Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
2.
Nucleic Acids Res ; 50(1): 244-258, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34904670

ABSTRACT

Loss of genome stability leads to reduced fitness, fertility and a high mutation rate. Therefore, the genome is guarded by the pathways monitoring its integrity and neutralizing DNA lesions. To analyze the mechanism of DNA damage induction by cytidine analog zebularine, we performed a forward-directed suppressor genetic screen in the background of Arabidopsis thaliana zebularine-hypersensitive structural maintenance of chromosomes 6b (smc6b) mutant. We show that smc6b hypersensitivity was suppressed by the mutations in EQUILIBRATIVE NUCLEOSIDE TRANSPORTER 3 (ENT3), DNA METHYLTRANSFERASE 1 (MET1) and DECREASE IN DNA METHYLATION 1 (DDM1). Superior resistance of ent3 plants to zebularine indicated that ENT3 is likely necessary for the import of the drug to the cells. Identification of MET1 and DDM1 suggested that zebularine induces DNA damage by interference with the maintenance of CG DNA methylation. The same holds for structurally similar compounds 5-azacytidine and 2-deoxy-5-azacytidine. Based on our genetic and biochemical data, we propose that zebularine induces enzymatic DNA-protein crosslinks (DPCs) of MET1 and zebularine-containing DNA in Arabidopsis, which was confirmed by native chromatin immunoprecipitation experiments. Moreover, zebularine-induced DPCs accumulate preferentially in 45S rDNA chromocenters in a DDM1-dependent manner. These findings open a new avenue for studying genome stability and DPC repair in plants.


Subject(s)
Cytidine/analogs & derivatives , Heterochromatin/metabolism , Mutagens/toxicity , RNA, Ribosomal/genetics , Arabidopsis , Arabidopsis Proteins/genetics , Cell Cycle Proteins/genetics , Cytidine/toxicity , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Drug Resistance , Heterochromatin/drug effects , Membrane Transport Proteins/genetics , Mutation , RNA, Ribosomal/drug effects , Transcription Factors/genetics
3.
Nucleic Acids Res ; 49(18): 10431-10447, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34551439

ABSTRACT

Transposable elements (TEs) have long been known to be major contributors to plant evolution, adaptation and crop domestication. Stress-induced TE mobilization is of particular interest because it may result in novel gene regulatory pathways responding to stresses and thereby contribute to stress adaptation. Here, we investigated the genomic impacts of stress induced TE mobilization in wild type Arabidopsis plants. We find that the heat-stress responsive ONSEN TE displays an insertion site preference that is associated with specific chromatin states, especially those rich in H2A.Z histone variant and H3K27me3 histone mark. In order to better understand how novel ONSEN insertions affect the plant's response to heat stress, we carried out an in-depth transcriptomic analysis. We find that in addition to simple gene knockouts, ONSEN can produce a plethora of gene expression changes such as: constitutive activation of gene expression, alternative splicing, acquisition of heat-responsiveness, exonisation and genesis of novel non-coding and antisense RNAs. This report shows how the mobilization of a single TE-family can lead to a rapid rise of its copy number increasing the host's genome size and contribute to a broad range of transcriptomic novelty on which natural selection can then act.


Subject(s)
Arabidopsis/genetics , Heat-Shock Response/genetics , Retroelements , Transcriptome , Arabidopsis/drug effects , Arabidopsis/metabolism , Arabidopsis Proteins/analysis , Cytidine/analogs & derivatives , Cytidine/toxicity , Epigenesis, Genetic , Exons , Gene Expression Regulation, Plant , Genome, Plant , Histone Code , Histones/analysis , Phenotype
4.
Eur J Med Chem ; 215: 113212, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33582576

ABSTRACT

The emergence of drug-resistant strains of pathogenic microorganisms necessitates the creation of new drugs. In order to find new compounds that effectively inhibit the growth of pathogenic bacteria and fungi, we synthesized a set of N4-derivatives of cytidine, 2'-deoxycytidine and 5-metyl-2'-deoxycytidine bearing extended N4-alkyl and N4-phenylalkyl groups. The derivatives demonstrate activity against a number of Gram-positive bacteria, including Mycobacterium smegmatis (MIC = 24-200 µM) and Staphylococcus aureus (MIC = 50-200 µM), comparable with the activities of some antibiotics in medical use. The most promising compound appeared to be N4-dodecyl-5-metyl-2'-deoxycytidine 4h with activities of 24 and 48 µM against M. smegmatis and S. aureus, respectively, and high inhibitory activity of 0.5 mM against filamentous fungi that can, among other things, damage works of art, such as tempera painting. Noteworthy, some of other synthesized compounds are active against fungal growth with the inhibitory concentration in the range of 0.5-3 mM.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Cytidine/analogs & derivatives , Cytidine/pharmacology , A549 Cells , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/toxicity , Antifungal Agents/chemical synthesis , Antifungal Agents/toxicity , Bacteria/drug effects , Cytidine/toxicity , Drug Discovery , Fungi/drug effects , Humans , Microbial Sensitivity Tests
5.
DNA Repair (Amst) ; 57: 116-124, 2017 09.
Article in English | MEDLINE | ID: mdl-28732309

ABSTRACT

Zebularine is a second-generation, highly stable hydrophilic inhibitor of DNA methylation with oral bioavailability that preferentially target cancer cells. It acts primarily as a trap for DNA methyl transferases (DNMTs) protein by forming covalent complexes between DNMT protein and zebularine-substrate DNA. It's well documented that replication-blocking DNA lesions can cause replication fork collapse and thereby to the formation of DNA double-strand breaks (DSB). DSB are dangerous lesions that can lead to potentially oncogenic genomic rearrangements or cell death. The two major pathways for repair of DSB are non-homologous end joining (NHEJ) and homologous recombination (HR). Recently, multiple functions for the HR machinery have been identified at arrested forks. Here we investigate in more detail the importance of the lesions induced by zebularine in terms of DNA damage and cytotoxicity as well as the role of HR in the repair of these lesions. When we examined the contribution of NHEJ and HR in the repair of DSB induced by zebularine we found that these breaks were preferentially repaired by HR. Also we show that the production of DSB is dependent on active replication. To test this, we determined chromosome damage by zebularine while transiently inhibiting DNA synthesis. Here we report that cells deficient in single-strand break (SSB) repair are hypersensitive to zebularine. We have observed more DSB induced by zebularine in XRCC1 deficient cells, likely to be the result of conversion of SSB into toxic DSB when encountered by a replication fork. Furthermore we demonstrate that HR is required for the repair of these breaks. Overall, our data suggest that zebularine induces replication-dependent DSB which are preferentially repaired by HR.


Subject(s)
Cytidine/analogs & derivatives , DNA Breaks, Double-Stranded , DNA Replication/drug effects , Recombinational DNA Repair , Animals , Cell Line , Cricetulus/genetics , Cricetulus/metabolism , Cytidine/pharmacology , Cytidine/toxicity , DNA/drug effects , DNA/metabolism , DNA Breaks, Single-Stranded , DNA Modification Methylases/antagonists & inhibitors , DNA Repair , Female
6.
Antiviral Res ; 143: 151-161, 2017 07.
Article in English | MEDLINE | ID: mdl-28412183

ABSTRACT

Recent cases of severe toxicity during clinical trials have been associated with antiviral ribonucleoside analogs (e.g. INX-08189 and balapiravir). Some have hypothesized that the active metabolites of toxic ribonucleoside analogs, the triphosphate forms, inadvertently target human mitochondrial RNA polymerase (POLRMT), thus inhibiting mitochondrial RNA transcription and protein synthesis. Others have proposed that the prodrug moiety released from the ribonucleoside analogs might instead cause toxicity. Here, we report the mitochondrial effects of several clinically relevant and structurally diverse ribonucleoside analogs including NITD-008, T-705 (favipiravir), R1479 (parent nucleoside of balapiravir), PSI-7851 (sofosbuvir), and INX-08189 (BMS-986094). We found that efficient substrates and chain terminators of POLRMT, such as the nucleoside triphosphate forms of R1479, NITD-008, and INX-08189, are likely to cause mitochondrial toxicity in cells, while weaker chain terminators and inhibitors of POLRMT such as T-705 ribonucleoside triphosphate do not elicit strong in vitro mitochondrial effects. Within a fixed 3'-deoxy or 2'-C-methyl ribose scaffold, changing the base moiety of nucleotides did not strongly affect their inhibition constant (Ki) against POLRMT. By swapping the nucleoside and prodrug moieties of PSI-7851 and INX-08189, we demonstrated that the cell-based toxicity of INX-08189 is mainly caused by the nucleoside component of the molecule. Taken together, these results show that diverse 2' or 4' mono-substituted ribonucleoside scaffolds cause mitochondrial toxicity. Given the unpredictable structure-activity relationship of this ribonucleoside liability, we propose a rapid and systematic in vitro screen combining cell-based and biochemical assays to identify the early potential for mitochondrial toxicity.


Subject(s)
Antiviral Agents/toxicity , Mitochondria/drug effects , Ribonucleosides/chemistry , Ribonucleosides/toxicity , Adenosine/analogs & derivatives , Amides/toxicity , Cell Line/drug effects , Cytidine/analogs & derivatives , Cytidine/toxicity , DNA-Directed RNA Polymerases/drug effects , Guanosine Monophosphate/analogs & derivatives , Guanosine Monophosphate/toxicity , Humans , Inhibitory Concentration 50 , Mitochondrial Proteins/metabolism , Nucleosides/toxicity , Prodrugs/pharmacology , Protein Biosynthesis/drug effects , Pyrazines/toxicity , RNA/metabolism , RNA, Mitochondrial , Sofosbuvir/toxicity , Structure-Activity Relationship , Transcription Initiation Site/drug effects , Transcription, Genetic/drug effects
7.
Proc Natl Acad Sci U S A ; 113(48): 13774-13779, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27849610

ABSTRACT

Routine dietary consumption of foods that contain aflatoxins is the second leading cause of environmental carcinogenesis worldwide. Aflatoxin-driven mutagenesis is initiated through metabolic activation of aflatoxin B1 (AFB1) to its epoxide form that reacts with N7 guanine in DNA. The resulting AFB1-N7-dG adduct undergoes either spontaneous depurination or imidazole-ring opening yielding formamidopyrimidine AFB1 (AFB1-Fapy-dG). Because this latter adduct is known to persist in human tissues and contributes to the high frequency G-to-T mutation signature associated with many hepatocellular carcinomas, we sought to establish the identity of the polymerase(s) involved in processing this lesion. Although our previous biochemical analyses demonstrated the ability of polymerase ζ (pol ζ) to incorporate an A opposite AFB1-Fapy-dG and extend from this mismatch, biological evidence supporting a unique role for this polymerase in cellular tolerance following aflatoxin exposure has not been established. Following challenge with AFB1, survival of mouse cells deficient in pol ζ (Rev3L-/-) was significantly reduced relative to Rev3L+/- cells or Rev3L-/- cells complemented through expression of the wild-type human REV3L. Furthermore, cell-cycle progression of Rev3L-/- mouse embryo fibroblasts was arrested in late S/G2 following AFB1 exposure. These Rev3L-/- cells showed an increase in replication-dependent formation of γ-H2AX foci, micronuclei, and chromosomal aberrations (chromatid breaks and radials) relative to Rev3L+/- cells. These data suggest that pol ζ is essential for processing AFB1-induced DNA adducts and that, in its absence, cells do not have an efficient backup polymerase or a repair/tolerance mechanism facilitating survival.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Liver Neoplasms/genetics , Aflatoxin B1/analogs & derivatives , Aflatoxin B1/genetics , Aflatoxin B1/toxicity , Aflatoxins/toxicity , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Chromosome Aberrations/drug effects , Cytidine/analogs & derivatives , Cytidine/genetics , Cytidine/toxicity , DNA Adducts/drug effects , DNA Adducts/genetics , DNA Damage/drug effects , DNA Repair/genetics , DNA-Directed DNA Polymerase/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Mice , Mutagenesis/drug effects , Mutagenesis/genetics , Mutation
8.
J Bacteriol ; 198(20): 2776-83, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27457718

ABSTRACT

UNLABELLED: We tested pairwise combinations of classical base analog mutagens in Escherichia coli to study possible mutagen synergies. We examined the cytidine analogs zebularine (ZEB) and 5-azacytidine (5AZ), the adenine analog 2-aminopurine (2AP), and the uridine/thymidine analog 5-bromodeoxyuridine (5BrdU). We detected a striking synergy with the 2AP plus ZEB combination, resulting in hypermutability, a 35-fold increase in mutation frequency (to 53,000 × 10(-8)) in the rpoB gene over that with either mutagen alone. A weak synergy was also detected with 2AP plus 5AZ and with 5BrdU plus ZEB. The pairing of 2AP and 5BrdU resulted in suppression, lowering the mutation frequency of 5BrdU alone by 6.5-fold. Sequencing the mutations from the 2AP plus ZEB combination showed the predominance of two new hot spots for A·T→G·C transitions that are not well represented in either single mutagen spectrum, and one of which is not found even in the spectrum of a mismatch repair-deficient strain. The strong synergy between 2AP and ZEB could be explained by changes in the dinucleoside triphosphate (dNTP) pools. IMPORTANCE: Although mutagens have been widely studied, the mutagenic effects of combinations of mutagens have not been fully researched. Here, we show that certain pairwise combinations of base analog mutagens display synergy or suppression. In particular, the combination of 2-aminopurine and zebularine, analogs of adenine and cytidine, respectively, shows a 35-fold increased mutation frequency compared with that of either mutagen alone. Understanding the mechanism of synergy can lead to increased understanding of mutagenic processes. As combinations of base analogs are used in certain chemotherapy regimens, including those involving ZEB and 5AZ, these results indicate that testing the mutagenicity of all drug combinations is prudent.


Subject(s)
Azacitidine/toxicity , Base Pairing/drug effects , Bromodeoxyuridine/toxicity , Cytidine/analogs & derivatives , Escherichia coli/drug effects , Mutagens/toxicity , Mutation/drug effects , Azacitidine/chemistry , Bromodeoxyuridine/chemistry , Cytidine/chemistry , Cytidine/toxicity , Drug Synergism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mutagens/chemistry
9.
Viruses ; 7(12): 6233-40, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26633464

ABSTRACT

The current outbreak of Ebola virus (EBOV) in West Africa has claimed the lives of more than 15,000 people and highlights an urgent need for therapeutics capable of preventing virus replication. In this study we screened known nucleoside analogues for their ability to interfere with EBOV replication. Among them, the cytidine analogue ß-d-N4-hydroxycytidine (NHC) demonstrated potent inhibitory activities against EBOV replication and spread at non-cytotoxic concentrations. Thus, NHC constitutes an interesting candidate for the development of a suitable drug treatment against EBOV.


Subject(s)
Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Ebolavirus/drug effects , Ebolavirus/physiology , Virus Replication/drug effects , Animals , Antiviral Agents/toxicity , Chlorocebus aethiops , Cytidine/pharmacology , Cytidine/toxicity , Vero Cells
10.
Chem Res Toxicol ; 25(11): 2443-50, 2012 Nov 19.
Article in English | MEDLINE | ID: mdl-22924387

ABSTRACT

The "mitochondrial Amidoxime Reducing Component" (mARC) is the newly discovered fourth molybdenum enzyme in mammals. All hitherto analyzed mammals express two mARC proteins, referred to as mARC1 and mARC2. Together with their electron transport proteins cytochrome b(5) and NADH cytochrome b(5) reductase, they form a three-component enzyme system and catalyze the reduction of N-hydroxylated prodrugs. Here, we demonstrate the reductive detoxification of toxic and mutagenic N-hydroxylated nucleobases and their corresponding nucleosides by the mammalian mARC-containing enzyme system. The N-reductive activity was found in all tested tissues with the highest detectable conversion rates in liver, kidney, thyroid, and pancreas. According to the presumed localization, the N-reductive activity is most pronounced in enriched mitochondrial fractions. In vitro assays with the respective recombinant three-component enzyme system show that both mARC isoforms are able to reduce N-hydroxylated base analogues, with mARC1 representing the more efficient isoform. On the basis of the high specific activities with N-hydroxylated base analogues relative to other N-hydroxylated substrates, our data suggest that mARC proteins might be involved in protecting cellular DNA from misincorporation of toxic N-hydroxylated base analogues during replication by converting them to the correct purine or pyrimidine bases, respectively.


Subject(s)
Adenine/analogs & derivatives , Cytidine/analogs & derivatives , Cytosine/analogs & derivatives , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Adenine/chemistry , Adenine/metabolism , Adenine/toxicity , Biocatalysis , Cytidine/chemistry , Cytidine/metabolism , Cytidine/toxicity , Cytosine/chemistry , Cytosine/metabolism , Cytosine/toxicity , Humans , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/isolation & purification , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
11.
DNA Repair (Amst) ; 9(2): 153-60, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-20005183

ABSTRACT

Most bacteria, including Escherichia coli, lack an enzyme that can phosphorylate deoxycytidine and its analogs. Consequently, most studies of toxicity and mutagenicity of cytosine analogs use ribonucleosides such as 5-azacytidine (AzaC) and zebularine (Zeb) instead of their deoxynucleoside forms, 5-aza-2'-deoxycytidine (AzadC) and 2'-deoxy-zebularine (dZeb). The former analogs are incorporated into both RNA and DNA creating complex physiological responses in cells. To circumvent this problem, we introduced into E. coli the Drosophila deoxynucleoside kinase (Dm-dNK), which has a relaxed substrate specificity, and tested these cells for sensitivity to AzadC and dZeb. We find that Dm-dNK expression increases substantially sensitivity of cells to these analogs and dZeb is very mutagenic in cells expressing the kinase. Furthermore, toxicity of dZeb in these cells requires DNA mismatch correction system suggesting a mechanism for its toxicity and mutagenicity. The fluorescence properties of dZeb were used to quantify the amount of this analog incorporated into cellular DNA of mismatch repair-deficient cells expressing Dm-dNK and the results showed that in a mismatch correction-defective strain a high percentage of DNA bases may be replaced with the analog without long term toxic effects. This study demonstrates that the mechanism by which Zeb and dZeb cause cell death is fundamentally different than the mechanism of toxicity of AzaC and AzadC. It also opens up a new way to study the mechanism of action of deoxycytidine analogs that are used in anticancer chemotherapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Deoxycytidine/toxicity , Drosophila melanogaster/enzymology , Escherichia coli/drug effects , Mutagenesis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Antineoplastic Agents/pharmacology , Azacitidine/chemistry , Azacitidine/toxicity , Cytidine/analogs & derivatives , Cytidine/chemistry , Cytidine/toxicity , DNA/metabolism , DNA Methylation/drug effects , DNA Mismatch Repair/drug effects , Drosophila melanogaster/drug effects , Fluorescence , Microbial Sensitivity Tests , Phosphorylation/drug effects
12.
J Med Chem ; 52(17): 5394-407, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19725579

ABSTRACT

The application of a phosphoramidate prodrug approach to 2'-C-methylcytidine (NM107), the first nucleoside inhibitor of the hepatitis C virus (HCV) NS5B polymerase, is reported. 2'-C-Methylcytidine, as its valyl ester prodrug (NM283), was efficacious in reducing the viral load in patients infected with HCV. Several of the phosphoramidates prepared demonstrated a 10- to 200-fold superior potency with respect to the parent nucleoside in the cell-based replicon assay. This is due to higher levels of 2'-C-methylcytidine triphosphate in the cells. These prodrugs are efficiently activated and converted to the triphosphate in hepatocytes of several species. Our SAR studies ultimately led to compounds that gave high levels of NTP in hamster and rat liver after subcutaneous dosing and that were devoid of the toxic phenol moiety usually found in ProTides.


Subject(s)
Amides/metabolism , Amides/therapeutic use , Antiviral Agents/metabolism , Cytidine/analogs & derivatives , Hepatitis C/drug therapy , Phosphoric Acids/metabolism , Phosphoric Acids/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Amides/pharmacology , Amides/toxicity , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Antiviral Agents/toxicity , Cell Line , Cytidine/metabolism , Cytidine/pharmacology , Cytidine/therapeutic use , Cytidine/toxicity , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Phosphoric Acids/pharmacology , Phosphoric Acids/toxicity , Polyphosphates/metabolism , Prodrugs/pharmacology , Prodrugs/toxicity , Structure-Activity Relationship , Virus Replication/drug effects
13.
Leuk Res ; 31(11): 1545-51, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17459472

ABSTRACT

The experimental cytotoxic drug cyclopentenyl cytosine (CPEC) is a non-competitive inhibitor of the enzyme cytidine triphosphate (CTP) synthethase. We evaluated the in vitro and in vivo antitumor activity of CPEC on human acute lymphoblastic leukemia (ALL) cell lines. CPEC displayed anti-leukemic activity with IC50 (after 3 days of incubation) ranging from 6 to 15 nM. Subsequently the in vivo activity of CPEC against primary human ALL was evaluated in a xenogeneic model of human ALL using NOD/scid mice inoculated with primary human ALL cells. In the model, only a marginal anti-leukemic activity was observed at 1.5 mg kg(-1) (5 days per week) and 5 mg kg(-1) (2 days per week), however, this activity was associated with severe systemic toxicity. The observed toxicity was not specific for the NOD/scid model, as toxicity at comparable treatment intensity was also observed in Balb/c mice. In conclusion, although CPEC showed antitumor activity against human ALL cells in vitro, its activity in the in vivo human leukemia model was only marginal and accompanied by severe toxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cytidine/analogs & derivatives , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Cytidine/toxicity , Humans , Karyotyping , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
14.
Antimicrob Agents Chemother ; 49(7): 2589-97, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980324

ABSTRACT

Beta-D-2',3'-dideoxy-3'-oxa-5-fluorocytidine (D-FDOC) is an effective inhibitor of human immunodeficiency virus 1 (HIV-1) and HIV-2, simian immunodeficiency virus, and hepatitis B virus (HBV) in vitro. The purpose of this study was to evaluate the intracellular metabolism of d-FDOC in human hepatoma (HepG2), human T-cell lymphoma (CEM), and primary human peripheral blood mononuclear (PBM) cells by using tritiated compound. By 24 h, the levels of D-FDOC-triphosphate (D-FDOC-TP) were 2.8 +/- 0.4, 6.7 +/- 2.3, and 2.0 +/- 0.1 pmol/10(6) cells in HepG2, CEM, and primary human PBM cells, respectively. Intracellular D-FDOC-TP concentrations remained greater than the 50% inhibitory concentration for HIV-1 reverse transcriptase for up to 24 h after removal of the drug from cell cultures. In addition to d-FDOC-monophosphate (D-FDOC-MP), -diphosphate (D-FDOC-DP), and -TP, D-FDOC-DP-ethanolamine and d-FDOC-DP-choline were detected in all cell extracts as major intracellular metabolites. D-FDOC was not a substrate for Escherichia coli thymidine phosphorylase. No toxicity was observed in mice given D-FDOC intraperitoneally for 6 days up to a dose of 100 mg/kg per day. Pharmacokinetic studies in rhesus monkeys indicated that D-FDOC has a t(1/2) of 2.1 h in plasma and an oral bioavailability of 38%. The nucleoside was excreted unchanged primary in the urine, and no metabolites were detected in plasma or urine. These results suggest that further safety and pharmacological studies are warranted to assess the potential of this nucleoside for the treatment of HIV- and HBV-infected individuals.


Subject(s)
Antiviral Agents , Cytidine , Cytidine/analogs & derivatives , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Antiviral Agents/toxicity , Cell Line, Tumor , Cytidine/administration & dosage , Cytidine/metabolism , Cytidine/pharmacokinetics , Cytidine/toxicity , Escherichia coli/enzymology , Female , Humans , Leukocytes, Mononuclear/metabolism , Macaca mulatta , Male , Mice , Thymidine Phosphorylase/metabolism
15.
Arch Toxicol ; 79(5): 268-76, 2005 May.
Article in English | MEDLINE | ID: mdl-15902424

ABSTRACT

The experimental anticancer drug cyclopentenyl cytosine (CPEC) was associated with cardiotoxicity in a phase I study. The aim of the present study was twofold; first we investigated whether the observed effects could be reproduced in in-vitro and in-vivo rat models. Second, we intended to investigate the underlying mechanism of the possible cardiotoxicity of CPEC. Effects on frequency and contractility were studied on the isolated atria of 18 male Wistar rats. Atria were incubated with 0.1 mmol L(-1) (n = 6) or 1 mmol L(-1) (n = 6) CPEC for 1.5 h and compared with control atria (incubation with buffer solution, n = 6). The cardiac apoptosis-inducing potential was studied in-vivo on 66 rats by 99mTc-Annexin V scintigraphy, followed by postmortem determination of radioactivity in tissues, histological confirmation with the TUNEL assay (late-phase apoptosis), and immunohistochemical staining for cleaved caspase 3 and cytochrome C (early-phase apoptosis). Serum levels of the necrotic cardiomyopathy marker troponin T were also determined. No effect on heart frequency was found in the isolated atria after CPEC treatment. A trend towards a decrease of contraction force was observed. However, the differences were not statistically significant. 99mTc-Annexin V scintigraphy showed no increase in cardiac uptake ratio upon CPEC treatment in the in-vivo rat model, which was confirmed by determination of radioactivity in heart versus blood ratios. At each section a few individual isolated late apoptotic cells (< 5) could be identified by the TUNEL assay in the highest CPEC dose group (90 mg kg(-1)) but not in controls or in rats treated with 60 mg kg(-1) CPEC. Staining for the early apoptosis markers cleaved caspase 3 and cytochrome C did not reveal any significant differences between treated and control rats. Cardiac troponin T levels were not increased after CPEC treatment. CPEC does not affect heart frequency or contraction force in our cardiotoxicity models. Moreover, we did not find an indication of CPEC-induced apoptosis in heart tissue.


Subject(s)
Antineoplastic Agents/toxicity , Cardiomyopathies/chemically induced , Cytidine/analogs & derivatives , Heart/drug effects , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Cardiomyopathies/blood , Cardiomyopathies/pathology , Cytidine/toxicity , Disease Models, Animal , Heart/diagnostic imaging , Heart Atria/diagnostic imaging , Heart Atria/drug effects , Heart Atria/pathology , Heart Atria/physiopathology , In Situ Nick-End Labeling , Male , Myocardial Contraction/drug effects , Myocardium/pathology , Organotechnetium Compounds , Rats , Rats, Wistar , Tomography, Emission-Computed, Single-Photon , Troponin T/blood
16.
Jpn J Cancer Res ; 92(3): 343-51, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11267946

ABSTRACT

We examined the effects of dosage schedule on antitumor activity in vitro and in vivo to determine the optimal administration schedule for a new nucleoside antimetabolite 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd, TAS-106). The cytotoxicity of TAS-106 in vitro against human tumors was evaluated at three drug exposure periods. TAS-106 exhibited fairly potent cytotoxicity even with 4 h exposure, and nearly equivalent and sufficiently potent cytotoxicity with 24 and 72 h exposures. These results suggest that long-term exposure to TAS-106 will not be required to achieve maximal cytotoxicity. The antitumor activity of TAS-106 in vivo was compared in nude rat models bearing human tumors on three administration schedules, once weekly, 3 times weekly, and 5 times weekly for 2 or 4 consecutive weeks. TAS-106 showed strong antitumor activity without serious toxicity on all three schedules, but the antitumor activity showed no obvious schedule-dependency in these models. When tumor-bearing nude rats were given a single i.v. dose of [(3)H]TAS-106, tumor tissue radioactivity tended to remain high for longer periods of time as compared to the radioactivity in various normal tissues. Furthermore, when the metabolism of TAS-106 in the tumor was examined, it was found that TAS-106 nucleotides (including the active metabolite, the triphosphate of TAS-106) were retained at high concentrations for prolonged periods. These pharmacodynamic features of TAS-106 may explain the strong antitumor activity without serious toxicity, observed on intermittent administration schedules, in nude rat models with human tumors. We therefore consider TAS-106 to be a promising compound which merits further investigation in patients with solid tumors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/therapeutic use , Cell Survival/drug effects , Cytidine/pharmacokinetics , Cytidine/therapeutic use , Lung Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Animals , Antimetabolites, Antineoplastic/toxicity , Breast Neoplasms , Colonic Neoplasms , Cytidine/analogs & derivatives , Cytidine/toxicity , Female , Humans , Male , Pancreatic Neoplasms , Rats , Rats, Inbred F344 , Rats, Nude , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured
17.
Nucleic Acids Symp Ser ; (42): 133-4, 1999.
Article in English | MEDLINE | ID: mdl-10780415

ABSTRACT

The molecular mechanism of cell death induced by 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd: Figure 1), a potent inhibitor of RNA synthesis, was performed using mouse mammary tumor FM3A cells and human fibrosarcoma HT1080 cells. ECyd induced the characteristics of apoptosis on these cells, such as morphological changes, DNA fragmentations (Figure 2), and caspase-3-like protease activation. General caspases inhibitor (Z-Asp-CH2-DCB) inhibited these changes and cell death. We also found that ECyd induced DNA and 28S ribosomal RNA (rRNA) fragmentations. Though the mechanisms of rRNA fragmentations haven't revealed, it suggests that translational function of the treated cells should be disturbed. These results indicate that antitumor mechanism of ECyd are characteristics of apoptosis on the cells and rRNA fragmentations is one of the death events resulted inhibition of RNA synthesis.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Cytidine/analogs & derivatives , Nucleic Acid Synthesis Inhibitors/toxicity , Transcription, Genetic/drug effects , Animals , Caspase 3 , Caspases/metabolism , Cytidine/toxicity , DNA Fragmentation , DNA, Neoplasm/drug effects , Enzyme Activation , Female , Fibrosarcoma , Humans , Mammary Neoplasms, Experimental , Mice , RNA, Ribosomal, 28S/genetics , RNA, Ribosomal, 28S/radiation effects , Tumor Cells, Cultured
18.
Mutat Res ; 402(1-2): 23-8, 1998 Jun 18.
Article in English | MEDLINE | ID: mdl-9675236

ABSTRACT

Previous studies in our laboratory have identified a set of mutations in the Escherichia coli dnaE gene that confer increased accuracy of DNA replication (antimutators). The dnaE gene encodes the polymerase subunit of DNA polymerase III holoenzyme that replicates the E. coli chromosome. Here, we have investigated their effect on mutagenesis by the base analog N4-aminocytidine (4AC). For three different mutational markers, rifampicin resistance, nalidixic acid resistance and lacI forward mutagenesis, the dnaE911 allele reduced 4AC-induced mutagenesis by approximately 2.5-fold, while the dnaE915 allele reduced it by 2.5-, 3.5- and 6.5-fold, respectively. We also investigated the dependence of 4AC mutagenesis on mutations in the MutHLS mismatch repair system and the UvrABC nucleotide excision repair system. The results show that mutagenesis by 4AC is unaffected by defects in either system. The combined results point to the critical role of the DNA polymerase in preventing mutations by base analogs.


Subject(s)
Antimutagenic Agents , Cytidine/analogs & derivatives , DNA Polymerase III/genetics , Escherichia coli/genetics , Mutagens/toxicity , Mutation , Alleles , Chromosomes, Bacterial , Cytidine/toxicity , Dose-Response Relationship, Drug
19.
Nucleic Acids Symp Ser ; (37): 137-8, 1997.
Article in English | MEDLINE | ID: mdl-9586037

ABSTRACT

The cytotoxic mechanisms of 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)cytosine (ECyd) and 1-(3-C-ethynyl-beta-D-ribo-pentofuranosyl)uracil (EUrd) were studied with mouse mammary tumor FM3A cells and human fibrosarcoma HT1080 cells. ECyd and EUrd are converted to ECyd 5'-triphosphate (ECTP) in the cells. ECTP has also outstanding stability in the cells; the half life of ECTP in FM3A cells was more than 3 days. The metabolisms and mechanisms of these analogues may play a key role in a potent antitumor activities against slow-growing solid tumors.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Cytidine/analogs & derivatives , Uridine/analogs & derivatives , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacokinetics , Biotransformation , Cell Death/drug effects , Cell Line , Cytidine/chemistry , Cytidine/pharmacokinetics , Cytidine/toxicity , Female , Fibrosarcoma , Humans , Mammary Neoplasms, Experimental , Mice , Models, Chemical , Tumor Cells, Cultured , Uridine/chemistry , Uridine/pharmacokinetics , Uridine/toxicity
20.
Biochem Pharmacol ; 49(2): 173-80, 1995 Jan 18.
Article in English | MEDLINE | ID: mdl-7840794

ABSTRACT

Among nine compounds surveyed, cytidine was found to be the most effective in reversing the antiproliferative effects of cyclopentenyl cytosine (CPEC) on human T-lymphoblasts (MOLT-4) in culture. Cytidine, at concentrations of 1-25 microM, enabled cells to maintain normal logarithmic growth when added up to 12 hr after exposure to a 200 nM concentration of the oncolytic nucleoside, CPEC. The most abundant CPEC metabolite, CPEC-5'-triphosphate, is a potent [K1 approximately 6 microM] inhibitor of CTP synthetase (EC 6.3.4.2). Accumulation of this inhibitor resulted in a depletion of CTP levels to 17% of their original cellular concentration. Exogenous cytidine reversed CPEC-induced cellular cytotoxicity by suppressing the formation of CPEC-5'-triphosphate by 70%, and by partially replenishing intracellular CTP to at least 60-70% of its original concentration. In vivo, cytidine (500 mg/kg) administered intraperitoneally 4 hr after each daily dose of CPEC (LD10-LD100) for 9 days reduced the toxicity and abolished the lethality of CPEC to non-tumored mice. Of greater practical importance is the finding that, under these experimental conditions, cytidine did not curtail the antineoplastic properties of CPEC in L1210 tumor-bearing mice. Moreover, the concentration range over which CPEC exhibited antineoplastic activity was extended with cytidine administration.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytidine/analogs & derivatives , Cytidine/pharmacology , Leukemia L1210/drug therapy , Animals , Cytidine/antagonists & inhibitors , Cytidine/blood , Cytidine/therapeutic use , Cytidine/toxicity , Cytidine Triphosphate/analysis , Drug Interactions , Leukemia L1210/blood , Male , Mice , Survival Analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...