Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Environ Sci Pollut Res Int ; 29(1): 779-789, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34341928

RESUMEN

Nonalcoholic fatty liver disease is a hepatic disorder with deposition of fat droplets and has a high risk of progression to steatosis-related hepatitis and irreversible hepatic cancer. Metronidazole (MNZ) is an antiprotozoal and antimicrobial agent widely used to treat patients infected with anaerobic bacteria and intestinal parasites; however, MNZ has also been shown to induce liver tumors in rodents. To investigate the effects of MNZ on steatosis-related early-stage hepatocarcinogenesis, male rats treated with N-nitrosodiethylamine following 2/3 hepatectomy at week 3 were received a control basal diet, high fat diet (HFD), or HFD containing 0.5% MNZ. The HFD induced obesity and steatosis in the liver, accompanied by altered expression of Pparg and Fasn, genes related to lipid metabolism. MNZ increased nuclear translocation of lipid metabolism-related transcription factor peroxisome proliferator-activated receptor gamma in hepatocytes, together with altered liver expression of lipid metabolism genes (Srebf1, Srebf2, Pnpla2). Furthermore, MNZ significantly increased the number of preneoplastic liver foci, accompanied by DNA double-strand breaks and late-stage autophagy inhibition, as reflected by increased levels of γ-H2AX, LC3, and p62. Therefore, MNZ could induce steatosis-related hepatocarcinogenesis by inducing DNA double-strand breaks and modulating autophagy in HFD-fed rats.


Asunto(s)
Dieta Alta en Grasa , Enfermedad del Hígado Graso no Alcohólico , Animales , Autofagia , ADN/metabolismo , Dieta Alta en Grasa/efectos adversos , Humanos , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Metronidazol , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratas
2.
J Toxicol Sci ; 44(8): 565-574, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31378768

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive deposition of droplets in hepatocytes. Patients with NAFLD can be at risk for nonalcoholic steatohepatitis, which can lead to hepatocellular carcinoma. Autophagy is a cellular pathway that is crucial for survival and homeostasis, and which protects against pathophysiological changes like obesity and cancer. We determined the expression of autophagy markers in preneoplastic hepatic lesions and the effects of an autophagy repressor chloroquine (CQ) or inducer amiodarone (AM) in a steatosis-related hepatocarcinogenesis model. Male F344 rats were fed a control diet or high fat diet (HFD), and subjected to initiation and promotion steps with N-nitrosodiethylamine injection at week 0 and a partial hepatectomy at week 3. Several HFD-fed rats were administered 0.1% CQ and 0.5% AM in their drinking water during week 2 and 8. CQ and AM did not improve HFD-induced obesity. AM, but not CQ, significantly decreased the number of glutathione S-transferase placental form-positive preneoplastic liver foci in the liver. Autophagosome markers LC3 and the LC3-binding protein p62 were heterogeneously expressed in the preneoplastic foci. CQ might inhibit autophagy by significantly increased p62/LC3 ratio, while AM might have a potential of inducing autophagy by showing an increased gene expression of the autophagy regulator, Atg5. These results suggest that preneoplastic lesions express autophagosome markers and that AM might decrease steatosis-related early hepatocarcinogenesis by potentially inducing autophagy in HFD-fed rats, while inhibition of autophagy by CQ did not alter the hepatocarcinogenesis. However, an immunohistochemical trial revealed a technical limitation in detecting autophagosome markers because there were variations in each preneoplastic lesion.


Asunto(s)
Autofagosomas , Autofagia/genética , Dieta Alta en Grasa/efectos adversos , Expresión Génica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Amiodarona/farmacología , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Cloroquina/farmacología , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Masculino , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ratas Endogámicas F344
3.
J Vet Med Sci ; 81(8): 1229-1233, 2019 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-31270282

RESUMEN

A female koala presented with hyperglycemia related to diabetes mellitus diagnosed at 9 years and treated with insulin. She presented with nasal hemorrhage, anemia, leukocytosis, and tachypnea at 10 years. A blood smear examination revealed scattered, atypical large myeloid cells and a clinical diagnosis of myelogenous leukemia was made. White blood cell count reached a maximum of 295 × 102/µl, with evidence of severe regenerative anemia and thrombocytopenia. Grossly, systemic lymph node enlargement, fragile liver with hemorrhage, and bloody ascites were observed. Histopathologically, atypical myeloid cells, including myelocytic and metamyelocytic cells, were scattered in the vasculature and surrounding tissues throughout the organs. The patient was infected with a koala retrovirus, which might have caused the myelogenous leukemia.


Asunto(s)
Complicaciones de la Diabetes/veterinaria , Diabetes Mellitus/veterinaria , Leucemia Mieloide Aguda/veterinaria , Phascolarctidae , Infecciones por Retroviridae/veterinaria , Animales , Complicaciones de la Diabetes/diagnóstico , Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/virología , Femenino , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/virología , Phascolarctidae/virología , Infecciones por Retroviridae/complicaciones
4.
J Toxicol Sci ; 43(10): 611-621, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30298849

RESUMEN

Mineralocorticoid receptor (MR)/NADPH oxidase (NOX) signaling is involved in the development of obesity, insulin resistance, and renal diseases; however, the role of this signaling on steatotic preneoplastic liver lesions is not fully elucidated. We determined the effects of the MR antagonist potassium canrenoate (PC) on MR/NOX signaling in hepatic steatosis and preneoplastic glutathione S-transferase placental form (GST-P)-positive liver foci. Rats were subjected to a two-stage hepatocarcinogenesis model and fed with basal diet or high fat diet (HFD) that was co-administered with PC alone or in combination with the antioxidant alpha-glycosyl isoquercitrin (AGIQ). PC reduced obesity and renal changes (basophilic tubules that expressed MR and p22phox) but did not affect blood glucose tolerance and non-alcoholic fatty liver disease activity score (NAS) in HFD-fed rats. However, the drug increased the area of GST-P-positive liver foci that expressed MR and p22phox as well as increased expression of NOX genes (p22phox, Poldip2, and NOX4). PC in combination with AGIQ had the potential of inhibiting the effects of PC on the area of GST-P-positive liver foci and the effects were associated with increasing expression of an anti-oxidative enzyme (Catalase). The results suggested that MR/NOX signaling might be involved in development of preneoplastic liver foci and renal basophilic changes in HFD-fed rats; however, the impacts of PC were different in each organ.


Asunto(s)
Ácido Canrenoico/farmacología , Dieta Alta en Grasa/efectos adversos , Riñón/patología , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/patología , Hígado/patología , Antagonistas de Receptores de Mineralocorticoides/farmacología , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Ácido Canrenoico/administración & dosificación , Catalasa/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Gutatión-S-Transferasa pi/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , NADPH Oxidasas/fisiología , Especificidad de Órganos , Quercetina/administración & dosificación , Quercetina/análogos & derivados , Quercetina/farmacología , Receptores de Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Toxicol Pathol ; 46(5): 530-539, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29843569

RESUMEN

Administration of the diuretic, spironolactone (SR), can inhibit chronic liver diseases. We determined the effects of SR alone or in combination with the antioxidant α-glycosyl isoquercitrin (AGIQ) on hyperlipidemia- and steatosis-related precancerous lesions in high-fat diet (HFD)-fed rats subjected to a two-stage hepatocarcinogenesis model. Rats were fed with control basal diet or HFD, which was administered with SR alone or in combination with an antioxidant AGIQ in drinking water. An HFD increased body weight, intra-abdominal fat (adipose) tissue weight, and plasma lipids, which were reduced by coadministration of SR and AGIQ. SR and AGIQ coadministration also reduced hepatic steatosis and preneoplastic glutathione S-transferase placental form-positive foci, in association with decrease in NADPH oxidase (NOX) subunit p22phox-positive cells and an increase in active-caspase-3-positive cells in the foci. Hepatic gene expression analysis revealed that the coadministration of SR and AGIQ altered mRNA levels of lipogenic enzymes ( Scd1 and Fasn), antioxidant-related enzymes ( Catalase), NOX component ( P67phox), and anti-inflammatory transcriptional factor ( Pparg). Our results indicated that SR in combination with AGIQ had the potential of suppressing hyperlipidemia- and steatosis-related early hepatocarcinogenesis through the reduced expression of NOX subunits.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/prevención & control , NADPH Oxidasas/metabolismo , Lesiones Precancerosas/prevención & control , Quercetina/análogos & derivados , Espironolactona/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Quimioterapia Combinada , Hígado Graso/complicaciones , Hígado Graso/patología , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Lesiones Precancerosas/patología , Quercetina/administración & dosificación , Quercetina/uso terapéutico , Ratas Endogámicas F344 , Espironolactona/administración & dosificación
6.
Toxicol Appl Pharmacol ; 334: 207-216, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28899750

RESUMEN

We previously found downregulation of ubiquitin-conjugating enzyme E2E 2 (UBE2E2) in GST-P-positive (+) proliferative lesions produced by tumor promotion from early hepatocarcinogenesis stages in rats. Here we investigated the role of UBE2E2 downregulation in preneoplastic lesions of the liver and other target organs produced by tumor promotion in rats. Increased number of UBE2E2-related ubiquitination target proteins, phosphorylated c-MYC, KDM4A and KMT5A, was found in the UBE2E2-downregulated GST-P+ foci, compared with GST-P+ foci expressing UBE2E2. However, p21WAF1/CIP1, another UBE2E2 target protein, did not increase in the positive cells. Furthermore, the numbers of PCNA+ cells and γH2AX+ cells were increased in UBE2E2-downregulated foci. These results suggest sustained activation of c-MYC and stabilization of KMT5A to result in c-MYC-mediated transcript upregulation and following KMT5A-mediated protein stabilization of PCNA in GST-P+ foci, as well as KDM4A stabilization resulting in slowing down of DNA damage response in these lesions. Similar results were also observed in GST-P+ foci produced by repeated treatment of rats with a hepatocarcinogen, thioacetamide, for 90days. Hepatocarcinogen treatment for 28 or 90days also increased the numbers of liver cells expressing UBE2E2-related ubiquitination target proteins, as well as PCNA+ or γH2AX+ cells. Conversely, UBE2E2 downregulation was lacking in PPARα agonist-induced hepatocarcinogenesis, as well as in carcinogenic processes targeting other organs, suggestive of the loss of UBE2E2-related ubiquitination limited to hepatocarcinogenesis producing GST-P+ proliferative lesions. Our results suggest that repeated hepatocarcinogen treatment of rats causes stabilization of UBE2E2-related ubiquitination target proteins in liver cells to promote carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Gutatión-S-Transferasa pi/metabolismo , Hepatocitos/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Reparación del ADN , Regulación hacia Abajo , Regulación de la Expresión Génica , Gutatión-S-Transferasa pi/genética , Lesiones Precancerosas/inducido químicamente , Distribución Aleatoria , Ratas , Ubiquitina-Proteína Ligasas/genética
7.
Toxicol Sci ; 159(1): 211-223, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28903497

RESUMEN

We previously observed downregulation of mitochondrial oxidative phosphorylation-related protein, TMEM70, which is suggestive of disrupted cellular senescence, in GST-P-expressing (+) proliferative lesions from early hepatocarcinogenesis stages in rats. The present study investigated the immunohistochemical relationship between TMEM70 downregulation and cellular metabolic changes in carcinogenic processes, as well as the onset of the liver cell respiratory changes after repeated hepatocarcinogen treatment in rats. At the early hepatocarcinogenesis stage in a 2-stage model, GST-P+ preneoplastic lesions showing TMEM70 downregulation also downregulated the mitochondrial ATPase, ATPB, but upregulated glycolysis-related glucose transporter member 1 (GLUT1) and glucose-6-phosphate dehydrogenase, suggesting a metabolic shift from oxidative phosphorylation to glycolysis, known as the Warburg effect. Combined downregulation of TMEM70 and ATPB increased proliferation activity in GST-P+ preneoplastic lesions, suggesting cell proliferation facilitation by reducing mitochondrial respiration. Concurrent GLUT1-upregulation and TMEM70-downregulation increased nuclear phosphorylated c-MYC+ cells in GST-P+ preneoplastic lesions, suggesting c-MYC-mediated transcription facilitation to promote glycolysis and cell proliferation. The TMEM70-related metabolic shift was enhanced in GST-P+ neoplastic lesions, suggesting a contribution to tumor progression. Conversely, the TMEM70-related metabolic shift was lacking in peroxisome proliferator-activated receptor-α agonist-induced hepatocarcinogenesis, as well as in carcinogenic processes targeting other organs. Transcript expression analysis following 28- and 90-day repeated hepatocarcinogen treatment showed downregulation of Tmem70 from day 28 and upregulation of Pkm and Myc at day 90, suggesting early onset of a catastrophic cellular senescence-related metabolic shift beginning from depressed mitochondrial respiration in the liver. These results suggest a contribution of TMEM70 downregulation to the Warburg effect, which directs tumor promotion and progression in GST-P+-linked hepatocarcinogenesis in rats.


Asunto(s)
Carcinógenos/toxicidad , Regulación hacia Abajo , Gutatión-S-Transferasa pi/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Hígado/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Oxígeno/metabolismo , Animales , Apoptosis , Proliferación Celular , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/enzimología , Neoplasias Hepáticas Experimentales/patología , Fosforilación Oxidativa , Ratas
8.
Toxicology ; 389: 55-66, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28688903

RESUMEN

This study was performed to compare the exposure effects of N-methyl-N-nitrosourea (MNU), a cytocidal agent of proliferating cells, on rat hippocampal neurogenesis between developmental and postpubertal periods. Developmental exposure through maternal drinking water from gestational day 6 to day 21 after delivery on weaning decreased GFAP-immunoreactive (+) stem cells and increased immunoreactive cells indicative of subsequent progenitor and postmitotic immature neuronal populations, TUNEL+ or p21Cip1/Waf1+ stem/progenitor cells and COX2+ granule cells, on postnatal day (PND) 21. On PND 77 after cessation of developmental exposure, NeuN+ postmitotic granule cells decreased in number. Postpubertal exposure by oral gavage for 28days decreased the numbers of all granule cell lineage populations and ARC+ or COX2+ granule cells and increased the number of TUNEL+ stem/progenitor cells. These results suggested that both developmental and postpubertal exposure caused apoptosis of stem/progenitor cells. However, developmental exposure increased COX2 expression to facilitate intermediate progenitor cell proliferation and increased neuronal plasticity. This effect was concurrent with the induction of p21Cip1/Waf1 that causes cell cycle arrest of stem/progenitor cells in response to accumulating DNA damage on weaning, resulting in a subsequent reduction of postmitotic granule cells. In contrast, postpubertal exposure suppressed neuronal plasticity as evidenced by downregulation of ARC and COX2. The COX2 downregulation was responsible for the lack of facilitating stem/progenitor cell proliferation. Induction of apoptosis and the lack of cell proliferation facilitation may be responsible for the overall reduction of neurogenesis caused by postpubertal exposure. Thus, the disrupted pattern of hippocampal neurogenesis induced by MNU is different between developmental and postpubertal exposure.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Hipocampo/efectos de los fármacos , Exposición Materna/efectos adversos , Metilnitrosourea/toxicidad , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Factores de Edad , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Hipocampo/enzimología , Hipocampo/patología , Masculino , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Plasticidad Neuronal/efectos de los fármacos , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
9.
Toxicol Lett ; 266: 13-22, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27914986

RESUMEN

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.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Epigénesis Genética/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Proteínas de la Membrana/genética , Tioacetamida/toxicidad , Ubiquitina-Proteína Ligasas/genética , Adenoma de Células Hepáticas/inducido químicamente , Animales , Carcinoma Hepatocelular/inducido químicamente , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hígado/patología , Ratas
10.
Neurotox Res ; 31(1): 46-62, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27566479

RESUMEN

Valproic acid (VPA) is used to establish models of experimental autism. The present study investigated the developmental exposure effect of VPA on postnatal hippocampal neurogenesis in accordance with the exposure scheme of OECD Test Guideline 426 adopted for developmental neurotoxicity. Pregnant rats were administered drinking water containing 0, 667, or 2000 ppm VPA from gestational day 6 until day 21 post-delivery. In the subgranular zone (SGZ) and granule cell layer (GCL) of offspring, the number of granule cell lineage subpopulations remained unchanged upon weaning. However, in the hilus of the dentate gyrus, the number of reelin+ interneurons decreased at ≥667 ppm, and the number of PVALB+ or GAD67+ interneurons decreased at 2000 ppm. Conversely, Reln and Gad1 transcript levels increased at 2000 ppm, but Pvalb and Grin2d decreased, in the dentate gyrus. At the adult stage, PCNA+ proliferating SGZ cells, NeuN+ postmitotic SGZ/GCL neurons, and ARC+ or COX2+ GCL neurons increased at ≥667 ppm. In the dentate hilus, decreases in GAD67+ interneuron subpopulations and Grin2d transcript levels sustained at 2000 ppm. These results suggested that VPA primarily targets interneurons by developmental exposure, and this is followed by late effects on granule cell lineages, likely by influencing SGZ cell proliferation and synaptic plasticity. A reduced population of reelin+ or PVALB+ interneurons did not affect distribution of granule cell lineage subpopulations upon weaning. The late effect on neurogenesis, which resulted in increased GCL neurons, might be the result of a sustained decrease in GAD67+ interneurons expressing NR2D encoded by Grin2d.


Asunto(s)
Giro Dentado/efectos de los fármacos , Interneuronas/efectos de los fármacos , Exposición Materna , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Ácido Valproico/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Movimiento Celular/efectos de los fármacos , Corteza Cerebelosa/efectos de los fármacos , Corteza Cerebelosa/crecimiento & desarrollo , Corteza Cerebelosa/metabolismo , Corteza Cerebelosa/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Giro Dentado/crecimiento & desarrollo , Giro Dentado/metabolismo , Giro Dentado/patología , Modelos Animales de Enfermedad , Agua Potable , Femenino , Interneuronas/metabolismo , Interneuronas/patología , Masculino , Neurogénesis/fisiología , Embarazo , Distribución Aleatoria , Ratas , Proteína Reelina , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/fisiología
11.
Exp Toxicol Pathol ; 69(1): 9-16, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27789131

RESUMEN

We determined effects of the NADPH oxidase (NOX) inhibitor apocynin (APO) or the antioxidant enzymatically modified isoquercitrin (EMIQ) on an early stage of hepatocarcinogenesis in the liver with steatosis. Male rats were given a single intraperitoneal injection of N-diethylnitrosamine (DEN) and fed a high-fat diet (HFD) to subject to a two-stage hepatocarcinogenesis model. Two weeks later, rats were fed a HFD containing the lipogenic substance malachite green (MG), which were co-administered with EMIQ or APO in drinking water for 6 weeks. Three after DEN initiation, rats were subjected to a two-third partial hepatectomy to enhance cell proliferation. The HFD increased total cholesterol and alkaline phosphatase levels, which were reduced by EMIQ co-administration. APO co-administration reduced MG-increased preneoplastic liver lesions, glutathione S-transferase placental form (GST-P)-positive, adipophilin-negative liver foci, and tended to decrease MG-increased Ki-67-positive or active caspase-3-positive cells in the liver foci. EMIQ or APO co-administration reduced the expression of a NOX subunit p22phox in the liver foci, but did not alter the numbers of LC3a-positive cells, an autophagy marker. We identified no treatment-related effects on p47phox and NOX4 expression in the liver foci. The results indicated that APO or EMIQ had the potential to suppress hyperlipidaemia and steatosis-preneoplastic liver lesions, through suppression of NOX subunit expression in rats.


Asunto(s)
Acetofenonas/farmacología , Inhibidores Enzimáticos/farmacología , Hígado Graso/enzimología , NADPH Oxidasas/biosíntesis , Quercetina/análogos & derivados , Animales , Carcinogénesis/inducido químicamente , Modelos Animales de Enfermedad , Hígado Graso/patología , Inmunohistoquímica , Hígado/efectos de los fármacos , Hígado/enzimología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Quercetina/farmacología , Ratas , Ratas Endogámicas F344
12.
Toxicol Appl Pharmacol ; 310: 20-31, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27523638

RESUMEN

Both developmental and postpubertal cuprizone (CPZ) exposure impairs hippocampal neurogenesis in rats. We previously found that developmental CPZ exposure alters the expression of genes related to neurogenesis, myelination, and synaptic transmission in specific brain regions of offspring. Here, we examined neuronal and glial toxicity profiles in response to postpubertal CPZ exposure by using expression microarray analysis in the hippocampal dentate gyrus, corpus callosum, cerebral cortex, and cerebellar vermis of 5-week-old male rats exposed to 0, 120, and 600mg/kg CPZ for 28days. Genes showing transcript upregulation were subjected to immunohistochemical analysis. We found transcript expression alterations at 600mg/kg for genes related to synaptic transmission, Ache and Prima1, and cell cycle regulation, Tfap4 and Cdkn1a, in the dentate gyrus, which showed aberrant neurogenesis in the subgranular zone. This dose downregulated myelination-related genes in multiple brain regions, whereas KLOTHO+ oligodendrocyte density was decreased only in the corpus callosum. The corpus callosum showed an increase in transcript levels for inflammatory response-related genes and in the number of CD68+ microglia, MT+ astrocytes, and TUNEL+ apoptotic cells. These results suggest that postpubertal CPZ exposure targets synaptic transmission and cell cycle regulation to affect neurogenesis in the dentate gyrus. CPZ suppressed myelination in multiple brain regions and KLOTHO-mediated oligodendrocyte maturation only in the corpus callosum. The increased number of CD68+ microglia, MT+ astrocytes, and TUNEL+ apoptotic cells in the corpus callosum may be involved in the induction of KLOTHO+ oligodendrocyte death and be a protective mechanism against myelin damage following CPZ exposure.


Asunto(s)
Encéfalo/metabolismo , Cuprizona/farmacología , Perfilación de la Expresión Génica , Vaina de Mielina/efectos de los fármacos , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Masculino , Ratas , Ratas Sprague-Dawley
13.
Exp Toxicol Pathol ; 68(7): 399-408, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27402199

RESUMEN

The present study aimed to determine the onset time of hepatocarcinogen/hepatocarcinogenic tumor promoter-specific cell proliferation, apoptosis and aberrant cell cycle regulation after post-initiation treatment. Six-week-old rats were treated with the genotoxic hepatocarcinogen, carbadox (CRB), the marginally hepatocarcinogenic leucomalachite green (LMG), the tumor promoter, ß-naphthoflavone (BNF) or the non-carcinogenic hepatotoxicant, acetaminophen, for 2, 4 or 6 weeks during the post-initiation phase using a medium-term liver bioassay. Cell proliferation activity, expression of G2 to M phase- and spindle checkpoint-related molecules, and apoptosis were immunohistochemically analyzed at week 2 and 4, and tumor promotion activity was assessed at week 6. At week 2, hepatocarcinogen/tumor promoter-specific aberrant cell cycle regulation was not observed. At week 4, BNF and LMG increased cell proliferation together with hepatotoxicity, while CRB did not. Additionally, BNF and CRB reduced the number of cells expressing phosphorylated-histone H3 in both ubiquitin D (UBD)(+) cells and Ki-67(+) proliferating cells, suggesting development of spindle checkpoint dysfunction, regardless of cell proliferation activity. At week 6, examined hepatocarcinogens/tumor promoters increased preneoplastic hepatic foci expressing glutathione S-transferase placental form. These results suggest that some hepatocarcinogens/tumor promoters increase their toxicity after post-initiation treatment, causing regenerative cell proliferation. In contrast, some genotoxic hepatocarcinogens may disrupt the spindle checkpoint without facilitating cell proliferation at the early stage of tumor promotion. This suggests that facilitation of cell proliferation and disruption of spindle checkpoint function are induced by different mechanisms during hepatocarcinogenesis. Four weeks of post-initiation treatment may be sufficient to induce hepatocarcinogen/tumor promoter-specific cellular responses.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Hígado/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Acetaminofén/toxicidad , Animales , Carbadox/toxicidad , Cocarcinogénesis , Dietilnitrosamina/toxicidad , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratas Endogámicas F344 , Colorantes de Rosanilina/toxicidad , Factores de Tiempo , beta-naftoflavona/toxicidad
14.
Reprod Toxicol ; 65: 113-122, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27346841

RESUMEN

To elucidate the developmental exposure effects of ochratoxin A (OTA) on postnatal hippocampal neurogenesis, pregnant SD rats were provided a diet containing 0, 0.12, 0.6, or 3.0ppm OTA from gestation day 6 to day 21 on weaning after delivery. Offspring were maintained through postnatal day (PND) 77 without OTA exposure. At 3.0ppm, offspring of both sexes showed a transient body weight decrease after weaning. Changes in hippocampal neurogenesis-related parameters as measured in male PND 21 offspring were observed at 3.0ppm. In the subgranular zone (SGZ) of the dentate gyrus, PAX6+ or TBR2+ cells were decreased, while GFAP+ or DCX+ cells did not fluctuate in number, suggesting decreased numbers of type-2 progenitor cells. On the other hand, SGZ cells accumulating malondialdehyde increased. In the hilus of the dentate gyrus, SST+ or CHRNB2+ γ-aminobutyric acid (GABA)-ergic interneurons decreased, accompanied with transcript downregulation of Chrnb2 in the dentate gyrus. These results suggest that maternal exposure to OTA decreased type-2 progenitor cells by reducing hilar GABAergic interneurons innervating type-2 progenitor cells via cholinergic signal downregulation and also by increasing oxidative stress in the SGZ. Transcript levels of neurotrophin (Bdnf), glutamatergic receptors (Gria1, Gria2, and Grin2a), serotonin-synthesizing enzyme, and serotonergic receptors (Tph2, Htr1a, and Htr4) increased in the dentate gyrus, suggesting multiple neuroprotective actions against OTA-induced aberrant neurogenesis. All observed fluctuations were reversed by PND 77. The no-observed-adverse-effect level for offspring neurogenesis was determined to be 0.6ppm, corresponding to 39.3-76.0µg/kg body weight/day.


Asunto(s)
Hipocampo/efectos de los fármacos , Intercambio Materno-Fetal , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Ocratoxinas/toxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Proteína Doblecortina , Regulación hacia Abajo , Femenino , Hipocampo/citología , Masculino , Células-Madre Neurales/citología , Nivel sin Efectos Adversos Observados , Estrés Oxidativo , Embarazo , Ratas Sprague-Dawley
15.
J Toxicol Sci ; 41(1): 91-104, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26763396

RESUMEN

We previously reported that 28-day exposure to hepatocarcinogens that facilitate cell proliferation specifically alters the expression of G1/S checkpoint-related genes and proteins, induces aberrant early expression of ubiquitin D (UBD) at the G2 phase, and increases apoptosis in the rat liver, indicating G1/S and spindle checkpoint dysfunction. The present study aimed to determine the time of onset of carcinogen-specific cell-cycle disruption after repeated administration of renal carcinogens for up to 28 days. Rats were orally administered the renal carcinogens nitrofurantoin (NFT), 1-amino-2,4-dibromoantraquinone (ADAQ), and 1,2,3-trichloropropane (TCP) or the non-carcinogenic renal toxicants 1-chloro-2-propanol, triamterene, and carboxin for 3, 7 or 28 days. Both immunohistochemical single-molecule analysis and real-time RT-PCR analysis revealed that carcinogen-specific expression changes were not observed after 28 days of administration. However, the renal carcinogens ADAQ and TCP specifically reduced the number of cells expressing phosphorylated-histone H3 at Ser10 in both UBD(+) cells and proliferating cells, suggestive of insufficient UBD expression at the M phase and early transition of proliferating cells from the M phase, without increasing apoptosis, after 28 days of administration. In contrast, NFT, which has marginal carcinogenic potential, did not induce such cellular responses. These results suggest that it may take 28 days to induce spindle checkpoint dysfunction by renal carcinogens; however, induction of apoptosis may not be essential. Thus, induction of spindle checkpoint dysfunction may be dependent on carcinogenic potential of carcinogen examined, and marginal carcinogens may not exert sufficient responses even after 28 days of administration.


Asunto(s)
Antraquinonas/administración & dosificación , Antraquinonas/toxicidad , Riñón/citología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Nitrofurantoína/administración & dosificación , Nitrofurantoína/toxicidad , Propano/análogos & derivados , Animales , Peso Corporal/efectos de los fármacos , Carboxina/administración & dosificación , Carboxina/toxicidad , Proliferación Celular/efectos de los fármacos , Clorhidrinas/administración & dosificación , Clorhidrinas/toxicidad , Histonas/metabolismo , Riñón/efectos de los fármacos , Masculino , Tamaño de los Órganos/efectos de los fármacos , Propano/administración & dosificación , Propano/toxicidad , Ratas Endogámicas F344 , Factores de Tiempo , Triantereno/administración & dosificación , Triantereno/toxicidad , Ubiquitinas/metabolismo
16.
Toxicol Appl Pharmacol ; 290: 10-20, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26577399

RESUMEN

Developmental cuprizone (CPZ) exposure impairs rat hippocampal neurogenesis. Here, we captured the developmental neurotoxicity profile of CPZ using a region-specific expression microarray analysis in the hippocampal dentate gyrus, corpus callosum, cerebral cortex and cerebellar vermis of rat offspring exposed to 0, 0.1, or 0.4% CPZ in the maternal diet from gestation day 6 to postnatal day (PND) 21. Transcripts of those genes identified as altered were subjected to immunohistochemical analysis on PNDs 21 and 77. Our results showed that transcripts for myelinogenesis-related genes, including Cnp, were selectively downregulated in the cerebral cortex by CPZ at ≥0.1% or 0.4% on PND 21. CPZ at 0.4% decreased immunostaining intensity for 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase) and CNPase(+) and OLIG2(+) oligodendrocyte densities in the cerebral cortex, whereas CNPase immunostaining intensity alone was decreased in the corpus callosum. By contrast, a striking transcript upregulation for Klotho gene and an increased density of Klotho(+) oligodendrocytes were detected in the corpus callosum at ≥0.1%. In the dentate gyrus, CPZ at ≥0.1% or 0.4% decreased the transcript levels for Gria1, Grin2a and Ptgs2, genes related to the synapse and synaptic transmission, and the number of GRIA1(+) and GRIN2A(+) hilar γ-aminobutyric acid (GABA)-ergic interneurons and cyclooxygenase-2(+) granule cells. All changes were reversed at PND 77. Thus, developmental CPZ exposure reversibly decreased mature oligodendrocytes in both cortical and white matter tissues, and Klotho protected white matter oligodendrocyte growth. CPZ also reversibly targeted glutamatergic signals of GABAergic interneuron to affect dentate gyrus neurogenesis and synaptic plasticity in granule cells.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Cuprizona/toxicidad , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Glucuronidasa/genética , Glucuronidasa/metabolismo , Proteínas Klotho , Masculino , Oligodendroglía/citología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica , Sustancia Blanca/citología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/metabolismo , Ácido gamma-Aminobutírico/metabolismo
17.
Arch Toxicol ; 90(8): 2009-24, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26314264

RESUMEN

To determine the developmental exposure effects of T-2 toxin on postnatal hippocampal neurogenesis, pregnant ICR mice were provided a diet containing T-2 toxin at 0, 1, 3, or 9 ppm from gestation day 6 to day 21 on weaning after delivery. Offspring were maintained through postnatal day (PND) 77 without T-2 toxin exposure. In the hippocampal dentate gyrus of male PND 21 offspring, GFAP(+) and BLBP(+) type-1 stem cells and PAX6(+) and TBR2(+) type-2 progenitor cells decreased in the subgranular zone (SGZ) at 9 and ≥3 ppm, respectively, in parallel with increased apoptosis at ≥3 ppm. In the dentate hilus, reelin(+) γ-aminobutyric acid (GABA)-ergic interneurons increased at 9 ppm, suggesting reflection of neuronal mismigration. T-2 toxin decreased transcript levels of cholinergic and glutamate receptor subunits (Chrna4, Chrnb2 and Gria2) and glutamate transporter (Slc17a6) in the dentate gyrus, suggesting decreased cholinergic signals on hilar GABAergic interneurons innervating type-2 cells and decreased glutamatergic signals on type-1 and type-2 cells. T-2 toxin decreased SGZ cells expressing stem cell factor (SCF) and increased cells accumulating malondialdehydes. Neurogenesis-related changes disappeared on PND 77, suggesting that T-2 toxin reversibly affects neurogenesis by inducing apoptosis of type-1 and type-2 cells with different threshold levels. Decreased cholinergic and glutamatergic signals may decrease type-2 cells at ≥3 ppm. Additionally, decreased SCF/c-Kit interactions and increased oxidative stress may decrease type-1 and type-2 cells at 9 ppm. The no-observed-adverse-effect level for offspring neurogenesis was determined to be 1 ppm (0.14-0.49 mg/kg body weight/day).


Asunto(s)
Hipocampo/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Toxina T-2/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Femenino , Edad Gestacional , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones Endogámicos ICR , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Nivel sin Efectos Adversos Observados , Estrés Oxidativo/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Receptores Colinérgicos/metabolismo , Proteína Reelina , Factor de Células Madre/metabolismo
18.
J Appl Toxicol ; 36(2): 223-37, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26011634

RESUMEN

We have previously reported that a 28-day treatment of carcinogens evoking target cell proliferation activates G1 /S checkpoint function and apoptosis, as well as induction of aberrant ubiquitin D (Ubd) expression, suggesting disruptive spindle checkpoint function, in rats. The present study aimed to determine the onset time of rat liver cells to undergo carcinogen-specific cell cycle aberration and proliferation. Animals were treated orally with a hepatocarcinogenic dose of methyleugenol or thioacetamide for 3, 7 or 28 days. For comparison, some animals were subjected to partial hepatectomy or treated with noncarcinogenic hepatotoxicants (acetaminophen, α-naphthyl isothiocyanate or promethazine). Carcinogen-specific liver cell kinetics appeared at day 28 as evident by increases of cell proliferation, p21(Cip1+) cells, phosphorylated-Mdm2(+) cells and cleaved caspase 3(+) cells, and upregulation of DNA damage-related genes. Hepatocarcinogens also downregulated Rbl2 and upregulated Cdkn1a and Mdm2, and decreased Ubd(+) cells co-expressing phosphorylated-histone H3 (p-Histone H3) and p-Histone H3(+) cell ratio within the Ki-67(+) proliferating population. These results suggest that it takes 28 days to induce hepatocarcinogen-specific early withdrawal of proliferating cells from M phase due to disruptive spindle checkpoint function as evidenced by reduction of Ubd(+) cells staying at M phase. Disruption of G1 /S checkpoint function reflected by downregulation of Rbl2 as well as upregulation of Mdm2 suggestive of sequestration of retinoblastoma protein is responsible for the facilitation of carcinogen-induced cell proliferation at day 28. Accumulation of DNA damage probably in association with facilitation of p53 degradation by activation of Mdm2 may be a prerequisite for aberrant p21(Cip1) activation, which is responsible for apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinógenos/toxicidad , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hepatectomía/efectos adversos , Hígado/efectos de los fármacos , Hígado/crecimiento & desarrollo , Animales , Carcinogénesis/efectos de los fármacos , Eugenol/análogos & derivados , Eugenol/toxicidad , Masculino , Ratas , Ratas Endogámicas F344 , Tioacetamida/toxicidad
19.
J Appl Toxicol ; 36(2): 211-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25943520

RESUMEN

Hexachlorophene (HCP) has been shown to induce myelin vacuolation due to intramyelinic edema of the nerve fibers in animal neural tissue. We investigated the maternal exposure effect of HCP on hippocampal neurogenesis in the offspring of pregnant mice supplemented with 0 (control), 33 or 100 ppm HCP in diet from gestational day 6 to day 21 after delivery. On postnatal day (PND) 21, offspring as examined in males exhibited decreased granule cell lineage populations expressing paired box 6, sex-determining region Y-box 2 and eomesodermin in the hippocampal subgranular zone (SGZ) accompanied by myelin vacuolation involving white matter tracts of the hippocampal fimbria at ≥ 33 ppm. However, SGZ cellular populations expressing brain lipid binding protein and doublecortin were unchanged at any dose. Transcript expression of cholinergic receptor genes, Chrna4 and Chrnb2, and glutamate receptor genes, Grm1 and Grin2d, examined at 100 ppm, decreased in the dentate gyrus. HCP exposure did not alter the number of proliferating or apoptotic cells in the SGZ, or reelin- or calcium-binding protein-expressing γ-aminobutyric acid (GABA)ergic interneurons in the dentate hilus, on PND 21 and PND 77. All neurogenesis-related changes observed in HCP-exposed offspring on PND 21 disappeared on PND 77, suggesting that maternal HCP exposure at ≥ 33 ppm reversibly decreased type 2 intermediate-stage progenitor cells in the hippocampal neurogenesis. Myelin vacuolation might be responsible for changes in neurogenesis possibly by reducing nerve conduction velocity of cholinergic inputs from the septal-hippocampal pathway to granule cell lineages and/or GABAergic interneurons, and of glutamatergic inputs to granule cell lineages.


Asunto(s)
Hexaclorofeno/toxicidad , Hipocampo/efectos de los fármacos , Exposición Materna/efectos adversos , Vaina de Mielina/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Células Madre/efectos de los fármacos , Animales , Colinérgicos/metabolismo , Fármacos actuantes sobre Aminoácidos Excitadores/metabolismo , Femenino , Masculino , Ratones , Embarazo , Proteína Reelina
20.
J Toxicol Sci ; 40(6): 855-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26558467

RESUMEN

We aimed to clarify the hepatocarcinogen-specific disruption of cell cycle checkpoint functions and its time course after repeated administration of hepatocarcinogens. Thus, rats were repeatedly administered with hepatocarcinogens (methapyrilene, carbadox and thioacetamide), a marginal hepatocarcinogen (leucomalachite green), hepatocarcinogenic promoters (oxfendazole and ß-naphthoflavone) or non-carcinogenic hepatotoxicants (promethazine and acetaminophen) for 7, 28 or 90 days, and the temporal changes in cell proliferation, expression of G1/S and spindle checkpoint-related molecules, and apoptosis were examined using immunohistochemistry and/or real-time RT-PCR analysis. Hepatocarcinogens facilitating cell proliferation at day 28 of administration also facilitated cell proliferation and apoptosis at day 90. Hepatocarcinogen- or hepatocarcinogenic promoter-specific cellular responses were not detected by immunohistochemical single molecule analysis even after 90 days. Expression of Cdkn1a, Mad2l1, Chek1 and Rbl2 mRNA also lacked specificity to hepatocarcinogens or hepatocarcinogenic promoters. In contrast, all hepatocarcinogens and the marginally hepatocarcinogenic leucomalachite green induced Mdm2 upregulation or increase in the number of phosphorylated MDM2(+) cells from day 28, irrespective of the lack of cell proliferation facilitation by some compounds. However, different Tp53 expression levels suggest different mechanisms of induction or activation of MDM2 among hepatocarcinogens. On the other hand, hepatocarcinogenic methapyrilene and carbadox downregulated the number of both ubiquitin D(+) cells and proliferating cells remaining in M phase at day 28 and/or day 90, irrespective of the lack of cell proliferation facilitation in the latter. These results suggest that hepatocarcinogens disrupt spindle checkpoint function after 28 or 90 days of administration, which may be induced ahead of cell proliferation facilitation.


Asunto(s)
Carbadox/toxicidad , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Hígado/citología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Metapirileno/toxicidad , Tioacetamida/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencimidazoles/administración & dosificación , Bencimidazoles/toxicidad , Carbadox/administración & dosificación , Proliferación Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Expresión Génica/efectos de los fármacos , Masculino , Metapirileno/administración & dosificación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Ratas Endogámicas F344 , Colorantes de Rosanilina/toxicidad , Factores de Tiempo , Ubiquitinas/genética , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA