Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Biochimie ; 95(2): 320-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23063694

ABSTRACT

In vitro assay of mammalian DNA replication has been variously approached. Using gapped circular duplex substrates containing a 500-base single-stranded DNA region, we have constructed a mammalian cell-free system in which physiological DNA replication may be reproduced. Reaction of the gapped plasmid substrate with crude extracts of human HeLaS3 cells induces efficient DNA synthesis in vitro. The induced synthesis was strongly inhibited by aphidicolin and completely depended on dNTP added to the system. In cell extracts in which PCNA was depleted step-wise by immunoprecipitation, DNA synthesis was accordingly reduced. These data suggest that replicative DNA polymerases, particularly pol delta, may chiefly function in this system. Furthermore, DNA synthesis is made quantifiable in this system, which enables us to evaluate the efficiency of DNA replication induced. Our system sensitively and quantitatively detected the reduction of the DNA replication efficiency in the DNA substrates damaged by oxidation or UV cross-linking and in the presence of a potent chain terminator, ara-CTP. The quantitative assessment of mammalian DNA replication may provide various advantages not only in basic research but also in drug development.


Subject(s)
Biological Assay , Cell-Free System/metabolism , DNA Replication/genetics , DNA, Single-Stranded , DNA-Directed DNA Polymerase/metabolism , Animals , Aphidicolin/pharmacology , Arabinofuranosylcytosine Triphosphate/pharmacology , Cell-Free System/drug effects , HeLa Cells , Humans , Kinetics , Nucleic Acid Synthesis Inhibitors , Plasmids/genetics , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism
2.
Mol Cancer Ther ; 7(10): 3373-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18852140

ABSTRACT

The therapeutic efficiency of anticancer nucleoside analogues (NA) strongly depends on their intracellular accumulation and conversion into 5'-triphosphates. Because active NATP cannot be directly administrated due to instability, we present here a strategy of nanoencapsulation of these active drugs for efficient delivery to tumors. Stable lyophilized formulations of 5'-triphosphates of cytarabine (araCTP), gemcitabine (dFdCTP), and floxuridine (FdUTP) encapsulated in biodegradable PEG-cl-PEI or F127-cl-PEI nanogel networks (NGC and NGM, respectively) were prepared by a self-assembly procedure. Cellular penetration, in vitro cytotoxicity, and drug-induced cell cycle perturbations of these nanoformulations were analyzed in breast and colorectal cancer cell lines. Cellular accumulation and NATP release from nanogel was studied by confocal microscopy and direct high-performance liquid chromatography analysis of cellular lysates. Antiproliferative effect of dFdCTP nanoformulations was evaluated in human breast carcinoma MCF7 xenograft animal model. Nanoencapsulated araCTP, dFdCTP, and FdUTP showed similar to NA cytotoxicity and cell cycle perturbations. Nanogels without drugs showed very low cytotoxicity, although NGM was more toxic than NGC. Treatment by NATP nanoformulations induced fast increase of free intracellular drug concentration. In human breast carcinoma MCF7 xenograft animal model, i.v. dFdCTP-nanogel was equally effective in inhibiting tumor growth at four times lower administered drug dose compared with free gemcitabine. Active triphosphates of NA encapsulated in nanogels exhibit similar cytotoxicity and cell cycle perturbations in vitro and faster cell accumulation and equal tumor growth-inhibitory activity in vivo at much lower dose compared with parental drugs, illustrating their therapeutic potential for cancer chemotherapy.


Subject(s)
Breast Neoplasms/pathology , Colorectal Neoplasms/pathology , Nucleosides/pharmacology , Polyethylene Glycols/metabolism , Polyethyleneimine/metabolism , Polymers/metabolism , Polyphosphates/metabolism , Animals , Antineoplastic Agents/pharmacology , Arabinofuranosylcytosine Triphosphate/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Cytarabine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Carriers , Drug Screening Assays, Antitumor , Flow Cytometry , Humans , Mice , Nanogels , Gemcitabine
3.
Int J Hematol ; 85(2): 108-15, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17321987

ABSTRACT

The combination of cytarabine (ara-C) with fludarabine is a common approach to treating resistant acute myeloid leukemia. Success depends on a fludarabine triphosphate (F-ara-ATP)-mediated increase in the active intracellular metabolite of ara-C, ara-C 5'-triphosphate (ara-CTP). Therapy-resistant leukemia may exhibit ara-C resistance, the mechanisms of which might induce cross-resistance to fludarabine with reduced F-ara-ATP formation. The present study evaluated the effect of combining ara-C and fludarabine on ara-C-resistant leukemic cells in vitro. Two variant cell lines (R1 and R2) were 8-fold and 10-fold more ara-C resistant, respectively, than the parental HL-60 cells. Reduced deoxycytidine kinase activity was demonstrated in R1 and R2 cells, and R2 cells also showed an increase in cytosolic 5'-nucleotidase II activity. Compared with HL-60 cells, R1 and R2 cells produced smaller amounts of ara-CTP. Both variants accumulated less F-ara-ATP than HL-60 cells and showed cross-resistance to fludarabine nucleoside (F-ara-A). R2 cells, however, accumulated much smaller amounts of F-ara-ATP and were more F-ara-A resistant than R1 cells. In HL-60 and R1 cells, F-ara-A pretreatment followed by ara-C incubation produced F-ara-ATP concentrations sufficient for augmenting ara-CTP production, thereby enhancing ara-C cytotoxicity. No potentiation was observed in R2 cells. Nucleotidase might preferentially degrade F-ara-A monophosphate over ara-C monophosphate, leading to reduced F-ara-ATP production and thereby compromising the F-ara-A-mediated potentiation of ara-C cytotoxicity in R2 cells. Thus, F-ara-A-mediated enhancement of ara-C cytotoxicity depended on F-ara-ATP accumulation in ara-C-resistant leukemic cells but ultimately was associated with the mechanism of ara-C resistance.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Vidarabine/analogs & derivatives , Arabinofuranosylcytosine Triphosphate/agonists , Arabinofuranosylcytosine Triphosphate/metabolism , Drug Synergism , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Vidarabine/agonists , Vidarabine/pharmacology
4.
Cancer Chemother Pharmacol ; 60(4): 467-77, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17171362

ABSTRACT

PURPOSE: The prodrug cytosinearabinoside (ara-C) is widely used in the treatment of acute leukemias. The active drug is the intracellular metabolite cytosine-arabinoside-5'-triphosphate (ara-CTP). The purpose of the present study was to investigate the relation between sensitivity and pharmacokinetic parameters Cmax, t1/2 and AUC of ara-CTP. The obtained results were compared to previous studies. EXPERIMENTAL DESIGN: Cmax, t1/2 and AUC of ara-CTP were assessed in leukemic cells of 17 pediatric patients with acute lymphoblastic leukemia (ALL) and in 6 lymphoblastic cell lines and compared with normal lymphocytes of 9 healthy donors by high pressure liquid chromatography (HPLC). The sensitivity of the cells against ara-C was determined by the MTT assay. RESULTS: The intracellular accumulation of ara-CTP was significantly lower in normal lymphocytes (Cmax 47.7-60.9 pmol/10(6) cells) compared to leukemic cell lines (Cmax 11-1128 pmol/10(6) cells) and leukemic cells of our patients (Cmax 85.9-631 pmol/10(6) cells). Similar results were found for the AUC. There was no significant difference between initial and relapsed leukemias in our small cohort. A correlation between sensitivity in terms of IC50 values and the intracellular ara-CTP accumulation was observed in cell lines, but not in leukemic cells and normal lymphocytes from healthy donors. CONCLUSIONS: Pharmacokinetic parameters varied tremendously in leukemic cells in contrast to normal lymphocytes without a difference in sensitivity. It is worthwhile to compare literature data to assess an optimal dosage of ara-C in pediatric patients.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacokinetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adolescent , Arabinofuranosylcytosine Triphosphate/metabolism , Cell Line , Cell Line, Tumor , Child , Child, Preschool , Chromatography, High Pressure Liquid , Cytarabine/pharmacology , Half-Life , Humans , Infant , Inhibitory Concentration 50 , Lymphocytes/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence
5.
J Neurosci ; 26(20): 5524-33, 2006 May 17.
Article in English | MEDLINE | ID: mdl-16707804

ABSTRACT

Genomic recombination requires cutting, processing, and rejoining of DNA by endonucleases, polymerases, and ligases, among other factors. We have proposed that DNA recombination mechanisms may contribute to long-term memory (LTM) formation in the brain. Our previous studies with the nucleoside analog 1-beta-D-arabinofuranosylcytosine triphosphate (ara-CTP), a known inhibitor of DNA ligases and polymerases, showed that this agent blocked consolidation of conditioned taste aversion without interfering with short-term memory (STM). However, because polymerases and ligases are also essential for DNA replication, it remained unclear whether the effects of this drug on consolidation were attributable to interference with DNA recombination or neurogenesis. Here we show, using C57BL/6 mice, that ara-CTP specifically blocks consolidation but not STM of context fear conditioning, a task previously shown not to require neurogenesis. The effects of a single systemic dose of cytosine arabinoside (ara-C) on LTM were evident as early as 6 h after training. In addition, although ara-C impaired LTM, it did not impair general locomotor activity nor induce brain neurotoxicity. Importantly, hippocampal, but not insular cortex, infusions of ara-C also blocked consolidation of context fear conditioning. Separate studies revealed that context fear conditioning training significantly induced nonhomologous DNA end joining activity indicative of DNA ligase-dependent recombination in hippocampal, but not cortex, protein extracts. Finally, unlike inhibition of protein synthesis, systemic ara-C did not block reconsolidation of context fear conditioning. Our results support the idea that DNA recombination is a process specific to consolidation that is not involved in the postreactivation editing of memories.


Subject(s)
Avoidance Learning/physiology , Conditioning, Psychological/physiology , Fear/physiology , Memory/physiology , Nucleic Acid Synthesis Inhibitors/pharmacology , Recombination, Genetic/physiology , Animals , Arabinofuranosylcytosine Triphosphate/pharmacology , Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , DNA/biosynthesis , DNA/genetics , DNA Ligases/antagonists & inhibitors , DNA Ligases/metabolism , Fear/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Memory/drug effects , Memory Disorders/chemically induced , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Recombination, Genetic/drug effects
6.
Biochem Pharmacol ; 68(1): 11-21, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15183113

ABSTRACT

We have previously reported on the potential use of a novel in vitro human cell-derived model system to investigate the mechanism of action of anticancer agents that directly affect the process of DNA replication. Our cell-free system uses a multiprotein DNA replication complex (designated the DNA synthesome) that has been isolated, characterized, and extensively purified from a wide variety of mammalian cells and tissues. The DNA synthesome is competent to orchestrate simian virus 40 (SV40) origin-specific and large T antigen-dependent DNA replication in vitro. In this study, the synthesome-based cell-free system was tested to evaluate the mechanism of action of 1-beta-d-arabinofuranosylcytosine (ara-C), camptothecin (CPT), and doxorubicin (DOX). Using a novel synthesome-based in vitro kinetic assay, we demonstrated that DNA replication mediated by the synthesome is initiated within the SV40 replication origin and proceeds bidirectionally in a manner analogous to that occurring within the cell. Ara-CTP, CPT, and DOX have been found to affect different stages of the in vitro DNA replication process mediated by the complex. Ara-CTP inhibited both the initiation and elongation stages, whereas CPT produced most of its effects by inhibiting the elongation phase of DNA replication. DOX inhibited the termination stage of DNA synthesis mediated by the synthesome. The data presented here support our contention that the DNA synthesome represents a highly effective in vitro model system for investigating the mechanism by which some anticancer agents can directly affect the process of DNA replication.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Replication/drug effects , DNA-Directed DNA Polymerase/drug effects , Multienzyme Complexes/drug effects , Arabinofuranosylcytosine Triphosphate/pharmacology , Camptothecin/pharmacology , Cytarabine/pharmacology , DNA, Viral/drug effects , DNA, Viral/metabolism , DNA-Directed DNA Polymerase/metabolism , Doxorubicin/pharmacology , Electrophoresis, Agar Gel , Humans , Multienzyme Complexes/metabolism , Simian virus 40/genetics , Simian virus 40/physiology
7.
Learn Mem ; 10(6): 503-9, 2003.
Article in English | MEDLINE | ID: mdl-14657261

ABSTRACT

We examined the hypothesis that processes related to DNA recombination and repair are involved in learning and memory. Rats received intracerebroventricular (i.c.v.) infusions of the antimetabolite 1-beta-D-arabinofuranosylcytosine triphosphate (ara-CTP) or its precursor cytosine arabinoside (ara-C) 30 min prior to conditioned taste aversion (CTA) training. Both ara-CTP and ara-C caused significant impairments in long-term memory (LTM) of CTA. Control experiments indicate that the effect of ara-CTP on CTA memory is related to interference with learning. Furthermore, as it was previously demonstrated for the protein synthesis inhibitor anisomycin, ara-CTP had no effect on CTA memory when it was injected 1 h after training. Importantly, although both ara-CTP and anisomycin significantly blocked LTM in the task, short-term memory (STM) measured 1 h after training was not affected by either of the drugs. Finally, ara-CTP had no effect on in vitro transcription, but it did effectively block nonhomologous DNA end joining (NHEJ) activity of brain protein extracts. We suggest that DNA ligase-mediated DNA recombination and repair processes are necessary for the expression of LTM in the brain.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Avoidance Learning/drug effects , Brain/drug effects , Conditioning, Classical/drug effects , Cytarabine/pharmacology , Memory/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Taste , Animals , Brain/physiology , Injections, Intraventricular , Male , Memory/physiology , Memory, Short-Term/drug effects , Rats , Rats, Long-Evans , Recombination, Genetic , Transcription, Genetic
8.
Int J Cancer ; 107(1): 149-54, 2003 Oct 20.
Article in English | MEDLINE | ID: mdl-12925971

ABSTRACT

Adequate intracellular concentrations of ara-CMP, the monophosphorylated derivative of ara-C, are essential for its cytotoxicity. The critical step for ara-CMP formation is intracellular phosphorylation of ara-C by deoxycytidine kinase (dCK). A common nucleoside resistance mechanism is mutation affecting the expression or the specificity of dCK. We describe the ability of a tert-butyl S-acyl-thioethyl (SATE) derivative of ara-CMP (UA911) to circumvent ara-C resistance in a dCK-deficient human follicular lymphoma cell line (RL-G). The RL-G cell line was produced by continuous exposure to gemcitabine and displayed low dCK mRNA and protein expression that conferred resistance both to ara-C (2,250-fold) and to gemcitabine (2,092-fold). RL-G cells were able to take up the UA911 pronucleotide by diffusion and metabolize it to the corresponding ara-CMP and ara-CTP nucleotides, exhibiting a 199-fold reduction in resistance ratios, and a similar cell cycle arrest to the parental RL-7 cells. Exposures to 10, 50 or 100 microM concentrations of UA911 produced 160 +/- 7, 269 +/- 8 and 318 +/- 62 pmol ara-CTP/mg protein in RL-7 cells, and 100 +/- 12, 168 +/- 10 and 217 +/- 39 pmol ara-CTP/mg protein in RL-G cells, respectively. Exposure of RL-G cells to underivatized, radiolabeled ara-C produced no detectable amounts of the active triphosphate metabolites. We conclude that the UA911 pronucleotide is capable of overcoming dCK-mediated resistance. This result can be attributed to the unique cellular metabolism of the SATE pronucleotides giving rise to the intracellular delivery of ara-CMP to dCK-deficient cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , Deoxycytidine Kinase/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Lymphoma, Follicular/drug therapy , Prodrugs/pharmacology , Antimetabolites, Antineoplastic/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacology , Cell Division/drug effects , Chromatography, High Pressure Liquid , Deoxycytidine/pharmacology , Humans , Lymphoma, Follicular/metabolism , Lymphoma, Follicular/pathology , Nucleic Acid Synthesis Inhibitors/pharmacology , Tumor Cells, Cultured/drug effects , Gemcitabine
9.
Hepatology ; 36(3): 710-22, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198665

ABSTRACT

To design combination strategies for chronic hepatitis B therapy, we evaluated in vitro the inhibitory activity of 4 nucleoside analogs, (-)FTC, L-FMAU, DXG, and DAPD, in comparison with lamivudine (3TC) and PMEA. In a cell-free assay for the expression of wild-type duck hepatitis B virus (DHBV) reverse transcriptase, DAPD-TP was found to be the most active on viral minus strand DNA synthesis, including the priming reaction, followed by 3TC-TP, (-)FTC-TP, and DXG-TP, whereas L-FMAU-TP was a weak inhibitor. In cell culture experiments, important differences in drug concentration allowing a 50% inhibition of viral replication or polymerase activity (IC50s) were observed depending on the cell type used, showing that antiviral effect of nucleoside analogs may depend on their intracellular metabolism. IC50s obtained for wild-type DHBV replication in primary duck hepatocytes were much lower than with DHBV transfected LMH cells. IC50s were also significantly lower in the 2.2.1.5 and HepG2 cells compared with HBV transfected HuH7 cells. Moreover, L-FMAU inhibited preferentially HBV plus strand DNA synthesis in these cell lines. The antiviral effect of these inhibitors was also evaluated against 3TC-resistant mutants of the DHBV and HBV polymerases. These mutants were found to be cross resistant to (-)FTC. By contrast, the double DHBV polymerase mutant was sensitive to DXG-TP and DAPD-TP. Moreover, both purine analogs remained active against DHBV and HBV 3TC-resistant mutants in transfected LMH and HepG2 cells, respectively. In conclusion, the unique mechanism of action of these new inhibitors warrants further evaluation in experimental models to determine their capacity to delay or prevent the selection of drug resistant mutants.


Subject(s)
Arabinofuranosylcytosine Triphosphate/analogs & derivatives , Cytidine Triphosphate/analogs & derivatives , Dioxolanes/pharmacology , Guanosine/analogs & derivatives , Hepadnaviridae Infections/drug therapy , Hepatitis B Virus, Duck/genetics , Hepatitis, Viral, Animal/drug therapy , Purine Nucleosides/pharmacology , Thiophenes , Animals , Arabinofuranosylcytosine Triphosphate/chemistry , Arabinofuranosylcytosine Triphosphate/pharmacology , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/chemistry , Arabinofuranosyluracil/pharmacology , Carcinoma, Hepatocellular , Cytidine Triphosphate/chemistry , Cytidine Triphosphate/pharmacology , DNA Replication/drug effects , DNA, Viral/genetics , DNA-Directed DNA Polymerase/metabolism , Dioxolanes/chemistry , Drug Resistance, Viral , Gene Expression Regulation, Viral/drug effects , Guanosine/chemistry , Guanosine/pharmacology , Hepatitis B virus , Hepatocytes/cytology , Hepatocytes/virology , Humans , Lamivudine/pharmacology , Liver Neoplasms , Mutation , Purine Nucleosides/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Tumor Cells, Cultured
10.
Biochem Pharmacol ; 64(3): 441-9, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12147295

ABSTRACT

We have investigated whether nucleoside drugs that induce or protect neurones against apoptosis might directly activate or inhibit mitochondrial permeability transition (mPT) since opening of the mPT pore can promote release of cytochrome c and apoptosis, while its closure can prevent these changes. We found that the pro-apoptotic pyrimidine analogues cytosine beta-D-arabinofuranoside and cytosine beta-D-arabinofuranoside 5'-triphosphate, which activated apoptosis in post-mitotic neurones without incorporation into nuclear DNA, induced rapid calcium-dependent mitochondrial swelling of isolated liver mitochondria in a dose-dependent manner. Induction of up to 50 and 80%, respectively, of maximal swelling induced by high calcium was obtained at 1mM concentrations, which also promoted a 17-fold increase in the release of cytochrome c. Both activities were inhibited by cyclosporine A to unstimulated levels; dCTP had no effect. In contrast, the anti-apoptotic adenine analogues, 3-methyladenine (3-MA) and olomoucine (but not iso-olomoucine), inhibited swelling induced by calcium or phenylarsine oxide in a dose-dependent manner at concentrations that protect neurones from apoptosis. Both compounds also inhibited the release of cytochrome c (by 82%, 20 mM 3-MA and 95%, 0.9 mM olomoucine), similar to the inhibition obtained with cyclosporine A, and 5mM ADP or ATP. Similar inhibitory effects with olomoucine and 3-MA were found in isolated heart mitochondria. These studies identify the mPT as an important target for hitherto untested pro- and anti-apoptotic nucleoside-based drugs and suggest that screening for mPT modulation is an important component in the validation of a drug's mechanism of action.


Subject(s)
Adenine/analogs & derivatives , Cytochrome c Group/metabolism , Ion Channels/metabolism , Mitochondria, Liver/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Adenine/pharmacology , Animals , Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacology , Drug Interactions , Enzyme Inhibitors/pharmacology , Ion Channels/antagonists & inhibitors , Kinetin , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Purines/pharmacology , Rats , Rats, Wistar
11.
Biochem Pharmacol ; 60(3): 403-11, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10856436

ABSTRACT

The antimetabolite 1-beta-D-arabinofuranosylcytosine (ara-C) has been used as a highly effective agent for the treatment of leukemia. The active metabolite 1-beta-D-arabinofuranosylcytosine triphosphate (ara-CTP) is a potent inhibitor of DNA polymerases alpha, delta, and epsilon, and is responsible for inhibiting intact cell DNA synthesis. We have shown that a multiprotein complex, exhibiting many of the properties expected of the human cell DNA replication apparatus, can be readily isolated from human cells and tissues and is capable of supporting origin-dependent DNA synthesis in vitro. DNA polymerases alpha, delta, and epsilon are components of this multiprotein complex, termed the DNA synthesome, and we report here that the activities of these DNA synthesome-associated DNA polymerases are inhibited differentially by ara-CTP. Inhibition of the DNA synthesome-associated DNA polymerase alpha increased in a concentration-dependent manner, and was correlated closely with the inhibition of simian virus 40 (SV40) origin-dependent in vitro DNA replication, whereas DNA synthesome-associated DNA polymerase delta activity was not inhibited significantly by ara-CTP at 100 microM. Recent work has shown that the synthesome-associated DNA polymerase epsilon does not function in in vitro SV40 DNA replication, suggesting that only polymerases alpha and delta drive the DNA replication fork. Therefore, our results suggest that inhibition of the activity of the mammalian cell DNA synthesome by ara-CTP is due primarily to the inhibition of the DNA synthesome-associated DNA polymerase alpha. This observation implies that the drug may target specific phases of the DNA synthetic process in human cells.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , DNA Polymerase III/antagonists & inhibitors , DNA Polymerase I/antagonists & inhibitors , DNA Replication/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Cell Line , DNA/biosynthesis , DNA/drug effects , DNA Polymerase I/metabolism , DNA Polymerase III/metabolism , HeLa Cells , Humans , Simian virus 40/physiology
12.
J Viral Hepat ; 7(2): 161-5, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10760047

ABSTRACT

Several nucleoside analogues (penciclovir, lobucavir, dioxalane guanine [DXG], 1-beta-2,6-diaminopurine dioxalane [DAPD], L-FMAU, lamivudine) and acyclic nucleoside phosphonate analogues (adefovir, tenofovir) that are in clinical use, in clinical trials or under preclinical development for the treatment of hepatitis B virus (HBV) infections, were evaluated for their inhibitory effect on the replication of a la- mivudine-resistant HBV variant containing the methionine --> valine substitution (M550V) in the polymerase nucleoside-binding domain. The antiviral activity was determined in the tetracycline-responsive HepAD38 and HepAD79 cells, which are stably transfected with either a cDNA copy of the wild-type pregenomic RNA or with cDNA containing the M550V mutation. As expected, lamivudine was much less ( approximately 200-fold) effective at inhibiting replication of the M550V mutant virus than the wild-type virus. In contrast, adefovir, tenofovir, lobucavir, L-FMAU, DXG and DAPD proved almost equally effective against both viruses. A second objective of this study was to directly compare the antiviral potency of the anti-HBV agents in HepG2 2.2.15 cells (which are routinely used for anti-HBV drug-screening purposes) with that in HepAD38 cells. HepAD38 cells produce much larger quantities of HBV than HepG2 2.2.15 cells, and thus allow drug screening in a multiwell plate format. All compounds were found to be almost equally effective at inhibiting HBV replication in HepAD38 cells (as in HepG2 2.2.15 cells), except for penciclovir, which was clearly less effective in HepAD38 cells.


Subject(s)
Acyclovir/analogs & derivatives , Adenine/analogs & derivatives , Antiviral Agents/pharmacology , Arabinofuranosylcytosine Triphosphate/analogs & derivatives , DNA-Directed DNA Polymerase/genetics , Dioxolanes/pharmacology , Guanine/analogs & derivatives , Hepatitis B virus/physiology , Organophosphonates , Organophosphorus Compounds/pharmacology , Purine Nucleosides/pharmacology , Virus Replication/drug effects , Acyclovir/pharmacology , Adenine/pharmacology , Arabinofuranosylcytosine Triphosphate/pharmacology , Cell Line , DNA, Viral/chemistry , Guanine/pharmacology , Hepatitis B virus/enzymology , Hepatitis B virus/genetics , Humans , Tenofovir
13.
Curr Opin Hematol ; 7(3): 197-202, 2000 May.
Article in English | MEDLINE | ID: mdl-10786659

ABSTRACT

During the past decade, investigators have evaluated the role of hematopoietic growth factors as cytokines that could potentiate the cytotoxic action of certain chemotherapeutic agents when administered simultaneously with induction therapy. Such cytokines included granulocyte-macrophage colony-stimulating factor, granulocyte colony-stimulating factor, interleukin 3, and stem cell factor. Phase I and II studies have confirmed a significant biologic activity of such priming therapy, and the aggregate of subsequent controlled studies have also established the safety of such manipulations. However, despite the plethora of clinical trials, data regarding clinical efficacy remain uncertain. It is fair to say that little progress in our clinical understanding has occurred during the past 24 months and at the present time, the concurrent use of growth factors with chemotherapy, as a method of enhancing the antileukemic action of cytotoxic therapy, remains very investigational.


Subject(s)
Antineoplastic Agents/therapeutic use , Growth Substances/therapeutic use , Leukemia/drug therapy , Acute Disease , Antineoplastic Agents/pharmacology , Antineoplastic Agents/standards , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacology , Clinical Trials as Topic , Colony-Stimulating Factors/pharmacology , Colony-Stimulating Factors/standards , Colony-Stimulating Factors/therapeutic use , Growth Substances/pharmacology , Growth Substances/standards , Humans , Leukemia/blood
14.
Cancer Chemother Pharmacol ; 45(4): 312-9, 2000.
Article in English | MEDLINE | ID: mdl-10755320

ABSTRACT

PURPOSE: An intact and fully functional multiprotein DNA replication complex (DNA synthesome) from human as well as from murine mammary carcinoma cells was first isolated and characterized in our laboratory. The human cell synthesome supports the in vitro origin-specific simian virus 40 (SV40) DNA replication reaction in the presence of the viral large T-antigen using a semiconservative mechanism and has been shown to contain all the proteins and enzymes required to support DNA synthesis. We are currently using the DNA synthesome as a unique model for analyzing the mechanism of action of anticancer drugs affecting DNA replication. The purpose of this study was to further investigate the mechanism of action of ara-C using the DNA synthesome isolated from the human breast cancer cell line MDA MB-468. METHODS: Synthesome-mediated SV40 DNA replication was performed in the presence of various concentrations of ara-CTP (the active metabolite of ara-C) and the types of daughter DNA molecules produced were analyzed lusing neutral and alkaline gel electrophoresis. We also examined the effect of ara-C on intact MDA MB-468 cell DNA synthesis and on cell proliferation. In addition, we studied the effect of ara-CTP on the activity of some of the synthesome target proteins (the DNA polymerases alpha and delta). RESULTS: Full-length daughter DNA molecules were obtained in the presence of low concentrations of ara-CTP while at higher concentrations, there was an inhibition of full-length daughter DNA synthesis. The findings suggest that specifically the initiation phase of DNA synthesis was inhibited by ara-CTP since the production of the short Okazaki fragments was suppressed at all concentrations of the drug above 10 microM. In addition, it was found that the IC50 of ara-CTP for inhibition of synthesome-mediated in vitro DNA replication was comparable to that required to inhibit intact cell DNA synthesis. Further experimentation has shown that ara-CTP preferentially inhibits the activity of the synthesome-associated DNA polymerase alpha enzyme while the DNA polymerase delta seems to be resistant to the inhibitory effect of that drug. CONCLUSIONS: Our results indicate that ara-C's action on DNA replication is mediated primarily through DNA polymerase alpha and suggest that this enzyme plays a key role in DNA synthetic initiation events. The results also provide definitive support for the use of the DNA synthesome as a unique and powerful model for analyzing the mechanism of action of anticancer drugs which directly affect DNA replication.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , Antigens, Polyomavirus Transforming/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacology , Breast Neoplasms/metabolism , Cell Division/drug effects , DNA Polymerase I/biosynthesis , DNA Polymerase III/biosynthesis , Humans , Replicon/drug effects , Tumor Cells, Cultured
15.
Cancer Chemother Pharmacol ; 45(4): 320-8, 2000.
Article in English | MEDLINE | ID: mdl-10755321

ABSTRACT

PURPOSE: Gemcitabine (dFdC) and cytarabine (araC) are both analogs of deoxycytidine. Gemcitabine is a relatively new drug that has been shown in both clinical trials and in vitro systems to have more potent antitumor activity than araC. We have previously isolated a fully functional multiprotein DNA replication complex from human cells and termed it the DNA synthesome. Using the DNA synthesome, we have successfully examined the mechanism of action of several anticancer drugs that directly affect DNA synthesis. In this study, we compared the effects of dFdC and araC on in vitro DNA synthesis mediated by the DNA synthesome with the effects of these drugs on intact MCF7 cell DNA synthesis. METHODS: We examined the effects of dFdC and araC on intact MCF7 cell DNA synthesis and clonogenicity. We also performed in vitro SV40 replication assays mediated by the MCF7 cell-derived DNA synthesome in presence of dFdCTP and araCTP. The types of daughter molecules produced in the assay were analyzed by neutral and alkaline agarose gel electrophoresis. Finally, we examined the effects ofdFdCTP and araCTP on the synthesome-associated DNA polymerase alpha and delta activities. RESULTS: Our results showed that dFdC was more potent than araC at inhibiting intact MCF7 cell DNA synthesis and clonogenicity. [3H]Thymidine incorporation was inhibited by 50% at a dFdC concentration of 10 microM, which was about tenfold lower than the concentration of araC required to inhibit intact cell DNA synthesis by the same amount. As examined by clonogenicity assay, dFdC was also significantly more cytotoxic than araC after a 24-h incubation. In vitro SV40 replication assays using the DNA synthesome derived from MCF7 cells demonstrated that the formation of full-length DNA along with replication intermediates were inhibited by dFdCTP in a concentration-dependent manner. Full-length DNA was produced in the in vitro DNA replication assay even when the dFdCTP was incubated in the assay at concentrations of up to 1 mM. We observed that in the presence of 10 microM dCTP, 3 microM dFdCTP and 60 microM araCTP were required to inhibit in vitro SV40 DNA synthesis by 50%. Although dFdCTP is more potent than araCTP at inhibiting in vitro SV40 DNA synthesis, there was no significant difference between the inhibitory effect of these two drugs on the activity of the MCF7 synthesome-associated DNA polymerases alpha and delta. It was found that the drug concentrations required to inhibit 50% of the synthesome-associated DNA polymerase delta activity were much higher than those required to inhibit 50% of DNA polymerase alpha activity for both dFdCTP and araCTP. CONCLUSION: Taken together, our results demonstrated that: (1) dFdC is a more potent inhibitor of intact cell DNA synthesis and in vitro SV40 DNA replication than araC; (2) the decrease in the synthetic activity of synthesome-mediated in vitro SV40 origin-dependent DNA synthesis by dFdCTP and araCTP correlates with the inhibition of DNA polymerase alpha activity; and (3) the MCF7 cell DNA synthesome can serve as a unique and relevant model to study the mechanism of action of anticancer drugs that directly affect DNA synthesis.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Cytarabine/pharmacology , DNA, Neoplasm/biosynthesis , Deoxycytidine/analogs & derivatives , Antigens, Polyomavirus Transforming/biosynthesis , Antigens, Polyomavirus Transforming/genetics , Arabinofuranosylcytosine Triphosphate/pharmacology , Breast Neoplasms/genetics , Cloning, Molecular , DNA Polymerase I/metabolism , DNA Polymerase III/metabolism , Deoxycytidine/pharmacology , Humans , Replicon/drug effects , Replicon/genetics , Tumor Cells, Cultured , Gemcitabine
16.
Biochem Pharmacol ; 55(8): 1181-7, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9719472

ABSTRACT

1-(2'-Deoxy-2'-fluoro-beta-L-arabinofuranosyl)-5-methyluracil (L-FMAU) was shown to have potent antiviral activity against Epstein-Barr virus (EBV) without any cellular toxicity at concentrations up to 200 microM (Yao et al., Biochem Pharmacol 51: 941-947, 1996). The 5'-triphosphate of L-FMAU was not a substrate for EBV or cellular DNA polymerases, but could inhibit the elongation reaction, 3'-to-5' exonuclease activity, and nucleotide turnover catalyzed by EBV DNA polymerase. DNA synthesis catalyzed by human DNA polymerases was inhibited to a lesser extent. The inhibition pattern of EBV DNA polymerase by L-FMAU-5'-triphosphate (L-FMAU-TP) was consistent with an uncompetitive mechanism when dNTP or template-primer were used as the variable substrates. The Ki values were 38+/-10 microM for the elongation reaction, and about 50+/-10 microM for both nucleotide exchange and 3'-to-5' exonuclease reactions, values that were 10-20 times less than that for GMP. L-FMAU-TP is the first nucleoside 5'-triphosphate shown to have such unique behavior toward DNA polymerases. EBV DNA polymerase could be one of the targets for the inhibitory effect of L-FMAU-TP on EBV replication.


Subject(s)
Antiviral Agents/pharmacology , Arabinofuranosylcytosine Triphosphate/analogs & derivatives , DNA-Binding Proteins , Herpesvirus 4, Human/drug effects , Nucleic Acid Synthesis Inhibitors , Viral Proteins , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacology , Cell Line , DNA Replication/drug effects , DNA-Directed DNA Polymerase , Exodeoxyribonuclease V , Exodeoxyribonucleases/antagonists & inhibitors , Exonucleases/antagonists & inhibitors , Humans , Substrate Specificity , Tumor Cells, Cultured
17.
J Cell Biochem ; 61(3): 444-51, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8761948

ABSTRACT

DNA replication machinery is an important target for chemotherapeutic drugs. We have used an in vitro system to study the effect of drugs on mammalian DNA replication, either by direct interaction with the DNA structure or with replication proteins and machinery. The anthracycline doxorubicin (Dox) showed a dose-dependent inhibitory effect on DNA replication, whether incubated with HeLa cell extracts or with DNA and nucleotides. Earliest-labeled fragment analysis revealed that inhibition of replication began within the origin-containing fragment in both control and Dox-containing reactions in vitro. AraC, a nucleoside analog, had no significant effect on DNA synthesis. In contrast, araCTP was able to inhibit DNA replication in vitro. Since metabolism is diminished in this in vitro system, the degree of phosphorylation of araC was apparently low. Progesterone showed an increase in nucleotide incorporation (sensitive to BuPdGTP inhibition of replication-specific polymerases alpha and delta) after preincubation with HeLa cell extracts, although progesterone receptors were not detectable in the HeLa cell extracts. In addition, we observed an inhibition in DNA replication when progesterone was preincubated with DNA and nucleotides. These results suggest that progesterone may have a mechanism of action that is different from any known to be mediated through progesterone receptors. In conclusion, these results indicate that this mammalian in vitro replication system will be useful for the study of mechanisms and design of therapeutic drugs that inhibit mammalian DNA replication.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Arabinofuranosylcytosine Triphosphate/pharmacology , Cytarabine/pharmacology , DNA Replication/drug effects , Doxorubicin/pharmacology , HeLa Cells , Humans , Phosphorylation , Plasmids , Progesterone/metabolism , Receptors, Progesterone/metabolism
18.
Pharmazie ; 50(3): 213-5, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7732054

ABSTRACT

Two DNA polymerase activities from Tetrahymena pyriformis were studied. The cytoplasmic polymerase was inhibited by cytosine arabinoside triphosphate (ara-CTP) and N-ethylmaleiimide (NEM) whereas the nuclear enzyme was not inhibited by araCTP and was inhibited by NEM to a lesser extent. The two enzymes could use CsCl, NaCl, and LiCl instead of KCl to varying degrees in the polymerase reaction. The two activities showed optimum activity at different concentrations of NaCl and KCl.


Subject(s)
Arabinofuranosylcytosine Triphosphate/pharmacology , Ethylmaleimide/pharmacology , Nucleic Acid Synthesis Inhibitors , Tetrahymena pyriformis/enzymology , Animals , Cytoplasm/drug effects , Cytoplasm/enzymology , Metals/pharmacology , Tetrahymena pyriformis/drug effects
19.
Bull Cancer ; 82 Suppl 1: 37s-41s, 1995.
Article in French | MEDLINE | ID: mdl-7626853

ABSTRACT

There are two distinct levels where dose intensity can be increased by pharmacomodulation. The first level implies the inhibition of clearly identified resistance mechanisms. Among them, MDR type resistance is the most studied currently. MDR resistance is mediated by GP170, a cell membrane protein, which can be inhibited by several pharmacological agents like verapamil, ciclosporine and S 9788 which are currently being clinically investigated. The inhibition of DNA repair mechanisms is another approach and a representative example is the inhibition of O6-alkylguanine DNA alkyltransferase by O6-methylguanine or O6-benzylguanine. Modulation of detoxification pathways has also been considered, with for instance, the use of buthionine sulfoximide (BSO) to deplete intracellular glutathione levels. The second level for pharmacomodulation concerns pharmacological interferences with metabolic pathways controlling the activity of anticancer agents and particularly the antimetabolites like ara-C and 5-FU. The use of hydroxyurea or PALA in the case of 5-FU are good illustrations of pharmacomodulation via specific metabolic routes. Finally, the 5-FU-folinic acid combination could characterise a third level of pharmacomodulation where cytotoxic activity is optimised at the site of interaction between activated drug (5FdUMP) and target (thymidilate synthase).


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/pharmacology , Antineoplastic Agents/metabolism , Arabinofuranosylcytosine Triphosphate/pharmacology , DNA Repair , DNA, Neoplasm/drug effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Multiple , Fluorouracil/pharmacology , Leucovorin/pharmacology
20.
Biochim Biophys Acta ; 1227(1-2): 92-100, 1994 Oct 21.
Article in English | MEDLINE | ID: mdl-7918688

ABSTRACT

The mammalian DNA polymerase inhibitors aphidicolin and 1-beta-D-arabinofuranosylcytosine (araC), when used in combination, inhibit the repair of DNA damage induced by gamma rays or 4-nitroquinoline 1-oxide in normal human fibroblasts to an extent 2- to 4-fold greater than that seen with each inhibitor alone. Thus either aphidicolin modulates the rate of intracellular accumulation of araC 5'-triphosphate (araCTP), the presumed rate-limiting step in the genotoxic action of araC, or aphidicolin and araC inhibit repair by different mechanisms. To explore these possibilities, we compared the effects of aphidicolin, araC, araCTP, and 2',3'-dideoxythymidine triphosphate (ddTTP) on repair of DNA damage induced by 60Co gamma radiation in intact versus permeable human fibroblasts. Both aphidicolin and araC strongly inhibited repair in permeable cells, as indicated by the accumulation of DNA strand breaks in irradiated cultures that were subsequently treated with saponin (25 micrograms/ml; 10 min) and incubated for 2 h with either chemical. The extent of repair inhibition by each drug was comparable in intact and permeable cells, amounting to approximately 1.1 sites/10(8) daltons/2 h upon exposure to 150 Gy. The active metabolite of araC, araCTP, did not inhibit repair in intact cells, but did so in permeable cells to an extent within the range of that seen with araC or aphidicolin alone. The incidence of DNA strand breaks accumulating in gamma-irradiated permeable cultures as a result of incubation with araCTP plus aphidicolin, or araC plus aphidicolin, was approximately 2-fold greater than that arising in parallel cultures which had been incubated with optimal concentrations of each of the three drugs alone. Although the resolution of our assays compelled us to monitor repair events in moribund cell populations, we have reason to be confident that within the short post-irradiation period considered here, the observed drug-accumulated breaks truly represent functional repair inhibition and not merely abortive pathological responses. We thus conclude that (1) the accumulation of araCTP in intact cells is not limiting the ability of araC to inhibit DNA repair; and (2) the mode of the inhibitory action of araC/araCTP on gamma ray repair is different from that of aphidicolin. In contrast to the observations with these chemicals, ddTTP (20 microM), a potent inhibitor of DNA polymerase beta, did not produce any measurable effect on DNA repair in gamma-irradiated permeable fibroblasts, nor did it enhance the efficacy of araC, araCTP or aphidicolin to inhibit repair. These results strongly suggest that DNA polymerase beta plays no significant role in the repair of gamma radioproducts in human fibroblasts.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aphidicolin/pharmacology , Cytarabine/pharmacology , DNA Repair/drug effects , Nucleic Acid Synthesis Inhibitors , Arabinofuranosylcytosine Triphosphate/pharmacology , Cells, Cultured/drug effects , Child , Drug Combinations , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Saponins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...