Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Acta sci., Health sci ; 44: e56960, Jan. 14, 2022.
Article in English | LILACS | ID: biblio-1367539

ABSTRACT

Colorectal cancer is the 4thcause of cancer death; with considering the growth process of this cancer and the necessity of early diagnosis, the purpose of the research is to state the LncRNA 00970, LncRNA UCAI,and the Wntgene before and after the treatment by 5-Azacytidine epigenetic medicine, to reach the biomarker in the very first steps of colorectal cancer. In this experiment, the human colon cancer cell line (HT29) treated with different concentrations of 5-aza-2'-deoxycytidine (5-aza-dC) was utilized to induce DNA demethylation; Quantitative PCR (qPCR) was used to measure LncRNA UCA1and LncRNA LINC00970 and Wntexpression. There was a significant relationship between the expression of LncRNA 00970, LncRNA UCAI,and the Wntgene and its effects on colorectal (p < 0.05). The Wntgene was treated by 1 and 10 of 5-Azacytidine epigenetic medicine, which then experienced decreases. In LncRNA UCAI and LncRNA00970 in dose 1 micromolar of 5-Azacytidine had decrement and increment of expressionrespectively that explains their efficiency but in treatment by dose 10 mM of this medicine, no significant LncRNA expression difference was detected, 5-azacitidine has a direct impact on its target genes and LncRNAs.Therefore, it can be used in the early diagnosis of colorectal cancer.


Subject(s)
In Vitro Techniques/methods , DNA/analysis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/therapy , Colonic Neoplasms/diagnosis , Early Diagnosis , Azacitidine/analysis , Azacitidine/antagonists & inhibitors , Biomarkers , Colorectal Neoplasms/mortality , Cell Line/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/therapy , Epigenomics , RNA, Long Noncoding , RNA, Long Noncoding/drug effects , Genes
2.
Toxicol Appl Pharmacol ; 396: 114996, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32278510

ABSTRACT

Antineoplastic drugs cause severe cytotoxicity for normal cells, especially hematopoietic stem cells (HSCs). However, bleomycin (BLM) is glycopeptide antibiotic that is effective on various cancers and has either low or no myelosuppression effects. The aim of the present study was to investigate the effect of BLM on 5-Azacitidine (5-AZA) induced cytotoxicity in bone marrow HSCs. 5-AZA reduced HSC cell viability in a time and dose-dependent manner with an IC50 value of 16 µM. However, pretreatment of the cells with BLM for 4 h induced an antagonistic cytotoxicity with an increased IC50 of 64 µM. 5-AZA decreased the colony formation ability of HSC cells in semi-solid agar culture and this effect was attenuated by BLM. 5-AZA significantly downregulated high mobility group Box1 (HMGB1) and Bcl-2 gene expression but upregulated Bax gene expression, while BLM impeded the action of 5-AZA. Pretreatment with BLM remarkably decreased HMGB1 release into culture media that was induced by 5-AZA. The cells were distribution at the sub/G1 phase. Annexin/PI staining of the cells, poly (ADP-ribose) polymerase (PARP) cleavage, and anion superoxide production indicated that BLM limited 5-AZA induced apoptotic cell death. In conclusion, BLM in combination with 5-AZA effectively reduces the adverse cytotoxic effects of 5-AZA on bone marrow hematopoietic stem cells, providing a new chemotherapeutic strategy.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Azacitidine/toxicity , Bleomycin/pharmacology , HMGB1 Protein/metabolism , Hematopoietic Stem Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism , Animals , Azacitidine/antagonists & inhibitors , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism
3.
Drug Metab Pharmacokinet ; 32(6): 301-310, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29174536

ABSTRACT

Decitabine (DAC), a nucleoside-related DNA methylation inhibitor, is taken up into cancer cells via equilibrative nucleoside transporter 1 (ENT1), and is then monophosphorylated by deoxycytidine kinase (dCK). In the present study, we examined the contribution of dCK to the uptake of DAC in HCT116 colon cancer cells. Irinotecan and etoposide inhibited the uptake of [3H]-uridine and [3H]-DAC at 10 s and 5 min, while cytarabine and gemcitabine only inhibited that of [3H]-DAC at 5 min. Irinotecan and etoposide inhibited [3H]-DAC uptake in negative control small interfering RNA (siRNA)- or dCK siRNA-transfected cells at 10 s, whereas cytarabine and gemcitabine did not. Cytarabine and gemcitabine inhibited DAC monophosphate generation by the cytosolic proteins of HCT116 cells and recombinant human dCK protein, assessed using polyethylenimine cellulose thin-layered chromatography. Simulations using simple kinetic models showed that apparent DAC uptake in dCK and ENT1 siRNA-treated cells was attributed to its conversion to monophosphates or a decrease in the cellular flux, respectively, and that the apparent uptake of DAC in dCK-knockdown and ENT1-knockdown cells was similar at longer times, but differed at a very short time. These results suggest that the apparent uptake of DAC is affected by ENT1 and dCK in HCT116 cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/analogs & derivatives , Colonic Neoplasms/drug therapy , Deoxycytidine Kinase/metabolism , Antimetabolites, Antineoplastic/metabolism , Azacitidine/antagonists & inhibitors , Azacitidine/metabolism , Azacitidine/pharmacokinetics , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Colonic Neoplasms/pathology , Cytarabine/pharmacology , Decitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Etoposide/pharmacology , HCT116 Cells , Humans , Irinotecan , Phosphorylation , Gemcitabine
4.
BMC Microbiol ; 16(1): 130, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27349222

ABSTRACT

BACKGROUND: Escherichia coli K-12 strains contain DNA cytosine methyltransferase (Dcm), which generates 5-methylcytosine at 5'CCWGG3' sites. Although the role of 5-methylcytosine in eukaryotic gene expression is relatively well described, the role of 5-methylcytosine in bacterial gene expression is largely unknown. RESULTS: To identify genes that are controlled by 5-methylcytosine in E. coli, we compared the transcriptomes of cells grown in the absence and presence of the DNA methylation inhibitor 5-azacytidine. We observed expression changes for 63 genes. The majority of the gene expression changes occurred at early stationary phase and were up-regulations. To identify gene expression changes due to a loss of DNA methylation, we compared the expression of selected genes in a wild-type and dcm knockout strain via reverse transcription quantitative PCR. CONCLUSIONS: Our data indicate that 5-azacytidine can influence gene expression by at least two distinct mechanisms: DNA methylation loss and a mechanism that is independent of DNA methylation loss. In addition, we have identified new targets of 5-methylcytosine-mediated regulation of gene expression. In summary, our data indicate that 5-azacytidine impacts the composition of the bacterial transcriptome, and the primary effect is increased gene expression at early stationary phase.


Subject(s)
Azacitidine/antagonists & inhibitors , DNA Methylation/drug effects , Escherichia coli K12/drug effects , Escherichia coli K12/genetics , Gene Expression Regulation, Bacterial/drug effects , Transcriptome/drug effects , 5-Methylcytosine/metabolism , 5-Methylcytosine/physiology , Base Sequence , Cell Culture Techniques , Cytosine , DNA, Bacterial , Escherichia coli K12/growth & development , Escherichia coli K12/metabolism , Escherichia coli Proteins/genetics , Gene Knockout Techniques , Genes, Bacterial , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Bacterial/isolation & purification , Sequence Analysis , Up-Regulation
5.
Mol Genet Metab ; 114(4): 604-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25661292

ABSTRACT

Due to the active inhibition of the adipogenic programming, the default destiny of the developing lung mesenchyme is to acquire a myogenic phenotype. We have previously shown that perinatal nicotine exposure, by down-regulating PPARγ expression, accentuates this property, culminating in myogenic pulmonary phenotype, though the underlying mechanisms remained incompletely understood. We hypothesized that nicotine-induced PPARγ down-regulation is mediated by PPARγ promoter methylation, controlled by DNA methyltransferase 1 (DNMT1) and methyl CpG binding protein 2 (MeCP2), two known key regulators of DNA methylation. Using cultured alveolar interstitial fibroblasts and an in vivo perinatal nicotine exposure rat model, we found that PPARγ promoter methylation is strongly correlated with inhibition of PPARγ expression in the presence of nicotine. Methylation inhibitor 5-aza-2'-deoxycytidine restored the nicotine-induced down-regulation of PPARγ expression and the activation of its downstream myogenic marker fibronectin. With nicotine exposure, a specific region of PPARγ promoter was significantly enriched with antibodies against chromatin repressive markers H3K9me3 and H3K27me3, dose-dependently. Similar data were observed with antibodies against DNA methylation regulatory factors DNMT1 and MeCP2. The knock down of DNMT1 and MeCP2 abolished nicotine-mediated increases in DNMT1 and MeCP2 protein levels, and PPARγ promoter methylation, restoring nicotine-induced down regulation of PPARγ and upregulation of the myogenic protein, fibronectin. The nicotine-induced alterations in DNA methylation modulators DNMT1 and MeCP2, PPARγ promoter methylation, and its down-stream targets, were also validated in perinatally nicotine exposed rat lung tissue. These data provide novel mechanistic insights into nicotine-induced epigenetic silencing of PPARγ that could be exploited to design novel targeted molecular interventions against the smoke exposed lung injury in general and perinatal nicotine exposure induced lung damage in particular.


Subject(s)
Epigenesis, Genetic , Lung/cytology , Nicotine/toxicity , PPAR gamma/genetics , Pulmonary Alveoli/metabolism , Animals , Azacitidine/analogs & derivatives , Azacitidine/antagonists & inhibitors , Base Sequence , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Decitabine , Disease Models, Animal , Down-Regulation , Female , Fibroblasts/metabolism , Fibronectins/genetics , Humans , Lung/ultrastructure , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Molecular Sequence Data , Pregnancy , Promoter Regions, Genetic , Pulmonary Alveoli/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Smoking/adverse effects , Up-Regulation
6.
Cancer Lett ; 266(2): 238-48, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18499340

ABSTRACT

2'-Deoxy-N4-[2-(4-nitrophenyl)ethoxycarbonyl]-5-azacytidine (NPEOC-DAC), decitabine with a modification of the N4 position of the azacitidine ring can be used to inhibit DNA methyltransferase. This modification protects the azacitidine ring and can be cleaved by carboxylesterase to release decitabine. NPEOC-DAC was 23-fold less potent at low doses (<10microM) than decitabine at inhibiting DNA methylation, and was also associated with a 3-day delay in its effect. However, at doses > or = 10microM NPEOC-DAC was more effective at inhibiting DNA methylation. Theses differences between decitabine and NPEOC-DAC are dependent on the cleavage of the carboxylester bond, and could be potentially exploited pharmacologically.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Carboxylic Ester Hydrolases/metabolism , DNA-Cytosine Methylases/antagonists & inhibitors , Prodrugs/pharmacology , Antimetabolites, Antineoplastic/chemistry , Azacitidine/antagonists & inhibitors , Azacitidine/chemistry , Azacitidine/pharmacology , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cell Line, Tumor , DNA Methylation/drug effects , Decitabine , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Silencing , Humans , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Prodrugs/chemistry , RNA, Messenger/metabolism , Time Factors
7.
Br J Haematol ; 138(5): 616-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17686055

ABSTRACT

Azacitidine and decitabine are cytidine analogues that inhibit DNA methylation, and are used to treat myeloid haematological malignancies. Hydroxycarbamide (HC) (also known as hydroxyurea), a ribonucleotide reductase (RR) inhibitor, blocks the conversion of ribonucleotides to deoxyribonucleotides, and is also used to treat leukaemia and sickle-cell disease. Azacitidine is a ribonucleoside and decitabine is a deoxyribonucleoside; therefore, we hypothesized that inhibition of RR by HC would be antagonistic to azacitidine and synergistic to decitabine. HL-60 and T24 cancer cell lines were treated with azacitidine or decitabine in combination with HC and DNA methylation of LRE1, MAGEA1 and CDKN2A was quantitatively measured by bisulphite-polymerase chain reaction pyrosequencing. Surprisingly, we found that HC blocked the ability of both azacitidine and decitabine to inhibit DNA methylation and this antagonistic effect was attributable to the arrest of the cell cycle induced by HC. However, this antagonism could be avoided with sequential treatment of HC followed by azacitidine or decitabine. This data suggest that concurrent combination of HC blocks the ability of azacitidine and decitabine to inhibit DNA methylation and therefore these drugs should be used sequentially.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/antagonists & inhibitors , DNA Methylation/drug effects , Hydroxyurea/pharmacology , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Aphidicolin/pharmacology , Azacitidine/administration & dosage , Azacitidine/pharmacology , Cell Cycle/drug effects , DNA, Neoplasm/metabolism , Decitabine , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Hydroxyurea/administration & dosage , Tumor Cells, Cultured
8.
J Exp Ther Oncol ; 1(3): 191-203, 1996 May.
Article in English | MEDLINE | ID: mdl-9414404

ABSTRACT

Fazarabine has shown activity in the panel of 60 cultured human tumor lines of the National Cancer Institute. COMPARE analyses relating correlation coefficients of other anticancer drugs with those of fazarabine suggest that this agent operates through a similar mode of action to that of cytarabine. Studies have been carried out both in culture and in vivo to examine the mechanism of action of fazarabine in P388 murine and Molt-4 human lymphoblasts. Authentic fazarabine nucleotide standards were prepared by chemical and enzymatic methods and characterized on HPLC by comparison to related pyrimidine nucleoside-5'-phosphates as well as by enzymatic digestion. Fazarabine inhibited the incorporation of labeled thymidine into DNA without influencing the synthesis of RNA or protein. Deoxycytidine overcomes this inhibition of DNA synthesis and also prevents the cytotoxicity of the drug to lymphoblasts, probably by competing for fazarabine uptake and metabolism. Fazarabine was rapidly phosphorylated in both cell lines; in P388 cells it was incorporated into DNA, where it continued to undergo the same type of ring opening and degradation as the free nucleoside. Alkaline elution studies demonstrated that exposure to the agent resulted in the formation of alkaline labile sites. Fazarabine also inhibited the methylation of deoxycytidine residues in DNA, but this effect was less pronounced than that produced by 5-azacytidine. Taken together, these studies suggest that fazarabine probably acts by arresting the synthesis and/or altering the structural integrity or functional competence of DNA.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/analogs & derivatives , Lymphocytes/drug effects , Animals , Antimetabolites, Antineoplastic/metabolism , Azacitidine/antagonists & inhibitors , Azacitidine/metabolism , Azacitidine/therapeutic use , Cell Survival/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Deoxycytidine/pharmacology , Humans , Leukemia P388/drug therapy , Lymphocytes/metabolism , Mice , Mice, Inbred Strains , Phosphorylation , Tumor Cells, Cultured
9.
Eur J Pharmacol ; 193(2): 173-7, 1991 Feb 07.
Article in English | MEDLINE | ID: mdl-1710991

ABSTRACT

The effects of 5-azacytidine and hydroxyurea on their independent ability to change adult hemoglobin proportions toward newborn proportions in adult rats were examined. The results revealed that both the chemotherapeutic agents were capable of switching certain hemoglobin components toward newborn values and required similar time-span to express their actions. However, the switching effect of these drugs was totally lost if aspirin was simultaneously administered into the rats, reflecting the need for concurrent prostaglandin synthesis.


Subject(s)
Aspirin/pharmacology , Azacitidine/antagonists & inhibitors , Fetal Hemoglobin/drug effects , Hydroxyurea/antagonists & inhibitors , Age Factors , Animals , Azacitidine/pharmacology , Fetal Hemoglobin/chemistry , Hydroxyurea/pharmacology , Male , Rats , Rats, Inbred Strains , Reference Values
10.
Teratog Carcinog Mutagen ; 11(5): 259-65, 1991.
Article in English | MEDLINE | ID: mdl-1726448

ABSTRACT

To clarify the mechanism underlying the digital teratogenesis of 5-azacytidine (5-AC), the DNA contents of mesenchymal cells were examined in the limb buds of the rat by microspectrophotometry. 5-AC induced hyperploid cells: 2.4% at 18 h after injection and 3.2% at 24 h. No hyperploid cells were noted 30 h after the treatment. Because posttreatment caffeine suppressed 5-AC-induced digital defects, the actions of caffeine on 5-AC-induced hyperploid cells were further studied; no hyperploid cells were detected at any sampling point. Caffeine suppressed hyperploid cells as well as digital defects. These results suggest that the causative factors of digital defects by 5-AC may involve hyperploid cells. However, in that there were too few hyperploid cells to cause the defects, further studies are required to clarify the nature of the action of 5-AC.


Subject(s)
Abnormalities, Drug-Induced/etiology , Azacitidine/toxicity , Polyploidy , Toes/abnormalities , Abnormalities, Drug-Induced/embryology , Abnormalities, Drug-Induced/genetics , Abnormalities, Drug-Induced/prevention & control , Animals , Azacitidine/antagonists & inhibitors , Caffeine/pharmacology , Female , Hindlimb/abnormalities , Hindlimb/embryology , Humans , Male , Rats , Rats, Inbred Strains/embryology , Specific Pathogen-Free Organisms , Toes/embryology
11.
Teratology ; 39(2): 163-72, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2467392

ABSTRACT

The present study investigated microscopically the process of 5-azacytidine (5-AC)-induced digital teratogenesis and caffeine's suppressive effect on this process. Three distinct zones of programmed cell death were observed in control and caffeine-treated embryos 3 hours after 5-AC injection: the preaxial and postaxial ectodermal regions and the central part of the mesodermal regions. 5-AC temporarily suppressed programmed cell death in the ectoderm and mesoderm 3 hours after it was injected. However, caffeine promoted programmed cell death; normal programmed cell death was observed in the limb buds of embryos whose dams were treated with 5-AC and caffeine. The percentage of total cell death in hindlimb buds of embryos treated with 5-AC and caffeine was higher than that from embryos treated with 5-AC, whereas 5-AC-induced digital malformations were reduced by post-treatment with caffeine. Cell death reached a maximum 12 hours after the injection in limb buds from 5-AC and caffeine-treated embryos and at 24 hours in the 5-AC treated embryos. Furthermore, in the 5-AC and caffeine-treated embryos, the frequency of cell deaths at 12 hours increased almost linearly with the doses of caffeine in parallel with the reduction of 5-AC-induced malformation frequency by caffeine. These results suggest that although induced cell death may be one of the factors leading to digital malformations produced by 5-AC, it is not essential, and the existence of other factors affecting the pattern formation of the limb bud is proposed.


Subject(s)
Abnormalities, Drug-Induced/pathology , Azacitidine/antagonists & inhibitors , Caffeine/pharmacology , Hindlimb/abnormalities , Animals , Azacitidine/adverse effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Embryo, Mammalian , Hindlimb/pathology , Rats , Rats, Inbred Strains , Time Factors
12.
Leukemia ; 1(12): 814-9, 1987 Dec.
Article in English | MEDLINE | ID: mdl-2447450

ABSTRACT

The effect of deoxycytidine (dCyd) on the inhibitory effects of two antileukemic nucleoside analogs, 5-aza-2'-deoxycytidine and ara-5-aza-Cyd, toward the clonogenic growth of normal human bone marrow progenitors (CFU-GM) and leukemic blast progenitors (L-CFU) was examined. Continuous exposure of cells to 10(-6)-10(-5) M 5-aza-deoxycytidine or 10(-5)-5 x 10(-5) M ara-5-aza-Cyd in conjunction with a 10- 100-fold excess of dCyd resulted in significantly greater restoration of CFU-GM growth than L-CFU colony formation at each dose relationship. Normal bone marrow mononuclear cells exposed to 10(-3) M dCyd for 4 hr (along with 5-aza-dCyd or ara-5-aza-Cyd) exhibited intracellular deoxycytidine triphosphate (dCTP) pools 20-fold higher than their leukemic counterparts. However, this finding was not associated with enhanced analog incorporation into leukemic cell DNA. These results suggest that high concentrations of dCyd preferentially protect normal versus leukemic progenitor cells from the inhibitory actions of 5-aza-dCyd and ara-5-aza-Cyd. They also raise the possibility that this in vitro selectivity may be related to enhanced expansion of dCTP pools in normal bone marrow elements and involves factors other than differential short-term analog incorporation into DNA.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/antagonists & inhibitors , Deoxycytidine/pharmacology , Hematopoietic Stem Cells/drug effects , Leukemia/pathology , Neoplastic Stem Cells/drug effects , Acute Disease , Azacitidine/pharmacology , Bone Marrow , DNA, Neoplasm/metabolism , Decitabine , Deoxycytosine Nucleotides/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Neoplastic Stem Cells/metabolism
13.
Br J Cancer ; 56(3): 261-5, 1987 Sep.
Article in English | MEDLINE | ID: mdl-2444244

ABSTRACT

The antiproliferative effects of 5-azacytidine (acaCyd) and 5-aza-2'-deoxycytidine (azadCyd) were studied in murine B16 melanoma and a series of B16 melanoma derived mutant strains with selective resistances to the respective drugs. The in vitro cytotoxicities of azaCyd and azadCyd on B16 wild type, expressed in terms of IC50 values, were found to be 5 microM and 0.2 microM, respectively. The in vitro cytotoxicity of both drugs was dependent on the duration of exposure. Uridine and cytidine were able to reverse the in vitro cytotoxicity of azaCyd, but not of azadCyd. Conversely, 2'-deoxycytidine was able to reverse the cytotoxic effect of azadCyd but not of azaCyd. Thymidine and 2'-deoxyuridine had no detectable effects on the in vitro cytotoxicity of either azaCyd or azadCyd. B16 melanoma mutant strains that were selected for resistance to azaCyd showed no cross-resistance to azadCyd, cytosine arabinoside or the fluorinated pyrimidine analogues FUrd, FCyd, FdUrd and FdCyd. Mutant strains that were selected for resistance to azadCyd showed no cross-resistance to azaCyd or fluorinated pyrimidine analogs, but only to cytosine arabinoside. The combined data suggest that azaCyd and azadCyd follow different routes of intracellular metabolic activation and exert their cytotoxic activity via different intracellular targets.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Melanoma/drug therapy , Animals , Azacitidine/antagonists & inhibitors , Cell Survival/drug effects , Cells, Cultured , Cytidine/pharmacology , Decitabine , Drug Resistance , Mice , Mutation , Uridine/pharmacology
14.
Teratology ; 35(2): 247-52, 1987 Apr.
Article in English | MEDLINE | ID: mdl-2440123

ABSTRACT

The effect of caffeine on 5-azacytidine (5-AC)-induced digital malformations in rat fetuses was investigated. Caffeine suppressed all types of digital defects in the fore- and hindlimbs except for syndactyly induced by 1.0 mg/kg of 5-AC; it was still effective when administered 24 hours after 5-AC treatment. However, fetal mortality increased as the frequency of malformations decreased. While the malformation results support the view that caffeine inhibits the processes leading to malformation expression, the relation between its suppressive effect on malformations and its enhancing effect on fetal mortality is unclear.


Subject(s)
Abnormalities, Drug-Induced/etiology , Azacitidine/toxicity , Caffeine/pharmacology , Limb Deformities, Congenital , Animals , Azacitidine/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Fetal Death/chemically induced , Methylation , Pregnancy , Rats
15.
Cancer Treat Rep ; 70(12): 1451-3, 1986 Dec.
Article in English | MEDLINE | ID: mdl-2431776

ABSTRACT

In this study we show that cytarabine given simultaneously with 5-aza-2'-deoxycytidine (Aza-dC) antagonized Aza-dC activity against L1210 mouse leukemia. This antagonism was seen after a single dose (on Day 3 or Day 5 after tumor implant) and after repeated doses. This observation suggests caution in combining cytarabine with Aza-dC in the treatment of human leukemias.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/analogs & derivatives , Leukemia L1210/drug therapy , Animals , Azacitidine/administration & dosage , Azacitidine/antagonists & inhibitors , Cytarabine/administration & dosage , Decitabine , Drug Interactions , Leukemia L1210/pathology , Male , Mice , Statistics as Topic
16.
Neoplasma ; 27(2): 121-7, 1980.
Article in English | MEDLINE | ID: mdl-6156420

ABSTRACT

Out of different nucleosides and deoxynucleosides testes for their ability to block the phosphorylation of labeled 5-aza-2'-deoxycytidine in the presence of ATP and the cell-free extract from mouse spleen only deoxycytidine and cytosine arabinoside depressed the reaction significantly. With the same system the phosphate donor specificity for 5-aza-2'-deoxycytidine and deoxycytidine was determined. All of the 5'-triphosphates used were less efficient donors with respect to the analogue than to the natural substrate. The apparent Michaelis constants for the phosphorylation of 5-aza-2'-deoxycytidine and deoxycytidine were 2.9 and 2.5 x 10(-5) M, respectively. Using the extract from L1210 leukemic cells the Km constant for 5-aza-2'-deoxycytidine was 6.4 x 10(-5) M and that for deoxycytidine 2.7 x 10(-5) M. The Ki constants with the spleen extract were 1.2 x 10(-3) M for 5-aza-2'-deoxycytidine and 5.8 x 10(-6) M for deoxycytidine. Both compounds acted as competitive inhibitors of one another.


Subject(s)
Azacitidine/analogs & derivatives , Leukemia L1210/metabolism , Spleen/metabolism , Animals , Azacitidine/antagonists & inhibitors , Azacitidine/metabolism , Binding, Competitive , Cell-Free System , Decitabine , Deoxycytidine Kinase/metabolism , Deoxyribonucleosides/pharmacology , Kinetics , Mice , Nucleosides/pharmacology , Phosphorylation
18.
Cancer Res ; 37(7 Pt 1): 1956-61, 1977 Jul.
Article in English | MEDLINE | ID: mdl-67884

ABSTRACT

5,6-Dihydro-5-azacytidine hydrochloride, a chemically stable, soluble analog of 5-azacytidine, has cytostatic activity against mouse leukemic L1210 cells grown in culture, but concentrations on the order of 10 micronM, 10-fold higher, than the parent drug, are necessary to inhibit cell growth. The addition of either cytidine or uridine protected against growth inhibition by 5-azacytidine and 5,6-dihydro-5-azacytidine, whereas thymidine potentiated the cytostatic action of both drugs. Deoxycytidine also enhanced the action of 5-azacytidine but had no effect with the reduced analog. Cell suspensions of L1210 cells were able to phosphorylate 5-azacytidine and, to a lesser extent, 5,6-dihydro-5-azacytidine. In cell-free extracts in the presence of ATP and Mg2+, both drugs were converted to nucleotides but at less than 5% the rate of cytidine. As a substrate for mouse kidney cytidine deaminase, the apparent Km value for 5,6-dihydro-5-azacytidine (33 micronM) is of the same order of magnitude as that for cytidine (37 micronM) but less than that for 5-azacytidine (2.1 X 10(3) micronM). The Vm for deamination of the reduced analog is one-tenth that for 5-azacytidine. 3,4,5,6-Tetrahydrouridine, a potent inhibitor of cytidine deaminase, is more effective in blocking deamination of 5-azacytidine than 5,6-dihydro-5-azacytidine.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Leukemia L1210/drug therapy , Animals , Azacitidine/antagonists & inhibitors , Azacitidine/metabolism , Cell Division/drug effects , Cells, Cultured , Cytidine/pharmacology , Cytidine Deaminase/metabolism , Deoxycytidine/pharmacology , Drug Synergism , Kinetics , Leukemia L1210/metabolism , Oxidative Phosphorylation , Tetrahydrouridine/pharmacology , Thymidine/pharmacology , Uridine/pharmacology
19.
Cancer Res ; 37(6): 1636-9, 1977 Jun.
Article in English | MEDLINE | ID: mdl-66984

ABSTRACT

The in vitro effect of 5-aza-2'-deoxycytidine (5-aza-CdR) on cytotoxicity and macromolecular synthesis in A(T1)C1-3 hamster fibrosarcoma cells was investigated. The in vitro concentrations that produce 50% cell kill for 5-aza-CdR were about 1.0 and 0.01 microng/ml for a 2- and 24-hr exposure, respectively. 5-aza-CdR inhibited the growth of the fibrosarcoma cells by 40% at a concentration of 0.05 microng/ml. Deoxycytidine, but not cytidine, was a potent antagonist of the cytotoxicity produced by 5-aza-CdR. At cytotoxic concentrations 5-aza-CdR did not appear to inhibit DNA, RNA, or protein synthesis during a 1-hr incubation as measured by the incorporation of radioactive thymidine, uridine,, or leucine into acid-insoluble material. At a concentration of 10 microng/ml, 5-aza-CdR stimulated the incorporation of radioactive thymidine into DNA by more than 50%. These results indicate that 5-aza-CdR is a very potent cytotoxic agent to tumor cells in vitro at concentrations that do not inhibit macromolecular synthesis.


Subject(s)
Azacitidine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Fibrosarcoma/drug therapy , Azacitidine/antagonists & inhibitors , Azacitidine/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytidine/pharmacology , DNA, Neoplasm/biosynthesis , Deoxycytidine/antagonists & inhibitors , Deoxycytidine/pharmacology , Fibrosarcoma/metabolism , Neoplasm Proteins/biosynthesis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , RNA, Neoplasm/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...