Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 13(11): 1960-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16557274

ABSTRACT

The present study was undertaken to determine the significance of histone acetylation versus DNA damage in drug-induced irreversible growth arrest (senescence) and apoptosis. Cellular treatment with the DNA-damaging drugs doxorubicin and cisplatin or with the histone deacetylase inhibitor trichostatin A, led to the finding that all the three drugs induced senescence at concentrations significantly lower than those required for apoptosis. However, only doxorubicin and cisplatin induced activation of H2AX, a marker for double-strand break formation. Interestingly, this occurred mainly at apoptosis and not senescence-inducing drug concentrations, suggesting that non-DNA-damage pathways may be implicated in induction of senescence by these drugs. In agreement with this, chromatin immunoprecipitation experiments indicated that doxorubicin was able to induce acetylation of histone H3 at the promoter of p21/WAF1 only at senescence-inducing concentrations. Collectively, these findings suggest that alteration of chromatin structure by cytotoxic drugs may represent a key mediator of senescence.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Cisplatin/pharmacology , DNA Damage/physiology , Doxorubicin/pharmacology , Histones/metabolism , Hydroxamic Acids/pharmacology , Acetylation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA/metabolism , Enzyme Activation/drug effects , Humans , Kinetics , Reproducibility of Results , Time Factors
2.
Apoptosis ; 11(3): 377-88, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16520893

ABSTRACT

Rapid CD4+ lymphocyte depletion due to cell death caused by HIV infection is one of the hallmarks of acquired immunodeficiency syndrome. HIV-1 viral protein R (Vpr) induces apoptosis and is believed to contribute to CD4+ lymphocyte depletion. Thus, identification of cellular factors that potentially counteract this detrimental viral effect will not only help us to understand the molecular action of Vpr but also to design future antiviral therapies. In this report, we describe identification of elongation factor 2 (EF2) as such a cellular factor. Specifically, EF2 protein level is responsive to vpr gene expression; it is able to suppress Vpr-induced apoptosis when it is overproduced beyond its physiological level. EF2 was initially identified through a genome-wide multicopy suppressor search for Vpr-induced apoptosis in a fission yeast model system. Overproduction of fission yeast Ef2 completely abolishes Vpr-induced cell killing in fission yeast. Similarly, overexpression of the human homologue of yeast Ef2 in a neuroblastoma SKN-SH cell line and two CD4+ H9 and CEM-SS T-cell lines also blocked Vpr-induced apoptosis. The anti-apoptotic property of EF2 is demonstrated by its ability to suppress caspase 9 and caspase 3-mediated apoptosis induced by Vpr. In addition, it also reduces cytochrome c release induced by Vpr, staurosporine and TNFalpha. The fact that overproduction of EF2 blocks Vpr-induced cell death both in fission yeast and human cells, suggested that EF2 posses a highly conserved anti-apoptotic activity. Moreover, the responsive elevation of EF2 to Vpr suggests a possible host innate antiviral response.


Subject(s)
Apoptosis/physiology , Gene Products, vpr/metabolism , HIV-1/metabolism , Peptide Elongation Factor 2/metabolism , CD4-Positive T-Lymphocytes/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Enzyme Inhibitors/metabolism , Gene Products, vpr/genetics , HIV-1/genetics , Humans , Peptide Elongation Factor 2/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Staurosporine/metabolism , Tumor Necrosis Factor-alpha/metabolism , vpr Gene Products, Human Immunodeficiency Virus
3.
Cell Prolif ; 35(2): 105-15, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11952645

ABSTRACT

The inhibitory action of gangliosides GT1B, GD1A, GM3 and GM1 on cell proliferation and epidermal growth factor receptor (EGFR) phosphorylation was determined in the N-myc amplified human neuroblastoma cell line NBL-W. The IC50 of each ganglioside was estimated from concentration-response regressions generated by incubating NBL-W cells with incremental concentrations (5-1000 microm) of GT1B, GD1A, GM3 or GM1 for 4 days. Cell proliferation was quantitatively determined by a colourimetric assay using tetrazolium dye and spectrophotometric analysis, and EGFR phosphorylation by densitometry of Western blots. All gangliosides assayed, with the exception of GM1, inhibited NBL-W cell proliferation in a concentration-dependent manner. The IC50s for gangliosides GT1B [molecular weight (MW) 2129], GM3 (MW 1236), and GD1A (MW 1838) were (mean +/- SEM) 117 +/- 26, 255 +/- 29, and 425 +/- 44 m, respectively. In contrast, the IC50 for GM1 (MW 1547) could not be determined. Incubation of NBL-W cells with epidermal growth factor (EGF) concentrations ranging from 0.1 to 1000 ng/ml progressively increased cell proliferation rate, but it plateaued at concentrations above 10 ng/ml. EGFR tyrosine phosphorylation, however, was incrementally stimulated by EGF concentrations from 1 to 100 ng/ml. The suppression of EGF-induced EGFR phosphorylation differed for each ganglioside, and their respective inhibitory potencies were as follows: EGFR phosphorylation [area under curve (+ EGF)/area under curve (- EGF)]: control (no ganglioside added) = 8.2; GM1 = 8.3; GD1A = 6.7; GM3 = 4.87, and GT1B = 4.09. The lower the ratio, the greater the inhibitory activity of the ganglioside. Gangliosides GD1A and GT1B, which have terminal N-acetyl neuraminic acid moieties, as well as one and two N-acetyl neuraminic acid residues linked to the internal galactose, respectively, both inhibited cell proliferation and EGFR phosphorylation. However, GD1A was a more potent suppressor of cell proliferation and GT1B most effective against EGFR phosphorylation. GM3, which only has a terminal N-acetyl neuraminic acid, inhibited cell proliferation and EGFR phosphorylation almost equivalently. These data suggest that gangliosides differ in their potency as inhibitors of NBL-W neuroblastoma cell proliferation and EGFR tyrosine phosphorylation, and that perturbations in the differential expression of membrane glycosphingolipids may play a role in modulating neuroblastoma growth.


Subject(s)
ErbB Receptors/metabolism , G(M1) Ganglioside/metabolism , G(M3) Ganglioside/metabolism , Neuroblastoma , Animals , Carbohydrate Sequence , Cell Division/drug effects , Cell Division/physiology , Dogs , Epidermal Growth Factor/pharmacology , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/pharmacology , G(M3) Ganglioside/chemistry , G(M3) Ganglioside/pharmacology , Gangliosides/chemistry , Gangliosides/metabolism , Gangliosides/pharmacology , Humans , Molecular Sequence Data , Phosphorylation , Structure-Activity Relationship , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Tyrosine/metabolism
4.
Cell Signal ; 13(7): 499-505, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11516625

ABSTRACT

The mitogen-activated protein kinase (MAPk) signaling pathway, which plays a critical role in the proliferation of mammalian cells, is frequently up-regulated in human tumors and may contribute to the transformed phenotype. Since a major limitation of current cancer chemotherapy is prevalent resistance to cytotoxic drugs, this study determined whether alterations in growth factor signaling through MAPk may contribute to this phenomenon in human neuroblastoma cell lines. Drug-resistant SKNSH cell lines were established by long-term incubation with increasing concentrations to 10(-6) M doxorubicin (SKNSH rDOX6) or MDL 28842 (SKNSH rMDL6). The expression of epidermal growth factor receptor (EGFR) and epidermal growth factor (EGF)-induced EGFR tyrosine phosphorylation were lower in drug-resistant SKNSH cells than their wild-type counterparts. In SKNSH rDOX6 cells, decreased activation and reduced nuclear translocation of MAPk in response to EGF, or lysophosphatidic acid (LPA), or phorbol 12-myristate 13-acetate (PMA), were observed. In SKNSH rMDL6 cells, although MAPk could be activated to wild-type levels by ligand stimulation, the translocation of active MAPk to the nucleus was also reduced. These results suggest that resistance to cytotoxic drugs in human neuroblastoma cell lines is associated with a decrease in growth factor signaling through the MAPk pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Growth Substances/pharmacology , MAP Kinase Signaling System , Neuroblastoma/drug therapy , Active Transport, Cell Nucleus , Adenosine/analogs & derivatives , Adenosine/pharmacology , Cell Nucleus/metabolism , Down-Regulation , Doxorubicin/pharmacology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Lysophospholipids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Neuroblastoma/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
5.
Mol Med ; 7(6): 393-400, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11474132

ABSTRACT

BACKGROUND: The transcription factor Stat3 has been reported to play a key role in protecting cells against apoptosis by up-regulating expression of the anti-apoptotic gene BCl-xL. This investigation analyzes the relationship between the development of resistance to doxorubicin-mediated apoptosis in neuroblastoma cells (SKN-SH) and activation of the Stat3 signaling pathway. MATERIALS AND METHODS: A drug-resistant cell line (SKN-SH/Dox6) was generated by continuous exposure to incremental concentrations of doxorubicin. Specific antibodies were utilized for Western blots and confocal microscopy to determine the nuclear localization of activated Stat3. RESULTS: Doxorubicin-mediated DNA fragmentation was inhibited and caspase-3 activity decreased in SKN-SH/Dox6 cells. Up-regulation of Stat3 phosphorylation and Bcl-xL expression, increased nuclear translocation of phospho-Stat3, and binding to DNA occurred only in resistant SKN-SH/Dox6 cells. The expression of Bcl-xL was inhibited by AG490, an inhibitor of the JAK/Stat3 signaling pathway, suggesting that the regulation of Bcl-xL and Stat3 involved a common mechanism. Activation of Stat3 in SKN-SH/Dox6 cells was contingent upon stimulation evoked by ligands secreted by the drug-resistant cells. Evidence to support this hypothesis was provided by experiments in which doxorubicin-sensitive SKN-SH cells were preincubated with conditioned media obtained from doxorubicin-resistant SKN-SH/Dox6 cells. This treatment increased Stat3 activation. It also rendered SKN-SH cells resistant to doxorubicin as demonstrated by a sharp decrease in doxorubicin-induced DNA degradation and cytotoxic potency. CONCLUSIONS: These findings suggest that the resistance of SKN-SH/Dox6 cells to doxorubicin may be mediated by anti-apoptotic factor(s) that are synthesized and secreted by tumor cells in response to cytotoxic agents.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Trans-Activators/metabolism , Animals , Culture Media, Conditioned , Humans , Neuroblastoma/chemistry , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT3 Transcription Factor , Signal Transduction , Tumor Cells, Cultured , Tyrphostins/pharmacology , bcl-X Protein
6.
Int J Oncol ; 18(2): 323-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11172599

ABSTRACT

We have previously reported that C-1300 murine neuroblastoma (rMNB) cells made resistant to the nucleoside analogue, (Z)-5'-fluoro-4', 5'-didehydro-5'deoxyadenosine (MDL), an irreversible inhibitor of S-adenosylhomocysteine (AdoHcy) hydrolase have an increased expression of the S-adenosylmethionine (AdoMet) synthetase gene. Results of the immunoblot analysis of DNA (cytosine) methyltransferase with anti-human DNA (cytosine) methyltransferase specific polyclonal antibody demonstrated a significant increase ( approximately 2-fold, p<0.01) in expression of DNA (cytosine) methyltransferase protein in rMNB/MDL cells compared to wild-type C1300 MNB (wMNB) cells. To rule out the possibility that multidrug resistance (MDR) genes are involved in development of acquired drug resistance in murine neuroblastoma (rMNB/MDL) cells made resistant to MDL, the expression of Mdr1a, Mdr1b, Mdr2 (multidrug resistance/P-glycoprotein), and Mrp-1 (multidrug resistance associated protein) was examined in rMNB-MDL cells. The analysis of Mdr and Mrp-1 expression was performed by RT-PCR using PCR specific primers to respective genes. No significant difference was observed in the expression of MDR1a, Mdr1b and Mrp-1 genes between wMNB and rMNB-MDL cells, however, a slight decrease was noticed in Mdr1 expression in some samples. Expression of the Mdr2 (human MDR3) gene, which is not associated with the acquired drug resistance phenotype, was significantly decreased in rMNB-MDL cells. These findings were also confirmed by the immunoblot analyses using specific monoclonal antibodies to Mdr1/3 proteins. Expression of N-Myc gene--a prognostic factor in neuroblastoma tumors was also not altered in rMNB-MDL cells. Results of the present study suggest that acquired drug resistance in rMNB-MDL cells to MDL is associated to the overexpression of DNA (cytosine) methyltransferase, and could be due to genetic or epigenetic changes in particular to DNA hypermethylation in response to an increased AdoMet synthetase gene expression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression/physiology , Neuroblastoma/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , Drug Resistance, Neoplasm/physiology , Genes, MDR/genetics , Genes, MDR/physiology , Humans , Mice , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neuroblastoma/genetics , Tumor Cells, Cultured/metabolism
7.
Cancer Chemother Pharmacol ; 48(6): 423-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11800021

ABSTRACT

UNLABELLED: The induction of p53 expression and stimulation of the Fas/caspase-8 pathway represent major mechanisms by which cytotoxic drugs induce apoptosis, but in neuroblastomas, the caspase-8 gene is often not expressed. PURPOSE: The aim of this study was to determine whether doxorubicin could induce apoptosis in caspase-8-deficient neuroblastoma cells and to define its mechanism of action. METHODS: The caspase-8-deficient human neuroblastoma cell line, SKN-SH, was incubated with doxorubicin and the apoptotic response, as well as expression of apoptotic molecules in the p53/ Fas/caspase-8 pathway, were determined. RESULTS: SKN-SH cells incubated with doxorubicin readily underwent apoptosis in a concentration-dependent manner. Western blot analyses with specific antibodies demonstrated that both p53 and Fas ligand were endogenously expressed in SKN-SH cells, but their expression was not stimulated by doxorubicin. Fas receptor was not detected in these cells and caspase-8 was totally absent. Electron microscopic analyses of SKN-SH cells treated with doxorubicin revealed pronounced alterations in mitochondrial structure. This treatment also induced the release of cytochrome c from mitochondria and activated the downstream apoptotic intermediate, caspase-3. CONCLUSION: These results indicate that the p53/Fas/caspase-8 system does not play a role in mediating the apoptotic action of doxorubicin in the human neuroblastoma cell line SKN-SH. Thus, mitochondria and downstream apoptotic signaling intermediates may be considered as key targets for doxorubicin-induced apoptosis in neuroblastoma tumors having deficiencies in the Fas/caspase-8 system.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Doxorubicin/pharmacology , Gene Expression Regulation , Mitochondria/physiology , Caspase 8 , Caspase 9 , Caspases/biosynthesis , Caspases/deficiency , Genes, p53 , Humans , Neuroblastoma/pathology , Signal Transduction , Tumor Cells, Cultured , fas Receptor
8.
J Neurosci Res ; 59(2): 265-75, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10650885

ABSTRACT

The response of wild-type and genetically engineered neuroectodermal tumor (NET) cells to exogenous and endogenously synthesized nerve growth factor (NGF) was investigated. Differences in cell proliferation rate, neurite formation, and expression of NGF binding sites were quantitatively determined. Ecotropic retroviral vectors were used to transfer the genes for beta-galactosidase (beta-GAL) and NGF into wild-type C-1300 and Neuro-2A murine neuroblastoma (MNB) and rat pheochromocytoma (PC-12) cells. Conditioned media obtained from NET cells infected with the NGF gene contained biologically active NGF, whereas media from beta-GAL infected cells did not. Infection with the NGF vector induced a short-term decrease in cell proliferation rate and increased neurite formation in wild-type, substrate-adherent PC-12 and Neuro-2A MNB cells (P > 0.05). Incubation of wild-type C-1300, Neuro-2A MNB, and PC-12 cells with NGF (0-200 ng/ml) for 5 days significantly reduced proliferation rates in a concentration-dependent manner and increased neurite extrusion. All NGF-NET cells had a significantly diminished response to the antiproliferative action of exogenous NGF. Ligand binding assays with 125I-NGF demonstrated a marked reduction in the number of NGF binding sites on NGF-NET cells compared to wild type. The attenuated response of NGF-NET cells to exogenous NGF correlated positively with the down-regulation of NGF binding sites. In conclusion, beta-NGF gene transfer into wild-type NET cells induces the synthesis and secretion of NGF, temporarily decreases cell proliferation rate, increases neurite extrusion, down-regulates NGF binding sites, and reduces NET cell responsiveness to NGF. A putative role for NGF may be the modulation of NET cell proliferation and differentiation.


Subject(s)
Gene Transfer Techniques , Nerve Growth Factor/genetics , Neurites/physiology , Retroviridae , Animals , Binding Sites , Brain Neoplasms , Cell Division/genetics , Cell Size , Fluorescent Antibody Technique , Gene Expression Regulation, Viral , Iodine Radioisotopes , Nerve Growth Factor/chemistry , Nerve Growth Factor/metabolism , Neuroblastoma , Neuroectodermal Tumors , PC12 Cells , Radioligand Assay , Rats , Retroviridae Infections , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , beta-Galactosidase/analysis , beta-Galactosidase/genetics
9.
Clin Cancer Res ; 5(11): 3661-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589784

ABSTRACT

Increased expression of the bisecting GlcNAc has been correlated with tumor progression in several experimental tumor models. Its expression and function in brain tumors are, however, not yet known. In this study, we investigated expression of the bisecting GlcNAc structure in a series of pediatric brain tumors and its relationship to tumor response to vinblastine. A plant lectin (E-PHA) that recognizes the bisecting GlcNAc structure was used for detection of this molecule in a total of 90 pediatric brain tumors and normal brain tissue specimens. Our results showed that, whereas E-PHA staining was undetectable in the normal brain tissue, pediatric brain tumor specimens exhibited different levels of reactivity. Lectin staining was particularly prominent in high-grade astrocytomas (73%) and ependymomas (72%). In astrocytomas, there was a positive correlation with the tumor grade, which suggests that the bisecting GlcNAc may be of particular interest as a tumor marker for diagnosis and/or prognosis. By using a human glioma cell culture model, we have found that treatment of these cells with E-PHA lectin enhances their sensitivity to vinblastine. E-PHA interacted directly with the drug transporter P-glycoprotein and inhibited its drug efflux function. In a drug-resistant glioma cell line transfected with the mdr1 gene, drug resistance was reversed by E-PHA. Our findings indicate that: (a) expression of the bisecting GlcNAc in pediatric brain tumors may have a potential relevance as a tumor marker; and (b) glioma response to chemotherapy may be modulated through the bisecting GlcNAc.


Subject(s)
Acetylglucosamine/metabolism , Brain Neoplasms/pathology , Cerebral Cortex/cytology , Phytohemagglutinins , Vinblastine/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acetylglucosamine/analysis , Astrocytoma/pathology , Carbohydrate Sequence , Cell Division/drug effects , Cerebellar Neoplasms/pathology , Cerebral Cortex/pathology , Ependymoma/pathology , Glioma/metabolism , Glioma/pathology , Glycosylation , Humans , Lectins , Medulloblastoma/pathology , Molecular Sequence Data , Oligosaccharides/biosynthesis , Oligosaccharides/chemistry , Phytohemagglutinins/pharmacology , Tumor Cells, Cultured
10.
Cancer Res ; 59(8): 1852-6, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10213491

ABSTRACT

S-Adenosylmethionine (AdoMet) synthetase (EC 2.5.1.6), which catalyzes the synthesis of AdoMet from methionine and ATP, is the major methyl donor for transmethylation reactions and propylamino donor for the biosynthesis of polyamines in biological systems. We have reported previously that wild-type C-1300 murine neuroblastoma (wMNB) cells, made resistant to the nucleoside analogue (Z)-5'-fluoro-4',5'-didehydro-5'-deoxyadenosine (MDL 28,842), an irreversible inhibitor of S-adenosylhomocysteine (AdoHcy) hydrolase (EC 3.3.1.1), express increased AdoMet synthetase activity (M. R. Hamre et al., Oncol. Res., 7: 487-492, 1995). In the present study, immunoblot analyses of AdoMet Synthetase with isoform-specific (MATII) antibodies demonstrated an elevation in the AdoMet synthetase immunoprotein in nucleoside analogue-resistant MNB cells (rMNB-MDL) when compared to wild-type, nonresistant MNB cells. An increase of 2.1-fold was observed in the alpha2/alpha2' catalytic subunit, which differed significantly from the much smaller increment in the noncatalytic beta-subunit of AdoMet synthetase. Densitometric analyses revealed that an increased expression of AdoMet synthetase in rMNB-MDL cells was due to overexpression of the alpha2 (Mr 53,000; 2.6-fold) and alpha2' (Mr 51,000; 1.8-fold) subunits. AdoMet synthetase mRNA expression in rMNB-MDL cells was remarkably greater than wMNB cells, as determined by quantitative competitive reverse transcription-PCR (QC-PCR) analysis. DNA (cytosine) methyl transferase expression, measured by reverse transcription-PCR analysis, was also elevated significantly in rMNB-MDL cells. In contrast, Western blot analyses demonstrated down-regulation (1.6-fold) of AdoMet synthetase in doxorubicin-resistant human leukemia cells (HL-60-R) expressing multidrug resistance protein when compared with wild-type, nonresistant HL-60 cells. The resistance of rMNB-MDL cells to nucleoside analogue inhibitors of S-adenosylhomocysteine hydrolase correlates directly with overexpression of the alpha2/alpha2' subunits of AdoMet synthetase. Cellular adaptation allows sufficient AdoMet to be synthesized, so that viability of the MNB cells can be maintained even in the presence of high AdoHcy concentrations. This novel mechanism of drug resistance does not appear to require multidrug resistance protein (P-glycoprotein) overexpression.


Subject(s)
Methionine Adenosyltransferase/biosynthesis , Neuroblastoma/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosylhomocysteinase , Animals , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Gene Expression , HeLa Cells , Humans , Hydrolases/antagonists & inhibitors , Methionine Adenosyltransferase/genetics , Mice , Neuroblastoma/enzymology , Neuroblastoma/metabolism , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
11.
Anticancer Res ; 15(3): 661-6, 1995.
Article in English | MEDLINE | ID: mdl-7645939

ABSTRACT

Exogenous gangliosides have been shown to exert a regulatory influence on the proliferation and differentiation of several cell lines in tissue culture. The effect of ganglioside GM3 on C-1300 murine neuroblastoma (MNB) cell proliferation and the modulating action of fetal calf serum (FCS) concentration in the culture media have been investigated. MNB cells were cultured in DMEM containing 1, 2.5, 5 or 10% FCS, and incubated with GM3 at concentrations ranging from 1.95 to 500 microM. Cell proliferation was assayed 4 days after the addition of GM3 using the CellTiter 96 Non-Radioactive Cell Proliferation Assay. GM3 inhibited MNB cell proliferation in a dose-dependent manner regardless of the FCS concentration in the culture media. However, the magnitude of this inhibitory effect was inversely proportional to the FCS concentration in the culture media. The addition of albumin to MNB cells cultured in DMEM containing 1% FCS exerted no effect on the antiproliferative action of GM3. FACS cell cycle analyses demonstrated that MNB cultured in DMEM containing 1% FCS had a higher proportion of cells in the G0/G1 compartment when compared to those cultured in 10% FCS. The enhanced response of MNB cells to GM3 observed in 1% FCS, may be due to a preferential action on cells in the G0/G1 stage of the cell cycle. These studies have demonstrated that the ganglioside GM3 inhibited MNB cell proliferation in tissue culture and this effect was modulated by FCS concentration in the culture media. Since protein-binding of GM3 by FCS does not appear to be the primary mechanism by which FCS exerts its antagonistic effects, we hypothesize that this may be due to the opposing action of stimulatory growth factors present in FCS.


Subject(s)
Cell Division/drug effects , G(M3) Ganglioside/pharmacology , Animals , Blood , Cattle , Cell Differentiation/drug effects , Culture Media , Culture Media, Serum-Free , Dose-Response Relationship, Drug , Kinetics , Mice , Neuroblastoma , Serum Albumin/pharmacology , Time Factors , Tumor Cells, Cultured
12.
Oncol Res ; 7(10-11): 487-92, 1995.
Article in English | MEDLINE | ID: mdl-8866660

ABSTRACT

Adaptive changes occurring in C1300 murine neuroblastoma cell lines developed for resistance to nucleoside analogue inhibitors of S-adenosyl-L-homocysteine hydrolase (AdoHcyase, EC 3.3.1.1) were investigated. Two drug-resistant cell lines, rMNB-MDL-7-2 and rMNB-Deaz-7-2, were established from wild-type C1300 neuroblastoma cells (wMNB) following incubation with the AdoHcyase inhibitors (Z)-4',5'-didehydro-5'-deoxy-5'-fluoroadenosine (MDL 28,842) and 3-deazaneplanocin A, respectively. The nucleoside analogue concentration required to inhibit cellular proliferation by 50% (IC50) was 3.2 x 10(2) to 4.3 x 10(3) fold higher in the resistant cells when compared with the wMNB cell line. The proliferation rates of the resistant cell lines under in vitro or in vivo conditions were significantly lower than the wMNB cell line. In contrast to wMNB, both resistant cell lines had slower doubling times in tissue culture (22% longer) and smaller tumor weights (55% smaller) 14 days after implantation in A/J mice. No significant differences in AdoHcyase activity were noted between the resistant and wild-type cell lines. The resistant cell lines had concentrations of S-adenosyl-L-methionine that were five times higher and methionine adenosyltransferase (MAT, EC 2.5.1.6) activities that were two to four times greater than the wMNB phenotype. These data indicate that neuroblastoma tumor cell resistance to AdoHcyase inhibitors is associated with an adaptive increase in MAT activity. This cellular response facilitates methylation by elevating intracellular concentrations of the methyl donor S-adenosyl-L-methionine, thereby sustaining tumor cell viability in the presence of MDL 28,842 and 3-deazaneplanocin A.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Methionine Adenosyltransferase/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Adenosine/analogs & derivatives , Adenosine/pharmacokinetics , Adenosine/pharmacology , Adenosylhomocysteinase , Animals , Cell Division/drug effects , Drug Resistance, Neoplasm , Male , Methylation/drug effects , Mice , Mice, Inbred A , Neuroblastoma/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Tritium , Tumor Cells, Cultured
13.
Cancer Res ; 53(24): 6058-60, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8261422

ABSTRACT

S-Adenosylmethionine-dependent transmethylation reactions are required for many critical pathways in human cells. The enzyme S-adenosylhomocysteine hydrolase converts S-adenosylhomocysteine, a potent endogenous inhibitor of S-adenosylmethione-mediated methyltransferase reactions, to adenosine and L-homocysteine. The effects of the inhibitor of S-adenosylhomocysteine hydrolase, Z-4',5'-didehydro-5'-deoxy-5'-fluoroadenosine (MDL 28,842), on the growth of cultured keratinocytes and cutaneous squamous cell carcinoma lines were investigated. MDL 28,842 suppressed the proliferation of all cells in a dose-dependent manner, and significantly increased keratinocyte differentiation at a concentration of 1 microM. Following incubation with MDL 28,842, the methylation indices (ratio of S-adenosylmethionine/S-adenosylhomocysteine) of undifferentiated keratinocytes and squamous cell carcinoma lines were significantly decreased. These data demonstrate that the inhibitory effect of MDL 28,842 on squamous carcinoma cells and keratinocyte proliferation may result directly from inhibition of S-adenosylhomocysteine hydrolase activity. The antiproliferative activity of MDL 28,842 against squamous carcinoma cells and keratinocytes suggests a potential role for MDL 28,842 as a novel therapeutic agent for neoplastic and hyperproliferative disorders of the skin.


Subject(s)
Adenosine/analogs & derivatives , Carcinoma, Squamous Cell/pathology , Hydrolases/antagonists & inhibitors , Keratinocytes/drug effects , Adenosine/pharmacology , Adenosylhomocysteinase , Carcinoma, Squamous Cell/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Methylation , Tumor Cells, Cultured
14.
Oncol Res ; 5(10-11): 433-9, 1993.
Article in English | MEDLINE | ID: mdl-8054704

ABSTRACT

The effect of the irreversible S-adenosyl-L-homocysteine hydrolase inhibitor, (Z)-5'-fluoro-4',5'-didehydro-5'-deoxyadenosine (MDL 28,842), on C-1300 murine neuroblastoma (MNB) cell proliferation in tissue culture and MNB tumor growth in vivo were investigated. MNB cells were incubated with MDL 28,842 at concentrations ranging from 8 x 10(-9) M to 1.6 x 10(-5) M for 3 days, and cell proliferation was determined by use of a CellTiter 96-well Proliferation Assay Kit. In tissue culture, MDL 28,842 inhibited MNB cell proliferation in a concentration-dependent manner, and the IC50 of MDL 28,842 against MNB in tissue culture was 1.8 x 10(-7) M. The response of in vivo tumor growth and host survival to MDL 28,842d was evaluated in a syngeneic mouse tumor model prepared by s.c. implantation of 1 x 10(6) MNB cells into A/J mice. Following palpation of a tumor mass, osmotic minipumps were implanted into each mouse. MDL 28,842 was infused at rates of 1.0 or 1.5 mg/kg/day over a 10-day period and 1.25 mg/kg/day over a 30-day period. The mean survival time of tumor-bearing mice significantly increased from 28.75 +/- 1.06 days (mean +/- 2 SEM) in the control group (diluent infusion) to 39.33 +/- 1.58 days, 44.11 +/- 1.74 days, and 41.0 +/- 1.30 days in the MDL 28,842-treated groups receiving 10-day infusions of 1.0 and 1.5 mg/kg/day or 30-day infusions of 1.25 mg/kg/day, respectively. No significant differences in survival rate were noted between groups receiving 10 vs 30 days of drug treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adenosine/analogs & derivatives , Enzyme Inhibitors/toxicity , Hydrolases/antagonists & inhibitors , Neuroblastoma/drug therapy , Adenosine/therapeutic use , Adenosine/toxicity , Adenosylhomocysteinase , Animals , Cell Division/drug effects , Cell Line , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred A , Neuroblastoma/pathology , Probability , Tumor Cells, Cultured
15.
Cancer Res ; 52(20): 5826-8, 1992 Oct 15.
Article in English | MEDLINE | ID: mdl-1356621

ABSTRACT

Inosine-5'-phosphate (IMP) dehydrogenase, a regulatory enzyme of guanine nucleotide biosynthesis, may play a role in cell proliferation and malignancy. To assess this role we examined IMP dehydrogenase expression in a series of human solid tumor tissues and tumor cell lines in comparison with their normal counterparts. Increased IMP dehydrogenase gene expression was observed in brain tumors relative to normal brain tissue and in sarcoma cells relative to normal fibroblasts. Similarly, in several B- and T-lymphoid leukemia cell lines, elevated levels of IMP dehydrogenase mRNA and cellular enzyme were observed in comparison with the levels in peripheral blood lymphocytes. These results are consistent with an association between increased IMP dehydrogenase expression and either enhanced cell proliferation or malignant transformation.


Subject(s)
Gene Expression Regulation, Neoplastic , IMP Dehydrogenase/genetics , Neoplasm Proteins/genetics , Animals , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , IMP Dehydrogenase/metabolism , Neoplasm Proteins/metabolism , Tumor Cells, Cultured
16.
J Pharmacol Exp Ther ; 262(3): 1070-5, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1527714

ABSTRACT

The relationship between 6-hydroxydopamine (6-OHDA)-induced ablation of central and peripheral adrenergic neurons and in situ C-1300 murine neuroblastoma (MNB) tumor growth and catecholamine concentration were investigated. Destruction of central and/or peripheral adrenergic neurons was produced by the intracerebral and/or s.c. administration of 6-OHDA to neonatal A/J mice. Disaggregated MNB cells (1 x 10(6)) were implanted s.c. into each mouse 3 weeks after treatment with 6-OHDA or diluent. Tumor onset time (the time interval between implantation of MNB cells and detection of palpable tumor), tumor weight, tumor weight to body weight ratio, tumor growth rate constant and tissue catecholamine concentrations were determined. Central axotomy caused a significant increase in tumor onset time and decrease in tumor weight when compared to controls. However, neither the tumor weight to body weight ratio or tumor growth rate constant were significantly lowered. In contrast, a reduction in all tumor growth parameters was produced by peripheral axotomy, which differed significantly from centrally axotomized and control animals. The catecholamine concentration of MNB tumors excised from control and 6-OHDA-treated mice 8 days after tumor onset were determined. Norepinephrine and dopamine levels were elevated above controls in MNB tumors obtained from mice that had been either peripherally or peripherally and centrally axotomized; whereas, no change in tumor catecholamine concentrations was noted in centrally axotomized mice. This investigation has demonstrated that ablation of central as well as peripheral adrenergic innervation exerts an inhibitory effect on MNB tumor growth.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Axons/drug effects , Catecholamines/chemistry , Neoplasms, Experimental/chemistry , Neuroblastoma/chemistry , Oxidopamine/pharmacology , Animals , Body Weight/drug effects , Chromatography, High Pressure Liquid , Injections, Intraventricular , Male , Mice , Mice, Inbred A , Neoplasms, Experimental/metabolism , Neuroblastoma/metabolism , Organ Size/drug effects , Tissue Distribution
17.
J Neurosci Res ; 28(3): 343-51, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1906944

ABSTRACT

The in situ C-1300 murine neuroblastoma (MNB) tumor model was used to investigate the influence of exogenously administered nerve growth factor (NGF) on tumor growth and tissue catecholamine concentration in mice sympathectomized with 6-hydroxy-dopamine (6-OHDA) on postnatal days 4-10. Mice were implanted with 1 x 10(6) disaggregated MNB cells 3 days after termination of 6-OHDA administration. NGF (12-15 micrograms/mouse/day) treatment was initiated at the time of MNB cell implantation and continued until sacrifice of the animal. The time interval between tumor cell implantation and detection of palpable tumor (tumor onset time), transverse tumor diameter, tumor weight, tumor weight to body weight ratio, and tumor catecholamine concentration were determined. Neonatal sympathectomy caused a decrease in myocardial norepinephrine concentration of 88% compared with vehicle-treated animals as well as a significant reduction in total body and organ weight. Average body, brain, heart, and spleen weights were decreased 31%, 16%, 25%, and 42%, respectively, below control values. The daily injection of NGF, from the time of MNB tumor implantation to sacrifice, did not prevent these effects of chemical sympathectomy from being expressed. Tumor onset time following implantation of MNB cells was significantly increased in neonatally sympathectomized mice and was not altered by treatment with NGF. In contrast, the decrease in MNB tumor growth rate observed in sympathectomized mice was reversed by administration of NGF. Mean tumor weight and mean tumor to body weight ratio were 89% and 115% of comparable control values, respectively, in sympathectomized mice receiving exogenous NGF.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Nerve Growth Factors/pharmacology , Neuroblastoma/pathology , Animals , Animals, Newborn , Hydroxydopamines/toxicity , Mice , Mice, Inbred A , Myocardium/chemistry , Neoplasm Transplantation , Norepinephrine/analysis , Oxidopamine , Sympathectomy, Chemical
18.
Cancer Lett ; 50(1): 11-8, 1990 Apr 09.
Article in English | MEDLINE | ID: mdl-2322923

ABSTRACT

The influence of host age and tumor load on survival time, tumor growth parameters and biochemical differentiation, as characterized by tumor catecholamine content, were investigated. A/J mice were implanted with tumor loads of 10(4), 10(5) and 10(6) disaggregated C-1300 murine neuroblastoma (MNB) cells at 1, 7, 14, 21 and 56 days of age. Studies performed in mice between 7 and 56 days of age demonstrated that MNB tumorigenicity, tumor growth rate, host survival and catecholamine content were independent of host age and tumor load whereas, tumor onset time was influenced by both factors. In contrast to older animals, tumor onset time and catecholamine content were decreased and tumor growth rate increased in 1-day-old mice. This difference may be due to the presence of endogenous growth factor(s) that modulate cell proliferation in the immediate post-natal period.


Subject(s)
Catecholamines/analysis , Neuroblastoma/pathology , Age Factors , Animals , Body Weight , Mice , Neuroblastoma/analysis
19.
Life Sci ; 47(24): 2251-9, 1990.
Article in English | MEDLINE | ID: mdl-2125097

ABSTRACT

The in situ growth characteristics of C-1300, N1E-115 and NS20Y murine neuroblastoma (MNB) tumor cell lines were compared in normal and sympathectomized A/J mice. Adrenergic nerve ablation was produced in neonatal mice by administration of 6-hydroxydopamine (6-OHDA) at a dose of 100 micrograms/gm body weight on post-natal days 4, 6, 8 and 10; controls received equivalent volumes of the vehicle solution (0.9% NaCl/0.1% Ascorbic Acid). All mice were inoculated subcutaneously with 10(6) viable MNB cells four to six weeks after treatment with 6-OHDA or vehicle. The growth rates of tumors produced by the adrenergic MNB cell lines, C-1300 and N1E-115, were significantly lower in sympathectomized mice when compared to control animals. In contrast, tumors induced by the cholinergic MNB cell line, NS20Y, grew at similar rates in both sympathectomized and control mice. All tumors obtained from control and sympathectomized mice regardless of whether they derived from cell lines characterized as cholinergic (NS20Y) or adrenergic (C-1300, N1E-115), contained both norepinephrine and dopamine. Depletion of adrenergic neurotransmitter in A/J mice was induced by administration of reserpine (5-10 micrograms/kg/day) beginning 30 days prior to implantation of the C-1300 MNB cell line and continuing until sacrifice of the animal. The effect of this treatment on organ and tumor catecholamine concentrations was confirmed by high-pressure liquid chromatography. Splenic catecholamine levels in reserpine-treated animals were reduced to 20% of controls as compared to 9% in the neonatally-sympathectomized group. However, there was no discernible effect on C-1300 MNB tumor growth in the reserpine-treated animals. C-1300 MNB tumor concentrations of nor-epinephrine and dopamine were significantly lower in the reserpine-treated animals than in controls. The suppression of tumor growth by adrenergic nerve ablation is selective for specific MNB tumor cell lines. An anatomically intact sympathetic nervous system appears to exert a greater influence than competency of adrenergic neuro-humoral transmission on MNB tumor growth. These data support the hypothesis that modulation of MNB tumor growth by the adrenergic nervous system is not mediated via catecholamines but may be modulated by endogenous growth factor(s).


Subject(s)
Neuroblastoma/pathology , Sympathetic Nervous System/physiology , Animals , Carcinogenicity Tests , Cell Division , Dopamine/metabolism , Female , Hydroxydopamines , Mice , Mice, Inbred A , Neuroblastoma/metabolism , Norepinephrine/metabolism , Oxidopamine , Reserpine , Sympathectomy, Chemical , Sympathetic Nervous System/metabolism , Tumor Cells, Cultured
20.
Life Sci ; 45(26): 2539-45, 1989.
Article in English | MEDLINE | ID: mdl-2559274

ABSTRACT

Attempts have been made to confirm previously reported results which demonstrated that the opioid antagonist, naltrexone, altered the in situ growth of murine neuroblastoma tumors. Adult male A/J mice were injected with tumor cells from three different cell lines of murine neuroblastoma; the spontaneously arising C-1300 line, the adrenergic clonal line N1E-115, and the cholinergic clonal line NS20Y. Naltrexone was administered daily in doses of 0.1, 0.4, or 10.0 mg/kg subcutaneously, to replicate the reported experimental design. In contrast to previous studies, we were unable to demonstrate any effect of naltrexone on in situ growth or other characteristics of tumors produced by the C-1300, N1E-115 or NS20Y murine neuroblastoma cell lines. Ligand binding studies have demonstrated the presence of high levels of opiate binding sites on membranes prepared from the NS20Y clonal cell line and low levels on the membranes of the C1300 tumor line.


Subject(s)
Endorphins/metabolism , Naltrexone/pharmacology , Neuroblastoma/drug therapy , Animals , Body Weight , Chi-Square Distribution , Endorphins/antagonists & inhibitors , Male , Mice , Mice, Inbred A , Naltrexone/metabolism , Neoplasm Transplantation , Neoplasms, Experimental , Neuroblastoma/metabolism , Receptors, Opioid/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...