Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mutagenesis ; 23(2): 131-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18267949

ABSTRACT

Cisplatin is an extremely effective chemotherapeutic agent used for the treatment of testicular and other solid tumours. It induces a variety of structural modifications in DNA, the most abundant being the GpG- and ApG-1,2-intrastrand cross links formed between adjacent purine bases. These cross links account for approximately 90% of cisplatin-induced DNA damage and are thought to be responsible for the cytotoxic activity of the drug. In human cells, the nucleotide excision repair (NER) process removes the intrastrand cross links from the genome, the efficiency of which is likely to be an important determinant of cisplatin cytotoxicity. We have investigated whether the p53 tumour suppressor status affects global NER of cisplatin-induced intrastrand cross links in human cells. We have used a (32)P-postlabelling method to monitor the removal of GpG- and ApG-intrastrand cross links from two human cell models (the 041TR system, in which p53 is regulated by a tetracycline-inducible promoter, together with WI38 fibroblasts and the SV40-transformed derivative VA13) that each differ in p53 status. We demonstrate that the absence of functional p53 leads to persistence of both cisplatin-induced intrastrand cross links in the genome, suggesting that p53 regulates NER of these DNA lesions. This observation extends the role of p53 in NER beyond enhancing the removal of environmentally induced DNA lesions to include those of clinical origin. Given the frequency of p53 mutations in human tumours, these results may have implications for the use of cisplatin in cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA Adducts/metabolism , DNA Repair/genetics , Tumor Suppressor Protein p53/physiology , Cell Extracts/chemistry , Cell Line, Tumor , DNA/drug effects , Dinucleoside Phosphates/metabolism , Humans , Isotope Labeling , Phosphorus Radioisotopes/chemistry , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/genetics
2.
Pharmacogenet Genomics ; 16(6): 391-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16708048

ABSTRACT

BACKGROUND: N-Acetyltransferases (NATs) and sulfotransferases (SULTs) are key phase II metabolizing enzymes that can be involved both in the detoxification and in the activation of many human promutagens and procarcinogens. METHODS AND RESULTS: We investigated the expression of NATs and SULTs in human prostate and tested their role in the activation the N-hydroxy (N-OH) metabolite of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), a dietary carcinogen, to form DNA adducts. Western blotting showed detectable levels of NAT1, SULT1A1 and SULT1A3 with marked inter-individual variation. NAT2 and other SULT enzymes were not detectable. NAT1 was localized by immunohistochemistry to the cytoplasm of epithelial cells. The presence of acetyl Co-enzyme A (acetyl CoA) and 3'-phosphoadenosine-5'-phosphosulfate (PAPS), NAT and SULT cofactors, respectively, significantly increased the level of DNA adducts, detected by P-postlabelling analysis, in calf thymus DNA incubated with N-OH-IQ and prostate cytosolic fractions. The enhancement in the level of DNA adducts in the presence of PAPS correlated with the level of SULT1A1 protein. A single prostate cytosol with the SULT1A1*2/*2 genotype produced less DNA adducts than cytosols with the *1/*2 and *1/*1 genotypes. No significant correlation was observed between NAT1 protein level and the formation of DNA adducts, even in the presence of acetyl CoA. CONCLUSIONS: In conclusion, we demonstrated that NAT1, SULT1A1 and SULT1A3 are present in human prostate and that both enzyme classes significantly contribute to the activation of N-hydroxylated heterocyclic amines to DNA-damaging species in this tissue. Variation in expression levels, in combination with dietary and/or environmental exposure to carcinogens, could be influential in determining individual susceptibility to prostate cancer.


Subject(s)
Arylamine N-Acetyltransferase/metabolism , Carcinogens/metabolism , Isoenzymes/metabolism , Prostate/enzymology , Sulfotransferases/metabolism , Adult , Arylamine N-Acetyltransferase/analysis , Cytoplasm/metabolism , DNA Adducts/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Immunohistochemistry , Isoenzymes/analysis , Male , Prostate/cytology , Sulfotransferases/analysis , Sulfotransferases/genetics
3.
Int J Cancer ; 119(4): 741-4, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16557573

ABSTRACT

It is controversial whether women have a higher lung cancer susceptibility compared to men. We previously reported higher levels of smoking-related bulky DNA adducts in female lungs. In a pilot study (27 cases), we also found a higher level of female lung cytochrome P4501A1 (CYP1A1) gene expression. In the present extended study we report on the pulmonary expression of several genes involved in polycyclic aromatic hydrocarbon bioactivation in relation to sex, smoking and DNA adducts. CYP1A1, CYP1B1, aryl hydrocarbon receptor and microsomal epoxide hydrolase gene expression was measured by quantitative real-time reverse transcriptase-PCR in 121 normal lung tissue samples. The expression of CYP1A1 and CYP1B1 was significantly higher among current smokers compared to ex-smokers and never-smokers. Among current smokers, females had a 3.9-fold higher median level of CYP1A1 compared to males (p = 0.011). CYP1B1 expression was not related to sex. Lung DNA adducts (measured by 32P-postlabeling) were highly significantly related to CYP1A1 (p < 0.0001) irrespective of smoking-status. Our results are consistent with the hypothesis that CYP1A1 plays a significant role in lung DNA adduct formation and support a higher susceptibility to lung cancer among females.


Subject(s)
DNA Adducts/genetics , Genetic Predisposition to Disease , Lung Neoplasms/genetics , Polycyclic Aromatic Hydrocarbons/pharmacology , Sex Characteristics , Smoking/genetics , Aged , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme System/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Risk Factors , Smoking/adverse effects
4.
Int J Cancer ; 118(11): 2899-902, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16381012

ABSTRACT

DNA mismatch repair (MMR) is essential for repair of single-base mismatches and insertion/deletion loops. MMR proteins also participate in cellular response to DNA damaging agents such as various alkylating agents. Mice deficient in the MMR gene Msh2 develop tumors earlier after exposure to alkylating agents when compared to unexposed mice. The interaction between the MMR system and polycyclic aromatic hydrocarbons such as benzo[a]pyrene (B[a]P) has not been investigated in vivo. Here, we show that treatment of Msh2-deficient mice with B[a]P enhances susceptibility to lymphomagenesis. Carrying at least one intact copy of the Msh2 gene had a protective effect. B[a]P treatment only induced lymphomas in 3 of the 40 (7.5%) mice with at least one intact copy of the Msh2 gene as compared to 13 of the 17 (76.5%) Msh2-deficient mice and occurs only after a much longer time period. The B[a]P-DNA adduct levels measured in lung, liver, spleen and forestomach of B[a]P-treated Msh2-/- mice were not significantly different from B[a]P-treated Msh2+/+ mice. In summary, the results suggest that B[a]P accelerates lymphomagenesis in Msh2-deficient mice. Furthermore, Msh2 deficiency does not have any significant effect on B[a]P-DNA adduct levels.


Subject(s)
Benzo(a)pyrene/toxicity , Lymphoma/chemically induced , MutS Homolog 2 Protein/genetics , Animals , DNA Adducts , DNA Damage , DNA Repair , Female , Lymphoma/genetics , Male , Mice , Mice, Knockout , MutS Homolog 2 Protein/physiology
5.
Mutagenesis ; 20(4): 297-303, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15928012

ABSTRACT

Tamoxifen is an anti-oestrogen widely used in the adjuvant therapy of breast cancer and is also used as a prophylactic to prevent the disease in high-risk women. An increased risk of endometrial cancer has been observed in both settings. In rats, tamoxifen potently induces liver carcinomas and also induces uterine tumours when given neonatally. It forms DNA adducts in rat liver via the formation of alpha-hydroxytamoxifen, the ultimately reactive form being generated by sulfotransferase. In order to investigate the formation of tamoxifen-derived DNA adducts in other rat tissues, female Fischer F344 or Sprague-Dawley rats were treated with tamoxifen or alpha-hydroxytamoxifen by gavage or by intraperitoneal injection, daily for 1, 4 or 7 days, and DNA adducts were detected by (32)P-postlabelling analysis. Tamoxifen formed DNA adducts in the liver but not in other tissues (uterus, stomach, kidney, spleen and colon). alpha-Hydroxytamoxifen also formed adducts at high levels in liver, but with the exception of single animals (1/8) in which a low level of adducts was detected in the stomach in one case, and in the kidney in the other; it also did not give rise to adducts in other tissues. The results suggest that tamoxifen is a genotoxic carcinogen in rat liver, but a non-genotoxic carcinogen in rat uterus, making it, uniquely, a carcinogen with more than one mechanism of action. Mutagenicity experiments conducted in Salmonella typhimurium strains expressing bacterial or human N,O-acetyltransferase did not provide evidence that either alpha-hydroxytamoxifen or alpha-hydroxy-N-desmethyltamoxifen undergoes metabolic activation by acetylation. The confinement of ST2A2, the isozyme of hydroxysteroid sulfotransferase that can activate the compounds, mainly to rat liver is the possible reason for the formation of ducts in the liver but not in other organs of the rat.


Subject(s)
DNA Adducts/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/adverse effects , Acetyltransferases/genetics , Acetyltransferases/metabolism , Animals , Biotransformation , DNA Adducts/analysis , Dose-Response Relationship, Drug , Female , Humans , Injections, Intraperitoneal , Organ Specificity , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Risk Assessment , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Tamoxifen/administration & dosage , Tamoxifen/pharmacokinetics
6.
Environ Pollut ; 138(2): 307-15, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15951082

ABSTRACT

There is a need to develop risk biomarkers during the remediation of contaminated land. We employed the earthworm, Aporrectodea longa (Ude), to determine whether genotoxicity measures could be applied to this organism's intestinal tissues. Earthworms were added, for 24h or 7 days, to soil samples spiked with benzo[a]pyrene (B[a]P) and/or lindane. After exposure, intestinal tissues (crop/gizzard or intestine) were removed prior to the measurement in disaggregated cells of DNA single-strand breaks (SSBs) by the alkaline comet assay. Damage was quantified by comet tail length (CTL, microm). B[a]P 24-h exposure induced dose-related increases (P<0.0001) in SSBs. Earthworm intestine was significantly (P<0.0001) more susceptible than crop/gizzard to B[a]P and/or lindane. However, both tissues appeared to acquire resistance following 7-day exposure. B[a]P-DNA adducts, measured by (32)P-postlabelling, showed a two-adduct-spot pattern. This preliminary investigation suggests that earthworm tissues may be incorporated into genotoxicity assays to facilitate hazard identification within terrestrial ecosystems.


Subject(s)
Benzo(a)pyrene/toxicity , DNA Adducts , DNA Damage , DNA, Single-Stranded , Oligochaeta , Soil Pollutants/toxicity , Animals , Biodegradation, Environmental , Comet Assay , Intestines , Risk Assessment , Toxicity Tests , Toxicogenetics/methods
7.
Mutagenesis ; 20(2): 115-24, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15755801

ABSTRACT

Liver homogenates from rats fed tamoxifen (TAM) in the diet were shared among four different laboratories. TAM-DNA adducts were assayed by high pressure liquid chromatography-electrospray tandem mass spectrometry (HPLC-ES-MS/MS), TAM-DNA chemiluminescence immunoassay (TAM-DNA CIA), and (32)P-postlabeling with either thin layer ((32)P-P-TLC) or liquid chromatography ((32)P-P-HPLC) separation. In the first study, rats were fed a diet containing 500 p.p.m. TAM for 2 months, and the values for measurements of the (E)-alpha-(deoxyguanosin-N(2)-yl)-tamoxifen (dG-N(2)-TAM) adduct in replicate rat livers varied by 3.5-fold when quantified using 'in house' TAM-DNA standards, or other approaches where appropriate. In the second study, rats were fed 0, 50, 250 or 500 p.p.m. TAM for 2 months, and TAM-DNA values were quantified using both 'in house' approaches as well as a newly synthesized [N-methyl-(3)H]TAM-DNA standard that was shared among all the participating groups. In the second study, the total TAM-DNA adduct values varied by 2-fold, while values for the dG-N(2)-TAM varied by 2.5-fold. Ratios of dG-N(2)-TAM:(E)-alpha-(deoxyguanosin-N(2)-yl)-N-desmethyltamoxifen (dG-N(2)-N-desmethyl-TAM) in the second study were approximately 1:1 over the range of doses examined. The study demonstrated a remarkably good agreement for TAM-DNA adduct measurements among the diverse methods employed.


Subject(s)
Antineoplastic Agents, Hormonal/toxicity , DNA Adducts/analysis , DNA Adducts/drug effects , Liver/chemistry , Liver/drug effects , Tamoxifen/toxicity , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/chemistry , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , DNA Adducts/chemistry , Dose-Response Relationship, Drug , Female , Luminescent Measurements , Rats , Rats, Inbred F344 , Spectrometry, Mass, Electrospray Ionization , Tamoxifen/administration & dosage , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry
8.
Methods Mol Biol ; 291: 3-12, 2005.
Article in English | MEDLINE | ID: mdl-15502206

ABSTRACT

32P-Postlabeling analysis is an ultrasensitive method for the detection of DNA adducts, such as those formed directly by the covalent binding of carcinogens and mutagens to bases in DNA, as well as other DNA lesions resulting from modification of bases by endogenous or exogenous agents (e.g., oxidative damage). The procedure involves four main steps: enzymatic digestion of a DNA sample; enrichment of the adducts; radiolabeling of the adducts by T4 kinase-catalyzed transference of 32P-orthophosphate from [gamma-32P]ATP; and chromatographic separation of labeled adducts and detection and quantification by means of their radioactive decay. Using 10 microg or less of DNA, this technique is capable of detecting adduct levels as low as one adduct in 10(9)-10(10) normal nucleotides. It is applicable to a wide range of investigations, including monitoring human exposure to environmental or occupational carcinogens, determining whether a chemical has genotoxic properties, analysis of the genotoxicity of complex mixtures, elucidation of the activation pathways of carcinogens, and monitoring of DNA repair.


Subject(s)
DNA Adducts/analysis , Isotope Labeling/methods , Phosphorus Radioisotopes , Animals , Carcinogenicity Tests , DNA Adducts/chemistry , DNA Damage , Humans , Mutagenicity Tests
9.
Lung Cancer ; 45(3): 289-97, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15301869

ABSTRACT

Epidemiological and biochemical studies have indicated that females may be at greater risk of smoking associated lung cancer compared with males. Among lung cancer patients, female smokers have been found to have higher levels of PAH-related DNA adducts and CYP1A1 gene expression in their normal lung tissue compared to male smokers. A possible role of steroid hormones in these sex differences via interactions between aryl hydrocarbon receptor and estrogen receptor mediated cellular effects has been suggested. In the present study the impact of the estrogen receptor (ERalpha) on CYP1A1 and CYP1B1 gene expression was studied in vitro in human bronchial epithelial cells. Transient transfection of the BEP2D cell line with ERalpha influenced neither constitutive expression of CYP1A1 or CYP1B1 nor induction of these genes by TCDD as measured by real-time RT-PCR. ERalpha had no effect on the constitutive or TCDD-induced enzymatic activity of CYP1A1 (EROD). We also studied the effect of steroid hormones on lung PAH metabolic activation in A/J mice. Intact and ovariectomized female mice were orally exposed to a single dose of benzo[a]pyrene. Ovariectomy did not influence the levels of either benzo[a]pyrene-derived protein or DNA adducts in the lung tissue measured by HPLC and 32P-postlabeling, respectively. In conclusion, the present data do not support the hypothesis of a role of estrogen or the ERalpha in regulating the metabolic activation of polycyclic aromatic hydrocarbons in lung.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1/biosynthesis , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/physiopathology , Polycyclic Aromatic Hydrocarbons/metabolism , Receptors, Estrogen/physiology , Administration, Oral , Animals , Aryl Hydrocarbon Hydroxylases/pharmacology , Benzo(a)pyrene/administration & dosage , Benzo(a)pyrene/toxicity , Cell Culture Techniques , Cytochrome P-450 CYP1A1/pharmacology , Cytochrome P-450 CYP1B1 , DNA, Neoplasm/analysis , Estrogen Receptor alpha , Female , Humans , Mice , Ovariectomy , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
10.
Environ Sci Technol ; 38(13): 3614-22, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15296313

ABSTRACT

Environmental factors are believed to play an important role in cancer aetiology. Whether environmental pollutants act in isolation or in combination within mixtures remains unclear. Four human milk-fat extracts (from resident U.K. women) were screened for levels of organochlorinated and brominated compounds prior to being tested (1-50 mg-equiv) for micronucleus (MN)-forming activity in MCF-7 cells. Using the cytokinesis-block micronucleus assay, micronuclei (MNi) were scored in 1000 binucleate cells per treatment. Cell viability (% plating efficiency) and immunohistochemical detection of p53 induction were also measured. The effects of treatment with 1 mg-equiv of extract in combination with benzo[a]pyrene (BP) were also examined. BP-DNA adducts were detected and quantified by 32P-postlabeling analysis. Dose-related increases in MNi independent of pollutant concentrations were induced by milk-fat extracts. All four extracts elevated the percentage of p53 positive cells, although not always in a dose-related fashion. Some combinations resulted in profound low-dose-induced increases in MNi and significant elevations in the percentage of p53 positive cells, which occurred without further reduction in cell viability or mitotic rate. When one particular extract was combined with BP, a 100-fold increase in BP-DNA adducts was detected as compared with the levels induced by BP alone; an effect not induced by other extracts. This adduct-enhancing extract was fractionated into 14 fractions that were subsequently tested (1 mg-equiv of original extract) in combination with 0.01 microM BP. Fraction 1, into which nonpolar pollutants mostly eluted, enhanced MN-forming activity with BP. Surprisingly, the more polar and less likely to contain fat-soluble pollutants fractions 5 and 8 also enhanced MN-forming activity. No identifiable pollutants were present in these fractions. The results suggest that different environmental pollutants present in human tissue may influence the susceptibility of target cells to initiating events.


Subject(s)
Benzo(a)pyrene/toxicity , Carcinogens, Environmental/toxicity , Hydrocarbons, Brominated/metabolism , Hydrocarbons, Chlorinated/metabolism , Lipid Metabolism , Milk, Human/metabolism , Carcinogens, Environmental/metabolism , Chemical Fractionation , DNA Adducts/drug effects , Dose-Response Relationship, Drug , Female , Gas Chromatography-Mass Spectrometry , Humans , Immunohistochemistry , Micronucleus Tests , Mutagenicity Tests , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , United Kingdom
11.
Chem Res Toxicol ; 17(8): 1092-101, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15310241

ABSTRACT

3-Nitrobenzanthrone (3-NBA) is a suspected human carcinogen found in diesel exhaust and ambient air pollution. The main metabolite of 3-NBA, 3-aminobenzanthrone (3-ABA), was recently detected in the urine of salt mining workers occupationally exposed to diesel emissions. Determining the capability of humans to metabolize 3-ABA and understanding which human enzymes are involved in its activation are important in the assessment of individual susceptibility. We compared the ability of eight human hepatic microsomal samples to catalyze DNA adduct formation by 3-ABA. Using the (32)P-postlabeling method, we found that all hepatic microsomes were competent to activate 3-ABA. DNA adduct patterns with multiple adducts, qualitatively similar to those formed in vivo in rats treated with 3-ABA, were observed. These patterns were also similar to those formed by the nitroaromatic counterpart 3-NBA and which derive from reductive metabolites of 3-NBA bound to purine bases in DNA. The role of specific cytochrome P450s (P450s) in the human hepatic microsomal samples in 3-ABA activation was investigated by correlating the P450-linked catalytic activities in each microsomal sample with the level of DNA adducts formed by the same microsomes. On the basis of this analysis, most of the hepatic microsomal activation of 3-ABA was attributable to P450 1A1 and 1A2 enzyme activity. Inhibition of DNA adduct formation in human liver microsomes by alpha-naphthoflavone and furafylline, inhibitors of P450 1A1 and 1A2, and P450 1A2 alone, respectively, supported this finding. Using recombinant human P450 1A1 and 1A2 expressed in Chinese hamster V79 cells and microsomes of baculovirus-transfected insect cells (Supersomes), we confirmed the participation of these enzymes in the formation of 3-ABA-derived DNA adducts. Moreover, essentially the same DNA adduct pattern found in microsomes was detected in metabolically competent human lymphoblastoid MCL-5 cells expressing P450 1A1 and 1A2. Using rat hepatic microsomes, we showed that both human and rat microsomes lead to the same 3-ABA-derived DNA adducts. Pretreatment of rats with beta-naphthoflavone or Sudan I, inducers of P450 1A1 and 1A2, and P450 1A1 alone, respectively, significantly stimulated the levels of 3-ABA-derived DNA adducts formed by rat liver microsomes. Utilizing purified rat recombinant P450 1A1, the participation of this enzyme in DNA adduct formation by 3-ABA was corroborated. In summary, our results strongly suggest a genotoxic potential of 3-ABA for humans. Moreover, 3-ABA is not only a suitable biomarker of exposure to 3-NBA but may also directly contribute to the high genotoxic potential of 3-NBA.


Subject(s)
Air Pollutants/metabolism , Benz(a)Anthracenes/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , DNA Adducts/metabolism , Microsomes, Liver/enzymology , Animals , Cell Line, Transformed , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Humans , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
J Natl Cancer Inst ; 96(14): 1099-104, 2004 Jul 21.
Article in English | MEDLINE | ID: mdl-15265972

ABSTRACT

BACKGROUND: Although the nonsteroidal antiestrogen tamoxifen is used as an adjuvant chemotherapeutic agent to treat hormone-dependent breast cancer and as a chemopreventive agent in women with elevated risk of breast cancer, it has also been reported to increase the risk of endometrial cancer. Reports of low levels of tamoxifen-DNA adducts in human endometrial tissue have suggested that tamoxifen induces endometrial cancer by a genotoxic mechanism. However, these findings have been controversial. We used electrospray ionization-tandem mass spectrometry (ES-MS/MS) and 32P-postlabeling analyses to investigate the presence of tamoxifen-DNA adducts in human endometrial tissue. METHODS: Endometrial DNA from eight tamoxifen-treated women and eight untreated women was hydrolyzed to nucleosides and assayed for (E)-alpha-(deoxyguanosin-N2-yl)-tamoxifen (dG-Tam) and (E)-alpha-(deoxyguanosin-N2-yl)-N-desmethyltamoxifen (dG-desMeTam), the two major tamoxifen-DNA adducts that have been reported to be present in humans and/or experimental animals treated with tamoxifen, using on-line sample preparation coupled with high-performance liquid chromatography (HPLC) and ES-MS/MS. The same DNA samples were assayed for the presence of dG-Tam and dG-desMeTam by (32)P-postlabeling methodology, using two different DNA digestion and labeling protocols, followed by both thin-layer chromatography and HPLC. RESULTS: We did not detect either tamoxifen-DNA adduct by HPLC-ES-MS/MS analyses (limits of detection for dG-Tam and dG-desMeTam were two adducts per 10(9) nucleotides and two adducts per 10(8) nucleotides, respectively) or by 32P-postlabeling analyses (limit of detection for both adducts was one adduct per 10(9) nucleotides) in any of the endometrial DNA samples. CONCLUSION: The initiation of endometrial cancer by tamoxifen is probably not due to a genotoxic mechanism involving the formation of dG-Tam or dG-desMeTam.


Subject(s)
Antineoplastic Agents, Hormonal/metabolism , DNA Adducts/analysis , Endometrial Neoplasms/chemically induced , Estrogen Receptor Modulators/metabolism , Spectrometry, Mass, Electrospray Ionization , Tamoxifen/metabolism , Adult , Aged , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/adverse effects , Chromatography, High Pressure Liquid , DNA Adducts/isolation & purification , Endometrial Neoplasms/diagnostic imaging , Endometrial Neoplasms/genetics , Estrogen Receptor Modulators/administration & dosage , Estrogen Receptor Modulators/adverse effects , Female , Humans , Middle Aged , Phosphorus Radioisotopes , Radionuclide Imaging , Tamoxifen/administration & dosage , Tamoxifen/adverse effects
13.
Biochem Biophys Res Commun ; 320(2): 297-302, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15219826

ABSTRACT

The nonsteroidal antiestrogen tamoxifen increases the risk of endometrial cancer; however, the mechanism for the induction of these tumors is not known. Recently, Sharma et al. [Biochem. Biophys. Res. Commun. 307 (2003) 157], using high performance liquid chromatography (HPLC) with online postcolumn photochemical activation and fluorescence detection, reported the presence of (E)-alpha-(deoxyguanosin- N2-yl)tamoxifen in DNA from human endometrial explants incubated with tamoxifen. Inasmuch as the methodology used by these investigators does not allow unambiguous characterization of tamoxifen-DNA adducts, we have used two additional techniques (HPLC coupled with electrospray ionization tandem mass spectrometry and 32P-postlabeling analyses) to assay for the presence of tamoxifen-DNA adducts in the human endometrial explant DNA. Tamoxifen-DNA adducts were not detected by either method.


Subject(s)
DNA Adducts/analysis , DNA/drug effects , Endometrium/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Tamoxifen/pharmacology , Chromatography, High Pressure Liquid , Female , Humans , Phosphorus Radioisotopes , Spectrometry, Fluorescence , Tamoxifen/analysis
14.
Chem Res Toxicol ; 17(5): 697-701, 2004 May.
Article in English | MEDLINE | ID: mdl-15144227

ABSTRACT

The antiestrogenic drug tamoxifen forms DNA adducts in rat liver through two genotoxic metabolites, alpha-hydroxytamoxifen and alpha-hydroxy-N-desmethyltamoxifen. These have now each been resolved into R- and S-enantiomers. The work with alpha-hydroxytamoxifen was published earlier [Osborne, et al. (2001) Chem. Res. Toxicol. 14, 888-893]. Here, we publish results with alpha-hydroxy-N-desmethyltamoxifen. We prepared the derivative N-ethoxycarbonyl-N-desmethyltamoxifen-alpha-S-camphanate, separated it into two diastereoisomers, and hydrolyzed them to give (+)- and (-)-alpha-hydroxy-N-desmethyltamoxifen. The configuration of the (-)-isomer was shown to be S- by degradation of the above ester to a derivative of (-)-2-hydroxy-1-phenyl-1-propanone, which has already been shown to have S-configuration. The two enantiomers have the same chemical properties and were equally reactive toward DNA in vitro at pH 6. However, on treatment of rat hepatocytes in culture, R-(+)-alpha-hydroxy-N-desmethyltamoxifen gave 10 times as many DNA adducts as the S-(-)-isomer. This suggests that the R-isomer more readily undergoes sulfate conjugation to generate a reactive carbocation that attacks DNA.


Subject(s)
DNA Adducts/metabolism , Hepatocytes/drug effects , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/pharmacology , Animals , DNA Adducts/chemistry , DNA Adducts/drug effects , Hydrolysis , Liver/metabolism , Rats , Stereoisomerism
15.
Mutat Res ; 560(2): 167-72, 2004 Jun 13.
Article in English | MEDLINE | ID: mdl-15157654

ABSTRACT

Several studies have identified tobacco smoking as a risk factor for anal cancer in both women and men. Samples of anal epithelium from haemorrhoidectomy specimens from current smokers (n = 20) and age-matched life-long non-smokers (n = 16) were analysed for DNA adducts by the nuclease P(1) digestion enhancement procedure of 32P-postlabelling analysis. The study included 14 men and 22 women. Both qualitative and quantitative differences in the adduct profiles were observed between the smokers and non-smokers. The mean adduct level was significantly higher in the smokers than in the non-smokers (1.88 +/- 0.71) (S.D.) versus 1.36 +/- 0.60 adducts per 10(8) nucleotides, P = 0.02, two-tailed unpaired t-test with Welch's correction); furthermore, the adduct pattern seen in two-dimensional chromatograms revealed the smoking-related diagonal radioactive zone in 17/20 smokers, but not in any of the non-smokers (P < 0.00001, Fisher's exact test). These results indicate that components of tobacco smoke inflict genotoxic damage in the anal epithelium of smokers and provide a plausible mechanism for a causal association between smoking and anal cancer.


Subject(s)
Anal Canal/chemistry , DNA Adducts/analysis , Smoking/adverse effects , Adult , Aged , Anus Neoplasms/etiology , Case-Control Studies , Female , Humans , Intestinal Mucosa/chemistry , Male , Middle Aged
16.
Carcinogenesis ; 25(5): 847-55, 2004 May.
Article in English | MEDLINE | ID: mdl-14729593

ABSTRACT

Red meat consumption is associated with endogenous metabolic generation of mutagenic N-nitroso compounds (NOC) and may be implicated in causation of colorectal cancer. Assessment of a biologically relevant dose of NOCs is hampered by imperfect understanding of NOC interactions with other dietary components. This study tests the hypothesis that NOC effects upon mutational biomarkers in mouse colon may be modulated by a non-genotoxic diet-related compound. N-methyl-N-nitrosourea (MNU) and undegraded lambda carrageenan (lambdaCgN) were selected as test chemicals, representing a NOC and a non-genotoxic agent, respectively. Study end-points included (i) DNA adduct formation and (ii) metallothionein (MT) crypt restricted immunopositivity indices (MTCRII) which are considered representative of crypt stem cell mutations. Frequency and size of MT immunopositive foci as well as total number of MT immunopositive crypts were assessed. Biologically effective doses of MNU and lambdaCgN were determined in model validation studies and the agents were then tested alone and in combination. Continuous lambdaCgN treatment for 10 weeks induced significantly greater colonic mucosal injury than a drinking water control. In combined treatment regimens, lambdaCgN treatment did not significantly affect MNU-induced DNA adduct formation. However, combinations of lambdaCgN with MNU significantly increased MTCRII in excess of those induced by MNU alone. Recurrent or continuous lambdaCgN regimens had greater interactive effects with MNU upon MTCRII than short-term lambdaCgN treatment. This study has shown that exposure to a non-genotoxic diet-related compound (lambdaCgN) modulates the effective NOC dosimetry for induction of MT crypt restricted immunopositivity.


Subject(s)
Alkylating Agents/toxicity , Colon/metabolism , Colonic Neoplasms/chemically induced , DNA/drug effects , Diet , Metallothionein/metabolism , Methylnitrosourea/toxicity , Animals , Carrageenan/toxicity , Colon/drug effects , DNA/metabolism , DNA Adducts , Drug Combinations , Feces/chemistry , Female , Metallothionein/immunology , Mice , Mice, Inbred BALB C , Stem Cells/drug effects , Stem Cells/metabolism
17.
Cancer Res ; 63(11): 2752-61, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12782579

ABSTRACT

Determining the capability of humans to metabolize the suspected carcinogen 3-nitrobenzanthrone (3-NBA) and understanding which human enzymes are involved in its activation are important in the assessment of individual susceptibility to this environmental contaminant found in diesel exhaust and ambient air pollution. We compared the ability of eight human hepatic microsomal samples to catalyze DNA adduct formation by 3-NBA. Using two enrichment procedures of the (32)P-postlabeling method, nuclease P1 digestion and butanol extraction, we found that all hepatic microsomes were competent to activate 3-NBA. DNA adduct patterns with multiple adducts, qualitatively similar to those found recently in vivo in rats, were observed. Additionally one major DNA adduct generated by human microsomes was detected. The role of specific cytochromes p450 (p450) and NADPH:p450 reductase in the human hepatic microsomal samples in 3-NBA activation was investigated by correlating the p450- and NADPH: p450 reductase-linked catalytic activities in each microsomal sample with the level of DNA adducts formed by the same microsomes. On the basis of this analysis, most of the hepatic microsomal activation of 3-NBA was attributed to NADPH: p450 reductase. Inhibition of DNA adduct formation in human liver microsomes by alpha-lipoic acid, an inhibitor of NADPH: p450 reductase, supported this finding. Using the purified rabbit enzyme and recombinant human NADPH: p450 reductase expressed in Chinese hamster V79 cells, we confirmed the participation of this enzyme in the formation of 3-NBA-derived DNA adducts. Moreover, essentially the same DNA adduct pattern found in microsomes was detected in metabolically competent human lymphoblastoid MCL-5 cells. The role of individual human recombinant p450s 1A1, 1A2, 1B1, 2A6, 2B6, 2D6, 2C9, 2E1, and 3A4 and of NADPH: p450 reductase in the metabolic activation of 3-NBA, catalyzing DNA adduct formation, was also examined using microsomes of baculovirus-transfected insect cells containing the recombinant enzymes (Supersomes). DNA adducts were observed in all Supersomes preparations, essentially similar to those found with human hepatic microsomes and in human cells. Of all of the recombinant human p450s, p450 2B6 and -2D6 were the most efficient to activate 3-NBA, followed by p450 1A1 and -1A2. These results demonstrate for the first time the potential of human NADPH: p450 reductase and recombinant p450s to contribute to the metabolic activation of 3-NBA by nitroreduction.


Subject(s)
Benz(a)Anthracenes/pharmacokinetics , Environmental Pollutants/pharmacokinetics , NADPH-Ferrihemoprotein Reductase/metabolism , Animals , Benz(a)Anthracenes/metabolism , Biotransformation , Cells, Cultured , Cricetinae , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , DNA Adducts/biosynthesis , Environmental Pollutants/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Lymphocytes/drug effects , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Microsomes, Liver/enzymology , NADPH-Ferrihemoprotein Reductase/antagonists & inhibitors , Oxidation-Reduction , Rabbits , Rats , Rats, Wistar , Recombinant Proteins/metabolism
18.
Biochem Biophys Res Commun ; 300(1): 107-14, 2003 Jan 03.
Article in English | MEDLINE | ID: mdl-12480528

ABSTRACT

Diesel exhaust is known to induce tumours in animals and is suspected of being carcinogenic in humans. Of the compounds found in diesel exhaust, 3-nitrobenzanthrone (3-NBA) is an extremely potent mutagen and suspected human carcinogen forming multiple DNA adducts in vitro. 3-Aminobenzanthrone (3-ABA), 3-acetylaminobenzanthrone (3-Ac-ABA), and N-acetyl-N-hydroxy-3-aminobenzanthrone (N-Ac-N-OH-ABA) were identified as 3-NBA metabolites. In order to gain insight into the pathways of metabolic activation leading to 3-NBA-derived DNA adducts we treated Wistar rats intraperitoneally with 2mg/kg body weight of 3-NBA, 3-ABA, 3-Ac-ABA, or N-Ac-N-OH-ABA and compared DNA adducts present in different organs. With each compound either four or five DNA adduct spots were detected by TLC in all tissues examined (lung, liver, kidney, heart, pancreas, and colon) using the nuclease P1 or butanol enrichment version of the 32P-postlabelling method, respectively. Using HPLC co-chromatographic analysis we showed that all major 3-NBA-DNA adducts produced in vivo in rats are derived from reductive metabolites bound to purine bases and lack an N-acetyl group. Our results indicate that 3-NBA metabolites (3-ABA, 3-Ac-ABA and N-Ac-N-OH-ABA) undergo several biotransformations and that N-hydroxy-3-aminobenzanthrone (N-OH-ABA) appears to be the common intermediate in 3-NBA-derived DNA adduct formation. Therefore, 3-NBA-DNA adducts are useful biomarkers for exposure to 3-NBA and its metabolites and may help to identify enzymes involved in their metabolic activation.


Subject(s)
Benz(a)Anthracenes/toxicity , DNA Adducts/biosynthesis , Environmental Pollutants/toxicity , Animals , Benz(a)Anthracenes/pharmacokinetics , Biomarkers , Biotransformation , Environmental Pollutants/pharmacokinetics , Female , Humans , Rats , Rats, Wistar , Vehicle Emissions/toxicity
19.
Oncogene ; 21(31): 4728-38, 2002 Jul 18.
Article in English | MEDLINE | ID: mdl-12101411

ABSTRACT

Tumour necrosis factor-alpha (TNF-alpha) deficient mice (TNF-alpha(-/-) mice) are resistant to skin carcinogenesis. Cellular signalling via the transcription factor complex AP-1 is thought to play a key role in tumour promotion. The induction of a specific subset of AP-1 responsive genes thought to be important for tumour development, namely GM-CSF, MMP-9 and MMP-3, was suppressed in TNF-alpha(-/-) compared to wild-type mouse skin in response to the tumour promotor TPA. The differential induction of these genes correlated with a temporal shift in AP-1 activation and c-Jun expression in TNF-alpha(-/-) compared to wild-type epidermis. The major receptor for TPA-induced signalling in basal keratinocytes, PKC alpha, was also differentially regulated in wild-type compared with TNF-alpha(-/-) epidermis. A marked delay in TPA-induced intracellular translocation and downregulation of PKC alpha was observed in TNF-alpha(-/-) epidermis, which correlated with the deregulated TPA-induced AP-1 activation and c-Jun expression. The frequency of DNA adduct formation and c-Ha-ras mutations was the same in wild-type and TNF-alpha(-/-) epidermis after DMBA treatment, suggesting that TNF-alpha was not involved in tumour initiation. These data suggest that the pro-inflammatory cytokine TNF-alpha is a critical mediator of tumour promotion, acting via a PKC alpha- and AP-1-dependent pathway. This may be one mechanism by which chronic inflammation increases susceptibility to cancer.


Subject(s)
Carcinogens , Isoenzymes/physiology , Protein Kinase C/physiology , Skin Neoplasms/metabolism , Transcription Factor AP-1/physiology , Tumor Necrosis Factor-alpha/physiology , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Carcinogens/pharmacology , Epidermal Cells , Epidermis/drug effects , Epidermis/metabolism , Keratinocytes/drug effects , Kinetics , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Protein Kinase C-alpha , RNA, Messenger/biosynthesis , Signal Transduction , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...