Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 154
1.
J Autoimmun ; 86: 75-92, 2018 01.
Article En | MEDLINE | ID: mdl-28931462

Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.


Diabetes Mellitus, Type 1/immunology , Muscles/immunology , Polymyositis/immunology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoantigens/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immune Tolerance , Mice , Mice, Inbred NOD , Mice, Transgenic , Organ Specificity , Transcription Factors/genetics , AIRE Protein
2.
J Toxicol Sci ; 39(6): 837-48, 2014.
Article En | MEDLINE | ID: mdl-25374375

Recent studies have shown that epigenetic alterations correlate with carcinogenesis in various tissues. Identification of these alterations might help characterize the early stages of carcinogenesis. We comprehensively analyzed DNA methylation and gene expression in livers obtained from rats exposed to nitrosodiethylamine (DEN) followed by a promoter of hepatic carcinogenesis, phenobarbital (PB). The combination of DEN and PB induced marked increases in number and area of glutathione S-transferase-placental form (GST-P)-positive foci in the liver. In the liver of rats that received 30 mg/kg of DEN, pathway analysis revealed alterations of common genes in terms of gene expression and DNA methylation, and that these alterations were related to immune responses. Hierarchical clustering analysis of the expression of common genes from public data obtained through the Toxicogenomics Project-Genomics Assisted Toxicity Evaluation system (TG-GATEs) showed that carcinogenic compounds clustered together. MBD-seq and GeneChip analysis indicated that major histocompatibility complex class Ib gene RT1-CE5, which has an important role in antigen presentation, was hypomethylated around the promoter region and specifically induced in the livers of DEN-treated rats. Further, immunohistochemical analysis indicated that the co-localization of GST-P and protein homologous to RT1-CE5 was present at the foci of some regions. These results suggest that common genes were altered in terms of both DNA methylation and expression in livers, with preneoplastic foci indicating carcinogenic potential, and that immune responses are involved in early carcinogenesis. In conclusion, the present study identified a specific profile of DNA methylation and gene expression in livers with preneoplastic foci. Early epigenetic perturbations of immune responses might correlate with the early stages of hepatocarcinogenesis.


Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , DNA Methylation , Diethylnitrosamine/toxicity , Epigenesis, Genetic/genetics , Gene Expression/genetics , Liver Neoplasms/genetics , Liver/metabolism , Animals , Carcinogenesis/immunology , Carcinogenesis/pathology , Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Glutathione Transferase/metabolism , Liver/enzymology , Liver Neoplasms/chemically induced , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Phenobarbital/toxicity , Rats, Sprague-Dawley
3.
J Toxicol Sci ; 39(5): 785-94, 2014.
Article En | MEDLINE | ID: mdl-25242409

We previously reported a toxicogenomics-based prediction model for hepatocarcinogens in which the expression patterns of signature genes following repeated doses of either genotoxic or non genotoxic compounds were similar. Based on the results of our prediction model, we hypothesized that repeated doses of non-genotoxic carcinogens might have initiating potential. Here, we conducted a two stage hepatocarcinogenesis study in rats exposed to the initiating agent nitrosodiethylamine (DEN), and hepatotoxic compounds thioacetamide (TAA), methapyrilene (MP) and acetaminophen (APAP) for 1-2 weeks followed by the liver tumor promoter phenobarbital (PB). The duration of initial treatment was determined based on positive results from our prediction model. Combined treatment of 3 or 30 mg/kg of genotoxic DEN and PB induced marked increases in altered hepatocellular foci and a DEN dose-dependent increase in the number and area of glutathione S-transferase-placental form (GST-P)-positive foci. A low number of altered hepatocellular foci were also observed in rats treated with TAA at a dose of 45 mg/kg.MP at a dose of 100 mg/kg induced a very low number of foci, but APAP did not. Hierarchical clustering analysis using gene expression data revealed that 2-week treatment with TAA at a dose of 30 mg/kg and MP at 45 mg/kg induced specific expression of DNA damage-related genes, similar to 1-week treatment with DEN at a dose of 30 mg/kg. These results suggest that TAA and MP induce DNA damage, which partially supports our hypothesis. Although this study does not indicate whether tumor growth in response to these compounds can be assessed in this model, our results suggest that cumulative treatment with non genotoxic TAA might have initiating potential in the liver.


Carcinoma, Hepatocellular/chemically induced , Liver Neoplasms/chemically induced , Methapyrilene/toxicity , Mutagenicity Tests/methods , Thioacetamide/toxicity , Acetaminophen/toxicity , Animals , Carcinoma, Hepatocellular/genetics , DNA Damage/drug effects , DNA Damage/genetics , Diethylnitrosamine/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Liver Neoplasms/genetics , Male , Oxidative Stress/genetics , Phenobarbital/toxicity , Rats, Sprague-Dawley
4.
J Toxicol Sci ; 39(1): 129-40, 2014 Feb.
Article En | MEDLINE | ID: mdl-24418717

Indole-3-carbinol (I3C) and phenobarbital (PB) are cytochrome P450 (CYP) 1A and CYP2B inducers, respectively, and have liver tumor-promoting effects in rats. In this study, we investigated the modifying effects on tumor promotion by I3C and PB co-administration. Six-week-old male F344 rats received a single intraperitoneal injection of N-diethylnitrosamine for initiation treatment. Two weeks after the initiation, rats were given no tumor-promoting agents (DEN alone), I3C (2,500 or 5,000 ppm in diet), PB (60 or 120 ppm in drinking water), or 2,500 ppm I3C + 60 ppm PB for 6 weeks. One week after the I3C/PB treatments, all animals underwent a two-thirds partial hepatectomy. The number and area of liver cell foci positive for glutathione S-transferase placental form (GST-P(+) foci) were not significantly fluctuated in the PB+I3C group in the isoadditive statistical model. On the contrary, the mRNA levels of Cyp2b1/2 and Nqo1 were suppressed and enhanced, respectively, in the PB+I3C group in the isoadditive model, but there was no enhancement in the microsomal reactive oxygen species (ROS) production, thiobarbituric acid-reactive substance levels, and Ki-67(+) cell ratio in this group. The results suggest that the co-administration of I3C and PB causes no modifying effects in liver tumor promotion in rats.


Anticarcinogenic Agents/pharmacology , Indoles/pharmacology , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Phenobarbital/pharmacology , Animals , Anticarcinogenic Agents/administration & dosage , Carcinogens , Cytochrome P-450 CYP2B1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Diethylnitrosamine , Enzyme Induction/drug effects , Glutathione Transferase/metabolism , Indoles/administration & dosage , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phenobarbital/administration & dosage , Rats , Rats, Inbred F344
5.
Exp Toxicol Pathol ; 66(1): 1-11, 2014 Jan.
Article En | MEDLINE | ID: mdl-23890812

We have previously reported that 28-day treatment with hepatocarcinogens increases liver cells expressing p21(Cip1), a G1/S checkpoint protein, and M phase proteins, i.e., nuclear Cdc2, Aurora B, phosphorylated-Histone H3 (p-Histone H3) and heterochromatin protein 1α (HP1α), in rats. To examine the roles of these markers in the early stages of carcinogenesis, we investigated their cellular distribution in several carcinogenic target organs using rat two-stage carcinogenesis models. Promoting agents targeting the liver (piperonyl butoxide and methapyrilene hydrochloride), thyroid (sulfadimethoxine), urinary bladder (phenylethyl isothiocyanate), and forestomach and glandular stomach (catechol) were administered to rats after initiation treatment for the liver with N-diethylnitrosamine, thyroid with N-bis(2-hydroxypropyl)nitrosamine, urinary bladder with N-butyl-N-(4-hydroxybutyl)nitrosamine, and forestomach and glandular stomach with N-methyl-N'-nitro-N-nitrosoguanidine. Numbers of cells positive for nuclear Cdc2, Aurora B, p-Histone H3 and HP1α increased within preneoplastic lesions as determined by glutathione S-transferase placental form in the liver or phosphorylated p44/42 mitogen-activated protein kinase in the thyroid, and hyperplastic lesions having no known preneoplastic markers in the urinary bladder, forestomach and glandular stomach. Immunoreactive cells for p21(Cip1) were decreased within thyroid preneoplastic lesions; however, they were increased within liver preneoplastic lesions and hyperplastic lesions in other organs. These results suggest that M phase disruption commonly occur during the formation of preneoplastic lesions and hyperplastic lesions. Differences in the expression patterns of p21(Cip1) between thyroid preneoplastic and proliferative lesions in other organs may reflect differences in cell cycle regulation involving G1/S checkpoint function between proliferative lesions in each organ.


Carcinogenesis/metabolism , Cell Division/physiology , Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Precancerous Conditions/metabolism , Animals , Carcinogens/toxicity , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Immunohistochemistry , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Neoplasms/chemically induced , Precancerous Conditions/chemically induced , Rats , Rats, Inbred F344 , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/metabolism , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/metabolism
6.
Arch Toxicol ; 88(2): 443-54, 2014 Feb.
Article En | MEDLINE | ID: mdl-23892646

To examine the developmental exposure effect of nicotine (NIC) on hippocampal neurogenesis, pregnant Sprague-Dawley rats were treated with (-)-NIC hydrogen tartrate salt through drinking water at 2, 10 or 50 ppm from gestational day 6 to day 21 after delivery. On postnatal day (PND) 21, immunohistochemically doublecortin (Dcx)(+) cells increased at ≥10 ppm in the dentate subgranular zone (SGZ) as examined in male offspring; however, dihydropyrimidinase-like 3 (TUC4)(+) cells decreased at 2 ppm, and T box brain 2 (Tbr2)(+) cells were unchanged at any dose. Double immunohistochemistry revealed decreases in TUC4(+)/Dcx(+) and TUC4(+)/Dcx(-) cells, an increase in TUC4(-)/Dcx(+) cells at 2 and 10 ppm and an increase in Tbr2(-)/Dcx(+) cells at 50 ppm, suggesting an increase in type-3 progenitor cells at ≥2 ppm and decrease in immature granule cells at 2 and 10 ppm. The number of mature neuron-specific NeuN(-) progenitor cells expressing nicotinic acetylcholine receptor α7 in the SGZ and mRNA levels of Chrna7 and Chrnb2 in the dentate gyrus was unchanged at any dose, suggesting a lack of direct nicotinic stimulation on progenitor cells. In the dentate hilus, glutamic acid decarboxylase 67(+) interneurons increased at ≥10 ppm. All changes disappeared on PND 77. Therefore, maternal exposure to NIC reversibly affects hippocampal neurogenesis targeting late-stage differentiation in rat offspring. An increase in interneurons suggested that their activation affected granule cell differentiation. The lowest observed adverse effect level was at 2 ppm (0.091 mg/kg/day as a free base) by the affection of hippocampal neurogenesis at ≥2 ppm.


Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Neurons/drug effects , Nicotine/toxicity , Animals , Animals, Newborn , Body Weight/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cotinine/urine , Dentate Gyrus/metabolism , Dose-Response Relationship, Drug , Doublecortin Domain Proteins , Doublecortin Protein , Eating/drug effects , Female , Male , Maternal Exposure/adverse effects , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism
7.
J Toxicol Sci ; 38(5): 679-88, 2013.
Article En | MEDLINE | ID: mdl-24025784

Phenobarbital (PB) is a cytochrome P450 (CYP) 2B inducer, and piperonyl butoxide (PBO) is a CYP1A/2B inducer. These inducers have liver tumor-promoting effects in rats. In this study, we performed a rat two-stage liver carcinogenesis bioassay to examine the tumor-promoting effect of PB and PBO co-administration. Male rats received an intraperitoneal injection of N-diethylnitrosamine (DEN) for initiation. Two weeks after DEN administration, rats were given PB (60 or 120 ppm in drinking water), PBO (1,250 or 2,500 ppm in diet) or 60 ppm PB+1,250 ppm PBO for 6 weeks. One week after the PB/PBO treatment, all rats were subjected to a two-thirds partial hepatectomy. To evaluate the effect of the combined administration, we used two statistical additive models. In the isoadditive model, the average values of the area of GST-P positive foci in the PB+PBO group were significantly lower than those in the High PB or High PBO groups. In the heteroadditive model, the net values of Cyp1a1 mRNA level and microsomal reactive oxygen species (ROS) production in the PB+PBO group were significantly lower than the sum of those in the Low PB or Low PBO groups. On the contrary, there was no interactive effect in the PCNA-positive hepatocyte ratio, mRNA levels of Cyp2b1/2, Gstm3, Gpx2 and Nqo1, and the level of thiobarbituric acid-reactive substances in the PB+PBO group. These results suggest that PB and PBO co-administration causes suppressive effects in liver tumor-promoting activity in rats resulting from inhibited microsomal ROS production because of suppression of CYP1A induction.


Carcinoma, Hepatocellular/prevention & control , Cytochrome P-450 CYP1A1/biosynthesis , Liver Neoplasms/prevention & control , Phenobarbital/administration & dosage , Piperonyl Butoxide/administration & dosage , Reactive Oxygen Species/metabolism , Animals , Carcinoma, Hepatocellular/chemically induced , Cell Proliferation/drug effects , Depression, Chemical , Diethylnitrosamine , Disease Models, Animal , Drug Therapy, Combination , Enzyme Induction/drug effects , Glutathione S-Transferase pi/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/etiology , Male , Microsomes, Liver/metabolism , Phenobarbital/pharmacology , Piperonyl Butoxide/pharmacology , Rats , Rats, Inbred F344
8.
Toxicol Lett ; 222(3): 295-302, 2013 Oct 09.
Article En | MEDLINE | ID: mdl-23968726

Disruptive epigenetic gene control has been shown to be involved in carcinogenesis. To identify key molecules in piperonyl butoxide (PBO)-induced hepatocarcinogenesis, we searched hypermethylated genes using CpG island (CGI) microarrays in non-neoplastic liver cells as a source of proliferative lesions at 25 weeks after tumor promotion with PBO using mice. We further performed methylation-specific polymerase chain reaction (PCR), real-time reverse transcription PCR, and immunohistochemical analysis in PBO-promoted liver tissues. Ebp4.1, Wdr6 and Cmtm6 increased methylation levels in the promoter region by PBO promotion, although Cmtm6 levels were statistically non-significant. These results suggest that PBO promotion may cause altered epigenetic gene regulation in non-neoplastic liver cells surrounding proliferative lesions to allow the facilitation of hepatocarcinogenesis. Both Wdr6 and Cmtm6 showed decreased expression in non-neoplastic liver cells in contrast to positive immunoreactivity in the majority of proliferative lesions produced by PBO promotion. These results suggest that both Wdr6 and Cmtm6 were spared from epigenetic gene modification in proliferative lesions by PBO promotion in contrast to the hypermethylation-mediated downregulation in surrounding liver cells. Considering the effective detection of proliferative lesions, these molecules could be used as detection markers of hepatocellular proliferative lesions and played an important role in hepatocarcinogenesis.


Carcinogens/toxicity , DNA Methylation/drug effects , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Piperonyl Butoxide/toxicity , Animals , Carcinogenicity Tests , CpG Islands/drug effects , Male , Mice , Mice, Inbred ICR , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Toxicol Sci ; 136(1): 154-65, 2013 Nov.
Article En | MEDLINE | ID: mdl-23976782

We have shown that maternal manganese (Mn) exposure caused sustained disruption of hippocampal neurogenesis of mouse offspring. To clarify the effects of maternal Mn exposure on epigenetic gene regulation contributing to the sustained disruption of hippocampal neurogenesis, we treated pregnant ICR mice with MnCl2 in diet from gestational day 10 through day 21 after delivery on weaning and searched epigenetically downregulated genes by global promoter methylation analysis in the hippocampal dentate gyrus of male offspring on postnatal day (PND) 21 and PND 77. By CpG promoter microarray analysis on PND 21 following 800-ppm Mn exposure, sustained promoter hypermethylation and transcript downregulation through PND 77 were confirmed with Mid1, Atp1a3, and Nr2f1, whereas Pvalb showed a transient hypermethylation only on weaning. The numbers of Pvalb⁺ and ATP1a3⁺ neurons suggestive of γ-aminobutyric acid (GABA)ergic interneurons, Mid1⁺ cells suggestive of late-stage granule cell lineage and GABAergic interneurons, and COUP-TF1⁺ cells suggestive of early-stage granule cell lineage were all reduced on PND 21, and reductions were sustained on PND 77 except for no change in Pvalb⁺ cells. Mid1⁺ cells showed asymmetric distribution with right-side predominance, and Mn exposure abolished it by promoter hypermethylation of the right side. These findings indicate epigenetic mechanisms as mediators, through which Mn exposure modulates neurogenesis involving both granule cell lineage and GABAergic interneurons with long-lasting and stable repercussions. Disruption of asymmetric cellular distribution of Mid1 suggests that higher brain functions specialized in the left or right side of the brain were affected.


Chlorides/toxicity , Dentate Gyrus/drug effects , Epigenesis, Genetic/drug effects , Manganese Poisoning/etiology , Neurogenesis/drug effects , Neurons/drug effects , Age Factors , Animals , Animals, Newborn , COUP Transcription Factor I/genetics , COUP Transcription Factor I/metabolism , CpG Islands , DNA Methylation/drug effects , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Developmental/drug effects , Gestational Age , Male , Manganese Compounds , Manganese Poisoning/genetics , Manganese Poisoning/physiopathology , Maternal Exposure , Mice , Mice, Inbred ICR , Neurogenesis/genetics , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Parvalbumins/genetics , Parvalbumins/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , RNA, Messenger/metabolism , Reproducibility of Results , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Ubiquitin-Protein Ligases , Weaning
10.
Chem Biol Interact ; 205(2): 108-18, 2013 Sep 25.
Article En | MEDLINE | ID: mdl-23830814

To investigate the protective effect of α-lipoic acid (a-LA) on the hepatocarcinogenic process promoted by thioacetamide (TAA), we used a two-stage liver carcinogenesis model in N-diethylnitrosamine (DEN)-initiated and TAA-promoted rats. We examined the modifying effect of co-administered a-LA on the liver tissue environment surrounding preneoplastic hepatocellular lesions, with particular focus on hepatic macrophages and the mechanism behind the decrease in apoptosis of cells surrounding preneoplastic hepatocellular lesions during the early stages of hepatocellular tumor promotion. TAA increased the number and area of glutathione S-transferase placental form (GST-P)(+) liver cell foci and the numbers of proliferating and apoptotic cells in the liver. Co-administration with a-LA suppressed these effects. TAA also increased the numbers of ED2(+), cyclooxygenase-2(+), and heme oxygenase-1(+) hepatic macrophages as well as the number of CD3(+) lymphocytes. These effects were also suppressed by a-LA. Transcript levels of some inflammation-related genes were upregulated by TAA and downregulated by a-LA in real-time RT-PCR analysis. Outside the GST-P(+) foci, a-LA reduced the numbers of apoptotic cells, active caspase-8(+) cells and death receptor (DR)-5(+) cells. These results suggest that hepatic macrophages producing proinflammatory factors may be activated in TAA-induced tumor promotion. a-LA may suppress tumor-promoting activity by suppressing the activation of these macrophages and the subsequent inflammatory responses. Furthermore, a-LA may suppress tumor-promoting activity by suppressing the DR5-mediated extrinsic pathway of apoptosis and the subsequent regeneration of liver cells outside GST-P(+) foci.


Anticarcinogenic Agents/pharmacology , Carcinogens/antagonists & inhibitors , Carcinogens/toxicity , Liver Neoplasms, Experimental/prevention & control , Thioctic Acid/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Animals , Apoptosis/drug effects , Cocarcinogenesis , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Diethylnitrosamine/toxicity , Glutathione S-Transferase pi/metabolism , Inflammation Mediators/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Rats , Rats, Inbred F344 , Thioacetamide/antagonists & inhibitors , Thioacetamide/toxicity , Thiobarbituric Acid Reactive Substances/metabolism
11.
J Toxicol Sci ; 38(3): 403-13, 2013.
Article En | MEDLINE | ID: mdl-23665939

Orphenadrine (ORPH), an anticholinergic agent, is a cytochrome P450 (CYP) 2B inducer. CYP2B inducers are known to have liver tumor-promoting effects in rats. In this study, we performed a rat two-stage liver carcinogenesis bioassay to examine the tumor-promoting effect of ORPH and to clarify its possible mechanism of action. Male rats were given a single intraperitoneal injection of N-diethylnitrosamine (DEN) as an initiation treatment. Two weeks after DEN administration, rats were fed a diet containing ORPH (0, 750, or 1,500 ppm) for 6 weeks. One week after the ORPH-administration rats were subjected to two-thirds partial hepatectomy for the acceleration of hepatocellular proliferation. The number and area of glutathione S-transferase placental form-positive foci significantly increased in the DEN-ORPH groups. Real-time RT-PCR revealed increased mRNA expression levels of Cyp2b1/2, Mrp2 and Cyclin D1 in the DEN-ORPH groups and of Gpx2 and Gstm3 in the DEN-High ORPH group. Microsomal reactive oxygen species (ROS) production and oxidative stress markers such as thiobarbituric acid-reactive substances and 8-hydroxydeoxyguanosine were increased in the DEN-High ORPH group. Immunohistochemically, constitutively active/androstane receptor (CAR) were clearly localized in the nuclei of hepatocytes in the DEN-ORPH groups. These results suggest that ORPH causes nuclear translocation of CAR resulting in the induction of the liver tumor-promoting activity. Furthermore, oxidative stress resulting from ROS production is also involved in the liver tumor-promoting activity of ORPH.


Cholinergic Antagonists/toxicity , Liver Neoplasms/chemically induced , Muscarinic Antagonists/toxicity , Orphenadrine/toxicity , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cell Nucleus/metabolism , Cholinergic Antagonists/administration & dosage , Constitutive Androstane Receptor , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Hepatocytes/cytology , Hepatocytes/metabolism , Male , Muscarinic Antagonists/administration & dosage , Orphenadrine/administration & dosage , Rats , Rats, Inbred F344
12.
J Toxicol Sci ; 38(3): 415-24, 2013.
Article En | MEDLINE | ID: mdl-23665940

Phenobarbital (PB) and orphenadrine (ORPH) are cytochrome P450 (CYP) 2B inducers and have liver tumor-promoting effects in rats. In this study, we performed a rat two-stage liver carcinogenesis bioassay to examine the tumor-promoting effect of PB and ORPH co-administration. Twelve male rats per group were given an intraperitoneal injection of N-diethylnitrosamine (DEN) for initiation. Two-week after DEN administration, rats were given PB (60 or 120 ppm in drinking water), ORPH (750 or 1,500 ppm in diet) or 60 ppm PB+750 ppm ORPH for 6-week. One-week after the PB/ORPH treatment, all rats were subjected to two-thirds partial hepatectomy. To evaluate the effect of the combined administration, we used two statistical models: a heteroadditive model and an isoadditive model. In the heteroadditive model, the net values of the number and area of glutathione S-transferase placental form (GST-P) positive foci, Cyp2b1/2, Gstm3 and Gpx2 mRNA levels, microsomal reactive oxygen species (ROS) production and thiobarbituric acid-reactive substances level in the PB+ORPH group were significantly higher than the sum of the net values of those in the Low PB and Low ORPH groups. In the isoadditive model, the average values of the area of GST-P positive foci and PCNA positive hepatocyte ratio and Gstm3 mRNA level in the PB+ORPH group were significantly higher than the average values of those in the High PB and High ORPH groups. These results suggest that PB and ORPH co-administration causes synergistic effects in liver tumor-promoting activity in rats resulting from oxidative stress due to enhanced microsomal ROS production.


Liver Neoplasms/chemically induced , Orphenadrine/toxicity , Phenobarbital/toxicity , Animals , Cytochrome P-450 CYP2B1/metabolism , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Drug Combinations , Drug Synergism , Enzyme Induction/drug effects , Male , Microsomes, Liver/metabolism , Orphenadrine/administration & dosage , Oxidative Stress/genetics , Phenobarbital/administration & dosage , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
13.
J Toxicol Sci ; 38(3): 431-43, 2013.
Article En | MEDLINE | ID: mdl-23665942

For molecular analysis in anatomically-specific brain regions for rodent studies, it is necessary to establish a fast and accurate procedure for tissue sampling to achieve high integrity and expression fidelity of extracted molecules. The present study was performed to examine suitability of whole brain fixation with methacarn and subsequent tissue sampling using punch-biopsy devices for gene expression analysis in rats. After fixation, each specific region, i.e., hippocampal dentate gyrus, corpus callosum, cingulate cortex or cerebellar vermis was collected, and the integrity and variability of expression data of extracted total RNAs and polypeptides were examined. Methacarn fixation, acetone fixation, and unfixed tissues were compared. Methacarn fixation resulted in high integrity of total RNAs sufficient for conducting global expression analysis and superior in terms of uniformity in the integrity among brain regions to that of acetone fixation. Extracted polypeptide after methacarn fixation revealed similar integrity to that without fixation or with acetone fixation. Methacarn fixation resulted in lower mRNA expression variability between samples than acetone fixation in microarray analysis. The fidelity of polypeptide expression was mostly equivalent between methacarn and acetone fixation in 2-dimensional differential in-gel electrophoresis, although the expression levels of a small number of polypeptides from acetone-fixed tissues were affected. These results suggest that whole brain fixation with methacarn retains advantages for global analyses of mRNAs and polypeptides in rodent studies.


Acetic Acid , Brain/metabolism , Chloroform , Fixatives , Gene Expression Profiling/methods , Methanol , Peptides/analysis , RNA, Messenger/analysis , Animals , Chlorobutanol , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Tissue Fixation
14.
Toxicol Sci ; 134(1): 140-54, 2013 Jul.
Article En | MEDLINE | ID: mdl-23596259

To investigate the neurotoxicity profile of glycidol and its effect on developmental hippocampal neurogenesis, pregnant Sprague Dawley rats were given drinking water containing 0, 100, 300, or 1000 ppm glycidol from gestational day 6 until weaning on day 21 after delivery. At 1000 ppm, dams showed progressively worsening gait abnormalities, and histopathological examination showed generation of neurofilament-L(+) spheroids in the cerebellar granule layer and dorsal funiculus of the medulla oblongata, central chromatolysis in the trigeminal nerve ganglion cells, and axonal degeneration in the sciatic nerves. Decreased dihydropyrimidinase-like 3(+) immature granule cells in the subgranular zone (SGZ) and increased immature reelin(+) or calbindin-2(+) γ-aminobutyric acid-ergic interneurons and neuron-specific nuclear protein (NeuN)(+) mature neurons were found in the dentate hilus of the offspring of the 1000 ppm group on weaning. Hilar changes remained until postnatal day 77, with the increases in reelin(+) and NeuN(+) cells being present at ≥ 300 ppm, although the SGZ change disappeared. Thus, glycidol caused axon injury in the central and peripheral nervous systems of adult rats, suggesting that glycidol targets the newly generating nerve terminals of immature granule cells, resulting in the suppression of late-stage hippocampal neurogenesis. The sustained hilar changes may be a sign of continued aberrations in neurogenesis and migration. The no-observed-adverse-effect level was determined to be 300 ppm (48.8mg/kg body weight/day) for dams and 100 ppm (18.5mg/kg body weight/day) for offspring. The sustained developmental exposure effect on offspring neurogenesis was more sensitive than the adult axonal injury.


Axons/drug effects , Cell Differentiation/drug effects , Epoxy Compounds/toxicity , Hippocampus/drug effects , Neurogenesis/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Propanols/toxicity , Aging/pathology , Animals , Apoptosis/drug effects , Axons/pathology , Dose-Response Relationship, Drug , Female , Hippocampus/embryology , Hippocampus/growth & development , Hippocampus/pathology , Male , Neurons/drug effects , Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley , Reelin Protein
15.
Toxicol Lett ; 219(3): 203-10, 2013 Jun 07.
Article En | MEDLINE | ID: mdl-23535288

We have previously reported that hepatocarcinogens increase liver cells expressing p21(Cip1), a G1 checkpoint protein and M phase proteins after 28-day treatment in rats. This study aimed to identify early prediction markers of carcinogens available in many target organs after 28-day treatment in rats. Immunohistochemical analysis was performed on Ki-67, p21(Cip1) and M phase proteins [nuclear Cdc2, phospho-Histone H3 (p-Histone H3), Aurora B and heterochromatin protein 1α (HP1α)] with carcinogens targeting different organs. Carcinogens targeting thyroid (sulfadimethoxine; SDM), urinary bladder (phenylethyl isothiocyanate), forestomach (butylated hydroxyanisole; BHA), glandular stomach (catechol; CC), and colon (2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine and chenodeoxycholic acid) were examined using a non-carcinogenic toxicant (caprolactam) and carcinogens targeting other organs as negative controls. All carcinogens increased Ki-67(+), nuclear Cdc2(+), p-Histone H3(+) or Aurora B(+) carcinogenic target cells, except for both colon carcinogens, which did not increase cell proliferation. On the other hand, p21(Cip1+) cells increased with SDM and CC. HP1α responded only to BHA. Results revealed carcinogens evoking cell proliferation concurrently induced cell cycle arrest at M phase or showing chromosomal instability reflecting aberration in cell cycle regulation, irrespective of target organs, after 28-day treatment. Therefore, M phase proteins may be early prediction markers of carcinogens evoking cell proliferation in many target organs.


Carcinogens/pharmacology , Cell Division/drug effects , Animals , Aurora Kinase B , Aurora Kinases , Biomarkers/analysis , Butylated Hydroxyanisole/pharmacology , CDC2 Protein Kinase , Catechols/pharmacology , Cell Cycle Proteins/analysis , Cell Division/physiology , Cell Proliferation/drug effects , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/analysis , Cyclin-Dependent Kinases , Histones/analysis , Imidazoles/pharmacology , Isothiocyanates/pharmacology , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/chemistry , Protein Serine-Threonine Kinases/analysis , Pyridines/pharmacology , Rats , Rats, Inbred F344 , Sulfadimethoxine/pharmacology
16.
Arch Toxicol ; 87(6): 1141-53, 2013 Jun.
Article En | MEDLINE | ID: mdl-23411599

Some hepatocarcinogens induce cytomegaly, which reflects aberrant cell cycling and increased ploidy, from the early stages of administration to animals. To clarify the regulatory molecular mechanisms behind cell cycle aberrations related to the early stages of hepatocarcinogenesis, we performed gene expression analysis using microarrays and real-time reverse transcription polymerase chain reaction followed by immunohistochemical analysis in the livers of rats treated with the cytomegaly inducing hepatocarcinogens thioacetamide (TAA), fenbendazole, and methyleugenol, the cytomegaly non-inducing hepatocarcinogen piperonyl butoxide (PBO), or the non-carcinogenic hepatotoxicants acetaminophen and α-naphthyl isothiocyanate, for 28 days. Gene expression profiling showed that cell cycle-related genes, especially those of G(2)/M phase, were mostly upregulated after TAA treatment. Immunohistochemical analysis was performed on cell cycle proteins that were upregulated by TAA treatment and on related proteins. All hepatocarcinogens, irrespective of their cytomegaly inducing potential, increased liver cells immunoreactive for p21(Cip1), which acts on cells arrested in G(1) phase, and for Aurora B or Incenp, which is suggestive of an increase in a cell population with chromosomal instability caused by overexpression. PBO did not induce cell proliferation after 28-day treatment. Hepatocarcinogens that induced cell proliferation after 28-day treatment also caused an increase in p53(+) cells in parallel with increased apoptotic cells, as well as increased population of cells expressing M phase-related proteins nuclear Cdc2, phospho-Histone H3, and HP1α. These results suggest that hepatocarcinogens may increase cellular populations arrested in G1 phase or showing chromosomal instability after 28-day treatment. Hepatocarcinogens that induce cell cycle facilitation may cause M phase arrest accompanied by apoptosis.


Carcinogens/toxicity , Cell Cycle Proteins/metabolism , Liver Neoplasms/chemically induced , Liver/drug effects , Animals , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Chromobox Protein Homolog 5 , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , M Phase Cell Cycle Checkpoints/drug effects , Male , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Reprod Toxicol ; 38: 25-36, 2013 Jul.
Article En | MEDLINE | ID: mdl-23428981

The effect of developmental exposure to chlorpyrifos (CPF) on hippocampal neurogenesis was examined in male mice after maternal dietary exposure to CPF at 0, 4, 20, or 100ppm from gestation day 10 to postnatal day (PND) 21. Cholinesterase activity was dose-dependently decreased in red blood cells at ≥4ppm and in the brain at 100ppm both in dams and offspring on PND 21. Immunohistochemically, doublecortin(+) cells were decreased at ≥20ppm in the subgranular zone (SGZ) of the dentate gyrus, and NeuN(+)-expressing mature neurons were decreased at 100ppm in the hilus on PND 21. There were no differences in the numbers of progenitor populations expressing Tbr2 or M1 muscarinic acetylcholine receptors. Transcript levels of Dcx also decreased at ≥20ppm, and those of Pcna, Casp3, Bax, Bcl2, Pax6 and Tbr2 were unchanged in the dentate gyrus by real-time RT-PCR. At PND 77, hippocampal neurogenesis was unchanged. These results suggest that developmental CPF exposure directly but transiently suppresses maturation of late-stage granule cell lineages in the SGZ and affects interneuron populations in the hilus.


Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Dentate Gyrus/drug effects , Insecticides/toxicity , Neurogenesis/drug effects , Animals , Cholinesterases/blood , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Doublecortin Protein , Female , Male , Maternal-Fetal Exchange , Mice , Pregnancy , Prenatal Exposure Delayed Effects , Prosencephalon/enzymology , Receptors, Muscarinic/metabolism , Stem Cells/metabolism
18.
Exp Toxicol Pathol ; 65(6): 845-52, 2013 Sep.
Article En | MEDLINE | ID: mdl-23290887

We previously reported that indole-3-carbinol (I3C) had hepatocellular tumor-promoting activity in a short-term (8 weeks) two-stage liver carcinogenesis model in rats. It was suggested that this effect was related to the production of reactive oxygen species (ROS) caused by cytochrome P450 1A (CYP1A) induction. In the present study, 0.5% I3C was administered to DEN-initiated rats for 26 weeks to examine the effect of prolonged administration of I3C and to clarify the possible mechanisms of I3C-induced hepatocarcinogenesis. The number and area of GST-P positive foci, ROS production, TBARS level, 8-OHdG content and mRNA levels of Ahr and Nrf2 gene batteries significantly increased in the DEN-I3C group compared with the DEN-alone group. Furthermore, some GST-P positive preneoplastic foci progressed to hepatocellular adenomas with the prolongation of I3C administration. Lack of PTEN and phospho-Smad2/3 expression and translocations of PDPK1 and phospho-Akt substrates to underneath the cell membrane were observed in the majority of hepatocellular adenomas. In addition, the number of Ki-67 positive cells increased in adenomas compared with the preneoplastic foci. These results suggest that the administration of I3C for 26 weeks in DEN-initiated rats induces tumor progression from hepatocellular altered foci to hepatocellular adenomas by ROS-mediated Akt activation that inhibits the TGF-ß/Smad signaling and results in the increased cell proliferation.


DNA Damage , Indoles/toxicity , Liver Neoplasms, Experimental/chemically induced , Oxidative Stress/drug effects , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Body Weight/drug effects , Cocarcinogenesis , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , Immunohistochemistry , Indoles/administration & dosage , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Organ Size/drug effects , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
19.
J Cancer Res Clin Oncol ; 139(3): 395-401, 2013 Mar.
Article En | MEDLINE | ID: mdl-23114882

PURPOSE: Some goitrogens promote thyroid carcinogenesis in rats in an initiation-promotion model; this model frequently produces carcinomas that invade fibrously thickened capsules, termed capsular invasive carcinomas (CICs). The present study tested a hypothesis that CICs originate from parenchymal proliferative lesions located beneath the capsule. METHODS: Cell proliferation activity, cell-cycle kinetics and cellular invasion were immunohistochemically examined in subcapsular proliferative lesions in male F344 rats treated with an anti-thyroid agent, propylthiouracil or sulfadimethoxine, during the tumor-promotion phase after initiation with N-bis(2-hydroxypropyl)nitrosamine. RESULTS: Focal follicular cell hyperplasias (FFCHs) were the most commonly observed parenchymal proliferative lesions. Subcapsular FFCHs located near CICs showed more Ki-67(+) cells in the capsular side than the contrary parenchymal center side. Most of these FFCHs located near CICs showed accumulated immunoreactivity for cyclin A, cyclin D, cyclin E and cyclin-dependent kinase-2, whereas most subcapsular FFCHs located elsewhere did not show such accumulated expression of cell-cycle molecules. Subcapsular FFCHs immunoreactive at the capsular front for tenascin-C, a tumor invasion marker of extracellular matrix protein, showed high proliferation activity. CONCLUSIONS: Subcapsular FFCH-forming cells can potentially spread directly into the fibrously thickened capsule to form CICs by accelerating cell-cycle activity.


Carcinoma/pathology , Disease Models, Animal , Hypothyroidism/complications , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Animals , Carcinogens , Carcinoma/chemically induced , Carcinoma/etiology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Disease Progression , Hyperplasia/etiology , Hyperplasia/pathology , Hypothyroidism/pathology , Male , Neoplasm Invasiveness , Nitrosamines , Rats , Rats, Inbred F344 , Thyroid Neoplasms/chemically induced , Thyroid Neoplasms/etiology
20.
Reprod Toxicol ; 35: 125-36, 2013 Jan.
Article En | MEDLINE | ID: mdl-23099338

To examine the effects of developmental exposure to chlorpyrifos (CPF) on neurogenesis in the hippocampal dentate gyrus, pregnant rats were treated with 2.8, 14 or 70 ppm CPF in the diet from gestational day 10 to day 21 after delivery. Dams had decreased cholinesterase (ChE) activities in red blood cells (RBC) at intakes of ≥2.8 ppm and in brain at 70 ppm. Offspring on postnatal day (PND) 21 had decreased ChE activities in the RBC and brain at 70 ppm. There were no behavioral abnormalities in the offspring. Immunohistochemical analysis showed decreases in the numbers of cells positive for proliferating cell nuclear antigen and T box brain 2 in the subgranular zone (SGZ) of the dentate gyrus on PND 21 at 70 ppm, while other progenitor cell populations and the apoptotic cell number were unaffected in this zone. However, on PND 77 all changes had disappeared. The distribution of the progenitor cell population expressing nicotinic acetylcholine receptor α7 and lacking expression of postmitotic neuron-specific nuclear protein was unchanged by CPF-exposure, suggesting no effect of cholinergic stimulation on neurogenesis. These results suggest that developmental exposure to CPF directly but transiently affect the proliferation of type-2 progenitor cell populations in the hippocampal neurogenesis. The lowest-observed-adverse-effect level (LOAEL) of CPF was determined to be 2.8 ppm (0.36 mg/kg body weight/day) for dams by the inhibition of ChE activity in the RBC at this dose. As for offspring, no-observed-adverse-effect level (NOAEL) was determined to be 14 ppm (1.86 mg/kg body weight/day) by the decrease of type-2 progenitor cell proliferation in the SGZ and the inhibition of ChE activity in the RBC and brain at 70 ppm. The NOAEL of dams based on the offspring's effects was approximately 2800 times higher than the estimated consumption of CPF through food in the general population and in pregnant women as examined in Japan.


Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Dentate Gyrus/cytology , Insecticides/toxicity , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation/drug effects , Cholinesterases/blood , Extracellular Matrix Proteins/metabolism , Female , Hand Strength , Interneurons/drug effects , Interneurons/metabolism , Male , Maternal-Fetal Exchange , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , No-Observed-Adverse-Effect Level , Pregnancy , Rats , Reelin Protein , Serine Endopeptidases/metabolism , Stem Cells/cytology , Thyroid Hormones/blood
...