Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Exp Neurobiol ; 30(2): 120-143, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33972466

ABSTRACT

Central neurocytoma (CN) has been known as a benign neuronal tumor. In rare cases, CN undergoes malignant transformation to glioblastomas (GBM). Here we examined its cellular origin by characterizing differentiation potential and gene expression of CN-spheroids. First, we demonstrate that both CN tissue and cultured primary cells recapitulate the hierarchal cellular composition of subventricular zone (SVZ), which is comprised of neural stem cells (NSCs), transit amplifying progenitors (TAPs), and neuroblasts. We then derived spheroids from CN which displayed EGFR+/ MASH+ TAP and BLBP+ radial glial cell (RGC) characteristic, and mitotic neurogenesis and gliogenesis by single spheroids were observed with cycling multipotential cells. CN-spheroids expressed increased levels of pluripotency and tumor stem cell genes such as KLF4 and TPD5L1, when compared to their differentiated cells and human NSCs. Importantly, Gene Set Enrichment Analysis showed that gene sets of GBM-Spheroids, EGFR Signaling, and Packaging of Telomere Ends are enriched in CN-spheroids in comparison with their differentiated cells. We speculate that CN tumor stem cells have TAP and RGC characteristics, and upregulation of EGFR signaling as well as downregulation of eph-ephrin signaling have critical roles in tumorigenesis of CN. And their ephemeral nature of TAPs destined to neuroblasts, might reflect benign nature of CN.

2.
Cell Rep ; 25(13): 3631-3646.e3, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30590038

ABSTRACT

A major mechanism contributing to synaptic plasticity involves alterations in the number of AMPA receptors (AMPARs) expressed at synapses. Hippocampal CA1 synapses, where this process has been most extensively studied, are highly heterogeneous with respect to their probability of neurotransmitter release, P(r). It is unknown whether there is any relationship between the extent of plasticity-related AMPAR trafficking and the initial P(r) of a synapse. To address this question, we induced metabotropic glutamate receptor (mGluR) dependent long-term depression (mGluR-LTD) and assessed AMPAR trafficking and P(r) at individual synapses, using SEP-GluA2 and FM4-64, respectively. We found that either pharmacological or synaptic activation of mGluR1 reduced synaptic SEP-GluA2 in a manner that depends upon P(r); this process involved an activity-dependent reduction in surface mGluR1 that selectively protects high-P(r) synapses from synaptic weakening. Consequently, the extent of postsynaptic plasticity can be pre-tuned by presynaptic activity.


Subject(s)
Cell Membrane/metabolism , Neurotransmitter Agents/metabolism , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Endocytosis/drug effects , Glutamates/metabolism , Long-Term Synaptic Depression/drug effects , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Probability , Protein Transport/drug effects , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism , Theta Rhythm/drug effects
3.
Mol Brain ; 7: 9, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24512690

ABSTRACT

BACKGROUND: Group I metabotropic glutamate receptors (mGlu1/5 receptors) have important roles in synaptic activity in the central nervous system. They modulate neuronal excitability by mobilizing intracellular Ca2+ following receptor activation. Also, accumulating evidence has indicated the association of Ca2+ signaling with lipid rafts. Caveolin, an adaptor protein found in a specialized subset of lipid rafts, has been reported to promote the localization of membrane proteins to lipid rafts. RESULTS: In the present study, we investigated the role of lipid rafts on the mGlu1α receptor-mediated Ca2+ signaling in association with caveolin in hippocampal primary neurons and HEK293 cells. We show that the disruption of lipid rafts using methyl-ß-cyclodextrin markedly decreased mGlu1α receptor-mediated Ca2+ transients and lipid rafts localization of the receptor. Furthermore, transfection of mGlu1α receptor with mutated caveolin-binding domain reduced localization of the receptor to lipid rafts. Also, application of a peptide blocker of mGlu1α receptor and caveolin binding reduced the Ca2+ signaling and the lipid rafts localization. CONCLUSIONS: Taken together, these results suggest that the binding of mGlu1α receptor to caveolin is crucial for its lipid rafts localization and mGlu1α receptor-mediated Ca2+ transients.


Subject(s)
Calcium Signaling , Caveolin 1/metabolism , Membrane Microdomains/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Calcium Signaling/drug effects , HEK293 Cells , Hippocampus/cytology , Humans , Membrane Microdomains/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacology
4.
FASEB J ; 27(12): 4776-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23982146

ABSTRACT

Mitochondrial dysfunction and synaptic damage are important features of Alzheimer's disease (AD) associated with amyloid ß (Aß) and tau. We reported previously that the scaffolding protein RanBP9, which is overall increased in brains of patients with AD and in mutant APP transgenic mice, simultaneously promotes Aß generation and focal adhesion disruption by accelerating the endocytosis of APP and ß1-integrin, respectively. Moreover, RanBP9 induces neurodegeneration in vitro and in vivo and mediates Aß-induced neurotoxicity. Here we show in primary hippocampal neurons that RanBP9 potentiates Aß-induced reactive oxygen species (ROS) overproduction, apoptosis, and calcium deregulation. Analyses of calcium-handling measures demonstrate that RanBP9 selectively delays the clearance of cytosolic Ca(2+) mediated by the mitochondrial calcium uniporter through a process involving the translocation of cofilin into mitochondria and oxidative mechanisms. Further, RanBP9 retards the anterograde axonal transport of mitochondria in primary neurons and decreases synaptic mitochondrial activity in brain. These data indicate that RanBP9, cofilin, and Aß mimic and potentiate each other to produce mitochondrial dysfunction, ROS overproduction, and calcium deregulation, which leads to neurodegenerative changes reminiscent of those seen in AD.


Subject(s)
Actin Depolymerizing Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Calcium Signaling , Cytoskeletal Proteins/metabolism , Mitochondria/metabolism , Nuclear Proteins/metabolism , Actin Depolymerizing Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis , Axonal Transport , Calcium/metabolism , Calcium Channels/metabolism , Cytoskeletal Proteins/genetics , Hippocampus/cytology , Membrane Potential, Mitochondrial , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Nuclear Proteins/genetics , Peptide Fragments/pharmacology , Reactive Oxygen Species/metabolism , Synapses/metabolism
5.
J Neurosci ; 32(37): 12909-14, 2012 Sep 12.
Article in English | MEDLINE | ID: mdl-22973014

ABSTRACT

In the cerebellum, synaptic strength at the synapses between parallel fibers and Purkinje cells is best known to be modulated via metabotropic glutamate receptor 1 (mGluR1)-dependent cerebellar long-term depression (LTD). An increase in intracellular calcium levels plays an important role in inducing mGluR1-dependent cerebellar LTD. Downstream of mGluR1, there are two major sources of calcium: transient receptor potential canonical (TRPC) channels and inositol trisphosphate receptors (IP(3)R). IP(3)R triggers a calcium release from the intracellular calcium store. Here, we show that TRPC channels mediate mGluR1-evoked slow currents to regulate cerebellar LTD in Sprague Dawley rats. We found that the inhibition of TRPC channels blocks the induction of cerebellar LTD. Moreover, we show that processes known to underlie cerebellar LTD induction, such as increases in intracellular calcium concentration, the activation of protein kinase C, and the internalization of GluR2, are also hindered by blocking TRPC. These results suggest that the mGluR1-evoked activation of TRPC channels is required for the induction of cerebellar LTD.


Subject(s)
Cerebellum/physiology , Long-Term Synaptic Depression/physiology , Receptors, Metabotropic Glutamate/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Female , Male , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Cell Physiol ; 301(1): C150-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21411724

ABSTRACT

Since the first isolation of endothelial progenitor cells (EPCs) from human peripheral blood in 1997, many researchers have conducted studies to understand the characteristics and therapeutic effects of EPCs in vascular disease models. Nevertheless, the electrophysiological properties of EPCs have yet to be clearly elucidated. The inward rectifier potassium channel (Kir) performs a major role in controlling the membrane potential and cellular events. Here, via the whole cell patch-clamp technique, we found inwardly rectifying currents in EPCs and that these currents were inhibited by Ba(2+) (100 µM) and Cs(+) (1 mM), known as Kir blockers, in a dose-dependent manner (Ba(2+), 91.2 ± 1.4% at -140 mV and Cs(+), 76.1 ± 6.9% at -140 mV, respectively). Next, using DiBAC(3), a fluorescence indicator of membrane potential, we verified that Ba(2+) induced an increase of fluorescence in EPCs (10 µM, 123 ± 2.8%), implying the depolarization of EPCs. At the mRNA and protein levels, we confirmed the existence of several Kir subtypes, including Kir2.x, 3.x, 4.x, and 6.x. In a functional experiment, we observed that, in the presence of Ba(2+), the number of tubes on Matrigel formed by EPCs was dose-dependently reduced (10 µM, 62.3 ± 6.5%). In addition, the proliferation of EPCs was increased in a dose-dependent fashion (10 µM, 157.9 ± 17.4%), and specific inhibition of Kir2.1 by small interfering RNA also increased the proliferation of EPCs (116.2 ± 2.5%). Our results demonstrate that EPCs express several types of Kir which may modulate the endothelial function and proliferation of EPCs.


Subject(s)
Endothelial Cells/metabolism , Membrane Potentials , Potassium Channels, Inwardly Rectifying/metabolism , Stem Cells/metabolism , Barium/pharmacology , Blotting, Western , Cell Proliferation , Cesium/pharmacology , Fetal Blood , Fluorescent Antibody Technique , Humans , Leukocytes, Mononuclear/metabolism , Membrane Potentials/drug effects , Patch-Clamp Techniques , Phenotype , Polymerase Chain Reaction , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/genetics , RNA Interference , RNA, Small Interfering
7.
PLoS One ; 5(5): e10359, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20479868

ABSTRACT

Store-operated Ca(2+) entry (SOCE) channels are the main pathway of Ca(2+) entry in non-excitable cells such as neural progenitor cells (NPCs). However, the role of SOCE channels has not been defined in the neuronal differentiation from NPCs. Here, we show that canonical transient receptor potential channel (TRPC) as SOCE channel influences the induction of the neuronal differentiation of A2B5(+) NPCs isolated from postnatal-12-day rat cerebrums. The amplitudes of SOCE were significantly higher in neural cells differentiated from proliferating A2B5(+) NPCs and applications of SOCE blockers, 2-aminoethoxy-diphenylborane (2-APB), and ruthenium red (RR), inhibited their rise of SOCE. Among TRPC subtypes (TRPC1-7), marked expression of TRPC5 and TRPC6 with turned-off TRPC1 expression was observed in neuronal cells differentiated from proliferating A2B5(+) NPCs. TRPC5 small interfering RNA (siRNA) blocked the neuronal differentiation from A2B5(+) NPCs and reduced the rise of SOCE. In contrast, TRPC6 siRNA had no significant effect on the neuronal differentiation from A2B5(+) NPCs. These results indicate that calcium regulation by TRPC5 would play a key role as a switch between proliferation and neuronal differentiation from NPCs.


Subject(s)
Cell Differentiation , Neurons/cytology , Stem Cells/cytology , Stem Cells/metabolism , TRPC Cation Channels/metabolism , Animals , Boron Compounds/pharmacology , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Gene Knockdown Techniques , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , TRPC Cation Channels/genetics , Thapsigargin/pharmacology
8.
J Neurochem ; 111(1): 61-71, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19627451

ABSTRACT

Agonist-induced internalization of metabotropic glutamate receptors (mGluRs) plays an important role in neuronal signaling. Although internalization of mGluRs has been reported to be mediated by clathrin-dependent pathway, studies describing clathrin-independent pathways are emerging. Here, we report that agonist-induced internalization of mGluR1alpha is mediated by caveolin. We show that two caveolin-binding motifs of mGluR1alpha interact with caveolin1/2. Using cell surface-immunoprecipitation and total internal reflection fluorescence imaging, we found that agonist-induced internalization of mGluR1alpha is regulated by caveolin-binding motifs of the receptor in heterologous cells. Moreover, in the cerebellum, group I mGluR agonist dihydroxyphenylglycol increased the interaction of phosphorylated caveolin with mGluR1alpha. This interaction was blocked by methyl-beta-cyclodextrin, known to disrupt caveolin/caveolae-dependent signaling by cholesterol depletion. Methyl-beta-cyclodextrin also blocked the agonist-induced internalization of mGluR1alpha. Thus, these findings represent the evidence for agonist-induced internalization of mGluR1alpha via caveolin and suggest that caveolin might play a role in synaptic metaplasticity by regulating internalization of mGluR1alpha in the cerebellum.


Subject(s)
Caveolins/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Excitatory Amino Acid Agonists/pharmacology , Quisqualic Acid/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Carcinoma , Cell Line, Transformed , Cell Line, Tumor , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Humans , Immunoprecipitation/methods , In Vitro Techniques , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Luminescent Proteins/genetics , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mutation/genetics , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Transfection/methods
9.
J Mater Sci Mater Med ; 19(8): 2953-62, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18360798

ABSTRACT

Stem cell-based therapy has recently emerged for use in novel therapeutics for incurable diseases. For successful recovery from neurologic diseases, the most pivotal factor is differentiation and directed neuronal cell growth. In this study, we fabricated three different widths of a micro-pattern on polydimethylsiloxane (PDMS; 1, 2, and 4 microm). Surface modification of the PDMS was investigated for its capacity to manage proliferation and differentiation of neural-like cells from umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs). Among the micro-patterned PDMS fabrications, the 1 microm-patterned PDMS significantly increased cell proliferation and most of the cells differentiated into neuronal cells. In addition, the 1 microm-patterned PDMS induced an increase in cytosolic calcium, while the differentiated cells on the flat and 4 microm-patterned PDMS had no response. PDMS with a 1 microm pattern was also aligned to direct orientation within 10 degrees angles. Taken together, micro-patterned PDMS supported UCB-MSC proliferation and induced neural like-cell differentiation. Our data suggest that micro-patterned PDMS might be a guiding method for stem cell therapy that would improve its therapeutic action in neurological diseases.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dimethylpolysiloxanes/pharmacology , Mesenchymal Stem Cells/drug effects , Umbilical Cord/cytology , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology , Microscopy, Electron, Scanning , Surface Properties
10.
Biochem Biophys Res Commun ; 337(2): 481-9, 2005 Nov 18.
Article in English | MEDLINE | ID: mdl-16223607

ABSTRACT

Neuronatin (Nnat) is selectively expressed in the neonatal brain and is involved in neuronal differentiation during brain development. However, Nnat also appears to be abundantly expressed in adipose tissue, and is conspicuously elevated in the adipose tissue of obese Zucker diabetic fatty rats compared with control lean Zucker lean control rats shown in our previous report. Here, we examined the expression of Nnat in adipose tissue and demonstrated that the ectopic expression of Nnat mediated by retroviral infection or stable transfection of 3T3-L1 pre-adipocytes stimulated differentiation into mature adipocytes with early induction of adipogenic transcription factors. Moreover, in 3T3-L1 cells overexpressing Nnat, increased intracellular free calcium levels and enhanced phosphorylation of cAMP-response element-binding protein (CREB) were observed, which appears to potentiate CCAAT/enhancer-binding protein (C/EBP)beta, C/EBPdelta, and C/EBPalpha transcriptional activities. Collectively, the data indicate that Nnat enhances CREB phosphorylation through increasing intracellular free calcium levels, which potentiates expression of adipogenic transcription factors resulting in heightened adipocyte differentiation. These findings contribute to a greater fundamental understanding of obesity, a clinically important risk factor in numerous diseases.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Extracellular Fluid/metabolism , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Biochem Biophys Res Commun ; 325(1): 320-7, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15522236

ABSTRACT

It has been recently shown that the expression of various types of neurotransmitter receptors is not restricted to neurons but also observed in a majority of glial cells. However, their function in glial cells is not known well in both physiological and pathological conditions. Here, we investigated the role of glutamate receptor on c-fos gene expression in primary cultured and BV-2 microglia. Our results demonstrated that both c-fos mRNA and protein were dramatically induced following treatment with various glutamate receptor agonists (500muM); N-methyl-d-aspartic acid, kainic acid, (S)-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and (RS)-3,5-dihydroxyphenylglycine. The responses were significantly suppressed by specific antagonists and also by calcium chelating agents EGTA and BAPTA-AM. Our results suggest that glutamate receptor activation regulates c-fos gene expression by modifying intracellular calcium levels in microglia. These findings might provide an insight in to understanding the function of microglial glutamate receptors in neuron-to-glial interaction under the excitotoxic conditions.


Subject(s)
Gene Expression Regulation , Microglia/physiology , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Glutamate/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Chelating Agents/metabolism , Chelating Agents/pharmacology , Excitatory Amino Acid Agonists/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Mice , Microglia/cytology , Microglia/drug effects , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...