Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Biophys Res Commun ; 495(1): 1214-1220, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180007

ABSTRACT

The high concentrations of nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) in activated glial cells in response to neuroinflammatory stimuli have neurotoxic effects on the brain. At basal levels, iNOS expression is low, and proinflammatory stimuli induce iNOS expression in astrocytes, microglia, and oligodendrocytes. Fyn, a non-receptor tyrosine kinase, regulates iNOS expression in several types of immune cells. However, its role in stimulated astrocytes is less clear. In this study, we investigated the role of Fyn in the regulation of lipopolysaccharide (LPS)-induced iNOS expression in astrocytes from mice and rats. Intracerebroventricular LPS injections in cortical regions enhanced iNOS mRNA and protein levels, which were increased in Fyn-deficient mice. Accordingly, LPS-induced nitrite production was enhanced in primary astrocytes cultured from Fyn-deficient mice or rats. Similar results were observed in cultured astrocytes after the siRNA-induced knockdown of Fyn expression. Finally, we observed increased LPS-induced extracellular signal-regulated protein kinase (ERK) activation in Fyn-deficient astrocytes. These results suggested that Fyn has a regulatory role in iNOS expression in astrocytes during neuroinflammatory responses.


Subject(s)
Astrocytes/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Gene Expression Regulation, Enzymologic/immunology , Inflammation Mediators/immunology , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/immunology , Proto-Oncogene Proteins c-fyn/immunology , Animals , Astrocytes/drug effects , Cells, Cultured , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
2.
Int J Mol Sci ; 16(5): 9217-35, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25915032

ABSTRACT

The aim of this study was to investigate the effect of ethyl acetate fraction of Euryale ferox seed extracts (Efse-EA) on melanogenesis in immortalized mouse melanocyte cell line, melan-a. Efse-EA showed strong dose-dependent mushroom tyrosinase inhibitory activity. Treatment of melan-a cells with 30 µg/mL Efse-EA produced strong inhibition of cellular tyrosinase and melanin synthesis. Efse-EA significantly reduced the levels of melanogenesis-related proteins, such as tyrosinase, tyrosinase-related proteins 1 and 2, and microphthalmia-associated transcription factor. Because Efse-EA treatment reduced tyrosinase protein levels without changing its mRNA expression, we investigated whether this decrease was related to proteasomal or lysosomal degradation of tyrosinase. We found that chloroquine, a lysosomal proteolysis inhibitor, almost completely abolished both the down-regulation of tyrosinase and the inhibition of melanin synthesis induced by Efse-EA. These results suggested that Efse-EA may contribute to the inhibition of melanogenesis by altering lysosomal degradation of tyrosinase, and that this extract may provide a new cosmetic skin-whitening agent.


Subject(s)
Lysosomes/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Plant Extracts/pharmacology , Seeds/chemistry , Streptophyta/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Chelating Agents/chemistry , Chelating Agents/pharmacology , Copper , Dose-Response Relationship, Drug , Levodopa/metabolism , Melanins/metabolism , Mice , Monophenol Monooxygenase/antagonists & inhibitors , Plant Extracts/chemistry
3.
J Neurosci Res ; 92(5): 658-70, 2014 May.
Article in English | MEDLINE | ID: mdl-24510599

ABSTRACT

Preconception exposure to EtOH through the paternal route may affect neurobehavioral and developmental features of offspring. This study investigates the effects of paternal exposure to EtOH before conception on the hyperactivity, inattention, and impulsivity behavior of male offspring in mice. Sire mice were treated with EtOH in a concentration range approximating human binge drinking (0-4 g/kg/day EtOH) for 7 weeks and mated with untreated females mice to produce offspring. EtOH exposure to sire mice induced attention deficit hyperactivity disorder (ADHD)-like hyperactive, inattentive, and impulsive behaviors in offspring. As a mechanistic link, both protein and mRNA expression of dopamine transporter (DAT), a key determinant of ADHD-like phenotypes in experimental animals and humans, were significantly decreased by paternal EtOH exposure in cerebral cortex and striatum of offspring mice along with increased methylation of a CpG region of the DAT gene promoter. The increase in methylation of DAT gene promoter was also observed in the sperm of sire mice, suggesting germline changes in the epigenetic methylation signature of DAT gene by EtOH exposure. In addition, the expression of two key regulators of methylation-dependent epigenetic regulation of functional gene expression, namely, MeCP2 and DNMT1, was markedly decreased in offspring cortex and striatum sired by EtOH-exposed mice. These results suggest that preconceptional exposure to EtOH through the paternal route induces behavioral changes in offspring, possibly via epigenetic changes in gene expression, which is essential for the regulation of ADHD-like behaviors.


Subject(s)
Attention Deficit Disorder with Hyperactivity/chemically induced , Central Nervous System Depressants/toxicity , Dopamine Plasma Membrane Transport Proteins/metabolism , Epigenesis, Genetic/drug effects , Ethanol/toxicity , Prenatal Exposure Delayed Effects/physiopathology , Animals , Avoidance Learning/physiology , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Drinking Behavior , Exploratory Behavior/physiology , Female , Gene Expression Regulation/drug effects , Male , Maze Learning/physiology , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred ICR , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced
4.
Cell Mol Neurobiol ; 34(2): 297-305, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24338128

ABSTRACT

Fragile X mental retardation protein (FMRP) is encoded by Fmr1 gene in which mutation is known to cause fragile X syndrome characterized by mental impairment and other psychiatric symptoms similar to autism spectrum disorders. FMRP plays important roles in cellular mRNA biology such as transport, stability, and translation as an RNA-binding protein. In the present study, we identified potential role of FMRP in the neural differentiation, using cortical neural progenitor cells from Sprague-Dawley rat. We newly found NeuroD1, an essential regulator of glutamatergic neuronal differentiation, as a new mRNA target interacting with FMRP in co-immunoprecipitation experiments. We also identified FMRP as a regulator of neuronal differentiation by modulating NeuroD1 expression. Down-regulation of FMRP by siRNA also increased NeuroD1 expression along with increased pre- and post-synaptic development of glutamatergic neuron, as evidenced by Western blot and immunocytochemistry. On the contrary, cells harboring FMRP over-expression construct showed decreased NeuroD1 expression. Treatment of cultured neural precursor cells with a histone deacetylase inhibitor, valproic acid known as an inducer of hyper-glutamatergic neuronal differentiation, down-regulated the expression of FMRP, and induced NeuroD1 expression. Our study suggests that modulation of FMRP expression regulates neuronal differentiation by interaction with its binding target mRNA, and provides an example of the gene and environmental interaction regulating glutamatergic neuronal differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neurons/cytology , Protein Biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glutamates/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Protein Binding/drug effects , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism , Valproic Acid/pharmacology
5.
Glia ; 61(5): 694-709, 2013 May.
Article in English | MEDLINE | ID: mdl-23378038

ABSTRACT

Tissue plasminogen activator (tPA) is expressed in several regions of brain and plays regulatory roles such as neurite outgrowth, synaptic plasticity and long term potentiation. The activity of tPA is regulated by an endogenous inhibitor plasminogen activator inhibitor-1 (PAI-1), which is expressed mainly in astrocytes. Valproic acid (VPA), a histone deacetylase inhibitor that is used for the treatment of epilepsy and bipolar disorders, promotes neurite extension, neuronal growth and has neuroprotective effect in neurodegenerative diseases. In this study, we examined whether the neurite extension effects of VPA is mediated by modulating tPA/PAI-1 system. VPA dose-dependently increased tPA activity and decreased PAI-1 activity in rat primary astrocytes but not in neurons. PAI-1 protein level secreted into the culture medium but not tPA per se was decreased by VPA. In co-culture system or in neuronal culture stimulated with astrocyte conditioned media but not in pure neuronal cell culture, VPA induced neurite outgrowth via increased tPA activity due to the decreased PAI-1 activity in astrocytes. The decrease in PAI-1 activity and increased neurite extension was regulated via JNK mediated post-transcriptional pathway. The essential role of tPA/PAI-1 system in the regulation of VPA-mediated neurite extension was further demonstrated by experiments using astrocyte conditioned media obtained from tPA or PAI-1 knockout mice. Regulation of PAI-1 activity in astrocyte by VPA may affect both physiological and pathological processes in brain by upregulating tPA activity.


Subject(s)
Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Cortex/cytology , Neurites/drug effects , Neurites/physiology , Plasminogen Activator Inhibitor 1/metabolism , Tissue Plasminogen Activator/metabolism , Valproic Acid/pharmacology , Animals , Astrocytes/pathology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Coculture Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Up-Regulation/physiology
6.
J Neurochem ; 124(6): 832-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23311691

ABSTRACT

Autism spectrum disorder (ASD) is a pervasive developmental disorder characterized by three main behavioral symptoms including social deficits, impaired communication, and stereotyped and repetitive behaviors. ASD prevalence shows gender bias to male. Prenatal exposure to valproic acid (VPA), a drug used in epilepsy and bipolar disorder, induces autistic symptoms in both human and rodents. As we reported previously, prenatally VPA-exposed animals at E12 showed impairment in social behavior without any overt reproductive toxicity. Social interactions were not significantly different between male and female rats in control condition. However, VPA-exposed male offspring showed significantly impaired social interaction while female offspring showed only marginal deficits in social interaction. Similar male inclination was observed in hyperactivity behavior induced by VPA. In addition to the ASD-like behavioral phenotype, prenatally VPA-exposed rat offspring shows crooked tail phenotype, which was not different between male and female groups. Both male and female rat showed reduced GABAergic neuronal marker GAD and increased glutamatergic neuronal marker vGluT1 expression. Interestingly, despite of the similar increased expression of vGluT1, post-synaptic marker proteins such as PSD-95 and α-CAMKII expression was significantly elevated only in male offspring. Electron microscopy showed increased number of post-synapse in male but not in female at 4 weeks of age. These results might suggest that the altered glutamatergic neuronal differentiation leads to deranged post-synaptic maturation only in male offspring prenatally exposed to VPA. Consistent with the increased post-synaptic compartment, VPA-exposed male rats showed higher sensitivity to electric shock than VPA-exposed female rats. These results suggest that prenatally VPA-exposed rats show the male preponderance of ASD-like behaviors including defective social interaction similar to human autistic patients, which might be caused by ectopic increase in glutamatergic synapses in male rats.


Subject(s)
Child Development Disorders, Pervasive/psychology , Disease Models, Animal , Interpersonal Relations , Sex Characteristics , Synapses/drug effects , Valproic Acid/toxicity , Animals , Child , Child Development Disorders, Pervasive/chemically induced , Child Development Disorders, Pervasive/pathology , Female , Humans , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology , Random Allocation , Rats , Rats, Sprague-Dawley , Synapses/pathology , Synapses/ultrastructure , Synaptic Potentials
7.
Neurochem Res ; 38(9): 1960-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824559

ABSTRACT

Cytoplasmic polyadenylation binding protein 1 (CPEB1) is a RNA binding protein, which regulates translation of target mRNAs by regulating polyadenylation status. CPEB1 plays important roles in the regulation of germline cell development by modulating cell cycle progression through the polyadenylation of target mRNAs such as cyclin B1. Similar mechanism is reported in proliferating astrocytes by us, although CPEB1 is involved in the transport of target mRNAs as well as local translation at dendritic spines. In this study, we found the expression of CPEB1 in cultured rat primary neural progenitor cells (NPCs). EGF stimulation of cultured NPCs induced rapid phosphorylation of CPEB1, a hallmark of CPEB1-dependent translational control along with cyclin B1 polyadenylation and translation. EGF-induced activation of ERK1/2 and Aurora A kinase was responsible for CPEB1 phosphorylation. Pharmacological inhibition studies suggested that ERK1/2 is involved in the activation of Aurora A kinase and regulation of CPEB1 phosphorylation in cultured NPCs. Long-term incubation in EGF resulted in the down-regulation of CPEB1 expression, which further increased expression of cyclin B1 and cell cycle progression. When we down-regulated the expression of CPEB1 in NPCs by siRNA transfection, the proliferation of NPCs was increased. Increased NPCs proliferation by down-regulation of CPEB1 resulted in eventual up-regulation of neuronal differentiation with increase in both pre- and post-synaptic proteins. The results from the present study may suggest the importance of translational control in the regulation of neuronal development, an emerging concept in many neurodevelopmental and psychiatric disorders such as autism spectrum disorder.


Subject(s)
Cell Proliferation , Neural Stem Cells/cytology , Neurons/cytology , RNA-Binding Proteins/physiology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Female , Pregnancy , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
8.
Neurochem Res ; 38(3): 620-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283698

ABSTRACT

Prenatal exposure to alcohol has consistently been associated with adverse effects on neurodevelopment, which is collectively called fetal alcohol spectrum disorder (FASD). Increasing evidence suggest that prenatal exposure to alcohol increases the risk of developing attention deficit/hyperactivity disorder-like behavior in human. In this study, we investigated the behavioral effects of prenatal exposure to EtOH in offspring mice and rats focusing on hyperactivity and impulsivity. We also examined changes in dopamine transporter and MeCP2 expression, which may underlie as a key neurobiological and epigenetic determinant in FASD and hyperactive, inattentive and impulsive behaviors. Mouse or rat offspring born from dam exposed to alcohol during pregnancy (EtOH group) showed hyper locomotive activity, attention deficit and impulsivity. EtOH group also showed increased dopamine transporter and norepinephrine transporter level compared to control group in the prefrontal cortex and striatum. Prenatal exposure to EtOH also significantly decreased the expression of MeCP2 in both prefrontal cortex and striatum. These results suggest that prenatal exposure to EtOH induces hyperactive, inattentive and impulsive behaviors in rodent offspring that might be related to global epigenetic changes as well as aberration in catecholamine neurotransmitter transporter system.


Subject(s)
Ethanol/toxicity , Animals , Attention Deficit Disorder with Hyperactivity/chemically induced , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Dopamine Plasma Membrane Transport Proteins/biosynthesis , Epigenesis, Genetic/drug effects , Female , Fetal Alcohol Spectrum Disorders/psychology , Impulsive Behavior/chemically induced , Methyl-CpG-Binding Protein 2/metabolism , Mice , Norepinephrine Plasma Membrane Transport Proteins/biosynthesis , Pregnancy , Prenatal Exposure Delayed Effects , Rats
9.
J Neurosci ; 31(34): 12118-28, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865454

ABSTRACT

Astrocytes are the most abundant cells in the brain, playing vital roles in neuronal survival, growth, and function. Understanding the mechanism(s) regulating astrocyte proliferation will have important implications in brain development, response to injury, and tumorigenesis. Cyclin B1 is well known to be a critical regulator of mitotic entry via its interaction with cyclin-dependent kinase 1. In rat astrocytes, we now show that the mRNA binding protein cytoplasmic polyadenylation element binding protein 1 (CPEB1) is associated with cyclin B1 mRNA and that this interaction is enriched at the centrosome. In addition, if growth-arrested astrocytes are stimulated to divide, CPEB1 is phosphorylated and cyclin B1 mRNA is polyadenylated, both hallmarks of CPEB1 activation, resulting in an increase in cyclin B1 protein. CPEB1 binding to mRNA initially inhibits translation; therefore, removing CPEB1 from mRNA should result in an increase in translation due to derepression. Indeed, when we either knocked down CPEB1 protein with siRNA or sequestered it from endogenous mRNA by expressing RNA containing multiple CPEB1 binding sites, cyclin B1 protein was increased and cell proliferation was stimulated. Our data suggest a mechanism wherein CPEB1 is bound and represses cyclin B1 mRNA translation until a signal to proliferate phosphorylates CPEB1, resulting in an increase in cyclin B1 protein and progression into mitosis. Our results demonstrate for the first time a role for CPEB1 in regulating cell proliferation in the brain.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Cyclin B1/biosynthesis , Cyclin B1/genetics , Gene Expression Regulation/physiology , Polyadenylation/genetics , RNA Stability/genetics , RNA-Binding Proteins/metabolism , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Centrosome/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Female , Male , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley
10.
Biochem Biophys Res Commun ; 409(4): 687-92, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21620800

ABSTRACT

Upon CNS damage, astrocytes undergo a series of biological changes including increased proliferation, production of inflammatory mediators and morphological changes, in a response collectively called reactive gliosis. This process is an essential part of the brains response to injury, yet much is unknown about the molecular mechanism(s) that induce these changes. In this study, we investigated the role of cytoplasmic polyadenylation element binding protein 1 (CPEB1) in the regulation of inflammatory responses in a model of reactive gliosis, lipopolysaccharide-stimulated astrocytes. CPEB1 is an mRNA-binding protein recently shown to be expressed in astrocytes that may play a role in astrocytes migration. After LPS stimulation, the expression and phosphorylation of CPEB1 was increased in rat primary astrocytes in a JNK-dependent process. siRNA-induced knockdown of CPEB1 expression inhibited the LPS-induced up-regulation of iNOS as well as NO and ROS production, a hallmark of immunological activation of astrocytes. The results from the study suggest that CPEB1 is actively involved in the regulation of inflammatory responses in astrocytes, which might provide new insights into the regulatory mechanism after brain injury.


Subject(s)
Brain Injuries/metabolism , Encephalitis/metabolism , Nitric Oxide Synthase Type II/biosynthesis , RNA-Binding Proteins/metabolism , Animals , Astrocytes , Down-Regulation , Gene Knockdown Techniques , Lipopolysaccharides , MAP Kinase Kinase 4/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Oxidative Stress , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Rats , Reactive Oxygen Species/metabolism
11.
J Biomed Sci ; 18(1): 48, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21722408

ABSTRACT

BACKGROUND: At the beginning of neurogenesis, massive brain cell death occurs and more than 50% of cells are eliminated by apoptosis along with neuronal differentiation. However, few studies were conducted so far regarding the regulation of neural progenitor cells (NPCs) death during development. Because of the physiological role of cell death during development, aberration of normal apoptotic cell death is detrimental to normal organogenesis.Apoptosis occurs in not only neuron but also in NPCs and neuroblast. When growth and survival signals such as EGF or LIF are removed, apoptosis is activated as well as the induction of differentiation. To investigate the regulation of cell death during developmental stage, it is essential to investigate the regulation of apoptosis of NPCs. METHODS: Neural progenitor cells were cultured from E14 embryonic brains of Sprague-Dawley rats. For in vivo VPA animal model, pregnant rats were treated with VPA (400 mg/kg S.C.) diluted with normal saline at E12. To analyze the cell death, we performed PI staining and PARP and caspase-3 cleavage assay. Expression level of proteins was investigated by Western blot and immunocytochemical assays. The level of mRNA expression was investigated by RT-PCR. Interaction of Bcl-XL gene promoter and NF-κB p65 was investigated by ChIP assay. RESULTS: In this study, FACS analysis, PI staining and PARP and caspase-3 cleavage assay showed that VPA protects cultured NPCs from cell death after growth factor withdrawal both in basal and staurosporine- or hydrogen peroxide-stimulated conditions. The protective effect of prenatally injected VPA was also observed in E16 embryonic brain. Treatment of VPA decreased the level of IκBα and increased the nuclear translocation of NF-κB, which subsequently enhanced expression of anti-apoptotic protein Bcl-XL. CONCLUSION: To the best of our knowledge, this is the first report to indicate the reduced death of NPCs by VPA at developmentally critical periods through the degradation of IκBα and the activation of NF-κB signaling. The reduced NPCs death might underlie the neurodevelopmental defects collectively called fetal valproate syndrome, which shows symptoms such as mental retardation and autism-like behavior.


Subject(s)
Apoptosis/drug effects , Cytoprotection , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Valproic Acid/pharmacology , bcl-X Protein/metabolism , Animals , Female , Neural Stem Cells/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
12.
J Biomed Sci ; 17: 85, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21073715

ABSTRACT

BACKGROUND: Prenatal ethanol exposure during pregnancy induces a spectrum of mental and physical disorders called fetal alcohol spectrum disorder (FASD). The central nervous system is the main organ influenced by FASD, and neurological symptoms include mental retardation, learning abnormalities, hyperactivity and seizure susceptibility in childhood along with the microcephaly. In this study, we examined whether ethanol exposure adversely affects the proliferation of NPC and de-regulates the normal ratio between glutamatergic and GABAergic neuronal differentiation using primary neural progenitor culture (NPC) and in vivo FASD models. METHODS: Neural progenitor cells were cultured from E14 embryo brain of Sprague-Dawley rat. Pregnant mice and rats were treated with ethanol (2 or 4 g/kg/day) diluted with normal saline from E7 to E16 for in vivo FASD animal models. Expression level of proteins was investigated by western blot analysis and immunocytochemical assays. MTT was used for cell viability. Proliferative activity of NPCs was identified by BrdU incorporation, immunocytochemistry and FACS analysis. RESULTS: Reduced proliferation of NPCs by ethanol was demonstrated using BrdU incorporation, immunocytochemistry and FACS analysis. In addition, ethanol induced the imbalance between glutamatergic and GABAergic neuronal differentiation via transient increase in the expression of Pax6, Ngn2 and NeuroD with concomitant decrease in the expression of Mash1. Similar pattern of expression of those transcription factors was observed using an in vivo model of FASD as well as the increased expression of PSD-95 and decreased expression of GAD67. CONCLUSIONS: These results suggest that ethanol induces hyper-differentiation of glutamatergic neuron through Pax6 pathway, which may underlie the hyper-excitability phenotype such as hyperactivity or seizure susceptibility in FASD patients.


Subject(s)
Cell Differentiation/drug effects , Ethanol/pharmacology , Fetal Alcohol Spectrum Disorders/physiopathology , Glutamic Acid/metabolism , Neural Stem Cells , Neurons/physiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Eye Proteins/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Neurogenesis/drug effects , Neurons/cytology , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Repressor Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
13.
Neuroimmunomodulation ; 16(6): 365-76, 2009.
Article in English | MEDLINE | ID: mdl-19609085

ABSTRACT

Neural progenitor cells (NPC) are self-renewing multipotent cells that generate neurons and glial cells in the brain. NPCs generate neurons and glia not only during development but also after neural injury. Recent studies have shown that endogenous NPCs are activated after brain injury and migrate toward damaged areas where astrocyte activation occurs. Considering the massive proliferation of astrocytes as well as the production of several kinds of cytoactive molecules after brain injury, such as NO, growth factors and cytokines, it is tempting to think that cytoactive molecules released by activated glial cells regulate neural progenitor differentiation and proliferation through inflammatory mediators. To test this hypothesis, we stimulated rat primary astrocytes with lipopolysaccharide (LPS) to induce the activation of astrocytes. After addition of the conditioned media from LPS-stimulated astrocytes to NPC culture, proliferation was examined by MTT assay and bromodeoxyuridine (BrdU) incorporation. The differentiation of NPC into neurons and astrocytes was examined by Western blot, ELISA and immunocytochemical staining with cell-type-specific markers. Conditioned media from LPS-stimulated astrocytes increased NPC proliferation as well as gliogenesis as compared with control conditioned media from astrocytes without LPS stimulation. In contrast, neurogenesis was decreased by LPS-conditioned media. To investigate the molecular mechanism mediating glial differentiation and proliferation of NPC by reactive astrocytes, we added inhibitors of the Erk and JNK pathways during LPS stimulation. These inhibitors - except for a p38 inhibitor - decreased NPC proliferation and glial differentiation. These results suggest that LPS stimulated astrocytes generate factors regulating NPC proliferation and gliogenesis via the Erk and JNK pathways.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cytokines/metabolism , Neurogenesis/physiology , Stem Cells/physiology , Animals , Astrocytes/cytology , Bromodeoxyuridine , Cell Communication/physiology , Cell Culture Techniques , Cells, Cultured , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Indicators and Reagents , Inflammation Mediators , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Tetrazolium Salts , Up-Regulation/physiology
14.
Biotechnol J ; 14(7): e1800606, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30927490

ABSTRACT

Active hemostatic agents can play a crucial role in saving patients' lives during surgery. Active hemostats have several advantages including utilization of natural blood coagulation and biocompatibility. Among them, although human neutrophil peptide-1 (HNP-1) has been previously reported with the hemostatic mechanism, which part of HNP-1 facilitates the hemostatic activity is not known. Here, a partial peptide (HNP-F) promoting hemostasis, originating from HNP-1, has been newly identified by the blood coagulation ability test. HNP-F shows the best hemostatic effect between the anterior half and posterior half of peptides. Moreover, microscopic images show platelet aggregation and an increase in the concentration of platelet factor 4, and the scanning electron microscope image of platelets support platelet activation by HNP-F. Thromboelastography indicates decreased clotting time and increased physical properties of blood clotting. Mouse liver experiments demonstrate improved hemostatic effect by treatment of peptide solution. Cell viability and hemolysis assays confirm the HNP-F's biosafety. It is hypothesized that the surface charge and structure of HNP-F could be favorable to interact with fibrinogen or thrombospondin-1. Collectively, because HNP-F as an active peptide hemostat has many advantages, it could be expected to become a potent hemostatic biomaterial, additive or pharmaceutical candidate for various hemostatic applications.


Subject(s)
Hemostasis/drug effects , alpha-Defensins , Animals , Cell Survival/drug effects , Hemolysis/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Thrombelastography , alpha-Defensins/chemistry , alpha-Defensins/genetics , alpha-Defensins/pharmacology
15.
Neuropsychopharmacology ; 44(2): 314-323, 2019 01.
Article in English | MEDLINE | ID: mdl-29899405

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopmental disorder, featuring social communication deficit and repetitive/restricted behaviors as common symptoms. Its prevalence has continuously increased, but, till now, there are no therapeutic approaches to relieve the core symptoms, particularly social deficit. In previous studies, abnormal function of the glutamatergic neural system has been proposed as a critical mediator and therapeutic target of ASD-associated symptoms. Here, we investigated the possible roles of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in autism symptoms using two well-known autistic animal models, Cntnap2 knockout (KO) mice and in utero valproic acid-exposed ICR (VPA) mice. We found that Cntnap2 KO mice displayed decreased glutamate receptor expression and transmission. Contrarily, VPA mice exhibited increased glutamate receptor expression and transmission. Next, we investigated whether AMPAR modulators (positive-allosteric-modulator for Cntnap2 KO mice and antagonist for VPA mice) can improve autistic symptoms by normalizing the aberrant excitatory transmission in the respective animal models. Interestingly, the AMPAR modulation specifically ameliorated social deficits in both animal models. These results indicated that AMPAR-derived excitatory neural transmission changes can affect normal social behavior. To validate this, we injected an AMPAR agonist or antagonist in control ICR mice and, interestingly, these treatments impaired only the social behavior, without affecting the repetitive and hyperactive behaviors. Collectively, these results provide insight into the role of AMPARs in the underlying pathophysiological mechanisms of ASD, and demonstrate that modulation of AMPAR can be a potential target for the treatment of social behavior deficits associated with ASD.


Subject(s)
Autism Spectrum Disorder/drug therapy , Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Receptors, AMPA/antagonists & inhibitors , Social Behavior , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/genetics , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Play and Playthings , Pregnancy , Prenatal Exposure Delayed Effects , Receptors, N-Methyl-D-Aspartate/metabolism , Valproic Acid
16.
Mol Cells ; 41(5): 486-494, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29696935

ABSTRACT

Recently, we have reported that animals with telomerase reverse transcriptase (TERT) overexpression exhibit reduced social interaction, decreased preference for novel social interaction and poor nest-building behaviors symptoms that mirror those observed in human autism spectrum disorders (ASD). Overexpression of TERT also alters the excitatory/inhibitory (E/I) ratio in the medial prefrontal cortex. However, the effects of TERT overexpression on hippocampal-dependent learning and synaptic efficacy have not been investigated. In the present study, we employed electrophysiological approaches in combination with behavioral analysis to examine hippocampal function of TERT transgenic (TERT-tg) mice and FVB controls. We found that TERT overexpression results in enhanced hippocampal excitation with no changes in inhibition and significantly impairs long-term synaptic plasticity. Interestingly, the expression levels of phosphorylated CREB and phosphory-lated CaMKIIα were significantly decreased while the expression level of CaMKIIα was slightly increased in the hippocampus of TERT-overexpressing mice. Our observations highlight the importance of TERT in normal synaptic function and behavior and provide additional information on a novel animal model of ASD associated with TERT overexpression.


Subject(s)
Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Neuronal Plasticity , Pyramidal Cells/physiology , Synaptic Transmission , Telomerase/physiology , Animals , Autism Spectrum Disorder/enzymology , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/genetics , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression , Hippocampus/enzymology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurotoxins/pharmacology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Recombinant Proteins/metabolism , Synaptic Transmission/drug effects , Telomerase/genetics , Tetrodotoxin/pharmacology
17.
Biomol Ther (Seoul) ; 26(5): 439-445, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29463073

ABSTRACT

T-type calcium channels are low voltage-activated calcium channels that evoke small and transient calcium currents. Recently, T-type calcium channels have been implicated in neurodevelopmental disorders such as autism spectrum disorder and neural tube defects. However, their function during embryonic development is largely unknown. Here, we investigated the function and expression of T-type calcium channels in embryonic neural progenitor cells (NPCs). First, we compared the expression of T-type calcium channel subtypes (CaV3.1, 3.2, and 3.3) in NPCs and differentiated neural cells (neurons and astrocytes). We detected all subtypes in neurons but not in astrocytes. In NPCs, CaV3.1 was the dominant subtype, whereas CaV3.2 was weakly expressed, and CaV3.3 was not detected. Next, we determined CaV3.1 expression levels in the cortex during early brain development. Expression levels of CaV3.1 in the embryonic period were transiently decreased during the perinatal period and increased at postnatal day 11. We then pharmacologically blocked T-type calcium channels to determine the effects in neuronal cells. The blockade of T-type calcium channels reduced cell viability, and induced apoptotic cell death in NPCs but not in differentiated astrocytes. Furthermore, blocking T-type calcium channels rapidly reduced AKT-phosphorylation (Ser473) and GSK3ß-phosphorylation (Ser9). Our results suggest that T-type calcium channels play essential roles in maintaining NPC viability, and T-type calcium channel blockers are toxic to embryonic neural cells, and may potentially be responsible for neurodevelopmental disorders.

18.
Neuropharmacology ; 113(Pt A): 71-81, 2017 02.
Article in English | MEDLINE | ID: mdl-27638451

ABSTRACT

Autism spectrum disorder (ASD) is an immensely challenging developmental disorder characterized primarily by two core behavioral symptoms of social communication deficits and restricted/repetitive behaviors. Investigating the etiological process and identifying an appropriate therapeutic target remain as formidable challenges to overcome ASD due to numerous risk factors and complex symptoms associated with the disorder. Among the various mechanisms that contribute to ASD, the maintenance of excitation and inhibition balance emerged as a key factor to regulate proper functioning of neuronal circuitry. Interestingly, our previous study involving the valproic acid animal model of autism (VPA animal model) has demonstrated excitatory-inhibitory imbalance (E/I imbalance) due to enhanced differentiation of glutamatergic neurons and reduced GABAergic neurons. Here, we investigated the potential of agmatine, an endogenous NMDA receptor antagonist, as a novel therapeutic candidate in ameliorating ASD symptoms by modulating E/I imbalance using the VPA animal model. We observed that a single treatment of agmatine rescued the impaired social behaviors as well as hyperactive and repetitive behaviors in the VPA animal model. We also observed that agmatine treatment rescued the overly activated ERK1/2 signaling in the prefrontal cortex and hippocampus of VPA animal models, possibly, by modulating over-excitability due to enhanced excitatory neural circuit. Taken together, our results have provided experimental evidence suggesting a possible therapeutic role of agmatine in ameliorating ASD-like symptoms in the VPA animal model of ASD.


Subject(s)
Agmatine/administration & dosage , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Agmatine/therapeutic use , Animals , Autism Spectrum Disorder/chemically induced , Disease Models, Animal , Grooming/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hyperkinesis/prevention & control , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Transgenic , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/prevention & control , Social Behavior , Valproic Acid
20.
Exp Neurobiol ; 26(5): 252-265, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29093634

ABSTRACT

The valproic acid (VPA)-induced animal model is one of the most widely utilized environmental risk factor models of autism. Autism spectrum disorder (ASD) remains an insurmountable challenge among neurodevelopmental disorders due to its heterogeneity, unresolved pathological pathways and lack of treatment. We previously reported that VPA-exposed rats and cultured rat primary neurons have increased Pax6 expression during post-midterm embryonic development which led to the sequential upregulation of glutamatergic neuronal markers. In this study, we provide experimental evidence that telomerase reverse transcriptase (TERT), a protein component of ribonucleoproteins complex of telomerase, is involved in the abnormal components caused by VPA in addition to Pax6 and its downstream signals. In embryonic rat brains and cultured rat primary neural progenitor cells (NPCs), VPA induced the increased expression of TERT as revealed by Western blot, RT-PCR, and immunostainings. The HDAC inhibitor property of VPA is responsible for the TERT upregulation. Chromatin immunoprecipitation revealed that VPA increased the histone acetylation but blocked the HDAC1 binding to both Pax6 and Tert genes. Interestingly, the VPA-induced TERT overexpression resulted to sequential upregulations of glutamatergic markers such as Ngn2 and NeuroD1, and inter-synaptic markers such as PSD-95, α-CaMKII, vGluT1 and synaptophysin. Transfection of Tert siRNA reversed the effects of VPA in cultured NPCs confirming the direct involvement of TERT in the expression of those markers. This study suggests the involvement of TERT in the VPA-induced autistic phenotypes and has important implications for the role of TERT as a modulator of balanced neuronal development and transmission in the brain.

SELECTION OF CITATIONS
SEARCH DETAIL