Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Arch Toxicol ; 84(3): 233-43, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20069279

RESUMEN

Short-chain chlorinated paraffins (SCCPs) cause kidney tumours in male rats, but not in female rats or mice of either sex. Male rat-specific tumours also occur in rats dosed with a range of compounds including 1,4- dichlorobenzene (DCB) and d-limonene (DL). These compounds bind to a male rat-specific hepatic protein, alpha-2-urinary globulin (α2u), and form degradationresistant complexes in the kidney. The resulting accumulation of α2u causes cell death and sustained regenerative cell proliferation, which in turn leads to the formation of renal tumours. To investigate whether the SCCP, Chlorowax 500C (C500C), causes tumours via the accumulation of α2u male rats were orally dosed with either C500C (625 mg/kg of body weight), DCB (300 mg/kg of body weight), or DL (150 mg/kg of body weight) for 28 consecutive days. An increase in renal α2u and cell proliferation was observed in DCB- and DL-treated rats but not in C500C-treated rats. C500C caused peroxisome proliferation and a down-regulation of α2u synthesis in male rat liver. This down-regulation occurred at the transcriptional level. Since less α2u was produced in C500C-treated rats, there was less available for accumulation in the kidney hence a typical α2u nephropathy did not appear. However, the administration of a radiolabelled SCCP, [14C]polychlorotridecane (PCTD), to male rats demonstrated its binding to renal α2u. Thus, it is possible that SCCPs bind to α2u and cause a slow accumulation of the protein in the kidney followed by delayed onset of α2u nephropathy. As a consequence of these findings in the current experiments, while evidence exists implicating α2u-globulin in the molecular mechanism of action of the C500C, the classic profile of a α2u-globulin nephropathy seen with other chemicals such as DCB and DL was not reproduced during this experimental protocol.


Asunto(s)
alfa-Globulinas/metabolismo , Carcinógenos/metabolismo , Hidrocarburos Clorados/metabolismo , Neoplasias Renales/metabolismo , alfa-Globulinas/genética , Animales , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Clorobencenos/metabolismo , Clorobencenos/toxicidad , Ciclohexenos/metabolismo , Ciclohexenos/toxicidad , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Hidrocarburos Clorados/toxicidad , Riñón/efectos de los fármacos , Riñón/metabolismo , Neoplasias Renales/inducido químicamente , Neoplasias Renales/patología , Limoneno , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Peroxisomas/efectos de los fármacos , Unión Proteica , Ratas , Ratas Endogámicas F344 , Terpenos/metabolismo , Terpenos/toxicidad , Transcripción Genética/efectos de los fármacos
2.
Arch Toxicol ; 84(10): 787-98, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20614104

RESUMEN

Ammonium perfluorooctanoate (APFO), a processing aid used in the production of fluoropolymers, produces hepatomegaly and hepatocellular hypertrophy in rodents. In mice, APFO-induced hepatomegaly is associated with increased activation of the xenosensor nuclear receptors, PPARα and CAR/PXR. Although non-genotoxic, chronic dietary treatment of Sprague-Dawley (S-D) rats with APFO produced an increase in benign tumours of the liver, acinar pancreas, and testicular Leydig cells. Most of the criteria for establishing a PPARα-mediated mode of action for the observed hepatocellular tumours have been previously established with the exception of the demonstration of increased hepatocellular proliferation. The present study evaluates the potential roles for APFO-induced activation of PPARα and CAR/PXR with respect to liver tumour production in the S-D rat and when compared to the specific PPARα agonist, 4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid (Wy 14,643). Male S-D rats were fed APFO (300 ppm in diet) or Wy 14,643 (50 ppm in diet) for either 1, 7, or 28 days. Effects of treatment with APFO included: decreased body weight; hepatomegaly, hepatocellular hypertrophy, hepatocellular hyperplasia (microscopically and by BrdU labelling index), and hepatocellular glycogen loss; increased activation of PPARα (peroxisomal ß-oxidation and microsomal CYP4A1 protein); decreased plasma triglycerides, cholesterol, and glucose; increased activation of CAR (CYP2B1/2 protein) and CAR/PXR (CYP3A1 protein). Responses to treatment with Wy 14,643 were consistent with increased activation of PPARα, specifically: increased CYP4A1 and peroxisomal ß-oxidation; increased hepatocellular hypertrophy and cell proliferation; decreased apoptosis; and hypolipidaemia. With the exception of decreased apoptosis, the effects observed with Wy 14,643 were noted with APFO, and APFO was less potent. These data clearly demonstrate an early hepatocellular proliferative response to APFO treatment and suggest that the hepatomegaly and tumours observed after chronic dietary exposure of S-D rats to APFO likely are due to a proliferative response to combined activation of PPARα and CAR/PXR. This mode of action is unlikely to pose a human hepatocarcinogenic hazard.


Asunto(s)
Caprilatos/toxicidad , Proliferación Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Hepatocitos/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/metabolismo , ADN/metabolismo , Hepatocitos/patología , Hepatomegalia/inducido químicamente , Hipertrofia , Hígado/enzimología , Hígado/metabolismo , Hígado/patología , Masculino , Tamaño de los Órganos/efectos de los fármacos , PPAR alfa/metabolismo , Peroxisomas/metabolismo , Receptor X de Pregnano , Ratas , Ratas Sprague-Dawley , Receptores de Esteroides/metabolismo
3.
Toxicol Sci ; 176(2): 329-342, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32458970

RESUMEN

Basimglurant (RG7090), a small molecule under development to treat certain forms of depression, demonstrated foci of altered hepatocytes in a long-term rodent-toxicity study. Additional evidence pointed toward the activation of the constitutive androstane receptor (CAR), an established promoter of nongenotoxic and rodent-specific hepatic tumors. This mode of action and the potential human relevance was explored in vivo using rodent and cynomolgus monkey models and in vitro using murine and human liver spheroids. Wild type (WT) and CAR/pregnane X receptor (PXR) knockout mice (CAR/PXR KO) were exposed to RG7090 for 8 consecutive days. Analysis of liver lysates revealed induction of Cyp2b mRNA and enzyme activity, a known activation marker of CAR, in WT but not in CAR/PXR KO animals. A series of proliferative genes were upregulated in WT mice only, and immunohistochemistry data showed increased cell proliferation exclusively in WT mice. In addition, primary mouse liver spheroids were challenged with RG7090 in the presence or absence of modified antisense oligonucleotides inhibiting CAR and/or PXR mRNA, showing a concentration-dependent Cyp2b mRNA induction only if CAR was not repressed. On the contrary, neither human liver spheroids nor cynomolgus monkeys exposed to RG7090 triggered CYP2B mRNA upregulation. Our data suggested RG7090 to be a rodent-specific CAR activator, and that CAR activation and its downstream processes were involved in the foci of altered hepatocytes formation detected in vivo. Furthermore, we demonstrated the potential of a new in vitro approach using liver spheroids and antisense oligonucleotides for CAR knockdown experiments, which could eventually replace in vivo investigations using CAR/PXR KO mice.


Asunto(s)
Imidazoles/farmacología , Piridinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores de Esteroides , Animales , Receptor de Androstano Constitutivo , Hepatocitos , Humanos , Hígado , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Organoides
4.
Toxicology ; 426: 152282, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31465819

RESUMEN

In a 2-year study the herbicide metazachlor (BAS 479H) was shown to significantly increase the incidence of liver tumours in female Wistar rats at a dietary level of 8000 ppm. As metazachlor is not a genotoxic agent, a series of in vivo and in vitro investigative studies were undertaken to elucidate the mode of action (MOA) for metazachlor-induced female rat liver tumour formation. Male and female Wistar rats were given diets containing 0 (control), 200 and 8000 ppm metazachlor for 3, 7, 14 and 28 days. The treatment of male rats with 200 and 8000 ppm metazachlor and female rats with 8000 ppm metazachlor resulted in significant increases in relative liver weight, which was associated with a centrilobular hepatocyte hypertrophy. Hepatocyte replicative DNA synthesis (RDS) was significantly increased in male rats given 8000 ppm metazachlor for 3 and 7 days and in female rats given 200 ppm metazachlor for 7-28 days and 8000 ppm metazachlor for 3-28 days. Significant increases in relative liver weight, centrilobular hepatocyte hypertrophy and hepatocyte RDS were also observed in male and female Wistar rats given and 500 ppm sodium phenobarbital (NaPB) for 3-28 days. The treatment of female Wistar rats with either 8000 ppm metazachlor for 7 days or with 500 ppm NaPB for 3 and 7 days resulted in the nuclear translocation of the hepatic constitutive androstane receptor (CAR). Treatment of male and female Wistar rats with 8000 ppm metazachlor for 14 days resulted in significant increases in hepatic microsomal total cytochrome P450 (CYP) content, CYP2B subfamily-dependent enzyme activities and mRNA levels, together with some increases in CYP3A enzyme activity and mRNA levels. The treatment of male Wistar rat hepatocytes with metazachlor (concentration range 0.5-50 µM) and NaPB (500 µM) for 4 days resulted in increased CYP2B enzyme activities and mRNA levels; with metazachlor and NaPB also producing significant increases in hepatocyte RDS levels. Studies were also performed with hepatocytes from male Sprague-Dawley wild type (WT) rats and CAR knockout (CAR KO) rats. While both treatment with metazachlor and NaPB for 4 days increased CYP2B enzyme activities and mRNA levels in WT rat hepatocytes, only minor effects were observed in CAR KO rat hepatocytes. Treatment with both metazachlor and NaPB only increased RDS in WT but not in CAR KO rat hepatocytes. The treatment of hepatocytes from two male human donors with 0.5-25 µM metazachlor or 500 µM NaPB for 4 days resulted in increases in CYP2B6 and CYP3A4 mRNA levels but had no effect on hepatocyte RDS. EGF as concurrently used positive control demonstrated the expected RDS response in all rat and human hepatocyte cultures. In conclusion, a series of in vivo and in vitro investigative studies have demonstrated that metazachlor is a CAR activator in rat liver, with similar properties to the prototypical CAR activator phenobarbital. A robust MOA for metazachlor-induced female rat liver tumour formation has been established. Based on the lack of effect of metazachlor on RDS in human hepatocytes, it is considered that the MOA for metazachlor-induced rat liver tumour formation is qualitatively not plausible for humans.


Asunto(s)
Acetamidas/toxicidad , Herbicidas/toxicidad , Neoplasias Hepáticas Experimentales/inducido químicamente , Animales , Células Cultivadas , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/efectos de los fármacos , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Replicación del ADN/efectos de los fármacos , Femenino , Técnicas de Inactivación de Genes , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hígado/patología , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Translocación Genética/efectos de los fármacos
5.
Toxicology ; 396-397: 23-32, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29425889

RESUMEN

Phenobarbital (PB), a constitutive androstane receptor (CAR) activator, produces liver tumours in rodents by a mitogenic mode of action involving CAR activation. In this study, the hepatic effects of sodium phenobarbital (NaPB) were compared in male C57BL/6J wild type (WT) mice and in humanized mice, where both the mouse CAR and pregnane X receptor (PXR) have been replaced by their human counterparts (hCAR/hPXR mice). Investigations were also performed in cultured male C57BL/6J and CD-1 mouse, male Sprague-Dawley rat and male and female human hepatocytes. The treatment of WT and hCAR/hPXR mice with 186-984 ppm NaPB in the diet for 7 days resulted in increased relative liver weight, hypertrophy and induction of cytochrome P450 (CYP) enzyme activities. Treatment with NaPB also produced dose-dependent increases in hepatocyte replicative DNA synthesis (RDS), with the effect being more marked in WT than in hCAR/hPXR mice. While the treatment of cultured C57BL/6J and CD-1 mouse, Sprague-Dawley rat and human hepatocytes with 100 and/or 1000 µM NaPB for 4 days induced CYP enzyme activities, increased RDS was only observed in mouse and rat hepatocytes. However, as a positive control, epidermal growth factor increased RDS in hepatocytes from all three species. In summary, although human hepatocytes are refractory to the mitogenic effects of NaPB, treatment with NaPB induced RDS in vivo in hCAR/hPXR mice, which is presumably due to the human CAR and PXR receptors operating in a mouse hepatocyte regulatory environment. As the response of the hCAR/hPXR mouse to the CAR activator NaPB differs markedly from that of human hepatocytes, the hCAR/hPXR mouse is thus not a suitable animal model for studies on the hepatic effects of nongenotoxic rodent CAR activators.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Fenobarbital/farmacología , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Receptores de Esteroides/efectos de los fármacos , Animales , Células Cultivadas , Receptor de Androstano Constitutivo , Citocromos/metabolismo , Replicación del ADN/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Hipnóticos y Sedantes/sangre , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Fenobarbital/sangre , Receptor X de Pregnano , Ratas , Ratas Sprague-Dawley
6.
Toxicology ; 293(1-3): 30-40, 2012 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-22239858

RESUMEN

In a prior 28-day dietary study in rats with 20 and 100 ppm K⁺ PFOS, activation of PPARα and CAR/PXR were concluded to be etiological factors in K⁺ PFOS-induced hepatomegaly and hepatic tumorigenesis. The objective of this study was to evaluate persistence/resolution of K⁺ PFOS-induced, liver-related effects in male Sprague Dawley rats following a 7-day dietary exposure to K⁺ PFOS at 20 or 100 ppm. Groups of 10 rats per treatment were observed on recovery Day(s) 1, 28, 56, and 84 following treatment. Changes consistent with hepatic PPARα and CAR/PXR activation noted on recovery Day 1 included: increased liver weight; decreased plasma cholesterol, alanine aminotransferase, and triglycerides; decreased liver DNA concentration and increased hepatocellular cytosolic CYP450 concentration; increased liver activity of acyl CoA oxidase, CYP4A, CYP2B, and CYP3A; increased liver proliferative index and decreased liver apoptotic index; decreased hepatocellular glycogen-induced vacuoles; increased centrilobular hepatocellular hypertrophy. Most effects resolved to control levels during recovery. Effects on plasma cholesterol, hepatocellular cytosolic CYP450 concentrations, liver apoptotic index, CYP3A, and centrilobular hepatocellular hypertrophy persisted through the end of the recovery period. Thyroid parameters (histology, apoptosis, and proliferation) were unaffected at all time points. Mean serum PFOS concentrations on recovery Day 1 were 39 and 140 µg/mL (20 ppm and 100 ppm K⁺ PFOS, respectively), decreasing to 4 and 26 µg/mL by recovery Day 84. Thus, hepatic effects in male rats resulting from K⁺ PFOS-induced activation of PPARα and CAR/PXR resolved slowly or were still present after 84-days following a 7-day dietary treatment, consistent with the slow elimination rate of PFOS.


Asunto(s)
Ácidos Alcanesulfónicos/farmacocinética , Ácidos Alcanesulfónicos/toxicidad , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Fluorocarburos/farmacocinética , Fluorocarburos/toxicidad , Hepatomegalia/inducido químicamente , Hígado/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinógenos/administración & dosificación , Carcinógenos/metabolismo , Proliferación Celular/efectos de los fármacos , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/metabolismo , Contaminantes Ambientales/farmacocinética , Contaminantes Ambientales/toxicidad , Fluorocarburos/administración & dosificación , Fluorocarburos/metabolismo , Semivida , Hepatomegalia/sangre , Hepatomegalia/metabolismo , Hepatomegalia/patología , Hígado/metabolismo , Hígado/patología , Masculino , PPAR alfa/metabolismo , Receptor X de Pregnano , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Glándula Tiroides/patología , Pruebas de Toxicidad Subaguda
7.
Toxicology ; 293(1-3): 16-29, 2012 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-22245121

RESUMEN

The present study investigated the potential role for activation of PPARα and CAR/PXR by potassium PFOS (K⁺ PFOS) with respect to the etiology of hepatic hypertrophy and hepatocellular adenoma in rats. Male Sprague-Dawley rats were fed K⁺ PFOS (20 or 100 ppm) for either 1, 7, or 28 days. Wyeth 14,643 (Wy 14,643, 50 ppm) and phenobarbital (PB, 500 ppm) were the controls for PPARα and CAR/PXR activation, respectively. Measurements included: plasma ALT, AST, cholesterol, triglycerides, and glucose; liver protein and DNA content; liver activities of palmitoyl CoA oxidase (ACOX), Cyp4A, CYP2B, and CYP3A; induction of liver CYP4A1, CYP2E1, CYP2B1/2, and CYP3A1 proteins (SDS-PAGE and Western blots); liver and thyroid microscopic histopathology, apoptotic index, and cell proliferation index. Terminal body weight was decreased by K⁺ PFOS (100 ppm) and Wy 14,643. All test-compound treatments increased liver weight. Plasma lipids were decreased by both PFOS and Wy 14,643. After treatment for 1 day, K⁺ PFOS (100 ppm), PB, and Wy 14,643 increased mean hepatic DNA concentration and total hepatic DNA, and total DNA remained elevated after treatment for 7 days and 28 days (PB and Wy 14,643 only). Hepatic P450 concentration was elevated after 7 and 28 days by K⁺ PFOS and by PB. K⁺ PFOS and Wy 14,643 increased liver activities of ACOX and CYP4A as well as increased liver CYP4A1 protein. By 28 days of treatment, K⁺ PFOS and PB increased liver activities of CYP2B and CYP3A as well as increased liver CYP2B1/2 and CYP3A1 proteins, and Wy 14,643 increased CYP2B enzyme activity to a slight extent. All test compounds increased the liver cell proliferative index and decreased the liver apoptotic index. No histological changes of the thyroid were noted; however, PB and WY increased thyroid follicular cell proliferation index (seven-day treatment only), while K⁺ PFOS did not. The thyroid follicular cell apoptotic index did not differ between groups. The hepatomegaly and hepatocellular adenoma observed after dietary exposure of Sprague-Dawley rats to K⁺ PFOS likely are due to the increased expression of xenosensor nuclear receptors PPARα and CAR/PXR. Given the markedly lower or absent response of human hepatocytes to the proliferative stimulus from activation of PPARα and CAR/PXR, the hepatocellular proliferative response from activation of these receptors by PFOS observed in rats is not expected to be of human relevance.


Asunto(s)
Adenoma de Células Hepáticas/inducido químicamente , Ácidos Alcanesulfónicos/toxicidad , Proliferación Celular/efectos de los fármacos , Fluorocarburos/toxicidad , Neoplasias Hepáticas/inducido químicamente , PPAR alfa/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Adenoma de Células Hepáticas/química , Adenoma de Células Hepáticas/metabolismo , Adenoma de Células Hepáticas/patología , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/análisis , Ácidos Alcanesulfónicos/farmacocinética , Animales , Apoptosis/efectos de los fármacos , Carcinógenos/administración & dosificación , Carcinógenos/análisis , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Receptor de Androstano Constitutivo , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/análisis , Contaminantes Ambientales/farmacocinética , Contaminantes Ambientales/toxicidad , Fluorocarburos/administración & dosificación , Fluorocarburos/análisis , Fluorocarburos/farmacocinética , Hepatomegalia/inducido químicamente , Hígado/química , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/química , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Proteínas de Neoplasias/metabolismo , Receptor X de Pregnano , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/patología , Pruebas de Toxicidad Subaguda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA