Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biosci Biotechnol Biochem ; 87(7): 771-776, 2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37133406

ABSTRACT

Diosmin (DSN) is found mainly in citrus fruits, and has potent antioxidant effects. This study aimed to evaluate pharmacokinetics of diosmetin-7-glucoside-γ-cyclodextrin (DIOSG-CD) inclusion complex. The area under the curve values from AUC0-24 of DIOSG-CD, prepared by reacting DSN and naringinase with γ-CD, were approximately 800-fold higher than those of DSN following their administration in Sprague-Dawley rats.


Subject(s)
Diosmin , gamma-Cyclodextrins , Rats , Animals , Rats, Sprague-Dawley , Diosmin/pharmacokinetics , Biological Availability
2.
J Toxicol Pathol ; 35(4): 313-320, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36406173

ABSTRACT

The carcinogenicity of 2,2'-[1,2-ethanediylbis(oxymethylene)]bis-oxirane (ethylene glycol diglycidyl ether; EGDE), 3-hydroxy-2-naphthoic acid (HNA), and acetoacetanilide (AAA) was investigated using a medium-term rat liver bioassay for an occupational safety assessment. F344 male rats were administered a single intraperitoneal injection of diethylnitrosamine (200 mg/kg body weight (bw)/day) and then starting 2 weeks later, they received EGDE at 6, 20, and 60 mg/kg bw/day, HNA at 20, 60, and 200 mg/kg bw/day, or AAA at 60, 200, and 600 mg/kg bw/day by oral gavage for 6 weeks. The animals in the positive control group received phenobarbital sodium solution (PB, 25 mg/kg bw/day) by oral gavage and those in the negative control group received a vehicle (water/corn oil) during the administration period of test substances in this model. All animals were subjected to two-thirds partial hepatectomy at week 3 and euthanized at week 8. Neither the number nor the area of hepatocellular foci positive for glutathione S-transferase placental form (GST-P) increased in any of the EGDE, HNA, or AAA treated groups. However, the number and area of GST-P-positive foci significantly increased in the positive control group treated with PB. The results indicate that EGDE, HNA, and AAA lack hepatocarcinogenicity in rats.

3.
Exp Toxicol Pathol ; 66(1): 19-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23988840

ABSTRACT

To explore the role of oxidative stress in chemical carcinogenesis driven by non-genotoxic mechanisms, nrf2-deficient (nrf2(-/-)) and nrf2-wild-type (nrf2(+/+)) mice were exposed to pentachlorophenol (PCP) at concentrations of 600 or 1200 ppm for 60 weeks, or piperonyl butoxide (PBO) at concentrations of 3000 or 6000 ppm in the diet for 52 weeks, respectively. Additional studies were performed to examine 8-hydroxydeoxyguanosine (8-OHdG) levels in liver DNA and hepatotoxicological parameters in serum following 8 weeks of exposure of each group to PBO at the same doses as in the long-term study. Exposure to 600 ppm PCP caused cholangiofibrosis (CF) only in nrf2(-/-) mice, while 1200 ppm PCP induced CF in both genotypes. Moreover, cholangiocarcinomas were found with significant incidence only in nrf2(-/-) mice treated with 1200 ppm PCP. Short-term exposure to 6000 ppm PBO caused significant elevation of 8-OHdG levels in both genotypes, while exposure to 3000 ppm caused a significant increase in 8-OHdG only in nrf2(-/-) mice. There were no inter-genotype changes in the incidences of regenerative hepatocellular hyperplasia (RHH) following long-term exposure to PBO. However, the incidence and multiplicity of hepatocellular adenomas, especially those observed in RHH, were much higher in nrf2-/- mice treated with 6000 ppm PBO than in nrf2+/+ mice treated with 6000 ppm PBO. Therefore, oxidative stress generated through PCP or PBO metabolism may promote the proliferation and progression of preneoplastic lesions to neoplasms.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , NF-E2-Related Factor 2/deficiency , Oxidative Stress/genetics , Precancerous Conditions/genetics , Animals , Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , Disease Progression , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism
4.
J Toxicol Pathol ; 26(2): 227-30, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23914067

ABSTRACT

Historical control data of tumor incidence were collected from the control groups (215 animals of each sex) in four recent carcinogenicity studies that were started between 2005 to 2009 (terminally sacrificed between 2007 and 2011) at BoZo Research Center Inc. (Gotemba, Shizuoka, Japan) using Fischer 344 rats (F344/DuCrlCrlj). These data were compared to the previous historical control data (from 1990 to 2004, previously reported) in the same facility. In the results, the incidence of C-cell adenoma in the thyroid tended to increase in both sexes in recent years (30.8% for males and 24.4% for females in 2005-2009) as compared with the previous data (17.4% and 20.1% for males and 11.5% and 11.8% for females in 1990-1999 and 2000-2004, respectively). In addition, the incidences of pancreatic islet cell adenoma in males and uterine adenocarcinoma tended to increase from around 2000 and remained high in recent years (incidences of islet cell adenoma in males of 10.5%, 17.1% and 20.5% in 1990-1999, 2000-2004 and 2005-2009; incidences of uterine adenocarcinoma of 3.3%, 12.0% and 13.5% in 1990-1999, 2000-2004 and 2005-2009, respectively). There was no apparent difference in the incidence of other tumors.

5.
J Appl Toxicol ; 33(12): 1433-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-22972318

ABSTRACT

Oxidative stress is thought to participate in chemical carcinogenesis and may trigger gene mutations. To accurately assess the carcinogenesis risk posed to humans by chemical exposure, it is important to understand the pathways by which reactive oxygen species (ROS) are generated and the effects of the resulting oxidative stress. In the present study, p53-proficient and -deficient gpt delta mice were given pentachlorophenol (PCP), phenobarbital (PhB) or piperonyl butoxide (PBO), which are classified as non-genotoxic hepatocarcinogens in rodents, at the respective carcinogenic doses for 13 weeks. Exposure to PCP or PBO, but not PhB, invoked significant increases in liver DNA 8-hydroxydeoxyguanosine (8-OHdG) levels. Treatment with PCP significantly increased mRNA levels of the gene encoding NAD(P):quinone oxidoreductase 1 (NQO1) in the liver, suggesting that redox cycling of the PCP metabolite tetrachlorohydroquinone gave rise to ROS. Exposure to PhB or PBO significantly elevated CYP 2B10 mRNA levels while NQO1 levels were also significantly increased in PBO-treated mice. Therefore, in addition to involvement of the CYP catalytic pathway in the ROS-generated system of PBO, catechol derivatives produced from the opening of the PBO functional group methylenedioxy ring probably resulted in ROS generation. However, PCP, PBO and PhB failed to increase gpt and red/gam gene mutations in the liver independently of p53. Overall, the action of oxidative stress by ROS derived from the metabolism of these carcinogens might be limited to cancer-promoting activity, which supports the previous classification of these carcinogens as non-genotoxic.


Subject(s)
Carcinogens/toxicity , DNA Damage , Escherichia coli Proteins/genetics , Liver/drug effects , Oxidative Stress/drug effects , Pentosyltransferases/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Organ Size/drug effects , Oxidative Stress/genetics , Prohibitins , Real-Time Polymerase Chain Reaction
7.
Environ Mol Mutagen ; 52(3): 244-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20740625

ABSTRACT

Tumor development in the skin may be a multistep process where multiple genetic alterations occur successively. The p53 gene is involved in genome stability and thus is referred to as "the guardian of the genome." To better understand the antigenotoxic effects of p53 in ultraviolet light B (UVB)-induced mutagenesis, mutations were measured in the epidermis of UVB-irradiated p53(+/+) and p53(-/-) gpt delta mice. In the mouse model, point mutations and deletions are separately identified by the gpt and Spi(-) assays, respectively. The mice were exposed to UVB at single doses of 0.5, 1.0, or 2.0 kJ/m(2) . The mutant frequencies (MFs) were determined 4 weeks after the irradiation. All doses of UVB irradiation enhanced gpt MFs by about 10 times than that of unirradiated mice. There were no significant differences in gpt MFs and the mutation spectra between p53(+/+) and p53(-/-) mice. The predominant mutations induced by UVB irradiation were G:C to A:T transitions at dipyrimidines. In contrast, in unirradiated p53(-/-) mice, the frequencies of Spi(-) large deletions of more than 1 kb and complex-type deletions with rearrangements were significantly higher than those of the Spi(-) large deletions in p53(+/+) counterparts. The specific Spi(-) mutation frequency of more than 1 kb deletions and complex types increased in a dose-dependent manner in the p53(+/+) mice. However, no increase of such large deletions was observed in irradiated p53(-/-) mice. These results suggest that the antigenotoxic effects of p53 may be specific to deletions and complex-type mutations induced by double-strand breaks in DNA.


Subject(s)
Base Sequence/radiation effects , Sequence Deletion , Skin/radiation effects , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Animals , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Skin/metabolism
8.
Arch Toxicol ; 83(11): 1021-30, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19669731

ABSTRACT

It was recently shown that 1-year chronic exposure of rats to tocotrienol (TT) induced highly proliferative liver lesions, nodular hepatocellular hyperplasia (NHH), and independently increased the number of glutathione S-transferase placental form (GST-P)-positive hepatocytes. Focusing attention on the pathological intrinsic property of NHH, a 104-week carcinogenicity study was performed in male and female Wistar Hannover rats given TT at concentrations of 0, 0.4 or 2% in the diet. The high-dose level was adjusted to 1% in both sexes from week 51 because the survival rate of the high-dose males dropped to 42% by week 50. At necropsy, multiple cyst-like nodules were observed, as in the chronic study, but were further enlarged in size, which consequently formed a protuberant surface with a partly pedunculated shape in the liver at the high dose in both sexes. Unlike the chronic study, NHH was not always accompanied by spongiosis, and instead angiectasis was prominent in some nodules. However, several findings in the affected hepatocytes such as minimal atypia, no GST-P immunoreactivity and heterogeneous proliferation, implied that NHH did not harbor neoplastic characteristics from increased exposure despite sustained high cell proliferation. On the other hand, in the high-dose females, the incidence of hepatocellular adenomas was significantly higher than in the control. There was no TT treatment-related tumor induction in any other organs besides the liver. Thus, the overall data clearly suggested that NHH is successively enlarged by further long-term exposure to TT, but does not become neoplastic. In contrast, TT induces low levels of hepatocellular adenomas in female rats.


Subject(s)
Focal Nodular Hyperplasia/chemically induced , Hepatocytes/drug effects , Tocotrienols/toxicity , Vitamins/toxicity , Adenoma, Liver Cell/chemically induced , Adenoma, Liver Cell/pathology , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Focal Nodular Hyperplasia/pathology , Hepatocytes/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Male , Rats , Rats, Wistar , Sex Factors , Tocotrienols/administration & dosage , Vitamins/administration & dosage
9.
Toxicology ; 250(2-3): 143-50, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18675878

ABSTRACT

Tocotrienol is an antioxidant which has found commercial application as a food additive and health supplement all over the world. Since there have been no reports regarding toxicological effects of long-term exposure, we performed a 52-week chronic study using Wistar Hannover rats of both sexes given the compound at doses of 0, 0.08, 0.4 or 2% in powdered basal diet. Since 6 animals in the 2% male group died of hemorrhage of several organs by week 50, the maximum dose level was changed to 1% in both sexes for the last 2 weeks. Decrease of body weight gain was observed in the 2% males from week 5 and females from week 10, this persisting to the end of the study. With the high dose, prolongation of prothrombin time and increase of serum ALT in males, and increase of serum ALP in both sexes were observed with statistical significance. In male and female rats receiving 0.4% or less, there were no toxicological changes in any of the parameters examined. At necropsy, multiple cyst-like nodules on the liver surface were macroscopically pronounced in both sexes receiving 2%. On histopathological examination, hepatocellular nodules were evident with distortion of hepatic cords and compression of the surrounding tissue, almost all including areas of spongiosis hepatis. The constituent hepatocytes were immunohistochemically stained with proliferation cell nuclear antigen at high rates. Nevertheless, they did not exhibit overt atypia and the basic lobular architecture remained intact. Additionally, they were consistently negative for glutathione S-transferase placental form (GST-P). Accordingly, we propose the newly categorized but previously used name 'nodular hepatocellular hyperplasia', which may not necessarily have a neoplastic or regenerative nature. However, quantitative GST-P analysis of the liver sections overall showed numbers of GST-P foci in the high dose females to be significantly elevated as compared to the control value. Based on the present data demonstrating nodular liver lesions only at the high dose of both sexes, we conclude that the no-observed-adverse-effect level (NOAEL) is 0.4% (303 mg/kg/day for males, and 472 mg/kg/day for females).


Subject(s)
Antioxidants/pharmacology , Cell Proliferation/drug effects , Diet , Hepatocytes/physiology , Tocotrienols/pharmacology , Animals , Blood Cell Count , Blood Chemical Analysis , Body Weight/drug effects , Eating/drug effects , Female , Glutathione Transferase/metabolism , Hepatocytes/drug effects , Immunohistochemistry , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Organ Size/drug effects , Rats , Rats, Wistar
10.
Toxicol Sci ; 104(2): 274-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18434339

ABSTRACT

Combination treatment with sodium nitrite (NaNO(2)) and ascorbic acid (AsA) is well known to promote forestomach carcinogenesis in rats and weakly enhance esophageal carcinogenesis under acid reflux conditions. Nitric oxide generation and oxidative DNA damage are considered to be related to the enhancement of carcinogenesis. The purpose of the present study was to investigate whether oxidative DNA damage-associated genotoxicity and tumor initiating potential are involved in the carcinogenesis. In the bacterial reverse mutation assay using Escherichia coli deficient in the mutM gene encoding 8-hydroxydeoxyguanosine (8-OHdG) DNA glycosylase, the combination with NaNO(2) and AsA increased the mutation frequency dramatically, slight increase being evident in the parental strain. In vivo, a significant increase in 8-OHdG levels in the rat forestomach epithelium occurred at 24 h after combined treatment. Six-week-old F344 male rats were given drinking water containing 0.2% NaNO(2) and a diet supplemented with 1% AsA in combination, or the chemicals individually or basal diet alone for 12 weeks. After an interval of 2 weeks, they received 1% butylated hydroxyanisole in the diet for promotion until the end of weeks 52 and 78. Although one squamous cell carcinoma was observed in the combined group, there was no significant variation in tumor development among the groups. The study indicated that the combination of NaNO(2) with AsA induces genotoxicity due to oxidative DNA damage in vitro, and elevates 8-OHdG levels in the forestomach epithelium, but lacks initiating activity in the rat two-stage carcinogenesis model.


Subject(s)
Antioxidants/toxicity , Ascorbic Acid/toxicity , Carcinogens/toxicity , DNA Damage , Mutagens/toxicity , Sodium Nitrite/toxicity , Stomach Neoplasms/chemically induced , 8-Hydroxy-2'-Deoxyguanosine , Animals , Butylated Hydroxyanisole/pharmacology , Cocarcinogenesis , DNA, Bacterial/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Drug Therapy, Combination , Escherichia coli/drug effects , Escherichia coli/genetics , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Male , Methylnitronitrosoguanidine/toxicity , Organisms, Genetically Modified , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
11.
Toxicol Pathol ; 36(2): 353-61, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18364461

ABSTRACT

To ascertain the possible roles of nuclear erythroid 2 p45-related factor 2 (Nrf2), a key transcription factor of phase 2 drug-metabolizing enzymes, in renal cellular defense against oxidative stress, wild-type and Nrf2-knockout -/- mice were treated with ferric nitrilotriacetate (Fe-NTA) at doses of 3 or 6 mg iron/kg body weight. After Fe-NTA treatment, Nrf2 -/- mice consistently showed lower levels of glutathione (GSH) in the kidney at the low dose and the liver at the high dose than the wild-type mice. Gamma-glutamylcysteine ligase (GCL) activity in the kidney and liver of Nrf2-/- mice was also consistently lower than in wild-type mice after the Fe-NTA treatment. Histopathological examination revealed that nephrotoxicity of Fe-NTA, reflected in necrosis of renal tubule epithelial cells following nuclear damage, was more severe in the Nrf2-/- mice than in their wild-type counterparts. Overall, the data suggest that Nrf2 -/- mice are unable to compensate for depletion of renal GSH because of oxidative stress, being more susceptible to Fe-NTA-induced nephrotoxicity. In conclusion, the present study showed that Nrf2 might play an important role in protecting cells from oxidative stress in the kidney through its regulation of antioxidant enzymes.


Subject(s)
Carcinogens/toxicity , Ferric Compounds/toxicity , Kidney/drug effects , NF-E2-Related Factor 2/physiology , Nitrilotriacetic Acid/analogs & derivatives , Animals , Cell Nucleus/drug effects , Cell Nucleus/pathology , Dipeptides/metabolism , Gene Silencing , Glutathione/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Lipid Peroxidation , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Necrosis , Nitrilotriacetic Acid/toxicity , Oxidative Stress , Thiobarbituric Acid Reactive Substances/metabolism
12.
Cancer Sci ; 99(1): 7-13, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17953708

ABSTRACT

Combined treatment with sodium nitrite (NaNO2) and ascorbic acid (AsA) has already been shown to promote rat forestomach carcinogenesis, possibly due to nitric oxide generation under acidic conditions. We hypothesized that a similar effect might occur in the esophagus when the luminal pH is decreased by acid reflux. To clarify this possibility, reflux esophagitis model rats (F344 male) were coadministered 0.2% NaNO2 in the drinking water and 1% AsA in the diet. After 32 weeks of the combined treatment, a significant increase in the incidence of epithelial hyperplasias of the lower-middle and lowest parts of the esophagus were observed compared with the basal-diet group, along with exacerbation of dysplasia and extension of the lesions. Additionally, one squamous cell papilloma was found only in the combined-treatment group. Subsequently, we confirmed the enhancing effects of NaNO2 and AsA cotreatment in the rat N-bis(2-hydroxypropyl)nitrosamine-initiated esophageal tumorigenesis model. The incidence of hyperplasia was enhanced in all segments, along with the incidence and multiplicity of squamous cell papillomas in the lowest segment of the esophagus. Thus, the data demonstrate that combined treatment with NaNO2 and AsA exerts promoting effects on rat esophageal carcinogenesis under acid reflux conditions, as in the forestomach. These findings suggest that the risk of excessive intake of a combination of nitrite and antioxidants for esophageal carcinogenesis is appreciable, particularly in patients with reflux esophagitis.


Subject(s)
Ascorbic Acid/toxicity , Carcinoma, Squamous Cell/etiology , Esophageal Neoplasms/etiology , Esophagitis, Peptic/complications , Sodium Nitrite/toxicity , Animals , Antioxidants/toxicity , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/pathology , Cocarcinogenesis , Disease Models, Animal , Esophageal Neoplasms/chemically induced , Esophageal Neoplasms/pathology , Esophagitis, Peptic/pathology , Food Preservatives/toxicity , Male , Rats , Rats, Inbred F344
13.
Mutat Res ; 633(1): 46-54, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17581771

ABSTRACT

To ascertain whether measurement of possible contributing factors to carcinogenesis concurrently with the transgenic mutation assay is useful to understand the mode of action underlying tumorigenesis of non-genotoxic carcinogens, male and female gpt delta mice were given dicyclanil (DC), a mouse hepatocarcinogen showing all negative results in various genotoxicity tests, at a carcinogenic dose for 13 weeks. Together with gpt and Spi(-) mutations, thiobarbituric acid-reactive substances (TBARS), 8-hydroxydeoxyguanosine (8-OHdG) and bromodeoxyuridine labeling indices (BrdU-LIs) in the livers were examined. Whereas there were no changes in TBARS levels among the groups, significant increases in 8-OHdG levels and centrilobular hepatocyte hypertrophy were observed in the treated mice of both genders. In contrast, BrdU-LIs and liver weights for the treated females, but not the males were significantly higher than those for the controls. Likewise, the gpt mutant frequencies (MFs) in the treated females were significantly elevated, GC:TA transversion mutations being predominant. No significant alterations were found in the gpt MFs of the males and the Spi(-) MFs of both sexes. The results for the transgenic mutation assays were consistent with DC carcinogenicity in terms of the sex specificity for females. Considering that 8-OHdG induces GC:TA transversion mutations by mispairing with A bases, it is likely that cells with high proliferation rates and a large amounts of 8-OHdG come to harbor mutations at high incidence. This is the first report demonstrating DC-induced genotoxicity, the results implying that examination of carcinogenic parameters concomitantly with reporter gene mutation assays is able to provide crucial information to comprehend the underlying mechanisms of so-called non-genotoxic carcinogenicity.


Subject(s)
DNA Damage/drug effects , Escherichia coli Proteins/physiology , Mutation/drug effects , Pentosyltransferases/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation , Cell Survival/drug effects , Cells, Cultured/drug effects , Dose-Response Relationship, Drug , Escherichia coli Proteins/genetics , Female , Juvenile Hormones/toxicity , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Mutagenicity Tests , Mutation/genetics , Pentosyltransferases/genetics , Thiobarbituric Acid Reactive Substances
14.
Toxicology ; 236(1-2): 61-75, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17498859

ABSTRACT

To clarify the possible mechanism of non-genotoxic hepatocarcinogenesis induced by piperonyl butoxide (PBO), male F344 rats were administered an i.p. injection of N-diethylnitrosamine (DEN) to initiate hepatocarcinogenesis. Two weeks later, the rats were administered a PBO-containing (0, 1, or 2%) diet for 6 weeks and subjected to a two-third partial hepatectomy 1 week later. After sacrificing them on week 8, their livers were histopathologically examined and analyzed for gene expression using a microarray and real-time RT-PCR. Reactive oxygen species (ROS) products were also measured using liver microsomes. Hepatocytes exhibited centrilobular hypertrophy and increased glutathione S-transferase placental form (GST-P) positive foci formation. ROS products increased significantly in liver microsomes. In the microarray analysis, the expressions of genes related to metabolism and oxidative stress - NAD(P)H dehydrogenase, quinone 1 (Nqo1), UDP-glucuronosyltransferase (UDPGTR-2), glutathione peroxidase 2 (Gpx2), glutathione reductase (GRx) - multidrug resistance associated protein 3 (Abcc3), and solute carrier family 7 (cationic amino acid transporter, y+ system) member 5 (Slc7a5) were up-regulated in the PBO group in comparison to the 0% PBO group; this was confirmed by real-time RT-PCR. Additionally, a significant up-regulation of stress response related genes such as CYP1A1 was observed in PBO-treated groups in real-time RT-PCR. HPLC analysis revealed that the level of 8-OHdG in the 2% PBO group was significantly higher than that in the 0% PBO group. This suggests that PBO has the potential to generate ROS via metabolic pathways and induce oxidative stress, including oxidative DNA damage, resulting in the induction of hepatocellular tumors in rats.


Subject(s)
Liver Neoplasms, Experimental/metabolism , Oxidative Stress , Pesticide Synergists , Piperonyl Butoxide , Animals , DNA/metabolism , Deoxyadenosines/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Cancer Sci ; 98(7): 949-57, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17428254

ABSTRACT

Combined treatment with several phenolic antioxidants and sodium nitrite (NaNO(2)) has already shown to enhance rat forestomach carcinogenesis. In the present experiment, effects of green tea catechins (GTC) alone or in combination with NaNO(2) on gastric carcinogenesis were investigated in a rat two-stage carcinogenesis model. Groups of eight, 6-week-old F344 male rats were given 0.01%N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) in their drinking water and 5% NaCl in the diet for 10 weeks for glandular stomach initiation and a single intragastric administration of 100 mg/kg/bodyweight of MNNG at week 9 for forestomach initiation. From week 11, they received either drinking water containing 0.2% NaNO(2) and a diet supplemented with 1% GTC in combination, each individual chemical alone or a basal diet until the end of week 42. In the forestomach, incidences and multiplicities of neoplastic lesions were clearly increased by the combined treatment, in spite of GTC alone suppressing the occurrence of papillomas. In a short-term experiment with similar protocol without MNNG pretreatment, a significant increase of 8-hydroxydeoxyguanosine (8-OHdG) levels in forestomach DNA occurred 24 h after the combined treatment, concomitant with erosion and inflammatory cell infiltration. In an in vitro study, electron spin resonance demonstrated hydroxyl radical formation after incubation of epigallocatechin gallate or epicatechin gallate with the NO generator, NOC-7. Thus, GTC alone showed a weak chemopreventive effect on forestomach carcinogenesis, but in the presence of NaNO(2) it exerted a promotive effect which might involve hydroxyl-radical-associated oxidative DNA damage. However, no influence was exerted in the glandular stomach.


Subject(s)
Catechin/toxicity , Methylnitronitrosoguanidine , Stomach Neoplasms/chemically induced , Stomach Neoplasms/pathology , Tea , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Disease Models, Animal , Epithelial Cells/pathology , Hyperplasia , Male , Rats , Rats, Inbred F344
16.
Toxicol Sci ; 97(1): 44-54, 2007 May.
Article in English | MEDLINE | ID: mdl-17264098

ABSTRACT

To investigate the relationship between fenofibrate (FF) and oxidative stress, enzymatic, histopathological, and molecular biological analyses were performed in the liver of male F344 rats fed 2 doses of FF (Experiment 1; 0 and 6000 ppm) for 3 weeks and 3 doses (Experiment 2; 0, 3000, and 6000 ppm) for 9 weeks. FF treatment increased the activity of enzymes such as carnitine acetyltransferase, carnitine palmitoyltransferase, fatty acyl-CoA oxidizing system, and catalase in the liver. However, it decreased those of superoxide dismutase in the liver in both experiments. Increased 8-hydroxy-2'-deoxyguanosine levels in liver DNA and lipofuscin accumulation were observed in the treated rats of Experiment 2. In vitro measurement of reactive oxygen species (ROS) in rat liver microsomes revealed a dose-dependent increase due to FF treatment. Microarray (only Experiment 1) or real-time reverse transcription-polymerase chain reaction analyses revealed that the expression levels of metabolism and DNA repair-related genes such as Aco, Cyp4a1, Cat, Yc2, Gpx2, Apex1, Xrcc5, Mgmt, Mlh1, Gadd45a, and Nbn were increased in FF-treated rats. These results provide evidence of a direct or indirect relationship between oxidative stress and FF treatment. In addition, increases in the expression levels of cell cycle-related genes such as Chek1, Cdc25a, and Ccdn1; increases in the expression levels of cell proliferation-related genes such as Hdgfrp3 and Vegfb; and fluctuations in the expression levels of apoptosis-related genes such as Casp11 and Trp53inp1 were observed in these rats. This suggests that cell proliferation induction, apoptosis suppression, and DNA damage due to oxidative stresses are probably involved in the mechanism of hepatocarcinogenesis due to FF in rats.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , DNA Damage , Fenofibrate/toxicity , Liver/drug effects , Oxidative Stress/drug effects , Peroxisome Proliferators/toxicity , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Biotransformation/genetics , Catalase/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Repair Enzymes/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling/methods , Ki-67 Antigen/metabolism , Lipid Metabolism/drug effects , Liver/enzymology , Liver/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism , Time Factors
17.
Toxicology ; 231(2-3): 234-42, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17254688

ABSTRACT

In order to examine whether the in vivo initiation activity of a new quinolone antimicrobial agent -- norfloxacin (NFLX) -- results in the induction of hepatocellular tumors, F344 male rats were subjected to two-thirds partial hepatectomy and oral administration of 1500 or 750mg/kg BW of NFLX or the vehicle once daily for 3 weeks. From 2 weeks after the completion of NFLX treatment, the rats were given 500ppm phenobarbital (PB) in their drinking water for 51 weeks. After the promotion treatment with PB for 17, 34, or 51 weeks, the rats were euthanized under ether anesthesia, and the NFLX-induced hepatic tumors were examined macroscopically by thoroughly sectioning the liver at 5-mm intervals. The liver slices, one each from all liver lobes, were fixed in 10% neutral buffered formalin for immunohistochemical examination of glutathione S-transferase placental form (GST-P) positive foci. NFLX increased neither the incidence of macroscopic hepatic tumors nor the mean number or area of GST-P positive foci. These results suggest that under the present study conditions, the initiation activity of NFLX does not result in the induction of hepatocellular tumors in rats; thus, the initiation activity of NFLX is extremely weak.


Subject(s)
Anti-Infective Agents/toxicity , Carcinogens/toxicity , Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Norfloxacin/toxicity , Administration, Oral , Animals , Carcinogenicity Tests , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Hepatectomy , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Phenobarbital/pharmacology , Rats , Rats, Inbred F344
18.
Arch Toxicol ; 81(1): 63-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16802149

ABSTRACT

Flumequine (FLU), an anti-bacterial quinolone agent, has been recognized as a non-genotoxic carcinogen for the mouse liver, but recent reports have suggested that some genotoxic mechanism involving oxidative DNA damage may be responsible for its hepatocarcinogenesis. In the present study, we investigated this possibility in the mouse liver using male and female B6C3F1 gpt delta mice fed diet containing 0.4% FLU, a carcinogenic dose, for 13 weeks. Measurements of 8-hydroxydeoxyguanosine levels in liver DNA, and gpt point and deletion mutations revealed no significant increases in any of these parameters in either sex. Histopathologically, centrilobular swelling of hepatocytes with vacuolation was apparent, however, together with significant increase in bromodeoxyuridine-labeling indices in the treated males and females. These results suggest that genotoxicity, including oxidative DNA damage, is not involved in mouse hepatocarcinogenesis by FLU, which might rather solely exert tumor-promoting effects in the liver.


Subject(s)
DNA Damage , Fluoroquinolones/toxicity , Liver/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Anti-Infective Agents/toxicity , Bromodeoxyuridine/analysis , Bromodeoxyuridine/metabolism , Chromatography, High Pressure Liquid , DNA/chemistry , DNA/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Deoxyguanosine/metabolism , Eating/drug effects , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Immunochemistry , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Mutagenicity Tests/methods , Mutation , Organ Size/drug effects , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Weight Loss/drug effects
19.
Cancer Sci ; 97(9): 829-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16805826

ABSTRACT

To clarify the role of 8-OHdG formation as a starting point for carcinogenesis, we examined the dose-dependence and time-course of changes of OGG1 mRNA expression, 8-OHdG levels and in vivo mutations in the kidneys of gpt delta rats given KBrO3 in their drinking water for 13 weeks. There were no remarkable changes in OGG1 mRNA in spite of some increments being statistically significant. Increases of 8-OHdG occurred after 1 week at 500 p.p.m. and after 13 weeks at 250 p.p.m. Elevation of Spi- mutant frequency, suggestive of deletion mutations, occurred after 9 weeks at 500 p.p.m. In a two-stage experiment, F344 rats were given KBrO3 for 13 weeks then, after a 2-week recovery, treated with 1% NTA in the diet for 39 weeks. The incidence and multiplicity of renal preneoplastic lesions in rats given KBrO3 at 500 p.p.m. followed by NTA treatment were significantly higher than in rats treated with NTA alone. Results suggest that a certain period of time might be required for 8-OHdG to cause permanent mutations. The two-step experiment shows that cells exposed to the alteration of the intranuclear status by oxidative stress including 8-OHdG formation might be able to form tumors with appropriate promotion.


Subject(s)
Bromates/toxicity , Carcinogens/toxicity , DNA Damage/drug effects , Kidney Neoplasms/genetics , Kidney/drug effects , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Animals, Genetically Modified , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , DNA Glycosylases/biosynthesis , DNA Glycosylases/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/biosynthesis , Dose-Response Relationship, Drug , Kidney/pathology , Kidney Neoplasms/chemically induced , Kidney Neoplasms/pathology , Male , Mutation , Nitrilotriacetic Acid/toxicity , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/pathology , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
20.
Cancer Sci ; 97(5): 368-73, 2006 May.
Article in English | MEDLINE | ID: mdl-16630133

ABSTRACT

The effects of IQ on the promotion stage of DHPN-induced lung carcinogenesis and contributions of oxidative stress were investigated in rats. Groups of 20 male 6-week-old F344 rats were given 0.1% DHPN in their drinking water for 2 weeks for initiation. From the age of 9 weeks, they were treated with 0, 150 and 300 p.p.m. of IQ in the diet for 27 weeks. Control rats were similarly fed 300 p.p.m. IQ or basal diet alone without the preceding initiation. IQ clearly (P < 0.01) enhanced the multiplicity of lung tumors in a dose-dependent manner (DHPN alone, 3.63 +/- 1.80; DHPN +150 p.p.m. IQ, 11.50 +/- 5.04; DHPN +300 p.p.m. IQ, 18.83 +/- 4.58 [no./rat]). In addition, the incidence of lung tumors in the 300 p.p.m. IQ alone group (25%) was significantly (P < 0.05) higher than that in the non-treatment group (0%). In a second experiment, male rats were given IQ at doses of 0 and 300 p.p.m. in the diet for one week in order to analyze 8-OHdG formation, levels of TBARS and BrdU-LI in the lungs. There were no changes in 8-OHdG or TBARS levels, but significant elevation of BrdU-LI occurred in the IQ administration group. The overall data clearly indicate that IQ is a potent lung carcinogen in rats, in which oxidative stress may not be involved in lung carcinogenesis.


Subject(s)
Carcinogens/toxicity , Lung Neoplasms/chemically induced , Nitrosamines/toxicity , Quinolines/toxicity , 8-Hydroxy-2'-Deoxyguanosine/analogs & derivatives , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation , DNA/metabolism , DNA Damage/drug effects , Disease Models, Animal , Guanine/analogs & derivatives , Guanine/metabolism , Lipid Peroxidation/drug effects , Lung/cytology , Lung/drug effects , Lung/metabolism , Male , Oxidative Stress/drug effects , Rats , Thiobarbituric Acid Reactive Substances/analysis , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...