Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Toxicol Pathol ; 45(4): 493-505, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28580885

RESUMO

We previously reported the contribution of constitutive androstane receptor (CAR) in cytotoxicity-related hepatocarcinogenesis induced by oxadiazon (OX) or acifluorfen (ACI), two pesticides categorized as protoporphyrinogen oxidase (PROTOX) inhibitors. The molecular characteristics of preneoplastic and neoplastic lesions induced by OX and ACI were immunohistochemically compared to those by phenobarbital (PB), a typical CAR activator, in wild-type (WT) and CAR knockout (CARKO) mice after diethylnitrosamine initiation. We focused on changes in ß-catenin and its transcriptional product glutamine synthetase (GS). In PB-promoted foci and adenomas, nuclear accumulation of mutated ß-catenin was increased with high frequency. PB treatment also increased the multiplicity and area of GS-positive foci and adenomas in WT mice. No foci and adenomas showed nuclear accumulation of ß-catenin and expression of GS in CARKO mice, similar to both genotypes of mice treated with OX and ACI. Interestingly, hepatocellular carcinoma induced in ACI-treated WT mice showed nuclear accumulation of ß-catenin and was positive for GS. Our results indicated that ß-catenin mutations were not involved in early-stage hepatocarcinogenesis induced by PROTOX inhibitors in mice, although activation of ß-catenin and CAR is important in PB-induced tumorigenesis. The significant differences in molecular profiles suggested involvements of multiple mode of actions for hepatocarcinogenesis induced by PROTOX inhibitors.


Assuntos
Carcinogênese/genética , Inibidores Enzimáticos/toxicidade , Neoplasias Hepáticas Experimentais/genética , Nitrobenzoatos/toxicidade , Oxidiazóis/toxicidade , beta Catenina/genética , Animais , Carcinogênese/induzido quimicamente , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , Receptor Constitutivo de Androstano , Glutamato-Amônia Ligase/genética , Glutamato-Amônia Ligase/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Mutação , Fenobarbital/toxicidade , Protoporfirinogênio Oxidase/antagonistas & inibidores , Protoporfirinogênio Oxidase/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-28464995

RESUMO

Polybrominated diphenyl ethers (PBDEs), a class of brominated flame retardants, have been widely used as additive flame retardants. Recently, the use of brominated flame retardants has been restricted or prohibited under various legislative acts because of the persistence, bioaccumulation potential, and toxicity of these compounds. However, there are also additional concerns regarding environmental contamination and human exposure to PBDEs resulting from informal recycling technology. Decabromodiphenyl ether (decaBDE), one type of PBDE, has carcinogenic potential in the livers of rodents. Although one study has shown that decaBDE exerts genotoxic effects, the other in vitro and in vivo studies were negative for such effects. Thus, it remains unknown whether genotoxic mechanisms are involved in decaBDE-induced hepatocarcinogenesis in rodents. In this study, to explore the genotoxicity of decaBDE in mice, particularly in the context of carcinogenesis, we performed micronucleus assays in the bone marrow and reporter gene mutation assays in the liver using gpt delta mice treated with decaBDE at carcinogenic doses for 28days. Our results demonstrated negative results in micronucleus tests and reporter gene mutation assays. Thus, decaBDE did not exert genotoxic effects at carcinogenic target sites and did not show positive results in conventional in vivo genotoxicity tests in mice for 4-week treatment. Overall, comprehensive evaluation using in vivo genotoxicity data in rats and our data indicated that nongenotoxic mechanisms may be responsible for decaBDE-induced hepatocarcinogenesis.


Assuntos
Retardadores de Chama/toxicidade , Genes Reporter , Éteres Difenil Halogenados/toxicidade , Animais , Bioensaio , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Testes para Micronúcleos , Mutação
3.
J Toxicol Sci ; 41(6): 801-811, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27853109

RESUMO

To clarify the major pathway of liver tumor development induced by imazalil (IMA), an imidazole fungicide, male constitutive androstane receptor (CAR)-knockout (CARKO) and wild-type (WT) mice were treated with IMA at 500 ppm in the diet up to 27 weeks after initiation by diethylnitrosamine. After 27 weeks of treatment, neither altered foci nor adenomas were significantly increased in CARKO mice, whereas both eosinophilic altered foci and adenomas were increased in WT mice. After 4 or 13 weeks of IMA treatment, liver hypertrophy was observed at the tumor-inducible dose without differences among genotypes or durations. Analysis of hepatic drug metabolite enzymes, performed after administration of multiple doses during a 1-week period, indicated that pregnane X receptor might be involved in liver hypertrophy because IMA markedly elevated Cyp3a11 and Cyp2b10 expression levels in a dose-dependent manner in both genotypes. Our results demonstrated that the CAR pathway was the main mechanism of liver tumor development induced by IMA. The carcinogenic pathway was different from that of liver hypertrophy.


Assuntos
Adenoma/induzido quimicamente , Fungicidas Industriais/toxicidade , Imidazóis/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Receptores Citoplasmáticos e Nucleares/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proliferação de Células/efeitos dos fármacos , Cocarcinogênese , Receptor Constitutivo de Androstano , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Família 2 do Citocromo P450/genética , Família 2 do Citocromo P450/metabolismo , Dietilnitrosamina/toxicidade , Relação Dose-Resposta a Droga , Genótipo , Hipertrofia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C3H , Camundongos Knockout , Fenótipo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Medição de Risco , Transdução de Sinais/efeitos dos fármacos , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Fatores de Tempo
4.
Toxicol Sci ; 151(2): 271-85, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26928356

RESUMO

Acifluorfen (ACI), a protoporphyrinogen oxidase (PROTOX) inhibitor herbicide, promotes the accumulation of protoporphyrin IX (PPIX), and induces tumors in the rodent liver. Porphyria is a risk factor for liver tumors in humans; however, the specific mechanisms through which ACI induces hepatocarcinogenesis in rodents are unclear. Here, we investigated the mode of action of ACI-induced hepatocarcinogenesis, focusing on constitutive androstane receptor (CAR, NR1I3), which is essential for the development of rodent liver tumors in response to certain cytochrome P450 (CYP) 2B inducers. Dietary treatment with 2500 ppm ACI for up to 13 weeks increased Cyp2b10 expression in the livers of wild-type (WT) mice, but not in CAR-knockout (CARKO) mice. Microscopically, ACI treatment-induced cytotoxic changes, including hepatocellular necrosis and inflammation, and caused regenerative changes accompanied by prolonged increases in the numbers of proliferating cell nuclear antigen-positive hepatocytes in WT mice. In contrast, these cytotoxic and regenerative changes in hepatocytes were significantly attenuated, but still observed, in CARKO mice. ACI treatment also increased liver PPIX levels similarly in both genotypes; however, no morphological evidence of porphyrin deposition was found in hepatocytes from either genotype. Treatment with 2500 ppm ACI for 26 weeks after initiation with diethylnitrosamine increased the incidence and multiplicities of altered foci and adenomas in hepatocytes from WT mice; these effects were significantly reduced in CARKO mice. These results indicated that prolonged cytotoxicity in the liver was a key factor for ACI-induced hepatocarcinogenesis, and that CAR played an important role in ACI-induced liver injury and tumor development in mice.


Assuntos
Adenoma/induzido quimicamente , Transformação Celular Neoplásica/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Nitrobenzoatos/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Receptor Constitutivo de Androstano , Família 2 do Citocromo P450/metabolismo , Dietilnitrosamina/toxicidade , Genótipo , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos C3H , Camundongos Knockout , Necrose , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Protoporfirinas/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Esteroide Hidroxilases/metabolismo , Fatores de Tempo
5.
Food Chem Toxicol ; 88: 75-86, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26710982

RESUMO

Oxadiazon (OX) is a protoporphyrinogen oxidase-inhibiting herbicide that induces porphyria and liver tumors in rodents. Although porphyria is generally considered to be a risk factor for liver tumor development, the mechanisms through which OX mediates tumor development are unclear. Therefore, in this study, we investigated the mechanisms of tumor development by focusing on constitutive active/androstane receptor (CAR), which is essential for the development of tumors in response to several chemicals. After 1, 4, or 13 weeks of dietary treatment with 1000 ppm OX, hepatic Cyp2b10 expression was induced in wild-type (WT) mice. However, this effect was blocked in CAR-knockout (CARKO) mice. Hepatic Cyp4a10 expression, indicative of peroxisome proliferator-activated receptor α (PPARα) activation, and cytotoxic changes in hepatocytes were also observed in both groups of mice. After initiation by diethylnitrosamine, 26-week treatment with OX resulted in an increase in proliferative lesions, including foci and adenomas, in both genotypes, and the incidence and multiplicity of proliferative lesions in CARKO mice were higher than those in control mice but lower than those in WT mice. These results suggested that CAR, PPARα activation, and cytotoxicity were involved in the development of liver tumors. Moreover, porphyrin was not apparently involved in OX-induced tumor development.


Assuntos
Neoplasias Hepáticas/induzido quimicamente , Oxidiazóis/toxicidade , PPAR alfa/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Carcinogênese/induzido quimicamente , Morte Celular , Células Cultivadas , Receptor Constitutivo de Androstano , Hepatócitos/efeitos dos fármacos , Herbicidas/toxicidade , Masculino , Camundongos , PPAR alfa/genética , Receptores Citoplasmáticos e Nucleares/genética
6.
Food Chem Toxicol ; 83: 201-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115596

RESUMO

Ginkgo biloba extract (GBE) is commonly used as a herbal supplement. The National Toxicology Program (NTP) study of GBE reported clear evidence of hepatocarcinogenicity in mice. To clarify the mode of action (MOA) for hepatocarcinogenesis by GBE, we investigated the involvement of the constitutive androstane receptor (CAR) in hepatocarcinogenesis induced by GBE using CAR-knockout (CARKO) and wild type (WT) mice. We used the same lot of GBE that was used for the NTP study. In 1-week GBE dietary treatment, hepatocellular DNA replication was increased in WT mice but not in CARKO mice. In 4- or 13-week treatment, greater hepatic Cyp2b10 induction and hepatocellular hypertrophy were observed in WT mice, whereas these effects of GBE were much smaller in CARKO mice. In a two-stage hepatocarcinogenesis model initiated by diethylnitrosamine, 27-week treatment with GBE resulted in an increase of eosinophilic altered foci and adenomas in WT mice. By contrast, foci and adenomas were clearly less evident in CARKO mice. These results indicate that GBE-induced hepatocarcinogenesis is mainly CAR-mediated. Since CAR-mediated MOA for hepatocarcinogenesis in rodents is considered to be qualitatively implausible for humans, our findings would be helpful to evaluate the carcinogenic characterization of GBE to humans.


Assuntos
Cocarcinogênese/metabolismo , Suplementos Nutricionais/efeitos adversos , Ginkgo biloba/química , Hepatomegalia/etiologia , Neoplasias Hepáticas/etiologia , Extratos Vegetais/efeitos adversos , Receptores Citoplasmáticos e Nucleares/agonistas , Adenoma de Células Hepáticas/induzido quimicamente , Adenoma de Células Hepáticas/etiologia , Adenoma de Células Hepáticas/metabolismo , Adenoma de Células Hepáticas/patologia , Animais , Hidrocarboneto de Aril Hidroxilases/química , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Carcinógenos/química , Carcinógenos/toxicidade , Cocarcinogênese/patologia , Receptor Constitutivo de Androstano , Indutores das Enzimas do Citocromo P-450/efeitos adversos , Família 2 do Citocromo P450 , Replicação do DNA , Dietilnitrosamina/agonistas , Dietilnitrosamina/toxicidade , Hepatomegalia/metabolismo , Hepatomegalia/patologia , Japão , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos C3H , Camundongos Knockout , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Esteroide Hidroxilases/química , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Testes de Toxicidade Subcrônica
7.
Food Chem Toxicol ; 78: 86-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25656644

RESUMO

We clarified the involvement of constitutive androstane receptor (CAR) in triazole-induced liver hypertrophy and tumorigenesis using CAR-knockout (CARKO) mice. Seven-week-old male CARKO and wild-type (WT) mice were treated with 200 ppm cyproconazole (Cypro), 1500 ppm tebuconazole (Teb), or 200 ppm fluconazole (Flu) in the diet for 27 weeks after initiation by diethylnitrosamine (DEN). At weeks 4 (without DEN) and 13 (with DEN), WT mice in all treatment groups and CARKO mice in Teb group revealed liver hypertrophy with mainly Cyp2b10 and following Cyp3a11 inductions in the liver. Teb also induced Cyp4a10 in both genotypes. Cypro induced slight and duration-dependent liver hypertrophy in CARKO mice. At week 27, Cypro and Teb significantly increased eosinophilic altered foci and/or adenomas in WT mice. These proliferating lesions were clearly reduced in CARKO mice administered both compounds. The eosinophilic adenomas caused by Flu decreased in CARKO mice. The present study indicates that CAR is the main mediator of liver hypertrophy induced by Cypro and Flu, but not Teb. In contrast, CAR played a crucial role in liver tumor development induced by all three triazoles.


Assuntos
Fluconazol/toxicidade , Hepatomegalia/patologia , Neoplasias Hepáticas Experimentais/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Triazóis/toxicidade , Alanina Transaminase/sangue , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptor Constitutivo de Androstano , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Família 2 do Citocromo P450 , Dietilnitrosamina/toxicidade , Fungicidas Industriais/toxicidade , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatomegalia/induzido quimicamente , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo
8.
Toxicol Sci ; 144(1): 65-76, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25636497

RESUMO

Carcinogenic doses of ochratoxin A (OTA) cause increases of mutant frequencies (MFs) of the red/gam gene (Spi(-)) in the kidneys of p53-deficient gpt delta mice, but not in p53-proficient mice. Here, we investigated the role of p53 in the progression from OTA-induced DNA damage to gene mutations. To this end, p53-proficient and -deficient mice were administered 5 mg/kg OTA for 3 days or 4 weeks by gavage. After 3 days of administration, comet assays were performed and there were no differences in the degrees of OTA-induced DNA damage between p53-proficient and -deficient mice. However, the frequencies of γ-H2AX-positive tubular epithelial cells in p53-deficient mice were significantly higher than those in p53-proficient mice, implying that p53 inhibited the progression from DNA damage to DNA double-strand breaks (DSBs). Evaluation of global gene expression and relevant mRNA/protein expression levels demonstrated that OTA increased the expression of Cdkn1a, which encodes the p21 protein, in p53-proficient mice, but not in p53-deficient mice. Moreover, in p53-deficient mice, mRNA levels of cell cycle progression and DSB repair (homologous recombination repair [HR])-related genes were significantly increased. Thus, G1/S arrest due to activation of the p53/p21 pathway may contribute to the prevention of DSBs in p53-proficient mice. In addition, single base deletions/insertions/substitutions were predominant, possibly due to HR. Overall, these results suggested that OTA induced DSBs at the carcinogenic target site in mice and that p53/p21-mediated cell cycle control prevented an increase in the formation of DSBs, leading to gene mutations.


Assuntos
Quebras de DNA de Cadeia Dupla , Neoplasias Renais/genética , Rim/metabolismo , Mutação , Ocratoxinas , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Western Blotting , Ensaio Cometa , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA Bacteriano/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Pontos de Checagem da Fase G1 do Ciclo Celular , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Histonas/metabolismo , Imuno-Histoquímica , Rim/patologia , Neoplasias Renais/induzido quimicamente , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
9.
Mutagenesis ; 30(2): 227-35, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25392148

RESUMO

Acrylamide (AA) is a contaminant in heated foods and is carcinogenic in multiple organs of rodents. There have been many reports regarding AA-induced DNA modification and genotoxicity. However, the data are insufficient to understand fully the relationship between the two events. A recent report demonstrated carcinogenicity in the mouse lung. The lung is advantageous for investigation of AA-induced genotoxicity because DNA adduct levels are relatively high in this organ. In the present study, reporter gene mutation assays and quantitative analyses of specific DNA adducts were performed in the lungs of mature gpt delta mice treated with AA at doses of 100, 200 and 400 p.p.m. in drinking water for 4 weeks. N7-GA-Gua was detected in all AA-treated mice in a dose-dependent manner. gpt mutant frequencies (MFs) were significantly increased in the middle- and high-dose groups. In the analysis of mutation spectra, significant increases in GC-TA transversions and single base deletion mutations were observed in the high-dose group. Spi(-) MFs were significantly increased in the high-dose group. Analysis of Spi(-) mutants revealed significant increases in the frequencies of single base deletion mutation in runs of G/C and A/T. Analyses of immature mice under the same experimental conditions showed that there were no differences of susceptibility to AA-induced genotoxicity in the two age classes. The overall data clearly show the causal relationship between AA-induced DNA adducts and the gene mutations at carcinogenic target sites.


Assuntos
Acrilamida/toxicidade , Carcinógenos/toxicidade , Adutos de DNA/metabolismo , Pulmão/efeitos dos fármacos , Mutação , Animais , Análise Mutacional de DNA , Pulmão/metabolismo , Masculino , Camundongos
10.
Asian Pac J Cancer Prev ; 15(17): 7149-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25227805

RESUMO

A large number of epidemiological studies have demonstrated that obesity is a risk factor for several human cancers. Several animal studies using rodents with diet-induced or genetic obesity have also demonstrated that obesity can promote tumor development. However, the effects of obesity on the early stages of carcinogenesis, and especially on the spontaneous occurrence of somatic gene mutations, remain unclear. To investigate the effects of obesity on the rate of spontaneous gene mutations, we performed reporter gene mutation assays in liver, kidney, and colon, organs in which obesity appears to be associated with cancer development on the basis of epidemiological or animal studies, in mice with high fat diet (HFD)-induced obesity. Six-week-old male and female C57BL/6 gpt delta mice were fed HFD or standard diet (STD) for 13 or 26 weeks. At the end of the experiments, reporter gene mutation assays of liver, kidney, and colon were performed. Final body weights and serum leptin levels of male and female mice fed HFD for 13 or 26 weeks were significantly increased compared with corresponding STD-fed groups. Reporter gene mutation assays of liver, kidney, and colon revealed that there were no significant differences in gpt or Spi- mutant frequencies between STD- and HFD-fed mice in either the 13-week or 26-week groups. These results indicate that HFD treatment and consequent obesity does not appear to influence the spontaneous occurrence of somatic gene mutations.


Assuntos
Colo/metabolismo , Dieta Hiperlipídica , Proteínas de Escherichia coli/genética , Genes Reporter/genética , Rim/metabolismo , Fígado/metabolismo , Mutação , Obesidade/genética , Pentosiltransferases/genética , Animais , Feminino , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/metabolismo
11.
Toxicol Pathol ; 42(8): 1174-87, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24862798

RESUMO

Medulloblastomas (MBs) are thought to be derived from granular cell precursors in the external granular layer (EGL) of the developing cerebellum. Heterozygous patched1 (Ptch1) knockout mice develop MBs that resemble those in humans when the sonic hedgehog (Shh) signaling pathway is activated. The present study was conducted to evaluate postnatal effects of a Shh signaling inhibitor, cyclopamine, on the development of MBs in Ptch1 mice. Ptch1 and wild-type mice were treated daily with subcutaneous cyclopamine at 40 mg/kg or vehicle from postnatal day (PND) 1 to PND14, and the subsequent development of MBs and preneoplastic lesions was examined up to week 12 (W12). Proliferative lesions in the cerebellum, MBs, and preneoplastic lesions were only detected in Ptch1 mice. Cyclopamine treatment resulted in a statistically significant reduction in the incidence and/or area of proliferative lesions at PND14 and 21. The trend of decreasing preneoplastic lesions persisted up to W12. At PND7, cyclopamine treatment reduced the width and proliferation of the EGL regardless of genotype. These results indicate that inhibition of Shh signaling during cerebellar development has prolonged inhibitory potential on MB development in Ptch1 mice. This inhibitory potential might be related to inhibition of EGL proliferation, including preneoplastic MB cells.


Assuntos
Cerebelo/efeitos dos fármacos , Proteínas Hedgehog/antagonistas & inibidores , Meduloblastoma/metabolismo , Alcaloides de Veratrum/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Cerebelo/química , Cerebelo/crescimento & desenvolvimento , Cerebelo/patologia , Meduloblastoma/química , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Patched , Receptor Patched-1 , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
12.
Toxicol Pathol ; 41(8): 1078-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23531792

RESUMO

The constitutive androstane receptor (CAR) is essential for Cyp2b induction, liver hypertrophy, and hepatocarcinogenesis in response to phenobarbital (PB). Liver hypertrophy with Cyp2b induction is a major mode of action of hepatocarcinogenesis in rodents. However, it remains unclear whether CAR is involved in the response to many other nongenotoxic hepatocarcinogens besides PB. In this study, we investigated CAR involvement in liver hypertrophy and hepatocarcinogenesis of Cyp2b-inducing nongenotoxic hepatocarcinogens, piperonyl butoxide (PBO), and decabromodiphenyl ether (DBDE), using wild-type and CAR knockout (CARKO) male mice. PB was used as the positive control. In the wild-type mice, 4-week treatment with PBO, DBDE, or PB induced hepatocellular hypertrophy with increased Cyp2b10 messenger RNA and Cyp2b protein expression. In CARKO mice, only PBO showed liver hypertrophy with Cyp2b10 and Cyp3a11 induction. After 27-week treatment following diethylnitrosamine initiation, PBO and PB generated many eosinophilic altered foci/adenomas in wild-type mice; however, the lesions were far less frequent in CARKO mice. DBDE increased the multiplicity of basophilic altered foci/adenomas in wild-type and CARKO mice. Our findings indicate that murine CAR plays major roles in hepatocarcinogenesis but not in liver hypertrophy of PBO. DBDE may act via CAR-independent pathways during hepatocarcinogenesis.


Assuntos
Éteres Difenil Halogenados/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/metabolismo , Butóxido de Piperonila/toxicidade , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptor Constitutivo de Androstano , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos
13.
Exp Toxicol Pathol ; 65(6): 863-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23369240

RESUMO

Patched1 (Ptch1) encodes a receptor for Sonic hedgehog (Shh) and is major gene related to human medulloblastoma (MB) in the Shh subgroup. MB is thought to arise from residual granule cell precursors (GCPs) located in the external granular layer (EGL) of the developing cerebellum. As the detailed preneoplastic changes of MB remain obscure, we immunohistochemically clarified the derived cell, early events of MBs, and the cerebellar developmental processes of Ptch1(+/-) (Ptch1) mice, an animal model of human MB of the Shh subgroup. In Ptch1 mice, the earliest proliferative lesions were detected at PND10 as focal thickened areas of outer layer of the EGL. This area was composed of GCP-like cells with atypia and nuclei disarrangement. In the latter cerebellar developmental period, GCP-like cell foci were detected at high incidence in the outermost area of the cerebellum. Their localization and morphological similarities indicated that the foci were derived from GCPs in the EGL. There were two types of the foci. A Ki-67-positive focus was found in Ptch1 mice only. This type resembled the GCPs in the outer layer of EGL characterized by having proliferating activity and a lack of neuronal differentiation. Another type of focus, Ki-67-negative, was observed in both genotypes and exhibited many of the same features of mature internal granule cells, suggesting that the focus had no preneoplastic potential. Due to morphological, immunohistochemical characteristics, our results indicate that the focal thickened area of EGL and Ki-67-positive foci are preneoplastic lesions of MB.


Assuntos
Cerebelo/patologia , Antígeno Ki-67/metabolismo , Meduloblastoma/patologia , Lesões Pré-Cancerosas/patologia , Receptores de Superfície Celular , Animais , Animais Recém-Nascidos , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Genótipo , Heterozigoto , Imuno-Histoquímica , Meduloblastoma/metabolismo , Camundongos , Camundongos Knockout , Receptores Patched , Receptor Patched-1 , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Receptores de Superfície Celular/genética
14.
J Toxicol Sci ; 37(5): 1077-82, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23038016

RESUMO

A major product formed during the Maillard reaction is 5-(hydroxymethyl)-2-furfural (HMF), which is present in various foods and beverages such as honey and fruit juice. HMF was shown to be a hepatocarcinogen in female mice using long-term bioassays. Although HMF is not a mutagen in conventional in vitro mutation assays, 5-sulfoxymethylfurfural (SMF), a reactive metabolite of HMF produced following sulfotransferase conjugation, does show mutagenicity. Thus, HMF-induced hepatocarcinogenesis likely involves genotoxic mechanisms. To clarify the mechanisms underlying HMF-induced hepatocarcinogenesis, female B6C3F(1) gpt delta mice were given HMF at carcinogenic doses (188 or 375 mg/kg b.w.) by gavage for 5 days per week for 4 weeks. This treatment produced no significant differences in mutant frequencies (MFs) of gpt and red/gam (Spi(-)) genes among the groups. These results suggest that genotoxicity does not contribute to HMF-induced hepatocarcinogenesis. Parameters related to cell proliferation, such as proliferation cell nuclear antigen-labeling index and Cyclin D1 and E1 mRNA expression, exhibited no significant changes in the livers of HMF-treated groups. In view of the lack of carcinogenicity in rats, HMF may be considered to be a weak carcinogen. These results help us to understand the underlying mechanisms of action of HMF carcinogenesis.


Assuntos
Carcinógenos/toxicidade , Furaldeído/análogos & derivados , Animais , Peso Corporal/efeitos dos fármacos , Ciclina D1/genética , Ciclina E/genética , Proteínas de Escherichia coli/genética , Feminino , Contaminação de Alimentos , Furaldeído/toxicidade , Genes Reporter , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Mutação , Proteínas Oncogênicas/genética , Tamanho do Órgão/efeitos dos fármacos , Pentosiltransferases/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro/metabolismo
15.
Cancer Sci ; 103(12): 2051-5, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22937732

RESUMO

Mice heterozygous for the ptch1 gene (ptch1 mice) are known as a valuable model of medulloblastoma, a common brain tumor in children. To increase the incidence and reduce the time required for tumor development, allowing for evaluation of modifier effects on medulloblastoma in a short time, we attempted to develop an early induction model of medulloblastoma in ptch1 mice initiated with N-ethyl-N-nitrosourea (ENU). Ptch1 mice and their wild-type littermates received a single intraperitoneal injection of ENU (10, 50 or 100 mg/kg) on postnatal day 1 (d1) or 4 (d4), and histopathological assessment of brains was conducted at 12 weeks of age. The width of the external granular layer (EGL), a possible origin of medulloblastoma, after injection of 100 mg ENU on d1 or d4 was measured in up to 21-day-old mice. Cerebellar size was apparently reduced at the 50 mg dose and higher regardless of genotype. Microscopically, early lesions of medulloblastomas occurred with a high incidence only in ptch1 mice receiving 10 mg on d1 or d4, but a significant increase was not observed in other groups. Persistent EGL cells and misalignment of Purkinje cells were increased dose-dependently. Although EGL was strikingly decreased after ENU injection, strong recovery was observed in mice of the d1-treated group. In summary, neonatal treatment with ENU is available for the induction of medulloblastoma in ptch1 mice, and 10 mg of ENU administered on d1 appeared to be an appropriate dose to induce medulloblastoma.


Assuntos
Alquilantes/administração & dosagem , Neoplasias Encefálicas/induzido quimicamente , Etilnitrosoureia , Meduloblastoma/induzido quimicamente , Receptores de Superfície Celular/genética , Alquilantes/farmacologia , Animais , Neoplasias Encefálicas/patologia , Heterozigoto , Meduloblastoma/patologia , Camundongos , Camundongos Knockout , Receptores Patched , Receptor Patched-1 , Células de Purkinje/patologia
16.
Mutat Res ; 749(1-2): 23-8, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-22885592

RESUMO

Estragole (ES), a natural organic compound, is frequently used as a flavoring in food even though it is a hepatocarcinogen in mice. Although formation of ES-specific DNA adducts following conversion from ES to the nucleophilic metabolite by sulfotransferase 1A1 (SULT1A1) has been reported, the modes of action underlying ES-induced hepatocarcinogenesis remain uncertain because conventional genotoxicity tests for ES yield negative results. In the present study, taking notice of the fact that there is a sex difference in SULT1A1 activity in the mouse liver, we assessed the frequency of micronuclei in polychromatic erythrocytes and the mutant frequency (MF) of reporter genes in female gpt delta mice treated with ES at doses of 0 (corn oil), 37.5, 75, 150 or 300mg/kg body weight (bw) by gavage for 13 weeks. Results were compared with those obtained in males. Since one female was found dead at week one, the highest dose was reduced to 250mg/kg bw in females from week two. As reported previously in C57BL/6 mice, the mRNA levels of Sult1a1 in female gpt delta mice were significantly higher than those in the males. The levels of ES-specific DNA adducts in the females were higher than those in the males at all doses except the highest dose. In addition, MFs of the gpt gene were significantly increased from doses of 75mg/kg bw of females, but the increment was observed only at the highest dose in males. There were no changes in the micronucleus test among the groups. Thus, the overall data suggest that specific DNA modifications by the SULT1A1-mediated carbocation formation and the resultant genotoxicity are key events in the early stage of ES-induced hepatocarcinogenesis of mice.


Assuntos
Anisóis/toxicidade , Arilsulfotransferase/metabolismo , Dano ao DNA/efeitos dos fármacos , Aromatizantes/toxicidade , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Mutagênicos/toxicidade , Derivados de Alilbenzenos , Animais , Adutos de DNA , Relação Dose-Resposta a Droga , Feminino , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes para Micronúcleos
17.
J Toxicol Sci ; 35(5): 709-20, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20930465

RESUMO

Chronic stimulus subsequent to cell injury plays an important role in cancer development, but the precise mechanisms remain unknown partly because appropriate animal models are lacking. In the present study, the effects of hepatotoxicant carbon tetrachloride (CCl(4)) on in vivo mutagenicity were investigated using gpt delta mice with or without p53. Female B6C3F(1) p53-proficient or -deficient gpt delta mice were given a diet containing 300 ppm of 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) for 13 weeks, concurrently with intraperitoneal injection of 1 ml/kg CCl(4) solution once a week. Mutant frequencies of gpt and red/gam in p53-proficient mice fed MeIQx were both significantly elevated by CCl(4)co-treatment. Enhancing effects of CCl(4) treatment were also noted in p53-deficient mice. In the mutation spectra analysis of gpt mutant colonies, G:C to T:A transversions were predominantly observed regardless of CCl(4) injection, and clonal expansion of gpt colonies were increased in the co-treated group as compared with MeIQx alone group. The present data showing no significant changes in mRNA expression levels of CYP1A2 and GSTa4 between MeIQx-treated groups with and without CCl(4). In the Western blotting analysis, CYP1A2 protein levels were significantly decreased in the co-treated group as compared to MeIQx alone group, and GSTα protein levels were not changed among any groups. It is suggested that the mutant frequency by co-treatment with CCl(4) might result from some factors other than p53 or MeIQx metabolism/excretion. Thus, our data clearly demonstrate that this model could be a powerful tool for identifying the mechanisms underlying combinatorial effects on carcinogenesis.


Assuntos
Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/genética , Fígado/efeitos dos fármacos , Mutagênicos/toxicidade , Quinoxalinas/toxicidade , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Sinergismo Farmacológico , Proteínas de Escherichia coli/genética , Feminino , Genótipo , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Pentosiltransferases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
18.
J Toxicol Sci ; 35(5): 731-41, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20930467

RESUMO

To investigate the effects of dextran sulfate sodium (DSS) and/or 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline (MeIQx) on in vivo genotoxicity in the colon, male C57BL/6 p53 (+/+), p53 (+/-) or p53 (-/-) gpt delta mice were twice given 1-week treatment with DSS, 2 weeks apart, and then sacrificed after 2 and 14 weeks. Although colon length was significantly shortened after DSS treatment in all genotypes at each time point, no significant difference in gpt mutant frequency (MF) and tumorigenicity was found between DSS and control groups regardless of genotype. Then, male B6C3F(1) p53 (+/+) or p53 (+/-) gpt delta mice were given DSS as described above and/or fed 300 ppm MeIQx for 7 weeks. Colon length was significantly shortened with DSS in either genotype at weeks 7 and 26, but no effects of co-treatment with MeIQx or p53 deficiency were evident. MeIQx showed a tendency to increase gpt MF in the colon of mice with either genotype, but co-treatment with DSS did not affect these increments. Appreciable incidences of colonic aberrant crypt foci (ACFs) were reported in DSS as well as co-treatment groups of each genotype. Colonic adenomas were observed in co-treatment groups of both genotypes as well as the DSS-only group of p53 (+/+). No effects of the combination of DSS and MeIQx on colon pre- and neoplastic lesions were reported. Our results indicate that MeIQx may take more than 7 weeks to induce genotoxicity in the colon and that the co-treatment of mice did not enhance colon tumorigenicity even in p53-deficient mice.


Assuntos
Cocarcinogênese , Neoplasias do Colo , Sulfato de Dextrana/toxicidade , Mutagênicos/toxicidade , Quinoxalinas/toxicidade , Proteína Supressora de Tumor p53/deficiência , Animais , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Proteínas de Escherichia coli/genética , Masculino , Camundongos , Camundongos Transgênicos , Pentosiltransferases/genética , Proteína Supressora de Tumor p53/genética
19.
Toxicol Sci ; 110(2): 293-306, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19478238

RESUMO

This research focused on three major questions regarding benzene-induced hematopoietic neoplasms (HPNs). First, why are HPNs induced equivocally and at only threshold level with low-dose benzene exposure despite the significant genotoxicity of benzene even at low doses both in experiments and in epidemiology? Second, why is there no linear increase in incidence at high-dose exposure despite a lower acute toxicity (LD(50) > 1000 mg/kg body weight; WHO, 2003, Benzene in drinking-water. Background document for development of WHO Guidelines for Drinking-Water Quality)? Third, why are particular acute myeloid leukemias (AMLs) not commonly observed in mice, although AMLs are frequently observed in human cases of occupational exposure to benzene? In this study, we hypothesized that the threshold-like equivocal induction of HPNs at low-dose benzene exposure is based on DNA repair potential in wild-type mice and that the limited increase in HPNs at a high-dose exposure is due to excessive apoptosis in wild-type mice. To determine whether Trp53 deficiency satisfies the above hypotheses by eliminating or reducing DNA repair and by allowing cells to escape apoptosis, we evaluated the incidence of benzene-induced HPNs in Trp53-deficient C57BL/6 mice with specific regard to AMLs. We also used C3H/He mice, AML prone, with Trp53 deficiency to explore whether a higher incidence of AMLs on benzene exposure might explain the above human-murine differences. As a result, heterozygous Trp53-deficient mice of both strains showed a nonthreshold response of the incidence of HPNs at the lower dose, whereas both strains showed an increasing HPN incidence up to 100% with increasing benzene exposure dose, including AMLs, that developed 38% of heterozygous Trp53-deficient C3H/He mice compared to only 9% of wild-type mice exposed to the high dose. The detection of AMLs in heterozygous Trp53-deficient mice, even in the C57BL/6 strain, implies that benzene may be a potent inducer of AMLs also in mice with some strain differences.


Assuntos
Benzeno/toxicidade , Carcinógenos/toxicidade , Neoplasias Hematológicas/induzido quimicamente , Leucemia Mieloide Aguda/induzido quimicamente , Proteína Supressora de Tumor p53/deficiência , Animais , Apoptose/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Heterozigoto , Exposição por Inalação , Dose Letal Mediana , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Fatores de Tempo , Proteína Supressora de Tumor p53/genética
20.
Arch Toxicol ; 83(8): 795-803, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19326098

RESUMO

Benzene is a well-known environmental pollutant that can induce hematotoxicity, aplastic anemia, acute myelogenous leukemia, and lymphoma. However, although benzene metabolites are known to induce oxidative stress and disrupt the cell cycle, the mechanism underlying lympho/leukemogenicity is not fully understood. Caspase-4 (alias caspase-11) and -12 are inflammatory caspases implicated in inflammation and endoplasmic reticulum stress-induced apoptosis. The objectives of this study were to investigate the altered expression of caspase-4 and -12 in mouse bone marrow after benzene exposure and to determine whether their alterations are associated with benzene-induced bone marrow toxicity, especially cellular apoptosis. In addition, we evaluated whether the p53 gene is involved in regulating the mechanism, using both wild-type (WT) mice and mice lacking the p53 gene. For this study, 8-week-old C57BL/6 mice [WT and p53 knockout (KO)] were administered a benzene solution (150 mg/kg diluted in corn oil) via oral gavage once daily, 5 days/week, for 1 or 2 weeks. Blood and bone marrow cells were collected and cell counts were measured using a Coulter counter. Total mRNA and protein extracts were prepared from the harvested bone marrow cells. Then qRT-PCR and Western blotting were performed to detect changes in the caspases at the mRNA and protein level, respectively. A DNA fragmentation assay and Annexin-V staining were carried out on the bone marrow cells to detect apoptosis. Results indicated that when compared to the control, leukocyte number and bone marrow cellularity decreased significantly in WT mice. The expression of caspase-4 and -12 mRNA increased significantly after 12 days of benzene treatment in the bone marrow cells of benzene-exposed p53KO mice. However, apoptosis detection assays indicated no evidence of apoptosis in p53KO or WT mice. In addition, no changes of other apoptosis-related caspases, such as caspase-3 and -9, were found in WT or p53KO mice at the level of mRNA and proteins. These results indicated that upregulation of caspase-4 and -12 in mice lacking the p53 gene is not associated with cellular apoptosis. In conclusion, caspase-4 and -12 can be activated by benzene treatment without inducing cell apoptosis in mouse bone marrow, which are partly under the regulation of the p53 gene.


Assuntos
Apoptose/efeitos dos fármacos , Benzeno/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Caspase 12/genética , Caspases/genética , Genes p53 , Animais , Proteínas Reguladoras de Apoptose/análise , Proteínas Reguladoras de Apoptose/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Caspases Iniciadoras , Fragmentação do DNA/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Leucócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA