Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 7: e2194, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27077810

ABSTRACT

Triple-negative breast cancers (TNBCs) are aggressive forms of breast carcinoma associated with a high rate of recidivism. In this paper, we report the production of mammospheres from three lines of TNBC cells and demonstrate that both parthenolide (PN) and its soluble analog dimethylaminoparthenolide (DMAPT) suppressed this production and induced cytotoxic effects in breast cancer stem-like cells, derived from dissociation of mammospheres. In particular, the drugs exerted a remarkable inhibitory effect on viability of stem-like cells. Such an effect was suppressed by N-acetylcysteine, suggesting a role of reactive oxygen species (ROS) generation in the cytotoxic effect. Instead z-VAD, a general inhibitor of caspase activity, was ineffective. Analysis of ROS generation, performed using fluorescent probes, showed that both the drugs stimulated in the first hours of treatment a very high production of hydrogen peroxide. This event was, at least in part, a consequence of activation of NADPH oxidases (NOXs), as it was reduced by apocynin and diphenylene iodinium, two inhibitors of NOXs. Moreover, both the drugs caused downregulation of Nrf2 (nuclear factor erythroid 2-related factor 2), which is a critical regulator of the intracellular antioxidant response. Prolonging the treatment with PN or DMAPT we observed between 12 and 24 h that the levels of both superoxide anion and hROS increased in concomitance with the downregulation of manganese superoxide dismutase and catalase. In addition, during this phase dissipation of mitochondrial membrane potential occurred together with necrosis of stem-like cells. Finally, our results suggested that the effect on ROS generation found in the first hours of treatment was, in part, responsible for the cytotoxic events observed in the successive phase. In conclusion, PN and DMAPT markedly inhibited viability of stem-like cells derived from three lines of TNBCs by inducing ROS generation, mitochondrial dysfunction and cell necrosis.


Subject(s)
Mitochondria/drug effects , Oxidative Stress/drug effects , Sesquiterpenes/toxicity , Acetophenones/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Oligopeptides/pharmacology , Onium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Cell Death Dis ; 4: e891, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24176849

ABSTRACT

Triple-negative breast cancers (TNBCs) are clinically aggressive forms associated with a poor prognosis. We evaluated the cytotoxic effect exerted on triple-negative MDA-MB231 breast cancer cells both by parthenolide and its soluble analogue dimethylamino parthenolide (DMAPT) and explored the underlying molecular mechanism. The drugs induced a dose- and time-dependent decrement in cell viability, which was not prevented by the caspase inhibitor z-VAD-fmk. In particular in the first hours of treatment (1-3 h), parthenolide and DMAPT strongly stimulated reactive oxygen species (ROS) generation. The drugs induced production of superoxide anion by activating NADPH oxidase. ROS generation caused depletion of thiol groups and glutathione, activation of c-Jun N-terminal kinase (JNK) and downregulation of nuclear factor kB (NF-kB). During this first phase, parthenolide and DMAPT also stimulated autophagic process, as suggested by the enhanced expression of beclin-1, the conversion of microtubule-associated protein light chain 3-I (LC3-I) to LC3-II and the increase in the number of cells positive to monodansylcadaverine. Finally, the drugs increased RIP-1 expression. This effect was accompanied by a decrement of pro-caspase 8, while its cleaved form was not detected and the expression of c-FLIPS markedly increased. Prolonging the treatment (5-20 h) ROS generation favoured dissipation of mitochondrial membrane potential and the appearance of necrotic events, as suggested by the increased number of cells positive to propidium iodide staining. The administration of DMAPT in nude mice bearing xenografts of MDA-MB231 cells resulted in a significant inhibition of tumour growth, an increment of animal survival and a marked reduction of the lung area invaded by metastasis. Immunohistochemistry data revealed that treatment with DMAPT reduced the levels of NF-kB, metalloproteinase-2 and -9 and vascular endothelial growth factor, while induced upregulation of phosphorylated JNK. Taken together, our data suggest a possible use of parthenolide for the treatment of TNBCs.


Subject(s)
Autophagy/drug effects , Reactive Oxygen Species/metabolism , Sesquiterpenes/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Fas-Associated Death Domain Protein/metabolism , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Sesquiterpenes/therapeutic use , Xenograft Model Antitumor Assays
3.
Int J Oncol ; 43(6): 1895-900, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24126683

ABSTRACT

The sesquiterpene lactone parthenolide (PN) has recently attracted considerable attention because of its anti-microbial, anti-inflammatory and anticancer effects. However, the mechanism of its cytotoxic action on tumor cells remains scarcely defined. We recently provided evidence that the effect exerted by PN in MDA-MB-231 breast cancer cells was mediated by the production of reactive oxygen species (ROS). The present study shows that PN promoted the phosphorylation of EGF receptor (phospho-EGFR) at Tyr1173, an event which was observed already at 1 h of incubation with 25 µM PN and reached a peak at 8-16 h. This effect seemed to be a consequence of ROS production, because N-acetylcysteine (NAC), a powerful ROS scavenger, prevented the increment of phospho-EGFR levels. In addition fluorescence analyses performed using dihydroethidium demonstrated that PN stimulated the production of superoxide anion already at 2-3 h of incubation and the effect further increased prolonging the time of treatment, reaching a peak at 8-16 h. Superoxide anion production was markedly hampered by apocynin, a well known NADPH oxidase (NOX) inhibitor, suggesting that the effect was dependent on NOX activity. The finding that AG1478, an EGFR kinase inhibitor, substantially blocked both EGFR phosphorylation and superoxide anion production strongly suggested that phosphorylation of EGFR can be responsible for the activation of NOX with the consequent production of superoxide anion. Therefore, EGFR phosphorylation can exert a key role in the production of superoxide anion and ROS induced by PN in MDA-MB-231 cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Sesquiterpenes/pharmacology , Superoxides/metabolism , Acetophenones/pharmacology , Acetylcysteine/metabolism , Antioxidants/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Humans , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , NF-kappa B/antagonists & inhibitors , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Quinazolines/pharmacology , Tyrphostins/pharmacology
4.
Biochimie ; 94(2): 287-99, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21835222

ABSTRACT

SAHA, an inhibitor of histone deacetylase activity, has been shown to sensitize tumor cells to apoptosis induced by TRAIL, a member of TNF-family. In this paper we investigated the effect of SAHA/TRAIL combination in two breast cancer cell lines, the ERα-positive MCF-7 and the ERα-negative MDA-MB231. Treatment of MDA-MB231 and MCF-7 cells with SAHA in combination with TRAIL caused detachment of cells followed by anoikis, a form of apoptosis which occurs after cell detachment, while treatment with SAHA or TRAIL alone did not produce these effects. The effects were more evident in MDA-MB231 cells, which were chosen for ascertaining the mechanism of SAHA/TRAIL action. Our results show that SAHA decreased the level of c-FLIP, thus favouring the interaction of TRAIL with the specific death receptors DR4 and DR5 and the consequent activation of caspase-8. These effects increased when the cells were treated with SAHA/TRAIL combination. Because z-IEDT-fmk, an inhibitor of caspase-8, prevented both the cleavage of the focal adhesion-kinase FAK and cell detachment, we suggest that activation of caspase-8 can be responsible for both the decrement of FAK and the consequent cell detachment. In addition, treatment with SAHA/TRAIL combination caused dissipation of ΔΨ(m), activation of caspase-3 and decrement of both phospho-EGFR and phospho-ERK1/2, a kinase which is involved in the phosphorylation of BimEL. Therefore, co-treatment also induced decrement of phospho-BimEL and a concomitant increase in the dephosphorylated form of BimEL, which plays an important role in the induction of anoikis. Our findings suggest the potential application of SAHA in combination with TRAIL in clinical trials for breast cancer.


Subject(s)
Anoikis/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hydroxamic Acids/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Anoikis/genetics , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspases/genetics , Caspases/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Vorinostat
5.
Mol Pharmacol ; 77(5): 854-63, 2010 May.
Article in English | MEDLINE | ID: mdl-20159939

ABSTRACT

In this article, we demonstrate that the synthetic cannabinoid R-(+)-(2,3-dihydro-5-methyl-3-[(4-morpholinyl)methyl]pyrol[1,2,3-de]-1,4-benzoxazin-6-yl)-(1-naphthalenyl) methanone mesylate (WIN 55,212-2) sensitizes human hepatocellular carcinoma (HCC) cells to apoptosis mediated by tumor necrosis-related apoptosis inducing ligand (TRAIL). The apoptotic mechanism induced by treatment with WIN/TRAIL combination involved the loss of the mitochondrial transmembrane potential and led to the activation of caspases. In HCC cells, WIN treatment induced the up-regulation of TRAIL death receptor DR5, an effect that seemed to be related to the increase in the level of p8 and CHOP, two factors implicated in cellular stress response and apoptosis. This relationship was suggested by the observation that the down-regulation of p8 or CHOP by specific small interfering RNAs attenuated both WIN-mediated DR5 up-regulation and the cytotoxicity induced by WIN/TRAIL cotreatment. Moreover, WIN induced a significant decrease in the levels of some survival factors (survivin, c-inhibitor of apoptosis protein 2, and Bcl-2) and in particular in that of the active phosphorylated form of AKT. This event seemed to be dependent on the transcription factor peroxisome proliferator-activated receptor-gamma whose level significantly increased after WIN treatment. Therefore, both the induction of DR5 via p8 and CHOP and the down-regulation of survival factors seem to be crucial for the marked synergistic effects induced by the two drugs in HCC cells. Taken together, the results reported in this article indicate that WIN/TRAIL combination could represent a novel important tool for the treatment of HCC.


Subject(s)
Apoptosis/physiology , Benzoxazines/pharmacology , Cannabinoids/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transcription Factor CHOP/physiology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/physiology , DNA Primers , DNA, Complementary/drug effects , DNA, Complementary/genetics , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , Flow Cytometry , Gene Amplification , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/drug effects
6.
Int J Oncol ; 32(1): 177-84, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18097557

ABSTRACT

This report shows that histone deacetylase inhibitors (HDACIs) induced apoptosis in human hepatoma HepG2 cells in a dose- and time-dependent manner. Trichostatin A (TSA), ITF2357 and suberoylanilide hydroxamic acid (SAHA), which were very effective agents, caused apoptotic effects after a lag phase of 12-16 h. In order to elucidate the mechanism of HDACIs action in HepG2 cells we have studied the effects of TSA, ITF2357 and SAHA on acetylation of p53 and histones H2A, H2B, H3 and H4. It was observed that HDACIs rapidly induced acetylation of these proteins, being the effects clearly visible already at 30 min of treatment at the same doses which caused apoptosis. Analysis of the immunocomplexes, obtained from nuclear extracts using an antibody against p53, revealed the presence of acetylated p53 together with acetylated forms of histones and histone acetyltransferases p300 and PCAF. Experiments performed using pifithrin-alpha, a reversible inhibitor of p53, showed a correlation between acetylation of p53 and induction of apoptosis. In addition treatment with siRNA against p53 indicated that p53 is involved in the acetylation of histones. In conclusion, this report suggests that complexes constituted by acetylated p53, acetylated histones and coactivators can play a central role in HDACI-induced apoptosis in HepG2 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/metabolism , Liver Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Acetylation , Benzothiazoles/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA Damage , Humans , Hydroxamic Acids/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , RNA, Small Interfering/pharmacology , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Vorinostat
7.
Apoptosis ; 12(7): 1327-38, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17351739

ABSTRACT

Histone deacetylase (HDAC) inhibitors represent a promising group of anticancer agents. This paper shows that the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) stimulated at 5-10 microM apoptosis in human hepatoma HepG2 and Huh6 cells, but was ineffective in primary human hepatocytes (PHH). In HepG2 cells SAHA induced the extrinsic apoptotic pathway, increasing the expression of both FasL and FasL receptor and causing the activation of caspase-8. Moreover, SAHA enhanced the level of Bim proteins, stimulated alternative splicing of the Bcl-X transcript with the expression of the proapoptotic Bcl-Xs isoform, induced degradation of Bid into the apoptotic factor t-Bid and dephosphorylation and inactivation of the anti-apoptotic factor Akt. Consequently, SAHA caused loss of mitochondrial transmembrane potential, release of cytochrome c from mitochondria, activation of caspase-3 and degradation of PARP. Interestingly, a combination of suboptimal doses of SAHA (1 microM) and bortezomib (5-10 nM), a potent inhibitor of 26S proteasome, synergistically induced apoptosis in both HepG2 and Huh6 cells, but was ineffective in PHH. Combined treatment increased with synergistic effects the expression levels of c-Jun, phospho-c-Jun and FasL and the production of Bcl-Xs. These effects were accompanied by activation of Bid, caspase-8 and 3. In conclusion, SAHA stimulated apoptosis in hepatoma cells and exerted a synergistic apoptotic effect when combined with bortezomib. In contrast, these treatments were quite ineffective in inducing apoptosis in PHH. Thus, our results suggest the potential application of the SAHA/bortezomib combination in clinical trials for liver cancer.


Subject(s)
Apoptosis/drug effects , Boronic Acids/metabolism , Carcinoma, Hepatocellular/metabolism , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Pyrazines/metabolism , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Hepatocellular/pathology , Caspase 8/metabolism , Cell Line, Tumor/cytology , Cell Line, Tumor/metabolism , Fas Ligand Protein/drug effects , Fas Ligand Protein/metabolism , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Vorinostat , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
8.
Apoptosis ; 11(4): 607-25, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16528474

ABSTRACT

The proteasome inhibitor bortezomib is an efficacious apoptotic agent in many tumor cells. This paper shows that bortezomib induced apoptosis in human hepatoma HepG2 cells associated with many modifications in the expression of survival or death factors. Although bortezomib increased the level of the protective factors HSP70 and HSP27, the effects of the drug that favour cell death were predominant. These events include accumulation of c-Jun, phospho-c-Jun and p53; increase in FasL level with activation of caspase-8; changes related to members of Bcl-2 family with increase in the level of pro-apoptotic members and decrease in that of anti-apoptotic ones; dissipation of mitochondrial potential with cytochrome c release and activation of caspase-3. In contrast, Chang liver cells exhibited a very low susceptibility to bortezomib-induced apoptosis, which was accompanied by modest modifications in the expression of apoptotic factors. In HepG2 cells bortezomib markedly increased AP-1 activity and the expression of its transcriptional targets such as c-Jun, FasL, BimEL, which are involved in apoptosis. Moreover, AP-1 induced its own production by increasing c-Jun content in the composition of the same AP-1 complex. In addition, bortezomib caused activation of JNK1, which in turn increased the level of phospho-c-Jun as well as stimulated the activation of caspase-3 and t-Bid, two fundamental apoptotic factors. Interestingly, siRNA silencing of c-Jun or JNK1 reduced HepG2 cell susceptibility to apoptosis and prevented the increase in AP-1 activity. Both JNK-1 and AP-1 thus exerted a crucial role in bortezomib-induced apoptosis. Differently, in Chang liver cells the different composition of AP-1 complex as well as the failure of JNK activation seemed to be responsible for the low susceptibility to apoptosis. Given the high susceptibility of hepatoma cells to bortezomib, our results suggest the potential application of this compound in clinical trials for liver cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Boronic Acids/pharmacology , Liver Neoplasms/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Transcription Factor AP-1/metabolism , Bortezomib , Caspase 8 , Caspases/metabolism , Cell Line , Cell Line, Tumor , Fas Ligand Protein , Heat-Shock Proteins/metabolism , Humans , Liver/drug effects , Liver/metabolism , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Tumor Necrosis Factors/metabolism
10.
FEBS Lett ; 579(3): 615-20, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15670817

ABSTRACT

This study describes the molecular mechanism by which treatment with 3-AB, a potent inhibitor of PARP, allows human osteosarcoma MG-63 cells to restrict growth and enter differentiation. Our findings show that in MG-63 cells, aberrant gene expression keeps Rb protein constitutively inactivated through hyperphosphorylation and this promotes uncontrolled proliferation of the cells. After 3-AB-treatment, the poly(ADP-ribosyl)ation of nuclear proteins markedly decreases and this results in an increase in both the hypophosphorylated active form of Rb and pRb/E2F complexes. These effects are accompanied by G1 arrest, downregulation of gene products required for proliferation (cyclin D1, beta-catenin, c-Jun, c-Myc and Id2) and upregulation of those implicated in the osteoblastic differentiation (p21/Waf1, osteopontin, osteocalcin, type I collagen, N-cadherins and alkaline phosphatase). Our study suggests that use of PARP inhibitors may induce a remodeling of chromatin with the reprogramming of gene expression and the activation of differentiation.


Subject(s)
Benzamides/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Osteosarcoma/pathology , Poly(ADP-ribose) Polymerase Inhibitors , Adenosine Diphosphate Ribose/metabolism , Base Sequence , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Primers , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Flow Cytometry , G1 Phase/drug effects , Gene Expression Regulation , Humans , Immunoprecipitation , Osteosarcoma/enzymology , Phosphorylation , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
11.
Eur J Cancer ; 40(9): 1441-52, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15177505

ABSTRACT

Butyrate can promote programmed cell death in a number of tumour cells in vitro. This paper provides evidence that butyrate induces apoptosis in human hepatoma HuH-6 and HepG2 cells but is ineffective in Chang liver cells, an immortalised non-tumour cell line. In both HuH-6 and HepG2 cells, apoptosis appeared after a lag period of approximately 16 h and increased rapidly during the second day of treatment. In particular, the effect was stronger in HuH-6 cells, which were, therefore, chosen for ascertaining the mechanism of butyrate action. In HuH-6 cells, beta-catenin seemed to exert an important protective role against apoptosis, since pretreatment with beta-catenin antisense ODN reduced the content of beta-catenin and anticipated the onset of apoptosis at 8 h of exposure to butyrate. Moreover, in HuH-6 cells, butyrate induced loss of mitochondrial membrane potential, release of cytochrome c from mitochondria, activation of caspase 9 and caspase 3, and degradation of poly(ADP-ribose) polymerase. In addition, during the second day of treatment, beta-catenin, pRb, and cyclins D and E were diminished and the phosphorylated form of pRb disappeared. Also, the content of the anti-apoptotic factor Bcl-XL fell markedly during this period, while that of the pro-apoptotic factor Bcl-Xs increased. These effects were accompanied by an increase in both Bcl-XL and Bcl-Xs mRNA transcripts, as ascertained by reverse transcriptase-polymerase chain reaction. Our results suggest that caspases have a crucial role in butyrate-induced apoptosis. This conclusion is supported by the observation that the inhibitors of caspases, benzyloxy carbonyl-Val-Ala-Asp-fluoromethylketone and benzyloxy carbonyl-Asp-Glu-Val-Asp-fluoromethylketone, prevented apoptosis and the decrease in Bcl-XL, pRb, cyclins and beta-catenin. These effects were most probably responsible for the increased sensitivity of the cells to butyrate-induced apoptosis, which was observed on the second day of treatment.


Subject(s)
Apoptosis , Butyrates/pharmacology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Apoptosis/drug effects , Blotting, Western/methods , Caspases/metabolism , Cell Line/drug effects , Cyclin D , Cyclin E/metabolism , Cyclins/metabolism , Cytoskeletal Proteins/metabolism , Humans , Membrane Potentials/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/metabolism , bcl-X Protein , beta Catenin
13.
Int J Oncol ; 21(4): 857-65, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239627

ABSTRACT

This report is focused on the apoptotic effect induced by MG132, an inhibitor of 26S proteasome, in human hepatoma HepG2 cells. The results were compared with those obtained with non-transformed human Chang liver cells. MG132 reduced the viability of HepG2 cells in a time- and dose-dependent manner. The effect was in tight connection with the induction of apoptosis, as indicated by fluorescence microscopy and cytometric analysis, and was accompanied by a remarkable increase in the production of H2O2 and a reduction in mitochondrial transmembrane potential (Deltapsim). In addition cell death was prevented by antioxidants such as GSH, N-acetylcysteine or catalase. Western blot analysis showed that HepG2 cells contain a very low level of Bcl-2 and a much higher level of Bcl-XL, another antiapoptotic factor of the same family. When the cells were exposed to MG132 the level of Bcl-XL diminished, while a new band, corresponding to the expression of the proapoptotic protein Bcl-XS was detected. MG132 also caused the release of cytochrome c from mitochondria and the activation of caspase-3 with the consequent degradation of poly-ADP ribose polymerase (PARP). The observation that the broad spectrum caspase inhibitor z-VAD markedly reduced the apoptotic effect of the drug clearly demonstrated that caspases play an important role in MG132-induced apoptosis. MG132 exerted a modest effect on the viability of Chang liver cells which primarily depended on the G2/M arrest of cell cycle while only a small percentage of apoptotic cells was found. The remarkable differences in the effects induced by MG132 in Chang liver cells and HepG2 cells made us hypothesise the potential use of proteasome inhibitors in hepatocarcinoma therapy.


Subject(s)
Apoptosis , Caspases/metabolism , Cysteine Endopeptidases/pharmacology , Hepatoblastoma/pathology , Leupeptins/pharmacology , Liver Neoplasms/pathology , Multienzyme Complexes/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Antineoplastic Agents/pharmacology , Blotting, Western , Caspase 3 , Cell Cycle , Cell Survival , Cytosol/metabolism , Enzyme Activation , Flow Cytometry , G2 Phase/drug effects , Humans , Hydrogen Peroxide/metabolism , Liver Neoplasms/metabolism , Membrane Potentials , Mitochondria/metabolism , Mitosis/drug effects , Oxidative Stress , Proteasome Endopeptidase Complex , Time Factors , Tumor Cells, Cultured , bcl-X Protein
14.
FEBS Lett ; 499(1-2): 191-7, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11418138

ABSTRACT

This paper studies the cytotoxic effect induced by the topoisomerase I inhibitor camptothecin in human osteosarcoma Saos-2 cells, which lack p53 and contain a non-functional form of the product of the retinoblastoma gene, pRb. Cytotoxicity induced by camptothecin was dose- and time-dependent; the treatment with 100 nM camptothecin reduced cell viability by 50% at 32 h and by 75% at 72 h of exposure. The cytotoxic effect was caused by apoptosis, as ascertained by morphological evidence, acridine orange-ethidium bromide staining and flow cytometric analysis. Apoptosis was accompanied by both the activation of caspase-3 and the fragmentation of poly(ADP-ribose) polymerase. Treatment with camptothecin caused a threefold increase in the activity of c-Jun N-terminal kinase (JNK) and an eightfold increase in the level of phosphorylated c-Jun. The introduction of the RB gene into Saos-2 cells reduced the rate of cell growth. Moreover, stable clones of transfected cells were resistant to camptothecin. Exposure to 100 nM camptothecin for 72 h reduced the viability of transfected cells by only 10%; moreover, very modest effects were observed on the activity of JNK as well as on the level of phosphorylated c-Jun. The results reported in this paper support the conclusion that the expression of wild-type pRb in Saos-2 cells exerts an anti-apoptotic influence through the control of JNK activity.


Subject(s)
Apoptosis/drug effects , Camptothecin/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Blotting, Western , Camptothecin/toxicity , Caspase 3 , Caspases/metabolism , Cell Cycle/drug effects , Cell Size/drug effects , Cell Survival/drug effects , DNA Topoisomerases, Type I/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Flow Cytometry , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Retinoblastoma Protein/genetics , Time Factors , Topoisomerase I Inhibitors , Transfection , Tumor Cells, Cultured
15.
Int J Oncol ; 18(6): 1233-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11351256

ABSTRACT

Our results demonstrate that sodium phenylbutyrate, a compound with a low degree of toxicity, exerted a cytotoxic effect on human retinoblastoma Y79 cells in a time- and dose-dependent manner. Treatment of Y79 cells for 72 h with phenylbutyrate reduced cell viability by 63% at 2 mM and 90% at 4 mM. Cell death caused by phenylbutyrate exhibited the typical features of apoptosis, as shown by light and fluorescent microscopy. Western blot analysis demonstrated that exposure of Y79 cells to phenylbutyrate decreased the level of the antiapoptotic factor Bcl-2 and induced the activation of caspase-3, a key enzyme in the execution phase of apoptosis. Moreover, treatment with phenylbutyrate markedly increased the level of acetylated histone-H3. Combined treatment with phenylbutyrate and topotecan, a topoisomerase I-inhibitor, resulted in a clear synergistic effect. We suggest that the effects exerted by phenylbutyrate on Y79 cells essentially depend on modifications of gene expression consequent to histone hyperacetylation.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Phenylbutyrates/pharmacology , Retinoblastoma/drug therapy , Topoisomerase I Inhibitors , Topotecan/pharmacology , Tumor Cells, Cultured/drug effects , Acetylation , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Drug Synergism , Drug Therapy, Combination , Enzyme Activation , Histones/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoblastoma/enzymology , Retinoblastoma/pathology , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology , Tumor Suppressor Protein p53/metabolism
16.
Tumour Biol ; 21(6): 337-48, 2000.
Article in English | MEDLINE | ID: mdl-11006574

ABSTRACT

This paper studies the effects caused in human retinoblastoma Y79 cells by treatment with combinations of sodium butyrate, the inhibitor of topoisomerase I camptothecin and the inhibitor of 26S proteasome MG132. The combination of sodium butyrate and camptothecin resulted in a strong synergistic cytotoxicity, as revealed by combination indices of 0.77 and 0.52 calculated at IC(50) and IC(75). Synergistic interactions were also demonstrated for combinations of sodium butyrate and MG132, camptothecin and MG132 and for a combination of all three compounds. The cytotoxic effects observed after the combined treatments can be considered a consequence of apoptosis, as suggested by the appearance of morphological signals of apoptosis and by the activation of caspase-3 with degradation of poly-ADP ribose polymerase and lamin B. Treatment of Y79 cells with sodium butyrate alone lowered the levels of p53, E2F-1 and Bcl-2. The addition of MG132 to sodium butyrate counteracted the effect on p53 only, while the addition of camptothecin to sodium butyrate counteracted the effect on both p53 and E2F-1. The treatment of Y79 cells with the triple combination increased the level of p53, decreased that of Bcl-2, while the level of E2F-1 was not modified. We suggest that the effects exerted on the levels of these regulatory proteins can explain the synergistic interactions demonstrated between sodium butyrate, camptothecin and MG132.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Retinoblastoma/pathology , Tumor Cells, Cultured/drug effects , Antineoplastic Combined Chemotherapy Protocols/toxicity , Apoptosis/drug effects , Butyrates/administration & dosage , Butyrates/pharmacology , Butyrates/toxicity , Camptothecin/administration & dosage , Camptothecin/pharmacology , Camptothecin/toxicity , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/administration & dosage , Cysteine Proteinase Inhibitors/pharmacology , Cysteine Proteinase Inhibitors/toxicity , Drug Synergism , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Humans , Leupeptins/administration & dosage , Leupeptins/pharmacology , Leupeptins/toxicity , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoblastoma/drug therapy , Retinoblastoma/metabolism , Topoisomerase I Inhibitors , Tumor Suppressor Protein p53/biosynthesis
17.
Exp Eye Res ; 70(4): 503-17, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10865999

ABSTRACT

Arachidonic acid administration caused apoptosis in Y79 cells, as shown by typical morphological changes, phosphatidylserine externalization, chromatin condensation, processing and activation of caspase-3 and cleavage of the endogenous caspase substrate poly-(ADP-ribose)-polymerase. Arachidonic acid also caused lamin B cleavage, suggesting caspase-6 activation. Arachidonic acid treatment was accompanied by increased formation of the lipid peroxidation end products malondialdehyde and 4-hydroxy-2-nonenal, lowering in reduced glutathione content and in mitochondrial membrane potential. Inhibiting glutathione synthesis sensitized Y79 cells to apoptosis-inducing stimuli, whilst replenishing reduced glutathione attenuated arachidonic acid toxicity. Similar findings were obtained using hydroperoxyeicosatetranoic acids (oxygenated metabolites of arachidonic acid which deplete the reduced glutathione pool) and nordihydroguaretic acid, a general inhibitor of lipooxygenase pathway. which may also trigger rapid depletion of reduced glutathione. Melittin, which is known to activate phospholipase A2, also potently induced apoptosis. Arachidonic acid toxicity was inversely related to cell density. This could depend on an increased production of molecules with antiapoptotic effect; insulin-like growth factors could most likely be one of these molecules. These results propose a role for oxidative stress in the cytotoxicity induced by arachidonic acid in Y79 cells and suggest that these cells could be protected from such toxicity as long as sufficient levels of reduced glutathione and survival factors are present.


Subject(s)
Apoptosis/drug effects , Arachidonic Acid/therapeutic use , Oxidative Stress/drug effects , Retinoblastoma/drug therapy , Blotting, Western , Caspases/drug effects , Cell Count , Cell Survival , Colorimetry , Dose-Response Relationship, Drug , Flow Cytometry , Glutathione/analysis , Humans , Membrane Potentials , Mitochondria/physiology , Poly(ADP-ribose) Polymerases/drug effects , Retinoblastoma/physiopathology , Trypan Blue , Tumor Cells, Cultured
18.
Cancer Res ; 59(21): 5586-95, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10554039

ABSTRACT

This study deals with the apoptotic effect exerted on human retinoblastoma Y79 cells by both sodium butyrate and an inhibitor of 26S proteasome [z-Leu-Leu-Leu-CHO (MG132)] and their synergistic effect. Exposure to sodium butyrate (1-4 mM) induced an accumulation of cells in the G2-M phase that was already visible after 24 h of treatment, when morphological and biochemical signs of apoptosis appeared only in a small number of cells (5-10%). Thereafter, the apoptotic effects increased progressively with slow kinetics, reaching a maximum after 72 h of exposure, when they concerned a large fraction of cells (>75% with 4 mM sodium butyrate). Sodium butyrate stimulated the conversion of procaspase-3 into caspase-3 and also induced the cleavage of poly-(ADP-ribose) polymerase and lamin B, two hallmarks of apoptosis. All of the apoptotic signals were suppressed by benzyloxy carbonyl-Val-Ala-Asp-fluoromethylketone (a general inhibitor of caspase activities), whereas acetyl-Asp-Glu-Val-Asp aldehyde, a specific inhibitor of caspase-3 activity, only induced a partial reversion of the apoptotic effects. Sodium butyrate also decreased the Bcl-2 level, whereas it increased the Bax level and stimulated the release of cytochrome c from the mitochondria, an event that was most likely responsible for the activation of caspase-3. Finally, sodium butyrate activated 26S proteasome, the major extralysosomal degradative machinery, which is responsible for the degradation of short-lived proteins. Consequently, the levels of p53, N-myc, and IkappaBalpha (factors that play regulatory roles in apoptosis) diminished, whereas the nuclear level of nuclear factor kappaB concomitantly increased. Treatment of Y79 cells with MG132 induced apoptosis with more rapid kinetics than with sodium butyrate. The effects appeared after 8 h of incubation, reaching a maximum at 24 h, and they were accompanied by increased levels of N-myc, p53, and IkappaBalpha. MG132 also favored the release of cytochrome c from the mitochondria and increased the activity of caspase-3. When Y79 cells were exposed to combinations of sodium butyrate and MG132, the latter compound suppressed the decreasing effect induced by sodium butyrate on the levels of p53, N-myc, and IkappaBalpha and the increasing effect on the nuclear level of nuclear factor kappaB. Moreover, an increase in the level of Bax and an enhancement in the release of cytochrome c from the mitochondria were observed. Clear synergistic effects concerning the activation of both caspase-3 and apoptosis were induced by a combination of suboptimal doses of sodium butyrate and MG132. The results support the conclusion that MG132 potentiates the apoptotic effect of sodium butyrate by suppressing its stimulatory effect on 26S proteasome activity. Synergistic interactions between butyrate and inhibitors of proteasome could represent a new important tool in tumor therapy and, in particular, the treatment of retinoblastoma.


Subject(s)
Apoptosis , Butyrates/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Proteasome Endopeptidase Complex , Retinoblastoma/pathology , Caspase 3 , Caspases/metabolism , Cell Cycle/drug effects , Cytochrome c Group/metabolism , Flow Cytometry , Humans , I-kappa B Proteins/metabolism , Kinetics , Lamin Type B , Lamins , Mitochondria/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Peptide Hydrolases/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
19.
Mol Cell Biochem ; 185(1-2): 7-15, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9746206

ABSTRACT

C2-ceramide, a cell-permeable analogue of ceramide, induced significant, dose- and time-dependent death in human retinoblastoma Y79 cells. Dying cells strongly displayed the morphology of apoptosis as characterized by microscopic evidence of cell shrinkage, membrane blebbing, nuclear and chromatin condensation and degeneration of the nucleus into membrane-bound apoptotic bodies. Upon induction of apoptosis Y79 cells evidence early phosphatidylserine externalization, as shown by annexin V-FITC. Apoptosis was also assessed by monitoring changes in cell granularity by staining with the combined fluorescent dyes acridine orange and ethidium bromide. C2-ceramide induced these morphological changes without a concomitant production of oligonucleosomal fragments responsible for the DNA ladder and without changes in p53 protein level. Apoptosis was accompanied by accumulation of a modified Bcl-2 protein with a slower-mobility form, and by proteolytic cleavage of PARP. The effect seemed to be specific for C2-ceramide, as C2-dihydroceramide, or other amphiphilic lipid analogues, or products of ceramide hydrolysis were ineffective. The effect also depended on mRNA and protein synthesis as it was markedly inhibited by actinomycin D and cycloheximide. Sphingomyelinase and interleukin-1beta, which are known to activate the sphingomyelin turnover leading to ceramide generation, also induced apoptosis mimicking the effects of ceramide. These findings propose ceramide as an activator of the suicidal program in Y79 cells.


Subject(s)
Apoptosis/drug effects , Retinoblastoma/metabolism , Sphingosine/analogs & derivatives , Apoptosis/physiology , Bacterial Proteins/pharmacology , Blotting, Western , Cell Survival/drug effects , Cell Survival/physiology , Ceramides/physiology , DNA Fragmentation/drug effects , DNA Fragmentation/physiology , Humans , Interleukin-1/pharmacology , Nucleosomes/chemistry , Nucleosomes/drug effects , Nucleosomes/physiology , Okadaic Acid/pharmacology , Proteins/analysis , Proteins/drug effects , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/drug effects , Retinoblastoma/pathology , Sphingomyelin Phosphodiesterase/pharmacology , Sphingosine/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/drug effects
20.
Tumour Biol ; 19(5): 356-63, 1998.
Article in English | MEDLINE | ID: mdl-9701726

ABSTRACT

This paper deals with the apoptotic effect exerted in human retinoblastoma Y79 cells by a number of compounds. A remarkable effect was observed after treatment with DNA-damaging agents, such as camptothecin, etoposide, cisplatin and carboplatin; camptothecin was found to be the most efficacious. Treatment with these compounds induced the appearance of morphological features of apoptosis in the cells together with the distinct fragmentation of DNA, as shown by agarose gel electrophoresis. These effects were also accompanied by a remarkable increase in the level of p53. Many other compounds, which are not DNA-damaging agents, induced the morphological features of apoptosis but none of them were capable of increasing the level of p53. Among these compounds, Taxol, suramin and sodium butyrate also stimulated the oligonucleosomal fragmentation of DNA, while C2-ceramide, a cell-permeable analogue of ceramide, and vitamin D3 were not effective in the induction of DNA laddering in Y79 cells. Apoptosis was dependent on macromolecular synthesis with all the compounds tested.


Subject(s)
Apoptosis/drug effects , Retinoblastoma/drug therapy , Retinoblastoma/pathology , Amsacrine/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Butyrates/pharmacology , Butyric Acid , Camptothecin/pharmacology , Carboplatin/pharmacology , Cisplatin/pharmacology , DNA Damage , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Paclitaxel/pharmacology , Suramin/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...