Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
FASEB J ; 31(2): 781-792, 2017 02.
Article in English | MEDLINE | ID: mdl-27836987

ABSTRACT

Reactive oxygen species (ROS) play a role in intracellular signaling during osteoclastogenesis. We previously reported that transcriptional factor nuclear factor E2-related factor 2 (Nrf2) was exported from the nucleus to the cytoplasm by receptor activator of nuclear factor-κB ligand (RANKL), and that Nrf2 negatively regulated osteoclastogenesis via antioxidant enzyme up-regulation. Knockout mice of BTB and CNC homology 1 (Bach1)-the competitor for Nrf2 in transcriptional regulation-was known to attenuate RANKL-mediated osteoclastogenesis, although the mechanism remains unclear. Therefore, we hypothesized that RANKL could be involved in the nuclear translocation of Bach1, which would attenuate Nrf2-mediated antioxidant enzymes, thereby augmenting intracellular ROS signaling in osteoclasts. RANKL induced Bach1 nuclear import and Nrf2 nuclear export. Induction of Bach1 nuclear export increased Nrf2 nuclear import, augmented antioxidant enzyme expression, and, thus, diminished RANKL-mediated osteoclastogenesis via attenuated intracellular ROS signaling. Finally, an in vivo mouse bone destruction model clearly demonstrated that induction of Bach1 nuclear export inhibited bone destruction. In this study, we report that RANKL favors osteoclastogenesis via attenuation of Nrf2-mediated antioxidant enzyme expression by competing with Bach1 nuclear accumulation. Of importance, induction of Bach1 nuclear export activates Nrf2-dependent antioxidant enzyme expression, thereby attenuating osteoclastogenesis. Bach1 nuclear export might be a therapeutic target for such bone destructive diseases as rheumatoid arthritis, osteoporosis, and periodontitis.-Kanzaki, H., Shinohara, F., Itohiya, K., Yamaguchi, Y., Katsumata, Y., Matsuzawa, M., Fukaya, S., Miyamoto, Y., Wada, S., Nakamura, Y. RANKL induces Bach1 nuclear import and attenuates Nrf2-mediated antioxidant enzymes, thereby augmenting intracellular reactive oxygen species signaling and osteoclastogenesis in mice.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , NF-E2-Related Factor 2/metabolism , Osteoclasts/physiology , RANK Ligand/metabolism , Reactive Oxygen Species/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Survival , Gene Expression Regulation/physiology , Mice , NF-E2-Related Factor 2/genetics , RANK Ligand/genetics , RAW 264.7 Cells , Signal Transduction/physiology
2.
Sci Rep ; 6: 32259, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27573075

ABSTRACT

Interferon-gamma (IFN-γ) is a pleiotropic cytokine that exerts anti-tumor and anti-osteoclastogenic effects. Although transcriptional and post-transcriptional regulation of IFN-γ is well understood, subsequent modifications of secreted IFN-γ are not fully elucidated. Previous research indicates that some cancer cells escape immune surveillance and metastasize into bone tissue by inducing osteoclastic bone resorption. Peptidases of the a-disintegrin and metalloproteinase (ADAM) family are implicated in cancer cell proliferation and tumor progression. We hypothesized that the ADAM enzymes expressed by cancer cells degrades IFN-γ and attenuates IFN-γ-mediated anti-tumorigenic and anti-osteoclastogenic effects. Recombinant ADAM17 degraded IFN-γ into small fragments. The addition of ADAM17 to the culture supernatant of stimulated mouse splenocytes decreased IFN-γ concentration. However, ADAM17 inhibition in the stimulated mouse T-cells prevented IFN-γ degradation. ADAM17-expressing human breast cancer cell lines MCF-7 and MDA-MB-453 also degraded recombinant IFN-γ, but this was attenuated by ADAM17 inhibition. Degraded IFN-γ lost the functionality including the inhibititory effect on osteoclastogenesis. This is the first study to demonstrate the extracellular proteolytic degradation of IFN-γ by ADAM17. These results suggest that ADAM17-mediated degradation of IFN-γ may block the anti-tumorigenic and anti-osteoclastogenic effects of IFN-γ. ADAM17 inhibition may be useful for the treatment of attenuated cancer immune surveillance and/or bone metastases.


Subject(s)
ADAM17 Protein/metabolism , Interferon-gamma/metabolism , Recombinant Proteins/metabolism , T-Lymphocytes/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Humans , Interferon-gamma/genetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Proteolysis/drug effects , RAW 264.7 Cells , RNA Interference , T-Lymphocytes/drug effects
3.
Redox Biol ; 8: 186-91, 2016 08.
Article in English | MEDLINE | ID: mdl-26795736

ABSTRACT

It has been reported that reactive oxygen species (ROS), such as hydrogen peroxide and superoxide, take part in osteoclast differentiation as intra-cellular signaling molecules. The current assumed signaling cascade from RANK to ROS production is RANK, TRAF6, Rac1, and then Nox. The target molecules of ROS in RANKL signaling remain unclear; however, several reports support the theory that NF-κB signaling could be the crucial downstream signaling molecule of RANKL-mediated ROS signaling. Furthermore, ROS exert cytotoxic effects such as peroxidation of lipids and phospholipids and oxidative damage to proteins and DNA. Therefore, cells have several protective mechanisms against oxidative stressors that mainly induce cytoprotective enzymes and ROS scavenging. Three well-known mechanisms regulate cytoprotective enzymes including Nrf2-, FOXO-, and sirtuin-dependent mechanisms. Several reports have indicated a crosslink between FOXO- and sirtuin-dependent regulatory mechanisms. The agonists against the regulatory mechanisms are reported to induce these cytoprotective enzymes successfully. Some of them inhibit osteoclast differentiation and bone destruction via attenuation of intracellular ROS signaling. In this review article, we discuss the above topics and summarize the current information available on the relationship between cytoprotective enzymes and osteoclastogenesis.


Subject(s)
Cell Differentiation/genetics , Osteogenesis/genetics , Oxidative Stress/genetics , Signal Transduction , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Heme Oxygenase-1/genetics , Humans , Lipid Peroxidation/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Sirtuins/genetics
4.
Free Radic Biol Med ; 77: 239-48, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224039

ABSTRACT

It has been reported that reactive oxygen species (ROS) play a role as intracellular signaling molecules in RANKL stimulation. Previously we demonstrated that induction of cytoprotective enzyme expression by Nrf2-gene transfer successfully ameliorated RANKL-dependent osteoclastogenesis. In the present study, we hypothesized that Nrf2 activation by inhibiting ubiquitination and degradation of Nrf2 by ETGE-peptide would induce Nrf2-dependent cytoprotective enzyme expression, attenuate ROS signaling, and thereby inhibit RANKL-dependent osteoclastogenesis. ETGE-peptide containing a cell-permeable sequence (seven consecutive arginine; 7R-ETGE) was applied to a mouse macrophage cell-line RAW 264.7 cell or a primary macrophage culture. ETGE-peptide prevents Keap1 from binding to Nrf2. Nrf2 nuclear translocation and Nrf2-dependent cytoprotective enzyme induction was observed. The effects of 7R-ETGE on RANKL-dependent induction of intracellular ROS levels and osteoclastogenesis were examined. Finally, the protective effect of 7R-ETGE on RANKL-mediated bone destruction was investigated in mice. 7R-ETGE dose-dependently induced nuclear Nrf2, followed by the induction of cytoprotective enzyme expression at both the gene and protein level. 7R-ETGE inhibited upregulation of intracellular ROS levels by RANKL stimulation, and osteoclastogenesis was attenuated. Of particular interest was that local injection of 7R-ETGE ameliorated RANKL-mediated bone destruction. Local induction of nuclear Nrf2 by protein transduction is a potential novel therapeutic target for bone destruction diseases such as periodontitis and rheumatoid arthritis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Cell Nucleus/metabolism , NF-E2-Related Factor 2/metabolism , Oligopeptides/pharmacology , Osteoclasts/physiology , Active Transport, Cell Nucleus , Animals , Cell Line , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , RANK Ligand/pharmacology , RANK Ligand/physiology , Reactive Oxygen Species/metabolism
5.
J Biol Chem ; 288(32): 23009-20, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23801334

ABSTRACT

Reactive oxygen species (ROS) act as intracellular signaling molecules in the regulation of receptor activator of nuclear factor-κB ligand (RANKL)-dependent osteoclast differentiation, but they also have cytotoxic effects that include peroxidation of lipids and oxidative damage to proteins and DNA. Cellular protective mechanisms against oxidative stress include transcriptional control of cytoprotective enzymes by the transcription factor, nuclear factor E2-related factor 2 (Nrf2). This study investigated the relationship between Nrf2 and osteoclastogenesis. Stimulation of osteoclast precursors (mouse primary peritoneal macrophages and RAW 264.7 cells) with RANKL resulted in the up-regulation of kelch-like ECH-associated protein 1 (Keap1), a negative regulator of Nrf2. It also decreased the Nrf2/Keap1 ratio, and it down-regulated cytoprotective enzymes (heme oxygenase-1, γ-glutamylcysteine synthetase, and glucose-6-phosphate dehydrogenase). Nrf2 overexpression up-regulated the expression of cytoprotective enzymes, decreased ROS levels, decreased the number of tartrate-resistant acid phosphatase-positive multinucleated cells, reduced marker genes for osteoclast differentiation, and attenuated bone destruction in both in vitro and in vivo models. Overexpression of Keap1 or RNAi knockdown of Nrf2 exerted the opposite actions. In addition, in vivo local Nrf2 overexpression attenuated lipopolysaccharide-mediated RANKL-dependent cranial bone destruction in vivo. This is the first study to show that the Keap1/Nrf2 axis regulates RANKL-dependent osteoclastogenesis through modulation of intracellular ROS signaling via expression of cytoprotective enzymes. This raises the exciting possibility that the Keap1-Nrf2 axis may be a therapeutic target for the treatment of bone destructive disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Cytoskeletal Proteins/metabolism , Macrophages, Peritoneal/metabolism , NF-E2-Related Factor 2/metabolism , Osteoclasts/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Cytoskeletal Proteins/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Kelch-Like ECH-Associated Protein 1 , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/pathology , Mice , NF-E2-Related Factor 2/genetics , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/metabolism , Osteolysis/pathology , RANK Ligand/biosynthesis
6.
Cancer Chemother Pharmacol ; 67(5): 1129-36, 2011 May.
Article in English | MEDLINE | ID: mdl-20680277

ABSTRACT

PURPOSE: Although selenium compounds possess chemotherapeutic features by inducing apoptosis in cancer cells with trivial side effects on normal cells, the mechanisms underlying its anti-cancer activity are insufficiently understood at the present. In this study, we investigated the effects of rapamycin on apoptosis induced by seleno-L-methionine (SeMet) or selenite in A549 cells. METHODS: The effects of Se compounds, SeMet and selenite, on cell proliferation, apoptosis and its signaling pathway were investigated in established human adenocarcinoma cell line (A549). Cancer cells were treated with each Se during different periods. Cell apoptosis and signaling molecules were analyzed by flow cytometry (TUNEL method) or immunoblotting, respectively. RESULTS: SeMet induces reactive oxygen species generation associated with the induction of apoptosis, because pretreatment of cells with N-acetyl-L-cysteine completely blocked SeMet-induced apoptosis. We also found that rapamycin completely suppressed the apoptosis of cells treated by SeMet, but not selenite. SeMet-induced apoptosis is significantly downregulated in combination with PI3 K family inhibitors (LY294002, wortmannin, PI-103, and 3-methyladenine). In addition, ROS generation was included in downstream signaling events associated with the phosphorylation of mTOR, because pretreatment of cells with rapamycin inhibited ROS generation. CONCLUSION: These results suggest that SeMet-induced apoptosis is affected by the Akt/mTOR/ROS pathway in A549 cells. Akt serves an anti-survival function in the system of SeMet-treated lung cancer cells, but autophagic signaling remained unsolved.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Selenomethionine/pharmacology , Sirolimus/pharmacology , Adenocarcinoma , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Lung Neoplasms , Microtubule-Associated Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Sodium Selenite/pharmacology , TOR Serine-Threonine Kinases/metabolism
7.
Cancer Chemother Pharmacol ; 66(3): 475-84, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19940991

ABSTRACT

PURPOSE: Selenium (Se) compounds are well known to inhibit cell proliferation and induce cell death in human cancer cells. Respective chemical forms of Se are intracellularly metabolized via complicated pathways, which target distinct molecules and exhibit varying degrees of anti-carcinogenicity in different cancer types; however, the precise mechanisms by which Se activates apoptosis remain poorly understood. METHODS: The effects of Se compounds, Se-methylselenocysteine (MSC), selenomethionine (SeMet), and selenite on cell proliferation, apoptosis and its pathway in established human carcinoma cell lines (HSC-3, -4, A549, and MCF-7) were investigated. Cancer cells were treated with each Se compound during different periods. Cell apoptosis, caspase activity and ER stress markers were analyzed by flow cytometric or immunoblotting analysis, respectively. RESULTS: We examined four cell lines for their sensitivity to MSC and SeMet in comparison with selenite. SeMet increased apoptotic cells in p53-positive A549 cells, whereas MSC increased apoptotic cells in p53-mutated HSC-3 cells. High activities of caspase-3, -8 and -9 were observed during apoptosis, and a pan-caspase inhibitor, z-VAD-fmk, rescued the cell viability of HSC-3 cells exposed to MSC. In addition, the occurrence of endoplasmic reticulum (ER) stress was suggested by the observation that levels of phosphorylated eIF2alpha and caspase-12 activity are increased in Se-treated cells. Selenite and MSC were accompanied with the concurrent reduction of phosphorylated Akt levels, and the inhibitory effects of these Se compounds on vascular endothelial growth factor expression were observed with identical patterns. CONCLUSION: The present findings demonstrate that Se-induced apoptosis in carcinoma cells is basically a caspase-dependent process involving complicated mechanisms. Activation of both the intrinsic apoptotic pathway and ER stress pathway plays a major and concurrent role, while p53 activation seems to have only a functional role in SeMet.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cysteine/analogs & derivatives , Neoplasms/drug therapy , Organoselenium Compounds/pharmacology , Selenomethionine/pharmacology , Sodium Selenite/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 3/drug effects , Caspase 8/drug effects , Caspase 8/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cysteine/pharmacology , Flow Cytometry , Humans , Neoplasms/pathology , Selenocysteine/analogs & derivatives , Tumor Suppressor Protein p53/metabolism
8.
Cancer Chemother Pharmacol ; 64(6): 1115-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19280190

ABSTRACT

PURPOSE: The histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances cisplatin [cis-diammine dichloroplatinum (II)] (CDDP)-induced apoptosis in the oral squamous cell carcinoma (OSCC) cell line by complex, multifunctional mechanisms. We investigated the role of endoplasmic reticulum (ER) stress in the enhancing effect of SAHA on CDDP, compared with the ER stressor thapsigargin. METHODS: We chose OSCC cell line HSC-3 to ascertain the mechanism of SAHA-enhanced cytotoxicity among various cell lines. HSC-3 cells were incubated with CDDP/SAHA for 48 h, followed by the assessment of cell chemosensitivity to CDDP with MTT and TUNEL assays. Western blot analysis was used to detect the expressions of ER-related molecules, and flow cytometry was used to monitor caspase activity. RESULTS: Treatment with CDDP/SAHA potently induced apoptosis in HSC-3 cells with a significant increase in caspase-4 and -12 functions. For example, 60% of cells became apoptotic after 48 h of treatment with CDDP/SAHA. In addition, SAHA alone rapidly induced sustained phosphorylation of eukaryotic translation initiation factor-2 (eIF2)alpha, which is up-regulated during ER stress. Inhibition of ER stress by salubrinal, an inhibitor of eIF2alpha dephosphorylation, abrogated SAHA's enhancement of CDDP cytotoxicity. Levels of phospho-Akt are decreased in SAHA-treated cells, and this is in turn associated with increased activity of protein phosphatase 1 (PP1) by SAHA, the phosphatase upstream of Akt. CONCLUSION: These data indicate that up-regulation of specific-ER stress-associated events is an integral part of the mechanism by which SAHA enhances CDDP-induced apoptosis, and PP1 up-regulation followed by Akt dephosphorylation plays an important role in SAHA-enhanced CDDP apoptosis.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cisplatin/pharmacology , Endoplasmic Reticulum/drug effects , Hydroxamic Acids/pharmacology , Mouth Neoplasms/pathology , Apoptosis/physiology , Carcinoma, Squamous Cell/metabolism , Caspase 12/metabolism , Caspase Inhibitors , Caspases, Initiator/metabolism , Cell Line, Tumor , Cinnamates/pharmacology , Cisplatin/toxicity , Drug Synergism , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Eukaryotic Initiation Factor-2/metabolism , Female , Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors , Humans , In Situ Nick-End Labeling , Membrane Glycoproteins/metabolism , Mouth Neoplasms/metabolism , Phosphorylation/drug effects , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thapsigargin/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Tunicamycin/pharmacology , Valproic Acid/pharmacology , Vorinostat
9.
Cancer Chemother Pharmacol ; 64(2): 223-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18830594

ABSTRACT

PURPOSE: During tumorigenesis, tumor suppressor and tumor-related genes are commonly silenced by aberrant DNA methylation in their promoter regions, which is one of the important determinants of susceptibility to 5-fluorouracil (5-FU) in oral squamous cell carcinoma (OSCC) cells. Here, we examine the chemotherapeutic efficacy of epigenetic agents on 5-FU cytotoxicity. METHOD: We investigated the effect of a DNA methyltransferase (DNMT) inhibitor, zebularine (Zeb), on the chemosensitivity of 5-FU and cisplatin (CDDP) by MTT and TUNEL methods, and compared the molecular mechanism of action with those of a GSK3beta inhibitor, LiCl, and an Hsp90 inhibitor, 17-AAG. RESULTS: A significant apoptotic effect by a combination of Zeb or 17-AAG was found in CDDP treatment; however, considerable suppression of 5-FU-induced apoptosis was observed after incubation with Zeb, 17-AAG, or LiCl. Zeb's suppressive effects were associated with activation of the cAMP/PKA/CREB pathway, differing from mechanisms of 17-AAG and LiCl. Suppression of 5-FU-induced apoptosis by Zeb was not associated with increased Bcl-2 and Bcl-xL expressions dependent on transcription factor CREB, and with the expression level of thymidylate synthase. CONCLUSIONS: In the present study, we identified a more detailed mechanism of action by which Zeb suppresses 5-FU-induced apoptosis. These results indicate that combination therapies have to be carefully investigated due to potential harmful effects in the clinical application of DNMT inhibitors.


Subject(s)
Apoptosis/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytidine/analogs & derivatives , Fluorouracil/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Adjuvants, Immunologic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytidine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Fluorouracil/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , In Situ Nick-End Labeling , Lactams, Macrocyclic/pharmacology , Lithium Chloride/pharmacology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , NF-kappa B/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects
10.
Mol Med Rep ; 1(4): 465-71, 2008.
Article in English | MEDLINE | ID: mdl-21479433

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent inducer of angiogenesis in oral squamous cell carcinoma (OSCC). In this study, we used the novel DNA methyltransferase inhibitor zebularine (Zeb) to investigate epigenetic influences on the secretion of VEGF-A in the OSCC cell line HSC-3. Under normoxic conditions, we found that Zeb inhibited secretion in a dose-dependent manner by reducing the activity of hypoxia-inducible factor-1α (HIF-1α). Treatment of the cells with the proteasome inhibitor MG132 protected the HIF-1α protein from Zeb-mediated epigenetic regulation. In addition, our study revealed that neither the PI3K/Akt nor the p53 signaling pathway is required for Zeb-induced HIF-1α degradation. In short, Zeb influenced the stability of the HIF-1α protein and the activity of its targets, such as VEGF, in HSC-3 cells in normoxic conditions. This study has laid the foundation for a novel anti-cancer approach, which may find applications in molecular staging.

11.
Int J Oncol ; 31(6): 1449-56, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982671

ABSTRACT

Epigenetic alterations such as histone acetylation and DNA methylation play an important role in the regulation of gene expression for cell cycles and apoptosis that may affect the chemosensitivity of cancers. Previously, we have reported that the combination of suberoylanilide hydroxamic acid (SAHA), a newly developed histone deacetylase inhibitor, with cisplatin (CDDP) possessed synergistic cytotoxicity against human oral squamous cell carcinoma (OSCC) cell line HSC-3. In this study, we used a novel DNA methyltransferase inhibitor, zebularine (Zeb), to investigate the epigenetic influence on the sensitivity of carcinoma cell lines to 5-fluorouracil (5-FU) or CDDP by evaluating apoptotic inducibility. Treatment with CDDP or 5-FU either alone or in combination with Zeb or SAHA continued for 48 or 72 h. In HSC-3 cells, Zeb had chemosensitive efficacy with CDDP, but not 5-FU, whereas SAHA showed efficacy with both CDDP and 5-FU. We showed that Zeb has strong anti-proliferative activity against HSC-3 cells, shown by decreased cellular growth and G2/M cell cycle phase accumulation. Furthermore, DNA methylation could be a regulatory mechanism for dihydropyrimidine dehydrogenase (DPD), known to be a principal factor in 5-FU resistance. CDHP (5-chloro-2,4-dihydroxypyridine), an inhibitor of DPD, had an enhancing effect on the apoptotic ability of 5-FU alone or 5-FU/Zeb combination. In conclusion, the present study suggests that low-dose (IC20) Zeb may sensitize cancer cells to CDDP, which may be an important characteristic for solid cancer treatment, and that DPD and other agents activated by Zeb in cancer cells could be an inhibitory factor in the response to apoptosis induced by 5-FU.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Cytidine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic , Fluorouracil/pharmacology , Mouth Neoplasms/drug therapy , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cytidine/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mouth Neoplasms/pathology , Vorinostat
12.
Int J Oncol ; 30(5): 1181-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17390020

ABSTRACT

In the present study, the precise mechanism of the enhancing action of histone deacetylase (HDAC) inhibitors on cisplatin (CDDP)-induced apoptosis was investigated using suberoylanilide hydroxamic acid (SAHA) in human oral squamous cell carcinoma cells (HSC-3). HDAC inhibitors are promising novel compounds for the treatment of cancer, which cooperate with chemotherapeutic agents to induce apoptosis. Apoptosis enhancement of HSC-3 cells by SAHA was accompanied by the activation of caspase-3, -8 and -9, suggesting a mitochondrial-dependent amplification loop. Concomitant treatment (CDDP/SAHA) of cells resulted in the most effective enhancement of apoptosis compared to other timing combinations. By means of cell-cycle synchronization, G0/G1-phase cells proved to be a more sensitive fraction to SAHA action than their synchronized counterparts in other phases. Furthermore, cells treated with SAHA revealed a decrease in intracellular reduced glutathione (GSH) contents. Of importance, the GSH synthesis inhibitor, diethyl maleate, decreased intracellular GSH and enhanced CDDP-induced apoptosis in a similar pattern of timing to SAHA. Thus, SAHA appears to disrupt the intracellular redox balance, which causes maximal apoptosis at the G0/G1 phase arrested by CDDP in HSC-3 cells. These results demonstrate that HDAC inhibitors not only cause a change in the histone acetylation status, but are also able to influence the apoptotic process at several levels, and GSH plays a key role in governing SAHA-dependent enhancement of CDDP-induced apoptosis.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Mouth Neoplasms/drug therapy , Apoptosis , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Drug Interactions , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Humans , Vorinostat
13.
Int J Oncol ; 27(2): 489-95, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010432

ABSTRACT

Selenium (Se) is a very effective anti-cancer agent. We studied the effects of inorganic Se compounds on induction of apoptosis by which Se compounds exert cancer chemopreventive activity. With the use of HSC-3 human oral squamous cell carcinoma cells, the present study showed that treatment with Se for 72 h, in the form of SeO2 and Na2SeO3, but not Na2SeO4, markedly induced apoptosis in a dose-dependent manner. Treatment of HSC-3 cells with 100 microM SeO2 resulted in the caspase-3-like and -9-like activation. Se compounds induced a loss of mitochondrial membrane potential (DeltaPsim), but did not induce the generation of reactive oxygen species. Treatment with SeO2 for 18 h resulted in 80% loss of reduced glutathione (GSH), which is known to be involved in the metabolism of Se. Treatment with N-acetyl-L-cysteine, or exogenous GSH, prevented the SeO2-induced apoptosis. Treatment with GSH led to the partial reverse in reduction of DeltaPsim caused by SeO2, while buthionine sulfoximine augmented the SeO2- or Na2SeO3-induced apoptosis. These results suggest that modulation of the mitochondrial redox equilibrium by Se contributes to the mitochondrial pathway, regulating caspase-9-mediated apoptosis without a concurrent increase in ROS.


Subject(s)
Apoptosis/drug effects , Glutathione/physiology , Mitochondria/drug effects , Selenium Compounds/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/physiopathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Glutathione/metabolism , Glutathione/pharmacology , Humans , In Situ Nick-End Labeling , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Mitochondria/physiology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/physiopathology , Reactive Oxygen Species/metabolism
14.
Int J Mol Med ; 15(4): 667-73, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15754030

ABSTRACT

Apoptosis induced by docetaxel that interferes with microtubule polymerization dynamics and is used clinically to treat advanced cancers, has not been fully defined in squamous cell carcinoma. In this study, apoptotic events involved in docetaxel treatment were investigated. When the human oral squamous cell carcinoma cell line HSC-3 was exposed to docetaxel for 72 h, a dose-dependent effect was observed in apoptosis using the TUNEL method. We observed activation of caspase cascade including activities like caspase-3, -8, and -9. And the pan-caspase inhibitor z-VAD-fmk prevented apoptosis induced by docetaxel (0.1 microM), showing participation of caspases in this process. Since an antagonistic CD95-antibody (ZB4) exerted no effect on docetaxel-induced apoptosis, CD95/CD95L interaction was not involved in this pathway. The caspase-8-like activity was inhibited not only by IETD-fmk (caspase-8) but also by DEVD-fmk (caspase-3). The results indicate that the caspase-8-like activation occurred downstream of DEVDase. Docetaxel promoted the formation of reactive oxygen species (ROS) in mitochondria, and preincubation of cells with anti-oxidants such as N-acetyl cysteine and pyrrolidine dithiocarbamate, protected against apoptosis mediated by docetaxel. Furthermore, treatment with docetaxel elicited reduction of mitochondrial membrane potential, and release of cytochrome c to cytosol, after 48 h of treatment. We observed binding activity to NF-kappaB consensus site and interference with the mitochondrial function via NF-kappaB after docetaxel treatment. Preventing pro-apoptotic property of NF-kappaB inhibited docetaxel-induced apoptosis. Thus, these results suggest that, following the activation of NF-kappaB by docetaxel, apoptosis is elicited through a mitochondria-dependent pathway.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Mitochondria/metabolism , Mouth Neoplasms/drug therapy , NF-kappa B/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Carcinoma, Squamous Cell/metabolism , Caspase 8 , Caspases/metabolism , Cytochromes c/metabolism , Docetaxel , Humans , Membrane Potentials/drug effects , Mitochondria/drug effects , Mouth Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Taxoids/pharmacology , Tumor Cells, Cultured
15.
Int J Mol Med ; 14(2): 241-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254772

ABSTRACT

In this study, we demonstrated evidence for the induction of CD80+ monocytes by staphylococcal enterotoxin B (SEB) via caspase actions. Pre-treatment with pyrrolidine dithiocarbamate (PDTC), a potent inhibitor of NF-kappaB, resulted in a significant reduction in the percentage of SEB- and interferon-gamma (IFN-gamma) (produced by SEB) -induced CD80+ monocytes. SEB and IFN-gamma activated NF-kappaB, which was inhibited by PDTC. SEB induced the activation of caspase-3 and -8, and pre-treatment with z-VAD-fmk, a broad-spectrum inhibitor of caspases, prevented the induction of apoptosis. Treating with z-VAD-fmk resulted in a reduction of the generation of CD80+ monocytes. These results indicated that CD80 driven by NF-kappaB is regulated by the enzymatic actions of caspases, which allows monocytes to participate in massive T-cell activation.


Subject(s)
B7-1 Antigen/biosynthesis , Caspases/physiology , Monocytes/metabolism , Superantigens/metabolism , Adult , Amino Acid Chloromethyl Ketones/pharmacology , Antibodies, Monoclonal/chemistry , Apoptosis , B7-1 Antigen/chemistry , Caspase 3 , Caspase 8 , Caspases/metabolism , Cells, Cultured , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enterotoxins/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/metabolism , Middle Aged , NF-kappa B/metabolism , Pyrrolidines/pharmacology , RNA/chemistry , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Thiocarbamates/pharmacology , Up-Regulation
16.
Leuk Res ; 28(1): 63-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14630082

ABSTRACT

One of the mechanisms repressing apoptosis in tumor cells can involve the expression of anti-apoptotic NF-kappaB target genes. In this study, we demonstrated that a potent NF-kappaB inhibitor, Nalpha-tosyl-L-lysinyl chloromethyl ketone (TLCK), inhibits apoptosis of THP-1 cells triggered by etoposide (VP16), and actinomycin D (ACT D) or cycloheximide inhibits apoptosis. However, persistent activation of NF-kappaB by lipopolysaccharide (LPS) led to the survival of leukemic cells against VP16-induced apoptosis. Thus, the molecular events (Bax/X-chromosome-linked IAP (XIAP)) occurring downstream of NF-kappaB activation during VP16 and/or LPS stimulation may become important to understand the multiple effects of NF-kappaB.


Subject(s)
Apoptosis , Etoposide/pharmacology , Leukemia/pathology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Humans , Inhibitor of Apoptosis Proteins , Leukemia/metabolism , NF-kappa B/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tosyllysine Chloromethyl Ketone/pharmacology , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...