Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Arch Toxicol ; 87(3): 505-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23052197

ABSTRACT

The traditional 2-year cancer bioassay needs replacement by more cost-effective and predictive tests. The use of toxicogenomics in an in vitro system may provide a more high-throughput method to investigate early alterations induced by carcinogens. Recently, the differential gene expression response in wild-type and cancer-prone Xpa (-/-) p53 (+/-) primary mouse hepatocytes after exposure to benzo[a]pyrene (B[a]P) revealed downregulation of cancer-related pathways in Xpa (-/-) p53 (+/-) hepatocytes only. Here, we investigated pathway regulation upon in vivo B[a]P exposure of wild-type and Xpa (-/-) p53 (+/-) mice. In vivo transcriptomics analysis revealed a limited gene expression response in mouse livers, but with a significant induction of DNA replication and apoptotic/anti-apoptotic cellular responses in Xpa (-/-) p53 (+/-) livers only. In order to be able to make a meaningful in vivo-in vitro comparison we estimated internal in vivo B[a]P concentrations using DNA adduct levels and physiologically based kinetic modeling. Based on these results, the in vitro concentration that corresponded best with the internal in vivo dose was chosen. Comparison of in vivo and in vitro data demonstrated similarities in transcriptomics response: xenobiotic metabolism, lipid metabolism and oxidative stress. However, we were unable to detect cancer-related pathways in either wild-type or Xpa (-/-) p53 (+/-) exposed livers, which were previously found to be induced by B[a]P in Xpa (-/-) p53 (+/-) primary hepatocytes. In conclusion, we showed parallels in gene expression responses between livers and primary hepatocytes upon exposure to equivalent concentrations of B[a]P. Furthermore, we recommend considering toxicokinetics when modeling a complex in vivo endpoint with in vitro models.


Subject(s)
Benzo(a)pyrene/toxicity , Carcinogenicity Tests/methods , Carcinogens/toxicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes/drug effects , Liver Neoplasms/chemically induced , Liver/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzo(a)pyrene/pharmacokinetics , Carcinogens/pharmacokinetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Computer Simulation , DNA Adducts/metabolism , DNA Replication/drug effects , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Hepatocytes/pathology , High-Throughput Screening Assays , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Primary Cell Culture , Risk Assessment , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/genetics , Xeroderma Pigmentosum Group A Protein/genetics
2.
Carcinogenesis ; 22(4): 619-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11285198

ABSTRACT

The effects of the food mutagen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) were studied in DNA repair deficient XPA(-/-) mice. The nullizygous XPA-knockout mice, which lack a functional nucleotide excision repair (NER) pathway, were exposed to dietary concentrations ranging from 10 to 200 p.p.m. The results show that PhIP is extremely toxic to XPA(-/-) mice, even at doses 10-fold lower than tolerated by wild-type C57BL/6 mice. XPA(-/-) mice rapidly lost weight and died within 2 and 6 weeks upon administration of 200 and 100 p.p.m., respectively. Intestinal abnormalities like distended and overfilled ileum and caecum, together with clear signs of starvation, suggests that the small intestines were the primary target tissue for the severe toxic effects. Mutation analysis in XPA(-/-) mice carrying a lacZ reporter gene, indicated that the observed toxicity of PhIP might be caused by genotoxic effects in the small intestine. LacZ mutant frequencies appeared to be selectively and dose-dependently increased in the intestinal DNA of treated XPA(-/-) mice. Furthermore, DNA repair deficient XPC(-/-) mice, which are still able to repair DNA damage in actively transcribed genes, did not display any toxicity upon treatment with PhIP (100 p.p.m.). This suggests that transcription coupled repair of DNA damage (PhIP adducts) in active genes plays a crucial role in preventing the intestinal toxicity of PhIP. Finally, PhIP appeared to be carcinogenic for XPA(-/-) mice at subtoxic doses. Upon treatment of the mice for 6 months with 10 or 25 p.p.m. PhIP, significantly increased tumour incidences were observed after a total observation period of one year. At 10 p.p.m. only lymphomas were found, whereas at 25 p.p.m. some intestinal tumours (adenomas and adenocarcinomas) were also observed.


Subject(s)
Carcinogens/toxicity , DNA Repair/genetics , Imidazoles/toxicity , Intestines/drug effects , Mutagens/toxicity , Adenocarcinoma/chemically induced , Adenoma/chemically induced , Animals , Body Weight/drug effects , DNA Mutational Analysis , Dose-Response Relationship, Drug , Female , Genes, Reporter , Genotype , Ileum/drug effects , Intestinal Neoplasms/chemically induced , Intestine, Small/drug effects , Lac Operon , Lymphoma/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors , Time Factors , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...