Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Acta cir. bras ; 32(10): 862-872, Oct. 2017. graf
Article in English | LILACS | ID: biblio-886174

ABSTRACT

Abstract Purpose: To investigate whether the neuroprotective effect of TSA on cerebral ischemia reperfusion injury is mediated by the activation of Akt/GSK-3β signaling pathway. Methods: Mice were randomly divided into four groups (n=15): sham group (S); ischemia reperfusion group (IR); ischemia reperfusion and pretreated with TSA group (IR+T); ischemia reperfusion and pretreated with TSA and LY294002 group (IR+T+L). The model of cerebral ischemia reperfusion was established by 1h of MCAO following 24h of reperfusion. TSA (5mg/kg) was intraperitoneally given for 3 days before MCAO, Akt inhibitor, LY294002 (15 nmol/kg) was injected by tail vein 30 min before the MCAO. Results: TSA significantly increased the expression of p-Akt, p-GSK-3β proteins and the levels of SOD, Bcl-2, reduced the infarct volume and the levels of MDA, ROS, TNF-α, IL-1β, Bax, Caspase-3, TUNEL and attenuated neurological deficit in mice with transient MCAO, LY294002 weakened such effect of TSA dramatically. Conclusions: TSA could significantly decrease the neurological deficit and reduce the cerebral infarct volume, oxidative stress, inflammation, as well as apoptosis during cerebral ischemia reperfusion injury, which was achieved by activation of the Akt/GSK-3β signaling pathway.


Subject(s)
Animals , Male , Rats , Signal Transduction/drug effects , Ischemic Attack, Transient/metabolism , Neuroprotective Agents/pharmacology , Glycogen Synthase Kinase 3/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Histone Deacetylase Inhibitors/pharmacology , Signal Transduction/physiology , Ischemic Attack, Transient/physiopathology , Glycogen Synthase Kinase 3/metabolism , Disease Models, Animal , Mice, Inbred BALB C
2.
Biol. Res ; 48: 1-9, 2015. ilus, graf
Article in English | LILACS | ID: biblio-950808

ABSTRACT

BACKGROUND: The fetus is surrounded by the amniotic fluid (AF) contained by the amniotic sac of the pregnant female. The AF is directly conveyed to the fetus during pregnancy. Although AF has recently been reported as an untapped resource containing various substances, it remains unclear whether the AF could influence fetal neurodevelopment. RESULTS: We used AF that was extracted from embryos at 16 days in pregnant SD rat and exposed the AF to the neural cells derived from the embryos of same rat. We found that the treatment of AF to cortical neurons increased the phosphorylation in ERK1/2 that is necessary for fetal neurodevelopment, which was inhibited by the treatment of MEK inhibitors. Moreover, we found the subsequent inhibition of glycogen synthase kinase-3 (GSK-3), which is an important determinant of cell fate in neural cells. Indeed, AF increased the neural clustering of cortical neurons, which revealed that the clustered cells were proliferating neural progenitor cells. Accordingly, we confirmed the ability of AF to increase the neural progenitor cells through neurosphere formation. Furthermore, we showed that the ERK/GSK-3 pathway was involved in AF-mediated neurosphere enlargement. CONCLUSIONS: Although the placenta mainly supplies oxygenated blood, nutrient substances for fetal development, these findings further suggest that circulating-AF into the fetus could affect fetal neurodevelopment via MAP kinases-derived GSK-3 pathway during pregnancy. Moreover, we suggest that AF could be utilized as a valuable resource in the field of regenerative medicine.


Subject(s)
Animals , Female , Pregnancy , Rats , MAP Kinase Signaling System/physiology , Glycogen Synthase Kinase 3/metabolism , Neural Stem Cells/physiology , Amniotic Fluid/physiology , Phosphorylation/drug effects , Signal Transduction/physiology , Cell Differentiation , Rats, Sprague-Dawley , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neural Stem Cells/cytology
3.
Experimental & Molecular Medicine ; : e60-2013.
Article in English | WPRIM | ID: wpr-152455

ABSTRACT

Alzheimer's disease (AD) is the most common cause of age-related dementia. The neuropathological hallmarks of AD include extracellular deposition of amyloid-beta peptides and neurofibrillary tangles that lead to intracellular hyperphosphorylated tau in the brain. Soluble amyloid-beta oligomers are the primary pathogenic factor leading to cognitive impairment in AD. Neural stem cells (NSCs) are able to self-renew and give rise to multiple neural cell lineages in both developing and adult central nervous systems. To explore the relationship between AD-related pathology and the behaviors of NSCs that enable neuroregeneration, a number of studies have used animal and in vitro models to investigate the role of amyloid-beta on NSCs derived from various brain regions at different developmental stages. However, the Abeta effects on NSCs remain poorly understood because of conflicting results. To investigate the effects of amyloid-beta oligomers on human NSCs, we established amyloid precursor protein Swedish mutant-expressing cells and identified cell-derived amyloid-beta oligomers in the culture media. Human NSCs were isolated from an aborted fetal telencephalon at 13 weeks of gestation and expanded in culture as neurospheres. Human NSCs exposure to cell-derived amyloid-beta oligomers decreased dividing potential resulting from senescence through telomere attrition, impaired neurogenesis and promoted gliogenesis, and attenuated mobility. These amyloid-beta oligomers modulated the proliferation, differentiation and migration patterns of human NSCs via a glycogen synthase kinase-3beta-mediated signaling pathway. These findings contribute to the development of human NSC-based therapy for AD by elucidating the effects of Abeta oligomers on human NSCs.


Subject(s)
Animals , Humans , Mice , Amyloid beta-Peptides/pharmacology , Apoptosis , Cellular Senescence , Cell Movement , Cell Proliferation , Culture Media, Conditioned/chemistry , Fetus/cytology , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Signal Transduction , Telomere Shortening
4.
Experimental & Molecular Medicine ; : 340-349, 2012.
Article in English | WPRIM | ID: wpr-153073

ABSTRACT

In this study, we showed the direct interaction between Mycobacterium avium subsp. paratuberculosis fibronectin attachment protein (FAP) and toll-like receptor4 (TLR4) via co-localization and binding by using confocal microscopy and co-immunoprecipitation assays. FAP triggered the expression of pro- and anti-inflammatory cytokines in a TLR4-dependent manner. In addition, FAP-induced cytokine expression in bone marrow-derived dendritic cells (BMDCs) was modulated in part by glycogen synthase kinase-3 (GSK-3). FAP-induced expression of CD80, CD86, major histocompatibility complex (MHC) class I, and MHC class II in TLR4+/+ BMDCs was not observed in TLR4-/- BMDCs. Furthermore, FAP induced DC-mediated CD8+ T cell proliferation and cytotoxic T lymphocyte (CTL) activity, and suppressed tumor growth with DC-based tumor vaccination in EG7 thymoma murine model. Taken together, these results indicate that the TLR4 agonist, FAP, a potential immunoadjuvant for DC-based cancer vaccination, improves the DC-based immune response via the TLR4 signaling pathway.


Subject(s)
Animals , Humans , Mice , Adhesins, Bacterial/genetics , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/therapeutic use , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/cytology , Disease Models, Animal , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Mice, Inbred C57BL , Mycobacterium avium/genetics , Paratuberculosis/metabolism , Protein Binding , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , Thymoma/genetics , Toll-Like Receptor 4/agonists
5.
Journal of Korean Medical Science ; : 572-575, 2012.
Article in English | WPRIM | ID: wpr-119890

ABSTRACT

Sauchinone has been known to have anti-inflammatory and antioxidant effects. We determined whether sauchinone is beneficial in regional myocardial ischemia/reperfusion (I/R) injury. Rats were subjected to 20 min occlusion of the left anterior descending coronary artery, followed by 2 hr reperfusion. Sauchinone (10 mg/kg) was administered intraperitoneally 30 min before the onset of ischemia. The infarct size was measured 2 hr after resuming the perfusion. The expression of cell death kinases (p38 and JNK) and reperfusion injury salvage kinases (phosphatidylinositol-3-OH kinases-Akt, extra-cellular signal-regulated kinases [ERK1/2])/glycogen synthase kinase (GSK)-3beta was determined 5 min after resuming the perfusion. Sauchinone significantly reduced the infarct size (29.0% +/- 5.3% in the sauchinone group vs 44.4% +/- 6.1% in the control, P < 0.05). Accordingly, the phosphorylation of JNK and p38 was significantly attenuated, while that of ERK1/2, Akt and GSK-3beta was not affected. It is suggested that sauchinone protects against regional myocardial I/R injury through inhibition of phosphorylation of p38 and JNK death signaling pathways.


Subject(s)
Animals , Rats , Benzopyrans/pharmacology , Dioxoles/pharmacology , Glycogen Synthase Kinase 3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Reperfusion Injury/metabolism , Phosphorylation , Protective Agents/pharmacology , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Journal of Veterinary Science ; : 235-244, 2012.
Article in English | WPRIM | ID: wpr-65170

ABSTRACT

Akt/protein kinase B is a well-known cell survival factor and activated by many stimuli including mechanical stretching. Therefore, we evaluated the cardioprotective effect of a brief mechanical stretching of rat hearts and determined whether activation of Akt through phosphatidylinositol 3-kinase (PI3K) is involved in stretch-induced cardioprotection (SIC). Stretch preconditioning reduced infarct size and improved post-ischemic cardiac function compared to the control group. Phosphorylation of Akt and its downstream substrate, GSK-3beta, was increased by mechanical stretching and completely blocked by wortmannin, a PI3K inhibitor. Treatment with lithium or SB216763 (GSK-3beta inhibitors) before ischemia induction mimicked the protective effects of SIC on rat heart. Gadolinium (Gd3+), a blocker of stretch-activated ion channels (SACs), inhibited the stretch-induced phosphorylation of Akt and GSK-3beta. Furthermore, SIC was abrogated by wortmannin and Gd3+. In vivo stretching induced by an aorto-caval shunt increased Akt phosphorylation and reduced myocardial infarction; these effects were diminished by wortmannin and Gd3+ pretreatment. Our results showed that mechanical stretching can provide cardioprotection against ischemia-reperfusion injury. Additionally, the activation of Akt, which might be regulated by SACs and the PI3K pathway, plays an important role in SIC.


Subject(s)
Animals , Male , Rats , Androstadienes/pharmacology , Gadolinium/pharmacology , Glycogen Synthase Kinase 3/metabolism , Indoles/pharmacology , Ischemic Preconditioning, Myocardial , Lithium/pharmacology , Maleimides/pharmacology , Myocardial Reperfusion Injury/enzymology , Phosphatidylinositol 3-Kinase/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms
7.
Experimental & Molecular Medicine ; : 53-61, 2011.
Article in English | WPRIM | ID: wpr-48414

ABSTRACT

Clusterin is a secretory glycoprotein, which is highly up-regulated in a variety of normal and injury tissues undergoing apoptosis including infarct region of the myocardium. Here, we report that clusterin protects H9c2 cardiomyocytes from H2O2-induced apoptosis by triggering the activation of Akt and GSK-3beta. Treatment with H2O2 induces apoptosis of H9c2 cells by promoting caspase cleavage and cytochrome c release from mitochondria. However, co-treatment with clusterin reverses the induction of apoptotic signaling by H2O2, thereby recovers cell viability. The protective effect of clusterin on H2O2-induced apoptosis is impaired by PI3K inhibitor LY294002, which effectively suppresses clusterin-induced activation of Akt and GSK-3beta. In addition, the protective effect of clusterin is independednt on its receptor megalin, because inhibition of megalin has no effect on clusturin-mediated Akt/GSK-3beta phosphoylation and H9c2 cell viability. Collectively, these results suggest that clusterin has a role protecting cardiomyocytes from oxidative stress and the Akt/GSK-3beta signaling mediates anti-apoptotic effect of clusterin.


Subject(s)
Animals , Humans , Rats , Apoptosis , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Chromones/pharmacology , Clusterin/metabolism , Glycogen Synthase Kinase 3/metabolism , Hydrogen Peroxide/pharmacology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Morpholines/pharmacology , Myocytes, Cardiac/metabolism , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Reactive Oxygen Species/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
8.
Experimental & Molecular Medicine ; : 82-90, 2011.
Article in English | WPRIM | ID: wpr-186265

ABSTRACT

It is not yet understood how the enhanced expression of pancreatic adenocarcinoma up-regulated factor (PAUF; a novel oncogene identified in our recent studies), contributes to the oncogenesis of pancreatic cells. We herein report that PAUF up-regulates the expression and transcriptional activity of beta-catenin while the suppression of PAUF by shRNA down-regulates beta-catenin. The induction of beta-catenin by PAUF is mediated by the activities of Akt and GSK-3beta, but inhibition of downstream ERK does not reduce beta-catenin expression. To test whether PAUF emulates either the Wnt3a-mediated or the protein kinase A-mediated signaling pathway for the stabilization of beta-catenin, we examined the phosphorylation status of beta-catenin in the presence of PAUF compared with that of beta-catenin during treatment with Wnt3a or dibutyryl cAMP, a cell permeable cyclic AMP analogue. PAUF expression induces phosphorylation at Ser-33/37/Thr-41 and Ser-675 of beta-catenin but no phosphorylation at Ser-45, indicating that a unique phosphorylation pattern of beta-catenin is caused by PAUF. Finally, the expression of PAUF up-regulates both cyclin-D1 and c-Jun, target genes of beta-catenin, leading to a rapid proliferation of pancreatic cells; conversely decreased PAUF expression (by shRNA) results in the reduced proliferation of pancreatic cells. Treatment with hexachlorophene (an inhibitor of beta-catenin) reduces the proliferation of pancreatic cells despite the presence of PAUF. Taken together, we propose that PAUF can up-regulate and stabilize beta-catenin via a novel pattern of phosphorylation, thereby contributing to the rapid proliferation of pancreatic cancer cells.


Subject(s)
Humans , Adenocarcinoma/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Lectins/genetics , Pancreatic Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Up-Regulation , beta Catenin/genetics
9.
Experimental & Molecular Medicine ; : 503-513, 2010.
Article in English | WPRIM | ID: wpr-214629

ABSTRACT

2-deoxy-D-glucose (2DG) is known as a synthetic inhibitor of glucose. 2DG regulates various cellular responses including proliferation, apoptosis and differentiation by regulation of glucose metabolism in cancer cells. However, the effects of 2DG in normal cells, including chondrocytes, are not clear yet. We examined the effects of 2DG on dedifferentiation with a focus on the beta-catenin pathway in rabbit articular chondrocytes. The rabbit articular chondrocytes were treated with 5 mM 2DG for the indicated time periods or with various concentrations of 2DG for 24 h, and the expression of type II collagen, c-jun and beta-catenin was determined by Western blot, RT-PCR, immunofluorescence staining and immunohistochemical staining and reduction of sulfated proteoglycan synthesis detected by Alcain blue staining. Luciferase assay using a TCF (T cell factor)/LEF (lymphoid enhancer factor) reporter construct was used to demonstrate the transcriptional activity of beta-catenin. We found that 2DG treatment caused a decrease of type II collagen expression. 2DG induced dedifferentiation was dependent on activation of beta-catenin, as the 2DG stimulated accumulation of beta-catenin, which is characterized by translocation of beta-catenin into the nucleus determined by immunofluorescence staining and luciferase assay. Inhibition of beta-catenin degradation by inhibition of glycogen synthase kinase 3-beta with lithium chloride (LiCl) or inhibition of proteasome with z-Leu-Leu-Leu-CHO (MG132) accelerated the decrease of type II collagen expression in the chondrocytes. 2DG regulated the post-translational level of beta-catenin whereas the transcriptional level of beta-catenin was not altered. These results collectively showed that 2DG regulates dedifferentiation via beta-catenin pathway in rabbit articular chondrocytes.


Subject(s)
Animals , Rabbits , Cartilage, Articular/cytology , Cell Dedifferentiation/drug effects , Cell Nucleus/drug effects , Chondrocytes/cytology , Deoxyglucose/pharmacology , Endoplasmic Reticulum/drug effects , Glycogen Synthase Kinase 3/metabolism , Mutant Proteins/metabolism , Protein Transport/drug effects , Proteoglycans/metabolism , Signal Transduction/drug effects , beta Catenin/metabolism
10.
Journal of Korean Medical Science ; : 1626-1632, 2010.
Article in English | WPRIM | ID: wpr-44279

ABSTRACT

Oxidative stress induced by chronic hyperglycemia in type 2 diabetes plays a crucial role in progressive loss of beta-cell mass through beta-cell apoptosis. Glucagon like peptide-1 (GLP-1) has effects on preservation of beta-cell mass and its insulin secretory function. GLP-1 possibly increases islet cell mass through stimulated proliferation from beta-cell and differentiation to beta-cell from progenitor cells. Also, it probably has an antiapoptotic effect on beta-cell, but detailed mechanisms are not proven. Therefore, we examined the protective mechanism of GLP-1 in beta-cell after induction of oxidative stress. The cell apoptosis decreased to ~50% when cells were treated with 100 microM H2O2 for up to 2 hr. After pretreatment of Ex-4, GLP-1 receptor agonist, flow cytometric analysis shows 41.7% reduction of beta-cell apoptosis. This data suggested that pretreatment of Ex-4 protect from oxidative stress-induced apoptosis. Also, Ex-4 treatment decreased GSK3beta activation, JNK phosphorylation and caspase-9, -3 activation and recovered the expression of insulin2 mRNA in beta-cell lines and secretion of insulin in human islet. These results suggest that Ex-4 may protect beta-cell apoptosis by blocking the JNK and GSK3beta mediated apoptotic pathway.


Subject(s)
Animals , Cricetinae , Humans , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Flow Cytometry , Glucagon-Like Peptide 1/pharmacology , Glycogen Synthase Kinase 3/metabolism , Hydrogen Peroxide/toxicity , Insulin/genetics , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Peptides/pharmacology , Phosphorylation , Receptors, Glucagon/agonists , Signal Transduction , Venoms/pharmacology
11.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 483-6, 2007.
Article in English | WPRIM | ID: wpr-634903

ABSTRACT

To examine the role of glycogen synthase kinase 3 (GSK-3) in the apoptosis of pancreatic beta-cells to better understand the pathogenesis and to find new approach to the treatment of type 2 diabetes, apoptosis was induced by oleic acid (OA) in INS-1 cells and the activity of GSK-3 was inhibited by LiCl. The PI staining and flow cytometry were employed for the evaluation of apoptosis. The phosphorylation level of GSK-3 was detected by Western blotting. The results showed that OA at 0.4 mmol/L could cause conspicuous apoptosis of INS-1 cells and the activity of GSK-3 was significantly increased. After the treatment with 24 mmol/L of LiCl, a inhibitor of GSK-3, the OA-induced apoptosis of INS-1 cells was lessened and the phosphorylation of GSK-3 was increased remarkably. It is concluded that GSK-3 activation plays an important role in OA-induced apoptosis in pancreatic beta-cells and inhibition of the GSK-3 activity can effectively protect INS-1 cells from the OA-induced apoptosis. Our study provides a new experimental basis and target for the clinical treatment of type-2 diabetes.


Subject(s)
Apoptosis/drug effects , Cell Line , Fatty Acids, Nonesterified/pharmacology , Glycogen Synthase Kinase 3/metabolism , Insulin-Secreting Cells/cytology , Oleic Acid/pharmacology , Phosphorylation
12.
Experimental & Molecular Medicine ; : 353-360, 2007.
Article in English | WPRIM | ID: wpr-201419

ABSTRACT

Glycogen synthase kinase 3 (GSK3) was recently suggested to be a potential target of psychotropics used in psychiatric illnesses such as schizophrenia and bipolar disorder. Relevant studies have found that antipsychotic drugs regulate GSK3 activity via an increase in either inhibitory serine phosphorylation or amount of GSK3 after acute or subchronic treatment. Recent evidence shows that GSK3 is regulated by dopaminergic or serotonergic systems implicated in the pathophysiology and treatment mechanisms of schizophrenia and bipolar disorder. Therefore, antipsychotics may regulate GSK3 via antagonizing dopaminergic or serotonergic activity. However, the signaling pathway that is involved in GSK3 regulation by dopaminergic or serotonergic systems has not been well established. Haloperidol is a typical antipsychotic with potent dopamine D(2) receptor antagonism. Clozapine is an atypical antipsychotic with potent serotonin 5HT(2) receptor antagonism. We injected rats with haloperidol or clozapine and examined the phosphorylation and amount of GSK3alpha/beta and its well-known upstream regulators Akt and Dvl in the rat frontal cortex by Western blotting. Both haloperidol and clozapine induced Ser21/9 phosphorylation of GSK3GSK3alpha/beta. Haloperidol increased the Ser473 phosphorylation of Akt transiently, whereas clozapine maintained the increase for 1 h. Haloperidol did not affect the phosphorylation and amount of Dvl, whereas clozapine increased both phosphorylation and the amount of Dvl. Our results suggest that GSK3 activity may be regulated by both typical and atypical antipsychotics and that Akt or Dvl, depending on the D(2)- or 5HT(2)- receptor antagonism properties of typical and atypical antipsychotics, mediate the regulation differently.


Subject(s)
Animals , Male , Rats , Adaptor Proteins, Signal Transducing/metabolism , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Dopamine Antagonists/pharmacology , Frontal Lobe/drug effects , Glycogen Synthase Kinase 3/metabolism , Haloperidol/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL