 (HSV-1) infects several types of cells, including neurons, but has not previously been shown to form such an organized factory in the nonneuronal cells in which its assembly has been best studied. Here, we show that HSV-1 forms an organized assembly factory in neuronal cells, and we identify some of the viral and host cell factors that are important for its formation.


Subject(s)
Cell Membrane/physiology , Herpesvirus 1, Human/physiology , Neurons/virology , Viral Proteins/metabolism , Virus Assembly/physiology , Actin Cytoskeleton/metabolism , Animals , Capsid/metabolism , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoplasm/virology , Golgi Apparatus/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Vero Cells , Viral Structural Proteins/metabolism , Virion/metabolism
3.
Brain Res ; 1729: 146627, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31883849

ABSTRACT

The locus coeruleus (LC) is a nucleus within the brainstem that consists of norepinephrine-releasing neurons. It is involved in broad processes including cognitive and emotional functions. Understanding the mechanisms that control the excitability of LC neurons is important because they innervate widespread brain regions. One of the key regulators is cytosolic calcium concentration ([Ca2+]c), the increases in which can be amplified by calcium-induced calcium release (CICR) from intracellular calcium stores. Although the electrical activities of LC neurons are regulated by changes in [Ca2+]c, the extent of CICR involvement in this regulation has remained unclear. Here we show that CICR hyperpolarizes acutely dissociated LC neurons of the rat and demonstrate the underlying pathway. When CICR was activated by extracellular application of 10 mM caffeine, LC neurons were hyperpolarized in the current-clamp mode of patch-clamp recording, and the majority of neurons showed an outward current in the voltage-clamp mode. This outward current was accompanied by increased membrane conductance, and its reversal potential was close to the K+ equilibrium potential, indicating that it is mediated by opening of K+ channels. The outward current was generated in the absence of extracellular calcium and was blocked when the calcium stores were inhibited by applying ryanodine. Pharmacological blockers indicated that it was mediated by Ca2+-activated K+ channels of the non-small conductance type. The application of caffeine increased [Ca2+]c, as visualized by fluorescence microscopy. These findings show CICR suppresses LC neuronal activity, and indicate its dynamic role in modulating the LC-mediated noradrenergic tone in the brain.


Subject(s)
Adrenergic Neurons/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Locus Coeruleus/metabolism , Adrenergic Neurons/drug effects , Animals , Calcium Signaling/drug effects , Central Nervous System Stimulants/pharmacology , Locus Coeruleus/drug effects , Rats , Rats, Sprague-Dawley
4.
PLoS One ; 13(11): e0206123, 2018.
Article in English | MEDLINE | ID: mdl-30403723

ABSTRACT

Autosomal-dominant, early-onset DYT1 dystonia is associated with an in-frame deletion of a glutamic acid codon (ΔE) in the TOR1A gene. The gene product, torsinA, is an evolutionarily conserved AAA+ ATPase. The fact that constitutive secretion from patient fibroblasts is suppressed indicates that the ΔE-torsinA protein influences the cellular secretory machinery. However, which component is affected remains unclear. Prompted by recent reports that abnormal protein trafficking through the Golgi apparatus, the major protein-sorting center of the secretory pathway, is sometimes associated with a morphological change in the Golgi, we evaluated the influence of ΔE-torsinA on this organelle. Specifically, we examined its structure by confocal microscopy, in cultures of striatal, cerebral cortical and hippocampal neurons obtained from wild-type, heterozygous and homozygous ΔE-torsinA knock-in mice. In live neurons, the Golgi was assessed following uptake of a fluorescent ceramide analog, and in fixed neurons it was analyzed by immuno-fluorescence staining for the Golgi-marker GM130. Neither staining method indicated genotype-specific differences in the size, staining intensity, shape or localization of the Golgi. Moreover, no genotype-specific difference was observed as the neurons matured in vitro. These results were supported by a lack of genotype-specific differences in GM130 expression levels, as assessed by Western blotting. The Golgi was also disrupted by treatment with brefeldin A, but no genotype-specific differences were found in the immuno-fluorescence staining intensity of GM130. Overall, our results demonstrate that the ΔE-torsinA protein does not drastically influence Golgi morphology in neurons, irrespective of genotype, brain region (among those tested), or maturation stage in culture. While it remains possible that functional changes in the Golgi exist, our findings imply that any such changes are not severe enough to influence its morphology to a degree detectable by light microscopy.


Subject(s)
Dystonia/genetics , Gene Deletion , Golgi Apparatus/metabolism , Molecular Chaperones/genetics , Animals , Autoantigens/metabolism , Brefeldin A/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Ceramides/metabolism , Dendrites/drug effects , Dendrites/metabolism , Fluorescence , Gene Knock-In Techniques , Golgi Apparatus/drug effects , Hippocampus/pathology , Image Processing, Computer-Assisted , Membrane Proteins/metabolism , Mice , Molecular Chaperones/metabolism , Neostriatum/pathology , Neurons/drug effects , Neurons/metabolism
5.
J Neurosci ; 36(40): 10245-10256, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27707963

ABSTRACT

Dystonia type 1 (DYT1) is a dominantly inherited neurological disease caused by mutations in TOR1A, the gene encoding the endoplasmic reticulum (ER)-resident protein torsinA. Previous work mostly completed in cell-based systems suggests that mutant torsinA alters protein processing in the secretory pathway. We hypothesized that inducing ER stress in the mammalian brain in vivo would trigger or exacerbate mutant torsinA-induced dysfunction. To test this hypothesis, we crossed DYT1 knock-in with p58(IPK)-null mice. The ER co-chaperone p58(IPK) interacts with BiP and assists in protein maturation by helping to fold ER cargo. Its deletion increases the cellular sensitivity to ER stress. We found a lower generation of DYT1 knock-in/p58 knock-out mice than expected from this cross, suggesting a developmental interaction that influences viability. However, surviving animals did not exhibit abnormal motor function. Analysis of brain tissue uncovered dysregulation of eiF2α and Akt/mTOR translational control pathways in the DYT1 brain, a finding confirmed in a second rodent model and in human brain. Finally, an unbiased proteomic analysis identified relevant changes in the neuronal protein landscape suggesting abnormal ER protein metabolism and calcium dysregulation. Functional studies confirmed the interaction between the DYT1 genotype and neuronal calcium dynamics. Overall, these findings advance our knowledge on dystonia, linking translational control pathways and calcium physiology to dystonia pathogenesis and identifying potential new pharmacological targets. SIGNIFICANCE STATEMENT: Dystonia type 1 (DYT1) is one of the different forms of inherited dystonia, a neurological disorder characterized by involuntary, disabling movements. DYT1 is caused by mutations in the gene that encodes the endoplasmic reticulum (ER)-resident protein torsinA. How mutant torsinA causes neuronal dysfunction remains unknown. Here, we show the behavioral and molecular consequences of stressing the ER in DYT1 mice by increasing the amount of misfolded proteins. This resulted in the generation of a reduced number of animals, evidence of abnormal ER protein processing and dysregulation of translational control pathways. The work described here proposes a shared mechanism for different forms of dystonia, links for the first time known biological pathways to dystonia pathogenesis, and uncovers potential pharmacological targets for its treatment.


Subject(s)
Dystonia/genetics , Dystonia/physiopathology , Endoplasmic Reticulum/genetics , Molecular Chaperones/genetics , Animals , Behavior, Animal , Calcium Signaling/genetics , Cerebellum/physiopathology , Dystonia/psychology , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation/genetics , Gene Knock-In Techniques , Genotype , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Knockout , Neurons/physiology , Signal Transduction/genetics
6.
J Vis Exp ; (95)2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25742545

ABSTRACT

High-resolution analysis of the morphology and function of mammalian neurons often requires the genotyping of individual animals followed by the analysis of primary cultures of neurons. We describe a set of procedures for: labeling newborn mice to be genotyped, rapid genotyping, and establishing low-density cultures of brain neurons from these mice. Individual mice are labeled by tattooing, which allows for long-term identification lasting into adulthood. Genotyping by the described protocol is fast and efficient, and allows for automated extraction of nucleic acid with good reliability. This is useful under circumstances where sufficient time for conventional genotyping is not available, e.g., in mice that suffer from neonatal lethality. Primary neuronal cultures are generated at low density, which enables imaging experiments at high spatial resolution. This culture method requires the preparation of glial feeder layers prior to neuronal plating. The protocol is applied in its entirety to a mouse model of the movement disorder DYT1 dystonia (ΔE-torsinA knock-in mice), and neuronal cultures are prepared from the hippocampus, cerebral cortex and striatum of these mice. This protocol can be applied to mice with other genetic mutations, as well as to animals of other species. Furthermore, individual components of the protocol can be used for isolated sub-projects. Thus this protocol will have wide applications, not only in neuroscience but also in other fields of biological and medical sciences.


Subject(s)
Brain/cytology , Cell Culture Techniques/methods , Genotyping Techniques/methods , Neurons/cytology , Animals , Animals, Newborn , Disease Models, Animal , Dystonic Disorders/genetics , Mice , Molecular Chaperones/genetics , Rats
7.
Article in English | MEDLINE | ID: mdl-25279252

ABSTRACT

BACKGROUND: An in-frame deletion leading to the loss of a single glutamic acid residue in the protein torsinA (ΔE-torsinA) results in an inherited movement disorder, DYT1 dystonia. This autosomal dominant disease affects the function of the brain without causing neurodegeneration, by a mechanism that remains unknown. METHODS: We evaluated the literature regarding the subcellular localization of torsinA. RESULTS: Efforts to elucidate the pathophysiological basis of DYT1 dystonia have relied partly on examining the subcellular distribution of the wild-type and mutated proteins. A typical approach is to introduce the human torsinA gene (TOR1A) into host cells and overexpress the protein therein. In both neurons and non-neuronal cells, exogenous wild-type torsinA introduced in this manner has been found to localize mainly to the endoplasmic reticulum, whereas exogenous ΔE-torsinA is predominantly in the nuclear envelope or cytoplasmic inclusions. Although these outcomes are relatively consistent, findings for the localization of endogenous torsinA have been variable, leaving its physiological distribution a matter of debate. DISCUSSION: As patients' cells do not overexpress torsinA proteins, it is important to understand why the reported distributions of the endogenous proteins are inconsistent. We propose that careful optimization of experimental methods will be critical in addressing the causes of the differences among the distributions of endogenous (non-overexpressed) vs. exogenously introduced (overexpressed) proteins.

9.
Neurosci Res ; 84: 68-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24815515

ABSTRACT

Ischemic tolerance resulting from preconditioning ischemia is a neuroprotective mechanism. In cultured astrocytes, its development depends on regulation of the expression of glucose transporter 3 (GLUT3) by the stress sensor/effector AMP-activated protein kinase (AMPK). Here we demonstrate that GLUT3 is upregulated during preconditioning and then downregulated during recovery. We also found that, although AMPK inhibition during preconditioning initially suppressed the upregulation of GLUT3 as shown previously, this was followed by a period of GLUT3 upregulation, enhanced glycogen accumulation, and enhanced tolerance to a subsequent ischemic challenge. These results reveal that AMPK has a complex influence on ischemic tolerance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Gene Expression Regulation/physiology , Glucose Transporter Type 3/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Cerebral Cortex/cytology , Gene Expression Regulation/drug effects , Glucose/deficiency , Glucose Transporter Type 3/genetics , Hypoxia/pathology , L-Lactate Dehydrogenase/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats
10.
J Vis Exp ; (85)2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24747983

ABSTRACT

Synaptic vesicles in functional nerve terminals undergo exocytosis and endocytosis. This synaptic vesicle recycling can be effectively analyzed using styryl FM dyes, which reveal membrane turnover. Conventional protocols for the use of FM dyes were designed for analyzing neurons following stimulated (evoked) synaptic activity. Recently, protocols have become available for analyzing the FM signals that accompany weaker synaptic activities, such as spontaneous or miniature synaptic events. Analysis of these small changes in FM signals requires that the imaging system is sufficiently sensitive to detect small changes in intensity, yet that artifactual changes of large amplitude are suppressed. Here we describe a protocol that can be applied to evoked, spontaneous, and miniature synaptic activities, and use cultured hippocampal neurons as an example. This protocol also incorporates a means of assessing the rate of photobleaching of FM dyes, as this is a significant source of artifacts when imaging small changes in intensity.


Subject(s)
Chromosome Pairing/physiology , Fluorescent Dyes/chemistry , Pyridinium Compounds/chemistry , Quaternary Ammonium Compounds/chemistry , Synaptic Vesicles/physiology , Animals , Mice , Photobleaching , Presynaptic Terminals/chemistry , Presynaptic Terminals/physiology , Rats , Synaptic Transmission/physiology , Synaptic Vesicles/chemistry
11.
J Neurosci Methods ; 221: 15-21, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24025262

ABSTRACT

BACKGROUND: Although recent developments in methodologies for light microscopy have enabled imaging of fine biological structures, such imaging is often accompanied by two types of problems. One is a tilting of the specimen with respect to the x-y plane (i.e. rotation around the x- or y-axis) such that the sample is not perpendicular to the optical z-axis, and the other is rotation around the z-axis that precludes optimal orientations for imaging and experimentation. These rotation problems can cause optical aberrations and hamper imaging experiments, even when the angular difference from the ideal position is small. NEW METHOD: In order to correct for these practical issues, we have developed a specimen holder with 3-axis (x-y-z) rotation for an inverted light microscope. This allows for full-range rotations of 2-4° for x-, y-axes, ~24° for z-axis, and a small-angle control of <0.1° for either axis. RESULTS: Using this device, we observed the cultured hippocampal neurons stained by immunofluorescence for a dendritic marker, or the sub-resolution fluorescent beads plated on a glass coverslip. The rotations and associated problems could be manipulated, while viewing the specimens by laser-scanning confocal microscopy. COMPARISON WITH EXISTING METHODS: This tilting/rotation device is easily manufactured and installed on a conventional microscope stage without requiring changes to the existing optical components. Similar devices with full capability have not been available. CONCLUSIONS: It will be useful for imaging experiments with biomedical applications.


Subject(s)
Hippocampus/cytology , Microscopy, Confocal/instrumentation , Microscopy/instrumentation , Neurons/cytology , Animals , Fluorescent Antibody Technique , Mice
12.
J Neurosci Methods ; 223: 92-113, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24333471

ABSTRACT

BACKGROUND: Images in biomedical imaging research are often affected by non-specific background noise. This poses a serious problem when the noise overlaps with specific signals to be quantified, e.g. for their number and intensity. A simple and effective means of removing background noise is to prepare a filtered image that closely reflects background noise and to subtract it from the original unfiltered image. This approach is in common use, but its effectiveness in identifying and quantifying synaptic puncta has not been characterized in detail. NEW ANALYSIS: We report on our assessment of the effectiveness of isolating punctate signals from diffusely distributed background noise using one variant of this approach, "Difference of Gaussian(s) (DoG)" which is based on a Gaussian filter. RESULTS: We evaluated immunocytochemically stained, cultured mouse hippocampal neurons as an example, and provided the rationale for choosing specific parameter values for individual steps in detecting glutamatergic nerve terminals. The intensity and width of the detected puncta were proportional to those obtained by manual fitting of two-dimensional Gaussian functions to the local information in the original image. COMPARISON WITH EXISTING METHODS: DoG was compared with the rolling-ball method, using biological data and numerical simulations. Both methods removed background noise, but differed slightly with respect to their efficiency in discriminating neighboring peaks, as well as their susceptibility to high-frequency noise and variability in object size. CONCLUSIONS: DoG will be useful in detecting punctate signals, once its characteristics are examined quantitatively by experimenters.


Subject(s)
Fluorescent Antibody Technique , Image Interpretation, Computer-Assisted , Neurons/cytology , Normal Distribution , Synapses/metabolism , Animals , Animals, Newborn , Cells, Cultured , Hippocampus/cytology , Mice , Microscopy, Confocal , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
13.
PLoS One ; 8(11): e80793, 2013.
Article in English | MEDLINE | ID: mdl-24260480

ABSTRACT

DYT1 dystonia is the most common hereditary form of primary torsion dystonia. This autosomal-dominant disorder is characterized by involuntary muscle contractions that cause sustained twisting and repetitive movements. It is caused by an in-frame deletion in the TOR1A gene, leading to the deletion of a glutamic acid residue in the torsinA protein. Heterozygous knock-in mice, which reproduce the genetic mutation in human patients, have abnormalities in synaptic transmission at the principal GABAergic neurons in the striatum, a brain structure that is involved in the execution and modulation of motor activity. However, whether this mutation affects the excitability of striatal GABAergic neurons has not been investigated in this animal model. Here, we examined the excitability of cultured striatal neurons obtained from heterozygous knock-in mice, using calcium imaging as indirect readout. Immunofluorescence revealed that more than 97% of these neurons are positive for a marker of GABAergic neurons, and that more than 92% are also positive for a marker of medium spiny neurons, indicating that these are mixed cultures of mostly medium spiny neurons and a few (~5%) GABAergic interneurons. When these neurons were depolarized by field stimulation, the calcium concentration in the dendrites increased rapidly and then decayed slowly. The amplitudes of calcium transients were larger in heterozygous neurons than in wild-type neurons, resulting in ~15% increase in cumulative calcium transients during a train of stimuli. However, there was no change in other parameters of calcium dynamics. Given that calcium dynamics reflect neuronal excitability, these results suggest that the mutation only slightly increases the excitability of striatal GABAergic neurons in DYT1 dystonia.


Subject(s)
Calcium/metabolism , Corpus Striatum/metabolism , Cytoplasm/metabolism , Dystonia Musculorum Deformans/genetics , Dystonic Disorders/genetics , GABAergic Neurons/metabolism , Action Potentials , Animals , Cells, Cultured , Cholinergic Neurons/metabolism , Disease Models, Animal , Heterozygote , Interneurons/metabolism , Mice , Mice, Transgenic , Mutation
14.
Neuroscience ; 253: 316-29, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24025868

ABSTRACT

Presynaptic functions of the mammalian central neurons are regulated by a network of protein interactions. Synaptic vesicle recycling in and neurotransmitter release from the presynaptic nerve terminals are altered when a glutamate-deleting mutation is present in the torsinA protein (ΔE-torsinA). This mutation is linked with a hereditary form of the movement disorder dystonia known as DYT1 dystonia. Although torsinA expression is prevalent throughout the central nervous system, its subcellular localization - in particular with respect to presynaptic nerve terminals - remains unclear. This information would be useful in narrowing down possible models for how wild-type torsinA affects presynaptic function, as well as the nature of the presynaptic dysfunction that arises in the context of ΔE-torsinA mutation. Here we report on an analysis of the presynaptic localization of torsinA in cultured neurons obtained from a knock-in mouse model of DYT1 dystonia. Primary cultures of neurons were established from heterozygous and homozygous ΔE-torsinA knock-in mice, as well as from their wild-type littermates. Neurons were obtained from the striatum, cerebral cortex and hippocampus of these mice, and were subjected to immunocytochemistry. This analysis revealed the expression of both proteins in the somata and dendrites. However, neither the nerve terminals nor axonal shafts were immunoreactive. These results were confirmed by fluorogram-based quantitation. Our findings indicate that neither the wild-type nor the ΔE-torsinA mutant protein is present at substantial levels in the presynaptic structures of cultured neurons. Thus, the effects of torsinA, in wild-type and mutant forms, appear to influence presynaptic function indirectly, without residing in presynaptic structures.


Subject(s)
Hippocampus/cytology , Molecular Chaperones/metabolism , Nerve Endings/metabolism , Neurons/cytology , Analysis of Variance , Animals , Cadmium Chloride/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Fluorescent Antibody Technique , Mice , Mice, Inbred BALB C , Neurons/drug effects , Potassium/pharmacology , omega-Agatoxin IVA/pharmacology , omega-Conotoxin GVIA/pharmacology
15.
Neurosci Lett ; 548: 61-6, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23748075

ABSTRACT

Increased activities of cytoplasmic calcium and the excitatory neurotransmitter glutamate have been independently implicated in dystonia pathophysiology. However, cellular-level evidence linking these two features is not available. Here we show that glutamate-dependent changes in neuronal calcium dynamics occur in a knock-in mouse model of DYT1 dystonia, the most common hereditary form of this disorder. Fluorescence-based analysis of the dynamics of cytoplasmic calcium concentration ([Ca(2+)]c) in cultured hippocampal neurons shows that electrical stimulation depolarizes the neurons and increases the dendritic [Ca(2+)]c, which then decays slowly to the pre-stimulus level. Whereas the peak amplitude of [Ca(2+)]c was not affected, the decay period was prolonged in neurons of heterozygous mice whose genotype reflects the human condition. We found that this effect was blocked by the antagonists of ionotropic glutamate receptors, and confirmed that glutamate receptors are present in these neurons. As the [Ca(2+)]c is readout and regulator of neuronal excitability, its abnormality represents an important cellular phenotype of dystonia.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cytoplasm/metabolism , Dystonia/metabolism , Hippocampus , Neurons/metabolism , Receptors, Glutamate/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout
16.
PLoS One ; 7(10): e48034, 2012.
Article in English | MEDLINE | ID: mdl-23110166

ABSTRACT

The astrocyte is a major glial cell type of the brain, and plays key roles in the formation, maturation, stabilization and elimination of synapses. Thus, changes in astrocyte condition and age can influence information processing at synapses. However, whether and how aging astrocytes affect synaptic function and maturation have not yet been thoroughly investigated. Here, we show the effects of prolonged culture on the ability of astrocytes to induce synapse formation and to modify synaptic transmission, using cultured autaptic neurons. By 9 weeks in culture, astrocytes derived from the mouse cerebral cortex demonstrated increases in ß-galactosidase activity and glial fibrillary acidic protein (GFAP) expression, both of which are characteristic of aging and glial activation in vitro. Autaptic hippocampal neurons plated on these aging astrocytes showed a smaller amount of evoked release of the excitatory neurotransmitter glutamate, and a lower frequency of miniature release of glutamate, both of which were attributable to a reduction in the pool of readily releasable synaptic vesicles. Other features of synaptogenesis and synaptic transmission were retained, for example the ability to induce structural synapses, the presynaptic release probability, the fraction of functional presynaptic nerve terminals, and the ability to recruit functional AMPA and NMDA glutamate receptors to synapses. Thus the presence of aging astrocytes affects the efficiency of synaptic transmission. Given that the pool of readily releasable vesicles is also small at immature synapses, our results are consistent with astrocytic aging leading to retarded synapse maturation.


Subject(s)
Astrocytes/physiology , Cellular Senescence/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/physiology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cells, Cultured , Coculture Techniques , Excitatory Postsynaptic Potentials/physiology , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Immunohistochemistry , Mice , Mice, Inbred ICR , Microscopy, Confocal , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Vesicles/metabolism , Time Factors , beta-Galactosidase/metabolism
17.
Synapse ; 66(9): 807-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22588999

ABSTRACT

TorsinA is an evolutionarily conserved AAA+ ATPase, and human patients with an in-frame deletion of a single glutamate (ΔE) codon from the encoding gene suffer from autosomal-dominant, early-onset generalized DYT1 dystonia. Although only 30-40% of carriers of the mutation show overt motor symptoms, most experience enhanced excitability of the central nervous system. The cellular mechanism responsible for this change in excitability is not well understood. Here we show the effects of the ΔE-torsinA mutation on miniature neurotransmitter release from neurons. Neurotransmitter release was characterized in cultured hippocampal neurons obtained from wild-type, heterozygous, and homozygous ΔE-torsinA knock-in mice using two approaches. In the first approach, patch-clamp electrophysiology was used to record glutamate-mediated miniature excitatory postsynaptic currents (mEPSCs) in the presence of the Na⁺ channel blocker tetrodotoxin (TTX) and absence of GABA(A) receptor antagonists. The intervals between mEPSC events were significantly shorter in neurons obtained from the mutant mice than in those obtained from wild-type mice. In the second approach, the miniature exocytosis of synaptic vesicles was detected by imaging the unstimulated release of FM dye from the nerve terminals in the presence of TTX. Cumulative FM dye release was higher in neurons obtained from the mutant mice than in those obtained from wild-type mice. The number of glutamatergic nerve terminals was also assessed, and we found that this number was unchanged in heterozygous relative to wild-type neurons, but slightly increased in homozygous neurons. Notably, in both heterozygous and homozygous neurons, the unitary synaptic charge during each mEPSC event was unchanged. Overall, our results suggest more frequent miniature glutamate release in neurons with ΔE-torsinA mutations. This change may be one of the underlying mechanisms by which the excitability of the central nervous system is enhanced in the context of DYT1 dystonia. Moreover, qualitative differences between heterozygous and homozygous neurons with respect to certain synaptic properties indicate that the abnormalities observed in homozygotes may reflect more than a simple gene dosage effect.


Subject(s)
Exocytosis/physiology , Glutamic Acid/metabolism , Hippocampus/physiology , Miniature Postsynaptic Potentials/physiology , Molecular Chaperones/genetics , Neurons/physiology , Animals , Disease Models, Animal , Dystonia Musculorum Deformans/genetics , Excitatory Postsynaptic Potentials/physiology , Heterozygote , Hippocampus/cytology , Homozygote , Inhibitory Postsynaptic Potentials/physiology , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Mutation , Receptors, GABA-A/metabolism , Sodium Channels/drug effects , Sodium Channels/metabolism , Synaptic Vesicles/metabolism , Tetrodotoxin/pharmacology , Vesicular Glutamate Transport Protein 1/metabolism
18.
Synapse ; 66(5): 453-64, 2012 May.
Article in English | MEDLINE | ID: mdl-22213465

ABSTRACT

Early-onset generalized dystonia, DYT1, is caused by a mutation in the gene encoding the evolutionarily conserved AAA+ ATPase torsinA. Synaptic abnormalities have been implicated in DYT1 dystonia, but the details of the synaptic pathophysiology are only partially understood. Here, we demonstrate a novel role for torsinA in synaptic vesicle recycling, using cultured hippocampal neurons from a knock-in mouse model of DYT1 dystonia (ΔE-torsinA) and live-cell imaging with styryl FM dyes. Neurons from heterozygous ΔE-torsinA mice released a larger fraction of the total recycling pool (TRP) during a single round of electrical stimulation than did wild-type neurons. Moreover, when the neurons were subjected to prior high activity, the time course of release was shortened. In neurons from homozygous mice, these enhanced exocytosis phenotypes were similar, but in addition the size of the TRP was reduced. Notably, when release was triggered by applying a calcium ionophore rather than electrical stimuli, neither a single nor two ΔE-torsinA alleles affected the time course of release. Thus, the site of action of ΔE-torsinA is at or upstream of the rise in calcium concentration in nerve terminals. Our results suggest that torsinA regulates synaptic vesicle recycling in central neurons. They also indicate that this regulation is influenced by neuronal activity, further supporting the idea that synaptic abnormalities contribute to the pathophysiology of DYT1 dystonia.


Subject(s)
Dystonic Disorders/metabolism , Molecular Chaperones/metabolism , Neurons/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Dystonic Disorders/genetics , Endocytosis/physiology , Exocytosis/physiology , Gene Knock-In Techniques , Heterozygote , Mice , Models, Animal , Molecular Chaperones/genetics , Synaptic Vesicles/genetics
19.
Neuropharmacology ; 45(6): 797-813, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14529718

ABSTRACT

At synapses of the mammalian central nervous system, release of neurotransmitter occurs at rates transiently as high as 100 Hz, putting extreme demands on nerve terminals with only tens of functional vesicles at their disposal. Thus, the presynaptic vesicle cycle is particularly critical to maintain neurotransmission. To understand vesicle cycling at the most fundamental level, we studied single vesicles undergoing exo/endocytosis and tracked the fate of newly retrieved vesicles. This was accomplished by minimally stimulating boutons in the presence of the membrane-fluorescent styryl dye FM1-43, then selecting for terminals that contained only one dye-filled vesicle. We then observed the kinetics of dye release during single action potential stimulation. We found that most vesicles lost only a portion of their total dye during a single fusion event, but were able to fuse again soon thereafter. We interpret this as direct evidence of "kiss-and-run" followed by rapid reuse. Other interpretations such as "partial loading" and "endosomal splitting" were largely excluded on the basis of multiple lines of evidence. Our data placed an upper bound of <1.4 s on the lifetime of the kiss-and-run fusion event, based on the assumption that aqueous departitioning is rate limiting. The repeated use of individual vesicles held over a range of stimulus frequencies up to 30 Hz and was associated with neurotransmitter release. A small percentage of fusion events did release a whole vesicle's worth of dye in one action potential, consistent with a classical picture of exocytosis as fusion followed by complete collapse or at least very slow retrieval.


Subject(s)
Endocytosis/physiology , Exocytosis/physiology , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Action Potentials/physiology , Animals , Cells, Cultured , Hippocampus/metabolism , Hippocampus/ultrastructure , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/ultrastructure
20.
J Neurosci ; 21(23): 9185-93, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11717352

ABSTRACT

The expansion of polyglutamine tracts encoded by CAG trinucleotide repeats is a common mutational mechanism in inherited neurodegenerative diseases. Spinocerebellar ataxia type 6 (SCA6), an autosomal dominant, progressive disease, arises from trinucleotide repeat expansions present in the coding region of CACNA1A (chromosome 19p13). This gene encodes alpha(1A), the principal subunit of P/Q-type Ca(2+) channels, which are abundant in the CNS, particularly in cerebellar Purkinje and granule neurons. We assayed ion channel function by introduction of human alpha(1A) cDNAs in human embryonic kidney 293 cells that stably coexpressed beta(1) and alpha(2)delta subunits. Immunocytochemical analysis showed a rise in intracellular and surface expression of alpha(1A) protein when CAG repeat lengths reached or exceeded the pathogenic range for SCA6. This gain at the protein level was not a consequence of changes in RNA stability, as indicated by Northern blot analysis. The electrophysiological behavior of alpha(1A) subunits containing expanded (EXP) numbers of CAG repeats (23, 27, and 72) was compared against that of wild-type subunits (WT) (4 and 11 repeats) using standard whole-cell patch-clamp recording conditions. The EXP alpha(1A) subunits yielded functional ion channels that supported inward Ca(2+) channel currents, with a sharp increase in P/Q Ca(2+) channel current density relative to WT. Our results showed that Ca(2+) channels from SCA6 patients display near-normal biophysical properties but increased current density attributable to elevated protein expression at the cell surface.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Protein Subunits , Spinocerebellar Ataxias/etiology , Trinucleotide Repeat Expansion/genetics , Blotting, Northern , Calcium/metabolism , Calcium Channels, P-Type/genetics , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/genetics , Calcium Channels, Q-Type/metabolism , Cell Line , Cell Membrane/metabolism , Chromosomes, Human, Pair 19/genetics , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression , Genes, Dominant , Humans , Immunohistochemistry , Ion Transport/genetics , Kidney/cytology , Kidney/metabolism , Patch-Clamp Techniques , RNA, Messenger/metabolism , Spinocerebellar Ataxias/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...