Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Cancer Prev ; 16(6): 505-10, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18090122

ABSTRACT

Sulforaphane is an antioxidant and a potent stimulator of natural detoxifying enzyme and associated with lowered risk of cancer that is associated with the consumption of cruciferous vegetables. The chemopreventive effects of SFN was investigated using the MCF-7 human breast cancer cells and the M13SV1-immortalized human breast luminal epithelial cells. Sulforaphane reduced proliferation in MCF-7 cells and inhibited cyclooxygenase-2 expression in M13SV1 cells treated with 12-O-tetradecanoylphorbol-13-acetate (TPA). The chemopreventive effects of sulforaphane were associated with p38 mitogen-activated protein kinase suggest its important role in cell survival/apoptosis regulation and stabilization of cyclooxygenase-2. Sulforaphane upregulates p38 in MCF-7 cells and prevented TPA-reduced phosphorylation of p38 in M13SV1 cells, but activated caspase-7 associated with apoptosis in MCF-7 cells. These results suggest that sulforaphane may be an alternative candidate for targeted prevention of ER-positive and cyclooxygenase-2-induced phenotypes and breast cancer.


Subject(s)
Breast Neoplasms/prevention & control , Caspase 7/metabolism , Cyclooxygenase 2/genetics , Receptors, Estrogen/genetics , Thiocyanates/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Transformed , Cell Nucleus Shape/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoprotection/drug effects , Cytoprotection/genetics , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isothiocyanates , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/prevention & control , Poly(ADP-ribose) Polymerases/metabolism , Sulfoxides , Thiocyanates/pharmacology , Treatment Outcome , Tumor Cells, Cultured
2.
Eur J Cancer Prev ; 16(4): 342-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17554207

ABSTRACT

Oligonol is a novel catechin-rich biotechnology product. The role of oligonol in modulating intracellular signaling mechanisms was investigated with the view of demonstrating its potential chemopreventive effect and the ability to inhibit cell proliferation using the estrogen-responsive MCF-7 and the estrogen-unresponsive MDA-MB-231 human breast cancer cell lines. Cell survival assay indicated that Oligonol was cytotoxic to both cells. Oligonol triggered apoptosis as revealed by the morphological features typical of nucleus staining and the accumulation of sub-G1 peak. Treatment with 25 microg/ml Oligonol resulted in an activation of caspase-7 and up-regulation of Bad on MCF-7 cells, while the Oligonol (20 microg/ml) induced up-regulation of Bcl-2 protein in a time-response manner on MDA-MB-231 cells. ERK1/2 in both cells were inactivated after Oligonol treatment in a time-dependent manner, and also inactivated upstream MEK1/2. Oligonol triggers apoptosis in MCF-7 and MDA-MB-231 cells through the modulation of pro-apoptotic Bcl-2 family proteins and MEK/ERK signaling pathway.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Catechin/analogs & derivatives , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Phenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Caspase 7/metabolism , Catechin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA, Neoplasm/metabolism , Female , Humans , Phosphorylation/drug effects , bcl-Associated Death Protein/metabolism
3.
Cancer Res ; 66(12): 6063-71, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778178

ABSTRACT

The epithelial components of the mammary gland are thought to arise from stem cells with a capacity for self-renewal and multilineage differentiation. Furthermore, these cells and/or their immediate progeny may be targets for transformation. We have used both in vitro cultivation and a xenograft mouse model to examine the role of hedgehog signaling and Bmi-1 in regulating self-renewal of normal and malignant human mammary stem cells. We show that hedgehog signaling components PTCH1, Gli1, and Gli2 are highly expressed in normal human mammary stem/progenitor cells cultured as mammospheres and that these genes are down-regulated when cells are induced to differentiate. Activation of hedgehog signaling increases mammosphere-initiating cell number and mammosphere size, whereas inhibition of the pathway results in a reduction of these effects. These effects are mediated by the polycomb gene Bmi-1. Overexpression of Gli2 in mammosphere-initiating cells results in the production of ductal hyperplasia, and modulation of Bmi-1 expression in mammosphere-initiating cells alters mammary development in a humanized nonobese diabetic-severe combined immunodeficient mouse model. Furthermore, we show that the hedgehog signaling pathway is activated in human breast "cancer stem cells" characterized as CD44+CD24-/lowLin-. These studies support a cancer stem cell model in which the hedgehog pathway and Bmi-1 play important roles in regulating self-renewal of normal and tumorigenic human mammary stem cells.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Hedgehog Proteins , Humans , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction , Trans-Activators/agonists , Trans-Activators/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
4.
Eur J Cancer Prev ; 15(1): 57-63, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16374231

ABSTRACT

Sodium butyrate (NaBu) is known to exhibit anti-cancer effects via the differentiation and apoptosis of various carcinoma cells. However, the mechanism by which NaBu induces apoptosis and the involvement of protein kinases during apoptosis is not completely understood. To investigate the underlying pathways, we performed cell culture experiments in androgen-independent human prostate cancer (DU145 cells) focusing on various protein kinases. NaBu causes concentration-dependent cell detachment and growth inhibition. Exposure of DU145 cells to NaBu for 24 h caused a strong apoptotic effect with 26% nuclear fragmentation and condensation. In addition, NaBu induced caspase-3 and poly-ADP ribose polymerase cleavage and up-regulation of bax, suggesting that mitochondrial damage is involved in NaBu-induced caspase-dependent apoptosis. Interestingly, NaBu stimulated p38 mitogen-activated protein kinase (MAPK) and c-Jun NH2-terminal kinase (JNK) activation, but not extracellular signal-regulated kinase 1/2 activation during apoptosis. Furthermore, NaBu up-regulated total protein levels and phospho forms of MAPK kinase 3 (MKK3) and MAPK kinase 4 (MKK4) as the upstream kinases of p38 MAPK and JNK independently of oxidative stress. Taken together, it is suggested that NaBu can be a promising chemopreventive agent for prostate cancer and the p38 MAPK and JNK pathways have critical roles in NaBu-induced apoptosis in DU145 cells.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Cell Proliferation/drug effects , Histamine Antagonists/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Butyrates/therapeutic use , Caspase 3 , Caspases/metabolism , Dose-Response Relationship, Drug , Enzyme Precursors/metabolism , Humans , Immunoblotting , Male , Prostatic Neoplasms/prevention & control , Tumor Cells, Cultured
5.
Cancer Lett ; 241(2): 301-8, 2006 Sep 28.
Article in English | MEDLINE | ID: mdl-16337085

ABSTRACT

The histone deacetylase (HDAC) inhibitors, trichostatin A (TSA) and sodium butyrate (NaBu) are considered as potent therapeutic agents for cancer treatment presenting therapeutic benefits with less risk of side effects. The microbial metabolite, TSA is a potent reversible and highly specific inhibitor of mammalian histone deacetylases. NaBu causes hyperacetylation of core histones with effects similar to TSA but it is not a specific inhibitor of HDACs. The gap junction is a channel in the plasma membrane of most cell types which allows direct communication (gap junctional intercellular communication; GJIC) of small molecules and ions. Modulation of GJIC is a known cellular event associated with tumor promotion. The effects of NaBu and TSA on the H(2)O(2)- and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced GJIC inhibition of WB cells and the mechanisms involved in the process were assessed. TSA and NaBu exerted differential preventive effects on the H(2)O(2) and TPA-induced inhibition of GJIC as well as hyperphosphorylation of connexin43 (Cx43) in WB-F344 rat liver epithelial cells (WB cells). NaBu prevented the TPA-induced GJIC inhibition via ERK1/2 inactivation whilst TSA restored the H(2)O(2)-induced GJIC inhibition and Cx43 hyperphosphorylation by preventing p38 MAP kinase. The inhibition of tyrosine phosphorylation and down-regulation of src protein observed may also contribute to Connexin 43 dephosphorylation and GJIC restoration by TSA and NaBu partly through depletion of src protein pool. Thus, TSA and NaBu exert differential effects on chemically induced GJIC inhibition via modulation of MAP kinases and partly, tyrosine kinases.


Subject(s)
Butyric Acid/pharmacology , Gap Junctions/drug effects , Histone Deacetylase Inhibitors , Hydrogen Peroxide/pharmacology , Hydroxamic Acids/pharmacology , Liver/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Animals , Carcinogens/pharmacology , Cell Communication/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Histamine Antagonists/pharmacology , Liver/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oxidants/pharmacology , Phosphorylation/drug effects , Rats
6.
Antioxid Redox Signal ; 7(11-12): 1767-72, 2005.
Article in English | MEDLINE | ID: mdl-16356138

ABSTRACT

Sodium butyrate (NaBu) has an inhibitory effect on histone deacetylases (HDACs). The mitogen-activated protein (MAP) kinases, such as extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 MAP, kinase are known to be modulated during NaBu-induced apoptosis. In the present study, we showed that low concentrations of NaBu could induce apoptosis synergistically with the inhibition of p38 MAP kinase as proven by using specific p38 MAP kinase inhibitor and dominant negative p38 transfection in a ras-transformed rat liver epithelial cell line (WB-ras). There were no changes in HDAC1, suggesting that NaBu might be able to kill transformed cells bypassing the HDAC inhibitory effect. We further demonstrated that inhibition of p38 MAP kinase potentiated apoptotic cascades, including cleavage of poly(ADP-ribose) polymerase, caspase-3, and decrease in Bcl-2/Bax ratio even at a lower concentration of NaBu. Thus, p38 MAP kinase played inhibitory roles in NaBu-induced apoptosis, and simultaneous modulation of MAP kinases in NaBu treatment could increase the efficiency of the chemotherapeutic effect of NaBu.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Liver/cytology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Mutat Res ; 579(1-2): 189-99, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16051281

ABSTRACT

Although 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has a variety of carcinogenic and noncarcinogenic effects in experimental animals, its role in human carcinogenicity remain controversial. A simian virus 40-immortalized cell line from normal human breast epithelial cells with stem cells and luminal characteristics (M13SV1) was used to study whether TCDD can induce AIG positive colony formation and cause increased cell numbers in a inverted U-shaped dose-response manner. TCDD activated Akt, ERK2, and increased the expression of CYP1A1, PAI-2, IL-lb mRNA, and ERK2 protein levels. TCDD was able to increased phosphorylation and expression of ERK2 in same dose-response manner as AIG positive colony formation. Thus, TCDD induced tumorigenicity in M13SV1, possibly through the phosphorylation of ERK2 and/or Akt. Further, cDNA microarray with 7448 sequence-verified clones was used to profile various gene expression patterns after treatment of TCDD. Three clear patterns could be delineated: genes that were dose-dependently up-regulated, genes expressed in either U-shape and/or inverted U-shape. The fact that these genes are intrinsically related to breast epithelial cell proliferation and survival clearly suggests that they may be involved in the TCDD-induced breast tumorigenesis.


Subject(s)
Breast/cytology , Polychlorinated Dibenzodioxins/toxicity , Stem Cells/cytology , Breast/drug effects , Breast/metabolism , Carcinogens/toxicity , Cell Line, Transformed , Cell Proliferation/drug effects , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/genetics , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Interleukin-1/genetics , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Plasminogen Activator Inhibitor 2/genetics , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/drug effects , Tumor Stem Cell Assay , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mutat Res ; 579(1-2): 47-57, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16046226

ABSTRACT

The two distinct members of the mitogen-activated protein (MAP) kinase family c-Jun N-terminal protein kinase (JNK) and p38 MAP kinase, play an important role in central nervous system (CNS) development and differentiation. However, their role and functions are not completely understood in CNS. To facilitate in vitro study, we have established an immortal stem cell line using SV40 from fetal rat embryonic day 17. In these cells, MAP kinase inhibitors (SP600125, SB202190, and PD98059) were treated for 1, 24, 48, and 72 h to examine the roles of protein kinases. Early inhibition of JNK did not alter phenotypic or morphological changes of immortalized cells, however overexpression of Bax and decrease of phosphorylated AKT was observed. The prolonged inhibition of JNK induced polyploidization of immortalized cells, and resulted in differentiation and inhibition of cell proliferation. Moreover, JNK and p38 MAP kinase but not ERK1/2 was activated, and p21, p53, and Bax were overexpressed by prolonged inhibition of JNK. These results indicate that JNK and p38 MAP kinase could play dual roles on cell survival and apoptosis. Furthermore, this established cell line could facilitate study of the role of JNK and p38 MAP kinase on CNS development or differentiation/apoptosis.


Subject(s)
Cell Differentiation/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Stem Cells/cytology , Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anthracenes/pharmacology , Apoptosis/physiology , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Telencephalon/cytology , Telencephalon/embryology , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Cancer Lett ; 225(2): 199-206, 2005 Jul 28.
Article in English | MEDLINE | ID: mdl-15978324

ABSTRACT

Histone deacetylase inhibitors such as TSA, SAHA, and NaBu etc. are prospective cancer therapeutics of growing interest. Here, we demonstrated that oncogenic ras-transformed rat liver epithelial (WB-ras) cells were specifically undergone apoptosis by 48 h treatment of NaBu. During this, inhibition of ras proteins, especially farnesylated form of ras, and down-regulation of ERK1/2 were observed, which suggest ras/raf/MEK/ERK down-regulation, while p38 MAP kinase was maintained up-regulated. In addition, up-regulation of pro-apoptotic proteins such as p53 and p21CIP1/WAF1, and down-regulation of cell cycle regulator/anti-apoptotic proteins such as cdk2, -4 and phosphorylated Akt were observed concurrently with an increase in apoptotic cell portion. A phosphatase inhibitor, sodium orthovanadate (SOV), efficiently blocked apoptosis and restored responsible proteins for each phenomenon including ERK1/2 while SB203580, a specific p38 MAP kinase inhibitor, showed minor effect on them. Thus, ras/ERK signaling pathway can be considered in chemotherapeutic strategies of NaBu regardless of its inhibitory action on histone deacetylase.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism , Animals , Butyrates/antagonists & inhibitors , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Imidazoles/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation/drug effects , Pyridines/pharmacology , Rats , Substrate Specificity
10.
J Vet Med Sci ; 67(3): 291-4, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15805733

ABSTRACT

Gap junctional intercellular communications (GJIC) contributes to neural function in development and differentiation of CNS. In this study, we have investigated the expression of GJIC during the differentiation of neuronal stem cells and 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced neuronal stem cell-derived cells from rat brain. During neuronal stem cell differentiation, expressions of Cx43 and 32 were increased for the duration of 72 hr, however the effect were decreased on the 7d. In the neuronal stem cell-derived cells, pretreatments with p38 MAP kinase inhibitor, SB203580, and MEK inhibitor, PD98059, could protect GJIC against TPA-induced inhibition of GJIC. Our data suggest that GJIC plays an important role during neuronal stem cell differentiation, and ERK1/2 and p38 MAP kinase signaling pathway may be closely related functionally to regulate gap junction in rat neuronal stem cell-derived cells.


Subject(s)
Cell Communication/drug effects , Cell Differentiation/physiology , Gap Junctions/physiology , Neurons/cytology , Rats/embryology , Stem Cells/physiology , Animals , Blotting, Western , Cell Communication/physiology , Connexin 43/metabolism , Connexins/metabolism , Flavonoids/pharmacology , Imidazoles/pharmacology , Pyridines/pharmacology , Tetradecanoylphorbol Acetate , Gap Junction beta-1 Protein
11.
Int J Cancer ; 102(3): 250-3, 2002 Nov 20.
Article in English | MEDLINE | ID: mdl-12397644

ABSTRACT

Although the pharmacology and clinical application of water extracts of Ganoderma lucidum have been extensively documented, little is known regarding its alcohol extract. In the present study, the anti-tumor effect of an alcohol extract of Ganoderma lucidum was investigated using MCF-7 cells. We found that the alcohol extract of Ganoderma lucidum inhibited cell proliferation in a dose- and time-dependent manner, which might be mediated through up-regulation of p21/Waf1 and down-regulation of cyclin D1. Furthermore, this compound can directly induce apoptosis in MCF-7 cells, which might be mediated through up-regulation of a pro-apoptotic Bax protein and not by the immune system. Our findings suggest that there are multiple mechanisms underlying the anti-tumor effects of Ganoderma lucidum.


Subject(s)
Apoptosis , Breast Neoplasms/drug therapy , Cell Cycle Proteins , DNA-Binding Proteins , Plant Extracts/therapeutic use , Proto-Oncogene Proteins , Reishi/metabolism , Alcohols/pharmacology , Blotting, Western , Caspase 7 , Caspases/metabolism , Cell Division , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Down-Regulation , E2F Transcription Factors , Genes, p53 , Humans , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Time Factors , Transcription Factors/metabolism , Tumor Cells, Cultured , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...