Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
Child Health Nurs Res ; 26(2): 296-308, 2020 Apr.
Article in Korean | MEDLINE | ID: mdl-35004473

ABSTRACT

PURPOSE: The purpose of this study was to identify the noise level and frequency experienced by premature infants receiving incubator care in the neonatal intensive care unit (NICU). METHODS: The participants were 20 premature infants receiving incubator care in the NICU of a university hospital in Daejeon Metropolitan city. The noise level was measured using a professional sound-level meter (ET-958, FLUS, Shenzhen, China) based on a noise classification table developed by the author. The data were analyzed with descriptive statistics, the t-test, analysis of variance, and Pearson correlation coefficients using SPSS for Windows version 22.0. RESULTS: The average noise level experienced by premature infants receiving incubator care in the NICU was 51.25 dB (range: 45.0~81.7 dB). The frequency of noises was highest for factors related to nursing activities (40.3%), followed by human factors (29.1%), machine alarm sounds (20.1%), incubator operation (6.6%), and internal environmental factors (3.9%). CONCLUSION: According to the above results, the noise level experienced by premature infants receiving incubator care in the NICU exceeded the recommendations of the American Academy of Pediatrics. Therefore, it is necessary to develop an interventional program to reduce noise in the NICU, and to conduct follow-up studies to verify its effectiveness.

3.
Arch Pharm Res ; 38(4): 556-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25030857

ABSTRACT

Propofol, a widely used anesthetic, regulates neurological processes including neurotoxicity, neuroprotection, glial activation, synaptic plasticity and neuronal maturation. Tissue plasminogen activator/tissue plasminogen activator inhibitor-1 (tPA/PAI-1) in CNS acts as a neuromodulator regulating synaptic plasticity, neurite outgrowth, seizure spreading and cell survival. Here, we investigated the effects of propofol on tPA/PAI-1 system using cultured neurons and astrocytes and their role in the regulation of neurite extension. Cultured rat primary astrocytes were treated with propofol (1-10 µM) and LPS (10 ng/ml). The expression of functional tPA/PAI-1 was examined by casein zymography, Western blot and RT-PCR. Alternatively, culture supernatants were added to cultured rat primary neuron to investigate the effects on neurite extension. Propofol alone did not affect tPA activity in rat primary cortical neuron. Similarly, propofol alone changed neither tPA nor PAI-1 activity in rat primary astrocytes. In immunologically challenged situation using LPS, propofol synergistically increased expression of PAI-1 in rat primary astrocytes without affecting tPA expression in a manner dependent on MAPKs activation. Increased expression of PAI-1 reduced tPA activity in LPS plus propofol-treated rat primary astrocytes. Consistent with the critical role of tPA activity in the regulation of neurite extension (Cho et al. 2013), the diminished tPA activity in astrocyte culture supernatants resulted in decreased neurite extension when administered to cultured rat primary cortical neuron. The results from the present study suggest that propofol, especially in immunologically-challenged situation, dysregulates tPA/PAI-1 system in brain. Whether the dysregulated tPA/PAI-1 activity adversely affects neural differentiation as well as regeneration of neuron in vivo should be empirically determined in the future.


Subject(s)
Astrocytes/immunology , Immunity, Cellular/immunology , Neurites/immunology , Plasminogen Activator Inhibitor 1/physiology , Propofol/pharmacology , Animals , Astrocytes/drug effects , Cells, Cultured , Immunity, Cellular/drug effects , Neurites/drug effects , Rats , Rats, Sprague-Dawley
4.
Mol Neurobiol ; 52(1): 8-25, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25106729

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is an endogenous inhibitor of tissue plasminogen activator (tPA) that acts as a neuromodulator in various neurophysiological and pathological conditions. Several researchers including us reported the induction of PAI-1 during inflammatory condition; however, the mechanism regulating PAI-1 induction is not yet clear. In this study, we investigated the role of non-receptor tyrosine kinase Fyn in the regulation of lipopolysaccharide (LPS)-induced upregulation of PAI-1 in rat primary astrocyte. The activation of toll-like receptor 4 (TLR4) signaling, induced by its ligand LPS, stimulated a physical interaction between TLR4 and Fyn along with phosphorylation of tyrosine residue in both molecules as determined by co-immunoprecipitation experiments. Immunofluorescence staining also showed increased co-localization of TLR4-Fyn on cultured rat primary astrocytes after LPS treatment. The increased TRLR4-Fyn interaction induced expression of PAI-1 through the activation of PI3k/Akt/NFĸB pathway. Treatment with Src kinase inhibitor (PP2) or transfection of Fyn small interfering RNA (siRNA) into cultured rat primary astrocytes inhibited phosphorylation of tyrosine residue of TLR4 and blocked the interaction between TLR4 and Fyn resulting to the inhibition of LPS-induced expression of PAI-1. The activation of PI3K/Akt/NFĸB signaling cascades was also inhibited by Fyn knockdown in rat primary astrocytes. The induction of PAI-1 in rat primary astrocytes, which resulted in downregulation of tPA activity in culture supernatants, inhibited neurite outgrowth in cultured rat primary cortical neuron. The inhibition of neurite extension was prevented by PP2 or Fyn siRNA treatment in rat primary astrocytes. These results suggest the critical physiological role of TRL4-Fyn interaction in the modulation of PAI-1-tPA axis in astrocytes during neuroinflammatory responses such as ischemia/reperfusion injuries.


Subject(s)
Astrocytes/metabolism , Lipopolysaccharides/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Toll-Like Receptor 4/metabolism , Animals , Astrocytes/drug effects , Cattle , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Gene Knockdown Techniques , Injections, Intraventricular , Male , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , Neurites/drug effects , Neurites/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphotyrosine/metabolism , Protein Binding/drug effects , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation/drug effects , src-Family Kinases/metabolism
5.
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
6.
J Ginseng Res ; 37(4): 401-12, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24235858

ABSTRACT

Korean Red Ginseng (KRG) is an oriental herbal preparation obtained from Panax ginseng Meyer (Araliaceae). To expand our understanding of the action of KRG on central nervous system (CNS) function, we examined the effects of KRG on tissue plasminogen activator (tPA)/plasminogen activator inhibitor-1 (PAI-1) expression in rat primary astrocytes. KRG extract was treated in cultured rat primary astrocytes and neuron in a concentration range of 0.1 to 1.0 mg/mL and the expression of functional tPA/PAI-1 was examined by casein zymography, Western blot and reverse transcription-polymerase chain reaction. KRG extracts increased PAI-1 expression in rat primary astrocytes in a concentration dependent manner (0.1 to 1.0 mg/mL) without affecting the expression of tPA itself. Treatment of 1.0 mg/mL KRG increased PAI-1 protein expression in rat primary astrocytes to 319.3±65.9% as compared with control. The increased PAI-1 expression mediated the overall decrease in tPA activity in rat primary astrocytes. Due to the lack of PAI-1 expression in neuron, KRG did not affect tPA activity in neuron. KRG treatment induced a concentration dependent activation of PI3K, p38, ERK1/2, and JNK in rat primary astrocytes and treatment of PI3K or MAPK inhibitors such as LY294002, U0126, SB203580, and SP600125 (10 µM each), significantly inhibited 1.0 mg/mL KRG-induced expression of PAI- 1 and down-regulation of tPA activity in rat primary astrocytes. Furthermore, compound K but not other ginsenosides such as Rb1 and Rg1 induced PAI-1 expression. KRG-induced up-regulation of PAI-1 in astrocytes may play important role in the regulation of overall tPA activity in brain, which might underlie some of the beneficial effects of KRG on CNS such as neuroprotection in ischemia and brain damaging condition as well as prevention or recovery from addiction.

7.
Biomol Ther (Seoul) ; 21(3): 222-8, 2013 May 30.
Article in English | MEDLINE | ID: mdl-24265868

ABSTRACT

Although the role of α-synuclein aggregation on Parkinson's disease is relatively well known, the physiological role and the regulatory mechanism governing the expression of α-synuclein are unclear yet. We recently reported that α-synuclein is expressed and secreted from cultured astrocytes. In this study, we investigated the effect of valproic acid (VPA), which has been suggested to provide neuroprotection by increasing α-synuclein in neuron, on α-synuclein expression in rat primary astrocytes. VPA concentrationdependently increased the protein expression level of α-synuclein in cultured rat primary astrocytes with concomitant increase in mRNA expression level. Likewise, the level of secreted α-synuclein was also increased by VPA. VPA increased the phosphorylation of Erk1/2 and JNK and pretreatment of a JNK inhibitor SP600125 prevented the VPA-induced increase in α-synuclein. Whether the increased α-synuclein in astrocytes is involved in the reported neuroprotective effects of VPA awaits further investigation.

8.
Biomol Ther (Seoul) ; 21(2): 107-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24009867

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) is a member of serine protease inhibitor family, which regulates the activity of tissue plasminogen activator (tPA). In CNS, tPA/PAI-1 activity is involved in the regulation of a variety of cellular processes such as neuronal development, synaptic plasticity and cell survival. To gain a more insights into the regulatory mechanism modulating tPA/PAI-1 activity in brain, we investigated the effects of proteasome inhibitors on tPA/PAI-1 expression and activity in rat primary astrocytes, the major cell type expressing both tPA and PAI-1. We found that submicromolar concentration of MG132, a cell permeable peptide-aldehyde inhibitor of ubiquitin proteasome pathway selectively upregulates PAI-1 expression. Upregulation of PAI-1 mRNA as well as increased PAI-1 promoter reporter activity suggested that MG132 transcriptionally increased PAI-1 expression. The induction of PAI-1 downregulated tPA activity in rat primary astrocytes. Another proteasome inhibitor lactacystin similarly increased the expression of PAI-1 in rat primary astrocytes. MG132 activated MAPK pathways as well as PI3K/Akt pathways. Inhibitors of these signaling pathways reduced MG132-mediated upregulation of PAI-1 in varying degrees and most prominent effects were observed with SB203580, a p38 MAPK pathway inhibitor. The regulation of tPA/PAI-1 activity by proteasome inhibitor in rat primary astrocytes may underlie the observed CNS effects of MG132 such as neuroprotection.

9.
Biochem Biophys Res Commun ; 438(2): 420-6, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23899524

ABSTRACT

Despite the extensive use of propofol in general anesthetic procedures, the effects of propofol on glial cell were not completely understood. In lipopolysaccharide (LPS)-stimulated rat primary astrocytes and BV2 microglial cell lines, co-treatment of propofol synergistically induced inflammatory activation as evidenced by the increased production of NO, ROS and expression of iNOS, MMP-9 and several cytokines. Propofol augmented the activation of JNK and p38 MAPKs induced by LPS and the synergistic activation of glial cells by propofol was prevented by pretreatment of JNK and p38 inhibitors. When we treated BV2 cell culture supernatants treated with LPS plus propofol on cultured rat primary neuron, it induced a significant neuronal cell death. The results suggest that the repeated use of propofol in immunologically challenged situation may induce glial activation in brain.


Subject(s)
Lipopolysaccharides/metabolism , Neuroglia/cytology , Neuroglia/drug effects , Propofol/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Astrocytes/cytology , Cell Death , Cell Survival , Cells, Cultured , Cerebral Cortex/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Interleukin-1beta/metabolism , Interleukin-6/metabolism , MAP Kinase Kinase 4/metabolism , Matrix Metalloproteinase 9/metabolism , Neurons/cytology , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Life Sci ; 92(17-19): 929-37, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23562854

ABSTRACT

AIMS: Tissue plasminogen activator (tPA) is an essential neuromodulator whose involvement in multiple functions such as synaptic plasticity, cytokine-like immune function and regulation of cell survival mandates rapid and tight tPA regulation in the brain. We investigated the possibility that a transient metabolic challenge induced by glucose deprivation may affect tPA activity in rat primary astrocytes, the main cell type responsible for metabolic regulation in the CNS. MAIN METHODS: Rat primary astrocytes were incubated in serum-free DMEM without glucose. Casein zymography was used to determine tPA activity, and tPA mRNA was measured by RT-PCR. The signaling pathways regulating tPA activity were identified by Western blotting. KEY FINDINGS: Glucose deprivation rapidly down-regulated the activity of tPA without affecting its mRNA level in rat primary astrocytes; this effect was mimicked by translational inhibitors. The down-regulation of tPA was accompanied by increased tPA degradation, which may be modulated by a proteasome-dependent degradation pathway. Glucose deprivation induced activation of PI3K-Akt-GSK3ß, p38 and AMPK, and inhibition of these pathways using LY294002, SB203580 and compound C significantly inhibited glucose deprivation-induced tPA down-regulation, demonstrating the essential role of these pathways in tPA regulation in glucose-deprived astrocytes. SIGNIFICANCE: Rapid and reversible regulation of tPA activity in rat primary astrocytes during metabolic crisis may minimize energy-requiring neurologic processes in stressed situations. This effect may thereby increase the opportunity to invest cellular resources in cell survival and may allow rapid re-establishment of normal cellular function after the crisis.


Subject(s)
Astrocytes/metabolism , Down-Regulation , Glucose/deficiency , Proteasome Endopeptidase Complex/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Blotting, Western , Chromones/pharmacology , Imidazoles/pharmacology , Morpholines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
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
12.
Toxicol Res ; 29(3): 173-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24386517

ABSTRACT

In-utero exposure to valproic acid (VPA) has been known as a potent inducer of autism spectrum disorder (ASD), not only in humans, but also in animals. In addition to the defects in communication and social interaction as well as repetitive behaviors, ASD patients usually suffer from gastrointestinal (GI) problems. However, the exact mechanism underlying these disorders is not known. In this study, we examined the gross GI tract structure and GI motility in a VPA animal model of ASD. On embryonic day 12 (E12), 4 pregnant Sprague-Dawley (SD) rats were subcutaneously injected with VPA (400 mg/kg) in the treatment group, and with phosphate buffered saline (PBS) in the control group; the resulting male offspring were analyzed at 4 weeks of age. VPA exposure decreased the thickness of tunica mucosa and tunica muscularis in the stomach and ileum. Other regions such as duodenum, jejunum, and colon did not show a significant difference. In high-resolution microscopic observation, atrophy of the parietal and chief cells in the stomach and absorptive cells in the ileum was observed. In addition, decreased staining of the epithelial cells was observed in the hematoxylin and eosin (H&E)-stained ileum section. Furthermore, decreased motility in GI tract was also observed in rat offspring prenatally exposed to VPA. However, the mechanism underlying GI tract defects in VPA animal model as well as the association between abnormal GI structure and function with ASD is yet to be clearly understood. Nevertheless, the results from the present study suggest that this VPA ASD model undergoes abnormal changes in the GI structure and function, which in turn could provide beneficial clues pertaining to the pathophysiological relevance of GI complications and ASD phenotypes.

13.
J Adolesc ; 35(4): 969-80, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22418452

ABSTRACT

The purpose of this study was to test the relationships between financial socialization agents, financial experiences, money attitudes, demographic characteristics, and the financial literacy of Korean adolescents. Using the 2006 Korean National Financial Literacy Test Survey for Adolescents (N = 1185), a series of regression analyses were performed to determine the factors related to financial literacy. It was found that those who chose media as their primary financial socialization agent, and those who had a bank account, exhibited higher levels of financial literacy. Among the sample, those who saw money as good or as a reward for efforts tended to report higher levels of financial literacy, while those perceiving money in terms of avoidance or achievement had lower levels of financial literacy. Students with mid-range monthly allowances showed higher levels of financial literacy compared to the highest allowance group. Implications for financial educators, policy makers, and researchers are provided.


Subject(s)
Attitude , Financing, Personal , Socialization , Achievement , Adolescent , Economics , Educational Status , Female , Humans , Male , Republic of Korea , Socioeconomic Factors
14.
Biomol Ther (Seoul) ; 20(2): 226-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-24116300

ABSTRACT

Ethanol exposure during gestational period is related to growth retardation, morphological abnormality, and even in neurological abnormalities including attention deficit/hyperactivity disorder (ADHD)-like behaviors on offspring. However, relatively little is known about the effects of maternal ethanol consumption prior to conception on their offspring. In this study, we investi-gated whether maternal ethanol administration during preconceptional phase produces ADHD-like behaviors in the rat offspring. Sprague-Dawley (SD) female rats were administrated ethanol via intragastric intubation with dosing regimen of 6 g/kg daily for 10 consecutive days and treated female rats then mated with non-treated male SD rats after 8 weeks. Another group subjected to the same procedure as those conducted on ethanol treated group except the saline administration instead of ethanol. Offspring was tested for their ADHD-like behaviors using open field test, Y maze test and impulsivity test that is performed in the aversive electronic foot shock paradigm. Offspring of preconceptional ethanol treated (EtOH) group showed hyperlocomotive activity, attention deficit and impulsivity. And reduction of striatal dopamine transporter (DAT) level was observed by Western blot in the EtOH group, compared to control (Con) group, while the immunohistochemical analysis exhibited increased expression of norepinephrine transporter (NET) in the frontal cortex. These results suggest that maternal ethanol consumption in the preconceptional phase induces ADHD-like behaviors in offspring that might be related to the abnormal expression of DAT and NET in rat.

15.
J Neurosci Res ; 89(7): 1059-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21412817

ABSTRACT

Although originally known as a plasma serine protease involved in clot dissolution, tPA and its primary inhibitor, PAI-1, play crucial roles in synaptic reorganization and plasticity in the central nervous system. In contrast to the wide array of work conducted in neural cells, relatively little is known about the regulatory mechanism governing tPA/PAI-1 expression in astrocytes. Glucocorticoids (GCs) such as hydrocortisone regulate the expression of tPA/PAI-1 in various biological systems in a tissue-specific manner. However, little is known about GC-mediated regulation of tPA/PAI-1 system in CNS. The aims of the present study were to investigate whether tPA/PAI-1 expression is regulated by hydrocortisone in rat primary astrocytes. Enzyme activity of tPA was decreased in a concentration-dependent manner by hydrocortisone treatment, and the activity of PAI-1 was increased by hydrocortisone. Hydrocortisone did not affect the level of tPA mRNA, which suggests that transcriptional down-regulation of tPA mRNA is not involved in the down-regulation of tPA enzyme activity in astrocytes. However, the level of PAI-1 mRNA and protein was increased. Both hydrocortisone and a tPA-Stop treatment prevented glutamate-induced neurotoxicity in rat cortical primary mixed astrocyte-neuron culture, which suggests a neurotoxic role for tPA in our culture system. Interestingly, hydrocortisone further increased LPS-induced up-regulation of PAI-1 while inhibiting the up-regulation of iNOS and COX-2 expression. Our data show that hydrocortisone up-regulated PAI-1 expression along with down-regulation of tPA activity in both normal and inflammatory conditions.


Subject(s)
Astrocytes/metabolism , Hydrocortisone/physiology , Plasminogen Activator Inhibitor 1/metabolism , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Down-Regulation/physiology , Enzyme Activation/physiology , Female , Glutamic Acid/toxicity , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Tissue Plasminogen Activator/genetics , Up-Regulation/physiology
16.
Biochem Biophys Res Commun ; 402(1): 48-53, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20888796

ABSTRACT

A new family of cytokine IL-32 has been implicated in pro-inflammatory immune responses several human diseases such as rheumatoid arthritis, inflammatory bowel diseases and vasculitis. In this study, we investigated the role of IL-32 in the inflammatory activation of cultured rat primary astrocytes. Treatment of IL-32 increased ROS production and augmented lipopolysaccharide-induced increased production of nitric oxide as well as the expression of iNOS. IL-32 also induced the expression of MMP-9 but not MMP-2 in rat primary astrocytes. The increased expression of these inflammatory mediators was accompanied by the increased mRNA expression encoding iNOS, MMP-9 and TNF-α. ERK1/2 and p38, two essential regulators of pro-inflammatory signaling in rat primary astrocytes were activated by IL-32 as evidenced by increased phosphorylation. The results from the present study suggest that IL-32 may play a role in the regulation of neuroinflammatory responses in several neurological disease conditions such as ischemia and Alzheimer's disease.


Subject(s)
Astrocytes/metabolism , Encephalitis/metabolism , Interleukins/physiology , Alzheimer Disease/metabolism , Animals , Astrocytes/drug effects , Brain Ischemia/metabolism , Cells, Cultured , Humans , Interleukins/pharmacology , Lipopolysaccharides/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Rats , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Neurosci Lett ; 469(3): 352-6, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20026244

ABSTRACT

It is increasingly evident that neuroinflammatory response is involved in the pathogenesis of Parkinson's disease. In this study, we examined whether alpha-synuclein, a major components of Lewy body that has been implicated in the modulation of neuroinflammation, regulates MMP-9 and tPA activity, which plays important roles in neurodegeneration as well as regeneration processes, in cultured rat primary glial cells. Monomeric alpha-synuclein dose-dependently increased MMP-9 but not MMP-2 activity as well as mRNA level from cultured rat primary astrocytes and microglial cells. Maximal stimulation was observed at 50 nM alpha-synuclein. In contrast, the activity of tPA was decreased by alpha-synuclein with only marginal changes in the level of mRNA encoding tPA, if any. Interestingly, same concentration of alpha-synuclein aggregates did not induce MMP-9 activity. Overexpression of alpha-synuclein in rat primary astrocytes similarly increased MMP-9 activity. Treatment of alpha-synuclein increased the phosphorylation of ERK1/2 and the inhibition of ERK1/2 reversed the changes in MMP-9 and tPA activity. These results suggest further functional role of alpha-synuclein via regulation of protease systems through modulation of ERK1/2 activity in brain.


Subject(s)
Astrocytes/metabolism , Matrix Metalloproteinase 9/metabolism , Microglia/metabolism , Tissue Plasminogen Activator/metabolism , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Frontal Lobe/metabolism , Matrix Metalloproteinase 2/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Neurochem Res ; 33(11): 2324-34, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18493852

ABSTRACT

We investigated the effect of the cAMP system on lipopolysaccharide (LPS)-induced changes in the activity of matrix metalloproteinases (MMPs) and tissue plasminogen activator (tPA) in rat primary astrocytes. LPS stimulation increased MMP-9 and decreased tPA activity in rat primary astrocytes. Co-treatment with a cAMP analog, dibutyryl-cAMP (db-cAMP), or the cAMP elevating beta-adrenergic agonist, isoproterenol, concentration-dependently inhibited LPS-induced MMP-9 activity. In contrast, db-cAMP concentration-dependently increased tPA activity in both basal and LPS-stimulated rat primary astrocytes. To confirm the effect of cAMP on MMP-9 and tPA activity, we treated LPS-stimulated astrocytes with cAMP phosphodiesterase inhibitors, IBMX or rolipram, and they exhibited similar effects to db-cAMP, namely decreasing MMP-9 activity and increasing tPA activity. RT-PCR analysis of MMP-9 mRNA expression and MMP-9 promoter luciferase reporter assays revealed transcriptional upregulation by LPS stimulation and downregulation by db-cAMP. In contrast, the level of tPA mRNA expression was increased both by LPS and by cAMP treatment. Consistent with RT-PCR analysis, tPA promoter reporter assays showed increased activity by both LPS and cAMP stimulation. Interestingly, the level of mRNA encoding plasminogen activator inhibitor-1 (PAI-1) was increased by LPS stimulation and decreased back to control level after co-treatment with db-cAMP, suggesting that PAI-1 expression plays a major role in the regulation of tPA activity. To examine PKA involvement in the effects of db-cAMP on MMP-9 and tPA activity, we added the PKA inhibitors, H89 or rp-cAMP, along with db-cAMP, and they inhibited db-cAMP-mediated changes in tPA activity without affecting MMP-9 activity. These data suggest that cAMP differentially modulates MMP-9 and tPA activity through a mechanism related to PKA activation. The differential regulation of MMP-9 and tPA by the cAMP system may confer more sophisticated regulation of physiological processes, such as extracellular matrix remodeling and cell migration, by activated astrocytes.


Subject(s)
Astrocytes/drug effects , Cyclic AMP/metabolism , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Enzyme Activation , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Plasminogen Activator/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...