Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
ACS Chem Neurosci ; 7(11): 1488-1498, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27442785

ABSTRACT

Nerve growth factor (NGF)-induced neuronal regeneration has emerged as a strategy to treat neuronal degeneration-associated disorders. However, direct NGF administration is limited by the occurrence of adverse effects at high doses of NGF. Therefore, development of a therapeutic strategy to promote the NGF trophic effect is required. In view of the lack of understanding of the mechanism for potentiating the NGF effect, this study investigated molecular targets of DA-9801, a well-standardized Dioscorea rhizome extract, which has a promoting effect on NGF. An increase in intracellular calcium ion level was induced by DA-9801, and chelation of extracellular calcium ions with ethylene-bis(oxyethylenenitrilo)tetraacetic acid (EGTA) suppressed the potentiating effect of DA-9801 on NGF-induced neurite outgrowth. In addition, EGTA treatment reduced the DA-9801-induced phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2), the major mediators of neurite outgrowth. To find which calcium ion-permeable channel contributes to the calcium ion influx induced by DA-9801, we treated PC12 cells with various inhibitors of calcium ion-permeable channels. NF449, a P2X1 receptor selective antagonist, significantly abolished the potentiating effect of DA-9801 on NGF-induced neurite outgrowth and abrogated the DA-9801-induced ERK1/2 phosphorylation. In addition, transfection with siRNA of P2X1 receptor significantly reduced the DA-9801-enhanced neurite outgrowth. In conclusion, calcium ion influx through P2X1 receptor mediated the promoting effect of DA-9801 on NGF-induced neurite outgrowth via ERK1/2 phosphorylation.


Subject(s)
Nerve Growth Factor/metabolism , Neurites/drug effects , Neuronal Outgrowth/drug effects , Plant Preparations/pharmacology , Purinergic Agents/pharmacology , Receptors, Purinergic P2X1/metabolism , Animals , Calcium/metabolism , Calcium Chelating Agents/pharmacology , Cations, Divalent/metabolism , Egtazic Acid/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurites/physiology , Neuronal Outgrowth/physiology , Neuroprotective Agents/pharmacology , PC12 Cells , Phosphorylation/drug effects , RNA, Small Interfering , Rats , Receptors, Purinergic P2X1/genetics
2.
Mol Neurobiol ; 49(1): 512-28, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24030726

ABSTRACT

Imbalance in excitatory/inhibitory signal in the brain has been proposed as one of the main pathological features in autism spectrum disorders, although the underlying cellular and molecular mechanism is unclear yet. Because excitatory/inhibitory imbalance can be induced by aberration in glutamatergic/GABAergic neuronal differentiation, we investigated the mechanism of dysregulated neuronal differentiation between excitatory and inhibitory neurons in the embryonic and postnatal brain of prenatally valproic acid-exposed rat offspring, which is often used as an animal model of autism spectrum disorders. Transcription factor Pax6, implicated in glutamatergic neuronal differentiation, was transiently increased in embryonic cortex by valproate exposure, which resulted in the increased expression of glutamatergic proteins in postnatal brain of offspring. Chromatin immunoprecipitation showed increased acetylated histone binding on Pax6 promoter region, which may underlie the transcriptional up-regulation of Pax6. Other histone deacetylase (HDAC) inhibitors including TSA and SB but not valpromide, which is devoid of HDAC inhibitor activity, induced Pax6 up-regulation. Silencing Pax6 expression in cultured rat primary neural progenitor cells demonstrated that up-regulation of Pax6 plays an essential role in valproate-induced glutamatergic differentiation. Blocking glutamatergic transmission with MK-801 or memantine treatment, and to a lesser extent with MPEP treatment, reversed the impaired social behaviors and seizure susceptibility of prenatally valproate-exposed offspring. Together, environmental factors may contribute to the imbalance in excitatory/inhibitory neuronal activity in autistic brain by altering expression of transcription factors governing glutamatergic/GABAergic differentiation during fetal neural development, in conjunction with the genetic preload.


Subject(s)
Autistic Disorder/metabolism , Cerebral Cortex/metabolism , Eye Proteins/physiology , Glutamic Acid/physiology , Homeodomain Proteins/physiology , Paired Box Transcription Factors/physiology , Prenatal Exposure Delayed Effects/metabolism , Repressor Proteins/physiology , Valproic Acid/toxicity , Animals , Autistic Disorder/chemically induced , Autistic Disorder/pathology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Eye Proteins/biosynthesis , Female , Homeodomain Proteins/biosynthesis , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Repressor Proteins/biosynthesis , Up-Regulation/physiology
3.
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
4.
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
5.
Neuropharmacology ; 63(6): 1028-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22841957

ABSTRACT

Autism is a spectrum of neurodevelopmental disorders characterized by social isolation and lack of interaction. Anatomically, autism patients often show macrocephaly and high neuronal density. To investigate the mechanism underlying the higher neuronal populations seen in ASD, we subcutaneously injected VPA (400 mg/kg) into pregnant Sprague-Dawley rats on E12, an animal model often used in ASD study. Alternatively, cultured rat neural progenitor cells were treated with VPA. Until E18, VPA induced NPC proliferation and delayed neurogenesis in fetal brain, but the subsequent differentiation of NPCs to neurons increased brain neuronal density afterward. Similar findings were observed with NPCs treated with VPA in vitro. At a molecular level, VPA enhanced Wnt1 expression and activated the GSK-3ß/ß-catenin pathway. Furthermore, inhibition of this pathway attenuated the effects of VPA. The findings of this study suggest that an altered developmental process underlies the macrocephaly and abnormal brain structure observed in the autistic brain.


Subject(s)
Anticonvulsants/toxicity , Glycogen Synthase Kinase 3/physiology , Megalencephaly/chemically induced , Neural Stem Cells/drug effects , Valproic Acid/toxicity , beta Catenin/physiology , Animals , Antimetabolites , Blotting, Western , Bromodeoxyuridine , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coloring Agents , Female , Flow Cytometry , Glycogen Synthase Kinase 3/genetics , Immunohistochemistry , Megalencephaly/pathology , Organ Size/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tetrazolium Salts , Thiazoles , Transfection , beta Catenin/genetics
6.
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
7.
Toxicol Lett ; 201(2): 137-42, 2011 Mar 05.
Article in English | MEDLINE | ID: mdl-21195144

ABSTRACT

Prenatal exposure to valproic acid (VPA) induces neural tube defects and impairment in social behaviors related to autistic spectrum disorder in newborns, which make it a useful animal model of autism. In this study, we compared the effects of different time window of prenatal valproic acid exposure for inducing the altered social behaviors relevant to autism from embryonic day 7 to embryonic day 15 in Sprague-Dawley rats to determine the critical periods for the impairment. Compared to E7, E9.5 and E15 exposure, VPA exposure at E12 showed most significant changes in behaviors over control animals with reduced sociability and social preference. E9.5 exposure to valproic acid showed strong reproductive toxicity including decrease in the number of live birth. In general, exposure at E15 showed only marginal effects on reproduction and social behaviors. Finally, VPA-exposed rats at E12 were more sensitive to electric shock than VPA-exposed rats at any other periods. These results suggested that E12 is the critical period in rats when valproate exposure has prominent effects for inducing the altered social behavior similar to human autistic behavior.


Subject(s)
Anticonvulsants/toxicity , Autistic Disorder/chemically induced , Fetus/drug effects , Valproic Acid/toxicity , Animals , Disease Models, Animal , Electroshock , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Neural Tube Defects/chemically induced , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, GABA/physiology , Social Behavior
8.
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
9.
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
10.
Brain Res Bull ; 76(4): 368-75, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18502312

ABSTRACT

Thrombin plays an important role in diverse neurological processes such as proliferation, cell migration, differentiation and neuroinflammation. In this study, we investigated the effect of thrombin on matrix metalloprotease-9 (MMP-9) expression in rat primary astrocytes. Thrombin (1-10U/ml) induced a significant increase in MMP-9 activity as measured by gelatin zymography. Thrombin also increased MMP-9 mRNA expression. Among three isotypes of thrombin receptor, i.e. protease-activated receptor (PAR)-1, -3 and -4, PAR1 agonist (1-100muM) but not PAR3 and PAR4 agonist induced MMP-9 expression. Inhibition of thrombin-induced MMP-9 production by SCH 79797 (10-50nM), a selective PAR1 receptor antagonist, confirmed that PAR1 is a main receptor for thrombin-induced MMP-9 expression. In astrocytes, thrombin activated Erk1/2, and it was inhibited by PD98059. In this study, thrombin-induced MMP-9 expression was inhibited by PD98059. PAR1 agonist activated Erk1/2 and PD98059 inhibited PAR1 agonist-induced MMP-9 expression. MMP-9 promoter reporter assay confirmed the positive effect of ERK1/2 on MMP-9 expression. These results suggest that the activation of PAR1 mediates thrombin-induced MMP-9 expression through the regulation of Erk1/2.


Subject(s)
Astrocytes/metabolism , Matrix Metalloproteinase 9/metabolism , Receptor, PAR-1/metabolism , Thrombin/metabolism , Animals , Astrocytes/drug effects , Cells, Cultured , Encephalitis/metabolism , Encephalitis/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Flavonoids/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , Rats , Receptor, PAR-1/agonists , Receptor, PAR-1/antagonists & inhibitors , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...