Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Arch Toxicol ; 87(6): 1141-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23411599

RESUMO

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.


Assuntos
Carcinógenos/toxicidade , Proteínas de Ciclo Celular/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Homólogo 5 da Proteína Cromobox , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator 6 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho do Órgão/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
2.
Arch Toxicol ; 86(9): 1453-64, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22411272

RESUMO

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.


Assuntos
Carcinógenos/toxicidade , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Histonas/metabolismo , Imuno-Histoquímica , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Córtex Renal/patologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Medula Renal/patologia , Neoplasias Renais/induzido quimicamente , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Túbulos Renais Proximais/patologia , Masculino , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344
3.
Toxicol Lett ; 266: 13-22, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27914986

RESUMO

The present study identified genes showing promoter region hypermethylation by CpG island microarrays in the liver of rats treated with hepatocarcinogen thioacetamide (TAA) for 28days. Among 47 hypermethylated genes, Hist1h2aa, Tmem70, Ube2e2, and Slk were confirmed to show hypermethylation by methylation-specific quantitative polymerase chain reaction (PCR) and pyrosequencing analyses as well as downregulation of transcript levels by real-time reverse transcription-PCR analysis in the livers of rats treated with TAA. All gene products of the 4 selected genes showed decreased immunoreactivity forming negative liver cell foci in a subpopulation of glutathione S-transferase placental form (GST-P)+ foci in TAA-promoted rat livers in a two-stage hepatocarcinogenesis model. Among them, TMEM70 and UBE2E2 showed increased incidences of negative foci in GST-P+ foci by promotion of all examined TAA, ß-naphthoflavone, piperonyl butoxide, fenbendazole and phenobarbital, while HIST1H2AA and SLK did not respond to all promotive treatments. In the late stage of tumor promotion by TAA, the incidence of GST-P+ proliferative lesions with downregulation of TMEM70 or UBE2E2 was higher in adenomas and carcinomas than liver cell foci. TMEM70 plays a role in mitochondrial oxidative phosphorylation, and UBE2E2 participates in the stabilization of cell cycle regulatory proteins. Therefore, our results indicate that aberrant epigenetic gene downregulation suggestive of a metabolic shift of cellular respiration from oxidative phosphorylation to glycolysis and aberrant cell cycle regulation facilitating cell proliferation from as early as 28days after hepatocarcinogen treatment contribute to tumor development.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/patologia , Epigênese Genética/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Proteínas de Membrana/genética , Tioacetamida/toxicidade , Ubiquitina-Proteína Ligases/genética , Adenoma de Células Hepáticas/induzido quimicamente , Animais , Carcinoma Hepatocelular/induzido quimicamente , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fígado/patologia , Ratos
4.
Exp Toxicol Pathol ; 66(1): 1-11, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23890812

RESUMO

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.


Assuntos
Carcinogênese/metabolismo , Divisão Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Lesões Pré-Cancerosas/metabolismo , Animais , Carcinógenos/toxicidade , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Masculino , Neoplasias/induzido quimicamente , Lesões Pré-Cancerosas/induzido quimicamente , Ratos , Ratos Endogâmicos F344 , Neoplasias da Glândula Tireoide/induzido quimicamente , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Bexiga Urinária/induzido quimicamente , Neoplasias da Bexiga Urinária/metabolismo
5.
Toxicol Lett ; 224(1): 64-72, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24120684

RESUMO

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.


Assuntos
Núcleo Celular/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Ocratoxinas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Glutationa/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Masculino , Ratos , Ratos Endogâmicos F344
6.
Exp Toxicol Pathol ; 65(7-8): 979-88, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23474136

RESUMO

Thioacetamide (TAA) induces oxidative stress and hepatocarcinogenicity in rats. We previously reported that TAA promotion caused various disruptions in cell cycle protein expression in rats, including downregulation of p16(Ink4a), which is associated with intraexonic hypermethylation in hepatocellular proliferative lesions. This study further investigated the contribution of cell cycle aberrations associated with early hepatocarcinogenic processes induced by TAA using antioxidants, enzymatically modified isoquercitrin (EMIQ) and α-lipoic acid (ALA), in a two-stage rat hepatocarcinogenesis model. TAA-promotion after initiation with N-diethylnitrosamine increased the number and area of hepatocellular foci immunoreactive for glutathione S-transferase placental form (GST-P) and the numbers of proliferating and apoptotic cells. Co-treatment with EMIQ and ALA suppressed these increases. TAA-induced formation of p16(Ink4a-) foci in concordance with GST-P(+) foci was not suppressed by co-treatment with EMIQ or ALA. TAA-promotion increased cellular distributions of cell proliferation marker Ki-67, G2/M and spindle checkpoint proteins (phosphorylated checkpoint kinase 1 and Mad2), the DNA damage-related protein phosphorylated histone H2AX, and G2-M phase-related proteins (topoisomerase IIα, phosphorylated histone H3 and Cdc2) within GST-P(+) foci, and co-treatment with EMIQ or ALA suppressed these increases. These results suggest that downregulation of p16(Ink4a) may allow selective proliferation of preneoplastic cells by TAA promotion. However, antioxidants did not counteract this gene control. Moreover, effective suppression of TAA-induced cellular population changes within preneoplastic lesions by antioxidants may reflect facilitation of cell cycling and accumulation of DNA damage causing the activation of cell cycle checkpoints, leading to G2 and M phase arrest at the early stages of hepatocarcinogenesis promoted by TAA.


Assuntos
Ciclo Celular/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Lesões Pré-Cancerosas/patologia , Animais , Carcinogênese/induzido quimicamente , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Estresse Oxidativo/efeitos dos fármacos , Lesões Pré-Cancerosas/induzido quimicamente , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tioacetamida/toxicidade
7.
J Cancer Res Clin Oncol ; 139(3): 395-401, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23114882

RESUMO

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.


Assuntos
Carcinoma/patologia , Modelos Animais de Doenças , Hipotireoidismo/complicações , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Animais , Carcinógenos , Carcinoma/induzido quimicamente , Carcinoma/etiologia , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/patologia , Progressão da Doença , Hiperplasia/etiologia , Hiperplasia/patologia , Hipotireoidismo/patologia , Masculino , Invasividade Neoplásica , Nitrosaminas , Ratos , Ratos Endogâmicos F344 , Neoplasias da Glândula Tireoide/induzido quimicamente , Neoplasias da Glândula Tireoide/etiologia
8.
Toxicol Lett ; 219(3): 203-10, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23535288

RESUMO

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.


Assuntos
Carcinógenos/farmacologia , Divisão Celular/efeitos dos fármacos , Animais , Aurora Quinase B , Aurora Quinases , Biomarcadores/análise , Hidroxianisol Butilado/farmacologia , Proteína Quinase CDC2 , Catecóis/farmacologia , Proteínas de Ciclo Celular/análise , Divisão Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/análise , Quinases Ciclina-Dependentes , Histonas/análise , Imidazóis/farmacologia , Isotiocianatos/farmacologia , Masculino , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/química , Proteínas Serina-Treonina Quinases/análise , Piridinas/farmacologia , Ratos , Ratos Endogâmicos F344 , Sulfadimetoxina/farmacologia
9.
Toxicol Lett ; 222(3): 295-302, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23968726

RESUMO

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.


Assuntos
Carcinógenos/toxicidade , Metilação de DNA/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Butóxido de Piperonila/toxicidade , Animais , Testes de Carcinogenicidade , Ilhas de CpG/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Reprod Toxicol ; 35: 125-36, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23099338

RESUMO

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.


Assuntos
Clorpirifos/toxicidade , Inibidores da Colinesterase/toxicidade , Giro Denteado/citologia , Inseticidas/toxicidade , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Moléculas de Adesão Celular Neuronais/metabolismo , Proliferação de Células/efeitos dos fármacos , Colinesterases/sangue , Proteínas da Matriz Extracelular/metabolismo , Feminino , Força da Mão , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Troca Materno-Fetal , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/efeitos dos fármacos , Nível de Efeito Adverso não Observado , Gravidez , Ratos , Proteína Reelina , Serina Endopeptidases/metabolismo , Células-Tronco/citologia , Hormônios Tireóideos/sangue
11.
J Toxicol Sci ; 38(5): 679-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24025784

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Citocromo P-450 CYP1A1/biossíntese , Neoplasias Hepáticas/prevenção & controle , Fenobarbital/administração & dosagem , Butóxido de Piperonila/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Animais , Carcinoma Hepatocelular/induzido quimicamente , Proliferação de Células/efeitos dos fármacos , Depressão Química , Dietilnitrosamina , Modelos Animais de Doenças , Quimioterapia Combinada , Indução Enzimática/efeitos dos fármacos , Glutationa S-Transferase pi/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/etiologia , Masculino , Microssomos Hepáticos/metabolismo , Fenobarbital/farmacologia , Butóxido de Piperonila/farmacologia , Ratos , Ratos Endogâmicos F344
12.
J Toxicol Sci ; 38(3): 403-13, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23665939

RESUMO

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.


Assuntos
Antagonistas Colinérgicos/toxicidade , Neoplasias Hepáticas/induzido quimicamente , Antagonistas Muscarínicos/toxicidade , Orfenadrina/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Núcleo Celular/metabolismo , Antagonistas Colinérgicos/administração & dosagem , Receptor Constitutivo de Androstano , Dietilnitrosamina/administração & dosagem , Dietilnitrosamina/toxicidade , Hepatócitos/citologia , Hepatócitos/metabolismo , Masculino , Antagonistas Muscarínicos/administração & dosagem , Orfenadrina/administração & dosagem , Ratos , Ratos Endogâmicos F344
13.
J Toxicol Sci ; 38(3): 415-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23665940

RESUMO

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.


Assuntos
Neoplasias Hepáticas/induzido quimicamente , Orfenadrina/toxicidade , Fenobarbital/toxicidade , Animais , Citocromo P-450 CYP2B1/metabolismo , Dietilnitrosamina/administração & dosagem , Dietilnitrosamina/toxicidade , Combinação de Medicamentos , Sinergismo Farmacológico , Indução Enzimática/efeitos dos fármacos , Masculino , Microssomos Hepáticos/metabolismo , Orfenadrina/administração & dosagem , Estresse Oxidativo/genética , Fenobarbital/administração & dosagem , Ratos , Ratos Endogâmicos F344 , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
14.
J Toxicol Sci ; 37(3): 517-26, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22687991

RESUMO

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.


Assuntos
Neoplasias Hepáticas Experimentais/patologia , Fígado/efeitos dos fármacos , beta-Naftoflavona/toxicidade , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2 , Citocromo P-450 CYP1B1 , Citocromos/genética , Citocromos/metabolismo , Dietilnitrosamina/toxicidade , Relação Dose-Resposta a Droga , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Hepatectomia/métodos , Imuno-Histoquímica , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ratos , Ratos Endogâmicos F344 , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Regulação para Cima
15.
J Toxicol Sci ; 37(6): 1113-26, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23208427

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Hepáticas/patologia , Neoplasias Gástricas/patologia , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Bexiga Urinária/patologia , Animais , Antígenos de Neoplasias/metabolismo , Carcinógenos/administração & dosagem , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Masculino , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo , Ubiquitinas/metabolismo
16.
J Toxicol Sci ; 37(6): 1093-111, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23208426

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , Fase G2/genética , Ubiquitinas/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Carcinógenos/administração & dosagem , Carcinógenos/farmacologia , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G2/fisiologia , Masculino , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo
17.
J Toxicol Sci ; 37(6): 1177-90, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23208433

RESUMO

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.


Assuntos
Antibacterianos/toxicidade , Citrinina/toxicidade , Ovário/efeitos dos fármacos , Administração Oral , Animais , Antibacterianos/administração & dosagem , Citrinina/administração & dosagem , Feminino , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Nível de Efeito Adverso não Observado , Tamanho do Órgão/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/patologia , Fatores de Tempo
18.
Toxicology ; 299(2-3): 146-54, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-22659510

RESUMO

Cell cycle aberration was immunohistochemically examined in relation to preneoplastic liver cell foci expressing glutathione S-transferase placental form (GST-P) at early stages of tumor-promotion in rats with thioacetamide (TAA), a hepatocarcinogen facilitating liver cell regeneration. Immunoexpression of p16(Ink4a) following exposure to other hepatocarcinogens/promoters and its DNA methylation status were also analyzed during early and late tumor-promotion stages. GST-P(+) liver cell foci increased cell proliferation and decreased apoptosis when compared with surrounding liver cells. In concordance with GST-P(+) foci, checkpoint proteins at G(1)/S (p21(Cip1), p27(Kip1) and p16(Ink4a)) and G(2)/M (phospho-checkpoint kinase 1, Cdc25c and phospho-Wee1) were either up- or downregulated. Cellular distribution within GST-P(+) foci was either increased or decreased with proteins related to G(2)-M phase or DNA damage (topoisomerase IIα, phospho-histone H2AX, phospho-histone H3 and Cdc2). In particular, p16(Ink4a) typically downregulated in GST-P(+) foci and regenerative nodules at early tumor-promotion stage with hepatocarcinogens facilitating liver cell regeneration and in neoplastic lesions at late tumor-promotion stage with hepatocarcinogens/promoters irrespective of regenerating potential. Hypermethylation at exon 2 of Cdkn2a was detected at both early- and late-stages. Thus, diverse disruptive expression of G(1)/S and G(2)/M proteins, which allows for clonal selection of GST-P(+) foci, results in the acquisition of multiple aberrant phenotypes to disrupt checkpoint function. Moreover, increased DNA-damage responses within GST-P(+) foci may be the signature of genetic alterations. Intraexonic hypermethylation may be responsible for p16(Ink4a)-downregulation, which facilitates cell cycle progression in early preneoplastic lesions produced by repeated cell regeneration and late-stage neoplastic lesions irrespective of the carcinogenic mechanism.


Assuntos
Ciclo Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Glutationa S-Transferase pi/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Regeneração Hepática/fisiologia , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação para Baixo , Epigênese Genética , Perfilação da Expressão Gênica , Imuno-Histoquímica , Masculino , Estadiamento de Neoplasias , RNA/química , RNA/genética , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Tioacetamida/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA