Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Toxicol Sci ; 46(8): 359-369, 2021.
Article in English | MEDLINE | ID: mdl-34334557

ABSTRACT

Predicting drug-induced side effects in the cardiovascular system is very important because it can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although chronic assessment of cardiac contractility is an important issue in safety pharmacology, an in vitro evaluation system has not been fully developed. We previously developed an imaging-based contractility assay system to detect acute cardiotoxicity using human iPS cell-derived cardiomyocytes (hiPSC-CMs). To extend the system to chronic toxicity assessment, we examined the effects of the anti-hepatitis C virus (HCV) drug candidate BMS-986094, a guanosine nucleotide analogue, which was withdrawn from phase 2 clinical trials because of unexpected contractility toxicities. Additionally, we examined sofosbuvir, another nucleotide analogue inhibitor of HCV that has been approved as an anti-HCV drug. Motion imaging analysis revealed the difference in cardiotoxicity between the cardiotoxic BMS-986094 and the less toxic sofosbuvir in hiPSC-CMs, with a minimum of 4 days of treatment. In addition, we found that BMS-986094-induced contractility impairment was mediated by a decrease in calcium transient. These data suggest that chronic treatment improves the predictive power for the cardiotoxicity of anti-HCV drugs. Thus, hiPSC-CMs can be a useful tool to assess drug-induced chronic cardiotoxicity in non-clinical settings.


Subject(s)
Antiviral Agents/toxicity , Cardiotoxicity/etiology , Guanosine Monophosphate/analogs & derivatives , Guanosine/analogs & derivatives , Guanosine/toxicity , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/drug effects , Antiviral Agents/adverse effects , Calcium/metabolism , Cells, Cultured , Chronic Disease , Guanosine/adverse effects , Guanosine Monophosphate/adverse effects , Guanosine Monophosphate/toxicity , Humans , Molecular Imaging , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Safety
2.
Chemistry ; 21(17): 6381-5, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25771988

ABSTRACT

Oxidation of RNA hairpin models corresponding to anticodon stem-loop (ASL) of transfer RNA led to RNA damage consisting solely of a unique loop guanine oxidation. Manganese porphyrin/oxone treatment of RNA resulted in dehydroguanidinohydantoin (DGh; major) and/or spiroiminodihydantoin (Sp) lesions. Ribose damage was not observed. This two-electron transfer oxidation reaction allowed the identification of guanine oxidation products for further study of RNA species carrying a unique lesion at a single G to investigate their biological impact.


Subject(s)
Guanidines/chemistry , Guanosine/analogs & derivatives , Guanosine/chemistry , Hydantoins/chemistry , Models, Chemical , RNA, Transfer/chemistry , Spiro Compounds/chemistry , Anticodon , Guanidines/toxicity , Guanosine/toxicity , Hydantoins/toxicity , Oxidation-Reduction , RNA, Transfer/metabolism , Ribose/toxicity , Spiro Compounds/toxicity
3.
Mutat Res ; 714(1-2): 11-6, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21704046

ABSTRACT

8-Oxo-7,8-dihydroguanine (8-hydroxyguanine) is oxidized more easily than normal nucleobases, which can produce spiroiminodihydantoin (Sp) and guanidinohydantoin (Gh). These secondary oxidation products of 8-oxo-7,8-dihydroguanine are highly mutagenic when formed within DNA. To evaluate the mutagenicity of the corresponding oxidation products of 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate (8-hydroxy-2'- deoxyguanosine 5'-triphosphate) in the nucleotide pool, Escherichia coli cells deficient in the mutT gene were treated with H(2)O(2), and the induced mutations were analyzed. Moreover, the 2'-deoxyriboside 5'-triphosphate derivatives of Sp and Gh were also introduced into competent E. coli cells. The H(2)O(2) treatment of mutT E. coli cells resulted in increase of G:C → T:A and A:T → T:A mutations. However, the incorporation of exogenous Sp and Gh 2'-deoxyribonucleotides did not significantly increase the mutation frequency. These results suggested that the oxidation product(s) of 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate induces G:C → T:A and A:T → T:A mutations, and that the 2'-deoxyriboside 5'-triphosphate derivatives of Sp and Gh exhibit quite weak mutagenicity, in contrast to the bases in DNA.


Subject(s)
Deoxyguanine Nucleotides/metabolism , Mutagens/toxicity , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Guanidines/toxicity , Guanine/analogs & derivatives , Guanine/metabolism , Guanosine/analogs & derivatives , Guanosine/toxicity , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Hydantoins/toxicity , Hydrogen Peroxide/toxicity , Oxidation-Reduction , Spiro Compounds/toxicity
4.
J Phys Chem B ; 115(12): 3263-70, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21384813

ABSTRACT

Patients treated with the immunosuppressant and anticancer drugs 6-thioguanine, azathioprine, or mercaptopurine can metabolize and incorporate them in DNA as 6-thioguanosine. The skin of these patients is sensitive to UVA radiation, and long-term treatment can result in extremely high incidence of sunlight-induced skin cancer. In this contribution the photophysics of 6-thioguanosine have been studied in aqueous buffer solution and in acetonitrile after excitation with UVA light to provide mechanistic insights about the origin of its phototoxicity. It is shown that most of the initial excited-state population in the S(2)(ππ*, L(a)) state decays by ultrafast intersystem crossing to the triplet manifold. A triplet quantum yield of 0.8 ± 0.2 is determined in aqueous buffer solution. A minor fraction of the S(2) population bifurcates on an ultrafast time scale to populate the S(1)(n(S)π*) state, which decays back to the ground state in tens of picoseconds. Quantum-chemical calculations that include solvent effects support the experimental results. The high triplet yield of 6-thioguanosine, which we argue can result in photosensitization of molecular oxygen and photooxidative DNA damage, is proposed to explain the high phototoxicity exhibited by these pro-drugs in patients upon sunlight exposure. Finally, the experimental and computational results for 6-thioguanosine are compared with those reported for the DNA/RNA guanine monomers.


Subject(s)
Guanosine/analogs & derivatives , Thionucleosides/chemistry , Absorption , Acetonitriles/chemistry , Buffers , Cell Death/drug effects , Cell Death/radiation effects , DNA/genetics , DNA/metabolism , DNA Damage , Electrons , Guanosine/chemistry , Guanosine/metabolism , Guanosine/toxicity , Models, Molecular , Molecular Conformation , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Photosensitizing Agents/toxicity , Solutions , Thionucleosides/metabolism , Thionucleosides/toxicity , Time Factors , Ultraviolet Rays , Water/chemistry
5.
Bioorg Med Chem Lett ; 20(16): 4850-4, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20637609

ABSTRACT

We herein report a novel double pro-drug approach applied to the anti-HCV agent 2'-beta-C-methyl guanosine. A phosphoramidate ProTide motif and a 6-O-methoxy base pro-drug moiety are combined to generate lipophilic prodrugs of the monophosphate of the guanine nucleoside. Modification of the ester and amino acid moieties lead to a compound INX-08189 that exhibits 10nM potency in the HCV genotype 1b subgenomic replicon, thus being 500 times more potent than the parent nucleoside. The potency of the lead compound INX-08189 was shown to be consistent with intracellular 2'-C-methyl guanosine triphosphate levels in primary human hepatocytes. The separated diastereomers of INX-08189 were shown to have similar activity in the replicon assay and were also shown to be similar substrates for enzyme processing. INX-08189 has completed investigational new drug enabling studies and has been progressed into human clinical trials for the treatment of chronic HCV infection.


Subject(s)
Antiviral Agents/chemical synthesis , Guanosine Monophosphate/analogs & derivatives , Hepacivirus/drug effects , Prodrugs/chemical synthesis , Amides/chemistry , Antiviral Agents/chemistry , Antiviral Agents/toxicity , Cells, Cultured , Drug Design , Guanosine/analogs & derivatives , Guanosine/chemical synthesis , Guanosine/toxicity , Guanosine Monophosphate/chemical synthesis , Guanosine Monophosphate/chemistry , Guanosine Monophosphate/pharmacology , Humans , Phosphoric Acids/chemistry , Prodrugs/chemistry , Prodrugs/toxicity
6.
Br J Pharmacol ; 159(6): 1247-63, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20132210

ABSTRACT

BACKGROUND AND PURPOSE: It is well known that adenine-based purines exert multiple effects on pain transmission. However, less attention has been given to the potential effects of guanine-based purines on pain transmission. The aim of this study was to investigate the effects of intraperitoneal (i.p.) and oral (p.o.) administration of guanosine on mice pain models. Additionally, investigation into the mechanisms of action of guanosine, its potential toxicity and cerebrospinal fluid (CSF) purine levels were also assessed. EXPERIMENTAL APPROACH: Mice received an i.p. or p.o. administration of vehicle (0.1 mM NaOH) or guanosine (up to 240 mg x kg(-1)) and were evaluated in several pain models. KEY RESULTS: Guanosine produced dose-dependent antinociceptive effects in the hot-plate, glutamate, capsaicin, formalin and acetic acid models, but it was ineffective in the tail-flick test. Additionally, guanosine produced a significant inhibition of biting behaviour induced by i.t. injection of glutamate, AMPA, kainate and trans-ACPD, but not against NMDA, substance P or capsaicin. The antinociceptive effects of guanosine were prevented by selective and non-selective adenosine receptor antagonists. Systemic administration of guanosine (120 mg x kg(-1)) induced an approximately sevenfold increase on CSF guanosine levels. Guanosine prevented the increase on spinal cord glutamate uptake induced by intraplantar capsaicin. CONCLUSIONS AND IMPLICATIONS: This study provides new evidence on the mechanism of action of the antinociceptive effects after systemic administration of guanosine. These effects seem to be related to the modulation of adenosine A(1) and A(2A) receptors and non-NMDA glutamate receptors.


Subject(s)
Analgesics/therapeutic use , Guanosine/therapeutic use , Pain/drug therapy , Administration, Oral , Analgesics/administration & dosage , Analgesics/cerebrospinal fluid , Analgesics/pharmacology , Analgesics/toxicity , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chromatography, High Pressure Liquid , Disease Models, Animal , Dose-Response Relationship, Drug , Edema/drug therapy , Guanosine/administration & dosage , Guanosine/cerebrospinal fluid , Guanosine/pharmacology , Guanosine/toxicity , Injections, Intraperitoneal , Lethal Dose 50 , Male , Mice , Motor Activity/drug effects , Pain/cerebrospinal fluid , Pain/physiopathology , Pain Threshold/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism
7.
Cancer Lett ; 262(2): 239-47, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18248787

ABSTRACT

8-Nitroguanosine is a nitratively modified nucleoside that is formed endogeneously under inflammatory conditions dependent on nitric oxide production, particularly associated with cancer risks. Here, we investigated the mutagenic potential of 8-nitroguanosine in mammalian cells. Treatment with 8-nitroguanosine (10-1000 microM) for 1h significantly increased (by 6-8 times) the mutation frequency of the xanthine-guanine phosphoribosyltransferase (gpt) gene in AS52 cells without cytotoxic effects. 8-Nitroguanosine treatment induced a G-to-T transversion in gpt gene at position 86. It also significantly increased levels of abasic sites in DNA. These observations suggest that formation of 8-nitroguanosine may contribute to the pathogenesis of inflammation-associated carcinogenesis.


Subject(s)
Guanosine/analogs & derivatives , Mutagens/toxicity , Nitro Compounds/toxicity , Animals , CHO Cells , Cricetinae , Cricetulus , DNA Damage , Guanosine/toxicity , Mutagenicity Tests , Nitrogen Oxides/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
8.
DNA Repair (Amst) ; 5(7): 761-72, 2006 Jul 13.
Article in English | MEDLINE | ID: mdl-16621731

ABSTRACT

In human and rodent cells, MTH1, an oxidized purine nucleoside triphosphatase, efficiently hydrolyzes oxidized dGTP, GTP, dATP and ATP such as 2'-deoxy-8-oxoguanosine triphosphate (8-oxo-dGTP) and 2'-deoxy-2-hydroxyadenosine triphosphate (2-OH-dATP) in nucleotide pools, thus avoiding their incorporation into DNA or RNA. MTH1 is expressed in postmitotic neurons as well as in proliferative tissues, and it is localized both in the mitochondria and nucleus, thus suggesting that MTH1 plays an important role in the prevention of the mutagenicity and cytotoxicity of such oxidized purines as 8-oxoG which are known to accumulate in the cellular genome. Our recent studies with MTH1-deficient mice or cells revealed that MTH1 efficiently minimizes accumulation of 8-oxoG in both nuclear and mitochondrial DNA in the mouse brain as well as in cultured cells, thus contributing to the protection of the brain from oxidative stress.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Purine Nucleotides/metabolism , Purine Nucleotides/toxicity , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Brain/metabolism , Cytotoxins/metabolism , Cytotoxins/toxicity , DNA Glycosylases/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , Guanosine/analogs & derivatives , Guanosine/metabolism , Guanosine/toxicity , Humans , Mice , Neurotoxins/metabolism , Neurotoxins/toxicity , Oxidation-Reduction , Parkinson Disease/metabolism , Parkinsonian Disorders/metabolism , Phosphoric Monoester Hydrolases/deficiency
9.
J Neurosci Res ; 79(1-2): 248-53, 2005.
Article in English | MEDLINE | ID: mdl-15558762

ABSTRACT

Oral and intraperitoneal administration of the nucleoside guanosine have been shown to prevent quinolinic acid- (QA) and alpha-dendrotoxin-induced seizures, impair memory, and impair anxiety in rats and mice. We investigated the effect of 2-weeks ad lib orally administered guanosine (0.5 mg/ml) on seizures induced by QA, inhibitory avoidance memory, and locomotor performance in rats. We also studied the mechanism of action of guanosine through the measurement of its concentration in the cerebrospinal fluid (CSF) and its effect on glutamate uptake in cortical slices of rats. QA produced seizures in 85% of rats, an effect partially prevented by guanosine (53% of seizures; P = 0.0208). Guanosine also impaired retention on the inhibitory avoidance task (P = 0.0278) and decreased locomotor activity on the open field test (P = 0.0101). The CSF guanosine concentration increased twofold in the treated group compared to that in the vehicle group (P = 0.0178). Additionally, QA promoted a 30% decrease in glutamate uptake as compared to that with intracerebroventricular saline administration, an effect prevented by guanosine in animals protected against QA-induced seizures. Altogether, these findings suggest a potential role of guanosine for treating diseases involving glutamatergic excitotoxicity such as epilepsy. These effects seem to be related to modulation of glutamate uptake.


Subject(s)
Behavior, Animal/drug effects , Glutamic Acid/metabolism , Guanosine/administration & dosage , Seizures/chemically induced , Analysis of Variance , Animals , Avoidance Learning/drug effects , Chromatography, High Pressure Liquid/methods , Drug Administration Schedule , Drug Interactions , Exploratory Behavior/drug effects , Guanosine/toxicity , Inhibition, Psychological , Male , Quinolinic Acid/toxicity , Rats , Rats, Wistar , Reaction Time/drug effects , Seizures/cerebrospinal fluid
10.
Fukuoka Igaku Zasshi ; 95(1): 17-30, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15031996

ABSTRACT

Oxygen radicals generated through normal cellular metabolism induce a variety of types of oxidative damage into DNA and its precursors. Among such types of oxidative damage, 7, 8-dihydro-8-oxoguanine (8-oxoG), an oxidized form of guanine, is known to be abundant and highly mutagenic. 8-OxoG can pair with adenine as well as cytosine, thus causing G: C to T: A transversions after DNA replication, if not repaired. Organisms are equipped with elaborate systems to avoid such mutations caused by 8-oxoG. In Escherichia coli, two DNA glycosylases have been identified to suppress these mutations. One is MutM, an 8-oxoguanine DNA glycosylase that removes 8-oxoG from 8-oxoG: C base pairs. The other is MutY, an adenine DNA glycosylase that excises adenine from 8-oxoG: A mismatches. Mammals also have such DNA glycosylases; OGG1 (Ogg1) is the functional counterpart of MutM, and MUTYH (Mutyh) is the MutY homologue. In order to investigate the roles of these two enzymes in the avoidance of 8-oxoG-related mutagenesis in mammals, we analyzed spontaneous mutagenesis in the small intestine of Ogg1-deficient (Ogg1-/-) and Ogg1-, Mutyh-double deficient (Ogg1-/-; Mutyh-/-) mice at the age of 4-5 weeks using the prokaryotic rpsL transgene as a reporter. The observed mutation frequency was 1.00 x 10(-5) in both wild type and Ogg1-/- mice, and 1.91 x 10(-5) in Ogg1-/-; Mutyh-/- mice, indicating that the overall spontaneous mutation frequency was increased in Ogg1-/-; Mutyh-/- mice, but not in Ogg1-/- mice. Analysis of the mutation spectrum revealed that the frequency of G: C to T: A transversions were significantly increased in both Ogg1-/- and Ogg1-/-; Mutyh-/- mice; a 5-fold increase in Ogg1-/- mice, and a 41-fold increase in Ogg1-/-; Mutyh-/- mice when compared with wild type mice. A previous study in our laboratory indicated that a defect in Mutyh caused a 4-fold increase in the frequency of G: C to T: A transversions in mice. Combined, these observations suggest that a cooperative function between Ogg1 and Mutyh exists to prevent 8-oxoG-related mutagenesis in mammals.


Subject(s)
DNA Glycosylases/physiology , Frameshift Mutation/genetics , Guanosine/analogs & derivatives , Mutagenesis/genetics , Animals , DNA Damage/genetics , DNA Replication/genetics , Escherichia coli Proteins , Female , Frameshift Mutation/drug effects , Gene Expression Regulation , Gene Frequency , Genotype , Guanosine/physiology , Guanosine/toxicity , Intestine, Small , Male , Mice , Mice, Transgenic , Mutagenesis/drug effects , Oxidative Stress/genetics , Ribosomal Protein S9
11.
Aquat Toxicol ; 67(1): 23-32, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15019248

ABSTRACT

7,8-Dihydro-8-oxodeoxyguanosine (8-oxo-dG) is a typical modification of DNA caused by oxygen free radicals and can be an useful biomarker for pollutants inducing oxidative stress. An immunoperoxidase method using monoclonal antibody 1F7 toward 8-oxo-dG was applied to tissues and smeared cells of marine organisms for detection and quantification of oxidative DNA damage in such models. The assay, previously employed on human cells, was assessed for the first time on Mediterranean mussels (Mytilus galloprovincialis) and European eels (Anguilla anguilla), exposed to model pro-oxidant chemicals, namely benzo[a]pyrene (B[a]P) and copper. Quantification of 8-oxo-dG was microscopically carried out and expressed as relative nuclear staining intensity. Higher levels of oxidative DNA damage were detected in the digestive glands of treated mussels compared to controls, while the effect was less pronounced in haemocytes, characterized by more elevated basal levels of 8-oxo-dG. The assay was suitable for detection of 8-oxo-dG also in fish liver sections indicating consistent damage after B[a]P exposure. The main advantage of the immunohistochemical approach is the elimination of DNA extraction which considerably reduces the processing of biological samples. In addition, the assay requires small amounts of frozen tissues or fixed cells for detection of 8-oxo-dG and is potentially able to discriminate variable susceptibility to oxidative stress in different cell types. Although further investigations are required for the improvement and the validation of the assay in field conditions, laboratory exposures provided useful indications on the consistency of the approach and the efficacy of antibody 1F7 in marine organisms for a rapid assessment of pollutant-induced oxidative DNA damage.


Subject(s)
Anguilla/metabolism , Bivalvia/metabolism , Guanosine/analogs & derivatives , Guanosine/toxicity , Immunoenzyme Techniques/methods , Oxidative Stress , Animals , Benzo(a)pyrene , Biomarkers , Copper , DNA Damage/drug effects , Guanosine/metabolism , Italy
12.
Bioorg Med Chem ; 10(4): 883-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11836094

ABSTRACT

(-)-5'-noraristeromycin (1) has shown antiviral activity towards, particularly cytomegalovirus, vaccinia virus and measles while its (+)-enantiomer (2) is effective towards hepatitis B virus. To determine if the antiviral characteristics of 1 and 2 extended to the guanine analogues (3 and 4), these enantiomers were prepared and evaluated against herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2), cytomegalovirus (CMV), varicella zoster virus (VZV), Epstein-Barr virus (EBV), human herpes virus type 6 (HHV-6), human herpes virus type 8 (HHV-8), vaccinia virus (VV), cowpox virus (CV), vesicular stomatitis virus (VSV), respiratory syncytial virus (RSV), hepatitis B virus (HBV), and human immunodeficiency virus type 1 (HIV-1) and type 2 (HIV-2). The only activity found for 3 was for Epstein-Barr virus in VCA Elisa (EC50 0.78 microg/mL), immunofluorescence assay for VCA or gp 350/250 (1.8-4.0 microg/mL) and DNA hybridization (EC50 0.82 microg/mL) assays with no accompanying toxicity seen in the host Daudi cells. No activity was noted for 4.


Subject(s)
Antiviral Agents/chemical synthesis , Guanosine/pharmacology , Herpesvirus 4, Human/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/toxicity , Guanosine/analogs & derivatives , Guanosine/chemical synthesis , Guanosine/toxicity , Humans , Microbial Sensitivity Tests , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured , Viruses/drug effects
13.
Proc Natl Acad Sci U S A ; 96(23): 13300-5, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557315

ABSTRACT

DNA damage generated by oxidant byproducts of cellular metabolism has been proposed as a key factor in cancer and aging. Oxygen free radicals cause predominantly base damage in DNA, and the most frequent mutagenic base lesion is 7,8-dihydro-8-oxoguanine (8-oxoG). This altered base can pair with A as well as C residues, leading to a greatly increased frequency of spontaneous G.C-->T.A transversion mutations in repair-deficient bacterial and yeast cells. Eukaryotic cells use a specific DNA glycosylase, the product of the OGG1 gene, to excise 8-oxoG from DNA. To assess the role of the mammalian enzyme in repair of DNA damage and prevention of carcinogenesis, we have generated homozygous ogg1(-/-) null mice. These animals are viable but accumulate abnormal levels of 8-oxoG in their genomes. Despite this increase in potentially miscoding DNA lesions, OGG1-deficient mice exhibit only a moderately, but significantly, elevated spontaneous mutation rate in nonproliferative tissues, do not develop malignancies, and show no marked pathological changes. Extracts of ogg1 null mouse tissues cannot excise the damaged base, but there is significant slow removal in vivo from proliferating cells. These findings suggest that in the absence of the DNA glycosylase, and in apparent contrast to bacterial and yeast cells, an alternative repair pathway functions to minimize the effects of an increased load of 8-oxoG in the genome and maintain a low endogenous mutation frequency.


Subject(s)
DNA Damage , Guanosine/analogs & derivatives , Mutagens/toxicity , Oxidative Stress , Animals , Base Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatography, High Pressure Liquid , DNA Repair , DNA-Formamidopyrimidine Glycosylase , Electrochemistry , Guanosine/toxicity , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Mutation , N-Glycosyl Hydrolases/genetics
14.
Leuk Lymphoma ; 33(1-2): 147-53, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10194132

ABSTRACT

Fludarabine has shown a definite clinical activity in B-cell chronic lymphocytic leukemia (B-CLL). Recently it has been demonstrated that loxoribine, a guanine ribonucleotide derivative, is able to increase the cytotoxicity of fludarabine in B-CLL cells, in vitro. We have here extended these findings by testing the activity of loxoribine in combination with fludarabine and mafosfamide. As we have previously demonstrated, loxoribine enhances the activity of fludarabine at all concentrations, while only lower doses of mafosfamide seem to be positively affected by loxoribine. The combination of fludarabine and mafosfamide is synergistic on CLL cells, and the cytotoxic activity is increased by the addition of loxoribine. We have also evaluated the pro-apoptotic activity of each drug, both alone and in combination; these results are concordant with the cytotoxicity data, thus demonstrating that, even though loxoribine is more active in combination with fludarabine than with mafosfamide, the efficacy of the triple combination is higher than that obtained with any other agent alone or in double combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/toxicity , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis , Cell Division/drug effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/toxicity , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Female , Guanosine/administration & dosage , Guanosine/analogs & derivatives , Guanosine/toxicity , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Tumor Cells, Cultured , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Vidarabine/toxicity
15.
Melanoma Res ; 8(2): 123-30, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9610864

ABSTRACT

Cystemustine (N'-(2-chloroethyl)-N-(2-(methylsulphonyl)ethyl)-N'-nitrosourea) is a new chloroethylnitrosourea (CENU) being used in phase II clinical trials of disseminated melanoma. Clinical results show that tumour regression has only been observed in 25% of melanomas treated by CENUs. Tumour resistance to CENU is known to be mainly due to a DNA repair protein, O6-methylguanine-DNA methyltransferase (MGMT). The poor remission rate of melanoma with CENUs is attributed to the fact that metastases contain high MGMT levels. Previously, we have shown that O6-benzyl-N2-acetylguanosine (BNAG), an MGMT inhibitor, can be combined with cystemustine by intravenous administration, and increases the antitumour effect of cystemustine in resistant human melanoma. In the work presented here, we investigated the in vitro pharmacological effect of this combination on the DNA of human melanoma cells (M3Dau cells). A quantitative polymerase chain reaction (QPCR) assay was used to measure DNA damage in a fragment (2.7 kb) of the hprt gene. The results show that treatment with BNAG enhances the number of lesions in the DNA of cystemustine-treated resistant malignant melanocytes, which may account for the high tumour-cell toxicity of the combination of cystemustine and BNAG.


Subject(s)
Antineoplastic Agents/toxicity , Guanosine/analogs & derivatives , Nitrosourea Compounds/toxicity , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Cell Survival/drug effects , DNA Damage , Drug Synergism , Enzyme Inhibitors/toxicity , Guanosine/toxicity , Humans , Melanocytes/enzymology , Melanoma , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Polymerase Chain Reaction , Tumor Cells, Cultured
16.
J Med Chem ; 40(18): 2902-9, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9288172

ABSTRACT

A series of O6-(alkyl/aralkyl)guanosines and 2'-deoxyguanosine analogs extended to peracetyl and N2-acetyl derivatives, potentially water soluble, was synthesized. Each was associated with N'-(2-chloroethyl)-N-[2-(methylsulfonyl)ethyl]-N'-nitrosourea for in vitro evaluation on M4Beu melanoma cells of their ability to enhance the cytotoxic effect of this chloroethylnitrosourea, which is frequently reduced by repairs performed by O6-alkylguanine-DNA-alkyltransferase. Structure-activity analysis revealed that (i) benzyl and 4-halobenzyl are the O6-substituents required to afford a significant activity, (ii) 2'-deoxyguanosine derivatives demonstrate greater potency than guanosine analogs, (iii) acetylation, especially at the N2 position, generally results in compounds with moderate ability but may prevent incorporation of such nucleosides into DNA. Accordingly, O6-(4-iodobenzyl)-N2-acetylguanosine (3b) and O6-benzylperacetyl-2'-deoxyguanosine (2a), as well as O6-benzyl-N2-acetylguanosine (1b) and O6-benzyl-N2-acetyl-2'-deoxyguanosine (2b), by far the most water soluble, exhibit a good profile for further in vivo trials by the intravenous route.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Antineoplastic Agents, Alkylating/toxicity , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/toxicity , Ethylnitrosourea/analogs & derivatives , Guanosine/analogs & derivatives , Guanosine/toxicity , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/chemistry , Cell Survival/drug effects , Computer Simulation , Deoxyguanosine/chemical synthesis , Deoxyguanosine/chemistry , Drug Synergism , Ethylnitrosourea/toxicity , Guanosine/chemical synthesis , Guanosine/chemistry , Hydrogen Bonding , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Structure , Static Electricity , Structure-Activity Relationship
17.
Eur J Pharmacol ; 260(2-3): 129-31, 1994 Aug 01.
Article in English | MEDLINE | ID: mdl-7988635

ABSTRACT

Several recent studies suggest that spinal cord levels of cyclic 3',5'-guanosine monophosphate (cGMP) may participate in the development of hyperalgesia. The purpose of this study was to directly evaluate whether cell permeable analogues of cGMP evoke a thermal hyperalgesia (using a hot-plate assay) when administered intrathecally in mice. Our results indicate that two cell permeable forms of cGMP evoke a dose dependent hyperalgesia when administered intrathecally in mice. Additionally, this hyperalgesia was selective since neither non-cell permeant cGMP nor guanosine had any effect on the latency of paw withdrawal when compared to the vehicle injected controls. These data indicate that cGMP is involved in the facilitation of thermal hyperalgesia at the level of the spinal cord.


Subject(s)
Cyclic GMP/analogs & derivatives , Dibutyryl Cyclic GMP/toxicity , Hyperalgesia/chemically induced , Animals , Cell Membrane Permeability , Cyclic GMP/administration & dosage , Cyclic GMP/pharmacokinetics , Cyclic GMP/toxicity , Dibutyryl Cyclic GMP/administration & dosage , Dose-Response Relationship, Drug , Guanosine/administration & dosage , Guanosine/toxicity , Hindlimb , Hot Temperature , Injections, Spinal , Male , Mice
18.
J Biochem Toxicol ; 9(1): 51-7, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8151632

ABSTRACT

Nucleoside phosphotransferase acting on inosine and deoxyinosine has been partially purified from cultured Chinese hamster lung fibroblasts (V79). The activity is associated with a cytosolic 5'-nucleotidase acting on IMP and deoxyIMP. The transfer of the phosphate group from IMP to inosine catalyzed by this enzyme was activated by ATP and 2,3-bisphosphoglycerate. Inosine, deoxyinosine, guanosine, deoxyguanosine, and the nucleoside analogs 2',3'-dideoxyinosine and 8-azaguanosine are substrates, while adenosine and deoxyadenosine are not. IMP, deoxyIMP, GMP, and deoxyGMP are the best phosphate donors. The cytosolic 5'-nucleotidase/phosphotransferase substrate, 8-azaguanosine, was found to be very toxic for cultured fibroblasts (LD50 = 0.32 microM). Mutants resistant to either 8-azaguanosine and the correspondent base 8-azaguanine were isolated and characterized. Our results indicated that the 8-azaguanosine-resistant cells were lacking both cytosolic 5'-nucleotidase and hypoxanthine-guanine phosphoribosyltransferase, while 8-azaguanine resistant cells were lacking only the latter enzyme. Despite this observation, both mutants displayed 8-azaguanosine resistance, thus indicating that cytosolic 5'-nucleotidase is not essential for the activation of this nucleoside analog.


Subject(s)
5'-Nucleotidase/metabolism , Azaguanine/toxicity , Cytosol/enzymology , Fibroblasts/enzymology , Guanosine/analogs & derivatives , Phosphotransferases/metabolism , 5'-Nucleotidase/isolation & purification , Adenosine Triphosphate/metabolism , Animals , Azaguanine/metabolism , Biotransformation , Cell Line , Cells, Cultured , Cricetinae , Cricetulus , Drug Resistance , Fibroblasts/cytology , Fibroblasts/drug effects , Guanosine/metabolism , Guanosine/toxicity , Hypoxanthine Phosphoribosyltransferase/metabolism , Inosine/metabolism , Inosine Monophosphate/metabolism , Male , Mutation , Phosphorylation , Phosphotransferases/isolation & purification
19.
In Vitro Cell Dev Biol Anim ; 29A(5): 362-70, 1993 May.
Article in English | MEDLINE | ID: mdl-8314731

ABSTRACT

Despite more than 50 attempts and the use of various methods, it has been impossible to establish homologous hybridomas between human mature macrophages and 8-azaguanine-resistant U-937 clones prepared in the laboratory. To rule out the possibility that these clones were unsuitable for the selection of hybrids, a study of their properties was done. It was shown that U-937 wild type cells were able to produce HPRT, whereas 8-azaguanine (8-aza)-resistant clones did not. Curiously, exonic and intronic HPRT sequences were undetectable both in wild type and in 8-aza-resistant cell genomes, under conditions where they were detected in control cells. Chromosome analysis of the clone UM9, one of the most frequently used in fusion experiments, revealed many qualitative and quantitative differences with the U-937 wild type cells. 8-aza-resistant U-937 cells were capable of fusion with human macrophages and gave rise to heterokaryons and probably to synkaryons, which survived for weeks without dividing in hypoxanthine-aminopterin-thymidine medium. The results could be interpreted in terms of the existence of a transacting negative regulatory mechanism of the macrophage genome on the proliferative capacity of homospecific hybridomas.


Subject(s)
Chromosome Aberrations , Hybrid Cells/cytology , Macrophages/cytology , Animals , Cell Division/drug effects , Cell Survival/drug effects , Clone Cells , DNA Probes , Drug Resistance , Ethyl Methanesulfonate/toxicity , Guanosine/analogs & derivatives , Guanosine/toxicity , Humans , Hybrid Cells/physiology , Hybrid Cells/ultrastructure , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Karyotyping , Kinetics , Lymphoma, Large B-Cell, Diffuse , Mice , Mutagenesis , Tumor Cells, Cultured
20.
Mutat Res ; 301(1): 45-9, 1993 Jan.
Article in English | MEDLINE | ID: mdl-7677943

ABSTRACT

The frequency of induced micronucleated polychromatic erythrocytes (MNPCEs) was compared in BALB/c, C57BL/6, and DBA/2 mice after intraperitoneal (i.p.) injection of 5-bromodeoxyuridine (BUdR), 5-fluorodeoxyuridine (FUdR), cytosine arabinoside (Ara-C), 6-mercaptopurine (6-MP), 5-bromouracil (5-BU), thymidine (TdR), uridine (UdR), adenosine (AdR) and guanosine (GdR). The experimental procedure was a single i.p. injection followed by harvest at 30 h. The frequency of MNPCEs was significantly increased in all strains by treatment with BUdR, FUdR, Ara-C and 6-MP compared to vehicle control. TdR and UdR induced MNPCEs slightly in BALB/c mice but showed no effect on C57BL/6 and DBA/2 mice. 5-BU, AdR, and GdR did not increase the frequency of MNPCEs in any mouse strain used. These results suggest that BALB/c mice are more susceptible to induction of MNPCEs by clastogenic base analogues and nucleosides than are C57BL/6 or DBA/2 mice.


Subject(s)
Mice/genetics , Micronucleus Tests , Mitogens/toxicity , Nucleosides/toxicity , Purines/toxicity , Pyrimidines/toxicity , Adenine/toxicity , Animals , Bromodeoxyuridine/toxicity , Bromouracil/toxicity , Cytarabine/toxicity , Erythrocytes/drug effects , Female , Floxuridine/toxicity , Guanosine/toxicity , Mercaptopurine/toxicity , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL/genetics , Mice, Inbred DBA/genetics , Species Specificity , Thymidine/toxicity , Uridine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...