Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Toxicol Pathol ; 43(5): 743-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25630681

ABSTRACT

Cell clusters were observed in the seminiferous tubules of C57BL/6J mice as a spontaneous lesion in a 2-week toxicity study, and they were demonstrated to be basically composed of Sertoli cells by immunohistochemistry for claudin-11 and GATA-4 (GATA-binding protein 4), which are both Sertoli cell markers. The clusters were composed of about 5 to 50 cells, which had eosinophilic and occasionally vacuolated cytoplasm with an unclear cell boundary. The cell clusters involved some sperm. No mitotic figures were observed and no immunoreactivity for proliferating cell nuclear antigen (PCNA) was detected in the clusters. In most cases, the cell clusters were observed in seminiferous tubules that also showed degenerative changes. In rare instances, cell aggregates immunohistochemically positive for claudin-11 were observed in the lumen of the epididymis, suggesting that some of the Sertoli cell clusters were sloughed off from the seminiferous epithelium into the epididymal ducts. To our knowledge, this is the first report of Sertoli cell clusters in any animal species except for transgenic or surgically altered animals.


Subject(s)
Seminiferous Tubules/cytology , Seminiferous Tubules/metabolism , Sertoli Cell Tumor/metabolism , Sertoli Cell Tumor/pathology , Sertoli Cells/chemistry , Animals , Claudins/analysis , Claudins/chemistry , GATA4 Transcription Factor/analysis , GATA4 Transcription Factor/chemistry , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Seminiferous Tubules/pathology , Sertoli Cell Tumor/chemistry
2.
Exp Toxicol Pathol ; 66(1): 1-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23890812

ABSTRACT

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.


Subject(s)
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
3.
Toxicol Lett ; 224(1): 64-72, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24120684

ABSTRACT

Ochratoxin A (OTA) is a renal carcinogen that induces karyomegaly in target renal tubular cells of the outer stripe of the outer medulla (OSOM). This study was performed to clarify the relationship between oxidative stress and the karyomegaly-inducing potential involving cell cycle aberration of OTA in the OSOM. Rats were treated with OTA for 28 days in combination with enzymatically modified isoquercitrin (EMIQ) or α-lipoic acid (ALA) as antioxidants. OTA increased the mRNA levels of the antioxidant enzyme-related genes Gpx1, Gpx2, Gstm1 and Nfe2l2, but did not increase the levels of Gsta5, Keap1, Nqo1, Hmox1, Aldh1a1, Por, Prdx1 and Txn1. OTA also did not change the levels of thiobarbituric acid-reactive substances, glutathione disulfide/reduced glutathione, and the immunoreactive tubular cell distribution of nuclear factor erythroid 2-related factor 2 in the OSOM. Co-treatment with EMIQ or ALA did not cause any changes in these parameters. As previously reported, OTA increased cell proliferation activity, apoptosis and immunohistochemical cellular distributions of molecules suggestive of induction of DNA damage and cell cycle aberrations involving spindle checkpoint disruption and cell cycle arrest. However, co-treatment with EMIQ or ALA did not suppress these changes, and ALA co-treatment increased the cell proliferation activity induced by OTA. These results suggest that OTA facilitates cell cycling involving cell cycle aberrations and apoptosis as a basis of the mechanism behind the development of karyomegaly and subsequent carcinogenicity targeting the OSOM, without relation to induction of oxidative stress. On the other hand, ALA may promote the OTA-induced proliferation of carcinogenic target cells.


Subject(s)
Cell Nucleus/drug effects , Kidney Tubules/drug effects , Ochratoxins/toxicity , Oxidative Stress/drug effects , Animals , Cell Cycle/drug effects , Cell Nucleus/pathology , Glutathione/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Rats , Rats, Inbred F344
4.
Toxicol Lett ; 219(3): 203-10, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23535288

ABSTRACT

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.


Subject(s)
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
5.
Arch Toxicol ; 87(6): 1141-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23411599

ABSTRACT

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.


Subject(s)
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
6.
J Toxicol Sci ; 37(6): 1113-26, 2012.
Article in English | MEDLINE | ID: mdl-23208427

ABSTRACT

We previously demonstrated that 28-day administration of carcinogens that evoked cell proliferation as determined by immunoreactivity for Ki-67 or minichromosome maintenance 3 (Mcm3), in many target organs, increased the numbers of topoisomerase (Topo) IIα(+), ubiquitin D (Ubd)(+), and TUNEL(+) apoptotic cells. We also found increased co-expression of Topo IIα and Ubd, suggestive of increased spindle checkpoint disruption at the M phase. To investigate the roles of these markers in the early stages of carcinogenesis, we examined distribution changes in several carcinogenic target organs using rat initiation-promotion models. Promoting agents targeting the liver (piperonyl butoxide, methapyrilene hydrochloride), thyroid (sulfadimethoxine), urinary bladder (phenylethyl isothiocyanate), and forestomach and glandular stomach (catechol) were administered to rats after initiation treatment for each target organ. Numbers of Ki-67(+), Mcm3(+), Topo IIα(+) and TUNEL(+) cells increased within preneoplastic lesions as determined by glutathione S-transferase placental form in the liver or phospho-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. On the other hand, Ubd(+) cells did not increase within these preneoplastic lesions, but increased within hyperplastic lesions. These results suggest that both cell proliferation and apoptosis may be involved in the formation of preneoplastic lesions in the liver and thyroid examined here; however, spindle checkpoint disruption may not be involved in this process. Changes in hyperplastic lesions of the urinary bladder, forestomach and glandular stomach are similar to the 28-day carcinogen-treated cases, suggestive of the hyperplastic cellular character before the preneoplastic state.


Subject(s)
Apoptosis/drug effects , Carcinogens/toxicity , Cell Cycle/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Stomach Neoplasms/pathology , Thyroid Neoplasms/pathology , Urinary Bladder Neoplasms/pathology , Animals , Antigens, Neoplasm/metabolism , Carcinogens/administration & dosage , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Male , Rats , Rats, Inbred F344 , Time Factors , Ubiquitins/metabolism
7.
J Toxicol Sci ; 37(6): 1093-111, 2012.
Article in English | MEDLINE | ID: mdl-23208426

ABSTRACT

We have previously reported that renal carcinogens examined in rats increase tubular cell proliferation and topoisomerase (Topo) IIα(+) cells. The present study was aimed at identifying early prediction markers of carcinogens after 28-day treatment in rats. Following gene expression screening by microarrays in renal tubules with renal carcinogens, immunohistochemical analysis and TUNEL-assay were performed with carcinogens targeting different organs. All renal carcinogens tested (ferric nitrilotriacetic acid, ochratoxin A (OTA), monuron, tris(2-chloroethyl) phosphate, and potassium bromate) increased tubular cells immunoreactive for minichromosome maintenance 3 (Mcm3) or ubiquitin D (Ubd) or those showing apoptosis, compared with untreated controls or non-carcinogenic renal toxicants. Carcinogens targeting the liver (thioacetamide (TAA), fenbendazole, piperonyl butoxide (PBO) and methyleugenol), thyroid (sulfadimethoxine), urinary bladder (phenylethyl isothiocyanate), forestomach (butylated hydroxyanisole), glandular stomach (catechol), and colon (chenodeoxycholic acid and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine) were examined for induction of Mcm3, Ubd, Topo IIα, Ki-67 and apoptosis using non-carcinogenic toxicants as negative controls. All carcinogens increased Mcm3(+), Ubd(+), Topo IIα(+), Ki-67(+) or TUNEL(+) cells, except for hepatocarcinogen PBO and both colon carcinogens, which did not increase cell proliferation. Ubd(+) cells co-expressing Topo IIα was increased without changing phospho-Histone H3-co-expressing cell population as examined with OTA and TAA. Results revealed cooperative responses of Topo IIα, Ubd and apoptosis by carcinogens inducing high proliferation activity, irrespective of target organs, examined here after a 28-day administration. Aberrant expression of Ubd at G(2) phase and increased apoptosis reflecting aberrant cell cycle regulation may be the common feature of these carcinogens.


Subject(s)
Apoptosis/drug effects , Carcinogens/toxicity , Cell Proliferation/drug effects , G2 Phase/genetics , Ubiquitins/metabolism , Animals , Antigens, Neoplasm/metabolism , Carcinogens/administration & dosage , Carcinogens/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , G2 Phase/physiology , Male , Rats , Rats, Inbred F344 , Time Factors
8.
J Toxicol Sci ; 37(6): 1177-90, 2012.
Article in English | MEDLINE | ID: mdl-23208433

ABSTRACT

The present study was performed to elucidate toxicity profile of citrinin (CTN) after repeated oral doses for 90 days, especially on the kidneys and female reproductive organs using female BALB/c mice. We first performed a 70-day repeated oral dose toxicity study of CTN by setting the doses at 1.25 and 7.5 ppm in the drinking water (Experiment 1). As a result, CTN did not produce any toxicity in the kidneys, liver, and female genital organs/tracts, except for a slight increase of relative ovary weight. We, next, performed 90-day repeated oral dose toxicity study of CTN by increasing the dose levels at 15 and 30 ppm in the drinking water. The results suggested that CTN did not produce any toxicity in the kidneys, liver, and female genital organs/tracts, except for increase of both absolute and relative ovary weights accompanying increase of large follicles at ≥ 15 ppm. On the basis of these findings, the lowest-observable-adverse-effect level of CTN was 15 ppm (2.25 mg/kg body weight/day) in the drinking water for female BALB/c mice after 90-day oral treatment.


Subject(s)
Anti-Bacterial Agents/toxicity , Citrinin/toxicity , Ovary/drug effects , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Citrinin/administration & dosage , Female , Kidney/drug effects , Liver/drug effects , Mice , Mice, Inbred BALB C , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Time Factors
9.
J Toxicol Sci ; 37(5): 969-85, 2012.
Article in English | MEDLINE | ID: mdl-23038005

ABSTRACT

Omeprazole (OPZ) and ß-naphthoflavone (BNF) are cytochrome P450 (CYP)1A inducers and have liver tumor promoting effects. In this study, we investigated the co-promoting and co-initiating effects of OPZ and BNF in rats. In Experiment 1, male rats were subjected to partial hepatectomy (PH), and given oral doses of 138 or 276 mg/kg OPZ, 0.125% or 0.25% BNF or 138 mg/kg OPZ+0.125% BNF (n = 9~12) for 6 weeks after N-diethylnitrosamine (DEN) initiation. In Experiment 2, male rats were treated with oral doses of 138 or 276 mg/kg OPZ, 0.03% or 0.06% BNF or 138 mg/kg OPZ+0.03% BNF (n = 11~12) for 9 days starting 1 week before initiating treatment. As an initiating treatment, 2-Amino-3,4-dimethylimidazo[4,5-f]quinolone (MeIQx) was orally administered 12 hr after PH. The rats were fed a basal diet for 15 days, followed by a diet containing 0.015% 2-acetylaminofluorene for the next 10 days with a single oral dose of carbon tetrachloride. In Experiment 1, the number and area of glutathione S-transferase placental form-positive foci in the OPZ+BNF group were significantly higher than the average values of the High OPZ or the High BNF group. The expression of cyclooxygenase-2 (Cox-2) and COX-2 protein in the liver significantly increased in the OPZ+BNF group. In Experiment 2, liver initiation activity was not enhanced by the co-administration of OPZ+BNF. The results of our studies suggest that the co-administration of OPZ and BNF results in synergistic effects in the liver tumor promotion probably owing to increased COX-2 expression, but no modifying effect in the liver initiation activity of MeIQx in rats.


Subject(s)
Carcinogens/toxicity , Cytochrome P-450 CYP1A1/metabolism , Liver Neoplasms, Experimental/chemically induced , Omeprazole/toxicity , Proton Pump Inhibitors/toxicity , beta-Naphthoflavone/toxicity , Animals , Carbon Tetrachloride , Carcinogens/administration & dosage , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Diethylnitrosamine , Drug Synergism , Lipid Peroxidation/drug effects , Liver Neoplasms, Experimental/metabolism , Male , Oligonucleotide Array Sequence Analysis , Omeprazole/administration & dosage , Omeprazole/blood , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/blood , Quinoxalines , Rats , Rats, Inbred F344 , beta-Naphthoflavone/administration & dosage , beta-Naphthoflavone/blood
10.
J Toxicol Pathol ; 25(2): 149-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22907981

ABSTRACT

Activators of tissue proteolysis including Stachybotrys microspora triprenyl phenol (SMTP)-7 are a new class of agents that are expected to be effective for amelioration of chronic tissue destructive diseases. The present study was performed to examine whether SMTP-7 is effective for the amelioration or protection of early-stage IgA nephropathy (IgAN) induced by nivalenol (NIV) in female BALB/c mice. In Experiment 1, mice were administered NIV at 24 ppm in diet for 8 weeks, and during the NIV treatment, they were intraperitoneally injected with SMTP-7 (10 mg/kg) three times a week. In Experiment 2, mice were injected similarly with SMTP-7 during the last 4 weeks of a 16-week NIV treatment. Immunofluorescence analysis revealed an inhibitory effect of SMTP-7 on the glomerular deposition of IgA in Experiment 1; however, it was ineffective in Experiment 2. On the other hand, SMTP-7 did not affect the serum concentration of IgA in both experiments. These results suggest that SMTP-7 has a potential to decrease the progression of IgAN induced by NIV through inhibition of local accumulation of IgA in the glomerular mesangium, while it was ineffective for suppression of IgA production. On the other hand, SMTP-7 was found to be ineffective for already deposited IgA, suggesting that SMTP-7 may not be effective for ameliorating advanced IgAN.

11.
J Toxicol Sci ; 37(3): 491-501, 2012.
Article in English | MEDLINE | ID: mdl-22687989

ABSTRACT

Omeprazole (OPZ), a proton pump inhibitor, is a cytochrome P450 (CYP) 1A1/2 inducer. Some CYP1A inducers are known to have liver tumor promoting effects in rats and the ability to enhance oxidative stress. In this study, we performed a two-stage liver carcinogenesis bioassay in rats to examine the tumor promoting effect of OPZ (Experiment 1) and to clarify a possible mechanism of action (Experiment 2). In Experiment 1, male F344 rats were subjected to a two-third partial hepatectomy, and treated with 0, 138 or 276 mg/kg OPZ by oral gavage once a day for six weeks after an intraperitoneal injection of N-diethylnitrosamine (DEN). Liver weights significantly increased in the DEN+OPZ groups, and the number and area of glutathione S-transferase placental form (GST-P) positive foci significantly increased in the DEN+276 mg/kg OPZ group. In Experiment 2, the same experiment as Experiment 1 was performed, but the dosage of OPZ was 0 or 276 mg/kg. The number and area of GST-P positive foci as well as liver weights significantly increased in the DEN+276 mg/kg OPZ group. The number of proliferative cell nuclear antigen (PCNA)-positive cells also significantly increased in the same group. Real-time RT-PCR showed that the expression of AhR battery genes including Cyp1a1, Cyp1a2, Ugt1a6 and Nqo1, and Nrf2 battery genes including Gpx2, Yc2, Akr7a3, Aldh1a1 Me1 and Ggt1 were significantly upregulated in this group. However, the production of microsomal reactive oxygen species (ROS) and formation of thiobarbituric acid-reactive substances (TBARS) decreased, and 8-hydroxydeoxyguanosine (8-OHdG) content remained unchanged in this group. These results indicate that OPZ, CYP1A inducer, is a liver tumor promoter in rats, but oxidative stress is not involved in the liver tumor promoting effect of OPZ.


Subject(s)
Liver Neoplasms, Experimental/pathology , Liver/pathology , Omeprazole/toxicity , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Administration, Oral , Aldehyde Dehydrogenase 1 Family , Animals , Carcinogens/toxicity , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2 , Cytochromes/genetics , Cytochromes/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Deoxyguanosine/metabolism , Diethylnitrosamine/toxicity , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Transferase/metabolism , Hepatectomy/methods , Injections, Intraperitoneal/methods , Liver/drug effects , Liver/metabolism , Male , NAD(P)H Dehydrogenase (Quinone) , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Up-Regulation
12.
J Toxicol Sci ; 37(3): 517-26, 2012.
Article in English | MEDLINE | ID: mdl-22687991

ABSTRACT

To determine the threshold dose of ß-Naphthoflavone (BNF) that induces hepatocellular tumor promoting effects, reactive oxygen species (ROS) generation and thiobarbituric acid-reactive substance (TBARS) formation, and drug-metabolizing enzymes that protect against ROS generation, two-stage liver carcinogenesis model was used. Partial hepatectomized rats (n = 11 to 12) were fed diets containing 0, 0.03, 0.06, 0.125 or 0.25% BNF for 6 weeks after an intraperitoneal injection of N-diethylnitrosamine (DEN) to initiate hepatocarcinogenesis. Histopathologically, glutathione S-transferase placental form (GST-P)-positive foci significantly increased in rats given 0.25% BNF. No marked changes in ROS production and TBARS contents were observed between the BNF treated and DEN alone groups. Real-time RT-PCR showed that the expression of Cyp1a1, Cyp1a2, Cyp1b1 and Nqo1 significantly increased in the groups given 0.03% BNF or more, but Ugt1a6, Akr7a3 and Gstm1 significantly increased in the groups given 0.125% BNF or more. Gpx2 and Yc2 significantly increased in the groups given 0.06% BNF or more and 0.25% BNF, respectively. Inflammation-related genes such as Ccl2, Mmp12, Serpine1 and Cox-2 significantly increased in the 0.25% BNF group. In immunohistochemistry, the number of cyclooxygenase-2 (COX-2)-positive cells increased in rats given 0.25% BNF. These results suggest that 0.25% BNF is the threshold dose for liver tumor promotion, and the fact that inflammation-related genes and COX-2 protein increased in the 0.25% BNF group strongly suggests that inflammation is involved in the liver tumor promoting effect of BNF in rats.


Subject(s)
Liver Neoplasms, Experimental/pathology , Liver/drug effects , beta-Naphthoflavone/toxicity , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2 , Cytochrome P-450 CYP1B1 , Cytochromes/genetics , Cytochromes/metabolism , Diethylnitrosamine/toxicity , Dose-Response Relationship, Drug , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hepatectomy/methods , Immunohistochemistry , Lipid Peroxidation/drug effects , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Male , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Thiobarbituric Acid Reactive Substances/metabolism , Up-Regulation
13.
Arch Toxicol ; 86(9): 1453-64, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22411272

ABSTRACT

Some renal carcinogens can induce karyomegaly, which reflects aberrant cell division in the renal tubules, from the early stages of exposure. To clarify the cell cycle-related changes during the early stages of renal carcinogenesis, we performed immunohistochemical analysis of tubular cells in male F344 rats treated with carcinogenic doses of representative renal carcinogens for 28 days. For this purpose, the karyomegaly-inducing carcinogens ochratoxin A (OTA), ferric nitrilotriacetic acid, and monuron, and the non-karyomegaly-inducing carcinogens tris(2-chloroethyl) phosphate and potassium bromate were examined. For comparison, a karyomegaly-inducing non-carcinogen, p-nitrobenzoic acid, and a non-carcinogenic non-karyomegaly-inducing renal toxicant, acetaminophen, were also examined. The outer stripe of the outer medulla (OSOM) and the cortex + OSOM were subjected to morphometric analysis of immunoreactive proximal tubular cells. Renal carcinogens, irrespective of their karyomegaly-inducing potential, increased proximal tubular cell proliferation accompanied by an increase in topoisomerase IIα-immunoreactive cells, suggesting a reflection of cell proliferation. Karyomegaly-inducing carcinogens increased nuclear Cdc2-, γH2AX-, and phosphorylated Chk2-immunoreactive cells in both areas, the former two acting in response to DNA damage and the latter one suggestive of sustained G2. OTA, an OSOM-targeting carcinogen, could easily be distinguished from untreated controls and non-carcinogens by evaluation of molecules responding to DNA damage and G2/M transition in the OSOM. Thus, all renal carcinogens examined facilitated proximal tubular proliferation by repeated short-term treatment. Among these, karyomegaly-inducing carcinogens may cause DNA damage and G2 arrest in the target tubular cells.


Subject(s)
Carcinogens/toxicity , Cell Cycle Proteins/metabolism , Cell Cycle/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Histones/metabolism , Immunohistochemistry , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Cortex/pathology , Kidney Medulla/drug effects , Kidney Medulla/metabolism , Kidney Medulla/pathology , Kidney Neoplasms/chemically induced , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/pathology , Male , Phosphoproteins/metabolism , Phosphorylation/drug effects , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Random Allocation , Rats , Rats, Inbred F344
14.
Arch Toxicol ; 86(5): 779-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22252605

ABSTRACT

We have recently shown that maternal exposure to acrylamide (AA) impaired neurogenesis in rat offspring measured by the increase in interneurons producing reelin, a molecule regulating migration and correct positioning of developing neurons, in the hippocampal dentate gyrus. To clarify the cellular target of AA on hippocampal neurogenesis and its reversibility after maternal exposure, pregnant Sprague-Dawley rats were given drinking water containing AA at 0, 4, 20, 100 ppm on day 10 of pregnancy through day 21 after delivery on weaning. Male offspring were examined immunohistochemically on postnatal day (PND) 21 and PND 77. For comparison, male pups of direct AA-injection control during lactation (50 mg/kg body weight, intraperitoneally, 3 times/week) were also examined. On PND 21, maternal AA-exposure decreased progenitor cell proliferation in the subgranular zone (SGZ) from 20 ppm accompanied with increased density of reelin-producing interneurons and NeuN-expressing mature neurons within the hilus at 100 ppm, similar to the direct AA-injection control. In the SGZ examined at 100 ppm, cellular populations immunoexpressing doublecortin or dihydropyrimidinase-like 3, suggesting postmitotic immature granule cells, were decreased. On PND 77, the SGZ cell proliferation and reelin-producing interneuron density recovered, while the hilar mature neurons sustained to increase from 20 ppm, similar to the direct AA-injection control. Thus, developmental exposure to AA reversibly affects hippocampal neurogenesis targeting the proliferation of type-3 progenitor cells resulting in a decrease in immature granule cells in rats. A sustained increase in hilar mature neurons could be the signature of the developmental effect of AA.


Subject(s)
Acrylamide/toxicity , Dentate Gyrus/drug effects , Neurogenesis/drug effects , Stem Cells/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation/drug effects , Dentate Gyrus/growth & development , Doublecortin Domain Proteins , Doublecortin Protein , Drinking Water , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Male , Maternal Exposure , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Reelin Protein , Serine Endopeptidases/metabolism , Stem Cells/metabolism
15.
J Toxicol Sci ; 36(6): 775-86, 2011.
Article in English | MEDLINE | ID: mdl-22129741

ABSTRACT

Indole-3-carbinol (I3C) has a liver tumor promoting activity in rats, and is also known as a cytochrome p450 1A (CYP1A) inducer. The generation of reactive oxygen species (ROS) resulting from CYP1A induction due to I3C, is probably involved in the tumor promotion. To clarify whether ROS generation contributes to I3C's induction of hepatocellular altered foci, partially hepatectomized rats were fed a diet containing 0.5% of I3C for 8 weeks with or without 0.3% N-acetyl-L-cysteine (NAC), an antioxidant, in their drinking water after N-diethylnitrosamine (DEN) initiation. Immunohistochemical analysis showed that the glutathione-S-transferase placental form (GST-P) positive foci promoted by I3C were suppressed by the administration of NAC. The mRNAs of members of the phase II nuclear factor, erythroid derived 2, like 2 (Nrf2) gene batteries, whose promoter region is called as antioxidant response element (ARE), were down-regulated in the DEN-I3C-NAC group compared to the DEN-I3C group, but Cyp1a1 was not suppressed in the DEN-I3C-NAC group compared to the DEN-I3C group. There was no marked difference in production of microsomal ROS and genomic 8-hydroxy-2'-deoxygunosine (8-OHdG) as an oxidative DNA marker between the DEN-I3C-NAC and DEN-I3C groups, while mapkapk3 and Myc were decreased by the NAC treatment. These results indicate that oxidative stress plays an important role for I3C's tumor promotion, and NAC suppresses induction of hepatocellular altered foci with suppressed cytoplasmic oxidative stress.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Carcinogens/toxicity , Indoles/toxicity , Liver Neoplasms, Experimental/metabolism , Reactive Oxygen Species/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Alkylating Agents/toxicity , Animals , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Diethylnitrosamine/toxicity , Gene Expression Profiling , Hepatectomy , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Weight Gain/drug effects
16.
J Vet Diagn Invest ; 23(4): 852-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21908338

ABSTRACT

A perianal subcutaneous tumor involving the anal sac developed in an 8-year-old male mixed Labrador Retriever dog. Histologically, this tumor showed typical features of the solid-type carcinoma of the apocrine glands of the anal sac. However, neoplastic cells were immunoreactive for cytokeratin 8, chromogranin A, vasoactive intestinal peptide, neuron-specific enolase, and synaptophysin, and negative for S-100 protein, α-smooth muscle actin, vimentin, glucagon, insulin, somatostatin, carcinoembryonic antigen, serotonin, and parathyroid hormone-related protein. Considering the distribution of chromogranin A-positive cells within the anal sac apocrine glands, this tumor was diagnosed as neuroendocrine carcinoma originating from the apocrine glands of the anal sac.


Subject(s)
Anal Sacs/pathology , Apocrine Glands/pathology , Carcinoma, Neuroendocrine/veterinary , Dog Diseases/pathology , Soft Tissue Neoplasms/veterinary , Animals , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/pathology , Dog Diseases/diagnosis , Dogs , Male , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/pathology
17.
Toxicology ; 283(2-3): 109-17, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21396975

ABSTRACT

The liver tumor-promoting effects of indole-3-carbinol (I3C), a cytochrome P450 (CYP) 1A inducer found in cruciferous vegetables, were investigated using a medium-term hepatocarcinogenesis model in rats. Six-week-old male F344 rats received an intraperitoneal injection of N-diethylnitrosamine (DEN) and were fed a diet containing 0 (DEN-alone), 0.25, 0.50 or 1.0% of I3C for 8 weeks from 2 weeks after DEN-initiation. The number and area of liver cell foci positive for glutathione S-transferase placental form (GST-P) significantly increased in the livers of rats given 0.5% I3C or more, compared to those in the DEN-alone group. The number of GST-P positive foci also increased in the 0.25% I3C group. The number of liver cells positive for proliferating cell nuclear antigen (PCNA) significantly increased in all I3C groups compared to that in the DEN-alone group. Real-time RT-PCR analysis showed that I3C increased transcript levels of not only Cyp1a1 but also aryl hydrocarbon receptor (AhR) and/or nuclear factor (erythroid-derived 2)-like 2 (Nrf2) gene batteries, such as Cyp1a2, Cyp1b1, Ugt1a6, Nrf2, Nqo1, Gsta5, Gstm2, Ggt1and Gpx2. Reactive oxygen species (ROS) in the microsomal fraction significantly increased in all I3C-treated groups compared to the DEN-alone group, and thiobarbituric acid-reactive substances (TBARS) levels and 8-hydroxy-2'-deoxyguanosine (8-OHdG) content significantly increased in all of the I3C-treated groups and 1.0% I3C group, respectively. These results suggest that I3C is an AhR activator and enhances microsomal ROS production resulting in the upregulation of Nrf2 gene batteries, but the oxidative stress generated overcomes the antioxidant effect of Nrf2-related genes. Such 'a redox imbalance' subsequently induces liver tumor-promoting effects by enhancing cellular proliferation in rats.


Subject(s)
Carcinogens/pharmacology , Diethylnitrosamine/pharmacology , Free Radical Scavengers/pharmacology , Indoles/pharmacology , Liver Neoplasms, Experimental/pathology , Oxidative Stress/drug effects , Precancerous Conditions/chemically induced , 8-Hydroxy-2'-Deoxyguanosine , Animals , Body Weight/drug effects , DNA Damage/drug effects , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Enzyme Induction/drug effects , Hepatectomy , Immunohistochemistry , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver Neoplasms, Experimental/metabolism , Male , Microarray Analysis , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Organ Size/drug effects , Precancerous Conditions/pathology , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thiobarbituric Acid Reactive Substances/metabolism
18.
Arch Toxicol ; 85(9): 1159-66, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21203749

ABSTRACT

To investigate liver tumor-promoting potentials of indole-3-carbinol (I3C) and flutamide (FLU), changes in mRNA expression of Cyp1a and genes encoding antioxidant/detoxifying enzymes in the liver, 6-week-old male F344 rats were subjected to medium-term liver bioassay. ß-Naphthoflavone (BNF), a strong CYP1A inducer, was also used for comparison. Two weeks after initiation with N-diethylnitrosamine (DEN), animals were fed a basal diet (untreated controls) or a diet containing 0.5% I3C, 0.1% FLU, or 0.5% BNF for 6 weeks. Each animal was subjected to a two-third partial hepatectomy 1 week after the start of promoter treatments. Histopathologically, I3C and BNF increased altered liver cell foci with the incidence (3.7- and 7.3-fold) and multiplicity (8.3- and 13.8-fold) compared with the DEN-alone group, respectively. Immunohistochemically, I3C significantly increased the number (3.1-fold; P < 0.01) and area (2.4-fold; P < 0.05) of foci positive for glutathione-S-transferase placental form (GST-P) compared with the DEN-alone group; FLU induced a slight but significant increase in the number of GST-P-positive foci (2.8-fold; P < 0.05) whereas BNF showed marked induction of the number and area of GST-P-positive foci (20- and 14-fold, respectively; P < 0.01). In parallel, I3C, FLU, and BNF markedly increased mRNA levels of Cyp1a1 (50-, 23-, 299-fold) and antioxidant/detoxifying enzymes such as Gpx2 and Nqo1 as shown by real-time reverse transcription-polymerase chain reaction analysis. These results suggest that I3C and FLU could promote hepatocellular tumors in parallel with that of CYP1A's potential to cause subsequent oxidative stress responses in rats.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Flutamide/toxicity , Indoles/toxicity , Liver Neoplasms, Experimental/enzymology , Liver/drug effects , Animals , Body Weight/drug effects , Cytochrome P-450 CYP1A1/genetics , Diethylnitrosamine/toxicity , Enzyme Induction , Gene Expression/drug effects , Glutathione S-Transferase pi/metabolism , Hepatectomy , Immunohistochemistry , Liver/enzymology , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Organ Size/drug effects , Oxidative Stress/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , beta-Naphthoflavone/pharmacology
19.
Arch Toxicol ; 85(8): 987-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21120452

ABSTRACT

To examine whether developmental exposure to acrylamide (AA) impairs neuronal development, pregnant Sprague-Dawley rats were treated with AA at 0, 25, 50 or 100 ppm in drinking water from gestational day 6 until weaning on postnatal day 21. Offspring were immunohistochemically examined at the end of exposure. We investigated the expression of Reelin (a molecule regulating neuronal migration and positioning) in the hilus of the hippocampal dentate gyrus. As a positive control for direct exposure, AA (50 mg/kg body weight) was administered to pups by intraperitoneal injection 3 times per week during the lactation period. As well as pups directly injected with AA, maternally exposed offspring decreased body weight at 100 ppm; increased dose-dependently the number of Reelin-immunoreactive cells (from 25 ppm AA) and glutamic acid decarboxylase 67-immunoreactive cells (from 50 ppm AA), confirming an increase in γ-aminobutyric acid-ergic interneurons. We also noted decreased apoptosis in the neuroblast-producing subgranular zone of the dentate gyrus of maternally exposed pups at 100 ppm, as well as in directly AA-injected pups. These results suggest that a compensatory regulatory mechanism exists to correct impaired neurogenesis and mismigration caused by maternal exposure to AA during neuronal development. The lowest-observed-adverse-effect level of AA was determined to be 25 ppm (3.72 mg/kg body weight/day).


Subject(s)
Acrylamide/toxicity , Dentate Gyrus/drug effects , Maternal Exposure , Neurons/drug effects , Acrylamide/administration & dosage , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/pathology , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/metabolism , Female , Glutamate Decarboxylase/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/metabolism , No-Observed-Adverse-Effect Level , Pregnancy , Rats , Rats, Sprague-Dawley , Reelin Protein , Serine Endopeptidases/metabolism
20.
Reprod Toxicol ; 30(4): 591-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20920577

ABSTRACT

Reelin signaling plays a role in neuronal migration and positioning during brain development. To clarify the effect of systemic growth retardation on the distribution of Reelin-expressing interneurons in the hilus of the hippocampal dentate gyrus, pregnant rats were fed a synthetic diet with either a normal (20% casein) or low (10% casein) protein concentration from gestational day 10 to postnatal day (PND) 21 at weaning. Male offspring were immunohistochemically examined at PND 21 and on PND 77. Protein-restricted offspring displayed systemic growth retardation through PND 77 and had decreased absolute brain weights and an increased number of external granular cells in the cerebellar cortex, suggestive of retarded brain growth at weaning. However, maternal protein restriction did not change the cellular distribution of immunoreactivity for Reelin, Calbindin-D-28K, or glutamic acid decarboxylase 67 or of NeuN-positive postmitotic neurons in the dentate hilus either at PND 21 or PND 77, which suggests that the population of γ-aminobutyric acid-ergic interneurons involving synthesis of Reelin was not affected. Furthermore, as well as the distribution of hilar neurons expressing neurogenesis-related FoxG1, cell proliferation and apoptosis in the subgranular zone were unaffected through PND 77. These results suggest that systemic growth retardation caused by maternal protein restriction does not affect neuronal migration and postnatal neurogenesis of the dentate gyrus resulting in unaltered distribution of Reelin-synthesizing interneurons.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Dentate Gyrus/metabolism , Extracellular Matrix Proteins/metabolism , Fetal Growth Retardation/metabolism , Interneurons/metabolism , Malnutrition/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Aging , Animals , Brain/pathology , Cell Movement , Dentate Gyrus/pathology , Diet, Protein-Restricted/adverse effects , Female , Fetal Growth Retardation/pathology , Male , Malnutrition/pathology , Maternal Nutritional Physiological Phenomena , Neurogenesis , Neurons/metabolism , Neurons/pathology , Organ Size , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley , Reelin Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...