Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters










Publication year range
1.
Pharmazie ; 70(7): 471-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26373208

ABSTRACT

Human choriocarcinoma has been used as a model to study trophoblast transcellular drug transport in the placenta. Previous models had limitations regarding low molecular weight drug transport through the intracellular gap junction. The purpose of this study was to evaluate placental carrier-mediated transport across a differentiating JEG-3 choriocarcinoma cell (DJEGs) layer model in which the intracellular gap junction was restricted. Cimetidine is the substrate of an efflux transporter, breast cancer resistance protein (BCRP). BCRP highly expressed in the placenta, and its function in the DJEGs model was investigated. In addition, the placental drug transport of another efflux transporter, multidrug resistance-associated proteins (MRPs), and an influx transporter, monocarboxylate transporter (MCT), were examined with various substrates. Cimetidine permeated from the fetal side to the maternal side at significantly high levels and saturated in a dose-dependent manner. The permeability coefficient of a MRP substrate, fluorescein, across the DJEGs model was significantly increased by inhibiting MRP function with probenecid. On the other hand, permeation in the influx direction to the fetal side with a substrate of MCT, valproic acid, had a gentle dose-dependent saturation. These findings suggest that the DJEGs model could be used to evaluate transcellular placental drug transport mediated by major placental transporters.


Subject(s)
Anticonvulsants/pharmacokinetics , Cimetidine/pharmacokinetics , Histamine H2 Antagonists/pharmacokinetics , Placenta/metabolism , Valproic Acid/pharmacokinetics , Adult , Algorithms , Carrier Proteins/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Female , Fluorescein , Humans , In Vitro Techniques , Pregnancy
2.
Eur J Histochem ; 57(1): e1, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23549458

ABSTRACT

Inorganic arsenic is a well-known human skin carcinogen. Chronic arsenic exposure results in various types of human skin lesions, including squamous cell carcinoma (SCC). To investigate whether mutant stem cells participate in arsenic-associated carcinogenesis, we repeatedly exposed the HaCaT cells line to an environmentally relevant level of arsenic (0.05 ppm) in vitro for 18 weeks. Following sodium arsenic arsenite administration, cell cycle, colony-forming efficiency (CFE), cell tumorigenicity, and expression of CD44v6, NF-κB and p53, were analyzed at different time points (0, 5, 10, 15, 20, 25 and 30 passages). We found that a chronic exposure of HaCaT cells to a low level of arsenic induced a cancer stem- like phenotype. Furthermore, arsenic-treated HaCaT cells also became tumorigenic in nude mice, their growth cycle was predominantly in G2/M and S phases. Relative to nontreated cells, they exhibited a higher growth rate and a significant increase in CFE. Western blot analysis found that arsenic was capable of increasing cell proliferation and sprouting of cancer stem-like phenotype. Additionally, immunohistochemical analysis demonstrated that CD44v6 expression was up-regulated in HaCaT cells exposed to a low level of arsenic during early stages of induction. The expression of CD44v6 in arsenic-treated cells was positively correlated with their cloning efficiency in soft agar (r=0.949, P=0.01). Likewise, the expressions of activating transcription factor NF-κB and p53 genes in the arsenic-treated HaCaT cells were significantly higher than that in non-treated cells. Higher expressions of CD44v6, NF-κB and p53 were also observed in tumor tissues isolated from Balb/c nude mice. The present results suggest that CD44v6 may be a biomarker of arsenic-induced neoplastic transformation in human skin cells, and that arsenic promotes malignant transformation in human skin lesions through a NF-κB signaling pathway-stimulated expression of CD44v6.


Subject(s)
Arsenites/adverse effects , Carcinoma, Squamous Cell , Cell Transformation, Neoplastic , Enzyme Inhibitors/adverse effects , Hyaluronan Receptors/biosynthesis , Keratinocytes , Skin Neoplasms , Skin , Sodium Compounds/adverse effects , Animals , Arsenic/adverse effects , Arsenic/pharmacology , Arsenites/pharmacology , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Inhibitors/pharmacology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Skin/metabolism , Skin/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Sodium Compounds/pharmacology , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
3.
Opt Lett ; 35(10): 1497-9, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20479787

ABSTRACT

Optical parametric chirped-pulse amplification (OPCPA) operation with low gain by seeding with high-energy, clean pulses is shown to significantly improve the contrast to better than 10(-10) to 10(-11) in a high-intensity Ti:sapphire laser system that is based on chirped-pulse amplification. In addition to the high-contrast broadband, high-energy output from the final amplifier is achieved with a flat-topped spatial profile of filling factor near 77%. This is the result of pump beam spatial profile homogenization with diffractive optical elements. Final pulse energies exceed 30 J, indicating capability for reaching peak powers in excess of 500 TW.


Subject(s)
Aluminum Oxide , Lasers , Titanium , Time Factors
4.
J Viral Hepat ; 15(7): 498-507, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18331251

ABSTRACT

8-Hydroxydeoxyguanosine (8-OHdG) is a promutagenic DNA lesion produced by hydroxyl radicals and is recognized as a useful marker in estimating DNA damage induced by oxidative stress. The aim of this study was to clarify the clinical significance of hepatic 8-OHdG levels in patients with chronic viral hepatitis. Hepatic 8-OHdG accumulation was investigated in patients with chronic hepatitis C (CH-C) (n = 77) and chronic hepatitis B (CH-B) (n = 34) by immunohistochemical staining of liver biopsy samples. 8-OHdG positive hepatocytes were significantly higher in patients with CH-C compared to CH-B (median 55.0 vs 18.8 cells/10(5) mum(2), P < 0.0001). The number of positive hepatocytes significantly increased with the elevation of serum aminotransferase levels, especially in CH-C patients (8-OHdG vs alanine aminotransferase (ALT)/aspartate aminotrasferase (AST) were r = 0.738/0.720 in CH-C and 0.506/0.515 in CH-B). 8-OHdG reactivity was strongly correlated with body and hepatic iron storage markers in CH-C (vs serum ferritin, r = 0.615; vs hepatic total iron score, r = 0.520; vs hepatic hepcidin mRNA levels, r = 0.571), although it was related to serum HBV-DNA titers (r = 0.540) and age of patients (r = -0.559) in CH-B. These results indicate that hepatic oxidative DNA damage is common in chronic viral hepatitis, in particular chronic HCV-infected patients, suggesting a possible link between chronic hepatic inflammation and hepatocarcinogenesis. The strong positive correlation between hepatic DNA damage and iron overload suggests that iron content is one of the most likely mediators of hepatic oxidative stress and iron reduction may be beneficial to reduce the incidence of hepatic cancer in CH-C patients.


Subject(s)
DNA Damage , Deoxyguanosine/analogs & derivatives , Hepatitis B, Chronic/metabolism , Hepatitis C, Chronic/metabolism , Iron Overload/metabolism , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Deoxyguanosine/metabolism , Female , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/genetics , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged
5.
Br J Cancer ; 98(3): 580-6, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18231107

ABSTRACT

Although the oxidative stress frequently occurs in patients with chronic hepatitis C, its role in future hepatocellular carcinoma (HCC) development is unknown. Hepatic 8-hydroxydeoxyguanosine (8-OHdG) was quantified using liver biopsy samples from 118 naïve patients who underwent liver biopsy from 1995 to 2001. The predictability of 8-OHdG for future HCC development and its relations to epidemiologic, biochemical and histological baseline characteristics were evaluated. During the follow-up period (mean was 6.7+/-3.3 years), HCC was identified in 36 patients (30.5%). Univariate analysis revealed that 16 variables, including 8-OHdG counts (65.2+/-20.2 vs 40.0+/-23.5 cells per 10(5) microm2, P<0.0001), were significantly different between patients with and without HCC. Cox proportional hazard analysis showed that the hepatic 8-OHdG (P=0.0058) and fibrosis (P=0.0181) were independent predicting factors of HCC. Remarkably, 8-OHdG levels were positively correlated with body and hepatic iron storage markers (vs ferritin, P<0.0001 vs hepatic iron score, P<0.0001). This study showed that oxidative DNA damage is associated with increased risk for HCC and hepatic 8-OHdG levels are useful as markers to identify the extreme high-risk subgroup. The strong correlation between hepatic DNA damage and iron overload suggests that the iron content may be a strong mediator of oxidative stress and iron reduction may reduce HCC incidence in patients with chronic hepatitis C.


Subject(s)
Carcinoma, Hepatocellular/etiology , DNA Damage , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/metabolism , Liver Neoplasms/etiology , Liver/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Female , Humans , Male , Middle Aged
6.
Opt Express ; 11(13): 1537-40, 2003 Jun 30.
Article in English | MEDLINE | ID: mdl-19466027

ABSTRACT

We demonstrate the generation of symmetrical supercontinuum of over 40 nm in the 1.55 m region (1540 - 1580 nm) by injecting 1562 nm, 2.2 ps, 40 GHz optical pulses into a 200 m-long, dispersion-flattened polarization-maintaining photonic crystal fiber. The chromatic dispersion and dispersion slope of the fiber at 1.55 m are -0.23 ps/km/nm and 0.01 ps/km/nm2, respectively. This is the first report of 1.55 m band supercontinuum generation in a dispersion-flattened and polarization-maintaining photonic crystal fiber.

7.
Free Radic Res ; 34(6): 595-604, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11697035

ABSTRACT

The mechanism of metal-mediated DNA damage by carcinogenic danthron (1,8-dihydroxyanthraquinone) and anthraquinone was investigated by the DNA sequencing technique using 32P-labeled human DNA fragments obtained from the human c-Ha-ras-1 protooncogene and the p53 tumor suppressor gene. Danthron caused DNA damage particularly at guanines in the 5'-GG-3', 5'-GGGG-3', 5'-GGGGG-3' sequences (damaged bases are underlined) in the presence of Cu(II), cytochrome P450 reductase and the NADPH-generating system. The DNA damage was inhibited by catalase and bathocuproine, suggesting the involvement of H2O2 and Cu(I). The formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine increased with increasing concentration of danthron. On the other hand, carcinogenic anthraquinone induced less oxidative DNA damage than danthron. Electron spin resonance study showed that the semiquinone radical could be produced by P450 reductase plus NADPH-mediated reduction of danthron, while little signal was observed with anthraquinone. These results suggest that danthron is much more likely to be reduced by P450 reductase and generate reactive oxygen species through the redox cycle, leading to more extensive Cu(II)-mediated DNA damage than anthraquinone. In the case of anthraquinone, its hydroxylated metabolites with similar reactivity to danthron may participate in DNA damage in vivo. We conclude that oxidative DNA damage by danthron and anthraquinone seems to be relevant for the expression of their carcinogenicity.


Subject(s)
Anthraquinones/pharmacology , Carcinogens/pharmacology , Copper/metabolism , DNA Damage , NADPH-Ferrihemoprotein Reductase/metabolism , NADP/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Cations, Divalent , Chelating Agents/pharmacology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/biosynthesis , Free Radical Scavengers/pharmacology , Metals , Phenanthrolines/pharmacology
8.
Jpn J Cancer Res ; 92(11): 1166-74, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714440

ABSTRACT

Hydroxyurea is a chemotherapeutic agent used for the treatment of myeloproliferative disorders (MPD) and solid tumors. The mutagenic and carcinogenic potential of hydroxyurea has not been established, although hydroxyurea has been associated with an increased risk of leukemia in MPD patients. To clarify whether hydroxyurea has potential carcinogenicity, we examined site-specific DNA damage induced by hydroxyurea using (32)P-5'-end-labeled DNA fragments obtained from the human p53 and p16 tumor suppressor genes and the c-Ha-ras-1 protooncogene. Hydroxyurea caused Cu(II)-mediated DNA damage especially at thymine and cytosine residues. NADH efficiently enhanced hydroxyurea-induced DNA damage. The DNA damage was almost entirely inhibited by catalase and bathocuproine, a Cu(I)-specific chelator, suggesting the involvement of hydrogen peroxide (H(2)O(2)) and Cu(I). Typical free hydroxyl radical scavengers did not inhibit DNA damage by hydroxyurea, but methional did. These results suggest that crypto-hydroxyl radicals such as Cu(I)-hydroperoxo complex (Cu(I)-OOH) cause DNA damage. Formation of 8-hydroxy-2'-deoxyguanosine (8-OHdG) was induced by hydroxyurea in the presence of Cu(II). An electron spin resonance spectroscopic study using N-(dithiocarboxy)sarcosine as a nitric oxide (NO)-trapping reagent demonstrated that NO was generated from hydroxyurea in the presence and absence of catalase. In addition, the generation of formamide was detected by both gas chromatography-mass spectrometry (GC-MS) and time-of-flight-mass spectrometry (TOF-MS). A high concentration of hydroxyurea induced depurination at DNA bases in an H(2)O(2)-independent manner, and endonuclease IV treatment led to chain cleavages. These results suggest that hydroxyurea could induce base oxidation as the major pathway of DNA modification and depurination as a minor pathway. Therefore, it is considered that DNA damage by hydroxyurea participates in not only anti-cancer activity, but also carcinogenesis.


Subject(s)
DNA Damage/drug effects , Hydrogen Peroxide/metabolism , Hydroxyurea/pharmacology , Nitric Oxide/metabolism , Animals , Catalase/metabolism , Cations, Divalent/pharmacology , Cattle , Copper/pharmacology , DNA/drug effects , DNA/genetics , DNA/metabolism , Electron Spin Resonance Spectroscopy , Free Radical Scavengers/pharmacology , Gas Chromatography-Mass Spectrometry , Genes, p16 , Genes, p53/genetics , Genes, ras/genetics , Humans , NAD/metabolism , Phenanthrolines/pharmacology , Substrate Specificity
9.
Biochem Biophys Res Commun ; 288(3): 552-7, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11676478

ABSTRACT

Phytic acid (myo-inositol hexaphosphate) is one of the most promising cancer chemopreventive agents. We investigated the mechanism by which phytic acid expresses preventive action to cancer. Phytic acid inhibited the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine in cultured cells treated with an H2O2-generating system, although it did not scavenge H2O2. Site-specific DNA damage by H2O2 and Cu(II) at GG and GGG sequences was inhibited by phytic acid, but not by myo-inositol. Phytic acid alone did not cause DNA damage and thus, it should not act as a prooxidant. We conclude that phytic acid acts as an antioxidant to inhibit the generation of reactive oxygen species from H2O2 by chelating metals, resulting in chemoprevention of cancer.


Subject(s)
DNA Damage , DNA/drug effects , Hydrogen Peroxide/pharmacology , Phytic Acid/pharmacology , Protective Agents/pharmacology , Binding Sites , Chemoprevention , Copper/pharmacology , DNA/metabolism , Deoxyadenosines/metabolism , Drug Interactions , Genes, ras/genetics , Glucose Oxidase/pharmacology , HL-60 Cells , Humans , Neoplasms/prevention & control , Oxidation-Reduction , Phytic Acid/therapeutic use , Protective Agents/therapeutic use , Tumor Suppressor Protein p53/genetics
10.
Chem Res Toxicol ; 14(10): 1421-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11599934

ABSTRACT

Acrylonitrile (ACN) is widely used as a monomer in the polymer industry. Studies on carcinogenicity in rats exposed to ACN showed increased incidences of tumors including glial cell tumors of central nervous system and increased production of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) in glial cells. Using a high performance liquid chromatograph equipped with an electrochemical detector, we revealed that ACN enhanced the formation of 8-oxo-dG induced by H2O2 and Cu(II) whereas ACN itself did not cause DNA damage. The enhancing effect of ACN was much more efficient in the double-stranded DNA than that in the single-stranded DNA. Experiments with 32P-labeled DNA revealed that addition of ACN enhanced the site-specific DNA damage at guanines, particularly at 5'-site of the GG and GGG sequences while H2O2/Cu(II) induced piperidine-labile sites at thymine, cytosine, and guanine residues. An electron spin resonance spectroscopy using alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone showed that a nitrogen-centered radical was generated from ACN in the presence of H2O2 and Cu(II). It is considered that ACN enhances H2O2-mediated DNA damage via nitrogen-centered radical formation. We will discuss the mechanism of the enhancing effect on oxidative DNA damage in relation to expression of ACN carcinogenicity.


Subject(s)
Acrylonitrile/adverse effects , Carcinogens/adverse effects , Copper/adverse effects , DNA Adducts , DNA Damage , Hydrogen Peroxide/adverse effects , Oxidants/adverse effects , Acrylonitrile/chemistry , Animals , Carcinogens/chemistry , Cattle , Chromatography, High Pressure Liquid , Electron Spin Resonance Spectroscopy , Guanine/chemistry , Hydrogen Peroxide/chemistry , Oxidants/chemistry , Thymus Gland
11.
Mutat Res ; 479(1-2): 101-11, 2001 Aug 08.
Article in English | MEDLINE | ID: mdl-11470485

ABSTRACT

2-Nitropropane (2-NP), a widely used industrial solvent, is carcinogenic to rats. To clarify the mechanism of carcinogenesis by 2-NP, we investigated DNA damage by 2-NP metabolites, N-isopropylhydroxylamine (IPHA) and hydroxylamine-O-sulfonic acid (HAS), using 32P-5'-end-labelled DNA fragments obtained from genes that are relevant to human cancer. In the presence of Fe(III) EDTA, both IPHA and HAS caused DNA damage at every nucleotide position without marked site preference. The damage was inhibited by free hydroxyl radical (-*OH) scavengers, catalase and deferoxamine mesilate, an iron chelating agent. These results suggest that the DNA damage was caused by -*OH generated via H(2)O(2) by both IPHA and HAS. In contrast, in the presence of Cu(II), IPHA frequently caused DNA damage at thymine. The Cu(II)-mediated DNA damage caused by IPHA was inhibited by catalase, methional and bathocuproine, a Cu(I)-specific chelator, suggesting the involvement of H(2)O(2) and Cu(I). These results suggest that the DNA damage induced by IPHA in the presence of Cu(II) was caused by a reactive oxygen species like the Cu(I)-hydroperoxo complex. On the other hand, HAS most frequently induced DNA damage at 5'-TG-3', 5'-GG-3' and 5'-GGG-3' sequences. Catalase and methional only partly inhibited the Cu(II)-mediated DNA damage caused by HAS, suggesting that the reactive oxygen species and another reactive species participate in this process. Formation of 8-oxodG by IPHA or HAS increased in the presence of metal ions. This study suggests that metal-mediated DNA damage caused by 2-NP metabolites plays an important role in the mutagenicity and the carcinogenicity of 2-NP.


Subject(s)
Carcinogens , DNA Damage , Deoxyguanosine/analogs & derivatives , Metals/toxicity , Nitroparaffins , Propane/analogs & derivatives , 8-Hydroxy-2'-Deoxyguanosine , Catalase/metabolism , Catalase/pharmacology , Chelating Agents/pharmacology , DNA/drug effects , Deferoxamine/pharmacology , Deoxyguanosine/biosynthesis , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Free Radical Scavengers , Free Radicals , Humans , Hydroxylamines/pharmacology , Ions , Iron/metabolism , Models, Chemical , Mutagens , Oxygen/metabolism
12.
Carcinogenesis ; 22(8): 1239-45, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11470755

ABSTRACT

Catechol, a naturally occurring and an important industrial chemical, has been shown to have strong promotion activity and induce glandular stomach tumors in rodents. In addition, catechol is a major metabolite of carcinogenic benzene. To clarify the carcinogenic mechanism of catechol, we investigated DNA damage using human cultured cell lines and 32P-labeled DNA fragments obtained from the human p53 and p16 tumor suppressor genes and the c-Ha-ras-1 proto-oncogene. Catechol increased the amount of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG), which is known to be correlated with the incidence of cancer, in a human leukemia cell line HL-60, whereas the amount of 8-oxodG in its hydrogen peroxide (H2O2)-resistant clone HP100 was not increased. The formation of 8-oxodG in calf thymus DNA was increased by catechol in the presence of Cu(2+). Catechol caused damage to 32P-labeled DNA fragments in the presence of Cu(2+). When NADH was added, DNA damage was markedly enhanced and clearly observed at relatively low concentrations of catechol (<1 microM). DNA cleavage was enhanced by piperidine treatment, suggesting that catechol plus NADH caused not only deoxyribose phosphate backbone breakage but also base modification. Catechol plus NADH frequently modified thymine residues. Bathocuproine, a specific Cu(+) chelator and catalase inhibited the DNA damage, indicating the participation of Cu(+) and H2O2 in DNA damage. Typical hydroxyl radical scavengers did not inhibit catechol plus Cu(2+)-induced DNA damage, whereas methional completely inhibited it. These results suggest that reactive species derived from the reaction of H2O2 with Cu(+) participates in catechol-induced DNA damage. Therefore, we conclude that oxidative DNA damage by catechol through the generation of H2O2 plays an important role in the carcinogenic process of catechol and benzene.


Subject(s)
Carcinogens/pharmacology , Catechols/pharmacology , DNA Damage , DNA/drug effects , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Animals , Catalase/pharmacology , Cattle , Cell Line , Copper/pharmacology , DNA Damage/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Free Radical Scavengers/pharmacology , Humans , Hydroxyl Radical , NAD/pharmacology , Phenanthrolines/pharmacology , Proto-Oncogene Mas
13.
Free Radic Biol Med ; 31(1): 108-16, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11425496

ABSTRACT

DNA damage in cultured cells and in lungs of rats induced by nickel compounds was investigated to clarify the mechanism of nickel carcinogenesis. DNA strand breaks in cultured cells exposed to nickel compounds were measured by using a pulsed field gel electrophoresis technique. Among nickel compounds (Ni(3)S(2), NiO (black), NiO (green), and NiSO(4)), only Ni(3)S(2), which is highly carcinogenic, induced lesions of both double- and single-stranded DNA in cultured human cells (Raji and HeLa cells). Treatment of cultured HeLa cells with Ni(3)S(2) (10 microg/ml) induced a 1.5-fold increase in 8-hydroxy-2'-deoxyguanosine (8-OH-dG) compared with control, whereas NiO (black), NiO (green), and NiSO(4) did not enhance the generation of 8-OH-dG. Intratracheal instillation of Ni(3)S(2), NiO(black), and NiO(green) to Wistar rats increased 8-OH-dG in the lungs significantly. NiSO(4) induced a smaller but significant increase in 8-OH-dG. Histological studies showed that all the nickel compounds used induced inflammation in lungs of the rats. Nitric oxide (NO) generation in phagocytic cells induced by Ni(3)S(2), NiO(black), and NiO(green) was examined using macrophage cell line RAW 264.7 cells. NO generation in RAW 264.7 cells stimulated with lipopolysaccharide was enhanced by all nickel particles. Two mechanisms for nickel-induced oxidative DNA damage have been proposed as follows: all the nickel compounds used induced indirect damage through inflammation, and Ni(3)S(2) also showed direct oxidative DNA damage through H(2)O(2) formation. This double action may explain relatively high carcinogenic risk of Ni(3)S(2).


Subject(s)
Carcinogens/toxicity , DNA Damage/drug effects , Lung/drug effects , Nickel/toxicity , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cattle , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Electrophoresis, Gel, Pulsed-Field , Humans , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Oxidative Stress , Rats , Rats, Wistar
14.
Chem Res Toxicol ; 14(6): 678-85, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11409938

ABSTRACT

Potassium bromate (KBrO3), a food additive, induces renal-cell tumors in rats. KBrO3 induced 8-oxo-7, 8-dihydro-2'-deoxyguanosine (8-oxodG) formation in human leukemia cell line HL-60 as well as in its H2O2-resistant clone, HP100, suggesting no involvement of H2O2. Depletion of GSH by buthionine sulfoximine (BSO) had a little inhibitory effect on KBrO3-induced 8-oxodG formation. However, the amount of 8-oxodG was still significantly higher than that in control, suggesting that intracellular Cys can affect KBrO3 to oxidize DNA, when GSH decreased. KBrO3 caused 8-oxodG in isolated DNA in the presence of GSH (tripeptide; gamma-GluCysGly), gamma-GluCys, CysGly, or Cys. Methional completely inhibited 8-oxodG formation induced by KBrO3 plus GSH, but typical hydroxyl radical scavengers, SOD and catalase, had little or no inhibitory effects. When bromine solution (BrO(-)) was used instead of BrO3(-), similar scavenger effects were observed. Experiments with 32P-labeled DNA fragments obtained from the human p53 tumor suppressor gene and the c-Ha-ras-1 protooncogene suggested that KBrO3 induced 8-oxodG formation at 5'-site guanine of GG and GGG sequences of double-stranded DNA in the presence of GSH and that treatment of formamidopyrimidine-DNA glycosylase led to chain cleavages at the guanine residues. ESR spin-trapping studies showed that 1:2:2:1 quartet DMPO (5,5-dimethyl-1-pyrroline N-oxide) spectrum similar to DMPO/hydroxy radical (*OH) adduct, but the signals were not inhibited by ethanol. Therefore, the signal seemed not to be due to *OH but byproduct due to oxidation of DMPO by the reactive species. The signals were suppressed by the addition of dGMP, but not by other mononucleotides, suggesting the specific reactivity with guanine. On the basis of our results and previous literature, it is speculated that reduction of KBrO3 by SH compounds in renal proximal tubular cells yields bromine oxides and bromine radicals, which are the reactive species that cause guanine oxidation, leading to renal carcinogenesis of KBrO3.


Subject(s)
Bromates/adverse effects , Carcinogens/adverse effects , Cysteine/chemistry , DNA Damage , Food Additives/adverse effects , Glutathione/chemistry , Guanine/chemistry , Kidney Neoplasms/chemically induced , Animals , Antineoplastic Agents/chemistry , Cell Transformation, Neoplastic , Deuterium/chemistry , Free Radicals/adverse effects , Genes, p53/genetics , Genes, ras/genetics , HL-60 Cells , Humans , Rats , Spin Trapping
15.
Mutat Res ; 494(1-2): 63-72, 2001 Jul 25.
Article in English | MEDLINE | ID: mdl-11423346

ABSTRACT

Norharman (9H-pyrido[3,4-b]indole), which is a heterocyclic amine included in cigarette smoke or cooked foodstuffs, is not mutagenic itself. However, norharman reacts with non-mutagenic aniline to form mutagenic aminophenylnorharman (APNH), of which DNA adducts formation and hepatocarcinogenic potential are pointed out. We investigated whether N-OH-APNH, an N-hydroxy metabolite of APNH, can cause oxidative DNA damage or not, using 32P-labeled DNA fragments. N-OH-APNH caused Cu(II)-mediated DNA damage. When an endogenous reductant, beta-nicotinamide adenine dinucleotide (NADH) was added, the DNA damage was greatly enhanced. Catalase and a Cu(I)-specific chelator inhibited DNA damage, suggesting the involvement of H(2)O(2) and Cu(I). Typical -*OH scavenger did not inhibit DNA damage. These results suggest that the main reactive species are probably copper-hydroperoxo complexes with DNA. We also measured 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) formation by N-OH-APNH in the presence of Cu(II), using an electrochemical detector coupled to a high-pressure liquid chromatograph. Addition of NADH greatly enhanced 8-oxodG formation. UV-VIS spectra and mass spectra suggested that N-OH-APNH was autoxidized to nitrosophenylnorharman (NO-PNH). We speculated that NO-PNH was reduced by NADH. Cu(II) facilitated the redox cycle. In the presence of NADH and Cu(II), very low concentrations of N-OH-APNH could induce DNA damage via redox reactions. We conclude that oxidative DNA damage, in addition to DNA adduct formation, may play an important role in the expression of genotoxicity of APNH.


Subject(s)
Aniline Compounds/metabolism , DNA Damage , Harmine/metabolism , Indoles/metabolism , Indoles/toxicity , Mutagens/toxicity , Pyridines/metabolism , Pyridines/toxicity , 8-Hydroxy-2'-Deoxyguanosine , Aniline Compounds/chemistry , Carbolines , Copper/metabolism , DNA/chemistry , DNA/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Harmine/analogs & derivatives , Harmine/chemistry , Models, Chemical , NAD/metabolism , Oxidation-Reduction
16.
FEBS Lett ; 495(3): 187-90, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11334889

ABSTRACT

Superoxide dismutases (SODs) are involved in the protection of cells from oxygen toxicity. However, several papers have reported that the overexpression of CuZn-SOD causes oxidative damage to cells. We investigated a mechanism by which an excess of SODs accelerates oxidative stress. The presence of CuZn-SOD, Mn-SOD or Mn(II) enhanced the frequency of DNA damage induced by hydrogen peroxide (H2O2) and Cu(II), and altered the site specificity of the latter: H2O2 induced Cu(II)-dependent DNA damage with high frequency at the 5'-guanine of poly G sequences; when SODs were added, the frequency of cleavages at thymine and cytosine residues increased. SODs also enhanced the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine by H2O2 and Cu(II). We conclude that SODs may increase carcinogenic risks, e.g. of tumors in Down syndrome.


Subject(s)
DNA Damage/drug effects , Deoxyguanosine/analogs & derivatives , Hydrogen Peroxide/pharmacology , Superoxide Dismutase/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cattle , Copper/chemistry , Deoxyguanosine/biosynthesis , Drug Synergism , Manganese/chemistry , Substrate Specificity , Zinc/chemistry
17.
Arch Biochem Biophys ; 389(1): 49-56, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11370671

ABSTRACT

The mechanism of DNA damage by a metabolite of the carcinogen o-anisidine in the presence of metals was investigated by the DNA sequencing technique using 32P-labeled human DNA fragments. The o-anisidine metabolite, o-aminophenol, caused DNA damage in the presence of Cu(II). The DNA damage was inhibited by catalase and bathocuproine, suggesting the involvement of H2O2 and Cu(I). The formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine by o-aminophenol increased in the presence of Cu(II). We conclude that Cu(II)-mediated oxidative DNA damage by this o-anisidine metabolite seems to be relevant for the expression of the carcinogenicity of o-anisidine. o-Aminophenol plus Cu(II) caused preferential DNA damage at the 5'-site guanine of GG and GGG sequences. When CuZn-SOD or Mn-SOD was added, the DNA damage was enhanced and its predominant cleavage sites were changed into thymine and cytosine residues. We consider that SOD may increase the frequency of mutations due to DNA damage induced by o-aminophenol and thus increase its carcinogenic potential.


Subject(s)
Aminophenols/pharmacology , Aniline Compounds/metabolism , Carcinogens/metabolism , DNA Damage , Superoxide Dismutase/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Aminophenols/metabolism , Animals , Catalase/pharmacology , Cattle , Chelating Agents/pharmacology , Copper/metabolism , Cytosine/chemistry , DNA/chemistry , DNA/drug effects , DNA Damage/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/biosynthesis , Deoxyguanosine/chemistry , Electron Spin Resonance Spectroscopy , Free Radical Scavengers/pharmacology , Genes, ras/drug effects , Genes, ras/genetics , Guanine/chemistry , Humans , Manganese/metabolism , Manganese/pharmacology , Oxidation-Reduction/drug effects , Phenanthrolines/pharmacology , Superoxide Dismutase/pharmacology , Thymine/chemistry , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics
18.
Biochemistry ; 40(15): 4763-8, 2001 Apr 17.
Article in English | MEDLINE | ID: mdl-11294644

ABSTRACT

Telomere shortening is associated with cellular senescence. We investigated whether UVA, which contributes to photoaging, accelerates telomere shortening in human cultured cells. The terminal restriction fragment (TRF) from WI-38 fibroblasts irradiated with UVA (365-nm light) decreased with increasing irradiation dose. Furthermore, UVA irradiation dose-dependently increased the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) in both WI-38 fibroblasts and HL-60 cells. To clarify the mechanism of the acceleration of telomere shortening, we investigated site-specific DNA damage induced by UVA irradiation in the presence of endogenous photosensitizers using (32)P 5'-end-labeled DNA fragments containing the telomeric oligonucleotide (TTAGGG)(4). UVA irradiation with riboflavin induced 8-oxodG formation in the DNA fragments containing telomeric sequence, and Fpg protein treatment led to chain cleavages at the central guanine of 5'-GGG-3' in telomere sequence. The amount of 8-oxodG formation in DNA fragment containing telomere sequence [5'-CGC(TTAGGG)(7)CGC-3'] was approximately 5 times more than that in DNA fragment containing nontelomere sequence [5'-CGC(TGTGAG)(7)CGC-3']. Catalase did not inhibit this oxidative DNA damage, indicating no or little participation of H(2)O(2) in DNA damage. These results indicate that the photoexcited endogenous photosensitizer specifically oxidizes the central guanine of 5'-GGG-3' in telomere sequence to produce 8-oxodG probably through an electron-transfer reaction. It is concluded that the site-specific damage in telomere sequence induced by UVA irradiation may participate in the increase of telomere shortening rate.


Subject(s)
DNA Damage , Deoxyguanosine/analogs & derivatives , Guanine/radiation effects , Telomere/genetics , Telomere/radiation effects , Ultraviolet Rays , 8-Hydroxy-2'-Deoxyguanosine , Base Sequence/radiation effects , Cell Line , Chromatography, High Pressure Liquid , DNA-Formamidopyrimidine Glycosylase , Deoxyguanosine/biosynthesis , Deoxyguanosine/radiation effects , Electrochemistry , Fibroblasts/physiology , Fibroblasts/radiation effects , HL-60 Cells , Humans , Light , N-Glycosyl Hydrolases/physiology , Photosensitizing Agents/pharmacology , Riboflavin/physiology , Telomere/metabolism
19.
Int J Cancer ; 92(3): 333-7, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11291067

ABSTRACT

Estrogen-induced carcinogenesis involves enhanced cell proliferation (promotion) and genotoxic effects (initiation). To investigate the contribution of estrogens and their metabolites to tumor initiation, we examined DNA damage induced by estradiol and its metabolites, the catechol estrogens 2-hydroxyestradiol (2-OHE(2)) and 4-hydroxyestradiol (4-OHE(2)). In the presence of Cu(II), catechol estrogens formed piperidine-labile sites at thymine and cytosine residues in (32)P 5'-end-labeled DNA fragments and induced the formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine. NADH markedly enhanced Cu(II)-dependent DNA damage mediated by nanomolar concentrations of catechol estrogens. Catalase and bathocuproine inhibited the DNA damage, suggesting the involvement of H(2)O(2) and Cu(I). These results suggest that H(2)O(2), generated during Cu(II)-catalyzed autoxidation of catechol estrogens, reacts with Cu(I) to form the Cu(I)-peroxide complex, leading to oxidative DNA damage, and that NADH enhanced DNA damage through the formation of redox cycle. To investigate the role of estrogens and their metabolites in tumor promotion, we examined their effects on proliferation of estrogen-dependent MCF-7 cells. Estradiol enhanced the proliferation of MCF-7 cells at much lower concentrations than catechol estrogens. These findings indicate that catechol estrogens play a role in tumor initiation through oxidative DNA damage, whereas estrogens themselves induce tumor promotion and/or progression by enhancing cell proliferation in estrogen-induced carcinogenesis.


Subject(s)
DNA Damage , Deoxyguanosine/analogs & derivatives , Estradiol/pharmacology , Estrogens, Catechol/pharmacology , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Cell Division/drug effects , Copper/pharmacology , DNA/drug effects , DNA/metabolism , DNA Fragmentation/drug effects , Deoxyguanosine/metabolism , Drug Interactions , Free Radical Scavengers/pharmacology , Humans , NAD/pharmacology , Phenanthrolines/pharmacology , Phosphorus Radioisotopes , Tumor Cells, Cultured
20.
Cancer Res ; 61(7): 2878-84, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306462

ABSTRACT

In most cases, apoptosis is considered to involve mitochondrial dysfunction with sequential release of cytochrome c from mitochondria, resulting in activation of caspase-3. However, we found that etoposide induced apoptosis in P39 cells, a myelodysplastic syndrome-derived cell line, without the release of cytochrome c. Furthermore, in etoposide-treated P39 cells, no changes in mitochondrial membrane potential (delta psi m) were detected by flow cytometry. Flow cytometry using a pH-sensitive probe demonstrated that lysosomal pH increased during early apoptosis in P39 cells treated with etoposide. A reduction in the ATP level preceded the elevation of lysosomal pH. In addition, specific inhibitors of vacuolar H+-ATPase induced apoptosis in P39 cells but not in HL60 cells. Although etoposide-induced activation of caspase-3 was followed by DNA ladder formation in P39 cells, E-64d, an inhibitor of lysosomal thiol proteases, specifically suppressed etoposide-induced activation of caspase-3. Western blotting analysis provided direct evidence for the involvement of a lysosomal enzyme, cathepsin L. These findings indicate that lysosomal dysfunction induced by a reduction in ATP results in leakage of lysosomal enzymes into the cytosolic compartment and that lysosomal enzyme(s) may be involved in activation of caspase-3 during apoptosis in P39 cells treated with etoposide.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cytochrome c Group/physiology , Endopeptidases , Leucine/analogs & derivatives , Macrolides , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology , Vacuolar Proton-Translocating ATPases , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Cathepsin L , Cathepsins/biosynthesis , Cell Line , Cysteine Endopeptidases , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/physiology , Leucine/pharmacology , Lysosomes/enzymology , Membrane Potentials/physiology , Mitochondria/physiology , Proton-Translocating ATPases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...