Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 165(8): 2771-86, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22029876

RESUMO

BACKGROUND AND PURPOSE: Drug candidates must be thoroughly investigated for their potential cardiac side effects. During the course of routine toxicological assessment, the compound RO5657, a CCR5 antagonist, was discovered to have the rare liability of inducing torsades de pointes (polymorphic ventricular arrhythmia) in normal, healthy animals. Studies were conducted to determine the molecular mechanism of this arrhythmia. EXPERIMENTAL APPROACH: Toxicological effects of repeat dosing were assessed in naïve monkeys. Cardiovascular effects were determined in conscious telemetry-implanted monkeys (repeat dosing) and anaesthetized instrumented dogs (single doses). Mechanistic studies were performed in guinea-pig isolated hearts and in cells recombinantly expressing human cardiac channels. KEY RESULTS: In cynomolgus monkeys, RO5657 caused a low incidence of myocardial degeneration and a greater incidence of ECG abnormalities including prolonged QT/QTc intervals, QRS complex widening and supraventricular tachycardia. In telemetry-implanted monkeys, RO5657 induced arrhythmias, including torsades de pointes and in one instance, degeneration to fatal ventricular fibrillation. RO5657 also depressed both heart rate (HR) and blood pressure (BP), with no histological evidence of myocardial degeneration. In the anaesthetized dog and guinea-pig isolated heart studies, RO5657 induced similar cardiovascular effects. RO5657 also inhibited Kv11.1 and sodium channel currents. CONCLUSIONS AND IMPLICATIONS: The molecular mechanism of RO5657 is hypothesized to be due to inhibition of cardiac sodium and Kv11.1 potassium channels. These results indicate that RO5657 is arrhythymogenic due to decreased haemodynamic function (HR/BP), decreased conduction and inhibition of multiple cardiac channels, which precede and are probably the causative factors in the observed myocardial degeneration.


Assuntos
Antagonistas dos Receptores CCR5 , Coração/efeitos dos fármacos , Compostos Orgânicos/farmacologia , Torsades de Pointes/induzido quimicamente , Animais , Pressão Sanguínea/efeitos dos fármacos , Carbamatos , Cães , Drogas em Investigação/efeitos adversos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Cobaias , Coração/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Humanos , Técnicas In Vitro , Macaca fascicularis , Masculino , Piperidinas , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Sódio/efeitos adversos , Canais de Sódio/fisiologia , Torsades de Pointes/fisiopatologia
2.
Toxicol Appl Pharmacol ; 257(1): 74-83, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21906609

RESUMO

Sunitinib, an oral tyrosine kinase inhibitor approved to treat advanced renal cell carcinoma and gastrointestinal stroma tumor, is associated with clinical cardiac toxicity. Although the precise mechanism of sunitinib cardiotoxicity is not known, both the key metabolic energy regulator, AMP-activated protein kinase (AMPK), and ribosomal S 6 kinase (RSK) have been hypothesized as causative, albeit based on rodent models. To study the mechanism of sunitinib-mediated cardiotoxicity in a human model, induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) having electrophysiological and contractile properties of native cardiac tissue were investigated. Sunitinib was cardiotoxic in a dose-dependent manner with an IC50 in the low micromolar range, observed by a loss of cellular ATP, an increase in oxidized glutathione, and induction of apoptosis in iPSC-CMs. Pretreatment of iPSC-CMs with AMPK activators AICAR or metformin, increased the phosphorylation of pAMPK-T172 and pACC-S79, but only marginally attenuated sunitinib mediated cell death. Furthermore, additional inhibitors of AMPK were not directly cytotoxic to iPSC-CMs up to 250 µM concentrations. Inhibition of RSK with a highly specific, irreversible, small molecule inhibitor (RSK-FMK-MEA) did not induce cytotoxicity in iPSC-CMs below 250 µM. Extensive electrophysiological analysis of sunitinib and RSK-FMK-MEA mediated conduction effects were performed. Taken together, these findings suggest that inhibition of AMPK and RSK are not a major component of sunitinib-induced cardiotoxicity. Although the exact mechanism of cardiotoxicity of sunitinib is not known, it is likely due to inhibition of multiple kinases simultaneously. These data highlight the utility of human iPSC-CMs in investigating the potential molecular mechanisms underlying drug-induced cardiotoxicity.


Assuntos
Indóis/toxicidade , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Pirróis/toxicidade , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Quinases Proteína-Quinases Ativadas por AMP , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Proteínas Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Sunitinibe
4.
Inflammopharmacology ; 12(1): 33-45, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15035777

RESUMO

Constitutive and inducible isoforms of nitric oxide synthase (NOS) catalyze the synthesis of nitric oxide (NO) from L-arginine in various tissues and in different pathophysiologic states. Short-term treatment with NOS inhibitors has been associated with pancreatic enzyme elevations and pancreatic acinar cell degeneration; however, long-term pancreatic effects of NOS inhibition are not known. The purpose of this study was to evaluate the subchronic pancreatic effects of L-nitro-arginine (LNA), a compound that preferentially inhibits constitutive NOS isoforms. LNA was administered orally at doses of 10 and 30 mg/kg per day to 6 female dogs/group for 4 weeks. To differentiate whether the pancreatic effects of LNA may be related to its arginine structure, an additional group was given L-arginine (L-Arg) at plasma concentrations similar to the high dose of LNA (30 mg/kg per day). Pancreatic effects were monitored by changes in serum levels of pancreatic enzymes at regular intervals and by microscopic examinations at the end of the study. Both LNA and L-Arg were systematically available throughout the 4-week study period. LNA produced dose-related elevations (1.3-10-fold above concurrent control) in serum levels of pancreatic enzymes (amylase, lipase and trypsin-like immunoreactivity) during the 4-week treatment period with peak elevations occurring during the first week. Histologic assessments of the pancreas conducted at the end of the 4-week dosing period were unremarkable. Additionally, LNA treatment resulted in reduction in heart rate (40%), gastric distension and gastric mucosal erosion and ulceration. No pancreatic, cardiac, or gastric effects were seen with L-Arg, indicating that above effects were likely due to NOS inhibition. Results of this study confirmed previous observations of acute pancreatic alterations following the inhibition of constitutive NOS isoforms. However, these pancreatic alterations appear to be only transient effects as elevations in serum enzymes declined over time and no structural acinar cell damage was seen after continuous treatment with LNA for 4 weeks, suggesting an adaptation to NOS inhibition over time.


Assuntos
Inibidores Enzimáticos/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Pâncreas/efeitos dos fármacos , Amilases/sangue , Animais , Arginina/farmacologia , Cães , Feminino , Frequência Cardíaca/efeitos dos fármacos , Lipase/sangue , Nitroarginina/farmacologia , Pâncreas/fisiologia , Tripsina/sangue
5.
J Toxicol Environ Health A ; 62(2): 127-41, 2001 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-11209821

RESUMO

Dieldrin-induced hepatocarcinogenesis, which is seen only in the mouse, apparently occurs through a nongenotoxic mechanism. Previous studies have demonstrated that dieldrin induces hepatic DNA synthesis in mouse, but not rat liver. A number of nongenotoxic hepatocarcinogens have been shown to increase hepatocyte nuclear ploidy following acute and subchronic treatment in rodents, suggesting that an induction of hepatocyte DNA synthesis may occur without a concomitant increase in cell division. The current study examined the effects of dieldrin on changes in hepatocyte DNA synthesis, mitosis, apoptosis, and ploidy in mouse liver (the sensitive strain and target tissue for dieldrin-induced carcinogenicity) and the rat liver (an insensitive species). Male F344 rats and B6C3F1 mice were treated with 0, 1, 3, or 10 mg dieldrin/kg diet and were sampled after 7, 14, 28, or 90 d on diet. Liver from mice fed 10 mg dieldrin/kg diet exhibited significantly increased DNA synthesis and mitosis at 14, 28, or 90 d on diet. In rats, no increase in DNA synthesis or mitotic index was observed. The apoptotic index in liver of mice and rats did not change over the 90-d study period. Exposure of mice to only the highest dose of dieldrin produced a significant increase in octaploid (8N) hepatocytes and a decrease in diploid (2N) hepatocytes, which were restricted primarily to centrilobular hepatocytes, with the periportal region showing little or no change from control. No changes in hepatocyte nuclear ploidy were observed in the rat. This study demonstrates that exposure to high concentrations of dieldrin is accompanied by increased nuclear ploidy and mitosis in mouse, but not rat, liver. It is proposed that the observed increase in nuclear ploidy in the mouse may reflect an adaptive response to dieldrin exposure.


Assuntos
Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dieldrin/efeitos adversos , Modelos Animais de Doenças , Exposição Ambiental/efeitos adversos , Hepatócitos/efeitos dos fármacos , Inseticidas/efeitos adversos , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/patologia , Ploidias , Animais , DNA/biossíntese , DNA/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Índice Mitótico , Ratos , Ratos Endogâmicos F344
6.
Toxicology ; 146(1): 15-22, 2000 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-10773359

RESUMO

Many nongenotoxic hepatocarcinogens can induce cell proliferation, and inhibit apoptosis and gap-junctional-intercellular communication (GJIC). GJIC, the movement of small molecules (less than 1.2 kD) through membrane channels, is important in regulating cellular homeostasis and differentiation. The inhibition of hepatic GJIC can increase cell proliferation and possibly, inhibit apoptosis. In this study, the relationship between hepatic GJIC, proliferation, and apoptosis was examined in rats treated for 7 days with tumor-promoting doses of the nongenotoxic hepatocarcinogens phenobarbital (PB; 800 ppm), pregnenolone-16alpha-carbonitrile (PCN; 1000 ppm), and Aroclor 1254 (PCB; 100 ppm). In addition, 3-methylcholanthrene (3MC) was included as a negative control. PB, PCN, and PCB increased parenchymal-cell proliferation and inhibited hepatic apoptosis, while no alteration in these growth parameters was observed in 3MC-treated rats. GJIC, as measured by fluorescent-dye transfer through intact liver, was decreased nearly 50% by PB, PCN, and PCB, yet no effect on GJIC was observed in liver from 3MC-treated rats. These data indicate that compounds that inhibit GJIC in liver may be nongenotoxic hepatocarcinogens, which occurs simultaneously during increased cell proliferation and inhibited apoptosis.


Assuntos
Carcinógenos/toxicidade , Comunicação Celular/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Fígado/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dextranos/química , Corantes Fluorescentes/química , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Isoquinolinas/química , Masculino , Metilcolantreno/toxicidade , Microscopia de Fluorescência , Fenobarbital/toxicidade , Carbonitrila de Pregnenolona/toxicidade , Antígeno Nuclear de Célula em Proliferação/análise , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Rodaminas/química , Organismos Livres de Patógenos Específicos
7.
Toxicology ; 143(2): 195-202, 2000 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-10755705

RESUMO

Propylthiouracil (PTU) or low-iodine diet (LID) treatment increases thyroid-follicular-cell proliferation, possibly by disrupting the movement of small molecules (< 1.2 kD) through membrane channels called gap junctions. Numerous tumor promoters and proliferative disease states exhibit inhibited gap-junctional-intercellular communication (GJIC) prior to the induction of cell proliferation, yet the association between GJIC and apoptosis is unclear. In the present study, we used an ex vivo method to examine whether GJIC is inhibited in the thyroid of PTU- or LID-treated rats. In addition, the effect of these models of hypothyroidism on thyroid-follicular-cell proliferation and apoptosis was examined to determine the association between GJIC and cell homeostasis. After 14 days of treatment of either PTU or LID (plus 1% KClO4 in the drinking water), serum tri-iodothyronine (T3) and thyroxine, (T4) was decreased to nearly undetectable levels and serum TSH was increased in PTU- and LID-treated rats. At the same time point, GJIC was decreased 30-35% in PTU- and LID-treated rats while thyroid-follicular-cell proliferation increased nearly threefold in both treatment groups. Interestingly, apoptosis increased twofold in both hypothyroid treatment groups. These data suggest that PTU or LID treatment inhibit thyroid GJIC during a state of increased thyroid-follicular-cell proliferation and apoptosis. While the increase in proliferation was anticipated, the paradoxical increase in apoptosis during decreased GJIC in thyroid-follicular cells warrants further examination.


Assuntos
Antitireóideos/farmacologia , Comunicação Celular/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Iodo/deficiência , Propiltiouracila/farmacologia , Glândula Tireoide/citologia , Glândula Tireoide/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dieta , Hipotireoidismo/fisiopatologia , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Tireotropina/sangue , Tiroxina/sangue , Tri-Iodotironina/sangue
8.
Toxicol Appl Pharmacol ; 161(1): 23-33, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10558920

RESUMO

The metabolism of CCl(4) initiates the peroxidation of polyunsaturated fatty acids producing alpha,beta-unsaturated aldehydes, such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA). The facile reactivity of these electrophilic aldehydic products suggests they play a role in the toxicity of compounds like CCl(4). To determine the rate at which CCl(4)-initiated lipid peroxidation results in the formation of 4-HNE and/or MDA hepatic protein adducts, rats were given an intragastric dose of CCl(4) (1.0 ml/kg) and euthanized 0-72 h after administration. Rabbit polyclonal antisera directed toward 4-HNE- or MDA-protein epitopes were employed in immuno-histochemical and immuno-precipitation/Western analyses to detect 4-HNE and MDA-protein adducts in paraffin-embedded liver sections and liver homogenates. As early as 6 h post CCl(4) exposure, 4-HNE and MDA adducts were detected immuno-histochemically in hepatocytes localized to zone 2 of the hepatic acinus. Liver injury was progressive to 24 h as lipid peroxidation and hepatocellular necrosis increased. The hallmark of CCl(4) hepatotoxicity, zone 3 necrosis, was observed 24 h after CCl(4) administration and immuno-positive hepatocytes were observed in zone 2 as well as zone 3. Immuno-positive cells were no longer visible by 36 to 72 h post CCl(4) administration. From 6 to 48 h after CCl(4) administration, at least four adducted proteins were immuno-precipitated from liver homogenates with the anti-MDA or anti-4HNE serum, which corresponded to molecular weights of 80, 150, 205, and greater than 205 kDa. These results demonstrate that 4-HNE and MDA alkylate specific hepatic proteins in a time-dependent manner, which appears to be associated with hepatocellular injury following CCl(4) exposure.


Assuntos
Aldeídos/metabolismo , Tetracloreto de Carbono/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Malondialdeído/metabolismo , Proteínas/metabolismo , Alanina Transaminase/metabolismo , Aldeídos/análise , Aldeídos/imunologia , Alquilação/efeitos dos fármacos , Animais , Western Blotting , Tetracloreto de Carbono/administração & dosagem , Ácidos Graxos Insaturados/metabolismo , Imuno-Histoquímica , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Masculino , Malondialdeído/análise , Malondialdeído/imunologia , Óleo Mineral , Peso Molecular , Necrose , Testes de Precipitina , Proteínas/análise , Proteínas/química , Proteínas/imunologia , Ratos , Substâncias Reativas com Ácido Tiobarbitúrico/análise , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Fatores de Tempo
9.
Toxicol Lett ; 106(2-3): 143-50, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10403658

RESUMO

Exposure to certain UDP-glucuronosyltransferase (UDP-GT) inducers leads to follicular cell hyperplasia, and ultimately thyroid gland tumors. These compounds decrease thyroid hormones, which increases serum concentrations of thyroid stimulating hormone (TSH). This induction of TSH enhances thyroid-follicular cell proliferation. In addition, treatment with classical goitrogenic compounds, such as propylthiouracil (PTU) and methimazole (MMI), induces TGF-beta1 in thyroid-follicular cells, presumably through increased TSH. In other tissues, increases in TGF-beta1 induce apoptosis, a particular form of programmed cell death. In this experiment, we sought to determine whether the UDP-GT inducers, phenobarbital (PB) and pregnenolone-16alpha-carbonitrile (PCN) modulate thyroid-follicular cell apoptosis. If so, are the induction of apoptosis and TGF-beta1 possibly linked? An additional group of rats treated with the thyroid goitrogen, PTU was included. Male Sprague-Dawley rats were treated with thyroid hormone disrupting doses of PB, PCN, or PTU for 3, 7, 14, 21, 28, 45, or 90 days. In this study, PTU treatment increased apoptosis and TGF-beta1 immunoreactive thyroid-follicular cells. PTU treatment of rats produced both a large increase number of TGF-beta1-positive cells (detected by immunohistochemistry), and apoptotic thyroid-follicular cells (detected by morphology). In PB- and PCN-treated rats, a moderate increase in apoptosis coincided with similar increases in TGF-beta1 immunoreactive thyroid-follicular cells. In summary, PB and PCN increase apoptosis and the percentage of TGF-beta1 positive thyroid-follicular cells. Thus, treatment with UDP-GT-inducing chemicals may increase the expression of TGF-beta1 and apoptosis in the thyroid to compensate for the thyroid hypertrophy and hyperplasia.


Assuntos
Apoptose/efeitos dos fármacos , Glucuronosiltransferase/biossíntese , Fenobarbital/toxicidade , Carbonitrila de Pregnenolona/toxicidade , Glândula Tireoide/efeitos dos fármacos , Fator de Crescimento Transformador beta/análise , Animais , Indução Enzimática/efeitos dos fármacos , Hiperplasia , Masculino , Ratos , Ratos Sprague-Dawley , Glândula Tireoide/patologia , Fator de Crescimento Transformador beta/imunologia
10.
Xenobiotica ; 29(6): 583-93, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10426557

RESUMO

1. The effect of partial hepatectomy on the expression of sulphotransferase (SULT) mRNA was studied. SULTs fall into two distinct classes based on substrate preference: phenol SULT1 family (SULT1A1, SULT1B1, SULT1C1 and SULT1E2) and hydroxysteroid SULT2 family (SULT20/21, SULT40/41 and SULT60). 2. Hepatic expression of SULT mRNA was analysed in the sham-hepatectomised rat (sham) and in the partially hepatectomised (PH) rat at various times after PH. Northern-blot analysis with [alpha-32P]dATP-labelled oligonucleotide probes specific for individual SULT mRNAs was used to monitor hepatic SULT mRNA expression. In general, SULT mRNAs underwent a decrease in expression after PH and the magnitude of decrease was dependent on the SULT isoform. 3. The decrease in SULT mRNA expression was resolved and even induced (SULT40/41 in the female rat) by 10-30 days after PH. Of the phenol SULT isoforms, both SULT1C1 and SULT1E2 mRNAs were significantly decreased by 18-24 h after PH in the male rat. The other phenol SULTs (SULT1A1 and SULT1B1) tended to decrease in the male rat after PH, but the decreases were not statistically significant. Expression of SULT20/21 mRNA was decreased in the female rat (80% at 24 h) and fully recovered by 10 days. SULT40/41 mRNA tended to decrease after PH; however, the decrease was not statistically significant. SULT 60 mRNA was decreased from 24 to 96 h after PH. 4. Thus, during the period of rapid liver growth that occurs after partial hepatectomy, SULT mRNA expression is decreased. The phenomenon of decreased SULT mRNA expression is similar to other states of rapid liver growth (e.g. cancer tissue and young animals) in which expression of SULT enzymes is characteristically low.


Assuntos
Hepatectomia , Regeneração Hepática/fisiologia , Fígado/enzimologia , RNA Mensageiro/metabolismo , Sulfotransferases/genética , Animais , Sequência de Bases , Northern Blotting , Sondas de DNA/química , Feminino , Fígado/cirurgia , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
11.
J Toxicol Environ Health A ; 53(6): 479-92, 1998 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-9537283

RESUMO

The effect of vitamin E on dieldrin-induced hepatic focal lesion growth in male B6C3F1 mice previously treated with diethylnitrosamine (DEN) was investigated. After hepatic focal lesions were formed, mice were placed into one of the following treatment groups: Group 1, 50 mg vitamin E/kg diet (control NIH-07 diet); Group 2, 10 mg dieldrin/kg NIH-07 diet; Group 3, 10 mg dieldrin and 450 mg vitamin E/kg NIH-07 diet; and Group 4, 450 mg vitamin E/kg NIH-07 diet. Mice were killed and necropsied after 30 and 60 d of dietary treatment. The effect of treatment on lesion growth was examined by measuring the number of focal lesions per liver and the relative hepatic focal lesion volume. In addition, the possible cellular mechanism of focal hepatocyte growth was investigated by examining both focal DNA synthesis and apoptosis. Dieldrin treatment alone (Group 2) increased the focal lesion volume, focal lesion number, and focal lesion labeling index. Supplementation with vitamin E (Group 3) blocked this effect. Vitamin E supplementation to the diet alone (Group 4) also enhanced focal lesion growth and increased the number of lesions per liver, the relative focal volume, and the labeling index in hepatic focal lesions. Interestingly, vitamin E supplementation inhibited apoptosis in normal liver but did not produce an observable decrease in apoptosis in hepatic focal lesions. The present study showed that dieldrin (Group 2) or vitamin E supplementation alone (Group 4) promoted the growth of hepatic focal lesions in mice. However, when vitamin E is supplemented to dieldrin-fed mice (Group 3), there is an inhibition of hepatic focal lesion growth.


Assuntos
Carcinógenos/toxicidade , Dieldrin/toxicidade , Inseticidas/toxicidade , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Lesões Pré-Cancerosas/tratamento farmacológico , Vitamina E/uso terapêutico , Animais , Apoptose , Doença Hepática Induzida por Substâncias e Drogas , Fígado/patologia , Hepatopatias/patologia , Masculino , Camundongos , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/patologia
12.
Environ Health Perspect ; 106 Suppl 1: 289-95, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9539021

RESUMO

Oxidative stress results when the balance between the production of reactive oxygen species (ROS) overrides the antioxidant capability of the target cell; oxidative damage from the interaction of reactive oxygen with critical cellular macromolecules may occur. ROS may interact with and modify cellular protein, lipid, and DNA, which results in altered target cell function. The accumulation of oxidative damage has been implicated in both acute and chronic cell injury including possible participation in the formation of cancer. Acute oxidative injury may produce selective cell death and a compensatory increase in cell proliferation. This stimulus may result in the formation of newly initiated preneoplastic cells and/or enhance the selective clonal expansion of latent initiated preneoplastic cells. Similarly, sublethal acute oxidative injury may produce unrepaired DNA damage and result in the formation of new mutations and, potentially, new initiated cells. In contrast, sustained chronic oxidative injury may lead to a nonlethal modification of normal cellular growth control mechanisms. Cellular oxidative stress can modify intercellular communication, protein kinase activity, membrane structure and function, and gene expression, and result in modulation of cell growth. We examined the role of oxidative stress as a possible mechanism by which nongenotoxic carcinogens may function. In studies with the selective mouse liver carcinogen dieldrin, a species-specific and dose-dependent decrease in liver antioxidant concentrations with a concomitant increase in ROS formation and oxidative damage was seen. This increase in oxidative stress correlated with an increase in hepatocyte DNA synthesis. Antioxidant supplementation prevented the dieldrin-induced cellular changes. Our findings suggest that the effect of nongenotoxic carcinogens (if they function through oxidative mechanisms) may be amplified in rodents but not in primates because of rodents' greater sensitivity to ROS. These results and findings reported by others support a potential role for oxidative-induced injury in the cancer process specifically during the promotion stage.


Assuntos
Carcinógenos/toxicidade , Dieldrin/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Estresse Oxidativo , Animais , Dano ao DNA , Humanos , Neoplasias Hepáticas Experimentais/metabolismo , Camundongos
13.
Toxicol Sci ; 46(1): 31-7, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9928666

RESUMO

Exposure to certain microsomal enzyme inducers that increase UDP-glucuronosyltransferase (UDP-GT) activity decreases thyroid hormone levels, which may lead to a subsequent increase in thyroid-stimulating hormone (TSH). This elevation of serum TSH has many effects on the thyroid, including increasing thyroid follicular cell proliferation, leading to hyperplasia. While induction of UDP-GT activity decreases thyroid hormone levels by enhancing biotransformation and subsequent biliary secretion, only certain UDP-GT inducers exhibit the ability to increase serum TSH levels. For example, phenobarbital (PB) and pregnenolone-16alpha-carbonitrile (PCN) increase serum levels of TSH, while 3-methylcholanthrene (3MC) and Aroclor 1254 (PCB) do not. Increased serum TSH concentration also enhances thyroid gland expression of TGF-beta1, an anti-proliferative, pro-apoptotic protein. In a previous study in our laboratory, rats were treated for various times (up to 90 days) with PB and PCN, which increased TGF-beta1 protein and apoptosis. The present study was designed to examine the dose-response effect of TSH-increasing (PB and PCN) and nonincreasing (3MC and PCB) UDP-GT inducers on apoptosis and TGF-beta1. PB and PCN, UDP-GT inducing compounds which increase serum TSH, increased the percentage of TGF-beta1-positive follicular cells and increased apoptosis. In contrast, UDP-GT inducers that did not increase TSH (3MC and PCB) did not alter cell death or TGF-beta production. These data suggest that the increase of TGF-beta by TSH may serve to regulate the growth of hyperplastic thyroid.


Assuntos
Apoptose/efeitos dos fármacos , Glucuronosiltransferase/biossíntese , Glândula Tireoide/citologia , Fator de Crescimento Transformador beta/biossíntese , Animais , Relação Dose-Resposta a Droga , Indução Enzimática , Imuno-Histoquímica , Masculino , Metilcolantreno/toxicidade , Fenobarbital/toxicidade , Carbonitrila de Pregnenolona/toxicidade , Ratos , Ratos Sprague-Dawley , Glândula Tireoide/efeitos dos fármacos
14.
Carcinogenesis ; 18(8): 1511-9, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9276624

RESUMO

The effect of rotenone treatment on [4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio] acetic acid (WY-14,643) hepatic lesion growth in male B6C3F1 mice was investigated. Following induction of hepatic focal lesions by diethylnitrosamine (DEN) 35 mg/kg twice a week for 8 weeks, mice were placed into one of the four treatment groups: group I, control NIH-07 diet (control diet), group II, rotenone (600 mg/kg diet), group III NIH-07 diet containing WY-14,643 (1000 mg/kg diet), and group IV, NIH-07 diet containing WY-14,643 (1000 mg/kg diet) and rotenone (600 mg/ kg diet). Mice were killed after 30 and 60 days of dietary treatment. The effect of treatment with WY-14,643 and rotenone on hepatic lesion growth was examined by estimating the number of focal lesions per liver and the relative volume of focal lesions. WY-14,643 (group III) increased both the number and the volume of focal lesions. In particular, an increase in number and volume of basophilic lesions was seen. Co-treatment with WY-14,643 and rotenone (group IV) decreased both the number and the volume of the total number of focal lesions and basophilic foci compared with WY-14,643 treatment alone (group II). Alterations in the growth of hepatic focal lesions was further investigated by examining DNA synthesis and apoptosis within individual lesions. WY-14,643 (group III) treatment increased the DNA synthetic labeling index in all foci. Co-treatment of rotenone and WY-14,643 (group IV) decreased focal DNA synthesis and mitosis and increased the incidence of apoptotic hepatocytes. These data suggest that rotenone's ability to inhibit WY-14,643-induced hepatic focal lesion growth was mediated through a decrease in hepatic focal proliferation and an increase in focal apoptosis.


Assuntos
Carcinógenos/antagonistas & inibidores , Neoplasias Hepáticas/prevenção & controle , Pirimidinas/antagonistas & inibidores , Rotenona/farmacologia , Adenoma/tratamento farmacológico , Adenoma/patologia , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Bromodesoxiuridina/análise , DNA/biossíntese , DNA/efeitos dos fármacos , Dietilnitrosamina , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos , Microcorpos/efeitos dos fármacos , Microcorpos/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Oxirredução/efeitos dos fármacos
15.
Ann Clin Lab Sci ; 27(3): 196-209, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9142372

RESUMO

The production of reactive oxygen species (ROS) by toxic chemicals has been implicated in acute and chronic disease states, including cancer. This increase in cellular ROS can lead to a state of oxidative stress. Many compounds selectively induce hepatic tumors in mice but not rats. The mechanism for the induction of hepatic cancer by these compounds and the observed species selectivity of this effect are not known but may be related to the induction of oxidative stress. Dieldrin is one such compound and is used in the present study to characterize the relationship between oxidative stress and the observed selective hepatotoxicity of dieldrin in mice. It was found that dieldrin induced oxidative stress in the mouse but not the rat, and the observed oxidative stress correlated with the induction of DNA S-phase synthesis. This evidence suggests that the induction of oxidative stress may be a mechanism by which dieldrin and other mouse specific compounds selectively induce their hepatic toxic effects in mice.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Dieldrin/toxicidade , Estresse Oxidativo , Animais , Antioxidantes , Carcinógenos/toxicidade , Fígado/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Masculino , Malondialdeído/metabolismo , Camundongos , Microssomos Hepáticos/metabolismo , Mutagênicos/toxicidade , Oxidantes , Ratos , Ratos Endogâmicos F344 , Espécies Reativas de Oxigênio/metabolismo
16.
Toxicol Appl Pharmacol ; 143(2): 380-7, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9144454

RESUMO

The effect of DL-alpha-tocopherol acetate (vitamin E) on hepatic focal lesion growth in male B6C3F1 mice previously treated with diethylnitrosamine (DEN) was investigated. After hepatic focal lesions were formed, mice were placed into one of the following dose groups: 0 mg vitamin E/kg NIH-07 diet, 50 mg vitamin E/kg NIH-07 diet (control diet), 250 mg vitamin E/kg NIH-07 diet, and 450 mg vitamin E/kg NIH-07 diet. Mice were euthanized after either 30 or 60 days of dietary treatment. In normal (nonlesion) liver, vitamin E deficiency (0 mg/kg diet) increased hepatic DNA synthesis. In addition, vitamin E supplementation (450 mg/kg diet) decreased the incidence of hepatic apoptosis, while vitamin E deficiency (0 mg/kg diet) increased the incidence of hepatic apoptosis. The effect of vitamin E-induced lesion growth was examined by measuring the number of focal lesions per liver and the relative focal lesion volume. High-dose vitamin E supplementation (450 mg/kg diet) appeared to enhance the growth of hepatic focal lesions. In particular, basophilic lesions appeared to be the most sensitive to high-dose vitamin E modulation (450 mg/kg diet) as evidenced by increased number, volume, and labeling index of hepatic focal lesions. Vitamin E deficiency also appeared to enhance the growth of hepatic focal lesions, though to a lesser extent than vitamin E supplementation (450 mg/kg diet). In the present study, both vitamin E supplementation (450 mg/kg diet) and deficiency (0 mg/kg diet) appeared to enhance focal lesion growth albeit neither treatment enhanced lesion growth as dramatically as known nongenotoxic hepatocarcinogens (e.g., phenobarbital and dieldrin). The data presented here suggest that oxidative stress in focal hepatocytes may be a component of the liver tumor promotion process.


Assuntos
Neoplasias Hepáticas Experimentais/induzido quimicamente , Fígado/efeitos dos fármacos , Vitamina E/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Carcinógenos/toxicidade , Contagem de Células , Divisão Celular/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , Dietilnitrosamina/toxicidade , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Deficiência de Vitamina E/metabolismo
17.
Carcinogenesis ; 17(8): 1657-64, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8761422

RESUMO

The effects of dietary restriction on the growth of hepatic focal lesions in phenobarbital (PB) promoted mice were examined. Dietary restriction which can inhibit many age-related diseases in rodents including hepatic cancer also decreases cell proliferation and increases apoptosis in the liver. In contrast, PB, a non-genotoxic rodent hepatocarcinogen, enhances the growth of hepatic focal lesions in mice and rats by increasing cell proliferation and inhibiting apoptosis. The present study examined the impact of dietary restriction on PB-induced hepatic tumor promotion. Focal lesions were produced by diethylnitrosamine (DEN) treatment (35 mg DEN/kg body weight injections, twice per week for 8 weeks). After lesions were produced, mice were placed into one of the following four groups: NIH-07 control diet/no PB (group 1); NIH-07 diet/500 mg PB per liter of drinking water (group 2); dietary restricted NIH-07 diet/no PB (group 3); and dietary restricted NIH-7 diet/ 500 mg PB per liter of drinking water (group 4). In this study, PB (500 mg/l) treatment to ad libitum-fed mice (group 2) enhanced focal lesion volume, number, and labeling index compared with group 1. In addition, PB treatment (group 2) inhibited apoptosis in normal and focal hepatocytes compared with untreated control mice (group 1). In contrast, in dietary restricted mice treated with PB (group 4) a significantly lower focal lesion volume, number and labeling index were seen compared with the ad libitum-fed/PB treatment group (group 2). PB treatment in dietary restricted mice (group 4) did not inhibit focal apoptosis, in fact, the incidence of focal apoptosis was increased in these mice compared with ad libitum and PB-treated mice (group 2). In dietary restricted mice treated with PB (group 4), the ability of PB to promote the growth of preneoplastic focal lesions was inhibited. These results show that dietary restriction can ablate the tumor promotional effects of PB in hepatic focal lesions and suggest that inhibition of focal lesion DNA synthesis and enhancement of apoptosis may be a mechanism for this effect.


Assuntos
Ingestão de Energia , Neoplasias Hepáticas Experimentais/prevenção & controle , Animais , Apoptose , Carcinógenos , Divisão Celular , Dietilnitrosamina , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos
18.
Carcinogenesis ; 17(7): 1403-9, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8706241

RESUMO

The effect of cessation of phenobarbital and dieldren treatment on hepatic focal lesion growth in male B6C3F1 mice was investigated. Following induction of lesions by diethylnitrosamine, mice were placed on control NIH-07 diet (control diet) or NIH-07 diet containing either dieldrin (10.0 mg/kg diet) or phenobarbital (500 mg/kg diet). Mice were sacrificed after 30 and 60 days of dietary treatment. Two additional groups of mice were fed either the dieldren- or phenobarbital-containing diet for 30 days followed by feeding of NIH-07-only diet for an additional 30 days. The effect of treatment and removal of dieldrin or phenobarbital on lesion growth was examined by measuring both the number of focal lesions per liver and the relative volume of focal lesions. In addition, the rate of cell proliferation and programmed cell death in focal lesion growth was investigated by examining DNA synthesis and apoptosis in the focal lesions. Dietary dieldrin or phenobarbital increased the number of focal lesions and the focal lesion volume. In both dieldrin- and phenobarbital-treated mice, an increased number of eosinophilic lesions were seen. The focal lesion volume was increased in both eosinophilic and basophilic lesions. Dieldrin and phenobarbital treatment also increased the DNA synthetic labeling index in both eosinophilic and basophilic lesions. Removal of dieldrin or phenobarbital from the diet after 30 days of promoter treatment decreased the total number and volume of hepatic focal lesions. The labeling index of the focal lesions was also decreased in these mice. At the terminal sacrifice, the percentage of apoptotic cells in focal lesions was higher in mice fed dieldrin- or phenobarbital-containing diets for the entire 60 days than in mice returned to control diet for the last 30 days. Eosinophilic lesions were more dependent on the presence of a promoting stimulus than the basophilic lesions. These data indicate that induction and maintenance of the growth of some preneoplastic lesions in the mouse may be dependent upon continuous tumor promoter treatment.


Assuntos
Carcinógenos/toxicidade , Dieldrin/toxicidade , Neoplasias Hepáticas/patologia , Fígado/patologia , Fenobarbital/toxicidade , Lesões Pré-Cancerosas/patologia , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Dieldrin/administração & dosagem , Dieta , Dietilnitrosamina , Eosinófilos/patologia , Fígado/efeitos dos fármacos , Neoplasias Hepáticas/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Fenobarbital/administração & dosagem , Lesões Pré-Cancerosas/induzido quimicamente
19.
Carcinogenesis ; 17(6): 1243-50, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8681438

RESUMO

Chronic exposure to a number of chlorinated pesticides, including dieldrin, results in an increased incidence and/or multiplicity of hepatocellular neoplasia in mice, with no such effect in similarly treated rats. One possible explanation of this observed selective carcinogenicity is species-specific hepatic tumor promotion. In the present study we examined the dose-response effect of dieldrin (at several doses) on focal lesion growth (tumor promotion), hepatocyte apoptosis and DNA synthesis in rat and mouse liver. Preneoplastic focal hepatic lesions were produced by diethylnitrosamine (DEN). After the lesions developed, mice and rats were placed into one of the following dose groups: control (NIH-07 diet) or 0.1, 1.0 or 10.0 mg dieldrin/kg diet. Increased focal lesion volume, number of foci per liver and focal DNA synthetic labeling index were observed in 10 mg dieldrin/kg diet-treated mice, but not in similarly treated rats. Dieldrin at dietary concentrations of 0.1 and 1.0 mg/kg diet produced an increase in the number of preneoplastic lesions (0.1 mg/kg diet at 7 days only) and focal volume (0.1 mg/kg diet at 7 and 30 days, 1.0 mg/kg diet at 30 days), but these concentrations did not increase focal DNA labeling index. At dietary concentrations of 0.1, 1.0 and 10 mg dieldrin/kg diet no significant change in lesion percent volume, number of preneoplastic lesions per liver or preneoplastic lesion DNA labeling index was seen in treated rats compared with control rats. Apoptosis, a form of programed cell death, was not decreased in foci by any concentration of dieldrin in either rats or mice. Thus our results suggest that dieldrin may function as a mouse-specific tumor promoter through increased lesion DNA synthesis.


Assuntos
Dieldrin/toxicidade , Inseticidas/toxicidade , Neoplasias Hepáticas Experimentais/induzido quimicamente , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Carcinógenos , Divisão Celular/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Dietilnitrosamina , Relação Dose-Resposta a Droga , Fígado/anatomia & histologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Tamanho do Órgão/efeitos dos fármacos , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/metabolismo , Ratos , Ratos Endogâmicos F344 , Especificidade da Espécie
20.
Carcinogenesis ; 17(5): 947-54, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8640942

RESUMO

Phenobarbital (PB), a non-genotoxic hepatocarcinogen in rodents, has been studied extensively but its mechanism of carcinogenic action is unclear. PB appears to function as a tumor promoter by selectively inducing the growth of preneoplastic hepatocytes. In the present study, the comparative effects of PB at tumor-promoting and non-promoting doses were examined in male B6C3F1 mice and male F344 rats. In addition, the mechanism by which PB produced the selective induction of preneoplastic cell growth (increased DNA synthesis/cell proliferation and/or decreased apoptosis) was investigated. Preneoplastic focal lesions were produced using diethylnitrosamine (DEN). After the lesions were histologically apparent, mice and rats were fed PB (10, 100, or 500 mg/kg NIH-07 diet) or control diet and sampled after 7, 30 and 60 days of treatment In both mice and rats, 100 and 500 mg PB/kg increased the number and the relative volume of focal lesions. In rats and mice, 10 mg PB/kg did not enhance focal lesion growth. The preneoplastic lesions that clonally expanded due to phenobarbital treatment were predominantly eosinophilic in appearance. In addition, DNA synthesis in focal hepatocytes was significantly increased in the 100 and 500 mg PB/kg diet. In PB-treated mice and rats, there also was a significant decrease in the rates of apoptosis in focal hepatocytes. Therefore, our data showed that PB at doses of 100 and 500 mg/kg diet promoted focal hepatic lesion growth both by increasing DNA synthesis and cell proliferation and by decreasing the rate of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , DNA/biossíntese , Neoplasias Hepáticas Experimentais/induzido quimicamente , Fenobarbital/toxicidade , Lesões Pré-Cancerosas/induzido quimicamente , Animais , Dietilnitrosamina , Relação Dose-Resposta a Droga , Masculino , Camundongos , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...