Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Biochem Mol Toxicol ; 37(3): e23267, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36524533

ABSTRACT

We identified a gene, subunit C3 (ATP5G3) of mitochondrial ATP synthase, that displayed changes in gene expression under oxidative stress. We examined the role of ATP5G3 and its molecular mechanisms in sodium nitroprusside (SNP)-induced cell death using ATP5G3 small interfering RNA (siATP5G3)-transfected HeLa cells. A significant increase in cytotoxicity was observed in the transfected cells treated with SNP, which suggests a protective role of ATP5G3 in SNP-induced cytotoxicity in the cells. The transfected cells treated with photodegraded SNP showed equal cytotoxicity to SNP, and pretreatment with deferoxamine (DFO) completely inhibited this cytotoxicity. Further, cytotoxicity was significantly inhibited by pretreatment with a p38 inhibitor and was accentuated by the p38 activator in cells. Pretreatment with the Bcl-xL inhibitor also significantly accentuated cytotoxicity. The increase in p38 phosphorylation was significantly higher in siATP5G3-transfected cells treated with SNP in immunoblotting, which was inhibited by pretreatment with DFO. The increase in cytotoxicity with siATP5G3 transfection was completely blocked by cotransfection with sip38, and the blocking effect disappeared by cotransfection with additional siBcl-xL, which suggests that the protective role of ATP5G3 is mediated by Bcl-xL via the inhibition of p38 activity. Cytotoxicity was completely blocked by the cotransfection of siATP5G3 with siBax. No change in apoptotic parameters was observed during cytotoxicity. However, pretreatment with lysosomal inhibitors significantly inhibited cytotoxicity and increased p62 protein levels. These findings suggest that ATP5G3 plays a protective role in autophagic cell death/lysosome-associated cell death induced by SNP via the sequential signaling of ROS/p38/Bcl-xL/Bax in HeLa cells.


Subject(s)
Carcinoma , Humans , Apoptosis , Cell Death , Cell Line, Tumor , HeLa Cells , Nitroprusside/pharmacology
2.
J Biochem Mol Toxicol ; 33(8): e22348, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31066958

ABSTRACT

Golgi S-nitro-N-acetylpenicillamine receptor complex 1 (GS28) has been implicated in Golgi vesicle transport. We examined the role of GS28 and its molecular mechanisms in sodium nitroprusside (SNP)-induced cell death using GS28 siRNA (siGS28)-transfected HeLa cells. Significant inhibition of cytotoxicity was observed in the cells treated with SNP, and photodegraded SNP showed equal cytotoxicity to SNP. Pretreatment with an ERK inhibitor or siErk1 cotransfection blocked the inhibition in cytotoxicity. Additionally, increased phosphorylation of ERK was maintained in the cells treated with SNP, and Nrf2 level was dependent on ERK phosphorylation. However, pretreatment with a pan-caspase inhibitor had no effect on cytotoxicity or procaspase-3 level. Pretreatment with an autophagy inhibitor or siATG5 cotransfection blocked the inhibition of cytotoxicity. The changes of LC3 corresponded to that in siErk1-cotransfected cells. These data suggest that GS28 has an inductive role in SNP-induced cell death via inhibition of ERK, leading to inhibition of autophagic processes in HeLa cells.


Subject(s)
Cell Death/drug effects , Nitroprusside/pharmacology , Qb-SNARE Proteins/metabolism , Uterine Cervical Neoplasms/pathology , Autophagy/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HeLa Cells , Humans , NF-E2-Related Factor 2/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/metabolism
3.
Exp Mol Med ; 50(8): 1-15, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30111834

ABSTRACT

The peroxisome proliferator-activated receptor-γ (PPARγ) improves whole-body insulin sensitivity by regulating the adipogenic and metabolic functions of mature adipocytes. We have previously demonstrated that an active splice variant of X-box binding protein 1 (XBP1s) enhances PPARγ expression during adipogenesis. In this study, we investigated the role of XBP1s, particularly with respect to PPARγ, in the mechanisms underlying insulin sensitivity in mature adipocytes. Insulin was able to stimulate XBP1s generation by activating inositol-requiring enzyme 1 (IRE1) α and was also able to increase its transcriptional activity by inducing nuclear translocation. XBP1s also upregulated the levels of phosphorylated IRS1 and AKT, demonstrating a positive feedback regulatory mechanism linking insulin and XBP1s. XBP1s enhanced the expression of fibroblast growth factor 21 and, in turn, increased PPARγ activity, translocation of GLUT4 to the cell surface, and glucose uptake rate in adipocytes. In addition, XBP1s abolished palmitate-induced insulin resistance in adipocytes by increasing adiponectin secretion, repressing the secretion of pro-inflammatory adipokines such as leptin, monocyte chemoattractant protein 1, and tumor necrosis factor α, and decreasing fatty acid release. These findings provide a novel mechanism by which XBP1s stimulate insulin sensitivity in adipocytes through fibroblast growth factor 21 induction and PPARγ activation.


Subject(s)
Adipocytes/metabolism , Endoribonucleases/metabolism , Glucose/metabolism , Insulin/pharmacology , PPAR gamma/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fibroblast Growth Factors/metabolism , Insulin Resistance , Mice , Models, Biological , Palmitic Acid , Protein Transport/drug effects , RNA Splicing/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , X-Box Binding Protein 1/genetics
4.
Int J Med Sci ; 14(6): 515-522, 2017.
Article in English | MEDLINE | ID: mdl-28638266

ABSTRACT

Aims: GS28 (Golgi SNARE protein, 28 kDa), a member of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) protein family, plays a critical role in mammalian endoplasmic reticulum (ER)-Golgi or intra-Golgi vesicle transport. To date, few researches on the GS28 protein in human cancer tissues have been reported. In this study, we assessed the prognostic value of GS28 in patients with colorectal cancer (CRC). Methods and results: We screened for GS28 expression using immunohistochemistry in 230 surgical CRC specimens. The CRCs were right-sided and left-sided in 28.3% (65/230) and 71.3% (164/230) of patients, respectively. GS28 staining results were available in 214 cases. Among these, there were 26 nuclear predominant cases and 188 non-nuclear predominant cases. Stromal GS28 expression was noted in 152 cases of CRC. GS28 nuclear predominant immunoreactivity was significantly associated with advanced tumour stage (p = 0.045) and marginally associated with perineural invasion (p = 0.064). Decreased GS28 expression in the stromal cells was significantly associated with lymph node metastasis (N stage; p = 0.036). GS28 expression was not associated with epidermal growth factor receptor (EGFR) immunohistochemical positivity or KRAS mutation status. Investigation of the prognostic value of GS28 with Kaplan-Meier analysis revealed a correlation with overall survival (p = 0.004). Cases with GS28 nuclear predominant expression had significantly poorer overall survival than those with a non-nuclear predominant pattern. Conclusions: Taken together, these results indicate that GS28 nuclear predominant expression could serve as a prognostic marker for CRC and may help in identifying aggressive forms of CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Prognosis , Qb-SNARE Proteins/genetics , Adult , Aged , Aged, 80 and over , Biological Transport/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease-Free Survival , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis
5.
Biochem Biophys Res Commun ; 488(3): 477-482, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28522296

ABSTRACT

In obese patients, free fatty acids ectopically accumulated in non-adipose tissues cause cell death. Saturated fatty acids are more deleterious to non-adipose cells, and supplementation with monounsaturated fatty acids has been proposed to rescue cells from saturated fatty acid-induced cytotoxicity; however, the mechanisms are not well understood. To understand the cytoprotective role of monounsaturated fatty acids in lipotoxic cell death of macrophages, we investigated the antagonizing effect of oleate and the underlying mechanisms in palmitate-treated RAW264.7 cells. Palmitate strongly induced apoptosis in macrophages by increasing CD36 expression, which was identified to mediate both endoplasmic reticulum stress and the generation of reactive oxygen species. Co-treatment with oleate significantly reduced CD36 expression and its downstream signaling pathways of apoptosis in palmitate-treated cells. These findings provide a novel mechanism by which oleate protects macrophages from palmitate-induced lipotoxicity.


Subject(s)
Apoptosis/drug effects , CD36 Antigens/biosynthesis , Down-Regulation/drug effects , Macrophages/drug effects , Oleic Acid/pharmacology , Palmitates/pharmacology , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Macrophages/metabolism , Mice , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
6.
PLoS One ; 11(9): e0162623, 2016.
Article in English | MEDLINE | ID: mdl-27611086

ABSTRACT

OBJECTIVE: The protein GS28 (28-kDa Golgi SNARE protein) has been described as a SNARE (Soluble N-ethylmaleimide-sensitive factor attachment protein receptors) protein family member that plays a critical role in mammalian ER-Golgi or intra-Golgi vesicle transport. Little is known about the possible roles of GS28 in pathological conditions. The purpose of this study was to evaluate GS28 expression in cervical cancer tissues and explore its correlation with clinicopathological features and prognosis. METHODS: We investigated GS28 expression in 177 cervical cancer tissues by using immunohistochemistry and evaluated the correlation of GS28 expression with clinicopathological features, the expression of p53 and Bcl-2, and prognosis of cervical cancer patients. Immunoblotting was performed using six freshly frozen cervical cancer tissues to confirm the subcellular localization of GS28. RESULTS: Immunoreactivity of GS28 was observed in both nuclear and cytoplasmic compartments of cervical cancer cells. High nuclear expression of GS28 was associated with advanced tumor stages (P = 0.036) and negative expression of p53 (P = 0.036). In multivariate analyses, patients with high nuclear expression of GS28 showed significantly worse overall survival (OS) (hazard ratio = 3.785, P = 0.003) and progression-free survival (PFS) (hazard ratio = 3.019, P = 0.008), compared to those with low or no nuclear expression. It was also a reliable, independent prognostic marker in subgroups of patients with early stage T1 and negative lymph node metastasis in OS (P = 0.008 and 0.019, respectively). The nuclear expression of GS28 was confirmed by immunoblotting. CONCLUSION: High nuclear expression of GS28 is associated with poor prognosis in early-stage cervical cancer patients. GS28 might be a novel prognostic marker and a potential therapeutic target in cervical cancer treatment.


Subject(s)
Cell Nucleus/metabolism , Qb-SNARE Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Immunoblotting , Lymphatic Metastasis , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Exp Mol Med ; 48: e226, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-27055562

ABSTRACT

Wnt10b, an endogenous inhibitor of adipogenesis, maintains preadipocytes in an undifferentiated state by suppressing adipogenic transcription factors. We have previously demonstrated that Wnt10b transcription during adipogenesis is negatively regulated by X-box-binding protein 1 (XBP1), an important transcription factor of the unfolded protein response. In this report, we demonstrate that XBP1s can directly induce the transcription of microRNA-148a, which in turn mediates the silencing of Wnt10b mRNA during adipogenic differentiation of 3T3-L1 cells. Stability of Wnt10b mRNA was found to be significantly increased by knockdown of XBP1s. Using computational algorithms, a set of microRNAs was predicted to bind Wnt10b mRNA, of which microRNA-148a was selected as a potential target for XBP1s. Our results revealed that microRNA-148a could bind to the 3'UTR of Wnt10b mRNA. Its ectopic expression significantly suppressed both Wnt10b expression and ß-catenin activity. When we altered the expression of XBP1 in 3T3-L1 cells, microRNA-148a levels changed accordingly. A potential XBP1 response element was found in the promoter region of microRNA-148a, and XBP1s directly bound to this response element as shown by point mutation analysis and chromatin immunoprecipitation assay. In addition, a microRNA-148a mimic significantly restored adipogenic potential in XBP1-deficient 3T3-L1 cells. These findings provide the first evidence that XBP1s can regulate Wnt10b by a post-transcriptional mechanism through directly inducing microRNA-148a.


Subject(s)
Adipogenesis/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Silencing , MicroRNAs/genetics , RNA Interference , Transcription Factors/genetics , Wnt Proteins/genetics , 3' Untranslated Regions , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Binding Sites , Humans , Mice , RNA Stability , Regulatory Factor X Transcription Factors , Transcription, Genetic , Wnt Signaling Pathway , X-Box Binding Protein 1
8.
FEBS J ; 281(22): 5132-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25223794

ABSTRACT

X-box binding protein 1 (XBP1), a transcription factor of the unfolded protein response, plays various roles in many biological processes. We examined its pro-adipogenic activity and target genes during adipogenic differentiation in wild-type and genetically modified 3T3-L1 cells. Signalling pathways that contribute to Xbp1 mRNA splicing, and the correlation of the transcriptionally active XBP1 isoform (XBP1s) level with body mass index and the level of peroxisome proliferator-activated receptor γ2 (PPARγ2) in human adipose tissues were also examined. The mRNA and nuclear protein expression levels of XBP1s increased immediately following hormonal induction of adipogenesis, reaching a peak at 6 h. Results from cDNA microarray and gene expression analyses using genetically modified cells indicated that PPARγ2 was a principal target of XBP1s. The XBP1s-specific binding motif, which is distinct from the CCAAT/enhancer-binding protein α binding site, was identified in the PPARγ2 promoter by site-directed mutagenesis. Fetal bovine serum, insulin, 3-isobutyl-1-methylxanthine and dexamethasone contributed independently to Xbp1 mRNA splicing. In human subcutaneous adipose tissues, the levels of both Xbp1s and Pparγ2 mRNA increased proportionally with body mass index, and there was a significant positive correlation between the two genes. These data suggest for the first time that positive regulation of PPARγ2 is a principal mechanism of XBP1s-mediated adipogenesis in 3T3-L1 cells.


Subject(s)
DNA-Binding Proteins/physiology , PPAR gamma/metabolism , Transcription Factors/physiology , Transcriptional Activation , 3T3-L1 Cells , Adipogenesis , Adult , Animals , Base Sequence , Binding Sites , Body Mass Index , Female , Gene Expression , Humans , Mice , Middle Aged , PPAR gamma/genetics , Promoter Regions, Genetic , Protein Binding , RNA Splicing , Regulatory Factor X Transcription Factors , Subcutaneous Fat , X-Box Binding Protein 1 , Young Adult
9.
FEBS Lett ; 587(11): 1644-9, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23603388

ABSTRACT

Differentiation of preadipocytes into adipocytes is controlled by various transcription factors. Recently, the pro-adipogenic function of XBP1, a transcription factor upregulated by endoplasmic reticulum stress, has been reported. In this study, we demonstrated that XBP1 suppresses the expression of Wnt10b, an anti-adipogenic Wnt, during the differentiation of 3T3-L1 preadipocytes. The expression pattern of XBP1 was reciprocal to that of Wnt10b during the early stage of adipogenesis. The intracellular protein levels of ß-catenin were negatively regulated by XBP1. Direct binding of XBP1 to the Wnt10b promoter and the subsequent decrease of the ß-catenin signalling pathway represent a novel adipogenic differentiation mechanism.


Subject(s)
Adipogenesis , DNA-Binding Proteins/metabolism , Down-Regulation , Gene Silencing , Transcription Factors/metabolism , Wnt Proteins/metabolism , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Promoter Regions, Genetic , Protein Binding , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription, Genetic , Wnt Proteins/genetics , Wnt Signaling Pathway , X-Box Binding Protein 1 , beta Catenin/genetics , beta Catenin/metabolism
10.
Exp Mol Med ; 44(9): 545-53, 2012 Sep 30.
Article in English | MEDLINE | ID: mdl-22809901

ABSTRACT

Brown adipose tissue is specialized to burn lipids for thermogenesis and energy expenditure. Second-generation antipsychotics (SGA) are the most commonly used drugs for schizophrenia with several advantages over first-line drugs, however, it can cause clinically-significant weight gain. To reveal the involvement of brown adipocytes in SGA-induced weight gain, we compared the effect of clozapine, quetiapine, and ziprasidone, SGA with different propensities to induce weight gain, on the differentiation and the expression of brown fat-specific markers, lipogenic genes and adipokines in a mouse brown preadipocyte cell line. On Oil Red-O staining, the differentiation was inhibited almost completely by clozapine (40 µM) and partially by quetiapine (30 µM). Clozapine significantly down-regulated the brown adipogenesis markers PRDM16, C/EBPß, PPARγ2, UCP-1, PGC-1α, and Cidea in dose- and time-dependent manners, whereas quetiapine suppressed PRDM16, PPARγ 2, and UCP-1 much weakly than clozapine. Clozapine also significantly inhibited the mRNA expressions of lipogenic genes ACC, SCD1, GLUT4, aP2, and CD36 as well as adipokines such as resistin, leptin, and adiponectin. In contrast, quetiapine suppressed only resistin and leptin but not those of lipogenic genes and adiponectin. Ziprasidone (10 µM) did not alter the differentiation as well as the gene expression patterns. Our results suggest for the first time that the inhibition of brown adipogenesis may be a possible mechanism to explain weight gain induced by clozapine and quetiapine.


Subject(s)
Antipsychotic Agents , Clozapine , Dibenzothiazepines , Piperazines , Thiazoles , Weight Gain/drug effects , Adipocytes, Brown/drug effects , Adipogenesis/drug effects , Adipokines/metabolism , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Clozapine/administration & dosage , Clozapine/adverse effects , Dibenzothiazepines/administration & dosage , Dibenzothiazepines/adverse effects , Gene Expression Regulation/drug effects , Humans , Mice , Piperazines/administration & dosage , Piperazines/adverse effects , Quetiapine Fumarate , Schizophrenia/drug therapy , Thiazoles/administration & dosage , Thiazoles/adverse effects
11.
Korean J Pain ; 25(2): 126-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22514783

ABSTRACT

Opioid analgesia is the primary pharmacologic intervention for managing pain. However, opioids can cause various adverse effects including pruritus, nausea, constipation, and sedation. Respiratory depression is the most fatal side effect. Therefore, cautious monitoring of respiratory status must be done after opioid administration. Here, we report a patient who suffered from respiratory depression with deep sedation and aspiration pneumonitis after intrathecal morphine administration.

12.
Korean J Physiol Pharmacol ; 15(3): 149-56, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21860593

ABSTRACT

Golgi SNAP receptor complex 1 (GS28) has been implicated in vesicular transport between intra-Golgi networks and between endoplasmic reticulum (ER) and Golgi. Additional role(s) of GS28 within cells have not been well characterized. We observed decreased expression of GS28 in rat ischemic hippocampus. In this study, we examined the role of GS28 and its molecular mechanisms in neuronal (SK-N-SH) cell death induced by hydrogen peroxide (H(2)O(2)). GS28 siRNA-transfected cells treated with H(2)O(2) showed a significant increase in cytotoxicity under glutathione (GSH)-depleted conditions after pretreatment with buthionine sulfoximine, which corresponded to an increase of intracellular reactive oxygen species (ROS) in the cells. Pretreatment of GS28 siRNA-transfected cells with p38 chemical inhibitor significantly inhibited cytotoxicity; we also observed that p38 was activated in the cells by immunoblot analysis. We confirmed the role of p38 MAPK in cotransfected cells with GS28 siRNA and p38 siRNA in the cell viability assay, flow cytometry, and immunoblot. Involvement of apoptotic or autophagic processes in the cells was not shown in the cell viability, flow cytometry, and immunoblot analyses. However, pretreatment of the cells with necrostatin-1 completely inhibited H(2)O(2)-induced cytotoxicity, ROS generation, and p38 activation, indicating that the cell death is necroptotic. Collectively these data imply that H(2)O(2) induces necroptotic cell death in the GS28 siRNA-transfected cells and that the necroptotic signals are mediated by sequential activations in RIP1/p38/ROS. Taken together, these results indicate that GS28 has a protective role in H(2)O(2)-induced necroptosis via inhibition of p38 MAPK in GSH-depleted neuronal cells.

13.
J Cell Biochem ; 112(8): 1985-96, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21433058

ABSTRACT

We examined the role of the c subunit (ATP6L) of vacuolar H(+) -ATPase and its molecular mechanisms in glial cell death induced by sodium nitroprusside (SNP). ATP6L siRNA-transfected cells treated with SNP showed a significant increase in cytotoxicity under glutathione (GSH)-depleted conditions after pretreatment with buthionine sulfoximine, but reduction of ATP6L did not affect the regulation of lysosomal pH in analyses with lysosomal pH-dependent fluorescence probes. Photodegraded SNP and ferrous sulfate induced cytotoxicity with the same pattern as that of SNP, but SNAP and potassium cyanide did not show activity. Pretreatment of the transfected cells with deferoxamine (DFO) reduced ROS production and significantly inhibited the cytotoxicity, which indicates that primarily iron rather than nitric oxide or cyanide from SNP contributes to cell death. Involvement of apoptotic processes in the cells was not shown. Pretreatment with JNK or p38 chemical inhibitor significantly inhibited the cytotoxicity, and we also confirmed that the MAPKs were activated in the cells by immunoblot analysis. Significant increase of LC3-II conversion was observed in the cells, and the conversions were inhibited by cotransfection of the MAPK siRNAs and pretreatment with DFO. Introduction of Atg5 siRNA inhibited the cytotoxicity and inhibited the activation of MAPKs and the conversion of LC3. We finally confirmed autophagic cell death and involvement of MAPKs by observation of autophagic vacuoles via electron microscopy. These data suggest that ATP6L has a protective role against SNP-induced autophagic cell death via inhibition of JNK and p38 in GSH-depleted glial cells.


Subject(s)
Autophagy/drug effects , Glutathione , Neuroglia/enzymology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Vacuolar Proton-Translocating ATPases/metabolism , Autophagy/genetics , Buthionine Sulfoximine/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Activation/genetics , Humans , Hydrogen-Ion Concentration , Lysosomes/enzymology , Lysosomes/ultrastructure , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuroglia/ultrastructure , Oxidation-Reduction/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/genetics
14.
Free Radic Res ; 45(4): 389-99, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21067284

ABSTRACT

A novel mechanism for H2O2-induced autophagic cell death in GSH-depleted RAW 264.7 cells, a murine macrophage cell line, is proposed. Under GSH-depleted conditions, H2O2-induced autophagic cell, characterized by an increased LC3-II/I ratio, a decreased level of p62 and the formation of autophagic vacuoles, was inhibited by bafilomycin A1 and by Atg5 siRNA transfection, whereas the cell death was not inhibited by zVAD-fmk, by PI3K inhibitors or by Beclin 1 siRNA transfection. In addition, H2O2 treatment reduced the activity of mTOR and promoted the ubiquitination and degradation of Rheb, a key upstream activator of mTOR. Furthermore, proteasome inhibition with MG132 restored the expression of Rheb and increased mTOR activity, resulting in an increased viability of H2O2-treated cells. Collectively, these findings demonstrate that H2O2 induces Beclin 1-independent autophagic cell death by suppressing the mTOR pathway via promoting the ubiquitination and degradation of Rheb in GSH-depleted RAW 264.7 cells.


Subject(s)
Autophagy/drug effects , Glutathione/deficiency , Hydrogen Peroxide/pharmacology , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Hydrogen Peroxide/metabolism , Lysosomes/metabolism , Macrolides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/metabolism , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism , Ubiquitination/drug effects
15.
J Biochem Mol Toxicol ; 24(5): 313-22, 2010.
Article in English | MEDLINE | ID: mdl-20201107

ABSTRACT

Previous studies reported that high levels of nitric oxide (NO) induce apoptotic cell death in osteoblasts. We examined molecular mechanisms of cytotoxic injury induced by sodium nitroprusside (SNP), a NO donor, in both glutathione (GSH)-depleted and control U2-OS osteoblasts. Cell viability was reduced by much lower effective concentrations of SNP in GSH-depleted cells compared to normal cells. The data suggest that the level of intracellular GSH is critical in SNP-induced cell death processes of osteoblasts. The level of oxidative stress due to SNP treatments doubled in GSH-depleted cells when measured with fluorochrome H2DCFDA. Pretreatment with the NO scavenger PTIO preserved the viability of cells treated with SNP. Viability of cells treated with SNP was recovered by pretreatment with Wortmannin, an autophagy inhibitor, but not by pretreatment with zVAD-fmk, a pan-specific caspase inhibitor. Large increases of LC3-II were shown by immunoblot analysis of the SNP-treated cells, and the increase was blocked by pretreatment with PTIO or Wortmannin; this implies that under GSH-depleted conditions SNP induces different molecular signaling that lead to autophagic cell death. The ultrastructural morphology of SNP-treated cells in transmission electron microscopy showed numerous autophagic vacuoles. These data suggest NO produces oxidative stress and cellular damage that culminate in autophagic cell death of GSH-depleted osteoblasts.


Subject(s)
Glutathione/metabolism , Nitroprusside/toxicity , Osteoblasts/cytology , Osteoblasts/physiology , Amino Acid Chloromethyl Ketones/metabolism , Apoptosis/physiology , Autophagy/physiology , Cell Death/physiology , Cell Line, Tumor , Cell Survival/physiology , Cells, Cultured , Humans , Nitric Oxide/metabolism , Nitric Oxide Donors , Osteoblasts/ultrastructure , Oxidative Stress/physiology , Sarcoma , Tetrazolium Salts/metabolism , Thiazoles/metabolism
16.
Prostaglandins Other Lipid Mediat ; 91(1-2): 30-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20044024

ABSTRACT

HL-60 cells treated by prostaglandin (PG) A(2) showed characteristics of apoptosis such as accumulation of hypodiploid and annexin V positive cells, condensed and fragmented nuclei, cytochrome c (Cyt C) release from mitochondria and activation of caspase-1, -2, -3, -7 and -9. PGA(2)-induced cell death was rescued by inhibitors of caspase-9 and -3, but PGA(2)-induced Cyt C release was not prevented by caspase inhibitors. During Cyt C release by PGA(2), mitochondrial transmembrane potential was maintained and mitochondrial permeability transition pore was not formed. In addition, anti-apoptotic BCL-2 family proteins like BCL-2 and BCL-XL, and ROS scavengers including ascorbic acid and 2,2,6,6-tetramethyl-1-piperidinyloxy were not able to inhibit Cyt C release as well as apoptosis by PGA(2). Finally, it was shown that PGA(2)-induced Cyt C release in vitro from purified mitochondria in the absence of cytosolic components. Furthermore, thiol-containing compounds such as N-acetylcysteine, l-cysteine and monothioglycerol prevented Cyt C release, and hence induction of apoptosis. Taken together, these results suggest that PGA(2) activates intrinsic apoptotic pathway by directly stimulating mitochondrial outer membrane permeabilization to release Cyt C, in which thiol-reactivity of PGA(2) plays a pivotal role.


Subject(s)
Apoptosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Prostaglandins A/metabolism , Prostaglandins A/pharmacology , Signal Transduction/drug effects , Animals , Caspase 3/metabolism , Cell Membrane Permeability/drug effects , Cytochromes c/metabolism , Enzyme Activation/drug effects , Free Radical Scavengers/metabolism , HL-60 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism
17.
Neurosci Lett ; 461(2): 131-5, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19539716

ABSTRACT

Oxidative stress by exposure to H(2)O(2) induces various types of cell death depending on cell type and conditions. We report herein on a study of the mechanisms underlying H(2)O(2)-induced cell death in C6 glioma cells. The findings show that H(2)O(2) triggers a caspase-independent autophagic cell death in these cells. The findings also show that H(2)O(2) induces the dephosphorylation of the mammalian target of rapamycin (mTOR) at Ser 2481 and the p70 ribosomal protein S6 kinase (p70S6K) at Thr389 in a Bcl-2/E1B 19kDa interacting protein 3 (BNIP3)-dependent manner. BNIP3 has the capacity to inhibit mTOR activity and mTOR inhibition plays a role in autophagic induction. This suggests that BNIP3 may mediate H(2)O(2)-induced autophagic cell death through the suppression of mTOR. The findings show that the down-regulation of BNIP3 by BNIP3 siRNA prevents C6 cells from undergoing H(2)O(2)-induced autophagic cell death. Collectively, these results suggest that H(2)O(2) induces autophagic cell death in C6 cells via the BNIP3-mediated suppression of the mTOR pathway.


Subject(s)
Autophagy , Hydrogen Peroxide/pharmacology , Membrane Proteins/physiology , Oxidative Stress , Protein Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Cell Line, Tumor , Glioma , Mitochondrial Proteins , Phosphorylation , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
18.
Exp Mol Med ; 41(6): 440-52, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19322020

ABSTRACT

When we treated rat bone marrow stromal cells (rBMSCs) with neuronal differentiation induction media, typical unfolded protein response (UPR) was observed. BIP/GRP78 protein expression was time-dependently increased, and three branches of UPR were all activated. ATF6 increased the transcription of XBP1 which was successfully spliced by IRE1. PERK was phosphorylated and it was followed by eIF2alpha phosphorylation. Transcription of two downstream targets of eIF2alpha, ATF4 and CHOP/GADD153, were transiently up-regulated with the peak level at 24 h. Immunocytochemical study showed clear coexpression of BIP and ATF4 with NeuN and Map2, respectively. UPR was also observed during the neuronal differentiation of mouse embryonic stem (mES) cells. Finally, chemical endoplasmic reticulum (ER) stress inducers, thapsigargin, tunicamycin, and brefeldin A, dose-dependently increased both mRNA and protein expressions of NF-L, and, its expression was specific to BIP-positive rBMSCs. Our results showing the induction of UPR during neuronal differentiations of rBMSCs and mES cells as well as NF-L expression by ER stress inducers strongly suggest the potential role of UPR in neuronal differentiation.


Subject(s)
Bone Marrow Cells/cytology , Embryonic Stem Cells/cytology , Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/drug effects , Cell Differentiation , Culture Media/pharmacology , DNA-Binding Proteins , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression/drug effects , Heat-Shock Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Nerve Tissue Proteins/genetics , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Folding , Rats , Stromal Cells
19.
Neurosci Lett ; 425(3): 183-7, 2007 Oct 02.
Article in English | MEDLINE | ID: mdl-17845832

ABSTRACT

We have isolated a gene, the c subunit (ATP6L) of vacuolar H(+)-ATPase, involved in oxidative stress response. In this study, we examined the role of ATP6L and its molecular mechanisms in glial cell death induced by H(2)O(2). Expression of the ATP6L gene was increased by H(2)O(2) treatment in C6 glial cells. ATP6L siRNA-transfected C6 cells treated with H(2)O(2) showed a significant decrease in viability. ATP6L siRNA-transfected cells that were pretreated with MEK1/2 inhibitor completely recovered cell viability. Pretreatment of the transfected cells with zVAD-fmk, a pan-specific caspase inhibitor, did not result in the recovery of cell viability, as determined by a H(2)O(2)-induced cytotoxicity assay. The ultrastructural morphology of the transfected cells as seen by the use of transmission electron microscopy showed numerous cytoplasmic autophagic vacuoles with double membrane. These results suggest that ATP6L has a protective role against H(2)O(2)-induced cytotoxicity via an inhibition of the Erk1/2 signaling pathway, leading to inhibition of autophagic cell death.


Subject(s)
Brain/enzymology , Hydrogen Peroxide/toxicity , Neuroglia/enzymology , Oxidative Stress/physiology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Autophagy/drug effects , Autophagy/physiology , Brain/physiopathology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cytoprotection/drug effects , Cytoprotection/physiology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Microscopy, Electron, Transmission , Neuroglia/drug effects , Oxidants/toxicity , Oxidative Stress/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering , Rats , Transfection , Vacuolar Proton-Translocating ATPases/genetics , Vacuoles/enzymology , Vacuoles/ultrastructure
20.
Exp Mol Med ; 38(2): 144-52, 2006 Apr 30.
Article in English | MEDLINE | ID: mdl-16672768

ABSTRACT

We have previously isolated a novel protein "B/K" that contains two C2-like domains. Here, we report the isolation and mRNA distribution of a human B/K isoform, and protein kinase A (PKA)-dependent phosphorylation of the B/K protein. The 1.5 kb human B/K cDNA clone exhibits 89% and 97% identities with rat B/K in the sequences of nucleotide and amino acid, respectively. Human B/K isoform encodes a 474 amino acid protein and shows structural features similar to the rat counterpart including two C2 domains, three consensus sequences for PKA, absence of a transmembrane region, and conservation of the N-terminal cysteine cluster. On Northern and dot blot analyses, a 3.0 kb B/K transcript was abundantly present in human brain, kidney, and prostate. Among the brain regions, strong signals were observed in the frontal and temporal lobes, the hippocampus, the hypothalamus, the amygdala, the substantia nigra, and the pituitary. Recombinant B/K proteins containing three consensus sites for PKA was very efficiently phosphorylated in vitro by PKA catalytic subunit. B/K protein which was overexpressed in LLC-PK1 cells was also strongly phosphorylated in vivo by vasopressin analog DDAVP, and PKA-specific inhibitor H 89 as well as type 2 vasopressin receptor antagonist specifically suppressed DDAVP-induced B/K phosphorylation. These results suggest that B/K proteins play a role as potential substrates for PKA in the area where they are expressed.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Phosphoproteins/metabolism , Adult , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Gene Expression Profiling , Humans , Male , Mice , Molecular Sequence Data , Phosphoproteins/genetics , Phosphorylation , Protein Isoforms/genetics , Rats , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Synaptotagmins
SELECTION OF CITATIONS
SEARCH DETAIL
...