Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Data Brief ; 45: 108567, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36124136

ABSTRACT

The data presented here are related to the research article entitled "Strengthening and deformation behavior of as-cast CoCrCu1.5MnNi high-entropy alloy (HEA) with micro-/nanoscale precipitation [1]". Non-equimolar CoCrCu1.5MnNi was cast by the conventional induction melting under a high-purity Ar atmosphere. Scanning electron microscopy equipped with energy dispersive spectroscopy (EDS), and transmission electron microscopy (TEM) were used for micro- and nanostructure characterization. Subsize tensile specimens with two different gage length to width ratio were tested at room and cryogenic temperatures to assess the accuracy of strength and ductility data in the as-cast CoCrCu1.5MnNi HEAs. The mixing enthalpy (ΔHmix) versus lattice elastic energy (ΔHel) criterion was used to predict the stable phases. The data on the effects of microstructural and nanostructural distribution of various phases on mechani-cal properties in the as-cast HEA could be used in designing high entropy alloys with excellent as-cast mechanical performance.

2.
Eur J Ophthalmol ; 29(1): 75-81, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29592777

ABSTRACT

PURPOSE:: Development of a new, fiber-free, single-use endo-illuminator for pars plana vitrectomy as a replacement for fiber-based systems with external light sources. The hand-guided intraocularly placed white micro light-emitting diode is evaluated for its illumination properties and potential photochemical and thermal hazards. METHODS:: A micro light-emitting diode was used to develop a single-use intraocular illumination system. The light-source-on-tip device was implemented in a prototype with 23G trocar compatible outer diameter of 0.6 mm. The experimental testing was performed on porcine eyes. All calculations of possible photochemical and thermal hazards during the application of the intraocular micro light-emitting diode were calculated according to DIN EN ISO 15007-2: 2014. RESULTS:: The endo-illuminator generated a homogeneous and bright illumination of the intraocular space. The color impression was physiologic and natural. Contrary to initial apprehension, the possible risk caused by inserting a light-emitting diode into the intraocular vitreous was much smaller when compared to conventional fiber-based illumination systems. The photochemical and thermal hazards allowed a continuous exposure time to the retina of at least 4.7 h. CONCLUSION:: This first intraocular light source showed that a light-emitting diode can be introduced into the eye. The system can be built as single-use illumination system. This light-source-on-tip light-emitting diode-endo-illumination combines a chandelier wide-angle illumination with an adjustable endo-illuminator.


Subject(s)
Light , Lighting/instrumentation , Semiconductors , Vitrectomy , Animals , Equipment Design , Miniaturization , Swine , Vitreoretinal Surgery
3.
J Pharmacol Exp Ther ; 349(2): 268-78, 2014 May.
Article in English | MEDLINE | ID: mdl-24472723

ABSTRACT

The present study was undertaken to determine the underlying mechanism of silibinin-induced cell death in human breast cancer cell lines MCF7 and MDA-MB-231. Silibinin-induced cell death was attenuated by antioxidants, N-acetylcysteine (NAC) and 6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid, suggesting that the effect of silibinin was dependent on generation of reactive oxygen species (ROS). Western blot analysis showed that silibinin induced downregulation of extracellular signal-regulated kinase (ERK) and Akt. When cells were transiently transfected with constitutively active (ca) mitogen-activated protein kinase (MEK), an upstream kinase of ERK and caAkt, they showed resistance to silibinin-induced cell death. Silibinin decreased the cleavage of Notch-1 mRNA and protein levels. Notch-1-overexpressed cells were resistant to the silibinin-induced cell death. Inhibition of Notch-1 signaling was dependent on ROSgeneration. Overexpression of Notch-1 prevented silibinin-induced inhibition of ERK and Akt phosphorylation. Silibinin-induced cell death was accompanied by increased cleavage of caspase-3 and was prevented by caspase-3 inhibitor in MDA-MB-231 cells but not in MCF7 cells. Silibinin induced translocation of apoptosis-inducing factor (AIF), which was blocked by NAC, and transfection of caMEK and caAkt. Silibinin-induced cell death was prevented by silencing of AIF expression using small interfering AIF RNA in MCF7 cells but not in MDA-MB-231 cells. In conclusion, silibinin induces cell death through an AIF-dependent mechanism in MCF7 cells and a caspase-3-dependent mechanism in MDA-MB-231 cells, and ROS generation and Notch-1 signaling act upstream of the ERK and Akt pathway. These data suggest that silibinin may serve as a potential agent for induction of apoptosis in human breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptor, Notch1/metabolism , Silymarin/pharmacology , Active Transport, Cell Nucleus , Antioxidants/pharmacology , Apoptosis Inducing Factor/metabolism , Breast Neoplasms , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , Proto-Oncogene Proteins c-akt/genetics , Receptor, Notch1/genetics , Signal Transduction , Silybin
4.
J Agric Food Chem ; 61(17): 4089-96, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23570653

ABSTRACT

Anticancer activity of silibinin, a flavonoid, has been demonstrated in various cancer cell types. However, the underlying mechanisms were not elucidated in human ovarian cancer cells. The present study was undertaken to examine the effect of silibinin in vitro and in vivo on tumor growth in human ovarian cancer cells. Silibinin decreased cell viability in a dose- and time-dependent manner. Silibinin caused an increase in reactive oxygen species (ROS) generation, and the silibinin-induced cell death was prevented by the antioxidant N-acetylcysteine (NAC). Western blot analysis showed silibinin-induced downregulation of extracellular signal-regulated kinase (ERK) and Akt. Transfection of constitutively active forms of MEK and Akt prevented the silibinin-induced cell death. Oral administration of silibinin in animals with subcutaneous A2780 cells reduced tumor volume. Subsequent tumor tissue analysis showed that silibinin treatment induced a decrease in Ki-67-positive cells, an increase in transferase-mediated dUTP nick end labeling (TUNEL)-positive cells, activation of caspase-3, and inhibition of p-ERK and p-Akt. These results indicate that silibinin reduces tumor growth through inhibition of ERK and Akt in human ovarian cancer cells. These data suggest that silibinin may serve as a potential therapeutic agent for human ovarian cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Silymarin/pharmacology , Acetylcysteine/adverse effects , Animals , Antioxidants/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Silybin
5.
Neurochem Res ; 37(12): 2667-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22878643

ABSTRACT

Adenosine A(3) receptor (A3AR) is coupled to G proteins that are involved in a variety of intracellular signaling pathways and physiological functions. 2-Chloro-N(6)-(3-iodobenzyl) adenosine-5'-N-methylcarboxamide (Cl-IB-MECA), an agonist of A3AR, has been reported to induce cell death in various cancer cells. However, the effect of CI-IB-MECA on glioma cell growth is not clear. This study was undertaken to examine the effect of CI-IB-MECA on glioma cell viability and to determine its molecular mechanism. CI-IB-MECA inhibited cell proliferation and induced cell death in a dose- and time-dependent manner. Treatment of CI-IB-MECA resulted in an increase in intracellular Ca(2+) followed by enhanced reactive oxygen species (ROS) generation. EGTA and N-acetylcysteine (NAC) blocked the cell death induced by CI-IB-MECA, suggesting that Ca(2+) and ROS are involved in the Cl-IB-MECA-induced cell death. Western blot analysis showed that CI-IB-MECA induced the down-regulation of extracellular signal-regulated kinases (ERK) and Akt, which was prevented by EGTA, NAC, and the A3AR antagonist MRS1191. Transfection of constitutively active forms of MEK, the upstream kinase of ERK, and Akt prevented the cell death. CI-IB-MECA induced caspase-3 activation and the CI-IB-MECA-induced cell death was blocked by the caspase inhibitors DEVD-CHO and z-VAD-FMK. In addition, expression of XIAP and Survivin were decreased in cells treated with Cl-IB-MECA. Collectively, these findings demonstrate that CI-IB-MECA induce a caspase-dependent cell death through suppression of ERK and Akt mediated by an increase in intracellular Ca(2+) and ROS generation in human glioma cells. These suggest that A3AR agonists may be a potential therapeutic agent for induction of apoptosis in human glioma cells.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Calcium/metabolism , Cell Death/drug effects , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Adenosine/pharmacology , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Primers , Humans , Reverse Transcriptase Polymerase Chain Reaction
6.
Oncol Rep ; 27(5): 1467-74, 2012 May.
Article in English | MEDLINE | ID: mdl-22266745

ABSTRACT

Lycii cortex radicis (LCR) is a traditional Korean medicinal herb. The present study was undertaken to examine the effect of an LCR extract on glioma cell growth and to determine its molecular mechanism in U87MG human glioma cells. The LCR extract resulted in apoptotic cell death in a dose- and time-dependent manner. The LCR extract-induced cell death was associated with generation of reactive oxygen species (ROS). Western blot analysis showed that the LCR extract caused downregulation of Akt and ERK. The LCR-induced cell death was prevented by transfection with the constitutively active forms of Akt and MEK. Oral administration of LCR extracts in subcutaneous U87MG xenograft models reduced glioma tumor volume. Taken together, these findings suggest that the LCR extract results in human glioma cell death through mechanisms involving ROS generation, downregulation of Akt and ERK, and caspase activation in vitro and reduces glioma tumor growth in vivo. These data suggest that the LCR extract may serve as a potential therapeutic agent for malignant human glioblastomas.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Glioma/enzymology , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Glioma/drug therapy , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
7.
J Hazard Mater ; 201-202: 7-15, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22169147

ABSTRACT

Palladized iron coated reactive geomembrane (Pd/Fe RGM) was developed to mitigate the mass transfer of chlorinated volatile organic compounds through HDPE geomembrane. It had good performance in mitigating the mass transfer of TCE at the preliminary study. This study focused on the effects of Pd/Fe coating on the mass transfer parameters and the reaction rate constant of RGM. Also, the effects of sulfide and aging on the RGM were evaluated. It was proved that the effect of Pd/Fe coating on partition and diffusion coefficients of toluene was not significant by batch immersion test results with student's t-test. The reactivity of Pd/Fe RGM had a linear correlation with an increase in the Pd/Fe loading rate on RGM. However, it was lower than the reactivity of the Pd/Fe powder by a factor of one hundred because the effective surface area of Pd/Fe was blocked partially by the geomembrane and the opportunity to contact Pd/Fe and TCE was reduced. The reactivity of Pd/Fe RGM decreased severely at 10mM sulfide concentration and with long-term aging of RGM in the atmosphere for three years. However, the reactivity of aged RGM was recovered slightly by acid treatment, but not completely.


Subject(s)
Environmental Pollution/prevention & control , Iron/chemistry , Membranes, Artificial , Palladium/chemistry , Trichloroethylene/analysis , Water Pollutants, Chemical/analysis , Microscopy, Electron, Scanning , Models, Chemical , Permeability , Surface Properties , Trichloroethylene/chemistry , Volatilization , Water Pollutants, Chemical/chemistry
8.
Cell Biol Int ; 36(4): 339-44, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22073986

ABSTRACT

The flavonoid, luteolin, has been shown to have anticancer activity in various cancer cells; however, the precise molecular mechanism of its action is not completely understood, and studies were conducted to find out how it induces apoptosis in breast cancer cells. Luteolin induced a reduction of viability in a dose- and time-dependent manner. The pro-apoptotic effect of luteolin was demonstrated by cell cycle measurement and Hoechst 3325 staining. Western blot analysis showed that luteolin activates ERK (extracellular-signal-regulated kinase) and p38. Pharmacological inhibition or knockdown of ERK and p38 protected against luteolin-induced cell death; however, the caspase-3-specific inhibitor had no effect. Immunocytochemical examination indicated that luteolin induced nuclear translocation of AIF (apoptosis-inducing factor), which was mediated by activation of ERK and p38. Transfection of a vector expressing the miRNA (microRNA) of AIF prevented luteolin-induced apoptosis. The data suggest that luteolin induces a caspase-dependent and -independent apoptosis involving AIF nuclear translocation mediated by activation of ERK and p38 in breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Inducing Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Luteolin/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Apoptosis Inducing Factor/genetics , Breast Neoplasms , Caspase 3/genetics , Caspase Inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Gene Knockdown Techniques , Genetic Vectors , Humans , MAP Kinase Signaling System/drug effects , MicroRNAs/genetics , Protein Transport/drug effects , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
9.
J Korean Med Sci ; 26(4): 499-506, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21468256

ABSTRACT

The purpose of this study was to establish a prediction rule for severe illness in adult patients hospitalized with pandemic influenza A (H1N1) 2009. At the time of initial presentation, the baseline characteristics of those with severe illness (i.e., admission to intensive care unit, mechanical ventilation, or death) were compared to those of patients with non-severe illnesses. A total of 709 adults hospitalized with pandemic influenza A (H1N1) 2009 were included: 75 severe and 634 non-severe cases. The multivariate analysis demonstrated that altered mental status, hypoxia (PaO(2)/FiO(2) ≤ 250), bilateral lung infiltration, and old age (≥ 65 yr) were independent risk factors for severe cases (all P < 0.001). The area under the ROC curve (0.834 [95% CI, 0.778-0.890]) of the number of risk factors were not significantly different with that of APACHE II score (0.840 [95% CI, 0.790-0.891]) (P = 0.496). The presence of ≥ 2 risk factors had a higher sensitivity, specificity, positive predictive value and negative predictive value than an APACHE II score of ≥ 13. As a prediction rule, the presence of ≥ 2 these risk factors is a powerful and easy-to-use predictor of the severity in adult patients hospitalized with pandemic influenza A (H1N1) 2009.


Subject(s)
Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza, Human/epidemiology , APACHE , Adult , Aged , Antiviral Agents/therapeutic use , Female , Hospitalization , Humans , Influenza, Human/drug therapy , Influenza, Human/mortality , Intensive Care Units , Male , Middle Aged , Pandemics , Predictive Value of Tests , ROC Curve , Respiration, Artificial , Risk Factors , Severity of Illness Index
10.
Arch Toxicol ; 85(9): 1057-65, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21259059

ABSTRACT

Apoptosis of stem cells may be related to certain degenerative conditions such as progressive tissue damage and an inability to repair. Ceramide induces cell death in various cell types. However, the underlying mechanisms of ceramide-induced cell death in stem cells are not explored. This study was designed to investigate the cell death process caused by cell-permeable ceramide and to determine the underlying mechanisms in mesenchymal stem cells derived from human adipose tissue (hASCs). Ceramide caused a loss of cell viability in a concentration- and time-dependent manner, which was largely attributable to apoptosis. Ceramide induced generation of reactive oxygen species (ROS) and disruption of the mitochondrial membrane potential. The ROS generation caused by ceramide was prevented by the antioxidant N-acetylcysteine (NAC). Although ceramide induced release of cytochrome c from mitochondria and activation of caspase-3, the ceramide-induced cell death was partially prevented by caspase inhibitors. Addition of ceramide caused apoptosis-inducing factor (AIF) nuclear translocation, which was prevented by antioxidant. Taken together, these data suggest that ceramide induces cell death through both caspase-dependent and caspase-independent mechanisms mediated by ROS generation in hASCs.


Subject(s)
Adipose Tissue/drug effects , Apoptosis/drug effects , Caspase 3/metabolism , Ceramides/pharmacology , Mesenchymal Stem Cells/drug effects , Adipose Tissue/enzymology , Adipose Tissue/pathology , Apoptosis Inducing Factor/metabolism , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Ceramides/metabolism , Ceramides/physiology , Cytochromes c/metabolism , Flow Cytometry , Humans , Membrane Potential, Mitochondrial/drug effects , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Reactive Oxygen Species/metabolism
11.
Neurochem Res ; 36(5): 722-31, 2011 May.
Article in English | MEDLINE | ID: mdl-21203833

ABSTRACT

Anticancer activity of proteasome inhibitors has been demonstrated in various cancer cell types. However, mechanisms by which they exert anticancer action were not fully understood. The present study was undertaken to examine the effect of the proteasome inhibitor MG-132 and the underlying mechanism in glioma cells. MG-132 caused alterations in mitochondrial membrane potential and apoptosis-inducing factor (AIF) nuclear translocation. MG-132 induced reduction in ERK and Akt activation. The transient transfection of constitutively active forms of MEK, an upstream of ERK, and Akt blocked the MG-132-induced cell death. Similarly to down-regulation of Akt, expression levels of mTOR were inhibited by MG-132. Addition of rapamycin, an inhibitor of mTOR, caused stimulation of the MG-132-induced cell death. There were no significant changes in levels of XIAP, survivin, and Bax. Overexpression of constitutively active forms of MEK and Akt blocked the MG-132-induced AIF nuclear translocation. These findings indicate that MG-132 induces AIF nuclear translocation through down-regulation of ERK and Akt/mTOR pathways. These data suggest that proteasome inhibitors may serve as potential therapeutic agents for malignant human gliomas.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Nucleus/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Leupeptins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Immunohistochemistry , Mitochondria/drug effects , Mitochondria/physiology , Protein Transport
12.
Phytother Res ; 25(3): 429-34, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20737659

ABSTRACT

The present study was undertaken to examine the effect of Fructus ligustri lucidi (FLL) extracts on glioma cell growth and to determine the underlying mechanism by which FLL extracts exert anticancer properties in human U87MG glioma cells. The FLL extracts resulted in cell death in a dose- and time-dependent manner. Western blot analysis showed that treatment with FLL extracts caused down-regulation of the phosphatidylinositol-3 kinase (PI3K)/Akt pathway. Overexpression of Akt prevented the cell death induced by the FLL extracts. The FLL extracts caused a decrease in the expression of mammalian target of rapamycin (mTOR) and the FLL extract-induced cell death was increased by the mTOR inhibitor rapamycin. The FLL extracts decreased the expression of survivin. Oral administration of FLL extracts in subcutaneous U87MG xenograft models reduced the glioma tumor volume. These findings indicate that the FLL extracts resulted in glioma cell death through regulation of the Akt/mTOR/survivin pathway in vitro and inhibited glioma tumor growth in vivo. These data suggest that the FLL extracts may serve as a potential therapeutic agent for malignant human gliomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Glioma/drug therapy , Ligustrum/chemistry , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Phosphatidylinositol 3-Kinases/drug effects , Survivin , Xenograft Model Antitumor Assays
13.
Nutr Cancer ; 62(3): 402-12, 2010.
Article in English | MEDLINE | ID: mdl-20358478

ABSTRACT

Mulberry has been reported to contain wide range of polyphenols and have chemopreventive activity. However, little has been known regarding the effect of mulberry fruit extracts on cell viability in vitro in human glioma cells and the anticancer efficacy in vivo. This study was undertaken to examine the effect of mulberry fruit (Moris fructus; MF) extracts on cell viability in vitro and anticancer efficacy in vivo. Cell viability and cell death were estimated by MTT assay and trypan blue exclusion assay, respectively. Reactive oxygen species (ROS) generation was measured using the fluorescence probe DCFH-DA. The mitochondrial transmembrane potential was measured with DiOC(6)(3). Bax expression and cytochrome c release were measured by Western blot analysis. Caspase activity was estimated using colorimetric kit. Cell migration was estimated using the scratched wound model. In vivo anticancer efficacy of MF extracts was evaluated using a subcutaneously injected mouse tumor model. Changes in proliferation and apoptosis were estimated by immunohistochemistric analysis. MF extracts resulted in apoptotic cell death in a dose- and time-dependent manner. MF extracts increased ROS generation, and the MF extract-induced cell death was also prevented by antioxidants, suggesting that ROS generation plays a critical role in the MF extract-induced cell death. Western blot analysis showed that treatment of MF extracts caused an increase in Bax expression, which was inhibited by the antioxidant N-acetylcysteine (NAC). MF extracts induced depolarization of mitochondrial membrane potential, and its effect was inhibited by the antioxidants NAC and catalase. MF extracts induced cytochrome c release, which was inhibited by NAC. Caspase activity was stimulated by MF extracts, and caspase inhibitors prevented the MF extract-induced cell death. Treatment of MF extracts inhibited cell migration. Oral MF extracts administration in animals with subcutaneous U87MG glioma cells reduced tumor volume. Subsequent tumor tissue analysis showed a decrease in PCNA-positive cells, an increase in TUNEL-positive cells, and caspase activation. From these data, we concluded that MF extracts reduce glioma tumor growth through inhibition of cell proliferation resulting from induction of apoptosis. These findings suggest that MF extracts result in human glioma cell death in vitro through ROS-dependent mitochondrial pathway and glioma tumor growth in vivo via reduction of tumor cell proliferation and induction of apoptosis.


Subject(s)
Apoptosis/drug effects , Glioma/drug therapy , Mitochondria/drug effects , Morus , Phytotherapy , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Caspases/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Glioma/pathology , Humans , Mitochondria/physiology , bcl-2-Associated X Protein/physiology
14.
Ren Fail ; 32(1): 103-11, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20113275

ABSTRACT

Rosiglitazone has been reported to exert the protective effect against acute renal failure in animal models. However, the underlying mechanisms by which it protects the damaged kidney cells are poorly understood. The present study was therefore undertaken to examine the effect of rosiglitazone on cell proliferation and to determine its molecular mechanism in opossum kidney (OK) cells, an established renal proximal tubular cell line. Rosiglitazone treatment inhibited cell proliferation in a dose- and time-dependent manner, and such effects were not associated with induction of cell death. The anti-proliferative effect of rosiglitazone was accompanied by the cell cycle arrest at the G1 phase. Western blot analysis data showed that rosiglitazone caused down-regulation of extracellular signal-regulated kinase (ERK) and Akt pathway. Transfection of constitutively active forms of MEK (an upstream kinase of ERK) and Akt prevented the proliferation inhibition induced by rosiglitazone. Rosiglitazone facilitated the recovery of cells after cisplatin-mediated injury. Taken together, these data suggest that rosiglitazone induces inhibition of cell proliferation through ERK and Akt-dependent cell cycle arrest at the G1 phase. The cell cycle arrest may play a protective role in kidney cells by preventing injured cells from progressing in the cell cycle.


Subject(s)
Cell Proliferation/drug effects , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/physiology , Thiazolidinediones/pharmacology , Cells, Cultured , Humans , Rosiglitazone
15.
Korean J Physiol Pharmacol ; 13(1): 15-22, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19885021

ABSTRACT

This study was undertaken to elucidate the underlying mechanisms of ATP depletion-induced membrane transport dysfunction and cell death in renal proximal tubular cells. ATP depletion was induced by incubating cells with 2.5 mM potassium cyanide (KCN)/0.1 mM iodoacetic acid (IAA), and membrane transport function and cell viability were evaluated by measuring Na(+)-dependent phosphate uptake and trypan blue exclusion, respectively. ATP depletion resulted in a decrease in Na(+)-dependent phosphate uptake and cell viability in a time-dependent manner. ATP depletion inhibited Na(+)-dependent phosphate uptake in cells, when treated with 2 mM ouabain, a Na(+) pump-specific inhibitor, suggesting that ATP depletion impairs membrane transport functional integrity. Alterations in Na(+)-dependent phosphate uptake and cell viability induced by ATP depletion were prevented by the hydrogen peroxide scavenger such as catalase and the hydroxyl radical scavengers (dimethylthiourea and thiourea), and amino acids (glycine and alanine). ATP depletion caused arachidonic acid release and increased mRNA levels of cytosolic phospholipase A(2) (cPLA(2)). The ATP depletion-dependent arachidonic acid release was inhibited by cPLA(2) specific inhibitor AACOCF(3). ATP depletion-induced alterations in Na(+)-dependent phosphate uptake and cell viability were prevented by AACOCF(3). Inhibition of Na(+)-dependent phosphate uptake by ATP depletion was prevented by antipain and leupetin, serine/cysteine protease inhibitors, whereas ATP depletion-induced cell death was not altered by these agents. These results indicate that ATP depletion-induced alterations in membrane transport function and cell viability are due to reactive oxygen species generation and cPLA(2) activation in renal proximal tubular cells. In addition, the present data suggest that serine/cysteine proteases play an important role in membrane transport dysfunction, but not cell death, induced by ATP depletion.

16.
Neurochem Res ; 34(8): 1479-90, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19263218

ABSTRACT

Anticancer activity of silibinin, a flavonoid, has been demonstrated in various cancer cell types. However, the underlying mechanism and in vivo efficacy in glioma were not elucidated. The present study was undertaken to determine the effect of silibinin on glioma cell proliferation in vitro and to examine whether silibinin inhibits tumor growth in vivo. Silibinin resulted in inhibition of proliferation in a dose- and time-dependent manner, which was largely attributed to cell death. Silibinin induced a transient increase in intracellular Ca2+ followed by an increase in reactive oxygen species (ROS) generation. The silibinin-induced cell death was prevented by EGTA, calpain inhibitor and antioxidants (N-acetylcysteine and Trolox). Western blot analysis showed that silibinin also induced ROS-dependent activation of extracellular signal-regulated kinase, p38 kinase, and c-Jun N-terminal kinase. Inhibitors of these kinases prevented the silibinin-induced cell death. Silibinin caused caspase activation and the silibinin-induced cell death was prevented by caspase inhibitors. Glioma cell migration was also decreased by silibinin treatment. Oral administration of silibinin in animals with subcutaneous U87MG glioma cells reduced tumor volume. Subsequent tumor tissue analysis showed a decrease in Ki-67 positive cells, an increase in TUNEL-positive cells, and caspase activation. These results indicate that silibinin induces a caspase-dependent cell death via Ca2+/ROS/MAPK-mediated pathway in vitro and inhibits glioma growth in vivo. These data suggest that silibinin may serve as a potential therapeutic agent for malignant human gliomas.


Subject(s)
Antioxidants/pharmacology , Brain Neoplasms/pathology , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Glioma/pathology , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Blotting, Western , Calcium/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Indicators and Reagents , Silybin , Silymarin/pharmacology
17.
Toxicology ; 258(1): 17-24, 2009 Apr 05.
Article in English | MEDLINE | ID: mdl-19167456

ABSTRACT

We have previously reported the cyclopentenone prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) induces renal proximal epithelial cell death through NF-kappaB inhibition. However, the upstream and down-stream signaling pathways that NF-kappaB inhibition mediates 15d-PGJ(2)-induced apoptosis remain to be defined. In the present study, we determined whether NF-kappaB inhibition induces cell death through the mitochondrial apoptotic pathway and whether protein kinase A (PKA) functions upstream of NF-kappaB inhibition by 15d-PGJ(2). The role of NF-kappaB inhibition in this apoptotic pathway was evaluated using NF-kappaB p65 transfected cells. 15d-PGJ(2) induced cell death by a PPARgamma-independent mechanism and the cell death was prevented by NF-kappaB p65 transfection. 15d-PGJ(2) treatment caused disruption of mitochondrial membrane potential, cytochrome c release, and caspase-3 activation, suggesting that 15d-PGJ(2) induces cell death through a mitochondria-dependent apoptotic mechanism. These changes by 15d-PGJ(2) were attenuated by NF-kappaB p65 transfection. 15d-PGJ(2) treatment resulted in an increase in Bax expression, which were blocked by NF-kappaB p65 transfection. 15d-PGJ(2) treatment caused PKA inhibition and 15d-PGJ(2)-induced cell death was enhanced by the PKA specific inhibitor H89. Inhibition of NF-kappaB by 15d-PGJ(2) was prevented by addition of forskolin, a PKA activator. Taken together, these results suggest that PKA-dependent NF-kappaB inhibition stimulates 15d-PGJ(2)-mediated mitochondrial apoptotic pathway through alterations in expression of the NF-kappaB target genes Bax.


Subject(s)
Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Epithelial Cells/drug effects , Kidney Tubules, Proximal/pathology , Mitochondria/physiology , NF-kappa B/antagonists & inhibitors , Prostaglandin D2/analogs & derivatives , Animals , Blotting, Western , Caspases/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, Reporter , Immunohistochemistry , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/metabolism , Luciferases/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , NF-kappa B/genetics , Opossums , Prostaglandin D2/pharmacology , Prostaglandin D2/physiology
18.
Neurochem Res ; 34(5): 991-1001, 2009 May.
Article in English | MEDLINE | ID: mdl-18949556

ABSTRACT

The present study was undertaken to determine the molecular mechanism by which kaempferol induces cell death in human glioma cells. Kaempferol resulted in loss of cell viability and inhibition of proliferation in a dose- and time-dependent manner, which were largely attributed to cell death. Kaempferol caused an increase in reactive oxygen species (ROS) generation and the kaempferol-induced cell death was prevented by antioxidants, suggesting that ROS generation is involved in kaempferol-induced cell death. Kaempferol caused depolarization of mitochondrial membrane potential. Western blot analysis showed that kaempferol treatment caused a rapid reduction in phosphorylation of extracellular signal-regulated kinase (ERK) and Akt. The ERK inhibitor U0126 and the Akt inhibitor LY984002 increased the kaempferol-induced cell death and overexpression of MEK, the upstream kinase of ERK, and Akt prevented the cell death. The expression of anti-apoptotic proteins XIAP and survivin was down-regulated by kaempferol and its effect was prevented by overexpression of MEK and Akt. Kaempferol induced activation of caspase-3 and kaempferol-induced cell death was prevented by caspase inhibitors. Taken together, these findings suggest that kaempferol results in human glioma cell death through caspase-dependent mechanisms involving down-regulation of XIAP and survivin regulating by ERK and Akt.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , Kaempferols/pharmacology , Microtubule-Associated Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/physiology , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , Brain Neoplasms , Caspase 3/metabolism , Caspase Inhibitors , Cell Death , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Glioma , Humans , Inhibitor of Apoptosis Proteins , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Survivin
19.
Toxicology ; 257(1-2): 1-9, 2009 Mar 04.
Article in English | MEDLINE | ID: mdl-19110029

ABSTRACT

We have previously demonstrated that the synthetic peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist ciglitazone induces apoptosis accompanied by activation of p38 mitogen-activated protein kinase (MAPK) and nuclear translocation of apoptosis inducing factor (AIF) in opossum kidney (OK) renal epithelial cells. However, the precise mechanism by which ciglitazone induces activation of p38 MAPK and the role of AIF in the induction of the apoptosis are not defined. This study was therefore undertaken to determine whether the roles of reactive oxygen species (ROS) generation and intracellular Ca(2+) in the ciglitazone-induced activation of p38 MAPK and whether AIF nuclear translocation is responsible for the ciglitazone-induced apoptosis in OK renal epithelial cells. Ciglitazone caused generation of ROS and an increase in intracellular Ca(2+). Ciglitazone-induced cell death was reduced by the antioxidant Trolox, the Ca(2+) chelator EGTA, and the store-operated Ca(2+) channels (SOCC) blocker lanthanum chloride (La(3+)), indicating involvement of ROS and Ca(2+) in the ciglitazone-induced cell death. Ciglitazone-induced intracellular Ca(2+) increase was decreased by Trolox, while ROS generation was not affected by EGTA and La(3+), suggesting that ROS generation promote the increase of intracellular Ca(2+). Transfection of small interfering RNA (siRNA) of p38 MAPK or vector expressing microRNA (miRNA) of AIF prevented the ciglitazone-induced cell death. Activation of p38 MAPK, mitochondrial membrane depolarization, and AIF nuclear translocation induced by ciglitazone were inhibited by Trolox, EGTA and La(3+). Taken together, these results suggest that ROS-dependent intracellular Ca(2+) increase is responsible for activation of p38 MAPK and nuclear translocation of AIF by ciglitazone.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Epithelial Cells/drug effects , Hypoglycemic Agents/toxicity , Kidney/drug effects , Reactive Oxygen Species/metabolism , Thiazolidinediones/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus , Animals , Antioxidants/pharmacology , Apoptosis Inducing Factor/genetics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Chromans/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Kidney/enzymology , Kidney/pathology , Lanthanum/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Opossums , PPAR gamma/agonists , RNA Interference , RNA, Small Interfering/metabolism , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/genetics
20.
J Bone Miner Metab ; 26(6): 551-60, 2008.
Article in English | MEDLINE | ID: mdl-18979154

ABSTRACT

The present study was undertaken to evaluate effects of quercetin, a major dietary flavonoid occurring in foods of plant origin, on cell viability and migration of osteoblastic cells. Quercetin inhibited cell viability, which was largely attributed to apoptosis, in a dose-and time-dependent manner in osteoblastic cells. Similar cytotoxicity of quercetin was observed in adipose tissue-derived stromal cells. Quercetin exerted a protective effect against H(2)O(2)-induced cell death, whereas it increased TNF-alpha-induced cell death. Western blot analysis showed that quercetin induced activation of ERK and p38, but not JNK. Quercetin-induced cell death was prevented by the ERK inhibitor PD98059, but not by inhibitors of p38 and JNK. Quercetin increased Bax expression and caused depolarization of mitochondrial membrane potential, which were inhibited by PD98059. Quercetin induced caspase-3 activation, and the quercetininduced cell death was prevented by caspase inhibitors. Quercetin inhibited cell migration, and its effect was prevented by inhibitors of ERK and p38. Taken together, these findings suggest that quercetin induces apoptosis through a mitochondria-dependent mechanism involving ERK activation and inhibits migration through activation of ERK and p38 pathways. Quercetin may exert both protective and deleterious effects in bone repair.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/drug effects , Osteoblasts/physiology , Quercetin/pharmacology , 3T3 Cells , Animals , Caspases/metabolism , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , Flavanones/pharmacology , Flavonoids/pharmacology , Hydrogen Peroxide/pharmacology , Mice , Osteoblasts/cytology , Oxidants/pharmacology , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...