Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Food Chem Toxicol ; 84: 125-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26303273

ABSTRACT

We investigated apoptosis induced by the green tea component the epigallocatechin-3-gallate (EGCG) and the pathways underlying its activity in a colon cancer cell line. A complete understanding of the mechanism(s) and molecules targeted by green tea polyphenols could be useful in developing novel therapeutic approaches for cancer treatment. EGCG, which is the major polyphenol in green tea, has cytotoxic effects and induced cell death in HT-29 cell death. In this study, we evaluated the effect EGCG on mitogen-activated protein kinase (MAPK) and Akt pathways. EGCG treatment increased phospho-ERK1/2, -JNK1/2 and -p38α, -p38γ and -p38δ, as well as phospho-Akt levels. Using a combination of kinase inhibitors, we found that EGCG-induced cell death is partially blocked by inhibiting Akt, ERK1/2 or alternative p38MAPK activity. Our data suggest that these kinase pathways are involved in the anti-cancer effects of EGCG and indicate potential use of this compound as chemotherapeutic agent for colon cancer treatment.


Subject(s)
Anticarcinogenic Agents/metabolism , Antioxidants/metabolism , Apoptosis , Catechin/analogs & derivatives , Colonic Neoplasms/metabolism , MAP Kinase Signaling System , Proto-Oncogene Proteins c-akt/agonists , Apoptosis/drug effects , Catechin/metabolism , Cell Line, Tumor , Colonic Neoplasms/chemically induced , Colonic Neoplasms/enzymology , Colonic Neoplasms/prevention & control , Food Handling , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Signaling System/drug effects , Osmolar Concentration , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Tea/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/chemistry , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Biol Chem ; 290(7): 4383-97, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25548290

ABSTRACT

p38 MAPKs regulate migration and invasion. However, the mechanisms involved are only partially known. We had previously identified fibulin 3, which plays a role in migration, invasion, and tumorigenesis, as a gene regulated by p38α. We have characterized in detail how p38 MAPK regulates fibulin 3 expression and its role. We describe here for the first time that p38α, p38γ, and p38δ down-regulate fibulin 3 expression. p38α has a stronger effect, and it does so through hypermethylation of CpG sites in the regulatory sequences of the gene. This would be mediated by the DNA methylase, DNMT3A, which is down-regulated in cells lacking p38α, but once re-introduced represses Fibulin 3 expression. p38α through HuR stabilizes dnmt3a mRNA leading to an increase in DNMT3A protein levels. Moreover, by knocking-down fibulin 3, we have found that Fibulin 3 inhibits migration and invasion in MEFs by mechanisms involving p38α/ß inhibition. Hence, p38α pro-migratory/invasive effect might be, at least in part, mediated by fibulin 3 down-regulation in MEFs. In contrast, in HCT116 cells, Fibulin 3 promotes migration and invasion through a mechanism dependent on p38α and/or p38ß activation. Furthermore, Fibulin 3 promotes in vitro and in vivo tumor growth of HCT116 cells through a mechanism dependent on p38α, which surprisingly acts as a potent inducer of tumor growth. At the same time, p38α limits fibulin 3 expression, which might represent a negative feed-back loop.


Subject(s)
Cell Movement , Colonic Neoplasms/pathology , DNA Methylation , Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Mitogen-Activated Protein Kinase 14/physiology , Animals , Blotting, Western , Cell Adhesion , Cell Proliferation , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Down-Regulation , Embryo, Mammalian/cytology , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Humans , Male , Mice , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Wound Healing , Xenograft Model Antitumor Assays
3.
Biochim Biophys Acta ; 1771(2): 164-76, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251057

ABSTRACT

We have previously shown that lovastatin, an HMG-CoA reductase inhibitor, induces apoptosis in rat brain neuroblasts. c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) are implicated in regulation of neuronal apoptosis. In this work, we investigated the role of JNK and p38 MAPK in neuroblast apoptosis induced by lovastatin. We found that lovastatin induced the activation of JNK, but not p38 MAPK. It also induced c-Jun phosphorylation with a subsequent increase in activator protein-1 (AP-1) binding, AP-1-mediated gene expression and BimEL protein levels. The effects of lovastatin were prevented by mevalonate. Pre-treatment with iJNK-I (a selective JNK inhibitor) prevented the effect of lovastatin on both neuroblast apoptosis and the activation of the JNK cascade. Furthermore, we found that the activation of the JNK signalling pathway triggered by lovastatin is accompanied by caspase-3 activation which is also inhibited by iJNK-I pre-treatment. Finally, a specific inhibitor of p38 MAPK, SB203580, had no effect on lovastatin-induced neuroblast apoptosis. Taken together, our data suggest that the activation of the JNK/c-Jun/BimEL signalling pathway plays a crucial role in lovastatin-induced neuroblast apoptosis. Our findings may also contribute to elucidate the intracellular mechanisms involved in the central nervous system side effects associated with statin therapy.


Subject(s)
Brain/metabolism , Lovastatin/pharmacology , Neurons/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Brain/cytology , Brain/drug effects , Cells, Cultured , Enzyme Activation , MAP Kinase Kinase 4/metabolism , Membrane Proteins/metabolism , Neurons/drug effects , Phosphorylation , Proto-Oncogene Proteins/metabolism , Rats , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Biochem J ; 401(1): 175-83, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16952276

ABSTRACT

We have shown previously that lovastatin, a 3-hydroxy-3-methyl- glutaryl coenzyme A reductase inhibitor, induces apoptosis in spontaneously immortalized rat brain neuroblasts. In the present study, we analysed the intracellular signal transduction pathways by which lovastatin induces neuroblast apoptosis. We showed that lovastatin efficiently inhibited Ras activation, which was associated with a significant decrease in ERK1/2 (extracellular-signal-regulated kinase 1/2) phosphorylation. Lovastatin also decreased CREB phosphorylation and CREB-mediated gene expression. The effects of lovastatin on the Ras/ERK1/2/CREB pathway were time- and concentration-dependent and fully prevented by mevalonate. In addition, we showed that two MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] inhibitors, PD98059 and PD184352, were poor inducers of apoptosis in serum-treated neuroblasts. However, these inhibitors significantly increased apoptosis induced by lovastatin treatment. Furthermore, we showed that pharmacological inhibition of both MEK and phosphoinositide 3-kinase activities was able to induce neuroblast apoptosis with similar efficacy as lovastatin. Our results suggest that lovastatin triggers neuroblast apoptosis by regulating several signalling pathways, including the Ras/ERK1/2 pathway. These findings might also contribute to elucidate the intracellular mechanisms involved in the central nervous system side effects associated with statin therapy.


Subject(s)
Brain/enzymology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Lovastatin/pharmacology , Neurons/enzymology , Animals , Cells, Cultured , Fetus , Genes, Reporter , Luciferases/metabolism , Neurons/cytology , Neurons/drug effects , Rats , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
FEBS J ; 273(1): 1-13, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16367743

ABSTRACT

We have previously shown that lovastatin induces apoptosis in spontaneously immortalized rat brain neuroblasts. Focal adhesion proteins and protein kinase Cdelta (PKCdelta) have been implicated in the regulation of apoptosis. We found that lovastatin exposure induced focal adhesion kinase, Crk-associated substrate (p130(Cas)), PKCdelta cleavage and caspase-3 activation in a concentration-dependent manner. Lovastatin effects were fully prevented by mevalonate. The cleavage of p130(Cas) was almost completely inhibited by z-DEVD-fmk, a specific caspase-3 inhibitor, and z-VAD-fmk, a broad spectrum caspase inhibitor, indicating that cleavage is mediated by caspase-3. In contrast, the lovastatin-induced cleavage of PKCdelta was only blocked by z-VAD-fmk suggesting that PKCdelta cleavage is caspase-dependent but caspase-3-independent. Additionally, z-VAD-fmk partially prevented lovastatin-induced neuroblast apoptosis. The present data show that lovastatin may induce neuroblast apoptosis by both caspase-dependent and independent pathways. These findings may suggest that the caspase-dependent component leading to the neuroblast cell death is likely to involve the cleavage of focal adhesion proteins and PKCdelta, which may be partially responsible for some biochemical features of neuroblast apoptosis induced by lovastatin.


Subject(s)
Apoptosis/physiology , Brain/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Lovastatin/pharmacology , Protein Kinase C-delta/metabolism , Rats/metabolism , Animals , Brain/drug effects , Brain/enzymology , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Flow Cytometry , Mevalonic Acid/pharmacology , Time Factors
6.
J Neurochem ; 94(5): 1277-87, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16111478

ABSTRACT

We previously showed that lovastatin, an HMG-CoA reductase inhibitor, suppresses cell growth by inducing apoptosis in rat brain neuroblasts. Our aim was to study intracellular signalling induced by lovastatin in neuroblasts. Lovastatin significantly decreases the phosphoinositide 3-kinase (PI3-K) activity in a concentration-dependent manner. Expression of p85 subunit and its association with phosphotyrosine-containing proteins are unaffected by lovastatin. Lovastatin decreases protein kinase B (PKB)/Akt phosphorylation, and its downstream effectors, p70S6K and the eukaryotic initiation factor 4E (eIF4E) regulatory protein 1, 4E-BP1, in a concentration-dependent manner, and reduces p70S6K expression. Lovastatin effects are fully prevented with mevalonate. Only the highest dose of PI3-K inhibitors that significantly reduce PI3-K kinase activity induces apoptosis in neuroblasts but to a lower degree than lovastatin. In summary, this work shows that treatment of brain neuroblasts with lovastatin leads to an inhibition of the main pathway that controls cell growth and survival, PI3-K/PKB and the subsequent blockade of downstream proteins implicated in the regulation of protein synthesis. This work suggests that inactivation of the antiapoptotic PI3-K appears insufficient to induce the degree of neuroblasts apoptosis provoked by lovastatin, which must necessarily involve other intracellular pathways. These findings might contribute to elucidate the molecular mechanisms of some statins effects in the central nervous system.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Neurons/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Stem Cells/enzymology , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Transformed , Cell Survival/drug effects , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Morpholines/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology , Tyrosine/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...